Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Oncology Letters
Join Editorial Board Propose a Special Issue
Print ISSN: 1792-1074 Online ISSN: 1792-1082
Journal Cover
January-2026 Volume 31 Issue 1

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
January-2026 Volume 31 Issue 1

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Progress in the treatment of esophageal neuroendocrine carcinoma (Review)

  • Authors:
    • Jie Shi
    • Bin He
    • Heng Di Zhang
    • Chao Yang Jiang
    • Ling Zhang
  • View Affiliations / Copyright

    Affiliations: Department of Oncology, The General Hospital of Western Theater Command of The Chinese People's Liberation Army, Chengdu, Sichuan 610083, P.R. China, Department of Ophthalmology, The General Hospital of Western Theater Command of The Chinese People's Liberation Army, Chengdu, Sichuan 610083, P.R. China
    Copyright: © Shi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 14
    |
    Published online on: October 31, 2025
       https://doi.org/10.3892/ol.2025.15367
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Esophageal neuroendocrine carcinoma (ENEC) is a rare and highly aggressive gastrointestinal malignancy with a markedly worse prognosis compared with other pathological types of esophageal cancer. The present study aimed to provide a systematic review of the pathological features, diagnostic strategies and advances in stratified treatment of ENEC, with a focus on current therapeutic approaches. The management of ENEC requires a multimodal approach. Among these modalities, surgery remains the cornerstone for achieving long‑term survival. For patients with initially unresectable disease, neoadjuvant therapy can convert cases to a resectable status. Additionally, combined chemoradiotherapy has been demonstrated to markedly improve survival rates. Beyond conventional treatments, the potential of targeted therapy in combination with chemotherapy has been suggested, and the synergy between immune checkpoint inhibitors and either radiotherapy or targeted drugs has achieved long‑term remission in certain cases.

Introduction

Neuroendocrine neoplasms (NENs) are tumors with neuroendocrine differentiation and expression of neuroendocrine markers, which can occur in various organs; however, they most commonly in the gastrointestinal tract, accounting for 55–70% of all NEN cases (1). Esophageal neuroendocrine carcinoma (ENEC) is a rare subtype within digestive tract NENs, comprising only 0.4–1% of gastroenteropancreatic NENs. ENEC is highly aggressive, prone to widespread metastasis and has a markedly worse prognosis compared with other common types of esophageal cancer, such as squamous cell carcinoma and adenocarcinoma (2).

Epidemiological studies have indicated that ENEC is more common in middle-aged and elderly men, and that it shows geographical clustering with higher incidence rates in certain regions, such as East Asia (including in China and Japan) (2,3). The risk factors for ENEC partially overlap with those of esophageal squamous cell carcinoma (ESCC), such as smoking, alcohol consumption, and a diet characterized by the frequent consumption of foods and beverages at very high temperatures (>65°C has been classified as probably carcinogenic to humans by the International Agency for Research on Cancer) (4), which may induce DNA damage and epigenetic changes in the esophageal mucosa (5,6). Certain patients with ENEC also present with Barrett's esophagus, which suggests that chronic inflammation might drive neuroendocrine differentiation through abnormal proliferative signaling pathways (7).

ENEC exhibits high biological heterogeneity. Early studies have suggested that it originates from amine precursor uptake and decarboxylation cells in the esophageal mucosa, derived from the neuroectoderm (8,9). However, more recent research has proposed that it likely originates from pluripotent basal epithelial stem cells in the esophagus, which can differentiate into squamous or glandular epithelium under normal conditions but may aberrantly differentiate into NEC under epigenetic or microenvironmental pressure, forming mixed tumors (10).

The present review particularly focuses on ENEC due to its distinct clinicopathological features and highly aggressive biological behavior. This focused approach is predicated on the distinct clinicopathological and molecular features of ENEC, which are justified by the fundamental differences between ENEC and other gastrointestinal NECs, particularly gastric NEC (GNEC). First, ENEC and GNEC possess unique molecular profiles; ENECs are characterized by a high frequency of co-mutations in tumor protein 53 and retinoblastoma gene 1, resembling small cell lung cancer, whereas GNECs exhibit considerable heterogeneity with alterations in genes such as low-density lipoprotein receptor-related protein 1B and dysregulation of pathways such as Wnt/β-catenin (11). Second, they arise from different epidemiological origins; ENEC shares strong associations with risk factors for ESCC (for example, smoking and alcohol) (12), whereas GNEC is often associated with chronic atrophic gastritis analogous to gastric adenocarcinoma (13). Notably, ENEC is associated with a markedly worse prognosis compared with GNEC, as confirmed by large-scale database analyses (3). Current evidence for the management of ENEC is often extrapolated from small cell lung cancer or aggregated with other NECs, underscoring the need for this particular synthesis of ENEC-specific data to provide clinicians with a nuanced overview of contemporary management and potential future directions.

While the seminal review by Ma et al (1) provided a key foundation to understand ENEC, the subsequent 8 years (2017–2025) have witnessed a paradigm shift in its management, which forms the core contribution of current research. The present review synthesizes these recent advances, which include the successful application of immune checkpoint inhibitors (for example, nivolumab and camrelizumab) (14–16), the emergence of combination immunotherapy strategies targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein-1 (PD-1) (17,18), and novel regimens combining anti-angiogenic tyrosine kinase inhibitors (TKIs) with immunotherapy (for example, anlotinib plus camrelizumab) (19–21). The present review also provides a key evaluation of refined chemotherapeutic approaches, such as the validation of folinic acid + fluorouracil + irinotecan (FOLFIRI) in second-line settings (22,23) and the exploration of modified folinic acid + fluorouracil + irinotecan + oxaliplatin (mFOLFIRINOX, which typically involves dose adjustments of the constituent drugs to improve tolerability) (22), moving beyond the traditional platinum-etoposide (EP) backbone. Furthermore, to translate evidence into practice, the present review offers structured guidance for clinicians, incorporating insights from recent trials (24–26) and summaries of ongoing clinical studies (for example, NCT04325425 and NCT04169672) (21,26). This comprehensive and up-to-date review aimed to equip clinicians with the knowledge to navigate the rapidly evolving therapeutic landscape of this aggressive malignancy in the future.

Pathological features and diagnosis

Pathological classification

According to the 2019 World Health Organization classification of digestive system NENs (27), NENs are classified into well-differentiated neuroendocrine tumors (NETs), poorly differentiated NECs and mixed neuroendocrine-non-NENs (MiNENs). NETs are graded G1 to G3 based on mitotic count and Ki-67 index (27). NECs are divided into small cell NEC (SCNEC) and large cell NEC (LCNEC), with SCNEC accounting for ~90% of all ENEC cases. Grossly, NECs often present as invasive submucosal masses or ulcerative lesions with esophageal lumen narrowing (28). Histologically, SCNEC exhibits oat cell-like nests with necrosis, whereas LCNEC features vesicular nuclei, prominent nucleoli and rosette structures. MiNENs contain ≥30% neuroendocrine components and may also exhibit adenocarcinoma or squamous differentiation (29). In China, MiNENs are usually squamous-dominant, whereas in Western countries, they often coexist with adenocarcinoma (30) (Table I).

Table I.

Classification of NENs.

Table I.

Classification of NENs.

Type of NENMorphological and molecular features
NET
  G1Mitotic count, <2/2 mm2 (or <2/10 HPF); Ki-67 index, ≤3%
  G2Mitotic count, 2–10/2 mm2 (or 2–20/10 HPF); Ki-67, 3–20%
  G3Mitotic count, >10/2 mm2 (or >20/10 HPF); Ki-67, >20%
NEC
  Small cell NEC (accounts forHigh mitotic count, >10/2 mm2 (or >20/10 HPF); Ki-67, >20% (often >55%)
  ~90% of esophageal NEC cases)
  Large cell NECHigh mitotic count, >10/2 mm2; Ki-67, >20% (often >55%)
MiNENBiphasic tumor with ≥30% each of neuroendocrine (NET/NEC) and non-neuroendocrine components

[i] HPF, high power field; MiNEN, mixed neuroendocrine-non-NEN; NEC, neuroendocrine carcinoma; NEN, neuroendocrine neoplasm; NET, neuroendocrine tumor.

