Abstract
The therapeutic landscape in nearly every therapeutic line in advanced/metastatic patients with squamous cell carcinoma (SCC) and esophagogastric adenocarcinoma (EGC) is enriched by recent approvals of immune checkpoint inhibitors (ICIs). In curative intended therapy, patients without pathological residual disease of SCC or GEJ (esophagogastric junction) cancer after preoperative chemoradiation and complete resection have access to adjuvant immunotherapy (independent of PD-L1 (programmed cell death protein 1) status, nivolumab, CHECKMATE 577). For metastatic SCC in the first-line, nivolumab combined with chemotherapy or with ipilimumab (TPS (tumor proportion score) ≥1 %, SCC, CHECKMATE 648) are approved, as well as second-line nivolumab alone regardless of PD-L1 status (ATTRACTION 03). For both, locally advanced or metastatic SCC and EGC, chemotherapy with pembrolizumab is available for patients with CPS (combined positive score) ≥10 (KEYNOTE 590) and for adenocarcinoma with nivolumab (CPS ≥5, CHECKMATE 649). Recent added approvals are chemotherapy with pembrolizumab in CPS ≥1 patients (KEYNOTE 859) and the addition of trastuzumab for personalized therapy in HER-2 positive/CPS ≥1 gastric and GEJ patients (KEYNOTE 811).
Introduction
Esophageal cancers are often advanced and metastatic at the time of diagnosis. Innovative multimodal therapy regimens have significantly improved the prognosis of esophageal carcinomas within the last years. Immune checkpoint inhibitors play an increasing role in every line of therapy and constitute a well tolerable and promising therapy option. Immune checkpoint inhibitors can be classified in gastrointestinal tumors depending on the target protein on the tumor or immune cells into three different groups: the anti-PD-1 antibodies nivolumab, pembrolizumab, tislelizumab, camrelizumab, sintilimab, toripalimab (T cells); the anti-PD-L1 antibodies atezolizumab, avelumab, and durvalumab (cancer cells, dendritic cells); and the anti-CTLA-4 antibody ipilimumab (T cells). Especially the combination of chemo- and immunotherapy with a possible synergistic effect of T-cell recruitment and activation is the object of current research.
Curative immunotherapy
Before immunotherapy entered the established standard therapy regimens, curative intended therapy for SCC consisted of definitive chemoradiation for esophageal cancer in the upper third and neoadjuvant chemoradiation followed by resection for esophageal cancer in the lower two thirds [1]. In EGC, perioperative chemotherapy was recommended, whereas therapy of locally advanced adenocarcinomas is currently independent of the HER2 status.
Based on the results of the Checkmate 577 study, for patients with SCC and GEJ-cancer who received neoadjuvant (radio-)chemotherapy and R0 resection with remaining residual pathologic disease (≥ypT1 or ≥ypN1), an adjuvant therapy with nivolumab is recommended and should be performed for 1 year. Placebo-controlled adjuvant application of nivolumab revealed both, a significant prolongation of the disease-free survival (DFS) (22.4 vs. 11.0 months, Hazard Ratio (HR) 0.69 [96.4 % CI 0.56–9.86]; p<0.001) and the metastatic-free survival [2].
Palliative first-line immunotherapy
In the past, systemic chemotherapy was recommended in the palliative situation. In SCC, chemotherapy with platin and 5-fluorouracil (FU) showed a median overall survival (OS) of about 10 months [3].
With the addition of immunotherapy, the prognosis could be improved significantly: in patients with advanced SCC, the combination of nivolumab and chemotherapy (cisplatin + FU) results in significantly longer OS than chemotherapy alone (13.2 vs. 10.7 months, HR 0.74 [99.1 % CI 0.58–0.96]; p=0.002). In patients with TPS (tumor proportion score) ≥1 % the benefit was even more pronounced with a median OS of 15.4 vs. 9.1 months, HR 0.54 [99.5 % CI 0.37–0.80], p<0.0001. Further, the dual immune checkpoint blockade with nivolumab and ipilimumab compared with chemotherapy only could achieve significantly longer overall survival among patients with TPS ≥1 % (13.7 vs. 9.1 months, HR 0.64 [98.6 % CI 0.46–0.90]; p=0.001) and in the overall population (12.7 vs. 10.7 months, HR 0.78 [98.2 % CI 0.62–0.98]; p=0.01) (CHECKMATE 648) [4]. Based on these data, first-line approval was granted in the EU recently in May 2022 for both nivolumab with platinum/fluoropyrimidine-based chemotherapy and the chemotherapy-free combination of nivolumab plus ipilimumab for patients with advanced, metastatic SCC with TPS ≥1 %.
