Primary small cell carcinoma of the esophagus: progression in the last decade
Review Article

Primary small cell carcinoma of the esophagus: progression in the last decade

Anqi Ji1#, Runsen Jin1#, Renquan Zhang2, Hecheng Li1

1Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;2Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230000, China

Contributions: (I) Conception and design: All authors; (II) Administrative support: None; (III) Provision of study materials: None; (IV) Collection and assembly of data: None; (V) Data analysis and interpretation: None; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Hecheng Li, MD, PhD. Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China. Email: lihecheng2000@hotmail.com.

Abstract: Primary small cell carcinoma of the esophagus (PSCCE) is a highly malignant tumor that is diagnosed by endoscopic biopsy and immunohistochemistry. Because of its low incidence, a high degree of malignancy, and rapid progress, it is difficult to conduct large, randomized controlled trials and to establish a standard treatment plan for this disease. In recent years, several retrospective studies have been reported, and with the rise of emerging therapies, PSCCE has gradually become a focus of thoracic surgery. This paper reviews progress in the diagnosis and treatment of PSCCE in recent years.

Keywords: Primary small cell carcinoma; esophagus; treatment


Submitted Feb 17, 2020. Accepted for publication Mar 18, 2020.

doi: 10.21037/atm.2020.03.214


Introduction

Esophageal cancer is one of the most common digestive tract carcinomas in the world; a small cell carcinoma is a unique form of esophageal cancer (1-3). In 1952, McKeown first reported primary small cell carcinoma of the esophagus (PSCCE) (4), which often occurs in the lower and middle segments of the esophagus and accounts for 0.4–2.8% of all esophageal cancers (5,6). Because PSCCE is highly invasive and tends to metastasize, most patients are diagnosed with distant metastasis that often leads to poor prognosis (7,8); the 5-year overall survival (OS) rate is approximately 6.7–18% (9-13). However, due to the low incidence of PSCCE, there have been no large, randomized controlled trials, and the standard treatment is controversial. Considering the systemic nature of PSCCE, multidisciplinary systemic treatments are typically utilized (9,14,15). This article will summarize the recent PSCCE literature, supply insights for the clinical management of PSCCE, and to focus on future treatment options.


General clinical characteristics

Clinical symptoms

The symptoms of patients with PSCCE are like those of esophageal squamous cell carcinoma (ESCC). The most common symptom is progressive dysphagia, and some patients present with symptoms such as pain during swallowing and weight loss (16,17). According to statistics, the average male to female ratio of PSCCE is approximately 3:1, although this ratio varies significantly around the globe (9,17). In China, PSCCE occurs most often in the middle esophagus, while in Western countries, PSCCE is most common in the lower esophagus (18). Similar to ESCC, besides drinking and smoking history, the risk factors for PSCCE are not defined (19-21).

Diagnosis

Most PSCCE cases present as ulcerative or diffuse types on gastroscopies; however, there are no specific differences between PSCCE, ESCC, and esophageal adenocarcinoma (EAC) in imaging examinations (5). Gastroscopy biopsy is the most used method for preoperative diagnosis. Unfortunately, the preoperative definite diagnosis rate is low, mainly due to the small volume of tissue collected during gastroscopy biopsies and the coexistence of small cell carcinoma, squamous cell carcinoma, and adenocarcinoma components in the same tumor tissue (22,23). Thus, when diagnosing complex small cell carcinomas, it may be difficult to distinguish them from poorly differentiated squamous cell carcinomas. To improve the diagnostic rate, some scholars recommend the use of fine-needle aspiration guided by ultrasonic gastroscopy to perform biopsies of submucosal lesions more accurately (5,24).

Tumor staging

Tumor staging is crucial for determining the best choice of treatment options. The American Joint Committee on Cancer (AJCC) staging system (25) and the Veterans Administration Lung Study Group staging system (VALSG) are generally used for PSCCE staging (26).

AJCC: According to the TNM staging system of esophageal cancer, patients are divided into T and M stages (8th edition 2017). Cases without regional lymph node metastasis are defined as N0, cases with one to three regional lymph node metastases are defined as N1, and cases with more than three regional lymph node metastases are defined as N2.

VALSG: the VALSG staging system for primary pulmonary small cell carcinomas divides cases of PSCCE into limited disease (LD) and extensive disease (ED). LD is defined as a tumor confined to the esophagus or surrounding tissue, with or without regional lymph node metastasis. ED is defined as tumors outside the locoregional boundaries (17).

Some studies have combined the AJCC and VALSG staging systems to further divide the LD group into two groups: local disease and regional disease. Localized disease is defined as tumors confined to the esophagus without lymph node metastasis (T1-4aN0M0). The regional disease is defined as tumors that have directly invaded surrounding organs or tissues or have regional lymph node metastases (T4b/N+M0). ED is defined as tumors that extend to distant lymph nodes or organs (M1) (18).

Prognosis

PSCCE is a rare and extremely aggressive malignancy. The median survival time (MST) is approximately 14 to 28 months, with a 1-year OS of 56-86%, a 3-year OS of 27.3–35.7%, and a 5-year OS of 6.7–18% (5,27,28). Most studies indicate that patients with PSCCE are prone to lymph node metastasis, which is associated with poor prognosis (29,30). According to a study in patients with no lymph node metastasis, the MST is 44.9 months, and the 3-year OS was 50.5%; for patients with lymph node metastasis, the MST is 17.8 months, and the 3-year OS was 31.6% (27). Besides, some studies found that therapy, tumor size, and infiltration depth were also associated with poor prognosis (5,31).