Diagnosis

Pathological confirmation of NENs necessitates a multimodal diagnostic approach. Immunohistochemically, synaptophysin (Syn) exhibits high sensitivity (>95%) and is expressed in virtually all NENs. Chromogranin A (CgA) is more specific but less expressed in poorly differentiated NECs, requiring additional markers such as CD56 and neuron-specific enolase (31). Insulinoma-associated protein 1, a sensitive and specific nuclear marker for neuroendocrine differentiation, is increasingly used in diagnostic panels, particularly when conventional markers are equivocal (31–33). However, the interpretation of immunohistochemical markers in NECs poses notable challenges. Key interpretative pitfalls include: i) Heterogeneous or weak expression levels of Syn and CgA in poorly differentiated tumors, which may lead to false-negative diagnoses; ii) non-specific staining of CD56 observed in a range of non-neuroendocrine malignancies, such as small cell lung carcinoma, lymphoma, melanoma and certain types of sarcoma; and iii) highly variable expression patterns in MiNENs, underscoring the necessity of extensive tumor sampling and the application of a comprehensive antibody panel to prevent misclassification (32).

Imaging features of ENEC markedly overlap with ESCC and esophageal adenocarcinoma (EAC), making radiological distinction challenging. Specifically, ENEC typically presents on X-ray barium swallow as irregular mucosal destruction, strictures or filling defects, similar to the appearances seen in ESCC and EAC. On CT and enhanced CT, ENEC commonly manifests as focal or circumferential wall thickening with heterogeneous enhancement, patterns that are also frequently observed in advanced ESCC and EAC. For ENEC, enhanced CT is crucial for assessing primary tumor location, local invasion and distant metastasis (particularly to the liver), with reported sensitivity for detecting liver metastases reaching up to 79% (34,35). Furthermore, endoscopic ultrasound accurately assesses tumor origin, size and invasion depth, although it cannot reliably differentiate ENEC from ESCC or EAC based solely on imaging characteristics (34). PET-CT is used for staging and recurrence detection (36). Somatostatin receptor (SSTR) scintigraphy, using 111In-labeled octreotide, is more sensitive for the detection of well-differentiated NETs and their metastases (particularly in the liver and lungs). The sensitivity of SSTR scintigraphy for poorly differentiated NECs is generally lower due to reduced or absent SSTR expression (37).

The assessment of SSTR expression via functional imaging also has potential therapeutic implications. Peptide receptor radionuclide therapy (PRRT), such as 177Lu-oxodotreotide, is a well-established treatment option for well-differentiated, SSTR+ NETs (38). However, to the best of our knowledge, its role in poorly differentiated NECs, including ENEC, remains limited and non-standard; this is primarily due to the frequent absence or low density of SSTR expression in poorly differentiated NECs, which precludes the use of SSTR-targeted therapies (36). Therefore, patient selection for PRRT in high-grade disease hinges on demonstrating adequate SSTR expression on functional imaging, a finding that is uncommon in NEC. Although anecdotal case reports exist (38,39), robust clinical trial data supporting the efficacy of PRRT in ENEC are currently lacking.

Treatment strategies

The management of ENEC involves a multimodal approach, including surgery, platinum-based chemotherapy, radiotherapy, targeted therapy and immunotherapy (Fig. 1).

Main treatment modalities for
esophageal neuroendocrine carcinoma. ESD, endoscopic submucosal
dissection.

Figure 1.

Main treatment modalities for esophageal neuroendocrine carcinoma. ESD, endoscopic submucosal dissection.

Endoscopic treatment

Endoscopic submucosal dissection (ESD) can be considered for highly selected cases of early ENEC confined to the mucosa (Tis or T1a according to the American Joint Committee on Cancer TNM staging system, 8th edition) (40) with a diameter of <1 cm (41). Although rare, such early cases have been successfully treated with ESD. Case reports have described patients achieving long-term disease-free survival following ESD without adjuvant therapy (42,43). For example, Fukui et al (42) documented a patient with pT1a ENEC (muscularis mucosae invasion) who declined adjuvant therapy post-ESD and remained recurrence-free during the 15-month follow-up. Similarly, Cheng et al (43) reported a 3-year disease-free survival in a case of NEC arising at the esophagogastric junction without lymphovascular invasion, treated solely by ESD.

Neoadjuvant and/or adjuvant combined surgery

Multimodal therapy combining neoadjuvant and/or adjuvant therapy with surgery is standard for locally advanced ENEC. Patients with resectable tumors and no lymph node metastasis may undergo surgery first, followed by adjuvant chemoradiotherapy if necessary (13). For initially unresectable cases, neoadjuvant therapy may enable surgery (1). Previous studies have suggested that neoadjuvant chemoradiotherapy followed by surgery markedly improves median overall survival (mOS) compared with surgery or chemoradiotherapy alone (44–46).

In patients with initially unresectable locally advanced ENEC, neoadjuvant therapy may facilitate surgical conversion, followed by personalized adjuvant regimens (47). A multicenter trial by Shapiro et al (48) demonstrated that neoadjuvant chemoradiotherapy combined with surgery markedly improved mOS in locally advanced ESCC (81.6 vs. 21.1 months) and adenocarcinoma (43.2 vs. 27.1 months), establishing this approach as the standard of care. Although ENEC was not included in this previous study, extrapolation of these principles supports guideline recommendations. The Clinical Practice Guidelines for Gastrointestinal and Pancreatic Neuroendocrine Tumors conditionally endorse neoadjuvant chemotherapy for borderline resectable ENEC (49).

A previous systematic review revealed notably increased survival outcomes with perioperative chemotherapy (neoadjuvant, 31 months; adjuvant, 25 months) versus surgery alone (9 months) in stage I–III ENEC, although no notable difference existed between the adjuvant and neoadjuvant groups (50). Notably, adding radiotherapy to neoadjuvant chemotherapy enhances surgical resectability and pathological complete response rates but does not improve survival (13). Awada et al (51) documented a case of poorly differentiated ENEC treated with neoadjuvant chemoradiotherapy and surgery, achieving >5 years of survival.

Radiation therapy

For patients with locally advanced, inoperable ENEC tumors, definitive chemoradiotherapy is recommended (52). Treatment plans should be based on the extent of invasion and lymph node involvement. Combined chemoradiotherapy yields a 3-year survival rate of ~31.6% (53). A Japanese retrospective study further supported chemoradiotherapy as a viable option for locally advanced ENEC (45). However, unlike in limited-stage small cell lung cancer where it is a standard of care, prophylactic brain irradiation is not routinely recommended due to the low incidence of brain metastases in ENEC (3).

Chemotherapy

First-line treatment

For advanced ENEC, systemic chemotherapy remains essential (46). First-line regimens are platinum-based doublets, either etoposide + platinum (EP) or irinotecan + platinum (IP). Capecitabine with temozolomide (CAPTEM), folinic acid + fluorouracil + oxaliplatin (FOLFOX), FOLFIRI and FOLFIRINOX have also demonstrated activity (Table II) (54). Previous studies have reported variable response rates [objective response rate (ORR), 14–75%] and a median progression-free survival (PFS) time of 1.8–8.9 months (25,54–56).

Table II.

Comparison between first-line chemotherapy regimens.

Table II.

Comparison between first-line chemotherapy regimens.

RegimenKey dataToxicity profileEvidence level
EP/ECmOS, 12.5 months (25); ORR, 14–75% (56,57); mPFS, 5.36 months (25)Neutropenia (90%), anemia and thrombocytopenia Guideline-recommended (69)
IPmOS, 10.9 months; no notable difference vs. EP (25)Diarrhea (50%), neutropenia (60%)Phase III trial
CAPTEMmOS, 12.6 months; mPFS, 2.43 months (markedly lower compared with EP) (24)Grade 3/4 adverse events, 29% (markedly lower compared with EP)Phase II trial
FOLFOXDCR, 91.3% (first-line); mPFS, 10 months (66)Mild toxicity (neurotoxicity and diarrhea)Retrospective study
FOLFIRIDCR, 91% (metastatic GI NEC) (67)Neutropenia and diarrheaSmall-sample study
FOLFIRINOXORR, 46%; mOS, 17.8 months (n=8) (62)High toxicity (hematological and gastrointestinal)Early exploratory
mFOLFIRINOXORR, 77%; mOS, 20.6 months (n=35) (22)Markedly higher severe toxicity risk in womenModified regimen study

[i] ORR, objective response rate; mPFS, median progression-free survival; mOS, median overall survival; DCR, disease control rate; FOLFIRI, folinic acid + fluorouracil + irinotecan; FOLFOX, folinic acid + fluorouracil + oxaliplatin; CAPTEM, capecitabine with temozolomide; FOLFIRINOX, folinic acid + fluorouracil + irinotecan + oxaliplatin; mFOLFIRINOX, modified FOLFIRINOX; EP/EC, etoposide with carboplatin; IP, irinotecan + platinum; GI, gastrointestinal; NEC, neuroendocrine carcinoma.