In patients with locally advanced or metastatic SCC and EGC, the combination of pembrolizumab and chemotherapy (cisplatin + 5-FU) indicates significant benefit in OS with 12.4 vs. 9.8 months, HR 0.73 [95 % CI 0.62–0.86]; p<0.0001) independently from PD-L1 CPS and histology. Indeed, the subgroup of SCC and adenocarcinomas with CPS (combined positivity score) ≥10 benefited most from combination therapy (KEYNOTE 590) [5]. In September 2020, EU approval was granted for pembrolizumab in combination with platinum and 5-FU for SCC and EGC with CPS ≥10.
In patients with gastric, gastroesophageal junction, and esophageal adenocarcinoma and CPS ≥5, the combination therapy of nivolumab plus chemotherapy (FOLFOX/XELOX) resulted in significant improvements in OS (14.4 vs. 11.1 months, HR 0.71 [98.4 % CI 0.59–0.86]; p<0.0001) and progression-free survival (PFS) (7.7 vs. 6.0 months, HR 0.68 [98 % CI 0.56–0.81]; p<0.0001) vs. chemotherapy alone (CHECKMATE 649) [6]. Subsequently, the combination of nivolumab plus fluoropyrimidine/platin-based chemotherapy was approved for EGC patients with CPS ≥5 in Europe. Latest 36-month follow-up results presented at ASCO 2023 promote the OS benefit in the CPS ≥5 population: median OS 14.4 vs. 11.1 months, HR 0.70 [0.61–0.81] and median PFS 8.3 vs. 6.1 months HR 0.70, [0.60–0.81] [7]. Recent added approvals are chemotherapy with pembrolizumab in CPS ≥1 patients (KEYNOTE 859) and the addition of trastuzumab for personalized therapy in HER-2 positive/CPS ≥1 gastric and GEJ patients (KEYNOTE 811).
Palliative second- and third-line immunotherapy
The multicenter phase III trial Attraction 03 evaluated the use of nivolumab in second-line therapy in SCC after progression on first-line therapy and demonstrates a significant survival benefit in overall survival for nivolumab monotherapy in comparison to chemotherapy (paclitaxel/docetaxel; median OS 10.9 vs. 8.4 months, HR 0.77 [95 % CI 0.62–0.96]; p=0.019) [8]. Based on these data, nivolumab was approved as a second-line treatment option for patients with advanced SCC regardless of PD-L1 expression status in Europe.
Except for the approval in MSI EGC patients, there is currently no approval for immunotherapy in patients with advanced EGC in second-line therapy. In the KEYNOTE 061 trial with pembrolizumab monotherapy vs. paclitaxel chemotherapy in patients with EGC and progression on first-line chemotherapy, the primary endpoint (superiority of OS) was not met. However, the data show a dependence of pembrolizumab efficacy on PD-L1-CPS score: patients with PD-L1-CPS ≥10 % expression demonstrate benefit of pembrolizumab therapy [9].
Potential therapy algorithms based on the current approvals for immune checkpoint inhibitors (ICI) therapy in Europe are presented in Figure 1 (SCC) and Figure 2 (EGC).

Potential therapy algorithm of SCC in Europe. PCR, Pathologic complete remission; CPS, Combined positive score; TPS, Tumor proportion score; Nivo, Nivolumab; Ipi, Ipilimumab; Pembro, Pembrolizumab; Nivo, Nivolumab; Tisleli, Tislelizumab.
![Figure 2:
Potential therapy algorithm of adenocarcinoma of the esophagus in Europe [25]. Additional approval has been given for Pembro in CPS ≥1 patients (KEYNOTE 859) in HER-2 positive/CPS ≥1 gastric and GEJ patients (KEYNOTE 811). Pembro, Pembrolizumab; Nivo, Nivolumab; CPS, Combined positive score; MSI, Microsatellite instability; EBV, Epstein–Barr virus; GEJ-cancer, Esophagogastric junction cancer.](/document/doi/10.1515/iss-2023-0023/asset/graphic/j_iss-2023-0023_fig_002.jpg)
Potential therapy algorithm of adenocarcinoma of the esophagus in Europe [25]. Additional approval has been given for Pembro in CPS ≥1 patients (KEYNOTE 859) in HER-2 positive/CPS ≥1 gastric and GEJ patients (KEYNOTE 811). Pembro, Pembrolizumab; Nivo, Nivolumab; CPS, Combined positive score; MSI, Microsatellite instability; EBV, Epstein–Barr virus; GEJ-cancer, Esophagogastric junction cancer.
