Pattern of tumor regression after neoadjuvant chemoimmunotherapy for esophageal squamous cell carcinoma
Original Article

Pattern of tumor regression after neoadjuvant chemoimmunotherapy for esophageal squamous cell carcinoma

Bin Li1,2,3#, Yichen Wang2,3,4#, Hui Yu2,3,5#, Haiqing Chen1,2,3#, Yihua Sun1,2,3, Hong Hu1,2,3, Yawei Zhang1,2,3, Jiaqing Xiang1,2,3, Yuan Li2,3,4, Haiquan Chen1,2,3

1Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China; 2Institute of Thoracic Oncology, Fudan University, Shanghai, China; 3Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; 4Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; 5Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China

Contributions: (I) Conception and design: Haiquan Chen, Y Li, B Li; (II) Administrative support: Haiquan Chen; (III) Provision of study materials or patients: Y Sun, H Hu, Y Zhang, Y Li, J Xiang, H Yu; (IV) Collection and assembly of data: Y Wang, H Yu; (V) Data analysis and interpretation: Y Wang, Haiqing Chen, B Li, Y Li, Haiquan Chen; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work as co-first authors.

Correspondence to: Haiquan Chen, MD. Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China; Institute of Thoracic Oncology, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong’an Rd., Shanghai 200032, China. Email: hqchen1@yahoo.com.

Background: Immune checkpoint inhibitors have been increasingly applied for esophageal cancer. The aims of this study were to evaluate the pattern of tumor regression after neoadjuvant chemoimmunotherapy.

Methods: From January 2020 to December 2021, 138 patients with esophageal squamous cell carcinoma who had esophagectomy after neoadjuvant chemoimmunotherapy were reviewed. Surgical and pathological results were analyzed, and tumor regression pattern was evaluated.

Results: Of the 138 patients, 65 (47.1%) patients had chemotherapy combined with camrelizumab, 48 (34.8%) with pembrolizumab, 13 (9.4%) with tislelizumab, and 12 (8.7%) with sintilimab. Sixty-four patients (46.4%) underwent McKewon procedure, and 74 (53.6%) Ivor-Lewis procedure, respectively. There were 131/138 patients (94.9%) who had R0 resections, and the median number of resected lymph nodes was 28. Pneumonia was the most common complication after surgery (14.5%). Pathological complete regression occurred in 28 patients (20.3%). Regarding to residual tumor, there were 50 patients (36.2%) with residual tumor in the mucosa, 81 (58.7%) in the submucosa, 85 (61.6%) in the muscularis propria, 47 (34.1%) in the adventitia and 71 (51.4%) in the lymph nodes. There were 88 patients with no residual tumor in the mucosa, of whom 60 (68.2%) had residual tumors in other layers or in the lymph nodes.

Conclusions: In this retrospective study, esophagectomy after neoadjuvant chemoimmunotherapy is safe with acceptable surgical risk. Preferential clearing of tumor cells in mucosa layer is common after immunotherapy, while the rate of complete pathological response is relatively low, indicating surgery is still necessary.

Keywords: Esophagectomy; chemoimmunotherapy; safety; tumor regression


Submitted May 31, 2023. Accepted for publication Sep 14, 2023. Published online Oct 07, 2023.

doi: 10.21037/jtd-23-882


Highlight box

Key findings

• Preferential clearing of tumor cells in the mucosa layer is common after chemoimmunotherapy. However, the rate of complete pathological response is relatively low in a real-world setting.

What is known and what is new?

• The use of immune checkpoint inhibitors as neoadjuvant regimen showed promising tumor response rate.

• Absent residual tumors in the mucosa layers were observed in approximately 28–29% of patients with residual tumors after chemoradiotherapy. Whereas in this study, higher rates of absent residual tumors in the mucosa were observed, accounting for 55% (60/110) of patients with residual tumors after chemoimmunotherapy.

What is the implication, and what should change now?

• Negative endoscopic biopsies should be carefully evaluated in patients after chemoimmunotherapy, and esophagectomy is still warranted.