The EP or etoposide and carboplatin (EC) regimen is widely used in NEC, while IP demonstrates comparable efficacy but distinct toxicity profiles. Retrospective studies have indicated variable response rates (ORR, 14–75%) and a median PFS time of 1.8–8.9 months (57). The phase III JCOG1213 TOPIC-NEC trial identified no notable difference in mOS between EP and IP regimens (EP group, 12.5 months vs. IP group, 10.9 months). The ENEC subgroup (15.5% in EP vs. 9.3% in IP) demonstrated no clear advantage in overall survival compared with the IP regimen. Toxicity profiles differed; for example, neutropenia occurred in 90% of patients treated with EP, whereas diarrhea affected ~50% of patients treated with IP, necessitating regimen selection based on individual tolerance (25). For patients who are cisplatin-intolerant, the EC regimen serves as an alternative, indicating efficacy comparable to EP (58). Notably, patients with Ki-67 >55% exhibit lower ORR to platinum-based chemotherapy but improved survival, a paradoxical phenomenon requiring further mechanistic study and context-specific clinical strategies (59).

Despite high response rates, the short PFS of first-line platinum-based regimens has prompted exploration of alternative therapies. It is important to distinguish high-grade NENs here; while the CAPTEM regimen is an option for advanced well-differentiated G3 NET (neoplasia), it is not a standard first-line therapy for poorly differentiated NEC. The role of CAPTEM has primarily been explored in later-line settings (24,60). Trials such as NCT04325425 are ongoing to compare mFOLFIRINOX and EP regimens (61). FOLFOX and FOLFIRI are being evaluated as alternatives or second-line regimens, with varying degrees of disease control (62–64). These exhibit antitumor activity in NEC; however, to the best of our knowledge, randomized prospective phase II studies remain limited and no international consensus exists. In a previous study by Merola et al (65) involving 72 patients with advanced cases (44.5% NET; 55.5% NEC), FOLFOX achieved a disease control rate (DCR) of 75% and mPFS of 8 months, with first-line DCR reaching 91.3% (mPFS, 10 months) and manageable toxicity. Extended treatment cycles were recommended for well-tolerated patients. Du et al (66) reported a DCR of 91% for FOLFIRI in 11 metastatic gastrointestinal NEC cases. An early small-sample study (n=8) of FOLFIRINOX demonstrated an ORR of 46% and mOS of 17.8 months in first-line treatment (61). A mFOLFIRINOX regimen (n=35) achieved an ORR of 77% (mOS, 20.6 months), although severe toxicity in female patients warrants caution (22). Most of these aforementioned studies were retrospective with small sample sizes (n, 8–72), which warrants the validation of FOLFOX efficacy. The ongoing randomized phase II trial NCT04325425 comparing mFOLFIRINOX and EP regimens may provide higher-level evidence for first-line NEC treatment (26).

Second and multiple lines of treatment

Second-line treatments have demonstrated limited effectiveness (ORR, ~18%) (56). Platinum rechallenge is considered for patients with progression >6 months after first-line therapy (67). Irinotecan-based (FOLFIRI) and oxaliplatin-based (FOLFOX) regimens are used in platinum-resistant cases (62,64). Temozolomide-based regimens have exhibited modest activity, particularly in O6-methylguanine-DNA methyltransferase (MGMT)-deficient tumors. Randomized trials are ongoing to refine second-line therapy strategies. Table III compares the main second-line chemotherapy regimens.

Table III.

Second-line chemotherapy regimens comparison.

Table III.

Second-line chemotherapy regimens comparison.

RegimenmOS, monthsmPFS, monthsORR, %DCR, %Evidence level
Platinum rechallenge11.7 (71)3.2 (71)17/31 (56,70)62 (71)Retrospective study
FOLFIRI5.9–18 (63,64,72)4.4–5.8 (63,64,72)-44-80 (63,64,72)Heterogeneous evidence
FOLFOX---64 (65)Retrospective study
CAPTEM12.1–22 (74,76)5.86 (76)26 (61)-Evidence from studies with conflicting results
Topoisomerase I inhibitors4.3 (78)1.8 (78)-15 (78)TLC388 trial

[i] ORR, objective response rate; mPFS, median progression-free survival; mOS, median overall survival; DCR, disease control rate; FOLFIRI, folinic acid + fluorouracil + irinotecan; FOLFOX, folinic acid + fluorouracil + oxaliplatin; CAPTEM, capecitabine with temozolomide.

Platinum rechallenge

Current consensus, as reflected in international guidelines such as those from the National Comprehensive Cancer Network (NCCN) and expert group recommendations, recommends regimen selection based on the duration of response to first-line chemotherapy (67,68). Patients with a time to progression of >6 months may consider EP rechallenge, whereas those with a time to progression of ≤6 months may benefit from regimens such as FOLFIRI or CAPTEM (67). Platinum-based rechallenge strategies have achieved ORRs of 17 and 31% in extrapulmonary NEC (55,69). In a nationwide multicenter study by Hadoux et al (70), platinum rechallenge chemotherapy demonstrated a DCR of 62% (mPFS, 3.2 months; mOS, 11.7 months). Patients with relapse-free intervals ≥3 months after first-line EP chemotherapy who received rechallenge therapy had markedly longer mOS (12 vs. 5.9 months). However, ENEC-specific subgroup analyses were lacking.

FOLFIRI program

Irinotecan-based regimens have been demonstrated to have heterogeneous efficacy. Previous studies have reported DCRs of 62–80% with FOLFIRI after platinum resistance (mPFS, 4–5.8 months; mOS, 11–18 months) (62,63). The PRODIGE 41-BEVANEC study validated FOLFIRI as a second-line option but identified no added benefit with bevacizumab (23). By contrast, Bardasi et al (71) demonstrated a lower DCR (44.1%; mOS, 5.9 months; mPFS, 4.4 months), highlighting discrepancies possibly due to study design or population characteristics. While FOLFIRI remains feasible after EP failure, its efficacy requires confirmation in prospective trials with controlled confounding factors. The NET-02 trial (n=102) compared liposomal irinotecan + 5-fluorouracil versus docetaxel in extrapulmonary NEC. Although no notable difference in mPFS or mOS was observed, the 6-month PFS rate doubled (29.6 vs. 13.8%), suggesting subgroup benefits. The trial failed its primary endpoint, Therefore, the potential advantage of the liposomal irinotecan combination regimen over docetaxel remains to be fully elucidated (72).

FOLFOX program

A retrospective study by Hadoux et al (64) demonstrated antitumor activity of FOLFOX as second- or third-line therapy in EP-refractory patients, with a DCR of 64% and <30% incidence of major hematological toxicity. These findings suggested that FOLFOX may offer survival benefits with manageable toxicity for aggressive ENEC lacking backline options.

CAPTEM program

Temozolomide efficacy remains debatable. Retrospective studies have reported an ORR of 26% and mOS of 22 months with CAPTEM in high-grade gastroenteropancreatic NEC after first-line failure (60,73). However, a prospective phase II study of temozolomide monotherapy (B160101021) demonstrated a lower ORR and mOS with an mPFS of 1.8 months, albeit with minimal toxicity (74). Notably, MGMT-deficient patients exhibited partial responses, suggesting that MGMT deficiency may serve as a potential predictive biomarker for sensitivity to temozolomide-based chemotherapy. The recent NCT04122911 trial reported improved outcomes (mPFS, 5.86 months; mOS, 12.1 months) for second-line temozolomide (75). These findings indicate modest efficacy and manageable toxicity, particularly in combination or MGMT-deficient subgroups.