Potential therapy options in the future
In the perioperative setting, the CROSS trial [10] and the FLOT regimen [11] represented the best perioperative multimodality concepts, which have been [2] and will further be improved by combinations with PD-1/PD-L1 inhibitors, currently under investigation: pembrolizumab in the perioperative treatment setting of EGC is being analyzed in the KEYNOTE 585 trial, where patients receive perioperative pembrolizumab plus chemotherapy (cisplatin + capecitabine/5-FU) vs. placebo plus chemotherapy [12]. A potential survival benefit of PD-L1 inhibitor durvalumab in the perioperative setting in patients with PD-L1-independent resectable EGC is investigated in a combination of durvalumab with FLOT vs. FLOT alone (MATTERHORN trial) [13]. Within the randomized phase II DANTE trial of the AIO (Arbeitsgemeinschaft Internistische Onkologie), patients with resectable localized EGC received the combination of atezolizumab (anti-PD-L1) plus chemotherapy (FLOT) compared with perioperative chemotherapy alone. First results show a feasible combination therapy and efficacy data are expected soon [14].
Various promising checkpoint inhibitors are under investigation in the palliative treatment of SCC and EGC: The combination of tislelizumab (PD-1 inhibitor) with chemotherapy (5-FU/platin-based) in the first-line therapy showed significant improvement of OS of 6.7 months in SCC patients with locally advanced/metastatic disease (median OS 17.3 vs. 10.6 months, HR 0.66 [95 % CI 0.54–0.80]; p<0.0001) (RATIONALE 306 trial) [15]. In the subgroup of patients with PD-L1-CPS ≥10 % expression, the OS benefit was particularly pronounced in the arm with checkpoint inhibition and chemotherapy vs. chemotherapy alone. Latest results of the interim analysis of the RATIONALE 305 trial at ASCO GI in January 2023 using tislelizumab placebo-controlled in first-line therapy in combination with chemotherapy (platin/5-FU) in patients with locally advanced or metastatic EGC demonstrated significant improvement in OS in the PD-L1 positive population (median OS 17.2 vs. 12.6 months, HR 0.74 [95 % CI 0.59–0.94], 1-sided p=0.0056). Combination of tislelizumab plus chemotherapy showed also longer PFS (median PFS 7.2 vs. 5.9 months, HR 0.67 [95 % CI 0.55–0.83]) [16]. Tislelizumab was also investigated in second-line therapy: after progression of SCC under prior chemotherapy with platinum and 5-FU, there was a significantly prolonged overall survival of tislelizumab monotherapy vs. chemotherapy (paclitaxel/docetaxel/irinotecan) (median OS of 8.6 vs. 6.3 months, HR 0.7 [95 % CI 0.57–0.85]; p=0.0001) (RATIONALE-302 trial). Again, the patient group with PD-L1-CPS ≥10 % expression showed greater benefit with a median OS of 10.3 vs. 6.8 months (HR 0.54 [95 % CI 0.36–0.79]; p=0.0006) [17].
As presented at ESMO 2021, further promising PD-1 inhibitors have potential to prolong OS in advanced SCC patients as investigated in Asian trials (Camrelizumab: ESCORT 1-trial, Sintilimab: ORIENT 15-trial, Toripalimab: JUPITER 06-trial) [18], [19], [20]. With respect to the Asian population, these results highlight the global efforts in implementing checkpoint inhibitors into standard first-line therapy regimens.
Another promising target for individualized therapy in advanced Her2-negative, Claudin-18.2-positive EGC (moderate-to-strong membrane staining in ≥75 % tumor cells) is the anti-Claudin-18.2 targeted antibody zolbetuximab (IMAB362), which specifically binds to the tight-junction component Claudin-18.2, which is expressed in malignant transformed gastric mucosa of diffuse and intestinal GC. At ASCO 2023, conclusive primary OS data of the combination of zolbetuximab with FOLFOX compared with placebo with chemotherapy were demonstrated (SPOTLIGHT trial). Median OS was improved by 2.7 months (median OS: 18.23 vs. 15.54 months, HR 0.750, p=0.0053), as well as PFS (median PFS: 10.61 vs. 8.67 months, HR 0.751, p=0.0066) [21].
