Next Article in Journal
The Flex Robotic System in Head and Neck Surgery: A Review
Next Article in Special Issue
MUC1 Expressions and Its Prognostic Values in US Gastric Cancer Patients
Previous Article in Journal
Managing Malignant Pleural Mesothelioma in the Age of Personalized Medicine: Where Are We and What Is Still Missing?
Previous Article in Special Issue
Cachexia Index as a Prognostic Indicator in Patients with Gastric Cancer: A Retrospective Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Surgery in the Management of Gastric Cancer: State of the Art

1
Digestive Surgery Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
2
Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
3
Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(22), 5542; https://doi.org/10.3390/cancers14225542
Submission received: 27 September 2022 / Revised: 4 November 2022 / Accepted: 9 November 2022 / Published: 11 November 2022
(This article belongs to the Special Issue Management of Gastric Cancer)

Abstract

:

Simple Summary

Gastric cancer remains the sixth most prevalent malignant tumor worldwide and the third leading cause of cancer-related death. Surgery is the pillar of its treatment at all stages, but the importance of endoscopic treatments and multimodal therapy is growing. The aim of this review is to provide a comprehensive description of the role of surgery for gastric cancer in the modern era.

Abstract

Surgery still represents the mainstay of treatment of all stages of gastric cancer (GC). Surgical resections represent potentially curative options in the case of early GC with a low risk of node metastasis. Sentinel lymph node biopsy and indocyanine green fluorescence are novel techniques which may improve the employment of stomach-sparing procedures, ameliorating quality of life without compromising oncological radicality. Nonetheless, the diffusion of these techniques is limited in Western countries. Conversely, radical gastrectomy with extensive lymphadenectomy and multimodal treatment represents a valid option in the case of advanced GC. Differences between Eastern and Western recommendations still exist, and the optimal multimodal strategy is still a matter of investigation. Recent chemotherapy protocols have made surgery available for patients with oligometastatic disease. In this context, intraperitoneal administration of chemotherapy via HIPEC or PIPAC has emerged as an alternative weapon for patients with peritoneal carcinomatosis. In conclusion, the surgical management of GC is still evolving together with the multimodal strategy. It is mandatory for surgeons to be conscious of the current evolution of the surgical management of GC in the era of multidisciplinary and tailored medicine.

1. Introduction

Despite its declining incidence over the last decades, gastric cancer (GC) remains the sixth most prevalent malignant tumor worldwide and the third leading cause of cancer-related death [1]. The most important factors strictly related to this decline are improved refrigeration [2] and effective therapy against Helicobacter pylori [3]. Nevertheless, GC is still burdened with high mortality and its prognosis remains dismal. Indeed, in Western countries, up to two-third of patients are diagnosed with advanced-stage disease (stage III–IV), and even in Eastern countries like Japan, where extensive screening programs exist, this percentage still reaches 50% [1]. Nowadays, a multidisciplinary approach is required for the management of GC. In this context, surgery still represents the mainstay of GC treatment, despite being burdened with significant morbidity (9.1–46%) and mortality (up to 13%), mostly in the case of radical gastrectomy [4]. In the last decades, technological improvements, the employment of innovative chemotherapy regimens, the spread of minimally invasive techniques and multidisciplinary decision-making have paved the way for patient-tailored strategies, with the aim of improving short- and long-term outcomes. The purpose of this narrative review is to provide a comprehensive description of the current surgical management of GC.

2. Early Gastric Cancer

Early gastric cancer (EGC) is defined as a cT1 tumor (i.e., limited to the mucosa or submucosa) independently of lymph node involvement [5]. The management of EGC is determined by the risk of lymph node metastasis, which is very low (1–5%) in the case of cT1aN0 differentiated GC, smaller than 2 cm, without ulceration and lymphovascular invasion [6,7]. Tumors with these features could be suitable for curative endoscopic resection, as reported by Eastern and Western authors [8,9,10]. Conversely, a propensity-matched study by Kamarajah et al. [7] showed that endoscopic resection was inferior to surgery regarding long-term survival for cT1aN0 and cT1bN0 GC, but the treatment selection was based only on the clinical T-stage. For these reasons, the Japanese Gastric Cancer Association (JGCA) [11], the European Society of Medical Oncology (ESMO) [12], the National Comprehensive Cancer Network (NCCN) [13], and the Italian Research Group for Gastric Cancer (GIRCG) [14] all currently recommend endoscopic resection as initial treatment for EGC that meets the aforementioned features. In the case of final pathology reporting poorly differentiated patterns, incomplete resection and/or lymphovascular invasion, the endoscopic resection cannot be considered curative due to the high nodal metastases rate (up to 14%) [15], and radical gastrectomy must be performed [11]. Additionally, surgical resection should be considered for gastric cT1b tumors, as the risk of nodal dissemination is 18–32% in these cases [7,16]. In the case of EGC with clinical evidence of lymph node metastases (cN+), patients should be referred to a multidisciplinary discussion for the definition of the appropriate multimodal therapy.
On the other hand, in recent years sentinel lymph node biopsy followed by function-preserving resection has emerged as viable option for EGC not suitable for endoscopic treatment or when curative endoscopic dissection is not achieved. The procedure of node mapping through submucosal injection of indocyanine green or radioactive colloid is routinely employed for breast cancer and melanoma and represents an innovative tool for GC in selected Eastern centers. A radical gastrectomy is warranted in the case of positive lymph nodes, while a stomach-preserving procedure without additional lymphadenectomy may be considered in the case of negative sentinel node biopsy [17]. Stomach-preserving procedures include gastric wedge resections or segmental gastric resections with gastro-gastric anastomosis [18], the latter being associated with better outcomes in terms of quality of life in Eastern centers [19]. To the best of our knowledge, the only trial addressing the long-term survival of organ-sparing gastric resections is the ongoing SENORITA trial [20].

3. Surgery for Hereditary Diffuse Gastric Cancer

Hereditary diffuse gastric cancer (HDGC) is an autosomal dominant syndrome associated with a mutation of the CDH1 gene. This syndrome leads to GC with a pathologic pattern characterized by the presence of signet ring cells and multifocal growth [21]. Prophylactic gastrectomy in early adulthood is recommended by the International Gastric Cancer Linkage Consortium (IGCLC) as a standard of care in patients with pathogenic CDH1 mutation associated with a family history of diffuse-type GC [22].

4. Non-Metastatic Gastric Cancer

A comprehensive and multimodal approach based on the triad surgery-chemotherapy-radiotherapy is indicated for non-metastatic GC that is more advanced than early GC and in the case of clinical evidence of nodal metastases.

4.1. Extent of Gastric Resection

The extent of surgical resection for GC hinges upon tumor size, location, and histological type. As a result, GC surgery may shift from the resection of the whole stomach including the cardia and pylorus, the so-called total gastrectomy, to subtotal gastrectomies. The latter encompasses different technical procedures depending on the resected anatomical region of the stomach:
Distal gastrectomy: up to two-thirds of the distal stomach and pylorus are resected, while the cardia is preserved;
Proximal gastrectomy: the resection includes the cardia and the upper third of the stomach, preserving the antrum and pylorus;
Pylorus-preserving gastrectomy: resection of the central part of the stomach preserving its upper third, the antro-pyloric region enclosing the infra-pyloric vessels and the hepatic branch of the vagus nerve.
According to the JGCA treatment guidelines, both proximal and pylorus-preserving gastrectomy may be considered as therapeutic options only for cT1N0 tumors [11]. Otherwise, the optimal surgical strategy for localized GC greater than EGC (T2–T4a) or with clinically positive lymph nodes (cN+) requires a subtotal or total gastrectomy with a D2 lymphadenectomy. However, the extent and type of surgical resection (subtotal vs. total gastrectomy) remains a subject of debate. The positive impact of organ-sparing surgery for GC on patients’ quality of life and nutritional status has led to the spread of less-demolitive procedures during the last years [23,24,25,26,27,28]. Furthermore, subtotal gastrectomy for distal GC resulted in similar mortality, overall and disease-free survival but less morbidity, compared to total gastrectomy [29,30,31,32,33,34]. The achievement of a complete resection with negative margins is the main goal of oncological gastric surgery, as it still plays the role of the only potentially curative intervention for GC. Specifically, the concept of R0 refers to a microscopically margin-negative resection with no evidence of macroscopic or microscopic tumor cells in the primary tumor site [35]. Distal gastrectomy is generally the privileged option for distal GC, strictly when an adequate proximal resection margin can be achieved [11,12,13]. Otherwise, total gastrectomy is mandatory. Currently, there is a considerable heterogeneity across Western and Eastern cancer guidelines on the proper distance between GC and resection margins to achieve R0 radical surgery (Table 1).
Interestingly, only the JGCA and the GIRCG guidelines conceive less-extensive gastric resections tailored on the histopathological growth pattern of GC [11,14]. Despite the aforementioned guidelines, several authors highlighted the absence of any significant influence of proximal margin length on GC overall survival [36,37,38,39], unlike others which advocated for a patient-tailored proximal margin length as an independent prognostic factor [40]. Recently, Maspero et al. assessed the impact of JGCA-recommended resection margins on long-term outcomes of 279 consecutive Western patients [41]. Analyzing 220 distal gastrectomies, the authors found that application of JGCA guidelines resulted in improved overall survival and a more organ-preserving surgical strategy, owing to a significant lower number of total gastrectomies compared with NCCN and ESMO guidelines (30% vs. 31% vs. 47%, respectively) without compromising oncological outcomes [41].