The NCT03387592 trial compared CAPTEM and FOLFIRI in metastatic NEC. At 12 weeks, DCRs were 39.1% (CAPTEM) and 28.0% (FOLFIRI), with no notable difference in 12-month survival (28.4 vs. 32.4%). Both regimens demonstrated mild toxicity (<35% incidence). High microRNA expression was associated with poor prognosis, offering insights for stratification. Early termination of the trial (n=53) limited conclusions, but safety and similar antitumor activity were confirmed (76).

Topoisomerase I inhibitors

Topoisomerase I inhibitor monotherapy lacks notable efficacy in multiple studies and is not recommended in in major oncology guidelines, such as the NCCN Guidelines for Neuroendocrine and Adrenal Tumors (26,68). The NCT02457273 trial evaluated the novel camptothecin analog TLC388 in EP-refractory metastatic NEC, demonstrating a DCR of 15% (mPFS, 1.8 months; mOS, 4.3 months). The trial was halted due to unmet efficacy endpoints. However, MutS homolog 6 mutations (40% of samples) in the studied cohort of poorly differentiated NEC (including cases from various primary sites) have been associated with tumor mutational burden, thus suggesting potential for targeted therapies (77).

Targeted therapy

Targeted therapy inhibits tumor growth and proliferation by interfering with specific molecular targets. While molecular-targeted drugs are approved for various solid tumors, such as non-small cell lung cancer, breast cancer and renal cell carcinoma (78–80), their efficacy in NEC remains to be elucidated. Current research focuses on mammalian target of rapamycin (mTOR) inhibitors and anti-angiogenic agents. It is considered that with the continuous development of research, traditional treatment combined with targeted therapy may provide hope for patients in the future.

mTOR inhibitors

mTOR, a key kinase regulating cell proliferation, metabolism and angiogenesis, promotes tumor progression and represents a potential therapeutic target. Everolimus, an mTOR inhibitor, has demonstrated limited efficacy as monotherapy in phase II trials (mPFS, 1.2–1.3 months), but has shown enhanced antitumor activity in combination regimens (81,82). The NCT02695459 study reported that everolimus combined with cisplatin was effective as first-line therapy for advanced extrapulmonary NEC, which improved quality of life by avoiding etoposide-related side effects. Subgroup analysis identified three patients with sustained remission >1 year, suggesting there may be molecular predictors of sensitivity that remain to be identified; however, their specific identities remain to be elucidated due to a lack of correlative biomarker analysis in the trial (83). Similarly, NCT01317615 reported that everolimus combined with carboplatin and paclitaxel was effective and well-tolerated in metastatic lung LCNEC (84). In contrast to the potential benefits of mTOR inhibitors, trials such as PRODIGE 41-BEVANEC did not report notable survival benefits from the addition of bevacizumab to FOLFIRI (85).

Antitumor neovascular drugs

Bevacizumab, an anti-angiogenic agent, exhibits variable efficacy in NEN. Early retrospective studies have suggested that bevacizumab combined with temozolomide may provide benefits for patients with poorly differentiated NEC, although incomplete data have limited conclusions (23,73). The PRODIGE 41-BEVANEC trial (NCT02820857) identified no survival difference between FOLFIRI with or without bevacizumab in platinum-resistant gastrointestinal-pancreatic NEC (6-month OS, 53 vs. 60%) (23). This may reflect the high Ki-67 index (>55%) and complex angiogenesis of NEC compared with vascular-dependent NET (G1-G2), which may demonstrate improved response to bevacizumab. Retrospective studies have supported combining bevacizumab with FOLFIRI, FOLFOX, FOLFIRINOX or temozolomide for NET (23,85).

Immunotherapy

Checkpoint inhibitors [PD-1, programmed cell death-ligand 1 (PD-L1) and CTLA-4] offer potential but lack large-scale ENEC-specific evidence (72). Case reports have described complete remission (CR) when combining immunotherapy with radiotherapy or targeted agents, such as anlotinib or apatinib (14,15,19,20). Dual immunotherapy (for example, nivolumab + ipilimumab) demonstrates promise in high-grade NENs (17,18).

Immunotherapy combined with radiotherapy

Takagi et al (14) reported complete response in a patient with metastatic ENEC treated with nivolumab and radiotherapy, maintaining relapse-free survival for 42 months despite ≤1% PD-L1 expression, which suggests radiotherapy may modulate the immune microenvironment. Hanzawa et al (15) described sustained disease control in unresectable esophagogastric NEC with nivolumab and radiotherapy, achieving >4.5-year survival.

Immunotherapy combined with chemotherapy

Based on previous trials of extensive-stage small cell lung cancer (86–89), immunotherapy combined with platinum-based chemotherapy has been hypothesized to benefit gastrointestinal NEC. Ongoing phase II trials (NCT03901378, NCT03147404 and NCT03352934) are evaluating pembrolizumab-chemotherapy and avelumab monotherapy. However, the NET-001/002 trial reported a DCR of only 21% for avelumab in grade 2–3 NEN (90), which underscores the need for optimized regimens.

Dual immunotherapy

Dual immunotherapy (CTLA-4 + PD-1 inhibition) targets complementary immune pathways. The SWOG S1609 DART trial reported a 26% ORR and 32% 6-month PFS with ipilimumab and nivolumab in high-grade NEN, with manageable toxicity (grade 3/4 alanine aminotransferase elevation was most common) (17). The GETNE 1601 trial achieved a 36.1% 9-month survival rate with durvalumab and tremelimumab in chemotherapy-refractory gastroenteropancreatic NEN (18). While promising, dual therapy requires caution due to potential toxicity.

Immunotherapy combined with targeted therapy

Combining checkpoint inhibitors with multi-target TKIs such as surufatinib, anlotinib or apatinib demonstrate promise. Previous studies have reported DCRs of >80% and durable remission in certain cases (18,21). The combination benefits from synergistic effects on the tumor immune microenvironment (91).

Sulfatinib, a multi-target kinase inhibitor [VEGF receptor VEGFR)1-3, fibroblast growth factor receptor 1 (FGFR1) and colony stimulating factor 1 receptor], approved for non-pancreatic NET in China, combined with toripalimab (PD-1 inhibitor) achieved an 80% DCR (mPFS, 4.1 months; mOS, 13.7 months) in advanced NEC with manageable toxicity in the NCT04169672 trial (21). No further subgroup analyses were performed in this trial.

Camrelizumab (a PD-1 inhibitor) has demonstrated efficacy in esophageal cancer, including ENEC. In a specific trial including patients with esophageal cancer, ORRs ranging from 17.4 to 73.1% and a mOS of 8.3 months were reported, with efficacy appearing to be influenced by PD-L1 expression and other biomarkers, such as tumor mutational burden or microsatellite instability (16). Liu et al (16) reported camrelizumab combined with apatinib (VEGFR-2 inhibitor) in third-line ENEC recurrence, achieving >10-month PFS, which suggests that tumor microenvironment modulation enhances efficacy.

Anlotinib, a multi-target TKI (VEGFR, platelet-derived growth factor receptor, FGFR and c-Kit), improved survival in the ALTER 1202 trial for small cell lung cancer (92). Zhou et al (19) described a patient with metastatic ENEC achieving 29-month PFS and >50-month OS with camrelizumab and anlotinib after chemoradiation failure, reaching CR on PET/CT.

Tislelizumab (PD-1 inhibitor), which has been approved for the treatment of multiple malignancies such as non-small cell lung cancer and hepatocellular carcinoma (93), combined with anlotinib in second-line metastatic ENEC achieved CR (PFS, 16 months; OS, 21 months) with minimal toxicity in an elderly patient (20).

Targeted-immunotherapy combinations synergistically regulate the tumor microenvironment, offering a novel strategy to overcome traditional treatment limitations. While current evidence derives from small studies or case reports (14–16,19,20), preliminary data have suggested manageable toxicity and survival benefits. Future high-quality trials and translational research are warranted to validate efficacy and optimize personalized treatment.

Immunotherapy summary

Immunotherapy exploration in ENEC highlights multidimensional advances. Radiotherapy combinations may enhance efficacy by modulating the immune microenvironment. Dual immunotherapy (CTLA-4 + PD-1 inhibition) improves ORR and survival in high-grade NEN but requires caution regarding toxicity. Chemotherapy combinations lack randomized trial validation in gastrointestinal NEC despite success in small cell lung cancer. Targeted-immunotherapy regimens remodel the tumor microenvironment, achieving survival benefits. To the best of our knowledge, current evidence is limited to small studies or case reports with heterogeneous populations and undefined biomarkers. Future efforts should prioritize prospective trials, tumor microenvironment dynamics and epigenetic analyses to establish precision treatment models and address drug resistance.