With respect to Her2 (human epidermal growth factor receptor 2)-targeted therapy, combination of trastuzumab and chemotherapy was the best first-line standard of care for advanced/metastatic HER2-positive cancers patients [22]. With recent impressive results of the phase 3 KEYNOTE 811, trastuzumab and chemotherapy combined with pembrolizumab showed superior overall response rates (ORR 74.4 % (66.2–81.6) vs. placebo (51.9 % (43.0–60.7), 95 % CI 11.2–33.7, p=0.00006). The complete response rate (CR) was also beneficial in the triple combination (11.3 vs. 3.1 %), as well as the disease control rate (DCR, 95 % CI 96.2 % (91.4–98.8) vs. 89.3 (82.7–94.0) [23]. Based on the most recent presented survival benefits, pembrolizumab with chemo and trastuzumab was approved in patients with HER-2 positive/CPS ≥1 gastric and GEJ patients by the FDA and EMA. Various further approaches of Her2-targeted therapy are currently investigated addressing Her2-positive patients who do not benefit from trastuzumab and show progressive disease under prior trastuzumab therapy, including the antibody-drug conjugate trastuzumab-deruxtecan (T-Dxd), now approved in second line or later for HER2 + adenocarinoma. Combination of T-Dxd with immune checkpoint inhibitors show first promising results (DESTINY Gastric 03-trial) [24].
Conclusions
Immunotherapy of SCC and EGC enriches the current therapy and is established as new standard treatment in almost every therapy line within a short period of time. Ongoing clinical trials further investigate different immune checkpoint inhibitors and address the remaining questions of individual responders/nonresponders to immunotherapy as well as the reuse of checkpoint inhibitors after progression to immunotherapy.
-
Research ethics: Not applicable.
-
Informed consent: Not applicable.
-
Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Competing interest: Yvonne Huber has no conflicts of interest. Markus Moehler: advisory roles for BMS, MSD, Merck, Amgen, BeiGene, Novartis, Lilly, Macrogenics, Sanofi, Servier and Roche. Anica Högner: advisory role for BMS.
-
Research funding: None declared.
-
Data availability: Not applicable.
References
1. Eyck, BM, van Lanschot, JJB, Hulshof, M, van der Wilk, BJ, Shapiro, J, van Hagen, P, 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. https://doi.org/10.1200/jco.20.03614.Search in Google Scholar
2. Kelly, RJ, Ajani, JA, Kuzdzal, J, Zander, T, Van Cutsem, E, Piessen, G, et al.. Adjuvant nivolumab in resected esophageal or gastroesophageal junction cancer. N Engl J Med 2021;384:1191–203. https://doi.org/10.1056/nejmoa2032125.Search in Google Scholar
3. Moehler, M, Maderer, A, Thuss-Patience, PC, Brenner, B, Meiler, J, Ettrich, TJ, et al.. Cisplatin and 5-fluorouracil with or without epidermal growth factor receptor inhibition panitumumab for patients with non-resectable, advanced or metastatic oesophageal squamous cell cancer: a prospective, open-label, randomised phase III AIO/EORTC trial (POWER). Ann Oncol 2020;31:228–35. https://doi.org/10.1016/j.annonc.2019.10.018.Search in Google Scholar PubMed
4. Doki, Y, Ajani, JA, Kato, K, Xu, J, Wyrwicz, L, Motoyama, S, et al.. Nivolumab combination therapy in advanced esophageal squamous-cell carcinoma. N Engl J Med 2022;386:449–62. https://doi.org/10.1056/nejmoa2111380.Search in Google Scholar
5. Sun, JM, Shen, L, Shah, MA, Enzinger, P, Adenis, A, Doi, T, 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. https://doi.org/10.1016/s0140-6736(21)01234-4.Search in Google Scholar PubMed
6. Janjigian, YY, Shitara, K, Moehler, M, Garrido, M, Salman, P, Shen, L, et al.. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet 2021;398:27–40. https://doi.org/10.1016/s0140-6736(21)00797-2.Search in Google Scholar PubMed PubMed Central