4.2. Lymphadenectomy

The extent of lymphadenectomy in radical gastrectomy has long been a source of controversy. The 8th International Union for Cancer Control/American Joint Committee on Cancer (IUCC/AJCC) classification recommends the analysis of at least 16 retrieved nodes for reliable staging [42,43]. Regional lymph nodes for GC were divided into 16 stations according to the JGCA classification [44,45] (Table 2).
Lymphadenectomy for GC is classified as follows:
D1 lymphadenectomy presupposes the removal of the perigastric lymph nodes;
D2 lymphadenectomy entails the resection of perigastric lymph nodes plus those along the left gastric, common hepatic and splenic arteries and the coeliac trunk.
In addition, the JGCA guidelines introduced the concept of D1+ lymphadenectomy as a node dissection more extended than D1 but not fulfilling the criteria of a D2 level [11]. Japanese surgeons considered D1+ lymphadenectomy for T1a tumors not suitable for endoscopic resection and for those of undifferentiated-type or greater than 1.5 cm T1b GC [11]. The NCCN, the ESMO, the JGCA and the GIRCG guidelines currently recommend D2 lymphadenectomy as the gold standard procedure for potentially curable cT2–T4a tumors and for cN+ tumors [11,12,13,14]. Additionally, only the Japanese Guidelines proposed a surgery-related and cancer-tailored technique of D2 lymphadenectomy. Indeed, the removal of distal splenic artery lymph nodes (JGCA node station 11d) is indicated for total gastrectomy, but not for distal gastrectomy, to fulfill the criteria of D2 level [11]. D2 lymphadenectomy, a technically more challenging procedure than D1 lymphadenectomy, represents the standard of care in Eastern countries [11], while it is performed only in experienced high-volume centers and medically fit patients in Western countries [12,13]. Experience from Japanese and Korean studies revealed excellent survivals associated with a lymph node dissection extended to the second D level [46,47,48,49]. Otherwise, several European randomized controlled trials comparing oncological outcomes of D2 versus D1 lymphadenectomy failed to prove any five-year survival superiority related to extensive node dissection [50,51,52,53]. The results of the Dutch Gastric Cancer Group Trial demonstrated a higher postoperative morbidity and mortality in the D2 group and comparable five-year overall survival [52]. Similar results were reported by the Italian Gastric Cancer Study Group (IGCSG) [53] and the Medical Research Council trial [50], despite the high rate of splenectomy, and distal pancreatectomy in D2 gastrectomy in the UK study should be considered as a confounding factor [51]. The 1five-year follow-up version of the Dutch trial showed a statistically significant difference in term of GC-related death (37% in D2 group vs. 48% in D1 group) and locoregional recurrence (12% in D2 group vs. 22% in D1 group) and even a not significantly higher overall survival (29% in D2 group vs. 21% in D1 group; p = 0.34) [54]. Moreover, after the exclusion of the subgroup of pancreatectomies and splenectomies, the 15-year overall survival increased to 32% in the D2 group (vs. 22% in D1 group; p = 0.006) [54]. Recently, the updated IGCSG experience also highlighted a significative improvement of disease-specific survival (51.4% in D2 group vs. 29.4% in D1 group) and GC-related mortality (43% in D2 group vs. 65% in D1 group) after a 15-year period in patients with advanced GC (pT > 1) and lymph node metastases who underwent D2 lymphadenectomy [55]. In conclusion, D2 lymphadenectomy is currently indicated in advanced GC [11,12,13] but in Western countries it should be performed only in selected patients by experienced surgeons in referral centers [12,13].

4.3. Minimally Invasive Procedures

The feasibility and safety of minimally invasive gastric surgery was first reported by Kitano et al. in 1995 [56]. Since then, laparoscopic gastrectomy has arisen as a non-inferior technique to conventional open gastrectomy [57,58] and its effectiveness has been analyzed by several randomized clinical trials (Table 3).
According to the results of a recent systematic review and meta-analysis, laparoscopic distal gastrectomy had less perioperative blood loss, fewer postoperative complication, faster recovery of bowel function, equivalent oncological surgical precision (defined by lymph node yield, resection margins and anastomotic leakage) compared with open surgery, at the cost of longer operative time [85]. Nevertheless, the technical complexity of laparoscopic gastric surgery is reflected by its steep learning curve, requiring 20 to 40 cases and up to 100 cases for laparoscopic distal and total gastrectomy, respectively, to achieve proficiency [86,87].
During the last years, robotic surgery has emerged as a new minimally invasive technique for oncological gastric surgery. Indeed, the robotic platform has overcome the limitations of laparoscopy through the introduction of several advantages, such as the wider range of motions and surgical dexterity, 3D high-definition view, tremor reduction, better ergonomics and a faster learning curve [88]. Despite higher costs and longer operative time than laparoscopy, robotic gastrectomy leads to similar or slightly improved complication rates and lymph node harvest [89,90,91,92,93], and comparable three-year overall and relapse-free survival [94], resulting in advantages also for patients with visceral fat obesity [95]. The benefits derived from the robotic platform were more conspicuous when considering only robotic distal gastrectomy (RDG) [96]. Recently, the safety and efficacy of minimally invasive distal gastrectomy were evaluated in a randomized controlled trial comparing robotic versus laparoscopy [97]. The robotic approach was then associated with faster recovery, lower postoperative complications and inflammatory response, higher dissection rates of extra/perigastric lymph nodes and shorter delay of adjuvant chemotherapy [97]. The higher effectiveness and safety of RDG in terms of surgical and oncological outcomes was ultimately confirmed by the Gong et al. meta-analysis [98].

4.4. Multimodal Treatment

All patients with a clinical T2 or more advanced disease (i.e., clinically positive lymph nodes) should be discussed in a multidisciplinary context to define the appropriate multimodal treatment [99]. Providing a comprehensive description of the pharmacological treatment of GC is far beyond the purpose of this review. However, given the positive impact of multimodal strategies in the treatment of locally advanced GC becoming clearer over time, it is mandatory to describe the efforts to improve the results obtained with surgery alone, including neoadjuvant and adjuvant strategies.

4.5. Neoadjuvant/Perioperative Therapy

According to the latest version of NCCN Guidelines [13], two main strategies are recommended as alternatives to upfront surgery for cT2 or higher locoregional disease, namely perioperative chemotherapy and preoperative chemoradiation. These recommendations derive from the results of several clinical trials which have directly compared surgery alone with neoadjuvant or perioperative chemotherapy, demonstrating a survival benefit for the multimodal approaches. Initially, the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) trial [100] randomly assigned 503 patients with potentially resectable gastric, distal esophageal or esophagogastric junction adenocarcinomas to upfront surgery or preoperative (three neoadjuvant plus three adjuvant cycles) chemotherapy with epirubicin, cisplatin and infusional fluorouracile (ECF), showing better overall and progression-free survival for patients with locoregional disease (cT2 or higher or cN > 0) who received perioperative chemotherapy. Notably, only 42% of these patients were able to complete the whole protocol treatment. Subsequently, the phase II/III FLOT4-AIO (Fluorouracil, Leucovorin, Oxaliplatin, and Docetaxel 4—Arbeitsgemeinschaft Internistische Onkologie) trial [101] demonstrated a better overall survival and three-year overall survival for patients with gastric or gastroesophageal cancer who received FLOT regimen (four preoperative and four post-operative Fluorouracil, Leucovorin, Oxaliplatin, and Docetaxel cycles) as compared to patients who received epirubicin-based therapy (ECF or ECX—epirubicin, cisplatin and capecitabine). These results made FLOT regimen the first choice for patients with excellent performance status. However, due to the considerable toxicity associated with FLOT, ECF is still recommended in selected patients with good performance status. Finally, for patients who have good to moderate performance status, the suggested perioperative regimen is fluorouracil and oxaliplatin (FOLFOX) or capecitabine and oxaliplatin (CAPOX) [13]. Ongoing trials are testing the addition of immune checkpoint inhibitors to the FLOT regimen for mismatch repair-deficient tumors that express programmed death-ligand 1, such as pembrolizumab in the KEYNOTE-585 trial [102], etezolizumab in the DANTE study [103], and durvalumab [104]. Moreover, ongoing studies are investigating the addition of trastuzumab to a current chemotherapy regimen in a perioperative setting for patients with ERBB2 (HER2 or HER2/neu)–positive tumors [105]. With regards to the use of chemoradiotherapy in the neoadjuvant setting, several small studies have demonstrated the possibility to produce a pathologic response in resectable GC [106,107,108,109]. Nonetheless, its clinical employment is still questionable and under investigation due to the lack of randomized controlled trials demonstrating a survival benefit in noncardia GC. Indeed, the only available trials analyzed esophageal, gastroesophageal junction (GEJ) and gastric cardia tumors only [110,111]. The answer to whether preoperative chemoradiotherapy is better than upfront surgery alone, neoadjuvant chemotherapy or adjuvant therapy after surgery will probably come from several ongoing trials, such as the Chemoradiotherapy after Induction Chemotherapy in Cancer of the Stomach II (CRITICS-II) [112], the ESOPEC trial [113] and the Trial of Preoperative Therapy for Gastric and Esophagogastric Junction Adenocarcinoma (TOPGEAR) trial [114].