Conclusion

ENEC is a rare, highly aggressive gastrointestinal tumor with diagnosis dependent on pathology and treatment requiring stratified management. Endoscopic or surgical resection is preferred in early stages, although curable cases are rare. For locally advanced disease, neoadjuvant/adjuvant therapy plus surgery or chemoradiotherapy improves survival. Platinum-based chemotherapy remains first-line in advanced stages, with individualized second-line regimens. Emerging therapies, particularly immunotherapy and targeted therapies, have achieved long-term remission in individual cases. Future large-scale clinical trials are warranted to optimize molecular subtyping, refine therapeutic strategies and improve outcomes for this high-grade malignancy. Finally, although the exclusive focus on ENEC in the present review is justified by its distinct biology, the omission of direct comparisons with other gastrointestinal NECs (such as GNEC) represents a limitation. Future studies integrating multi-origin NEC data may help refine both site-specific and common therapeutic strategies.

Acknowledgements

The authors would like to thank Dr Fangyuan Kong (Department of Oncology, The General Hospital of Western Theater Command, Chengdu, China) for their perspectives on the multidisciplinary management of esophageal neuroendocrine carcinoma and Dr Yong Diao (Department of Oncology, The General Hospital of Western Theater Command) for sharing their clinical experience regarding immunotherapy in rare malignancies. Their specialized knowledge enhanced the clinical viewpoints presented in the present review.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

JS and BH conceptualized the present review, curated the literature, devised the methodology and prepared the original draft. HZ and CJ contributed to the study conception and data acquisition, performed systematic literature retrieval, data extraction and validation, conducted the comparative analysis and interpretation of data from the included literature, and were responsible for the design and creation of all tables and figures. HZ and CJ also participated in drafting and critically reviewing the manuscript. LZ made substantial contributions to the conception of the work, participated in drafting the manuscript and provided critical revision for important intellectual content. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ma Z, Cai H and Cui Y: Progress in the treatment of esophageal neuroendocrine carcinoma. Tumour Biol. 39:10104283177113132017. View Article : Google Scholar : PubMed/NCBI

2 

Fraenkel M, Kim MK, Faggiano A and Valk GD: Epidemiology of gastroenteropancreatic neuroendocrine tumours. Best Pract Res Clin Gastroenterol. 26:691–703. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Dasari A, Mehta K, Byers LA, Sorbye H and Yao JC: Comparative study of lung and extrapulmonary poorly differentiated neuroendocrine carcinomas: A SEER database analysis of 162,983 cases. Cancer. 124:807–815. 2018. View Article : Google Scholar : PubMed/NCBI

4 

IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, . Drinking Coffee, Mate, and Very Hot Beverages. International Agency for Research on Cancer; Lyon: pp. 4882018

5 

Hirabayashi K, Zamboni G, Nishi T, Tanaka A, Kajiwara H and Nakamura N: Histopathology of gastrointestinal neuroendocrine neoplasms. Front Oncol. 3:22013. View Article : Google Scholar : PubMed/NCBI

6 

Nevárez A, Saftoiu A and Bhutani MS: Primary small cell carcinoma of the esophagus: Clinico- pathological features and therapeutic options. Curr Health Sci J. 37:31–34. 2011.PubMed/NCBI

7 

Huang Q, Fang DC, Yu CG, Zhang J and Chen MH: Barrett's esophagus-related diseases remain uncommon in China. J Dig Dis. 12:420–427. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Al Mansoor S, Ziske C and Schmidt-Wolf IGH: Primary small cell carcinoma of the esophagus: Patient data metaanalysis and review of the literature. Ger Med Sci. 11:Doc122013.PubMed/NCBI

9 

Imai T, Sannohe Y and Okano H: Oat cell carcinoma (apudoma) of the esophagus: A case report. Cancer. 41:358–364. 1978. View Article : Google Scholar : PubMed/NCBI

10 

Giannetta E, Guarnotta V, Rota F, de Cicco F, Grillo F, Colao A and Faggiano A; NIKE: A rare rarity: Neuroendocrine tumor of the esophagus. Crit Rev Oncol Hematol. 137:92–107. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Wu H, Yu Z, Liu Y, Guo L, Teng L, Guo L, Liang L, Wang J, Gao J, Li R, et al: Genomic characterization reveals distinct mutation landscapes and therapeutic implications in neuroendocrine carcinomas of the gastrointestinal tract. Cancer Commun (Lond). 42:1367–1386. 2022. View Article : Google Scholar : PubMed/NCBI

12 

Ji A, Jin R, Zhang R and Li H: Primary small cell carcinoma of the esophagus: Progression in the last decade. Ann Transl Med. 8:5022020. View Article : Google Scholar : PubMed/NCBI

13 

Gonzalez RS: Diagnosis and management of gastrointestinal neuroendocrine neoplasms. Surg Pathol Clin. 13:377–397. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Takagi K, Kamada T, Fuse Y, Kai W, Takahashi J, Nakashima K, Nakaseko Y, Suzuki N, Yoshida M, Okada S, et al: Nivolumab in combination with radiotherapy for metastatic esophageal neuroendocrine carcinoma after esophagectomy: A case report. Surg Case Rep. 7:2212021. View Article : Google Scholar : PubMed/NCBI

15 

Hanzawa S, Asami S, Kanazawa T, Oono S and Takakura N: Multimodal treatment with nivolumab contributes to long-term survival in a case of unresectable esophagogastric junction neuroendocrine carcinoma. Cureus. 16:e659812024.PubMed/NCBI

16 

Liu L, Liu Y, Gong L, Zhang M and Wu W: Salvage camrelizumab plus apatinib for relapsed esophageal neuroendocrine carcinoma after esophagectomy: A case report and review of the literature. Cancer Biol Ther. 21:983–989. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Patel SP, Othus M, Chae YK, Giles FJ, Hansel DE, Singh PP, Fontaine A, Shah MH, Kasi A, Baghdadi TA, et al: A phase II basket trial of dual anti-CTLA-4 and Anti-PD-1 blockade in rare tumors (DART SWOG 1609) in patients with nonpancreatic neuroendocrine tumors. Clin Cancer Res. 26:2290–2296. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Capdevila J, Teule A, López C, García-Carbonero R, Benavent M, Custodio A, Cubillo A, Alonso V, Gordoa TA, Carmona-Bayonas A, et al: 1157O A multi-cohort phase II study of durvalumab plus tremelimumab for the treatment of patients (pts) with advanced neuroendocrine neoplasms (NENs) of gastroenteropancreatic or lung origin: The DUNE trial (GETNE 1601). Ann Oncol. 31:S770–S771. 2020. View Article : Google Scholar

19 

Zhou L, Xu G, Chen T, Wang Q, Zhao J, Zhang T, Duan R and Xia Y: Anlotinib plus camrelizumab achieved long-term survival in a patient with metastatic esophageal neuroendocrine carcinoma. Cancer Rep (Hoboken). 6:e18552023.PubMed/NCBI

20 

Zhang Y, Liu X, Liang H, Liu W, Wang H and Li T: Late-stage esophageal neuroendocrine carcinoma in a patient treated with tislelizumab combined with anlotinib: A case report. J Int Med Res. 51:30006052311879422023. View Article : Google Scholar : PubMed/NCBI

21 

Zhang P, Shi S, Xu J, Chen Z, Song L, Zhang X, Cheng Y, Zhang Y, Ye F, Li Z, et al: Surufatinib plus toripalimab in patients with advanced neuroendocrine tumours and neuroendocrine carcinomas: An open-label, single-arm, multi-cohort phase II trial. Eur J Cancer. 199:1135392024. View Article : Google Scholar : PubMed/NCBI

22 

Borghesani M, Reni A, Lauricella E, Rossi A, Moscarda V, Trevisani E, Torresan I, Al-Toubah T, Filoni E, Luchini C, et al: Efficacy and toxicity analysis of mFOLFIRINOX in high-grade gastroenteropancreatic neuroendocrine neoplasms. J Natl Compr Canc Netw. 22:e2470052024. View Article : Google Scholar : PubMed/NCBI