Introduction

Esophageal cancer is one of the most common malignancies in the world. For locally advanced esophageal cancer, multidisciplinary treatment, involving surgery, chemotherapy, and radiotherapy, is required. However, long-term survival remains poor, with 5-year overall survival ranging from 47% to 55% (1,2).

Recent studies have shown the superiority of immune checkpoint inhibitors in improving survival among patients with esophageal cancer (3-5), justifying their use as neoadjuvant regimen. Published studies using immune checkpoint inhibitors as neoadjuvant regimen showed promising tumor response with objective response rate ranging from 22.2% to 55.6% (6-8). However, these studies were limited by small study numbers, and the type of tumor regression was unclear. The aim of the study was to assess the pattern of tumor regression after neoadjuvant chemoimmunotherapy. The safety of esophagectomy after neoadjuvant chemoimmunotherapy was also evaluated in this study. We present this article in accordance with the STROBE reporting checklist (available at https://jtd.amegroups.com/article/view/10.21037/jtd-23-882/rc).


Methods

Patients

Between January 1st, 2020 and December 30th, 2021, 1,829 patients underwent esophagectomy due to cancer at Fudan University Shanghai Cancer Center, of which 251 patients received preoperative therapy before esophagectomy due to advanced tumor stage. The institutional review board of Fudan University Shanghai Cancer Center approved the use of database for this study (No. 1612167-18). Informed consent was taken from all the patients. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013).

The records of all patients were reviewed, and the inclusion criteria for this analysis were as follows: (I) esophageal squamous cell carcinoma by pathological confirmation; (II) neoadjuvant chemotherapy combined with immunotherapy; and (III) no radiotherapy prior to surgery. Ultimately, 138 patients were included in this retrospective analysis.

As there is still no consensus on the optimal neoadjuvant regimen, different types of immune checkpoint inhibitors were used based on surgeons’ experience and preference in China. In Shanghai Cancer Center, chemoimmunotherapy was always applied to those with tumors of clinical stage T1–4aN2 or T3–4aN1 and no clinical evidence of metastatic spread (M0). Immunotherapy combined with chemoradiotherapy was not widely performed. Of the 138 patients in this study, there were 86 patients (62.3%) had immunotherapy in Shanghai Cancer Center, and 52 (37.7%) in their local hospitals.

Treatment

Preoperative workup assessing patient operability included a complete history, physical examination, endoscopy of the entire upper gastrointestinal tract, histologic confirmation of the carcinoma, ultrasonography of the neck, computed tomography (CT) of the chest and abdomen. Pulmonary function testing and electrocardiography were also performed to assess fitness. Positron emission tomography and endoscopic ultrasonography were not performed routinely.

Esophagectomy via the right thoracic approach with radical two-field lymphadenectomy was widely used after the trial compared to esophagectomy via the left thoracic approach with limited two-field lymphadenectomy at the Shanghai Cancer Center (9). The McKeown procedure with cervical anastomosis and Ivor-Lewis procedure with thoracic anastomosis, via open or minimally invasive approach, were performed depending on the surgeon’s preference.

Extended radial two-field lymphadenectomy is routinely performed. When lymph node metastasis in the neck was indicated on CT or ultrasonography, cervical lymph nodes dissection was performed. Patients were staged according to the tumor-node-metastasis classification of the eighth edition of the American Joint Committee for Cancer Staging Manual (10).

At the end of surgery, patients were extubated if physiologically stable and then admitted to a general surgical ward. On postoperative day 1, the patients were encouraged to move out of bed, and enteral nutrition was initiated via the feeding tube. Oral intake began on postoperative day 6 without evidence of anastomotic leakage, and the patients were discharged on postoperative day 7 or 8.

Morbidity and mortality were examined within 90 days of surgery. Postoperative complications were recorded, including anastomotic leakage, pulmonary infection, cardiovascular complications, chylothorax, and other complications (intestinal obstruction, wound infection, and vocal cord paralysis). The severity of complications was assessed based on the Clavien-Dindo classification (11).