4.6. Adjuvant Therapy

For patients who have undergone upfront gastric resection without receiving any neoadjuvant therapy, the current NCCN guidelines recommend adjuvant treatment rather than surgery alone in patients with pT3-4 and/or N + GC [13]. Options for adjuvant therapy include chemotherapy alone and chemoradiotherapy plus chemotherapy. In particular, postoperative chemotherapy with capecitabine and oxaliplatin or fluorouracil and oxaliplatin is indicated for patients who have undergone gastric resection with D2 lymph node dissection; postoperative chemoradiation plus chemotherapy (fluoropyrimidine before and after fluoropyrimidine-based chemoradiation) is indicated for patients who received less than D2 lymph node dissection; chemoradiotherapy alone for patients who received R1-R2 gastrectomy. Finally, for selected patients with high-risk features (including poorly differentiated or higher-grade cancer; lymphovascular invasion; neural invasion; <50 years of age; lymph node dissection less than D2), the NCCN guidelines recommend chemoradiation plus chemotherapy even in the case of pT2, N0 disease. With regards to chemotherapy, the optimal regime has not been established yet: data deriving from several phase III trials indicate CAPOX, FOLFOX and S-1 with or without docetaxel as the main alternatives. These indications derive from the results of several key randomized clinical trials, shown in Table 4.
Interestingly, a retrospective study based on data from the National Cancer Database [126] found a benefit for chemoradiotherapy with regard to five-year overall survival in patients with positive lymph nodes and lymphovascular invasion, but not in patients without lymphovascular invasion, suggesting the need for tailored approaches for selected groups of patients.

5. Metastatic Gastric Cancer

While chemotherapy still represents the mainstay of treatment for patients with metastatic gastric cancer, recent advances in systemic therapy are expanding the surgical indications even to patients with stage IV gastric cancer, leading to a more tailored approach.

5.1. Resectable Metastatic Disease

Several small retrospective studies analyzing the outcome of surgery in patients with limited metastatic disease have shown a possible survival benefit [127,128,129,130]. However, they are often characterized by an important selection bias, as the patients undergoing surgery often have better prognosis and a smaller disease burden than those who received gastrojejunostomy or no surgery at all. Moreover, many factors that influence survival, such as systemic chemotherapy, are not always considered. Resection of hepatic metastasis has been rarely performed in patients with isolated gastric cancer liver secondary lesions [131,132], and long-term survival in these patients represents a rare event [131,132,133,134,135]. For these reasons, no consensus regarding appropriate patient selection criteria currently exists. Pulmonary metastasectomy may also be taken into consideration for highly selected patients, potentially leading to long-term survival, but data are still insufficient [136,137,138,139]. However, encouraging results regarding potential long-term survival in selected patients with M+ gastric cancer after modern chemotherapy schemes followed by extensive surgery come from new and ongoing trials. The Arbeitsgemeinschaft Internistische Onkologie—Fluorouracil, Leucovorin, Oxaliplatin, and Docetaxel 3 (AIO-FLOT3) phase II trial [140] investigated the outcomes of perioperative FLOT plus surgery in limited metastatic disease (one metastatic organ site with or without retroperitoneal nodes) suitable for R0 and macroscopically complete (R1) resection of the primary tumor and metastatic lesions at restaging, respectively. The authors found a higher median overall survival in patients who underwent surgery than in patients who only received FLOT (31 vs. 16 months, respectively). The results of the follow-up RENAISSANCE (AIO-FLOT5) phase III trial [141], which is currently randomizing patients without progression after four cycles of FLOT to additional FLOT or surgery followed by postoperative FLOT, are awaited. Similar results were obtained from a large multicenter retrospective cohort study, namely the Conversion Therapy for Stage IV Gastric Cancer 1 (CONVO-GC-1) trial [142]. The authors included 1206 patients who received chemotherapy followed by radical surgical resection, reporting 56.6 months (95% CI, 46.4–74.5) of median overall survival for patients who underwent R0 resection, as compared to 36.7 months (95% CI, 34.4–40.0) for all patients.

5.2. Peritoneal Disease

After liver, the peritoneum represents the second most common site of gastric cancer metastatic spread [143].

5.2.1. Hyperthermic Intraperitoneal Chemotherapy (HIPEC)

For patients with isolated peritoneal metastasis, the role of cytoreductive surgery (CRS) with or without hyperthermic intraperitoneal chemotherapy (HIPEC; normally employing mitomycin C at 40 °C to 43 °C) still remains controversial and is not unanimously considered as the standard approach [144,145,146,147]. Indeed, trials comparing CRS with or without HIPEC and systemic chemotherapy are still lacking, and protocols vary largely by institution. Currently, the reported median overall survival after CRS plus HIPEC ranges from 11 to 19 months, and one of the main prognostic factors is represented by the quality and completeness of surgical cytoreduction [144,145]. The main evidence of improved survival derives from a phase III trial from China [145] and the CYTO-CHIP French study [144]. In particular, the latter analyzed data from nineteen French cancer treatment centers, investigating the impact of CRS alone as compared to CRS plus HIPEC in selected patients with isolated peritoneal metastases from gastric cancer. The authors found better median survival in the group of patients receiving HIPEC after CRS (19 vs. 12 months) and better five-year recurrence-free survival as well (17% vs. 4%). While demonstrating statistical significance, these results should not be overinterpreted, as less than one patient per center per year was recruited, which in fact represents a very highly selected population. Moreover, a significant percentage of patients had only positive cytology. With regard to patients with occult or minimal-volume peritoneal disease from GC (i.e., positive cytology only or radiographically occult peritoneal disease), occasional long-term survival has been reported in the case of curative-intent surgery following negative cytology achieved after neoadjuvant chemotherapy [148,149]. Moreover, a small phase II study from the US-tested HIPEC following systemic chemotherapy in 19 patients diagnosed with minimal peritoneal carcinomatosis showed 30.2 months of median overall survival [150]. Remarkably, some studies have shown that surgery itself can become a vehicle of iatrogenic peritoneal dissemination, especially through division of lymphatic channels in patients with high T and N disease [151,152]. In this context, the ongoing PILGRIM HIPEC-01 trial is currently testing this hypothesis, randomizing patients with T3–T4 Nx M0 gastric cancer undergoing curative-intent surgery to adjuvant chemotherapy or adjuvant HIPEC followed by adjuvant chemotherapy. Despite preliminary results showing a favorable safety profile for HIPEC, definitive results data are still awaited [153].