23 

Walter T, Lievre A, Coriat R, Malka D, Elhajbi F, Di Fiore F, Hentic O, Smith D, Hautefeuille V, Roquin G, et al: Bevacizumab plus FOLFIRI after failure of platinum-etoposide first-line chemotherapy in patients with advanced neuroendocrine carcinoma (PRODIGE 41-BEVANEC): A randomised, multicentre, non-comparative, open-label, phase 2 trial. Lancet Oncol. 24:297–306. 2023. View Article : Google Scholar : PubMed/NCBI

24 

Eads JR, Catalano PJ, Fisher GA, Rubin D, Iagaru A, Klimstra DS, Konda B, Kwong MS, Chan JA, de Jesus-Acosta A, et al: Randomized phase II study of platinum and etoposide (EP) versus temozolomide and capecitabine (CAPTEM) in patients (pts) with advanced G3 non-small cell gastroenteropancreatic neuroendocrine neoplasms (GEPNENs): ECOG-ACRIN EA2142. J Clin Oncol. 40 (16_suppl):S40202022. View Article : Google Scholar

25 

Morizane C, Machida N, Honma Y, Okusaka T, Boku N, Kato K, Nomura S, Hiraoka N, Sekine S, Taniguchi H, et al: Effectiveness of etoposide and cisplatin vs irinotecan and cisplatin therapy for patients with advanced neuroendocrine carcinoma of the digestive system: The TOPIC-nec phase 3 randomized clinical trial. JAMA Oncol. 8:1447–1455. 2022. View Article : Google Scholar : PubMed/NCBI

26 

Weaver JMJ, Hubner RA, Valle JW and McNamara MG: Selection of chemotherapy in advanced poorly differentiated extra-pulmonary neuroendocrine carcinoma. Cancers (Basel). 15:49512023. View Article : Google Scholar : PubMed/NCBI

27 

Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, Washington KM, Carneiro F and Cree IA; WHO Classification of Tumours Editorial Board, : The 2019 WHO classification of tumours of the digestive system. Histopathology. 76:182–188. 2020. View Article : Google Scholar : PubMed/NCBI

28 

Yang L, Sun X, Zou Y and Meng X: Small cell type neuroendocrine carcinoma colliding with squamous cell carcinoma at esophagus. Int J Clin Exp Pathol. 7:1792–1795. 2014.PubMed/NCBI

29 

Wilson CI, Summerall J, Willis I, Lubin J and Inchausti BC: Esophageal collision tumor (Large cell neuroendocrine carcinoma and papillary carcinoma) arising in a Barrett esophagus. Arch Pathol Lab Med. 124:411–415. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Maru DM, Khurana H, Rashid A, Correa AM, Anandasabapathy S, Krishnan S, Komaki R, Ajani JA, Swisher SG and Hofstetter WL: Retrospective study of clinicopathologic features and prognosis of high-grade neuroendocrine carcinoma of the esophagus. Am J Surg Pathol. 32:1404–1411. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Klöppel G: Classification and pathology of gastroenteropancreatic neuroendocrine neoplasms. Endocr Relat Cancer. 18 (Suppl 1):S1–S16. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Capelli P, Fassan M and Scarpa A: Pathology-grading and staging of GEP-NETs. Best Pract Res Clin Gastroenterol. 26:705–717. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Rooper LM, Bishop JA and Westra WH: INSM1 is a sensitive and specific marker of neuroendocrine differentiation in head and neck tumors. Am J Surg Pathol. 42:665–671. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Wang D, Zhang GB, Yan L, Wei XE, Zhang YZ and Li WB: CT and enhanced CT in diagnosis of gastrointestinal neuroendocrine carcinomas. Abdom Imaging. 37:738–745. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Schott M, Klöppel G, Raffel A, Saleh A, Knoefel WT and Scherbaum WA: Neuroendocrine neoplasms of the gastrointestinal tract. Dtsch Arztebl Int. 108:305–312. 2011.PubMed/NCBI

36 

Islam O, Sarti K, Verbruggen L, Vandersmissen V, Bulcke KV, Annys L, Verslype C, Van Laethem JL, Kalantari HR, Janssens J, et al: Management of high-grade neuroendocrine neoplasms: Impact of functional imaging. Endocr Relat Cancer. 32:e2402312025. View Article : Google Scholar : PubMed/NCBI

37 

Sahani DV, Bonaffini PA, Fernández-Del Castillo C and Blake MA: Gastroenteropancreatic neuroendocrine tumors: Role of imaging in diagnosis and management. Radiology. 266:38–61. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Strosberg J, El-Haddad G, Wolin E, Hendifar A, Yao J, Chasen B, Mittra E, Kunz PL, Kulke MH, Jacene H, et al: Phase 3 trial of 177Lu-dotatate for midgut neuroendocrine tumors. N Engl J Med. 376:125–135. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Chan DS, Kanagaratnam AL, Pavlakis N and Chan DL: Peptide receptor chemoradionuclide therapy for neuroendocrine neoplasms: A systematic review. J Neuroendocrinol. 37:e133552025. View Article : Google Scholar : PubMed/NCBI

40 

Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, Meyer L, Gress DM, Byrd DR and Winchester DP: The eighth edition AJCC cancer staging manual: Continuing to build a bridge from a population-based to a more ‘personalized’ approach to cancer staging. CA Cancer J Clin. 67:93–99. 2017.PubMed/NCBI

41 

Lee CG, Lim YJ, Park SJ, Jang BI, Choi SR, Kim JK, Kim YT, Cho JY, Yang CH, Chun HJ, et al: The clinical features and treatment modality of esophageal neuroendocrine tumors: A multicenter study in Korea. BMC Cancer. 14:5692014. View Article : Google Scholar : PubMed/NCBI

42 

Fukui H, Dohi O, Miyazaki H, Yasuda T, Yoshida T, Ishida T, Doi T, Hirose R, Inoue K, Harusato A, et al: A case of endoscopic submucosal dissection for neuroendocrine carcinoma of the esophagus with invasion to the muscularis mucosae. Clin J Gastroenterol. 15:339–344. 2022. View Article : Google Scholar : PubMed/NCBI

43 

Cheng YQ, Wang GF, Zhou XL, Lin M, Zhang XW and Huang Q: Early adenocarcinoma mixed with a neuroendocrine carcinoma component arising in the gastroesophageal junction: A case report. World J Gastrointest Oncol. 16:563–570. 2024. View Article : Google Scholar : PubMed/NCBI

44 

Vest M, Shah D, Nassar M and Niknam N: Neuroendocrine carcinoma of the esophagus with liver metastasis: A case report. Cureus. 14:e288422022.PubMed/NCBI

45 

Honma Y, Nagashima K, Hirano H, Shoji H, Iwasa S, Takashima A, Okita N, Kato K, Boku N, Murakami N, et al: Clinical outcomes of locally advanced esophageal neuroendocrine carcinoma treated with chemoradiotherapy. Cancer Med. 9:595–604. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Alese OB, Jiang R, Shaib W, Wu C, Akce M, Behera M and El-Rayes BF: High-grade gastrointestinal neuroendocrine carcinoma management and outcomes: A national cancer database study. Oncologist. 24:911–920. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Enjoji T, Kobayashi S, Hayashi K, Tetsuo H, Matsumoto R, Maruya Y, Araki T, Honda T, Akazawa Y, Kanetaka K, et al: Long-term survival after conversion surgery for an esophageal neuroendocrine carcinoma: A case report. Gen Thorac Cardiovasc Surg Cases. 3:282024.PubMed/NCBI

48 

Shapiro J, van Lanschot JJB, Hulshof MCCM, van Hagen P, van Berge Henegouwen MI, Wijnhoven BPL, van Laarhoven HWM, Nieuwenhuijzen GAP, Hospers GAP, Bonenkamp JJ, et al: Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): Long-term results of a randomised controlled trial. Lancet Oncol. 16:1090–1098. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Ito T, Masui T, Komoto I, Doi R, Osamura RY, Sakurai A, Ikeda M, Takano K, Igarashi H, Shimatsu A, et al: JNETS clinical practice guidelines for gastroenteropancreatic neuroendocrine neoplasms: diagnosis, treatment, and follow-up: A synopsis. J Gastroenterol. 56:1033–1044. 2021. View Article : Google Scholar : PubMed/NCBI

50 

Kikuchi Y, Shimada H, Yamaguchi K and Igarashi Y: Systematic review of case reports of Japanese esophageal neuroendocrine cell carcinoma in the Japanese literature. Int Cancer Conf J. 8:47–57. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Awada H, Ali AH, Bakhshwin A and Daw H: High-grade large cell neuroendocrine carcinoma of the esophagus: A case report and review of the literature. J Med Case Rep. 17:1442023. View Article : Google Scholar : PubMed/NCBI

52 

Shah MH, Goldner WS, Benson AB, Bergsland E, Blaszkowsky LS, Brock P, Chan J, Das S, Dickson PV, Fanta P, et al: Neuroendocrine and adrenal tumors, version 2.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 19:839–868. 2021. View Article : Google Scholar : PubMed/NCBI

53 

Wong AT, Shao M, Rineer J, Osborn V, Schwartz D and Schreiber D: Treatment and survival outcomes of small cell carcinoma of the esophagus: An analysis of the National cancer data base. Dis Esophagus. 30:1–5. 2017.