Biological characteristics

Molecular-related biological characteristics

The molecular-related biological characteristics of PSCCE is significant for diagnosis and prognosis. The immunohistochemistry of specific neuroendocrine markers, such as synaptophysin (Syn), chromogranin A (CgA), neuron-specific enolase (NSE), cytokeratin (CK), and neuronal cell adhesion molecules (CD56), can help to distinguish PSCCE from neuroendocrine cells, poorly differentiated squamous cells, or adenocarcinoma cells to improve diagnosis (32). However, these markers are also associated with the patient’s prognosis. A study on immunohistochemical markers included 73 patients with PSCCE and evaluation of thyroid transcription factor-1 (ttf-1), NSE, Syn, and CgA expression. Patients that were negative for all four markers had the worst prognosis, and patients with one positive marker had better prognoses than those with all negative markers (15.3 vs. 6.1 months, P=0.002) (33). Another study showed that patients with high CgA expression had increased survival times compared with those that were negative for CgA expression (34).

Moreover, the high expression of Ki-67 is a defining characteristic of small cell carcinoma. Ki-67 is generally labeling index is higher than 50% in small cell cancers and less than 25% in other types of tumors, which makes Ki-67 expression a valuable marker for the diagnosis of PSCCE (5,35,36). Besides, a retrospective study showed that high expression of Ki-67 was also a favorable prognostic factor in patients with PSCCE (P=0.012). Furthermore, in a stratified analysis, adjuvant therapy resulted in significant survival benefits only for patients with high Ki-67 expression (37).

Prognosis-related biological characteristics

Prognosis evaluation is particularly crucial for PSCCE patients because it affects treatment options. Besides molecular-related biological characteristics, inflammation biomarkers and microRNA also play an essential role in evaluating the prognosis of patients.

Increased attention has been paid to the role of systemic inflammatory responses in tumor genesis, development, and metastasis (38,39) because, in the tumor microenvironment, inflammatory cells can be involved in angiogenesis, viability, mobility, and invasion (40). Inflammatory biomarkers are associated with the prognosis of different types of cancers, such as liver cancer, lung cancer, and ESCC, and patients’ prognoses can be adequately assessed using pre-treatment hematologic biomarkers (40-42). A study retrospectively analyzed the independent prognostic factors for neutrophil-to-lymphocyte ratio in patients identified as having PSCCE (43). Another retrospective study found that a high platelet-to-lymphocyte ratio was an independent prognostic factor for poor OS (44).

With the in-depth study of microRNAs, evaluation of PSCCE patient prognosis via microRNA expression is receiving more attention. Okumura et al. used microarrays to detect the microRNA expression in PSCCE tumors. The expression of eight microRNAs (miR-4323, miR-625, miR-3619-3p, miR-4419b, miR-1249, miR-4648, miR-4664-3p, and miR-1203) was significantly correlated with tumor recurrence (P<0.01) (45).

Potential target-related molecules

Due to the low incidence of PSCCE, the study of esophageal cancer genes has focused on ESCC and EAC. However, with high-throughput genome technology, PSCCE has gradually received more attention. To understand the genetic basis of PSCCE, Wang et al. performed genomic profiling of 55 patients with PSCCE using whole-exome sequencing confirmed by ultra-deep targeted sequencing. Significant mutations were detected in eight genes (TP53, RB1, NOTCH1, FAT1, FBXW7, PDE3A, PTPRM, and CBLN2) that provided assistance for the development of new diagnostic and therapeutic tools for PSCCE therapy (46). Furthermore, Ishida et al. (47) found that loss of Rb1 gene expression and overexpression of the SOX gene were crucial for the pathogenesis and differentiation of PSCCE. The incidence of phosphatase and tensin homolog deleted on revision 10 (PTEN) mutations in Chinese patients with PSCCE is high, and the over-expression of p21-activated kinase-1 (PAK-1) is associated with poor prognosis of patients with PSCCE. Thus, PTEN and PAK-1 may be potential targets for precise treatment of patients with PSCCE (48,49).


Treatment strategies

Traditional treatment strategy

Due to the low incidence of esophageal small cell carcinoma, the rapid occurrence of lymph node metastasis, and poor prognosis, it is difficult to carry out a large-scale randomized controlled trial to establish standard treatment options. Thus, most studies on PSCCE treatment have focused on retrospective studies. Furthermore, it is controversial whether surgery can help PSCCE patients. Many scholars believe that chemotherapy (CT) or radiotherapy should be the main treatment for PSCCE patients (6,10,12,50-52). However, some scholars hold an opposing view that surgical treatment plays an important role in the treatment of PSCCE (29,30). We believe that different multidisciplinary treatment regimens should be adopted for patients with different stages of PSCCE (53). Therefore, traditional treatment strategies for distinct stages of PSCCE are summarized.