5.2.2. Pressurized Intraperitoneal Chemotherapy (PIPAC)

In the setting of unresectable peritoneal metastasis and malignant ascites from GC, a novel treatment using aerosolized chemotherapy, namely pressurized intraperitoneal chemotherapy (PIPAC), has been increasingly used in the last years with the goal of increasing patients’ survival. Reymond et al. first reported the significant impact of PIPAC on tumor response with a 25% complete pathological response [154]. Alyami et al. tested the outcomes of cisplatin and doxorubicin via PIPAC in 42 patients with unresectable peritoneal disease, showing a median overall survival of 19.1 months and only 6.1% morbidity [155]. Another study by Di Giorgio and colleagues reported a pathological response in 61.5% of 28 consecutive patients, with a median overall survival of 12.3 months for the entire cohort. Interestingly, the authors reported an overall survival of 15 months in the subgroup of patients undergoing more than one PIPAC procedure [156]. Several other studies have shown the safety and the effectiveness of PIPAC with cisplatin (7.5 mg/m2) and doxorubicin (1.5 mg/m2) in patients with unresectable peritoneal metastasis from GC [155,157,158]. However, definitive data regarding the safety and efficacy of PIPAC, as well as the optimal drugs and dose to be used, are still awaited. The PIPAC EstoK 01 [159] is an ongoing prospective randomized multicenter phase II trial assigning patients with peritoneal dissemination from gastric cancer to three cycles of PIPAC with oxaliplatin plus systemic intravenous chemotherapy or systemic chemotherapy alone. Two ongoing studies concerning the use of oxaliplatin PIPAC are aiming to find its optimal dose [144,160]. Finally, the PIPAC-GA01 trial is currently evaluating the safety and efficacy of PIPAC with three single doses of doxorubicin and cisplatin in patients with recurrent gastric cancer [161].

6. Palliation

The majority of patients diagnosed with GC will require palliative treatment due to the dismal prognosis of the disease [162]. While chemotherapy represents the most effective treatment for patients with metastatic disease, palliation of symptoms such as obstruction, bleeding or perforation may require multidisciplinary management employing surgery, endoscopy, radiotherapy, or other approaches.

6.1. Surgical Palliation

For patients diagnosed with advanced GC, palliative gastrectomy plays a role in rapidly relieving symptoms such as obstruction, perforation, bleeding, pain, and nausea, but did not show any survival benefit. Indeed, the Japan Clinical Oncology Group 0705 and Korean Gastric Cancer Association 01 (REGATTA) phase III trial [163] tested the survival benefit of gastrectomy in 175 patients with advanced GC and a single non-curable factor (such as peritoneum, liver or para-aortic metastasis), who were randomly assigned to S-1 plus cisplatin chemotherapy alone or to gastrectomy followed by the same systemic treatment. The study did not show any improvement in terms of overall survival, while increasing the incidence of serious adverse events related to chemotherapy. Palliative gastrojejunostomy also plays a role in symptom relief, especially when other nonsurgical methods cannot be used. Moreover, when performed laparoscopically, it can lead to less blood loss and shorter length of hospital stay than an open approach, as shown by several case reports and small studies [164]. Among surgical options, gastrectomy and gastrojejunostomy have not yet shown a significant difference in outcomes according to available data [165].

6.2. Non-Surgical Palliation

External beam radiation therapy plays a role in the control of obstruction, pain and especially bleeding in patients with unresectable gastric cancer [165,166,167,168,169,170,171,172], with a good tolerance of the treatment and a low rate of toxicity. While no controlled studies comparing radiotherapy with endoscopic or surgical treatments for symptomatic palliation currently exist, it is evident that the late onset of the effect of radiotherapy makes it not the ideal strategy for all the symptoms. In particular, in the case of obstruction, endoscopic stent placement represents a faster and less invasive alternative to surgery and radiotherapy. While not being inferior to palliative gastrojejunostomy in terms of efficacy and complications, it seems to lead to faster relief of obstructive symptoms and shorter length of hospital stay. However, it remains particularly indicated for patients with short life expectancy, due to a higher reintervention rate and a lower durability as compared to surgical palliation [173]. Additionally, endoscopy has a role in the relief of dysphagia through endoscopic laser treatment (especially in patients with GEJ tumors) [174,175,176], and in the treatment of hemorrhage through laser photocoagulation (in particular for large diffusely bleeding tumors) [177,178], argon plasma coagulation and application of hemostatic nanopowder [179].

7. Conclusions

Surgery still represents the mainstay of treatment for GC and its effectiveness has been enhanced by the introduction of novel minimally invasive surgical techniques and systemic therapies. The Eastern experience in the management of EGC underlined the importance of preserving quality of life without jeopardizing oncologic radicality, through the employment of endoscopic resection, sentinel lymph node biopsy and organ-sparing procedures. On the other side, more advanced non-metastatic GCs require a comprehensive and multimodal approach based on the surgery-chemotherapy-radiotherapy triad. Nevertheless, the lack of a global consensus on the optimal surgical strategy for GC has led to many discrepancies between Western and Eastern guidelines, mainly regarding the extent of surgical resection and lymphadenectomy. For this reason, a lymphadenectomy extended to the D2 level represents the standard of care in Eastern centers, while it is performed only in high-volume Western institutions for medically fit patients. In recent years, the spread of novel chemotherapy delivery topical systems, such as HIPEC and PIPAC, has changed the paradigm of end-stage GC with peritoneal dissemination. Similarly, the surgical management of metastatic GC disease has been allowed by the improvement of chemoradiotherapy regimens. Finally, the molecular classification of GC has recently been published, paving the way for new perspectives of tailored treatment options [180,181,182]. However, to date, its surgical implications are only speculative [183] and the integration in clinical practice is still awaited.

