Updates on the Treatment of Gastroesophageal Cancer

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Clinical Research of Cancer".

Deadline for manuscript submissions: closed (15 November 2022) | Viewed by 21981

Special Issue Editors


E-Mail Website
Guest Editor
Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
Interests: GI cancers; translational research; immunotherapy

E-Mail Website
Guest Editor
1st Department of Surgery, National and Kapodistrian University of Athens, Athens, Greece
Interests: esophageal cancer; gastric cancer; pancreatic cancer; immunotherapy; personalized medicine; surgery; medical education
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Gastroesophageal cancer remains a leading cause of death worldwide, despite the recent progress in early detection, diagnosis and treatment. Surgery has traditionally been the gold standard treatment for gastroesophageal cancer, offering the best possible long-term survival. However, the induction of neoadjuvant therapy has led to an increase in R0 resections, overall survival and disease free survival, so that nowdays multimodality treatment seems to represent the current standard of care. Furthermore, the application of novel surgical techniques, such as laparoscopic and robotic resections, have decreased postoperative morbidity and mortality and improved patients’ quality of life. Additionally, the introduction of immunotherapy and molecular targeted agents seem to enhance the efficacy of the traditionally administered chemotherapy in certain patient cohorts. New predictive biomarkers are being evaluated to identify the patients’ subgroups most benefited and set the optimal treatment algorithm in each clinical scenario.

The aim of this Special Issue is to highlight recent progress and future perspectives in multidisciplinary management including diagnosis, staging, surgery, chemotherapy, radiotherapy and immunotherapy in the adjuvant, neoadjuvant and metastatic setting.

Dr. Michalis V. Karamouzis
Dr. Dimitrios Schizas
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Gastric Cancer
  • Esophageal Cancer
  • Multimodality Treatment
  • Chemotherapy
  • Radiotherapy
  • Immunotherapy
  • Imaging
  • Minimally Invasive Surgery

Published Papers (11 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

11 pages, 2076 KiB  
Article
Assessment of Narrow Band Imaging Algorithm for Video Capsule Endoscopy Based on Decorrelated Color Space for Esophageal Cancer
by Kai-Yao Yang, Yu-Jen Fang, Riya Karmakar, Arvind Mukundan, Yu-Ming Tsao, Chien-Wei Huang and Hsiang-Chen Wang
Cancers 2023, 15(19), 4715; https://doi.org/10.3390/cancers15194715 - 25 Sep 2023
Cited by 3 | Viewed by 977
Abstract
Video capsule endoscopy (VCE) is increasingly used to decrease discomfort among patients owing to its small size. However, VCE has a major drawback of not having narrow band imaging (NBI) functionality. The current VCE has the traditional white light imaging (WLI) only, which [...] Read more.
Video capsule endoscopy (VCE) is increasingly used to decrease discomfort among patients owing to its small size. However, VCE has a major drawback of not having narrow band imaging (NBI) functionality. The current VCE has the traditional white light imaging (WLI) only, which has poor performance in the computer-aided detection (CAD) of different types of cancer compared to NBI. Specific cancers, such as esophageal cancer (EC), do not exhibit any early biomarkers, making their early detection difficult. In most cases, the symptoms are unnoticeable, and EC is diagnosed only in later stages, making its 5-year survival rate below 20% on average. NBI filters provide particular wavelengths that increase the contrast and enhance certain features of the mucosa, thereby enabling early identification of EC. However, VCE does not have a slot for NBI functionality because its size cannot be increased. Hence, NBI image conversion from WLI can presently only be achieved in post-processing. In this study, a complete arithmetic assessment of the decorrelated color space was conducted to generate NBI images from WLI images for VCE of the esophagus. Three parameters, structural similarity index metric (SSIM), entropy, and peak-signal-to-noise ratio (PSNR), were used to assess the simulated NBI images. Results show the good performance of the NBI image reproduction method with SSIM, entropy difference, and PSNR values of 93.215%, 4.360, and 28.064 dB, respectively. Full article
(This article belongs to the Special Issue Updates on the Treatment of Gastroesophageal Cancer)
Show Figures

