In Search of More Effective and Less Invasive Ways to Treat Esophagus and Stomach Cancer

A special issue of Current Oncology (ISSN 1718-7729). This special issue belongs to the section "Gastrointestinal Oncology".

Deadline for manuscript submissions: closed (31 January 2022) | Viewed by 34230

Special Issue Editors


E-Mail Website
Guest Editor
Department of Hematology, Oncology and Cancer-Immunology, Charité – University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
Interests: clinical trials in GI oncology; gastric cancer; esophageal cancer; palliative care

E-Mail Website
Guest Editor
Department of Surgery, Campus Virchow Klinikum/Campus Charite Mitte, Charite University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
Interests: surgical oncology; upper GI cancer; esophageal cancer; gastric cancer; clinical trials; minimally invasive surgery; robotic surgery
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Gastroenterology, Rheumatology und Infectious Diseases, Campus Benjamin Franklin, Charité – University Medicine Berlin, Hindenburgdamm 30, 12 203 Berlin, Germany
Interests: clinical trials in upper GI oncology; translational studies; response prediction; endoscopic diagnosis and treatment

Special Issue Information

Dear Colleagues,

In the last 3 to 5 years, treatment options for esophago-gastric cancer have been rapidly changing. New molecular subgroups with clinical significance have been identified, new targets have been discovered, and promising specific antibodies have been designed. Phase III data have changed treatment algorithms in perioperative, adjuvant and palliative settings. Technical innovations have optimized endoscopic treatment; minimally invasive and robotic surgery techniques are becoming more and more advanced.

This Special Issue is looking for original manuscripts and reviews to explore all areas of treatment, including:

  1. Screening and genetic syndromes;
  2. Molecular classification, prognostic and predictive biomarkers;
  3. New options for endoscopic treatment;
  4. New approaches to optimize perioperative and adjuvant strategies;
  5. New ways to optimize surgical techniques;
  6. Individualized treatment strategies in the palliative setting;
  7. Novel combinations for palliative therapy;
  8. Developments in immunotherapy and beyond.

Manuscript submissions are welcome from now until November 30th 2021 (deadline).

Dr. Peter Thuss-Patience
Prof. Dr. Matthias Biebl
Dr. Severin Daum
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. Current Oncology is an international peer-reviewed open access monthly 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 2200 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

  • esophageal cancer
  • gastric cancer
  • predictive biomarker
  • prognostic biomarker
  • molecular subgroups
  • checkpoint inhibitor
  • minimal invasive surgery
  • robotic surgery
  • endoscopy
  • perioperative treatment
  • chemoradiotherapy
  • adjuvant treatment
  • palliative treatment
  • immunotherapy
  • HER2-directed therapy

Published Papers (8 papers)

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

Research

Jump to: Review, Other

14 pages, 1452 KiB  
Article
Prognostic Relevance of Weight and Weight Loss during Multimodal Therapy for Oesophagogastric Tumours
by Alessandro Lorusso, Dmitry Bichev, Anica Högner, Prisca Bartels, Alexej Ballhausen, Christoph Treese, Matthias Biebl and Peter Thuss-Patience
Curr. Oncol. 2022, 29(4), 2706-2719; https://doi.org/10.3390/curroncol29040221 - 12 Apr 2022
Cited by 1 | Viewed by 1728
Abstract
The prognostic meaning of weight loss (WL) during standard treatment for operable oesophagogastric cancer is still unclear. The aim of this study is to analyse the prognostic effect of WL during perioperative chemotherapy (PC) for gastric cancer (GC) and oesophageal adenocarcinomas (OAC). We [...] Read more.
The prognostic meaning of weight loss (WL) during standard treatment for operable oesophagogastric cancer is still unclear. The aim of this study is to analyse the prognostic effect of WL during perioperative chemotherapy (PC) for gastric cancer (GC) and oesophageal adenocarcinomas (OAC). We retrospectively analysed data from 128 patients (pts) with GC and OAC who underwent surgery in the context of multimodal treatment with PC. We collected data on WL during different steps of therapy together with other histopathologic and demographic information. We analysed the effects on overall survival (OS) and disease-free survival (DFS). Results: Pts with WL ≥ 5% during neoadjuvant chemotherapy exhibited significantly worse OS compared with pts with WL < 5% (median OS: 23.6 months [95% CI: 4.4–42.9] vs. 63.5 months [95% CI: 50.7–76.2], p = 0.007) and DFS (median DFS: 12.5 months [95% CI: 2.9–22.1] vs. 63.5 months [95% CI: 31.6–95.4], p = 0.016). Pts with WL ≥ 14% during the whole treatment exhibited significantly worse OS compared with pts with WL < 14% (median OS: 43.7 months [95% CI: 13.2–74.2] vs. not reached, p = 0.028) and DFS (median DFS: 34.3 months [95% CI: 14.0–54.5] vs. not reached, p = 0.038). Conclusion: WL patterns during neoadjuvant chemotherapy and during the whole treatment correlate with a significantly worse prognosis in operated pts with curative GC or OAC in the context of a multimodal treatment with PC. A validation of this prognostic effect in prospective studies is warranted. Full article
Show Figures