Treatment strategies according to AJCC staging

Stages I and IIA

Some studies suggest that patients with early esophageal cancer could be considered for endoscopic treatment (54,55); however, there are currently no studies on endoscopic treatment of patients with early PSCCE. The FFCD 9901 trial showed that neoadjuvant therapy did not improve the productivity of patients with early esophageal cancer and that it could lead to an increase in postoperative mortality. Therefore, surgical treatment should be considered for early esophageal cancer (56). Similar conclusions have been reached in studies of stage I and IIA PSCCE, suggesting that early PSCCE should be treated first with surgery. Xu et al. (5) retrospectively analyzed 152 patients with PSCCE and found through stratified analysis that surgical treatment can result in survival benefits for patients with stage I and IIA; postoperative adjuvant therapy failed to improve patients’ OS (P=0.522) and disease-free survival (DFS) (P=0.368). Chen et al. (57) also suggested that surgery could lead to better MST for stage I and IIA patients (29 vs. 17.4 months, P=0.082). In conclusion, for patients with stage I and IIA, most scholars believe that surgical treatment should be the main treatment.

Stage IIB

The best treatment for patients with stage IIB PSCCE has been controversial. For patients with stage IIB PSCCE, Xu et al. (5) indicated that the OS and DFS were similar irrespective of whether the patients received only surgical treatment, postoperative CT, or chemoradiotherapy (CRT). However, Chen et al. (57) indicated that for patients with stage IIB PSCCE and above, postoperative CT can improve survival rates compared with surgical treatment alone (13 vs. 6.1 months, P=0.003), while the addition of RT also can improve survival benefits (16.8 vs. 9.5 months, P=0.076). A similar conclusion was reached by Ishida et al. (58), who concluded that non-surgical therapy was more beneficial for patients after stage II PSCCE than for patients with surgical therapy (16 vs. 11.5 months, P=0.097). Therefore, large-scale retrospective studies or randomized controlled trials are still needed to confirm the best treatment options for patients with stage IIB. Xu et al. (5) presented that the two patients who received neoadjuvant chemotherapy (nCT) had a longer survival time. However, due to the small sample size, it was not possible to further evaluate the efficacy of nCT in patients with stage IIB. Meanwhile, in the CROSS trial, van Hagen et al. (59) included a subset of patients with stage IIB esophageal cancer and showed that nCT improved survival (P=0.003). Hence, for patients with stage IIB PSCCE, the role of nCT in treatment must be further explored.

Stages III and IV

Most studies have shown CRT as the primary treatment for patients with stage III or above PSCCE. Chen et al. (57) showed that, compared with CT alone, CRT could improve the survival rate of stage IV patients (13.2 vs. 8.9 months, P=0.014). Similarly, a study by Wong et al. (27), which included PSCCE patients of whom 40% were stage IV, also found that CRT improved OS in patients with lymph node metastasis. However, a retrospective study by Hou et al. (9) involving 89 patients with stage III PSCCE found that surgical treatment combined with CRT resulted in longer survival times. Moreover, a randomized controlled study by Shapiro et al. (60) showed a greater survival benefit from nCT in patients with stage III or higher esophageal cancer than with surgery alone. A study of PSCCE patients by Xu et al. (5) reached a similar conclusion. However, the determination of the best scheme of nCT needs further discussion.

Treatment strategies according to VALSG staging

VALSG staging is also a commonly used tumor staging method that classifies patients into LD and ED stages according to whether the tumor has distant metastases. Zhu et al. (22) used VALSG for staging in 64 patients with PSCCE; multivariate analysis showed that treatment modality was an independent prognostic factor (P=0.008). The combination of surgical treatment and CT helped the survival of LD patients, while combined CT had a significant impact on the MST of ED patients (P=0.0001). Furthermore, a recent study by Chen et al. (50) demonstrated that, for patients with stage LD PSCCE, combined CT and RT could improve prognosis (P=0.046), as well as increased radiation doses (≥56 Gy, P=0.027). However, this study did not examine whether surgical treatment resulted in increased survival benefits. Jeene et al. (6) proposed a contrary view, suggesting that lower radiation doses (<45 Gy) were associated with improved survival; their multivariate analysis showed that only the number of CT cycles was associated with better survival (P=0.006). However, both studies included only a small number of patients, so large-scale retrospective studies or randomized controlled studies are needed to clarify further the role of RT in the treatment of LD-PSCCE.

Considering that more accurate tumor staging could enable patients to obtain more accurate treatment regimens, Zou et al. (31) further subdivided patients: patients with T1-2N0M0 were defined as LD I, while patients with T3-4N0M0 or T1-4N1-2M0 were defined as LD II. Stratified analysis showed that for patients with LD I, surgical treatment was recommended, and there was no significant improvement in survival in patients with postoperative adjuvant CT or CRT. Alternatively, adjuvant CT improved the survival rate of PSCCE patients with fully resected LD II disease.

After using propensity score matching to balance demographic factors (patients treated in China and the United States), Xiao et al. (18) used VALSG staging to classify PSCCE patients, dividing them into LD and ED. The further classification was performed according to the presence of lymph node metastasis, LD is divided into focal lesions (T1-4aN0M0), and regional lesions (T4b/N+M0). The stratified analysis showed that, for patients with focal lesions, surgery could be beneficial, but CT does not improve survival. In patients with regional lesions, ED, RT, and CT had improved OS.