Figure 1

16 pages, 3156 KiB  
Article
Preparing Well for Esophageal Endoscopic Detection Using a Hybrid Model and Transfer Learning
by Chu-Kuang Chou, Hong-Thai Nguyen, Yao-Kuang Wang, Tsung-Hsien Chen, I-Chen Wu, Chien-Wei Huang and Hsiang-Chen Wang
Cancers 2023, 15(15), 3783; https://doi.org/10.3390/cancers15153783 - 26 Jul 2023
Viewed by 1076
Abstract
Early detection of esophageal cancer through endoscopic imaging is pivotal for effective treatment. However, the intricacies of endoscopic diagnosis, contingent on the physician’s expertise, pose challenges. Esophageal cancer features often manifest ambiguously, leading to potential confusions with other inflammatory esophageal conditions, thereby complicating [...] Read more.
Early detection of esophageal cancer through endoscopic imaging is pivotal for effective treatment. However, the intricacies of endoscopic diagnosis, contingent on the physician’s expertise, pose challenges. Esophageal cancer features often manifest ambiguously, leading to potential confusions with other inflammatory esophageal conditions, thereby complicating diagnostic accuracy. In recent times, computer-aided diagnosis has emerged as a promising solution in medical imaging, particularly within the domain of endoscopy. Nonetheless, contemporary AI-based diagnostic models heavily rely on voluminous data sources, limiting their applicability, especially in scenarios with scarce datasets. To address this limitation, our study introduces novel data training strategies based on transfer learning, tailored to optimize performance with limited data. Additionally, we propose a hybrid model integrating EfficientNet and Vision Transformer networks to enhance prediction accuracy. Conducting rigorous evaluations on a carefully curated dataset comprising 1002 endoscopic images (comprising 650 white-light images and 352 narrow-band images), our model achieved exceptional outcomes. Our combined model achieved an accuracy of 96.32%, precision of 96.44%, recall of 95.70%, and f1-score of 96.04%, surpassing state-of-the-art models and individual components, substantiating its potential for precise medical image classification. The AI-based medical image prediction platform presents several advantageous characteristics, encompassing superior prediction accuracy, a compact model size, and adaptability to low-data scenarios. This research heralds a significant stride in the advancement of computer-aided endoscopic imaging for improved esophageal cancer diagnosis. Full article
(This article belongs to the Special Issue Updates on the Treatment of Gastroesophageal Cancer)
Show Figures

Figure 1

12 pages, 1298 KiB  
Article
Neoadjuvant Chemoradiotherapy versus Chemotherapy for Gastroesophageal Junction Adenocarcinoma; Which Is the Optimal Treatment Option?
by Eric Zandirad, Hugo Teixeira Farinha, Beatriz Barberá-Carbonell, Sandrine Geinoz, Nicolas Demartines, Markus Schäfer and Styliani Mantziari
Cancers 2022, 14(23), 5856; https://doi.org/10.3390/cancers14235856 - 28 Nov 2022
Viewed by 1337
Abstract
Background: Locally advanced gastroesophageal junction adenocarcinoma (GEJ) is treated with either perioperative chemotherapy (CT) or preoperative radiochemotherapy (RCT) followed by surgery. The aim of this study was to compare pathologic response and long-term outcomes in junction adenocarcinoma treated with neoadjuvant RCT versus [...] Read more.
Background: Locally advanced gastroesophageal junction adenocarcinoma (GEJ) is treated with either perioperative chemotherapy (CT) or preoperative radiochemotherapy (RCT) followed by surgery. The aim of this study was to compare pathologic response and long-term outcomes in junction adenocarcinoma treated with neoadjuvant RCT versus CT. Methods: All patients with locally advanced GEJ adenocarcinoma treated with neoadjuvant treatment (NAT) followed by surgery between 2009 and 2018 were retrospectively analyzed. Results: A total of 94 patients were included, 67 (71.2%) RCT and 27 (28.8%) CT. Complete pathologic response was more frequent in RCT patients (13.4% vs. 7.4%, p = 0.009) with a trend to better lymph node control (ypN0) (55.2% vs. 33.3%; p = 0.057). RCT offered no benefit in R0 resection (66.7% vs. 72.1% CT, p = 0.628) and was related to higher postoperative cardiovascular complications (35.8% vs. 11.1%; p = 0.017). Long-term overall and disease-free survival were similar (5-year OS 61.1% RCT vs. 75.7% CT, p = 0.259; 5-year DFS 33.5% RCT vs. 22.8% CT; p = 0.763). NAT type was neither independently associated with pathologic response nor long-term survival. Discussion: Patients with locally advanced GEJ adenocarcinoma treated with RCT had more postoperative cardiovascular complications but higher rates of complete pathologic response and a trend to superior locoregional lymph node control. This did not translate in a survival or recurrence benefit. Full article
(This article belongs to the Special Issue Updates on the Treatment of Gastroesophageal Cancer)
Show Figures