Figure 1

14 pages, 1852 KiB  
Article
Impact of Postoperative Chemotherapy in Patients with Gastric/Gastroesophageal Adenocarcinoma Treated with Perioperative Chemotherapy
by Alexej Ballhausen, Prisca Bartels, Ines Iacovella, Anica Hoegner, Alessandro Lorusso, Dmitry Bichev, Severin Daum and Peter Thuss-Patience
Curr. Oncol. 2022, 29(3), 1983-1996; https://doi.org/10.3390/curroncol29030161 - 14 Mar 2022
Viewed by 2100
Abstract
Perioperative chemotherapy is the standard of care for patients undergoing curative resection for gastroesophageal adenocarcinoma. However, less than 50% of patients complete postoperative chemotherapy, and the added benefit to preoperative chemotherapy remains unclear. The aim of this study was to compare disease-free and [...] Read more.
Perioperative chemotherapy is the standard of care for patients undergoing curative resection for gastroesophageal adenocarcinoma. However, less than 50% of patients complete postoperative chemotherapy, and the added benefit to preoperative chemotherapy remains unclear. The aim of this study was to compare disease-free and overall survival (DFS and OS) in patients with perioperative chemotherapy to those who received preoperative chemotherapy only. In addition, a current literature overview is included. This multicenter, retrospective case series included 124 patients with gastroesophageal adenocarcinoma undergoing potentially curative resection and receiving pre- or perioperative chemotherapy between 2006 and 2010. Histopathological, demographic, clinical, and survival data were used to identify the impact of perioperative vs. preoperative chemotherapy on DFS and OS. Patients with perioperative chemotherapy had significantly improved DFS and OS (median DFS 28.0 months; 95%CI 0–62.4 vs. 19.0 months; 95%CI 10.5–27.5; p = 0.008 and median OS 35.7 months; 95%CI 0–73.6 vs. 19.2 months; 95%CI 7.8–30.4; p = 0.002). However, in contrast to patients with tumor-free lymph nodes at the time of resection, patients with positive lymph node status did not significantly benefit from additional postoperative chemotherapy in subgroup analysis. Further studies are encouraged to investigate optimal adjuvant treatment strategies for primary chemotherapy-resistant patients. Full article
Show Figures

Figure 1

12 pages, 2339 KiB  
Article
HMGA1 Has Predictive Value in Response to Chemotherapy in Gastric Cancer
by Diana Pádua, Débora Filipa Pinto, Paula Figueira, Carlos Filipe Pereira, Raquel Almeida and Patrícia Mesquita
Curr. Oncol. 2022, 29(1), 56-67; https://doi.org/10.3390/curroncol29010005 - 23 Dec 2021
Cited by 5 | Viewed by 3345
Abstract
Gastric cancer is a serious health problem worldwide. Although its incidence is decreasing, the five-year survival rate remains low. Thus, it is essential to identify new biomarkers that could promote better diagnosis and treatment of patients with gastric cancer. High-mobility group AT-hook 1 [...] Read more.
Gastric cancer is a serious health problem worldwide. Although its incidence is decreasing, the five-year survival rate remains low. Thus, it is essential to identify new biomarkers that could promote better diagnosis and treatment of patients with gastric cancer. High-mobility group AT-hook 1 (HMGA1) is a non-histone, chromatin-binding protein that has been found overexpressed in several tumor types. It has been correlated with invasion, metastasis, and drug resistance, leading to worse patient survival. The aim of this work was to evaluate the clinical value of HMGA1 in gastric cancer. HMGA1 expression was analyzed by immunohistochemistry in a single hospital series (n = 323) of gastric adenocarcinoma cases (stages I to IV) with clinicopathological and treatment data. In this series, HMGA1 expression showed no significant relevance as a prognostic biomarker. Nevertheless, a significantly better overall survival was observed in cases with high levels of HMGA1 when they were treated with chemotherapy, compared to the nontreated ones, implying that they can benefit more from treatment than patients with low expression of HMGA1. We thereby show for the first time that HMGA1 expression has a substantial value as a biomarker of response to chemotherapy in gastric cancer. Full article
Show Figures