In conclusion, for patients with early surgically resectable PSCCE, most studies support the use of surgical treatment to improve patient survival. For some patients, CT and RT can also be considered, but the choice of CT options and radiation doses still is controversial. For patients with advanced PSCCE (stage III and IV or ED), CT combined with RT can improve the survival rate. These patients may also consider nCT, as it may reduce the tumor stage; When tumor staging decreases, surgical treatment can increase survival benefits.

Emerging therapies

With the rise of targeted therapy and immunotherapy, several clinical trials are underway to study the application of these therapies in tumor therapy. Although studies on targeted therapy and immunotherapy for esophageal cancer have focused on ESCC and EAC, these emerging therapies may also be right for PSCCE, which is often a mixture of squamous and adenocarcinoma cells. The following section will briefly introduce the emerging therapies for esophageal cancer.

Targeted therapy

Except for trastuzumab, the development of targeted therapies for esophageal cancer over the past decade has been disappointing (61). Several large, international randomized trials have investigated drugs targeting the EGFR, MET/hGF, mTOR, VEGF, and FGFR pathways without satisfactory results (62-64). For example, cetuximab, a humanized mouse EGFR monoclonal antibody, combined with CT, significantly improved OS in patients with esophageal cancer in the phase II clinical study (65). However, other studies found that cetuximab combined with CT has no advantage over CT alone (63). Moreover, a phase III clinical study involving 450 patients with esophageal cancer assessed the role of gefitinib in the progression of esophageal cancer after CT and also found no improvement in OS (66).

On the other hand, trastuzumab, a drug that targets human epidermal growth factor receptor 2 (HER-2), has gained attention in different types of HER-2-overexpressing cancers because of its success in targeted breast cancer therapy (67). In a Japanese clinical study, trastuzumab combined with capecitabine/cisplatin or 5-fluorouracil/cisplatin improved OS in patients with advanced esophageal cancer compared to CT alone (68). Meanwhile, a phase II study in Spain also found that trastuzumab combined with cisplatin as a first-line treatment for advanced esophageal cancer with positive HER-2 improved prognosis and had good safety (69). However, the overall development of targeted therapy for esophageal cancer has been prolonged. The emergence of next generation sequencing technology will supply a new direction for targeted therapy for esophageal cancer.

Immunotherapy

Immune checkpoint inhibitors

Programmed death ligand 1 (PD-L1) is expressed in up to 40% of esophageal cancers, and the expression of PD-L1 increases after conventional neoadjuvant CRT (19,70). Therefore, drugs targeting PD-L1 are considered useful in patients with PD-L1-positive esophageal cancer. Two PD-L1 inhibitors, pembrolizumab, and nivolumab have achieved satisfactory efficacy in esophageal cancer.

Pembrolizumab, a PD-1 inhibitor, was the first Food and Drug Administration (FDA)-approved immune checkpoint inhibitor for the treatment of advanced melanoma and was effective against esophageal cancer in several recent clinical trials. KEYNOTE-028 is a multi-cohort phase IB study designed to evaluate the safety and overall remission rate of pembrolizumab in advanced PD-L1-positive solid tumors. The study included 23 patients with PD-L1-positive advanced and metastatic esophageal cancer. The objective response rate was 30% [95% confidence interval (CI), 13–53%] with an average response duration of 15 months (ranging from 6 to ≥26 months). Treatment-related adverse events were present in 39% of patients (9 out of 23). The main adverse events included decreased appetite, decreased lymphocyte count, and a systemic rash. More than half of the patients showed tumor shrinkage. Overall, median progression-free survival was 1.8 months (95% CI, 1.7–2.9 months), and median OS was 7.0 months (95% CI, 4.3–17.7 months) (71,72). KEYNOTE-180 is a phase II single-arm study designed to evaluate the safety and efficacy of pembrolizumab. A total of 121 patients with advanced metastatic esophageal cancer who had progressed after a two-line treatment were enrolled. Compared to the previous traditional second-line treatment, the experimental results of this study were very satisfactory, with a median OS of 5.8 months and 6- and 12-month OS rates of 49% and 28%, respectively (62,72). The KEYNOTE-181 phase III clinical trial will assess pembrolizumab with CT as a second-line treatment of advanced esophageal cancer. Another phase III study (KEYNOTE-590) is comparing pembrolizumab combined with CT and placebo combined with CT (72).

Nivolumab is a human monoclonal IgG4 antibody that inhibits PD-1 expression on activated T cells. Nivolumab is FDA approved for the treatment of metastatic melanoma, non-small cell lung cancer, and renal cell carcinoma (72). A randomized, double-blind, multicenter phase III trial in Japan was designed to evaluate the efficacy and safety of nivolumab in patients with advanced gastric or gastroesophageal junction cancer, who had previously received two or more CT regimens. That study suggested that nivolumab can lead to longer survival times (5.26 vs. 4.14 months, P<0.0001) (73). Moreover, CheckMate-032 demonstrated the safety and efficacy of nivolumab combined with ipilimumab in patients with advanced esophageal cancer; the combination was superior to nivolumab monotherapy (74).

Other immunotherapies

Other emerging immunotherapies such as peptide vaccines, adoptive T-cell transfer, and oncolytic viruses are also receiving increasing attention. Clinical trials are studying their applications in the treatment of esophageal cancer.

Since several immunogenic cancer antigens (ICA) have been shown on esophageal cancer cells and therapeutic cancer vaccines, they have received increasing attention. Cancer vaccines are designed to effectively induce ICA-specific cytotoxic T lymphocytes to enhance the immune response (72).