Figure 1

12 pages, 2044 KiB  
Article
Fluorescent Lymphography-Guided Lymphadenectomy during Minimally Invasive Completion Total Gastrectomy for Remnant Gastric Cancer Patients
by Nasser Alrashidi, Ki-Yoon Kim, Sung Hyun Park, Sejin Lee, Minah Cho, Yoo Min Kim, Hyoung-Il Kim and Woo Jin Hyung
Cancers 2022, 14(20), 5037; https://doi.org/10.3390/cancers14205037 - 14 Oct 2022
Cited by 3 | Viewed by 1285
Abstract
No study has evaluated fluorescent lymphography for lymphadenectomy in remnant gastric cancer (RGC). This study aimed to assess the clinical application of fluorescent lymphography in minimally invasive completion total gastrectomy for RGC. Patients who had undergone minimally invasive completion total gastrectomy for RGC [...] Read more.
No study has evaluated fluorescent lymphography for lymphadenectomy in remnant gastric cancer (RGC). This study aimed to assess the clinical application of fluorescent lymphography in minimally invasive completion total gastrectomy for RGC. Patients who had undergone minimally invasive completion total gastrectomy for RGC from 2013 to 2020 were retrospectively reviewed. The perioperative outcomes and long-term prognosis were compared between patients who had undergone minimally invasive completion total gastrectomy with fluorescent lymphography (the FL group) and those without fluorescent lymphography (the non-FL group). The FL group comprised 32 patients, and the non-FL group comprised 36 patients. FL visualized lymphatics in all 32 patients without complications related to the fluorescent injection. The median number [the interquartile range] of LN retrieval was significantly higher in the FL group (17 [9.3–23.5]) than in the non-FL group (12.5 [4–17.8]); p = 0.016). The sensitivity of fluorescent lymphography in detecting metastatic LN stations was 75%, and the negative predictive value was 96.9% in the FL group. The overall relapse-free survivals were comparable between the groups (p = 0.833 and p = 0.524, respectively). FL is an effective tool to perform a more thorough lymphadenectomy during minimally invasive completion total gastrectomy for RGC. Using FL in RGC surgery may improve surgical quality and proper staging. Full article
(This article belongs to the Special Issue Updates on the Treatment of Gastroesophageal Cancer)
Show Figures

Figure 1

10 pages, 823 KiB  
Article
Prognostic Value of KRAS Mutations in Colorectal Cancer Patients
by Asimina Koulouridi, Michaela Karagianni, Ippokratis Messaritakis, Maria Sfakianaki, Alexandra Voutsina, Maria Trypaki, Maria Bachlitzanaki, Evangelos Koustas, Michalis V. Karamouzis, Anastasios Ntavatzikos, Anna Koumarianou, Nikolaos Androulakis, Dimitrios Mavroudis, Maria Tzardi and John Souglakos
Cancers 2022, 14(14), 3320; https://doi.org/10.3390/cancers14143320 - 07 Jul 2022
Cited by 17 | Viewed by 2423
Abstract
Colorectal cancer (CRC) remains a major public health issue. The detection of parameters that affect CRC prognosis is of great significance. KRAS mutations, play a crucial role in tumorigenesis with a strong predictive value. KRAS-mutated stage-IV CRC patients gain no benefit of [...] Read more.
Colorectal cancer (CRC) remains a major public health issue. The detection of parameters that affect CRC prognosis is of great significance. KRAS mutations, play a crucial role in tumorigenesis with a strong predictive value. KRAS-mutated stage-IV CRC patients gain no benefit of the anti-EGFR therapy. The KRAS G12C mutation subtype is under investigation for treatment regimens. The present study aimed to detect various RAS mutations in a cohort of 578 RAS-mutated CRC patients; 49% of them had de novo metastatic disease; 60% were male; 71.4% had left-sided tumors; and 94.6% had a good performance status. KRAS mutations were detected in 93.2% of patients, with KRAS G12D being the most common subtype (30.1%). KRAS mutations presented shorter progression-free (PFS) and overall survival (OS), compared with NRAS mutations, although not significantly (PFS: 13.8 vs. 18.5 months; p = 0.552; OS: 53.1 vs. 60.9 months; p = 0.249). KRAS G12D mutations presented better OS rates (p = 0.04). KRAS G12C mutation, even though not significantly, presented worse PFS and OS rates. KRAS exon 3 and 4 mutations presented different PFS and OS rates, although these were not significant. Concluding, KRAS G12D and G12C mutations lead to better and worst prognosis, respectively. Further studies are warranted to validate such findings and their possible therapeutic implication. Full article
(This article belongs to the Special Issue Updates on the Treatment of Gastroesophageal Cancer)
Show Figures