Author Contributions

Conceptualization, F.R. and S.A.; methodology, F.R., C.A.S. and V.L.; resources, F.R., C.A.S., V.L., G.Q., C.F., A.S., C.P. and V.P.; writing—original draft preparation, F.R., C.A.S. and V.L.; writing—review and editing, F.R., C.A.S. and V.L.; visualization, F.R., C.A.S., V.L., G.Q., C.F., A.S., C.P. and V.P.; supervision, S.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Coggon, D.; Barker, D.J.; Cole, R.B.; Nelson, M. Stomach cancer and food storage. J. Natl. Cancer Inst. 1989, 81, 1178–1182. [Google Scholar] [CrossRef] [PubMed]
  3. Parsonnet, J.; Vandersteen, D.; Goates, J.; Sibley, R.K.; Pritikin, J.; Chang, Y. Helicobacter pylori infection in intestinal- and diffuse-type gastric adenocarcinomas. J. Natl. Cancer Inst. 1991, 83, 640–643. [Google Scholar] [CrossRef]
  4. Papenfuss, W.A.; Kukar, M.; Oxenberg, J.; Attwood, K.; Nurkin, S.; Malhotra, U.; Wilkinson, N.W. Morbidity and mortality associated with gastrectomy for gastric cancer. Ann. Surg. Oncol. 2014, 21, 3008–3014. [Google Scholar] [CrossRef] [PubMed]
  5. Van Cutsem, E.; Sagaert, X.; Topal, B.; Haustermans, K.; Prenen, H. Gastric cancer. Lancet 2016, 388, 2654–2664. [Google Scholar] [CrossRef]
  6. Abdelfatah, M.M.; Barakat, M.; Lee, H.; Kim, J.J.; Uedo, N.; Grimm, I.; Othman, M.O. The incidence of lymph node metastasis in early gastric cancer according to the expanded criteria in comparison with the absolute criteria of the Japanese Gastric Cancer Association: A systematic review of the literature and meta-analysis. Gastrointest. Endosc. 2018, 87, 338–347. [Google Scholar] [CrossRef]
  7. Kamarajah, S.K.; Markar, S.R.; Phillips, A.W.; Salti, G.I.; Dahdaleh, F.S. Local Endoscopic Resection is Inferior to Gastrectomy for Early Clinical Stage T1a and T1b Gastric Adenocarcinoma: A Propensity-Matched Study. Ann. Surg. Oncol. 2021, 28, 2992–2998. [Google Scholar] [CrossRef] [PubMed]
  8. Manner, H.; Rabenstein, T.; May, A.; Pech, O.; Gossner, L.; Werk, D.; Manner, N.; Gunter, E.; Pohl, J.; Vieth, M.; et al. Long-term results of endoscopic resection in early gastric cancer: The Western experience. Am. J. Gastroenterol. 2009, 104, 566–573. [Google Scholar] [CrossRef] [PubMed]
  9. Uedo, N.; Iishi, H.; Tatsuta, M.; Ishihara, R.; Higashino, K.; Takeuchi, Y.; Imanaka, K.; Yamada, T.; Yamamoto, S.; Yamamoto, S.; et al. Longterm outcomes after endoscopic mucosal resection for early gastric cancer. Gastric Cancer 2006, 9, 88–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Quero, G.; Fiorillo, C.; Longo, F.; Laterza, V.; Rosa, F.; Cina, C.; Menghi, R.; Tortorelli, A.P.; Barbaro, F.; Pecere, S.; et al. Propensity score-matched comparison of short- and long-term outcomes between surgery and endoscopic submucosal dissection (ESD) for intestinal type early gastric cancer (EGC) of the middle and lower third of the stomach: A European tertiary referral center experience. Surg. Endosc. 2021, 35, 2592–2600. [Google Scholar] [CrossRef]
  11. Japanese Gastric Cancer Association. Japanese gastric cancer treatment guidelines 2018 (5th edition). Gastric Cancer 2021, 24, 1–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Smyth, E.C.; Verheij, M.; Allum, W.; Cunningham, D.; Cervantes, A.; Arnold, D.; Committee, E.G. Gastric cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2016, 27, v38–v49. [Google Scholar] [CrossRef] [PubMed]
  13. Ajani, J.A.; D’Amico, T.A.; Bentrem, D.J.; Chao, J.; Cooke, D.; Corvera, C.; Das, P.; Enzinger, P.C.; Enzler, T.; Fanta, P.; et al. Gastric Cancer, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2022, 20, 167–192. [Google Scholar] [CrossRef] [PubMed]
  14. De Manzoni, G.; Marrelli, D.; Baiocchi, G.L.; Morgagni, P.; Saragoni, L.; Degiuli, M.; Donini, A.; Fumagalli, U.; Mazzei, M.A.; Pacelli, F.; et al. The Italian Research Group for Gastric Cancer (GIRCG) guidelines for gastric cancer staging and treatment: 2015. Gastric Cancer 2017, 20, 20–30. [Google Scholar] [CrossRef] [Green Version]
  15. Jung, D.H.; Huh, C.W.; Kim, J.H.; Hong, J.H.; Park, J.C.; Lee, Y.C.; Youn, Y.H.; Park, H.; Choi, S.H.; Noh, S.H. Risk-Stratification Model Based on Lymph Node Metastasis After Noncurative Endoscopic Resection for Early Gastric Cancer. Ann. Surg. Oncol. 2017, 24, 1643–1649. [Google Scholar] [CrossRef]
  16. Bausys, R.; Bausys, A.; Vysniauskaite, I.; Maneikis, K.; Klimas, D.; Luksta, M.; Strupas, K.; Stratilatovas, E. Risk factors for lymph node metastasis in early gastric cancer patients: Report from Eastern Europe country- Lithuania. BMC Surg. 2017, 17, 108. [Google Scholar] [CrossRef]
  17. Takeuchi, H.; Kitagawa, Y. Sentinel lymph node biopsy in gastric cancer. Cancer J. 2015, 21, 21–24. [Google Scholar] [CrossRef]
  18. Furukawa, H.; Hiratsuka, M.; Imaoka, S.; Ishikawa, O.; Kabuto, T.; Sasaki, Y.; Kameyama, M.; Ohigashi, H.; Nakano, H.; Yasuda, T.; et al. Phase II study of limited surgery for early gastric cancer: Segmental gastric resection. Ann. Surg. Oncol. 1999, 6, 166–170. [Google Scholar] [CrossRef]
  19. Isozaki, H.; Matsumoto, S.; Murakami, S.; Takama, T.; Sho, T.; Ishihara, K.; Sakai, K.; Takeda, M.; Nakada, K.; Fujiwara, T. Diminished Gastric Resection Preserves Better Quality of Life in Patients with Early Gastric Cancer. Acta Med. Okayama 2016, 70, 119–130. [Google Scholar] [CrossRef] [PubMed]
  20. Park, J.Y.; Kim, Y.W.; Ryu, K.W.; Nam, B.H.; Lee, Y.J.; Jeong, S.H.; Park, J.H.; Hur, H.; Han, S.U.; Min, J.S.; et al. Assessment of laparoscopic stomach preserving surgery with sentinel basin dissection versus standard gastrectomy with lymphadenectomy in early gastric cancer-A multicenter randomized phase III clinical trial (SENORITA trial) protocol. BMC Cancer 2016, 16, 340. [Google Scholar] [CrossRef]
  21. Treese, C.; Siegmund, B.; Daum, S. Hereditary Diffuse Gastric Cancer-Update Based on the Current Consort Recommendations. Curr. Oncol. 2022, 29, 2454–2460. [Google Scholar] [CrossRef] [PubMed]
  22. Blair, V.R.; McLeod, M.; Carneiro, F.; Coit, D.G.; D’Addario, J.L.; van Dieren, J.M.; Harris, K.L.; Hoogerbrugge, N.; Oliveira, C.; van der Post, R.S.; et al. Hereditary diffuse gastric cancer: Updated clinical practice guidelines. Lancet Oncol. 2020, 21, e386–e397. [Google Scholar] [CrossRef]
  23. Brenkman, H.J.F.; Tegels, J.J.W.; Ruurda, J.P.; Luyer, M.D.P.; Kouwenhoven, E.A.; Draaisma, W.A.; van der Peet, D.L.; Wijnhoven, B.P.L.; Stoot, J.; van Hillegersberg, R.; et al. Factors influencing health-related quality of life after gastrectomy for cancer. Gastric Cancer 2018, 21, 524–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Rosania, R.; Chiapponi, C.; Malfertheiner, P.; Venerito, M. Nutrition in Patients with Gastric Cancer: An Update. Gastrointest. Tumors 2016, 2, 178–187. [Google Scholar] [CrossRef] [PubMed]
  25. Yamamoto, M.; Omori, T.; Shinno, N.; Hara, H.; Fujii, Y.; Mukai, Y.; Sugase, T.; Takeoka, T.; Asukai, K.; Kanemura, T.; et al. Laparoscopic Proximal Gastrectomy with Novel Valvuloplastic Esophagogastrostomy vs. Laparoscopic Total Gastrectomy for Stage I Gastric Cancer: A Propensity Score Matching Analysis. J. Gastrointest. Surg. 2022, 26, 2041–2049. [Google Scholar] [CrossRef]