The adoptive T-cell transfer is another research hotspot. This treatment removes T cells from the patient, alters the T cells in vitro to increase their immunologic activity, and then reintroduces them into the patient to improve specific immune responses. Related studies have shown preliminary clinical benefits in patients with esophageal cancer (75).

Many scholars believe that oncolytic virus therapy may be the next breakthrough in cancer immunotherapy. Oncolytic viruses can selectively replicate in tumor cells and then induce tumor cell lysis (76,77). Recently, a phase I/II study showed the efficiency and tolerance of a novel telomerase-specific oncolytic virus (OBP-301) in elderly patients with esophageal cancer (78).


Conclusions

The incidence of PSCCE is low, but the progress is rapid, and the prognosis is poor. Despite the growing interest in PSCCE, research has focused on retrospective studies. Significant advancements will require extensive, randomized controlled studies to determine the best treatment strategy. The survival rate of patients diagnosed with early PSCCE is notable; thus, early diagnosis is critical, requiring improved biopsy equipment and immunohistochemical tests. With a deepening understanding of the molecular biology and tumor microenvironment, the use of molecular biomarkers, inflammatory biomarkers, and microRNAs to help improve the diagnostic accuracy and evaluate the prognosis of PSCCE patients has received more attention.

Additionally, proper tumor staging is vital for the choice of treatment options for PSCCE patients. Surgical treatment is the preferred treatment for early-stage patients without lymph node metastasis. However, the benefits of neoadjuvant therapy in patients with partial lymph node metastasis or large tumors requires further study. For patients with advanced PSCCE, systemic treatment, such as radiotherapy and CT, should be considered first. If nCT can reduce tumor staging, surgical treatment may be considered to maximize the survival benefit. With targeted therapy and immunotherapy becoming the focus of cancer therapy and despite current research focusing on ESCC and EAC, future PSCCE patients will have new treatment options.


Acknowledgments

Funding: Our review was supported by Shanghai Municipal Education Commission-Gaofeng Clinical Medicine Grant (20172005), Shanghai Municipal Commission of Health and Family Planning Outstanding Academic Leaders Training Program (2017BR055) and Guangci Distinguished Young Scholars Training Program (GCQN-2018-A09).


Footnote

Conflicts of Interest: All authors have completed the ICMJE uniform (available at http://dx.doi.org/10.21037/atm.2020.03.214). 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.