Histopathological assessment

Esophageal resection specimens were histopathologically evaluated by experienced pathologists specializing in esophageal cancer. During the original pathological examination, the tumor type, differentiation grade, depth of invasion, number of involved lymph nodes, and presence of lymphovascular invasion were recorded. Sample margins, including the circumferential resection margin, were also recorded. Radical resection (R0) was defined as the absence of tumor cells at all resection margins; R1 resection involved microscopic residual tumors, and R2 resection involved macroscopic residual tumors.

For primary tumors, tumor regression grade (TRG) was evaluated using the modified Mandard scoring system. TRG 1 was defined as no residual tumor cells [pathological complete response (pCR)], TRG 2 as 1–10% residual tumor cells, TRG 3, as 11–50% residual tumor cells, and TRG 4 as more than 50% residual tumor cells (12).

All specimens were reviewed by a pathologist (Wang Y) who was blinded to the previous pathological records. Residual tumors in each layer of the esophageal wall were re-reviewed.

Statistical analysis

Data were presented as numbers and percentages, means and standard errors, or medians and interquartile ranges. Fisher’s exact test was used to compare the frequencies of pathological complete regression among different immune checkpoint inhibitors. All statistical analyses were performed using SPSS statistical software (SPSS 22.0, Chicago, IL, USA).


Results

Of the 138 patients, the mean age was 62±7 years. There were 126 male patients (91.3%) and 66 patients (47.8%) had the tumors located in the middle thoracic esophagus (Table 1).

Table 1

Patient characteristics of 138 patients

Characteristics Values
Age (years) 62±7
Gender
   Male 126 (91.3)
   Female 12 (8.7)
BMI (kg/m2) 23±3
Hypertension 41 (29.7)
Diabetes 11 (8.0)
Smokers 94 (68.1)
Drinkers 78 (56.5)
Tumor locations
   Upper 49 (35.5)
   Middle 66 (47.8)
   Lower 23 (16.7)
ASA fitness grade
   1 9 (6.5)
   2 122 (88.4)
   3 7 (5.1)
Immune checkpoint inhibitors
   Camrelizumab 65 (47.1)
   Pembrolizumab 48 (34.8)
   Tislelizumab 13 (9.4)
   Sintilimab 12 (8.7)
Clinical T-stage§
   1 34 (24.6)
   2 59 (42.8)
   3 44 (31.9)
   4 1 (0.7)
Clinical N-stage§
   0 48 (34.8)
   1 59 (42.8)
   2 23 (16.7)
   3 8 (5.8)

Data are shown as mean ± standard error or n (%). , at least ten cigarettes per week for more than 6 months, including former and current smoker; , any alcoholic beverage containing at least 20 g ethanol per week for more than 6 months, including former and current drinkers; §, clinical stage after chemoimmunotherapy. BMI, body mass index; ASA, American Society of Anaesthesiologist.

All patients received platinum-based chemotherapy, of which 120 (87.0%) combined with paclitaxel and 18 (13.0%) combined with docetaxel. There were 48 patients (34.8%) who received chemotherapy combined with pembrolizumab, 65 (47.1%) combined with camrelizumab, 13 (9.4%) combined with tislelizumab, and 12 (8.7%) combined with sintilimab. There were 10 patients (7.2%) having only one cycle, 102 (73.9%) having two cycles, and 26 (18.8%) having three cycles or more. There were 86 patients who had immunotherapy in Shanghai Cancer Center, of which 31 (36.0%) had treatment-related adverse events, and 14 leukocytopenia (16.3%) and 5 increased transaminase (5.8%), being the most common treatment-related adverse events.

Majority of patients had clinical T2 (59, 42.8%) or N1 (59, 42.8%) disease before surgery. Surgical outcomes are shown in Table 2. There were 64 patients (46.4%) who underwent McKewon procedure and 74 (53.6%) underwent Ivor-Lewis procedure. Majority of the surgeries were completed using open thoracic approach (130, 94.2%). A total of 131 patients (94.9%) underwent R0 resections, 4 (2.9%) R1 resections, and 3 (2.2%) R2 resections.