7. Janjigian, YY, Shitara, K, Moehler, MH, Garrido, M, Gallardo, C, Shen, L, et al.. Nivolumab (NIVO) plus chemotherapy (chemo) vs chemo as first-line (1L) treatment for advanced gastric cancer/gastroesophageal junction cancer/esophageal adenocarcinoma (GC/GEJC/EAC): 3-year follow-up from checkmate 649. J Clin Oncol 2023;41:291. https://doi.org/10.1200/jco.2023.41.4_suppl.291.Search in Google Scholar
8. Kato, K, Cho, BC, Takahashi, M, Okada, M, Lin, CY, Chin, K, et al.. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019;20:1506–17. https://doi.org/10.1016/s1470-2045(19)30626-6.Search in Google Scholar PubMed
9. Shitara, K, Özgüroğlu, M, Bang, YJ, Di Bartolomeo, M, Mandalà, M, Ryu, MH, et al.. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet 2018;392:123–33. https://doi.org/10.1016/s0140-6736(18)31257-1.Search in Google Scholar
10. van Hagen, P, Hulshof, MC, van Lanschot, JJ, Steyerberg, EW, van Berge Henegouwen, MI, Wijnhoven, BP, et al.. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 2012;366:2074–84. https://doi.org/10.1056/nejmoa1112088.Search in Google Scholar
11. Al-Batran, S-E, Homann, N, Pauligk, C, Goetze, TO, Meiler, J, Kasper, S, et al.. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. The Lancet 2019;393:1948–57. https://doi.org/10.1016/s0140-6736(18)32557-1.Search in Google Scholar PubMed
12. Bang, YJ, Van Cutsem, E, Fuchs, CS, Ohtsu, A, Tabernero, J, Ilson, DH, et al.. KEYNOTE-585: phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol 2019;15:943–52. https://doi.org/10.2217/fon-2018-0581.Search in Google Scholar PubMed
13. Janjigian, YY, Cutsem, EV, Muro, K, Wainberg, Z, Al-Batran, S-E, Hyung, WJ, et al.. Matterhorn: phase III study of durvalumab plus FLOT chemotherapy in resectable gastric/gastroesophageal junction cancer. Future Oncol 2022;18:2465–73. https://doi.org/10.2217/fon-2022-0093.Search in Google Scholar PubMed
14. Al-Batran, S-E, Lorenzen, S, Schenk, M, Thuss-Patience, PC, Goekkurt, E, Hofheinz, R, et al.. Safety of perioperative atezolizumab in combination with FLOT versus FLOT alone in patients with resectable esophagogastric adenocarcinoma: an interim safety analysis of the DANTE, a randomized, open-label phase II trial of the German gastric group at the AIO and the SAKK. J Clin Oncol 2020;38:398. https://doi.org/10.1200/jco.2020.38.4_suppl.398.Search in Google Scholar
15. Yoon, H, Kato, K, Raymond, E, Hubner, R, Shu, Y, Pan, Y, et al.. LBA-1 rationale-306: randomized, global, placebo-controlled, double-blind phase 3 study of tislelizumab plus chemotherapy versus chemotherapy as first-line treatment for advanced or metastatic esophageal squamous cell carcinoma (ESCC). Ann Oncol 2022;33:S375. https://doi.org/10.1016/j.annonc.2022.04.439.Search in Google Scholar
16. Moehler, MH, Kato, K, Arkenau, H-T, Oh, D-Y, Tabernero, J, Cruz-Correa, M, et al.. Rationale 305: phase 3 study of tislelizumab plus chemotherapy vs placebo plus chemotherapy as first-line treatment (1L) of advanced gastric or gastroesophageal junction adenocarcinoma (GC/GEJC). J Clin Oncol 2023;41:286. https://doi.org/10.1200/jco.2023.41.4_suppl.286.Search in Google Scholar
17. Shen, L, Kato, K, Kim, S-B, Ajani, JA, Zhao, K, He, Z, et al.. Tislelizumab versus chemotherapy as second-line treatment for advanced or metastatic esophageal squamous cell carcinoma (RATIONALE-302): a randomized phase III study. J Clin Oncol 2022;40:3065–76. https://doi.org/10.1200/jco.21.01926.Search in Google Scholar
18. Luo, H, Lu, J, Bai, Y, Mao, T, Wang, J, Fan, Q, 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. J Am Med Assoc 2021;326:916–25. https://doi.org/10.1001/jama.2021.12836.Search in Google Scholar PubMed PubMed Central