Figure 1

Review

Jump to: Research, Other

11 pages, 380 KiB  
Review
Immunotherapy in Squamous Cell Cancer of the Esophagus
by Peter Thuss-Patience and Alexander Stein
Curr. Oncol. 2022, 29(4), 2461-2471; https://doi.org/10.3390/curroncol29040200 - 30 Mar 2022
Cited by 18 | Viewed by 5169
Abstract
Treatment of esophageal carcinoma has changed dramatically following several landmark trials, which have proven the benefit of immunotherapy. The selective PD-1 (programmed cell death ligand-1)-inhibitor nivolumab has been shown to improve DFS in the adjuvant therapy setting (CheckMate-577). In the first-line treatment, PD-L1 [...] Read more.
Treatment of esophageal carcinoma has changed dramatically following several landmark trials, which have proven the benefit of immunotherapy. The selective PD-1 (programmed cell death ligand-1)-inhibitor nivolumab has been shown to improve DFS in the adjuvant therapy setting (CheckMate-577). In the first-line treatment, PD-L1 positive (CPS ≥ 10) squamous cell carcinoma patients (pts) have been shown to have an increased OS following treatment with the PD-1-inhibitor pembrolizumab in combination with chemotherapy (KEYNOTE-590). Nivolumab also improved overall survival in the first line setting either combined with ipilimumab or with chemotherapy (CheckMate 648) compared to chemotherapy alone. In Asian first-line patients, phase III trials investigating camrelizumab (ESCORT 1), toripalimab (JUPITER 06), or sintilimab (ORIENT 15) in addition to chemotherapy also showed significant survival benefits. In the second-line setting, monotherapy with nivolumab (ATTRACTION-03), pembrolizumab (KEYNOTE-181), camrelizumab (ESCORT), and tislelizumab (RATIONALE 302) demonstrated a benefit in OS in comparison to chemotherapy. Here we will review these trials and integrate them into the current treatment algorithm. Full article
Show Figures

Figure 1

24 pages, 3114 KiB  
Review
A Pleiotropic Role of Long Non-Coding RNAs in the Modulation of Wnt/β-Catenin and PI3K/Akt/mTOR Signaling Pathways in Esophageal Squamous Cell Carcinoma: Implication in Chemotherapeutic Drug Response
by Uttam Sharma, Masang Murmu, Tushar Singh Barwal, Hardeep Singh Tuli, Manju Jain, Hridayesh Prakash, Tea Kaceli, Aklank Jain and Anupam Bishayee
Curr. Oncol. 2022, 29(4), 2326-2349; https://doi.org/10.3390/curroncol29040189 - 26 Mar 2022
Cited by 7 | Viewed by 3844
Abstract
Despite the availability of modern techniques for the treatment of esophageal squamous cell carcinoma (ESCC), tumor recurrence and metastasis are significant challenges in clinical management. Thus, ESCC possesses a poor prognosis and low five-year overall survival rate. Notably, the origin and recurrence of [...] Read more.
Despite the availability of modern techniques for the treatment of esophageal squamous cell carcinoma (ESCC), tumor recurrence and metastasis are significant challenges in clinical management. Thus, ESCC possesses a poor prognosis and low five-year overall survival rate. Notably, the origin and recurrence of the cancer phenotype are under the control of complex cancer-related signaling pathways. In this review, we provide comprehensive knowledge about long non-coding RNAs (lncRNAs) related to Wnt/β-catenin and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway in ESCC and its implications in hindering the efficacy of chemotherapeutic drugs. We observed that a pool of lncRNAs, such as HERES, TUG1, and UCA1, associated with ESCC, directly or indirectly targets various molecules of the Wnt/β-catenin pathway and facilitates the manifestation of multiple cancer phenotypes, including proliferation, metastasis, relapse, and resistance to anticancer treatment. Additionally, several lncRNAs, such as HCP5 and PTCSC1, modulate PI3K/Akt/mTOR pathways during the ESCC pathogenesis. Furthermore, a few lncRNAs, such as AFAP1-AS1 and LINC01014, block the efficiency of chemotherapeutic drugs, including cisplatin, 5-fluorouracil, paclitaxel, and gefitinib, used for ESCC treatment. Therefore, this review may help in designing a better therapeutic strategy for ESCC patients. Full article
Show Figures