Table 2

Surgical outcomes of 138 patients

Surgical outcomes Values
Surgery procedure
   McKewon 64 (46.4)
   Ivor-Lewis 74 (53.6)
Surgical approach
   Minimally invasive 8 (5.8)
   Open 130 (94.2)
Extent of lymphadenectomy
   Three-field 11 (8.0)
   Two-field 127 (92.0)
Operative time (min) 200 [180–234]
Postoperative complications
   Anastomotic leak 8 (5.8)
   Pneumonia 20 (14.5)
   Arrhythmia 8 (5.8)
   Vocal cord paralysis 15 (10.9)
   Chylothorax 5 (3.6)
   Reintubation 5 (3.6)
   Uroschesis 1 (0.7)
   Wound infection 2 (1.4)
   Intestinal obstruction 1 (0.7)
   Re-operation 1 (0.7)
   90-day mortality 2 (1.4)
Hospital stay (days) 13 [10–18]
Clavien-Dindo classification
   I 9 (6.5)
   II 18 (13.0)
   III 4 (2.9)
   IV 6 (4.3)
   V 2 (1.4)

Data are shown as median [interquartile range] or n (%).

Of the 138 patients, 99 (71.7%) had no postoperative complications. Majority of postoperative complications (46.2%, 18/39) were classified as grade II complications. Two patients (1.4%) died during hospitalization, one due to severe pneumonia following an anastomotic leak on postoperative day 23 and one due to tracheoesophageal fistula on postoperative day 41.

Pathological outcomes are shown in Table 3. Pathological complete regression was observed in 28 patients (20.3%), whereas 110 patients (79.7%) had residual tumors. Pathological complete regression was observed in 9 patients (18.8%) using pembrolizumab, 14 (21.5%) using camrelizumab, 2 (15.4%) using tislelizumab, and 3 (25.0%) using sintilimab, P=0.919.

Table 3

Pathological outcomes of 138 patients

Pathological outcomes Values
Tumor length (cm) 2.0 [1.5–3.0]
T-stage
   T0 36 (26.1)
   T1 25 (18.1)
   T2 21 (15.2)
   T3 51 (37.0)
   T4 5 (3.6)
N-stage
   0 67 (48.6)
   1 38 (27.5)
   2 19 (13.8)
   3 14 (10.1)
Number of lymph nodes resected 28 [21–38]
Lymphovascular invasion
   Yes 49 (35.5)
   No 89 (64.5)
Tumor differentiation
   Well 3 (2.2)
   Moderate 56 (40.6)
   Poor 40 (29.0)
   Unknown 39 (28.3)
TNM classification
   YI 50 (36.2)
   YII 15 (10.9)
   YIIIa 21 (15.2)
   YIIIb 34 (24.6)
   YIVa 17 (12.3)
   YIVb 1 (0.7)
TRG
   1 36 (26.1)
   2 18 (13.0)
   3 31 (22.5)
   4 53 (38.4)

Data are shown as median [interquartile range] or n (%). TNM, tumor, node, and metastasis; TRG, tumor regression grade.

Regarding primary tumors, there were 36 patients (26.1%) who had TRG1 tumors, 18 (13.0%) with TRG2 tumors, 31 (22.5%) with TRG3 tumors, and 53 (38.4%) with TRG4 tumors. There were 50 patients (36.2%) with residual tumor in the mucosa, 81 (58.7%) in the submucosa, 85 (61.6%) in the muscularis propria, 47 (34.1%) in the adventitia, and 71 (51.4%) in the lymph nodes (Figure 1). There were 88 patients with no residual tumor in the mucosa, of whom 60 (68.2%) had residual tumors in other layers or in the lymph nodes. There were 54 patients with no residual tumor in the mucosa or submucosa, of which 26 (48.1%) had residual tumor in other layers or in the lymph nodes (Figure 2).