19. Shen, L, Lu, ZH, Wang, JY, Shu, YQ, Kong, L, Yang, L, et al.. LBA52 Sintilimab plus chemotherapy versus chemotherapy as first-line therapy in patients with advanced or metastatic esophageal squamous cell cancer: first results of the phase III ORIENT-15 study. Ann Oncol 2021;32:S1330. https://doi.org/10.1016/j.annonc.2021.08.2132.Search in Google Scholar
20. Wang, ZX, Cui, C, Yao, J, Zhang, Y, Li, M, Feng, J, et al.. Toripalimab plus chemotherapy in treatment-naïve, advanced esophageal squamous cell carcinoma (JUPITER-06): a multi-center phase 3 trial. Cancer Cell 2022;40:277–88.e273. https://doi.org/10.1016/j.ccell.2022.02.007.Search in Google Scholar PubMed
21. Shitara, K, Lordick, F, Bang, Y-J, Enzinger, PC, Ilson, DH, Shah, MA, et al.. Zolbetuximab + mFOLFOX6 as first-line (1L) treatment for patients (pts) withclaudin-18.2+ (CLDN18.2+) / HER2− locally advanced (LA) unresectable or metastatic gastric or gastroesophageal junction (mG/GEJ) adenocarcinoma: primary results from phase 3 SPOTLIGHT study. J Clin Oncol 2023;41:LBA292–2. https://doi.org/10.1200/jco.2023.41.4_suppl.lba292.Search in Google Scholar
22. Bang, Y-J, Van Cutsem, E, Feyereislova, A, Chung, HC, Shen, L, Sawaki, A, et al.. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010;376:687–97. https://doi.org/10.1016/s0140-6736(10)61121-x.Search in Google Scholar
23. Janjigian, YY, Kawazoe, A, Yañez, P, Li, N, Lonardi, S, Kolesnik, O, et al.. The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer. Nature 2021;600:727–30. https://doi.org/10.1038/s41586-021-04161-3.Search in Google Scholar PubMed PubMed Central
24. Janjigian, YY, Viglianti, N, Liu, F, Mendoza-Naranjo, A, Croydon, L. A phase Ib/II, multicenter, open-label, dose-escalation, and dose-expansion study evaluating trastuzumab deruxtecan (T-DXd, DS-8201) monotherapy and combinations in patients with HER2-overexpressing gastric cancer (DESTINY-Gastric03). J Clin Oncol 2021;39:TPS261–1. https://doi.org/10.1200/jco.2021.39.3_suppl.tps261.Search in Google Scholar
25. Högner, A, Thuss-Patience, P. Aktuelles und neue Perspektiven zum Plattenepithelkarzinom des Ösophagus. Trillium Krebsmed 2022:6–7.Search in Google Scholar
© 2024 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Editorial
- The evolving role of esophageal surgery – advancements, challenges, and the path forward
- Reviews
- Neoadjuvant treatment of esophageal cancer: chemotherapy, chemoradiation, immunotherapy, and future trends of therapy
- Active surveillance in patients with a complete clinical response after neoadjuvant chemoradiotherapy for esophageal- and gastroesophageal junction cancer
- Management of subepithelial esophageal tumors
- What is the best surgical approach for esophageal cancer?
- Immunotherapy as a new perspective for the therapy of esophageal cancer
- Voluminous hiatal hernias – the role of robotic surgery
- Oligometastatic disease – a renaissance for surgery?
Articles in the same Issue
- Frontmatter
- Editorial
- The evolving role of esophageal surgery – advancements, challenges, and the path forward
- Reviews
- Neoadjuvant treatment of esophageal cancer: chemotherapy, chemoradiation, immunotherapy, and future trends of therapy
- Active surveillance in patients with a complete clinical response after neoadjuvant chemoradiotherapy for esophageal- and gastroesophageal junction cancer
- Management of subepithelial esophageal tumors
- What is the best surgical approach for esophageal cancer?
- Immunotherapy as a new perspective for the therapy of esophageal cancer
- Voluminous hiatal hernias – the role of robotic surgery
- Oligometastatic disease – a renaissance for surgery?