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. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394-424. [Crossref] [PubMed]
  2. Chen W, Zheng R, Baade PD, et al. Cancer statistics in China, 2015. CA Cancer J Clin 2016;66:115-32. [Crossref] [PubMed]
  3. Medgyesy CD, Wolff RA, Putnam JB, et al. Small cell carcinoma of the esophagus: the University of Texas M. D. Anderson Cancer Center experience and literature review. Cancer 2000;88:262-7. [Crossref] [PubMed]
  4. McKeown F. Oat-cell carcinoma of the oesophagus. The Journal of Pathology and Bacteriology 1952;64:889-91. [Crossref] [PubMed]
  5. Xu L, Li Y, Liu X, et al. Treatment Strategies and Prognostic Factors of Limited-Stage Primary Small Cell Carcinoma of the Esophagus. J Thorac Oncol 2017;12:1834-44. [Crossref] [PubMed]
  6. Jeene PM, Geijsen ED, Muijs CT, et al. Small Cell Carcinoma of the Esophagus: A Nationwide Analysis of Treatment and Outcome at Patient Level in Locoregional Disease. Am J Clin Oncol 2019;42:534-8. [Crossref] [PubMed]
  7. Cicin I, Karagol H, Uzunoglu S, et al. Extrapulmonary small cell carcinoma compared with small cell lung carcinoma: a retrospective single-center study. Cancer 2007;110:1068-76. [Crossref] [PubMed]
  8. Brenner B, Tang LH, Klimstra DS, et al. Small cell carcinomas of the gastrointestinal tract: a review. J Clin Oncol 2004;22:2730-9. [Crossref] [PubMed]
  9. Hou X, Wei J-C, Wu J-X, et al. Multidisciplinary Modalities Achieve Encouraging Long-Term Survival in Resectable Limited-Disease Esophageal Small Cell Carcinoma. PLoS One 2013;8:e69259. [Crossref] [PubMed]
  10. Lv J, Liang J, Wang J, et al. Primary small cell carcinoma of the esophagus. J Thorac Oncol 2008;3:1460-5. [Crossref] [PubMed]
  11. Zhang BH, Yang WJ, Zhao L, et al. Surgical treatment and prognostic analysis of 109 patients with primary esophageal small cell carcinoma. Zhonghua Zhong Liu Za Zhi 2012;34:698-702. [PubMed]
  12. Meng MB, Zaorsky NG, Jiang C, et al. Radiotherapy and chemotherapy are associated with improved outcomes over surgery and chemotherapy in the management of limited-stage small cell esophageal carcinoma. Radiother Oncol 2013;106:317-22. [Crossref] [PubMed]
  13. Chen SB, Yang JS, Yang WP, et al. Treatment and prognosis of limited disease primary small cell carcinoma of esophagus. Dis Esophagus 2011;24:114-9. [Crossref] [PubMed]
  14. Wang HH, Zaorsky NG, Meng MB, et al. Multimodality therapy is recommended for limited-stage combined small cell esophageal carcinoma. Onco Targets Ther 2015;8:437-44. [PubMed]
  15. Ding J, Ji J, Zhu W, et al. A retrospective study of different treatments of limited-stage small cell esophageal carcinoma and associated prognostic factor analysis. Dis Esophagus 2013;26:696-702. [PubMed]
  16. Schuerle T, Aoun E, Farah K. Small cell carcinoma of the oesophagus: a rare cause of dysphagia. BMJ Case Rep 2013;2013. doi: 10.1136/bcr-2013-200468. [Crossref]
  17. Lu XJ, Luo JD, Ling Y, et al. Management of Small Cell Carcinoma of Esophagus in China. J Gastrointest Surg 2013;17:1181-7. [Crossref] [PubMed]
  18. Xiao Q, Xiao H, Ouyang S, et al. Primary small cell carcinoma of the esophagus: Comparison between a Chinese cohort and Surveillance, Epidemiology, and End Results (SEER) data. Cancer Med 2019;8:1074-85. [Crossref] [PubMed]
  19. Kojima T, Doi T. Immunotherapy for Esophageal Squamous Cell Carcinoma. Curr Oncol Rep 2017;19:33. [Crossref] [PubMed]
  20. Liu Y, Chen H, Sun Z, et al. Molecular mechanisms of ethanol-associated oro-esophageal squamous cell carcinoma. Cancer Lett 2015;361:164-73. [Crossref] [PubMed]
  21. Bagnardi V, Rota M, Botteri E, et al. Alcohol consumption and site-specific cancer risk: a comprehensive dose-response meta-analysis. Br J Cancer 2015;112:580-93. [Crossref] [PubMed]
  22. Zhu Y, Qiu B, Liu H, et al. Primary small cell carcinoma of the esophagus: review of 64 cases from a single institution. Dis Esophagus 2014;27:152-8. [Crossref] [PubMed]
  23. Nayal B, Vasudevan G, Rao ACK, et al. Primary Small Cell Carcinoma of The Esophagus - An Eight Year Retrospective Study. J Clin Diagn Res 2015;9:EC04-6. [PubMed]
  24. Moon JS. Endoscopic ultrasound-guided fine needle aspiration in submucosal lesion. Clinical Endoscopy 2012;45:117-23. [Crossref] [PubMed]
  25. Rice TW, Rusch VW, Ishwaran H, et al. Cancer of the esophagus and esophagogastric junction: data-driven staging for the seventh edition of the American Joint Committee on Cancer/International Union Against Cancer Cancer Staging Manuals. Cancer 2010;116:3763-73.
  26. Micke P, Faldum A, Metz T, et al. Staging small cell lung cancer: Veterans Administration Lung Study Group versus International Association for the Study of Lung Cancer--what limits limited disease? Lung Cancer 2002;37:271-6. [Crossref] [PubMed]
  27. Wong AT, Shao M, Rineer J, et al. Treatment and survival outcomes of small cell carcinoma of the esophagus: an analysis of the National Cancer Data Base. Dis Esophagus 2017;30:1-5. [PubMed]
  28. Tao H, Li F, Wang J, et al. Management of treatment-naïve limited-stage small cell esophagus carcinoma. Saudi Med J 2015;36:297-303. [Crossref] [PubMed]