Figure 1 Frequencies of residual tumor in each individual layer of the esophageal wall and all resected lymph nodes in 138 patients.
Figure 2 Distribution of residual tumors in 138 patients.

Regarding the possible regression pattern of primary tumors by Shapiro et al. [2013] (13). Of the 102 patients who had residual tumors in the esophageal wall (mucosa, submucosa, muscularis propria, and adventitia, as showed in Figure 2), 30 (29.4%) showed regression toward the lumen (more regression in the muscularis propria and the adventitia), 8 (7.8%) had regression toward the invasive front (more regression in the mucosa and the submucosa), 27 (26.5%) had concentric regression (more regression in the mucosa and the adventitia), and 37 (36.3%) had random regression (comparable extent of regression in all layers).


Discussion

In patients with esophageal squamous cell carcinoma, our results showed the safety of esophagectomy after neoadjuvant chemoimmunotherapy in a real-world scenario. Notably, preferential clearing of tumor cells in the mucosal layer is common after neoadjuvant chemoimmunotherapy.

Current guidelines recommend esophagectomy after neoadjuvant chemoradiotherapy for advanced esophageal squamous cell carcinoma. However, in the 10-year survival report of the ChemoRadiotherapy for Oesophageal Cancer Followed by Surgery Study (CROSS), chemoradiotherapy reduced locoregional recurrence compared to surgery alone, whereas distant metastases were similar between the two groups (14). In a trial comparing esophagectomies via the left versus right thoracic approaches, extended radical lymphadenectomy via the right thoracic approach reduced locoregional recurrence compared with inadequate lymphadenectomy via the left thoracic approach, whereas distant metastases were still the reason for treatment failure (9). Therefore, it is important to reduce distant metastases to improve the survival of patients with resectable esophageal cancer.

Recent studies have justified the perioperative use of immune checkpoint inhibitors in resectable esophageal cancer (6-8). However, in Keynote 590 study on advanced esophageal cancer (3), treatment-related adverse events of grade 3 or higher occurred in 72% of patients in the pembrolizumab plus chemotherapy group. Thus, one important concern is that immunotherapy would potentially increase the surgical risk. In the current study, most treatment-related adverse events were minor and manageable before surgery. No immunotherapy-related deaths were observed in the current cohort after esophagectomy. Moreover, both the incidence of postoperative complications and length of hospital stay were consistent with our previous report in patients who underwent upfront esophagectomy (15). This is probably due to fewer treatment cycles before surgery, and esophagectomy was always performed after 2–3 cycles of therapy. Therefore, we presume that the preoperative use of immunotherapy is safe without increased surgical complications.

Pathological complete response, which is a prognostic factor for patient survival in the era of neoadjuvant chemoradiotherapy (16), was the primary outcome in most published phase II trials referring to neoadjuvant immunotherapy, ranging from 21.7% to 39.2% (7,17,18). In this retrospective setting, pathological complete response was only 20.2%. To date, long-term survival data for immune checkpoint inhibitors used as neoadjuvant regimens are still rare. However, in a trial comparing neoadjuvant chemoradiotherapy versus neoadjuvant chemotherapy followed by minimally invasive esophagectomy, there was a significant difference in the pathological complete responses (35.7% vs. 3.8%), but with no significant difference in the 1-year overall survival (87.1% vs. 82.6%) (19). Therefore, it is still unclear whether a higher pathological complete response is related to better patient survival, and future prospective randomized trials are needed to clarify the value of immunotherapy by comparing with other neoadjuvant regimens.