  26. Park, S.; Chung, H.Y.; Lee, S.S.; Kwon, O.; Yu, W. Serial comparisons of quality of life after distal subtotal or total gastrectomy: What are the rational approaches for quality of life management? J. Gastric Cancer 2014, 14, 32–38. [Google Scholar] [CrossRef] [Green Version]
  27. Goh, Y.M.; Gillespie, C.; Couper, G.; Paterson-Brown, S. Quality of life after total and subtotal gastrectomy for gastric carcinoma. Surgeon 2015, 13, 267–270. [Google Scholar] [CrossRef]
  28. Kwon, O.K.; Yu, B.; Park, K.B.; Park, J.Y.; Lee, S.S.; Chung, H.Y. Advantages of Distal Subtotal Gastrectomy over Total Gastrectomy in the Quality of Life of Long-Term Gastric Cancer Survivors. J. Gastric Cancer 2020, 20, 176–189. [Google Scholar] [CrossRef]
  29. Qi, J.; Zhang, P.; Wang, Y.; Chen, H.; Li, Y. Does Total Gastrectomy Provide Better Outcomes than Distal Subtotal Gastrectomy for Distal Gastric Cancer? A Systematic Review and Meta-Analysis. PLoS ONE 2016, 11, e0165179. [Google Scholar] [CrossRef]
  30. Gouzi, J.L.; Huguier, M.; Fagniez, P.L.; Launois, B.; Flamant, Y.; Lacaine, F.; Paquet, J.C.; Hay, J.M. Total versus subtotal gastrectomy for adenocarcinoma of the gastric antrum. A French prospective controlled study. Ann. Surg. 1989, 209, 162–166. [Google Scholar] [CrossRef]
  31. Robertson, C.S.; Chung, S.C.; Woods, S.D.; Griffin, S.M.; Raimes, S.A.; Lau, J.T.; Li, A.K. A prospective randomized trial comparing R1 subtotal gastrectomy with R3 total gastrectomy for antral cancer. Ann. Surg. 1994, 220, 176–182. [Google Scholar] [CrossRef] [PubMed]
  32. Bozzetti, F.; Marubini, E.; Bonfanti, G.; Miceli, R.; Piano, C.; Crose, N.; Gennari, L. Total versus subtotal gastrectomy: Surgical morbidity and mortality rates in a multicenter Italian randomized trial. The Italian Gastrointestinal Tumor Study Group. Ann. Surg. 1997, 226, 613–620. [Google Scholar] [CrossRef] [PubMed]
  33. De Manzoni, G.; Verlato, G.; Roviello, F.; Di Leo, A.; Marrelli, D.; Morgagni, P.; Pasini, F.; Saragoni, L.; Tomezzoli, A.; Italian Research Group for Gastric Cancer (IRGGC). Subtotal versus total gastrectomy for T3 adenocarcinoma of the antrum. Gastric Cancer 2003, 6, 237–242. [Google Scholar] [CrossRef] [PubMed]
  34. Bozzetti, F.; Marubini, E.; Bonfanti, G.; Miceli, R.; Piano, C.; Gennari, L. Subtotal versus total gastrectomy for gastric cancer: Five-year survival rates in a multicenter randomized Italian trial. Italian Gastrointestinal Tumor Study Group. Ann. Surg. 1999, 230, 170–178. [Google Scholar] [CrossRef]
  35. Hermanek, P.; Wittekind, C. Residual tumor (R) classification and prognosis. Semin. Surg. Oncol. 1994, 10, 12–20. [Google Scholar] [CrossRef]
  36. Postlewait, L.M.; Squires, M.H., 3rd; Kooby, D.A.; Poultsides, G.A.; Weber, S.M.; Bloomston, M.; Fields, R.C.; Pawlik, T.M.; Votanopoulos, K.I.; Schmidt, C.R.; et al. The importance of the proximal resection margin distance for proximal gastric adenocarcinoma: A multi-institutional study of the US Gastric Cancer Collaborative. J. Surg. Oncol. 2015, 112, 203–207. [Google Scholar] [CrossRef]
  37. Lee, C.M.; Jee, Y.S.; Lee, J.H.; Son, S.Y.; Ahn, S.H.; Park, D.J.; Kim, H.H. Length of negative resection margin does not affect local recurrence and survival in the patients with gastric cancer. World J. Gastroenterol. 2014, 20, 10518–10524. [Google Scholar] [CrossRef]
  38. Kim, A.; Kim, B.S.; Yook, J.H.; Kim, B.S. Optimal proximal resection margin distance for gastrectomy in advanced gastric cancer. World J. Gastroenterol. 2020, 26, 2232–2246. [Google Scholar] [CrossRef]
  39. Berlth, F.; Kim, W.H.; Choi, J.H.; Park, S.H.; Kong, S.H.; Lee, H.J.; Yang, H.K. Prognostic Impact of Frozen Section Investigation and Extent of Proximal Safety Margin in Gastric Cancer Resection. Ann. Surg. 2020, 272, 871–878. [Google Scholar] [CrossRef]
  40. Wang, J.; Liu, J.; Zhang, G.; Kong, D. Individualized proximal margin correlates with outcomes in gastric cancers with radical gastrectomy. Tumour Biol. 2017, 39, 1010428317711032. [Google Scholar] [CrossRef]
  41. Maspero, M.; Sposito, C.; Benedetti, A.; Virdis, M.; Di Bartolomeo, M.; Milione, M.; Mazzaferro, V. Impact of Surgical Margins on Overall Survival after Gastrectomy for Gastric Cancer: A Validation of Japanese Gastric Cancer Association Guidelines on a Western Series. Ann. Surg. Oncol. 2022, 29, 3096–3108. [Google Scholar] [CrossRef] [PubMed]
  42. Amin, M.B.; Greene, F.L.; Edge, S.B.; Compton, C.C.; Gershenwald, J.E.; Brookland, R.K.; Meyer, L.; Gress, D.M.; Byrd, D.R.; Winchester, D.P. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J. Clin. 2017, 67, 93–99. [Google Scholar] [CrossRef] [PubMed]
  43. Lu, J.; Zheng, C.H.; Cao, L.L.; Li, P.; Xie, J.W.; Wang, J.B.; Lin, J.X.; Chen, Q.Y.; Lin, M.; Huang, C.M. The effectiveness of the 8th American Joint Committee on Cancer TNM classification in the prognosis evaluation of gastric cancer patients: A comparative study between the 7th and 8th editions. Eur. J. Surg. Oncol. 2017, 43, 2349–2356. [Google Scholar] [CrossRef] [PubMed]
  44. Japanese Gastric Cancer Association. Japanese classification of gastric carcinoma: 3rd English edition. Gastric Cancer 2011, 14, 101–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Rosa, F.; Costamagna, G.; Doglietto, G.B.; Alfieri, S. Classification of nodal stations in gastric cancer. Transl. Gastroenterol. Hepatol. 2017, 2, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Katai, H.; Ishikawa, T.; Akazawa, K.; Isobe, Y.; Miyashiro, I.; Oda, I.; Tsujitani, S.; Ono, H.; Tanabe, S.; Fukagawa, T.; et al. Five-year survival analysis of surgically resected gastric cancer cases in Japan: A retrospective analysis of more than 100,000 patients from the nationwide registry of the Japanese Gastric Cancer Association (2001–2007). Gastric Cancer 2018, 21, 144–154. [Google Scholar] [CrossRef]
  47. Wu, C.W.; Hsiung, C.A.; Lo, S.S.; Hsieh, M.C.; Chen, J.H.; Li, A.F.; Lui, W.Y.; Whang-Peng, J. Nodal dissection for patients with gastric cancer: A randomised controlled trial. Lancet Oncol. 2006, 7, 309–315. [Google Scholar] [CrossRef]
  48. Kim, E.Y.; Song, K.Y.; Lee, J. Does Hospital Volume Really Affect the Surgical and Oncological Outcomes of Gastric Cancer in Korea? J. Gastric Cancer 2017, 17, 246–254. [Google Scholar] [CrossRef] [Green Version]
  49. Lee, J.H.; Kim, J.G.; Jung, H.K.; Kim, J.H.; Jeong, W.K.; Jeon, T.J.; Kim, J.M.; Kim, Y.I.; Ryu, K.W.; Kong, S.H.; et al. Clinical practice guidelines for gastric cancer in Korea: An evidence-based approach. J. Gastric Cancer 2014, 14, 87–104. [Google Scholar] [CrossRef] [Green Version]
  50. Cuschieri, A.; Fayers, P.; Fielding, J.; Craven, J.; Bancewicz, J.; Joypaul, V.; Cook, P. Postoperative morbidity and mortality after D1 and D2 resections for gastric cancer: Preliminary results of the MRC randomised controlled surgical trial. The Surgical Cooperative Group. Lancet 1996, 347, 995–999. [Google Scholar] [CrossRef]
  51. Cuschieri, A.; Weeden, S.; Fielding, J.; Bancewicz, J.; Craven, J.; Joypaul, V.; Sydes, M.; Fayers, P. Patient survival after D1 and D2 resections for gastric cancer: Long-term results of the MRC randomized surgical trial. Surgical Co-operative Group. Br. J. Cancer 1999, 79, 1522–1530. [Google Scholar] [CrossRef] [PubMed]