54 

Moertel CG, Kvols LK, O'Connell MJ and Rubin J: Treatment of neuroendocrine carcinomas with combined etoposide and cisplatin. Evidence of major therapeutic activity in the anaplastic variants of these neoplasms. Cancer. 68:227–232. 1991. View Article : Google Scholar : PubMed/NCBI

55 

Yamaguchi T, Machida N, Morizane C, Kasuga A, Takahashi H, Sudo K, Nishina T, Tobimatsu K, Ishido K, Furuse J, et al: Multicenter retrospective analysis of systemic chemotherapy for advanced neuroendocrine carcinoma of the digestive system. Cancer Sci. 105:1176–1181. 2014. View Article : Google Scholar : PubMed/NCBI

56 

McNamara MG, Frizziero M, Jacobs T, Lamarca A, Hubner RA, Valle JW and Amir E: Second-line treatment in patients with advanced extra-pulmonary poorly differentiated neuroendocrine carcinoma: A systematic review and meta-analysis. Ther Adv Med Oncol. 12:17588359209152992020. View Article : Google Scholar : PubMed/NCBI

57 

Ooki A, Osumi H, Fukuda K and Yamaguchi K: Potent molecular-targeted therapies for gastro-entero-pancreatic neuroendocrine carcinoma. Cancer Metastasis Rev. 42:1021–1054. 2023. View Article : Google Scholar : PubMed/NCBI

58 

Imai H, Shirota H, Okita A, Komine K, Saijo K, Takahashi M, Takahashi S, Takahashi M, Shimodaira H and Ishioka C: Efficacy and safety of carboplatin and etoposide combination chemotherapy for extrapulmonary neuroendocrine carcinoma: A retrospective case series. Chemotherapy. 61:111–116. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Sorbye H, Welin S, Langer SW, Vestermark LW, Holt N, Osterlund P, Dueland S, Hofsli E, Guren MG, Ohrling K, et al: Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): The NORDIC NEC study. Ann Oncol. 24:152–160. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Chan DL, Bergsland EK, Chan JA, Gadgil R, Halfdanarson TR, Hornbacker K, Kelly V, Kunz PL, McGarrah PW, Raj NP, et al: Temozolomide in grade 3 gastroenteropancreatic neuroendocrine neoplasms: A multicenter retrospective review. Oncologist. 26:950–955. 2021. View Article : Google Scholar : PubMed/NCBI

61 

Butt BP, Stokmo HL, Ladekarl M, Tabaksblat EM, Sorbye H, Revheim ME and Hjortland GO: 1108P Folfirinox in the treatment of advanced gastroenteropancreatic neuroendocrine carsinomas. ESMO. 32:S9152021.

62 

Hentic O, Hammel P, Couvelard A, Rebours V, Zappa M, Palazzo M, Maire F, Goujon G, Gillet A, Lévy P, et al: FOLFIRI regimen: An effective second-line chemotherapy after failure of etoposide-platinum combination in patients with neuroendocrine carcinomas grade 3. Endocr Relat Cancer. 19:751–757. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Sugiyama K, Shiraishi K, Sato M, Nishibori R, Nozawa K and Kitagawa C: Salvage chemotherapy by folfiri regimen for poorly differentiated gastrointestinal neuroendocrine carcinoma. J Gastrointest Cancer. 52:947–951. 2021. View Article : Google Scholar : PubMed/NCBI

64 

Hadoux J, Malka D, Planchard D, Scoazec JY, Caramella C, Guigay J, Boige V, Leboulleux S, Burtin P, Berdelou A, et al: Post-first-line FOLFOX chemotherapy for grade 3 neuroendocrine carcinoma. Endocr Relat Cancer. 22:289–298. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Merola E, Dal Buono A, Denecke T, Arsenic R, Pape UF, Jann H, Wiedenmann B and Pavel ME: Efficacy and toxicity of 5-Fluorouracil-Oxaliplatin in gastroenteropancreatic neuroendocrine neoplasms. Pancreas. 49:912–917. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Du Z, Wang Y, Zhou Y, Wen F and Li Q: First-line irinotecan combined with 5-fluorouracil and leucovorin for high-grade metastatic gastrointestinal neuroendocrine carcinoma. Tumori. 99:57–60. 2013. View Article : Google Scholar : PubMed/NCBI

67 

Lamarca A, Frizziero M, Barriuso J, McNamara MG, Hubner RA and Valle JW: Urgent need for consensus: International survey of clinical practice exploring use of platinum-etoposide chemotherapy for advanced extra-pulmonary high grade neuroendocrine carcinoma (EP-G3-NEC). Clin Transl Oncol. 21:950–953. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Shah MH, Goldner WS, Benson AB, Bergsland E, Blaszkowsky LS, Brock P, Chan J, Das S, Dickson PV, Fanta P, et al: Neuroendocrine and adrenal tumors, version 2.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 19:839–868. 2021. View Article : Google Scholar : PubMed/NCBI

69 

Frizziero M, Spada F, Lamarca A, Kordatou Z, Barriuso J, Nuttall C, McNamara MG, Hubner RA, Mansoor W, Manoharan P, et al: Carboplatin in combination with oral or intravenous etoposide for extra-pulmonary, poorly-differentiated neuroendocrine carcinomas. Neuroendocrinology. 109:100–112. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Hadoux J, Walter T, Kanaan C, Hescot S, Hautefeuille V, Perrier M, Tauveron I, Laboureau S, Do Cao C, Petorin C, et al: Second-line treatment and prognostic factors in neuroendocrine carcinoma: the RBNEC study. Endocr Relat Cancer. 29:569–580. 2022. View Article : Google Scholar : PubMed/NCBI

71 

Bardasi C, Spallanzani A, Benatti S, Spada F, Laffi A, Antonuzzo L, Lavacchi D, Marconcini R, Ferrari M, Rimini M, et al: Irinotecan-based chemotherapy in extrapulmonary neuroendocrine carcinomas: Survival and safety data from a multicentric Italian experience. Endocrine. 74:707–713. 2021. View Article : Google Scholar : PubMed/NCBI

72 

McNamara MG, Swain J, Craig Z, Sharma R, Faluyi O, Wadsley J, Morgan C, Wall LR, Chau I, Reed N, et al: NET-02: A randomised, non-comparative, phase II trial of nal-IRI/5-FU or docetaxel as second-line therapy in patients with progressive poorly differentiated extra-pulmonary neuroendocrine carcinoma. EClinicalMedicine. 60:1020152023. View Article : Google Scholar : PubMed/NCBI

73 

Welin S, Sorbye H, Sebjornsen S, Knappskog S, Busch C and Oberg K: Clinical effect of temozolomide-based chemotherapy in poorly differentiated endocrine carcinoma after progression on first-line chemotherapy. Cancer. 117:4617–4622. 2011. View Article : Google Scholar : PubMed/NCBI

74 

Kobayashi N, Takeda Y, Okubo N, Suzuki A, Tokuhisa M, Hiroshima Y and Ichikawa Y: Phase II study of temozolomide monotherapy in patients with extrapulmonary neuroendocrine carcinoma. Cancer Sci. 112:1936–1942. 2021. View Article : Google Scholar : PubMed/NCBI