In the CROSS study, 150 of 184 (84%) patients in the chemoradiotherapy arm had esophageal cancer of clinical T3 staging before treatment, pathological complete response after chemoradiotherapy was 49% for esophageal squamous cell carcinoma, indicating wait-and-see strategy, and avoiding esophageal resection could be feasible in selected patients (1). Accurate detection of residual tumors is important to determine the appropriate management. Previous studies have reported absent residual tumors in the mucosa layers were observed in approximately 28–29% of patients with residual tumors after chemoradiotherapy (13,20). Whereas in this study, higher rates of absent residual tumors in the mucosa were observed, accounting for 55% (60/110) of patients with residual tumors after chemoimmunotherapy. Different results indicated different regression patterns after chemoimmunotherapy. Endoscopic bite-on-bite biopsies are recommended to improve the diagnostic accuracy. However, it might be challenging due to a preferential clearing of the tumor cells in the mucosa or the submucosa rather than in the muscle and adventitia layers, which were observed in approximately 48% of the patients in our study. Therefore, negative endoscopic biopsies should be carefully evaluated in patients after immunotherapy.

This study has some limitations. Firstly, the study was limited by its retrospective nature: only patients who underwent esophagectomy were included. The number of patients who did not undergo surgery due to tumor progression or adverse event was not available, and the incidence of treatment-related adverse events was underestimated. Secondly, the expression of programmed death-ligand 1 was not evaluated before treatment. Thirdly, different types of immune checkpoint inhibitors were used in this study. Although frequencies of pathological complete regression were comparable among different inhibitors, it is unclear whether the regression patterns were similar due to small study numbers of the four inhibitors. However, this study revealed the value of esophagectomy after neoadjuvant chemoimmunotherapy, particularly a high incidence of complete pathological response in the mucosa showing different regression patterns compared with that after neoadjuvant chemoradiotherapy.


Conclusions

In this retrospective study, esophagectomy after neoadjuvant chemoimmunotherapy can be safely performed. Preferential clearing of tumor cells in the mucosal layer is common after immunotherapy; however, the rate of complete pathological response is relatively low. Esophagectomy is still warranted after neoadjuvant chemoimmunotherapy.


Acknowledgments

Funding: This work was supported by the National Natural Science Foundation of China (No. 81930073), the Shanghai Science and Technology Innovation Action Project (No. 20JC1417200), and the Cooperation Project of Conquering Major Diseases in Xuhui District (No. XHLHGG202101).


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://jtd.amegroups.com/article/view/10.21037/jtd-23-882/rc

Data Sharing Statement: Available at https://jtd.amegroups.com/article/view/10.21037/jtd-23-882/dss

Peer Review File: Available at https://jtd.amegroups.com/article/view/10.21037/jtd-23-882/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://jtd.amegroups.com/article/view/10.21037/jtd-23-882/coif). Haiquan Chen serves as an unpaid editorial board member of Journal of Thoracic Disease from October 2022 to September 2024. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The institutional review board of Fudan University Shanghai Cancer Center approved the esophageal carcinoma database used in the present study (No. 1612167-18). Informed consent was taken from all the patients. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013).