  29. Xie MR, Xu SB, Sun XH, et al. Role of surgery in the management and prognosis of limited-stage small cell carcinoma of the esophagus. Dis Esophagus 2015;28:476-82. [Crossref] [PubMed]
  30. Situ D, Lin Y, Long H, et al. Surgical treatment for limited-stage primary small cell cancer of the esophagus. Ann Thorac Surg 2013;95:1057-62. [Crossref] [PubMed]
  31. Zou B, Li T, Zhou Q, et al. Adjuvant Therapeutic Modalities in Primary Small Cell Carcinoma of Esophagus Patients. Medicine 2016.95. [PubMed]
  32. Beasley MB. Immunohistochemistry of pulmonary and pleural neoplasia. Arch Pathol Lab Med 2008;132:1062-72. [PubMed]
  33. Zhang Y, Li C, Chen M. Prognostic value of immunohistochemical factors in esophageal small cell carcinoma (ESCC): analysis of clinicopathologic features of 73 patients. J Thorac Dis 2018;10:4023-31. [Crossref] [PubMed]
  34. Ku JW, Zhang DY, Song X, et al. Characterization of tissue chromogranin A (CgA) immunostaining and clinicohistopathological changes for the 125 Chinese patients with primary small cell carcinoma of the esophagus. Dis Esophagus 2017;30:1-7. [Crossref] [PubMed]
  35. Aslan DL, Gulbahce HE, Pambuccian SE, et al. Ki-67 immunoreactivity in the differential diagnosis of pulmonary neuroendocrine neoplasms in specimens with extensive crush artifact. Am J Clin Pathol 2005;123:874-8. [Crossref] [PubMed]
  36. Zheng G, Ettinger DS, Maleki Z. Utility of the quantitative Ki-67 proliferation index and CD56 together in the cytologic diagnosis of small cell lung carcinoma and other lung neuroendocrine tumors. Acta Cytol 2013;57:281-90. [Crossref] [PubMed]
  37. Deng HY, Chen ZH, Wang ZQ, et al. High expression of Ki-67 is an independent favorable prognostic factor for esophageal small cell carcinoma. Oncotarget 2017;8:55298-307. [Crossref] [PubMed]
  38. Grivennikov SI, Greten FR, Karin M. Immunity, Inflammation, and Cancer. Cell 2010;140:883-99. [Crossref] [PubMed]
  39. Thibodeau J, Bourgeois-Daigneault MC, Lapointe R. Targeting the MHC Class II antigen presentation pathway in cancer immunotherapy. Oncoimmunology 2012;1:908-16. [Crossref] [PubMed]
  40. Wang J, Jia Y, Wang N, et al. The clinical significance of tumor-infiltrating neutrophils and neutrophil-to-CD8+ lymphocyte ratio in patients with resectable esophageal squamous cell carcinoma. J Transl Med 2014;12:7. [Crossref] [PubMed]
  41. Aleksandrova K, Boeing H, Nöthlings U, et al. Inflammatory and metabolic biomarkers and risk of liver and biliary tract cancer. Hepatology 2014;60:858-71. [Crossref] [PubMed]
  42. Tang C, Liao Z, Gomez D, et al. Lymphopenia association with gross tumor volume and lung V5 and its effects on non-small cell lung cancer patient outcomes. Int J Radiat Oncol Biol Phys 2014;89:1084-91. [Crossref] [PubMed]
  43. Wang Y, Liu JF. A retrospective study on the prognostic value of preoperative neutrophil/lymphocyte ratio in patients with primary small cell carcinoma of the esophagus. Onco Targets Ther 2017;10:2453-9. [Crossref] [PubMed]
  44. Wang N, Li X, Luo H, et al. Prognostic value of pretreatment inflammatory biomarkers in primary small cell carcinoma of the esophagus. Thoracic Cancer 2019;10:1913-8. [Crossref] [PubMed]
  45. Okumura T, Shimada Y, Omura T, et al. MicroRNA profiles to predict postoperative prognosis in patients with small cell carcinoma of the esophagus. Anticancer Res 2015;35:719-27. [PubMed]
  46. Wang F, Liu D-B, Zhao Q, et al. The genomic landscape of small cell carcinoma of the esophagus. Cell Res 2018;28:771-4. [Crossref] [PubMed]
  47. Ishida H, Kasajima A, Kamei T, et al. SOX2 and Rb1 in esophageal small cell carcinoma: their possible involvement in pathogenesis. Mod Pathol 2017;30:660-71. [Crossref] [PubMed]
  48. Gan J, Zhang Y, Ke X, et al. Dysregulation of PAK1 Is Associated with DNA Damage and Is of Prognostic Importance in Primary Esophageal Small Cell Carcinoma. Int J Mol Sci 2015;16:12035-50. [Crossref] [PubMed]
  49. Zhang Z, Xiao H, Xie F, et al. High-incidence of PTEN mutations in Chinese patients with primary small cell carcinoma of the esophagus. BMC Cancer 2014;14:19. [Crossref] [PubMed]
  50. Chen B, Yang H, Ma H, et al. Radiotherapy for small cell carcinoma of the esophagus: outcomes and prognostic factors from a retrospective study. Radiat Oncol 2019;14:210. [Crossref] [PubMed]
  51. Song Y, Wang W, Tao G, et al. Survival benefit of radiotherapy to patients with small cell esophagus carcinoma - an analysis of Surveillance Epidemiology and End Results (SEER) data. Oncotarget 2016;7:15474-80. [PubMed]
  52. Gao R, Zhang Y, Wen XP, et al. Chemotherapy with cisplatin or carboplatin in combination with etoposide for small cell esophageal cancer: a systemic analysis of case series. Dis Esophagus 2014;27:764-9. [Crossref] [PubMed]
  53. Kukar M, Groman A, Malhotra U, et al. Small cell carcinoma of the esophagus: a SEER database analysis. Ann Surg Oncol 2013;20:4239-44. [Crossref] [PubMed]
  54. Domper Arnal MJ, Ferrández Arenas Á, Lanas Arbeloa Á. Esophageal cancer: Risk factors, screening and endoscopic treatment in Western and Eastern countries. World J Gastroenterol 2015;21:7933-43. [Crossref] [PubMed]
  55. Altorki NK, Lee PC, Liss Y, et al. Multifocal neoplasia and nodal metastases in T1 esophageal carcinoma: implications for endoscopic treatment. Ann Surg 2008;247:434-9. [Crossref] [PubMed]