  52. Bonenkamp, J.J.; Hermans, J.; Sasako, M.; van de Velde, C.J.; Welvaart, K.; Songun, I.; Meyer, S.; Plukker, J.T.; Van Elk, P.; Obertop, H.; et al. Extended lymph-node dissection for gastric cancer. N. Engl. J. Med. 1999, 340, 908–914. [Google Scholar] [CrossRef] [PubMed]
  53. Degiuli, M.; Sasako, M.; Ponti, A.; Vendrame, A.; Tomatis, M.; Mazza, C.; Borasi, A.; Capussotti, L.; Fronda, G.; Morino, M.; et al. Randomized clinical trial comparing survival after D1 or D2 gastrectomy for gastric cancer. Br. J. Surg. 2014, 101, 23–31. [Google Scholar] [CrossRef] [PubMed]
  54. Songun, I.; Putter, H.; Kranenbarg, E.M.; Sasako, M.; van de Velde, C.J. Surgical treatment of gastric cancer: 15-year follow-up results of the randomised nationwide Dutch D1D2 trial. Lancet Oncol. 2010, 11, 439–449. [Google Scholar] [CrossRef]
  55. Degiuli, M.; Reddavid, R.; Tomatis, M.; Ponti, A.; Morino, M.; Sasako, M.; Italian Gastric Cancer Study Group. D2 dissection improves disease-specific survival in advanced gastric cancer patients: 15-year follow-up results of the Italian Gastric Cancer Study Group D1 versus D2 randomised controlled trial. Eur. J. Cancer 2021, 150, 10–22. [Google Scholar] [CrossRef] [PubMed]
  56. Kitano, S.; Shimoda, K.; Miyahara, M.; Shiraishi, N.; Bandoh, T.; Yoshida, T.; Shuto, K.; Kobayashi, M. Laparoscopic approaches in the management of patients with early gastric carcinomas. Surg. Laparosc. Endosc. 1995, 5, 359–362. [Google Scholar]
  57. Jiang, L.; Yang, K.H.; Guan, Q.L.; Cao, N.; Chen, Y.; Zhao, P.; Chen, Y.L.; Yao, L. Laparoscopy-assisted gastrectomy versus open gastrectomy for resectable gastric cancer: An update meta-analysis based on randomized controlled trials. Surg. Endosc. 2013, 27, 2466–2480. [Google Scholar] [CrossRef]
  58. Wang, W.; Li, Z.; Tang, J.; Wang, M.; Wang, B.; Xu, Z. Laparoscopic versus open total gastrectomy with D2 dissection for gastric cancer: A meta-analysis. J. Cancer Res. Clin. Oncol. 2013, 139, 1721–1734. [Google Scholar] [CrossRef]
  59. Kitano, S.; Shiraishi, N.; Fujii, K.; Yasuda, K.; Inomata, M.; Adachi, Y. A randomized controlled trial comparing open vs. laparoscopy-assisted distal gastrectomy for the treatment of early gastric cancer: An interim report. Surgery 2002, 131, S306–S311. [Google Scholar] [CrossRef]
  60. Fujii, K.; Sonoda, K.; Izumi, K.; Shiraishi, N.; Adachi, Y.; Kitano, S. T lymphocyte subsets and Th1/Th2 balance after laparoscopy-assisted distal gastrectomy. Surg. Endosc. 2003, 17, 1440–1444. [Google Scholar] [CrossRef]
  61. Huscher, C.G.; Mingoli, A.; Sgarzini, G.; Sansonetti, A.; Di Paola, M.; Recher, A.; Ponzano, C. Laparoscopic versus open subtotal gastrectomy for distal gastric cancer: Five-year results of a randomized prospective trial. Ann. Surg. 2005, 241, 232–237. [Google Scholar] [CrossRef] [PubMed]
  62. Hayashi, H.; Ochiai, T.; Shimada, H.; Gunji, Y. Prospective randomized study of open versus laparoscopy-assisted distal gastrectomy with extraperigastric lymph node dissection for early gastric cancer. Surg. Endosc. 2005, 19, 1172–1176. [Google Scholar] [CrossRef] [PubMed]
  63. Lee, J.H.; Han, H.S.; Lee, J.H. A prospective randomized study comparing open vs. laparoscopy-assisted distal gastrectomy in early gastric cancer: Early results. Surg. Endosc. 2005, 19, 168–173. [Google Scholar] [CrossRef] [PubMed]
  64. Kim, Y.W.; Baik, Y.H.; Yun, Y.H.; Nam, B.H.; Kim, D.H.; Choi, I.J.; Bae, J.M. Improved quality of life outcomes after laparoscopy-assisted distal gastrectomy for early gastric cancer: Results of a prospective randomized clinical trial. Ann. Surg. 2008, 248, 721–727. [Google Scholar] [CrossRef] [PubMed]
  65. Kim, H.H.; Hyung, W.J.; Cho, G.S.; Kim, M.C.; Han, S.U.; Kim, W.; Ryu, S.W.; Lee, H.J.; Song, K.Y. Morbidity and mortality of laparoscopic gastrectomy versus open gastrectomy for gastric cancer: An interim report--a phase III multicenter, prospective, randomized Trial (KLASS Trial). Ann. Surg. 2010, 251, 417–420. [Google Scholar] [CrossRef] [PubMed]
  66. Kim, Y.W.; Yoon, H.M.; Yun, Y.H.; Nam, B.H.; Eom, B.W.; Baik, Y.H.; Lee, S.E.; Lee, Y.; Kim, Y.A.; Park, J.Y.; et al. Long-term outcomes of laparoscopy-assisted distal gastrectomy for early gastric cancer: Result of a randomized controlled trial (COACT 0301). Surg. Endosc. 2013, 27, 4267–4276. [Google Scholar] [CrossRef]
  67. Sakuramoto, S.; Yamashita, K.; Kikuchi, S.; Futawatari, N.; Katada, N.; Watanabe, M.; Okutomi, T.; Wang, G.; Bax, L. Laparoscopy versus open distal gastrectomy by expert surgeons for early gastric cancer in Japanese patients: Short-term clinical outcomes of a randomized clinical trial. Surg. Endosc. 2013, 27, 1695–1705. [Google Scholar] [CrossRef]
  68. Takiguchi, S.; Fujiwara, Y.; Yamasaki, M.; Miyata, H.; Nakajima, K.; Sekimoto, M.; Mori, M.; Doki, Y. Laparoscopy-assisted distal gastrectomy versus open distal gastrectomy. A prospective randomized single-blind study. World J. Surg. 2013, 37, 2379–2386. [Google Scholar] [CrossRef]
  69. Hu, Y.; Zhao, G.; Zheng, H. Therapeutic effects of laparotomy and laparoscopic surgery on patients with gastric cancer. Pak. J. Med. Sci. 2015, 31, 572–575. [Google Scholar] [CrossRef]
  70. Hu, Y.; Huang, C.; Sun, Y.; Su, X.; Cao, H.; Hu, J.; Xue, Y.; Suo, J.; Tao, K.; He, X.; et al. Morbidity and Mortality of Laparoscopic Versus Open D2 Distal Gastrectomy for Advanced Gastric Cancer: A Randomized Controlled Trial. J. Clin. Oncol. 2016, 34, 1350–1357. [Google Scholar] [CrossRef]
  71. Yamashita, K.; Sakuramoto, S.; Kikuchi, S.; Futawatari, N.; Katada, N.; Hosoda, K.; Moriya, H.; Mieno, H.; Watanabe, M. Laparoscopic versus open distal gastrectomy for early gastric cancer in Japan: Long-term clinical outcomes of a randomized clinical trial. Surg. Today 2016, 46, 741–749. [Google Scholar] [CrossRef] [PubMed]
  72. Katai, H.; Mizusawa, J.; Katayama, H.; Takagi, M.; Yoshikawa, T.; Fukagawa, T.; Terashima, M.; Misawa, K.; Teshima, S.; Koeda, K.; et al. Short-term surgical outcomes from a phase III study of laparoscopy-assisted versus open distal gastrectomy with nodal dissection for clinical stage IA/IB gastric cancer: Japan Clinical Oncology Group Study JCOG0912. Gastric Cancer 2017, 20, 699–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Park, Y.K.; Yoon, H.M.; Kim, Y.W.; Park, J.Y.; Ryu, K.W.; Lee, Y.J.; Jeong, O.; Yoon, K.Y.; Lee, J.H.; Lee, S.E.; et al. Laparoscopy-assisted versus Open D2 Distal Gastrectomy for Advanced Gastric Cancer: Results From a Randomized Phase II Multicenter Clinical Trial (COACT 1001). Ann. Surg. 2018, 267, 638–645. [Google Scholar] [CrossRef] [PubMed]
  74. Shi, Y.; Xu, X.; Zhao, Y.; Qian, F.; Tang, B.; Hao, Y.; Luo, H.; Chen, J.; Yu, P. Short-term surgical outcomes of a randomized controlled trial comparing laparoscopic versus open gastrectomy with D2 lymph node dissection for advanced gastric cancer. Surg. Endosc. 2018, 32, 2427–2433. [Google Scholar] [CrossRef] [PubMed]