Figure 1

Review

Jump to: Research, Other

11 pages, 274 KiB  
Review
Human Epidermal Growth Factor Receptor-2 Gastric Adenocarcinoma: Expanding Therapy of a Recognized Target
by Jane E. Rogers, Kohei Yamashita, Matheus Sewastjanow-Silva, Allison Trail, Rebecca E. Waters and Jaffer Ajani
Cancers 2023, 15(21), 5180; https://doi.org/10.3390/cancers15215180 - 27 Oct 2023
Cited by 2 | Viewed by 1068
Abstract
Human epidermal growth factor receptor-2 (HER2) is a well-known cancer target. Many HER2-targeted agents are marketed and being investigated. Unfortunately, these therapies lack consistent responses and outcomes amongst different tumors. Questions remain as to why HER2 biology is different in different tumor types. [...] Read more.
Human epidermal growth factor receptor-2 (HER2) is a well-known cancer target. Many HER2-targeted agents are marketed and being investigated. Unfortunately, these therapies lack consistent responses and outcomes amongst different tumors. Questions remain as to why HER2 biology is different in different tumor types. Gastric adenocarcinomas (GACs) demonstrate both intra- and inter-tumor HER2 expression heterogeneity and show discordance amongst primary and metastatic disease sites. This creates barriers in determining HER2 agents’ effectiveness and contributes to the failure of some HER2-targeted agents in the treatment of HER2-positive advanced GACs. Trastuzumab deruxtecan, an antibody drug conjugate of trastuzumab with a topoisomerase inhibitor, was recently approved for the treatment of refractory HER2-positive advanced GAC patients. There are exciting and newer therapies under investigation. Examining resistance patterns (both adaptive and acquired) along with establishing a better understanding of the intra- and inter-tumor heterogeneity is necessary to ensure successful progress. Here we review the current status of HER2-targeted therapy in GACs. We additionally review newer therapies under investigation and their potential role in HER2 GACs. Full article
(This article belongs to the Special Issue Updates on the Treatment of Gastroesophageal Cancer)
18 pages, 330 KiB  
Review
Recent Advances in the Systemic Treatment of Localized Gastroesophageal Cancer
by Hannah Christina Puhr, Thorsten J. Reiter, Matthias Preusser, Gerald W. Prager and Aysegül Ilhan-Mutlu
Cancers 2023, 15(6), 1900; https://doi.org/10.3390/cancers15061900 - 22 Mar 2023
Cited by 2 | Viewed by 1681
Abstract
The overall survival expectancy of localized gastroesophageal cancer patients still remains under 5 years despite advances in neoadjuvant and adjuvant treatment strategies in recent years. For almost a decade, immunotherapy has been successfully implemented as a first-line treatment for various oncological diseases in [...] Read more.
The overall survival expectancy of localized gastroesophageal cancer patients still remains under 5 years despite advances in neoadjuvant and adjuvant treatment strategies in recent years. For almost a decade, immunotherapy has been successfully implemented as a first-line treatment for various oncological diseases in advanced stages. In the case of advanced gastroesophageal cancer, 2021 witnessed several approvals of immune checkpoint inhibitor therapies by different authorities. Although it is still a debate whether this treatment should be restricted to a certain subgroup of patients based on biomarker selection, immunotherapy agents are making remarkable steps in resectable settings as well. The Checkmate-577 study demonstrated significant benefits of nivolumab as an adjuvant treatment for resectable esophageal and gastroesophageal junction tumors and thereby obtained approvals both from U.S. American and European authorities. First results of further potential practice-changing clinical trials are expected in 2023, which might change the treatment armamentarium for resectable gastroesophageal cancers significantly. This review aims to demonstrate the advances of immunotherapy and targeted therapies in treatment of localized gastric, gastroesophageal junction and esophageal tumors and gives a short summary on promising ongoing clinical trials. Full article
(This article belongs to the Special Issue Updates on the Treatment of Gastroesophageal Cancer)
12 pages, 280 KiB  
Review
Current Immune Checkpoint Inhibitor Genetic Biomarker Exploration in Gastrointestinal Tumors
by Jane E. Rogers, Kohei Yamashita, Matheus Sewastjanow Silva and Jaffer A. Ajani
Cancers 2022, 14(19), 4804; https://doi.org/10.3390/cancers14194804 - 30 Sep 2022
Viewed by 1386
Abstract
Immune checkpoint inhibitors have revolutionized cancer management. Some patients with gastrointestinal (GI) tract malignancy have experienced remarkable results. Here, in our review, we discuss predictive/prognostic GI tumor biomarkers that appear to correlate with benefits with this strategy. Remarkable progress has been made in [...] Read more.
Immune checkpoint inhibitors have revolutionized cancer management. Some patients with gastrointestinal (GI) tract malignancy have experienced remarkable results. Here, in our review, we discuss predictive/prognostic GI tumor biomarkers that appear to correlate with benefits with this strategy. Remarkable progress has been made in certain subsets of patients including the potential for solid tumor patients to avoid local therapies such as radiation and/or surgery (organ preservation), which come with acute and chronic risks that have historically been the only curable strategies for these GI tumors. These results provide new and exciting strategies for solid tumor management. Unfortunately, immune checkpoint inhibitors can correlate with biomarkers, but benefits occur in a small subset of patients with GI malignancies. Most frequently, immune checkpoint inhibitors fail to induce response in GI malignancies due to the “cold” tumor microenvironment that protects cancer. Translational strategies are needed to develop effective combination strategies and novel biomarkers to overcome the intrinsic resistance. Full article
(This article belongs to the Special Issue Updates on the Treatment of Gastroesophageal Cancer)