  56. Mariette C, Dahan L, Mornex F, et al. Surgery alone versus chemoradiotherapy followed by surgery for stage I and II esophageal cancer: final analysis of randomized controlled phase III trial FFCD 9901. J Clin Oncol 2014;32:2416-22. [Crossref] [PubMed]
  57. Chen WW, Wang F, Chen S, et al. Detailed analysis of prognostic factors in primary esophageal small cell carcinoma. Ann Thorac Surg 2014;97:1975-81. [Crossref] [PubMed]
  58. Ishida H, Kasajima A, Onodera Y, et al. A comparative analysis of clinicopathological factors between esophageal small cell and basaloid squamous cell carcinoma. Medicine 2019.98. [PubMed]
  59. van Hagen P, Hulshof MC, van Lanschot JJ, et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 2012;366:2074-84. [Crossref] [PubMed]
  60. Shapiro J, van Lanschot JJB, Hulshof MCCM, 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 2015;16:1090-8. [Crossref] [PubMed]
  61. Luo HQ, He YF, Yan Y, et al. The role of prostaglandin E receptor 2 and epidermal growth factor receptor in esophageal squamous cell carcinoma patients with (pN+) regional lymph node metastasis. Transl Cancer Res 2019;8:1233-41. [Crossref]
  62. Shah MA, Bang YJ, Lordick F, et al. Effect of Fluorouracil, Leucovorin, and Oxaliplatin With or Without Onartuzumab in HER2-Negative, MET-Positive Gastroesophageal Adenocarcinoma. JAMA Oncol 2017;3:620-7. [Crossref] [PubMed]
  63. Lordick F, Kang YK, Chung HC, et al. Capecitabine and cisplatin with or without cetuximab for patients with previously untreated advanced gastric cancer (EXPAND): a randomised, open-label phase 3 trial. Lancet Oncol 2013;14:490-9. [Crossref] [PubMed]
  64. Waddell T, Chau I, Cunningham D, et al. Epirubicin, oxaliplatin, and capecitabine with or without panitumumab for patients with previously untreated advanced oesophagogastric cancer (REAL3): a randomised, open-label phase 3 trial. Lancet Oncol 2013;14:481-9. [Crossref] [PubMed]
  65. Enzinger PC, Burtness BA, Niedzwiecki D, et al. CALGB 80403 (Alliance)/E1206: A Randomized Phase II Study of Three Chemotherapy Regimens Plus Cetuximab in Metastatic Esophageal and Gastroesophageal Junction Cancers. J Clin Oncol 2016;34:2736-42. [Crossref] [PubMed]
  66. Dutton SJ, Ferry DR, Blazeby JM, et al. Gefitinib for oesophageal cancer progressing after chemotherapy (COG): a phase 3, multicentre, double-blind, placebo-controlled randomised trial. Lancet Oncol 2014;15:894-904. [Crossref] [PubMed]
  67. Belkhiri A, El-Rifai W. Advances in targeted therapies and new promising targets in esophageal cancer. Oncotarget 2015;6:1348-58. [Crossref] [PubMed]
  68. Sawaki A, Ohashi Y, Omuro Y, et al. Efficacy of trastuzumab in Japanese patients with HER2-positive advanced gastric or gastroesophageal junction cancer: a subgroup analysis of the Trastuzumab for Gastric Cancer (ToGA) study. Gastric Cancer 2012;15:313-22. [Crossref] [PubMed]
  69. Grávalos C, Gómez-Martín C, Rivera F, et al. Phase II study of trastuzumab and cisplatin as first-line therapy in patients with HER2-positive advanced gastric or gastroesophageal junction cancer. Clin Transl Oncol 2011;13:179-84. [Crossref] [PubMed]
  70. Lim SH, Hong M, Ahn S, et al. Changes in tumour expression of programmed death-ligand 1 after neoadjuvant concurrent chemoradiotherapy in patients with squamous oesophageal cancer. Eur J Cancer 2016;52:1-9. [Crossref] [PubMed]
  71. Doi T, Piha-Paul SA, Jalal SI, et al. Safety and Antitumor Activity of the Anti-Programmed Death-1 Antibody Pembrolizumab in Patients With Advanced Esophageal Carcinoma. J Clin Oncol 2018;36:61-7. [Crossref] [PubMed]
  72. Tu CC, Hsu PK. The frontline of esophageal cancer treatment: questions to be asked and answered. Ann Transl Med 2018;6:83. [Crossref] [PubMed]
  73. Kang YK, Boku N, Satoh T, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390:2461-71. [Crossref] [PubMed]
  74. Janjigian YY, Bendell J, Calvo E, et al. CheckMate-032 Study: Efficacy and Safety of Nivolumab and Nivolumab Plus Ipilimumab in Patients With Metastatic Esophagogastric Cancer. J Clin Oncol 2018;36:2836-44. [Crossref] [PubMed]
  75. Kageyama S, Ikeda H, Miyahara Y, et al. Adoptive Transfer of MAGE-A4 T-cell Receptor Gene-Transduced Lymphocytes in Patients with Recurrent Esophageal Cancer. Clin Cancer Res 2015;21:2268-77. [Crossref] [PubMed]
  76. Moehler M, Goepfert K, Heinrich B, et al. Oncolytic virotherapy as emerging immunotherapeutic modality: potential of parvovirus h-1. Front Oncol 2014;4:92. [Crossref] [PubMed]
  77. Ungerechts G, Engeland CE, Buchholz CJ, et al. Virotherapy Research in Germany: From Engineering to Translation. Hum Gene Ther 2017;28:800-19. [Crossref] [PubMed]
  78. Tanabe S, Tazawa H, Kagawa S, et al. Phase I/II trial of endoscopic intratumoral administration of OBP-301, a novel telomerase-specific oncolytic virus, with radiation in elderly esophageal cancer patients. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75:Abstract nr CT123.
Cite this article as: Ji A, Jin R, Zhang R, Li H. Primary small cell carcinoma of the esophagus: progression in the last decade. Ann Transl Med 2020;8(7):502. doi: 10.21037/atm.2020.03.214

Download Citation