Figure 1

16 pages, 553 KiB  
Review
Immunotherapy in Gastric Cancer
by Anica Högner and Markus Moehler
Curr. Oncol. 2022, 29(3), 1559-1574; https://doi.org/10.3390/curroncol29030131 - 2 Mar 2022
Cited by 67 | Viewed by 10661
Abstract
Immune checkpoint inhibition is a new standard of targeted therapy in the treatment of advanced or metastatic gastric cancer (GC) and is represented in various combinations with and without chemotherapy in every therapy line within clinical trials. In advanced adenocarcinoma of GC, gastroesophageal [...] Read more.
Immune checkpoint inhibition is a new standard of targeted therapy in the treatment of advanced or metastatic gastric cancer (GC) and is represented in various combinations with and without chemotherapy in every therapy line within clinical trials. In advanced adenocarcinoma of GC, gastroesophageal junction cancer (GEJC) and esophageal cancer (EC), the combination of nivolumab and chemotherapy in first-line therapy improves overall survival (OS) in PD-L1 (programmed cell death protein 1)-positive patients with approval in Europe (PD-L1 CPS (combined positivity score) ≥ 5), USA and Taiwan (CHECKMATE-649) and pembrolizumab plus chemotherapy for GEJC and EC in Europe (CPS ≥ 10) and the USA (KEYNOTE-590). Furthermore, pembrolizumab plus trastuzumab and chemotherapy show clear benefits in OS and are approved as first-line treatment of Her2 (human epidermal growth factor receptor-2)-positive tumors in the USA (KEYNOTE-811). Nivolumab demonstrates superior OS regardless of PD-L1 expression in third-line therapy with approval in Japan (ATTRACTION-02) and pembrolizumab prolonged the duration of response in PD-L1 positive patients with approval in the USA in PD-L1 CPS ≥ 1 patients (KEYNOTE-059). This review reflects the rationale and current results of phase II and III clinical trials investigating various immune checkpoint inhibitors targeting PD-L1/1 and CTLA (anticytotoxic T-lymphocyte-associated antigen)-4 in combination with and without chemotherapy and Her2-targeted therapy in GC. Full article
Show Figures

Figure 1

12 pages, 506 KiB  
Review
Circulating Tumor DNA in Gastric and Gastroesophageal Junction Cancer
by Lisa Paschold and Mascha Binder
Curr. Oncol. 2022, 29(3), 1430-1441; https://doi.org/10.3390/curroncol29030120 - 25 Feb 2022
Cited by 7 | Viewed by 3317
Abstract
Tumor cells shed DNA into the plasma. “Liquid biopsy” analysis of mutations or other genomic alterations in circulating cell-free DNA (cfDNA) may provide us with a tool to detect minimal residual cancer, comprehensively profile the genomic tumor landscape in search of druggable targets, [...] Read more.
Tumor cells shed DNA into the plasma. “Liquid biopsy” analysis of mutations or other genomic alterations in circulating cell-free DNA (cfDNA) may provide us with a tool to detect minimal residual cancer, comprehensively profile the genomic tumor landscape in search of druggable targets, and monitor cancers non-invasively over time for treatment failure or emerging treatment-resistant tumor subclones. While liquid biopsies have not yet entered routine clinical management in patients with gastric and gastroesophageal junction cancers, this group of diseases may benefit from such advanced diagnostic tools due to their pronounced genetic spatiotemporal heterogeneity and limitations in imaging sensitivity. Moreover, as the armamentarium of targeted treatment approaches and immunotherapies expands, cfDNA analyses may reveal their utility not only as a biomarker of response but also for precision monitoring. In this review, we discuss the different applications of cfDNA analyses in patients with gastric and gastroesophageal junction cancer and the technical challenges that such liquid biopsies have yet to overcome. Full article
Show Figures

Figure 1

Other

Jump to: Research, Review

7 pages, 607 KiB  
Commentary
Hereditary Diffuse Gastric Cancer—Update Based on the Current Consort Recommendations
by Christoph Treese, Britta Siegmund and Severin Daum
Curr. Oncol. 2022, 29(4), 2454-2460; https://doi.org/10.3390/curroncol29040199 - 30 Mar 2022
Cited by 3 | Viewed by 2754
Abstract
Hereditary diffuse gastric cancer (HDGC) is an autosomal dominant inherited cancer syndrome that has been associated with a mutation of the CDH1, and rarely the CTNNA1 gene, respectively. HDGC is characterized histologically by multifocal growth and signet ring cells in the gastric [...] Read more.
Hereditary diffuse gastric cancer (HDGC) is an autosomal dominant inherited cancer syndrome that has been associated with a mutation of the CDH1, and rarely the CTNNA1 gene, respectively. HDGC is characterized histologically by multifocal growth and signet ring cells in the gastric mucosa and lobular type breast cancer. In cases of a proven pathogenic CDH1 mutation, a prophylactic gastrectomy, or alternatively, an annual surveillance gastroscopy in expert centers is recommended. Additionally, MR imaging of the breast should be performed annually starting from the age of 30, to detect lobular breast cancer. In 2020, the International Gastric Cancer Linkage Consortium (IGCLC) additionally defined new clinical groups with specific recommendations: (1) the group of patients with a proven mutation in the CDH1 gene, but exclusive manifestation as lobular breast cancer, was defined as hereditary lobular breast cancer (HLBC); (2) the group, which clinically fulfills familial HDGC criteria, in the absence of a relevant mutation, was designated as HDGC-like. This update summarizes relevant aspects of hereditary gastric cancer and the current recommendation criteria of the IGCLC published in 2020. Full article
Show Figures

Figure 1

Back to TopTop