Other

Jump to: Research, Review

22 pages, 4945 KiB  
Systematic Review
Robot-Assisted Minimally Invasive Esophagectomy versus Open Esophagectomy for Esophageal Cancer: A Systematic Review and Meta-Analysis
by Stepan M. Esagian, Ioannis A. Ziogas, Konstantinos Skarentzos, Ioannis Katsaros, Georgios Tsoulfas, Daniela Molena, Michalis V. Karamouzis, Ioannis Rouvelas, Magnus Nilsson and Dimitrios Schizas
Cancers 2022, 14(13), 3177; https://doi.org/10.3390/cancers14133177 - 29 Jun 2022
Cited by 12 | Viewed by 2109
Abstract
Robot-assisted minimally invasive esophagectomy (RAMIE) was introduced as a further development of the conventional minimally invasive esophagectomy, aiming to further improve the high morbidity and mortality associated with open esophagectomy. We aimed to compare the outcomes between RAMIE and open esophagectomy, which remains [...] Read more.
Robot-assisted minimally invasive esophagectomy (RAMIE) was introduced as a further development of the conventional minimally invasive esophagectomy, aiming to further improve the high morbidity and mortality associated with open esophagectomy. We aimed to compare the outcomes between RAMIE and open esophagectomy, which remains a popular approach for resectable esophageal cancer. Ten studies meeting our inclusion criteria were identified, including five retrospective cohort, four prospective cohort, and one randomized controlled trial. RAMIE was associated with significantly lower rates of overall pulmonary complications (odds ratio (OR): 0.38, 95% confidence interval (CI): [0.26, 0.56]), pneumonia (OR: 0.39, 95% CI: [0.26, 0.57]), atrial fibrillation (OR: 0.53, 95% CI: [0.29, 0.98]), and wound infections (OR: 0.20, 95% CI: [0.07, 0.57]) and resulted in less blood loss (weighted mean difference (WMD): −187.08 mL, 95% CI: [−283.81, −90.35]) and shorter hospital stays (WMD: −9.22 days, 95% CI: [−14.39, −4.06]) but longer operative times (WMD: 69.45 min, 95% CI: [34.39, 104.42]). No other statistically significant difference was observed regarding surgical and short-term oncological outcomes. Similar findings were observed when comparing totally robotic procedures only to OE. RAMIE is a safe and feasible procedure, resulting in decreased cardiopulmonary morbidity, wound infections, blood loss, and shorter hospital stays compared to open esophagectomy. Full article
(This article belongs to the Special Issue Updates on the Treatment of Gastroesophageal Cancer)
Show Figures