75 

von Arx C, Della Vittoria Scarpati G, Cannella L, Clemente O, Marretta AL, Bracigliano A, Picozzi F, Iervolino D, Granata V, Modica R, et al: A new schedule of one week on/one week off temozolomide as second-line treatment of advanced neuroendocrine carcinomas (TENEC-TRIAL): A multicenter, open-label, single-arm, phase II trial. ESMO Open. 9:1030032024. View Article : Google Scholar : PubMed/NCBI

76 

Bongiovanni A, Liverani C, Foca F, Bergamo F, Leo S, Pusceddu S, Gelsomino F, Brizzi MP, Di Meglio G, Spada F, et al: A randomized phase II trial of Captem or Folfiri as second-line therapy in neuroendocrine carcinomas. Eur J Cancer. 208:1141292024. View Article : Google Scholar : PubMed/NCBI

77 

Chen MH, Chou WC, Hsiao CF, Jiang SS, Tsai HJ, Liu YC, Hsu C, Shan YS, Hung YP, Hsich CH, et al: An open-label, single-arm, two-stage, multicenter, phase II study to evaluate the efficacy of TLC388 and genomic analysis for poorly differentiated neuroendocrine carcinomas. Oncologist. 25:e782–e788. 2020. View Article : Google Scholar : PubMed/NCBI

78 

Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, Sunpaweravong P, Han B, Margono B, Ichinose Y, et al: Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 361:947–957. 2009. View Article : Google Scholar : PubMed/NCBI

79 

Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, et al: Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 344:783–792. 2001. View Article : Google Scholar : PubMed/NCBI

80 

Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, et al: Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 356:115–124. 2007. View Article : Google Scholar : PubMed/NCBI

81 

Okuyama H, Ikeda M, Okusaka T, Furukawa M, Ohkawa S, Hosokawa A, Kojima Y, Hara H, Murohisa G, Shioji K, 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. 110:988–993. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Tarhini A, Kotsakis A, Gooding W, Shuai Y, Petro D, Friedland D, Belani CP, Dacic S and Argiris A: Phase II study of everolimus (RAD001) in previously treated small cell lung cancer. Clin Cancer Res. 16:5900–5907. 2010. View Article : Google Scholar : PubMed/NCBI

83 

Levy S, Verbeek WHM, Eskens FALM, van den Berg JG, de Groot DJA, van Leerdam ME and Tesselaar MET: First-line everolimus and cisplatin in patients with advanced extrapulmonary neuroendocrine carcinoma: A nationwide phase 2 single-arm clinical trial. Ther Adv Med Oncol. 14:175883592210770882022. View Article : Google Scholar : PubMed/NCBI

84 

Christopoulos P, Engel-Riedel W, Grohé C, Kropf-Sanchen C, von Pawel J, Gütz S, Kollmeier J, Eberhardt W, Ukena D, Baum V, et al: Everolimus with paclitaxel and carboplatin as first-line treatment for metastatic large-cell neuroendocrine lung carcinoma: a multicenter phase II trial. Ann Oncol. 28:1898–1902. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Ünal Ç and Sağlam S: Metronomic Temozolomide (mTMZ) and Bevacizumab-The Safe and Effective Frontier for Treating Metastatic Neuroendocrine Tumors (NETs): A Single-Center Experience. Cancers (Basel). 23:56882023. View Article : Google Scholar

86 

Horn L, Mansfield AS, Szczęsna A, Havel L, Krzakowski M, Hochmair MJ, Huemer F, Losonczy G, Johnson ML, Nishio M, et al: First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med. 379:2220–2229. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Liu SV, Reck M, Mansfield AS, Mok T, Scherpereel A, Reinmuth N, Garassino MC, De Castro Carpeno J, Califano R, Nishio M, et al: Updated overall survival and PD-L1 subgroup analysis of patients with extensive-stage small-cell lung cancer treated with atezolizumab, carboplatin, and etoposide (IMpower133). J Clin Oncol. 39:619–630. 2021. View Article : Google Scholar : PubMed/NCBI

88 

Paz-Ares L, Dvorkin M, Chen Y, Reinmuth N, Hotta K, Trukhin D, Statsenko G, Hochmair MJ, Özgüroğlu M, Ji JH, et al: Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): A randomised, controlled, open-label, phase 3 trial. Lancet. 394:1929–1939. 2019. View Article : Google Scholar : PubMed/NCBI

89 

Goldman JW, Dvorkin M, Chen Y, Reinmuth N, Hotta K, Trukhin D, Statsenko G, Hochmair MJ, Özgüroğlu M, Ji JH, et al: Durvalumab, with or without tremelimumab, plus platinum-etoposide versus platinum-etoposide alone in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): Updated results from a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 22:51–65. 2021. View Article : Google Scholar : PubMed/NCBI

90 

Chan DL, Rodriguez-Freixinos V, Doherty M, Wasson K, Iscoe N, Raskin W, Hallet J, Myrehaug S, Law C, Thawer A, et al: Avelumab in unresectable/metastatic, progressive, grade 2–3 neuroendocrine neoplasms (NENs): Combined results from NET-001 and NET-002 trials. Eur J Cancer. 169:74–81. 2022. View Article : Google Scholar : PubMed/NCBI

91 

Reddy SM, Reuben A and Wargo JA: Influences of BRAF inhibitors on the immune microenvironment and the rationale for combined molecular and immune targeted therapy. Curr Oncol Rep. 18:422016. View Article : Google Scholar : PubMed/NCBI

92 

Cheng Y, Wang Q, Li K, Shi J, Liu Y, Wu L, Han B, Chen G, He J, Wang J, et al: Anlotinib vs placebo as third- or further-line treatment for patients with small cell lung cancer: A randomised, double-blind, placebo-controlled Phase 2 study. Br J Cancer. 125:366–371. 2021. View Article : Google Scholar : PubMed/NCBI

93 

Wang J, Lu S, Yu X, Hu Y, Sun Y, Wang Z, Zhao J, Yu Y, Hu C, Yang K, et al: Tislelizumab plus chemotherapy vs chemotherapy alone as first-line treatment for advanced squamous non-small-cell lung cancer: A phase 3 randomized clinical trial. JAMA Oncol. 7:709–717. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Shi J, He B, Zhang HD, Jiang CY and Zhang L: Progress in the treatment of esophageal neuroendocrine carcinoma (Review). Oncol Lett 31: 14, 2026.
APA
Shi, J., He, B., Zhang, H.D., Jiang, C.Y., & Zhang, L. (2026). Progress in the treatment of esophageal neuroendocrine carcinoma (Review). Oncology Letters, 31, 14. https://doi.org/10.3892/ol.2025.15367
MLA
Shi, J., He, B., Zhang, H. D., Jiang, C. Y., Zhang, L."Progress in the treatment of esophageal neuroendocrine carcinoma (Review)". Oncology Letters 31.1 (2026): 14.
Chicago
Shi, J., He, B., Zhang, H. D., Jiang, C. Y., Zhang, L."Progress in the treatment of esophageal neuroendocrine carcinoma (Review)". Oncology Letters 31, no. 1 (2026): 14. https://doi.org/10.3892/ol.2025.15367
Copy and paste a formatted citation
x
Spandidos Publications style
Shi J, He B, Zhang HD, Jiang CY and Zhang L: Progress in the treatment of esophageal neuroendocrine carcinoma (Review). Oncol Lett 31: 14, 2026.
APA
Shi, J., He, B., Zhang, H.D., Jiang, C.Y., & Zhang, L. (2026). Progress in the treatment of esophageal neuroendocrine carcinoma (Review). Oncology Letters, 31, 14. https://doi.org/10.3892/ol.2025.15367
MLA
Shi, J., He, B., Zhang, H. D., Jiang, C. Y., Zhang, L."Progress in the treatment of esophageal neuroendocrine carcinoma (Review)". Oncology Letters 31.1 (2026): 14.
Chicago
Shi, J., He, B., Zhang, H. D., Jiang, C. Y., Zhang, L."Progress in the treatment of esophageal neuroendocrine carcinoma (Review)". Oncology Letters 31, no. 1 (2026): 14. https://doi.org/10.3892/ol.2025.15367
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team