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. 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]
  2. Ando N, Kato H, Igaki H, et al. A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol 2012;19:68-74. [Crossref] [PubMed]
  3. Sun JM, Shen L, Shah MA, et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet 2021;398:759-71. [Crossref] [PubMed]
  4. Luo H, Lu J, Bai Y, et al. Effect of Camrelizumab vs Placebo Added to Chemotherapy on Survival and Progression-Free Survival in Patients With Advanced or Metastatic Esophageal Squamous Cell Carcinoma: The ESCORT-1st Randomized Clinical Trial. JAMA 2021;326:916-25. [Crossref] [PubMed]
  5. Kelly RJ, Ajani JA, Kuzdzal J, et al. Adjuvant Nivolumab in Resected Esophageal or Gastroesophageal Junction Cancer. N Engl J Med 2021;384:1191-203. [Crossref] [PubMed]
  6. Fan M, Dai L, Yan W, et al. Efficacy of programmed cell death protein 1 inhibitor in resection transformation treatment of esophageal cancer. Thorac Cancer 2021;12:2182-8. [Crossref] [PubMed]
  7. Duan H, Wang T, Luo Z, et al. A multicenter single-arm trial of sintilimab in combination with chemotherapy for neoadjuvant treatment of resectable esophageal cancer (SIN-ICE study). Ann Transl Med 2021;9:1700. [Crossref] [PubMed]
  8. Li C, Zhao S, Zheng Y, et al. Preoperative pembrolizumab combined with chemoradiotherapy for oesophageal squamous cell carcinoma (PALACE-1). Eur J Cancer 2021;144:232-41. [Crossref] [PubMed]
  9. Li B, Hu H, Zhang Y, et al. Extended Right Thoracic Approach Compared With Limited Left Thoracic Approach for Patients With Middle and Lower Esophageal Squamous Cell Carcinoma: Three-year Survival of a Prospective, Randomized, Open-label Trial. Ann Surg 2018;267:826-32. [Crossref] [PubMed]
  10. Rice TW, Ishwaran H, Ferguson MK, et al. Cancer of the Esophagus and Esophagogastric Junction: An Eighth Edition Staging Primer. J Thorac Oncol 2017;12:36-42.
  11. Dindo D, Demartines N, Clavien PA. Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg 2004;240:205-13. [Crossref] [PubMed]
  12. Chirieac LR, Swisher SG, Ajani JA, et al. Posttherapy pathologic stage predicts survival in patients with esophageal carcinoma receiving preoperative chemoradiation. Cancer 2005;103:1347-55. [Crossref] [PubMed]
  13. Shapiro J, ten Kate FJ, van Hagen P, et al. Residual esophageal cancer after neoadjuvant chemoradiotherapy frequently involves the mucosa and submucosa. Ann Surg 2013;258:678-88; discussion 688-9. [Crossref] [PubMed]
  14. Eyck BM, van Lanschot JJB, Hulshof MCCM, et al. Ten-Year Outcome of Neoadjuvant Chemoradiotherapy Plus Surgery for Esophageal Cancer: The Randomized Controlled CROSS Trial. J Clin Oncol 2021;39:1995-2004. [Crossref] [PubMed]
  15. Li B, Hu H, Zhang Y, et al. Three-field versus two-field lymphadenectomy in transthoracic oesophagectomy for oesophageal squamous cell carcinoma: short-term outcomes of a randomized clinical trial. Br J Surg 2020;107:647-54. [Crossref] [PubMed]
  16. Shen J, Kong M, Yang H, et al. Pathological complete response after neoadjuvant treatment determines survival in esophageal squamous cell carcinoma patients (NEOCRTEC5010). Ann Transl Med 2021;9:1516. [Crossref] [PubMed]
  17. Zhang Z, Hong ZN, Xie S, et al. Neoadjuvant sintilimab plus chemotherapy for locally advanced esophageal squamous cell carcinoma: a single-arm, single-center, phase 2 trial (ESONICT-1). Ann Transl Med 2021;9:1623. [Crossref] [PubMed]
  18. Liu J, Yang Y, Liu Z, et al. Multicenter, single-arm, phase II trial of camrelizumab and chemotherapy as neoadjuvant treatment for locally advanced esophageal squamous cell carcinoma. J Immunother Cancer 2022;10:e004291. [Crossref] [PubMed]
  19. Wang H, Tang H, Fang Y, et al. Morbidity and Mortality of Patients Who Underwent Minimally Invasive Esophagectomy After Neoadjuvant Chemoradiotherapy vs Neoadjuvant Chemotherapy for Locally Advanced Esophageal Squamous Cell Carcinoma: A Randomized Clinical Trial. JAMA Surg 2021;156:444-51. [Crossref] [PubMed]
  20. Tang H, Jiang D, Zhang S, et al. Residual tumor characteristics of esophageal squamous cell carcinoma after neoadjuvant chemoradiotherapy. J Thorac Cardiovasc Surg 2021;162:1632-41. [Crossref] [PubMed]
Cite this article as: Li B, Wang Y, Yu H, Chen H, Sun Y, Hu H, Zhang Y, Xiang J, Li Y, Chen H. Pattern of tumor regression after neoadjuvant chemoimmunotherapy for esophageal squamous cell carcinoma. J Thorac Dis 2023;15(10):5517-5524. doi: 10.21037/jtd-23-882

Download Citation