Figure 1

15 pages, 1115 KiB  
Systematic Review
Role of Radiology in the Preoperative Detection of Arterial Calcification and Celiac Trunk Stenosis and Its Association with Anastomotic Leakage Post Esophagectomy, an Up-to-Date Review of the Literature
by Antonios Tzortzakakis, Georgios Kalarakis, Biying Huang, Eleni Terezaki, Emmanouil Koltsakis, Aristotelis Kechagias, Andrianos Tsekrekos and Ioannis Rouvelas
Cancers 2022, 14(4), 1016; https://doi.org/10.3390/cancers14041016 - 17 Feb 2022
Cited by 6 | Viewed by 2906
Abstract
Surgical resection of the esophagus remains a critical component of the multimodal treatment of esophageal cancer. Anastomotic leakage (AL) is the most significant complication following esophagectomy, in terms of clinical implications. Identifying risk factors for AL is important for modifying patient management and [...] Read more.
Surgical resection of the esophagus remains a critical component of the multimodal treatment of esophageal cancer. Anastomotic leakage (AL) is the most significant complication following esophagectomy, in terms of clinical implications. Identifying risk factors for AL is important for modifying patient management and improving surgical outcomes. This review aims to examine the role of radiological risk factors for AL after esophagectomy, and in particular, arterial calcification and celiac trunk stenosis. Eligible publications prior to 25 August 2021 were retrieved from Medline and Google Scholar using a predefined search algorithm. A total of 68 publications were identified, of which 9 original studies remained for in-depth analysis. The majority of these studies found correlations between calcifications in the aorta, celiac trunk, and right post-celiac arteries and AL following esophagectomy. Some studies suggest celiac trunk stenosis as a more appropriate surrogate. Our up-to-date review highlights the need for automated quantification of aortic calcifications, as well as the degree of celiac trunk stenosis in preoperative computed tomography in patients undergoing esophagectomy, to obtain robust and reproducible measurements that can be used for a definite correlation. Full article
(This article belongs to the Special Issue Updates on the Treatment of Gastroesophageal Cancer)
Show Figures

Figure 1

14 pages, 1693 KiB  
Systematic Review
Comparative Efficacy and Safety of Programmed Death-1 Pathway Inhibitors in Advanced Gastroesophageal Cancers: A Systematic Review and Network Meta-Analysis of Phase III Clinical Trials
by Laercio Lopes da Silva, Pedro Nazareth Aguiar, Jr., Robin Park, Eduardo Edelman Saul, Benjamin Haaland and Gilberto de Lima Lopes
Cancers 2021, 13(11), 2614; https://doi.org/10.3390/cancers13112614 - 26 May 2021
Cited by 12 | Viewed by 4560
Abstract
Background: The use of checkpoint inhibitors has changed the treatment landscape for gastroesophageal cancer in the third-line setting. However, success rates in earlier treatment lines are highly variable across trials. Herein, we compare the efficacy and safety of the different anti-PD-1/PD-L1 regimens [...] Read more.
Background: The use of checkpoint inhibitors has changed the treatment landscape for gastroesophageal cancer in the third-line setting. However, success rates in earlier treatment lines are highly variable across trials. Herein, we compare the efficacy and safety of the different anti-PD-1/PD-L1 regimens with or without chemotherapy; Methods: We performed a network meta-analysis (NMA) of anti-PD-1/PD-L1 monotherapy or combined with chemotherapy (chemoimmunotherapy) for gastroesophageal cancers without ERBB2 overexpression; Results: The first-line NMA included four trials (N = 3817), showing that chemoimmunotherapy improved OS and PFS without significant safety difference: Nivolumab-chemotherapy, OS (HR: 0.83 [95% CI, 0.75–0.92]), PFS (HR 0.68 [95% CI, 0.57–0.81]), Pembrolizumab-chemotherapy: OS (HR 0.77 [95% CI, 0.67–0.88]), PFS (HR: 0.72 [95% CI, 0.60–0.85]. Pembrolizumab monotherapy was the safest first-line treatment, SAE (OR 0.02 [95% CI, 0.00–0.2]) but showed no survival benefit. The second-line NMA encompassed four trials (N = 2087), showing that anti-PD-1 significantly improved safety but not survival: camrelizumab, SAE (OR 0.37; [95% CI, 0.24–0.56]); nivolumab, SAE (OR 0.13, [95% CI, 0.08–0.2]) pembrolizumab, SAE (OR 0.4; [95% CI, 0.30–0.53]); Conclusions: chemoimmunotherapy improves OS and PFS in previously untreated gastroesophageal cancers. Anti-PD-1 monotherapies improve safety in refractory disease, with no significant survival benefit. Full article
(This article belongs to the Special Issue Updates on the Treatment of Gastroesophageal Cancer)
Show Figures

Figure 1

Back to TopTop