  75. Shi, Y.; Xu, X.; Zhao, Y.; Qian, F.; Tang, B.; Hao, Y.; Luo, H.; Chen, J.; Yu, P. Long-term oncologic outcomes of a randomized controlled trial comparing laparoscopic versus open gastrectomy with D2 lymph node dissection for advanced gastric cancer. Surgery 2019, 165, 1211–1216. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, Z.; Xing, J.; Cai, J.; Zhang, Z.; Li, F.; Zhang, N.; Wu, J.; Cui, M.; Liu, Y.; Chen, L.; et al. Short-term surgical outcomes of laparoscopy-assisted versus open D2 distal gastrectomy for locally advanced gastric cancer in North China: A multicenter randomized controlled trial. Surg. Endosc. 2019, 33, 33–45. [Google Scholar] [CrossRef] [Green Version]
  77. Li, Z.; Shan, F.; Ying, X.; Zhang, Y.; Jian-Yu, E.; Wang, Y.; Ren, H.; Su, X.; Ji, J. Assessment of Laparoscopic Distal Gastrectomy After Neoadjuvant Chemotherapy for Locally Advanced Gastric Cancer: A Randomized Clinical Trial. JAMA Surg. 2019, 154, 1093–1101. [Google Scholar] [CrossRef] [Green Version]
  78. Lee, H.J.; Hyung, W.J.; Yang, H.K.; Han, S.U.; Park, Y.K.; An, J.Y.; Kim, W.; Kim, H.I.; Kim, H.H.; Ryu, S.W.; et al. Short-term Outcomes of a Multicenter Randomized Controlled Trial Comparing Laparoscopic Distal Gastrectomy With D2 Lymphadenectomy to Open Distal Gastrectomy for Locally Advanced Gastric Cancer (KLASS-02-RCT). Ann. Surg. 2019, 270, 983–991. [Google Scholar] [CrossRef]
  79. Yu, J.; Huang, C.; Sun, Y.; Su, X.; Cao, H.; Hu, J.; Wang, K.; Suo, J.; Tao, K.; He, X.; et al. Effect of Laparoscopic vs. Open Distal Gastrectomy on 3-Year Disease-Free Survival in Patients With Locally Advanced Gastric Cancer: The CLASS-01 Randomized Clinical Trial. JAMA 2019, 321, 1983–1992. [Google Scholar] [CrossRef] [Green Version]
  80. Liu, F.; Huang, C.; Xu, Z.; Su, X.; Zhao, G.; Ye, J.; Du, X.; Huang, H.; Hu, J.; Li, G.; et al. Morbidity and Mortality of Laparoscopic vs. Open Total Gastrectomy for Clinical Stage I Gastric Cancer: The CLASS02 Multicenter Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1590–1597. [Google Scholar] [CrossRef]
  81. Hyung, W.J.; Yang, H.K.; Park, Y.K.; Lee, H.J.; An, J.Y.; Kim, W.; Kim, H.I.; Kim, H.H.; Ryu, S.W.; Hur, H.; et al. Long-Term Outcomes of Laparoscopic Distal Gastrectomy for Locally Advanced Gastric Cancer: The KLASS-02-RCT Randomized Clinical Trial. J. Clin. Oncol. 2020, 38, 3304–3313. [Google Scholar] [CrossRef] [PubMed]
  82. van der Veen, A.; Brenkman, H.J.F.; Seesing, M.F.J.; Haverkamp, L.; Luyer, M.D.P.; Nieuwenhuijzen, G.A.P.; Stoot, J.; Tegels, J.J.W.; Wijnhoven, B.P.L.; Lagarde, S.M.; et al. Laparoscopic Versus Open Gastrectomy for Gastric Cancer (LOGICA): A Multicenter Randomized Clinical Trial. J. Clin. Oncol. 2021, 39, 978–989. [Google Scholar] [CrossRef] [PubMed]
  83. Huang, C.; Liu, H.; Hu, Y.; Sun, Y.; Su, X.; Cao, H.; Hu, J.; Wang, K.; Suo, J.; Tao, K.; et al. Laparoscopic vs. Open Distal Gastrectomy for Locally Advanced Gastric Cancer: Five-Year Outcomes From the CLASS-01 Randomized Clinical Trial. JAMA Surg. 2022, 157, 9–17. [Google Scholar] [CrossRef] [PubMed]
  84. Son, S.Y.; Hur, H.; Hyung, W.J.; Park, Y.K.; Lee, H.J.; An, J.Y.; Kim, W.; Kim, H.I.; Kim, H.H.; Ryu, S.W.; et al. Laparoscopic vs. Open Distal Gastrectomy for Locally Advanced Gastric Cancer: 5-Year Outcomes of the KLASS-02 Randomized Clinical Trial. JAMA Surg. 2022, 157, 879–886. [Google Scholar] [CrossRef] [PubMed]
  85. Hakkenbrak, N.A.G.; Jansma, E.P.; van der Wielen, N.; van der Peet, D.L.; Straatman, J. Laparoscopic versus open distal gastrectomy for gastric cancer: A systematic review and meta-analysis. Surgery 2022, 171, 1552–1561. [Google Scholar] [CrossRef]
  86. Kim, H.G.; Park, J.H.; Jeong, S.H.; Lee, Y.J.; Ha, W.S.; Choi, S.K.; Hong, S.C.; Jung, E.J.; Ju, Y.T.; Jeong, C.Y.; et al. Totally laparoscopic distal gastrectomy after learning curve completion: Comparison with laparoscopy-assisted distal gastrectomy. J. Gastric Cancer 2013, 13, 26–33. [Google Scholar] [CrossRef] [Green Version]
  87. Jung, D.H.; Son, S.Y.; Park, Y.S.; Shin, D.J.; Ahn, H.S.; Ahn, S.H.; Park, D.J.; Kim, H.H. The learning curve associated with laparoscopic total gastrectomy. Gastric Cancer 2016, 19, 264–272. [Google Scholar] [CrossRef]
  88. Kim, M.S.; Kim, W.J.; Hyung, W.J.; Kim, H.I.; Han, S.U.; Kim, Y.W.; Ryu, K.W.; Park, S. Comprehensive Learning Curve of Robotic Surgery: Discovery From a Multicenter Prospective Trial of Robotic Gastrectomy. Ann. Surg. 2021, 273, 949–956. [Google Scholar] [CrossRef]
  89. Kim, H.I.; Han, S.U.; Yang, H.K.; Kim, Y.W.; Lee, H.J.; Ryu, K.W.; Park, J.M.; An, J.Y.; Kim, M.C.; Park, S.; et al. Multicenter Prospective Comparative Study of Robotic Versus Laparoscopic Gastrectomy for Gastric Adenocarcinoma. Ann. Surg. 2016, 263, 103–109. [Google Scholar] [CrossRef]
  90. Li, Z.Y.; Zhou, Y.B.; Li, T.Y.; Li, J.P.; Zhou, Z.W.; She, J.J.; Hu, J.K.; Qian, F.; Shi, Y.; Tian, Y.L.; et al. Robotic Gastrectomy versus Laparoscopic Gastrectomy for Gastric Cancer: A Multicenter Cohort Study of 5402 Patients in China. Ann. Surg. 2021. [Google Scholar] [CrossRef]
  91. Wang, Z.K.; Lin, J.X.; Wang, F.H.; Xie, J.W.; Wang, J.B.; Lu, J.; Chen, Q.Y.; Cao, L.L.; Lin, M.; Tu, R.H.; et al. Robotic spleen-preserving total gastrectomy shows better short-term advantages: A comparative study with laparoscopic surgery. Surg. Endosc. 2022, 36, 8639–8650. [Google Scholar] [CrossRef] [PubMed]
  92. Omori, T.; Yamamoto, K.; Hara, H.; Shinno, N.; Yamamoto, M.; Fujita, K.; Kanemura, T.; Takeoka, T.; Akita, H.; Wada, H.; et al. Comparison of robotic gastrectomy and laparoscopic gastrectomy for gastric cancer: A propensity score-matched analysis. Surg. Endosc. 2022, 36, 6223–6234. [Google Scholar] [CrossRef] [PubMed]
  93. Kubo, N.; Sakurai, K.; Tamamori, Y.; Fukui, Y.; Kuroda, K.; Aomatsu, N.; Nishii, T.; Tachimori, A.; Maeda, K. Less Severe Intra-Abdominal Infections in Robotic Surgery for Gastric Cancer Compared with Conventional Laparoscopic Surgery: A Propensity Score-matched Analysis. Ann. Surg. Oncol. 2022, 29, 3922–3933. [Google Scholar] [CrossRef] [PubMed]
  94. Li, J.T.; Lin, J.X.; Wang, F.H.; Wang, J.B.; Lu, J.; Chen, Q.Y.; Cao, L.L.; Lin, M.; Tu, R.H.; Huang, Z.N.; et al. Comparison of long-term outcomes after robotic versus laparoscopic radical gastrectomy: A propensity score-matching study. Surg. Endosc. 2022, 6, 8047–8059. [Google Scholar] [CrossRef] [PubMed]
  95. Hikage, M.; Fujiya, K.; Waki, Y.; Kamiya, S.; Tanizawa, Y.; Bando, E.; Notsu, A.; Terashima, M. Advantages of a robotic approach compared with laparoscopy gastrectomy for patients with high visceral fat area. Surg. Endosc. 2022, 36, 6181–6193. [Google Scholar] [CrossRef] [PubMed]
  96. Isobe, T.; Murakami, N.; Minami, T.; Tanaka, Y.; Kaku, H.; Umetani, Y.; Kizaki, J.; Aoyagi, K.; Fujita, F.; Akagi, Y. Robotic versus laparoscopic distal gastrectomy in patients with gastric cancer: A propensity score-matched analysis. BMC Surg. 2021, 21, 203. [Google Scholar] [CrossRef]
  97. Lu, J.; Zheng, C.H.; Xu, B.B.; Xie, J.W.; Wang, J.B.; Lin, J.X.; Chen, Q.Y.; Cao, L.L.; Lin, M.; Tu, R.H.; et al. Assessment of Robotic Versus Laparoscopic Distal Gastrectomy for Gastric Cancer: A Randomized Controlled Trial. Ann. Surg. 2021, 273, 858–867. [Google Scholar] [CrossRef]
  98. Gong, S.; Li, X.; Tian, H.; Song, S.; Lu, T.; Jing, W.; Huang, X.; Xu, Y.; Wang, X.; Zhao, K.; et al. Clinical efficacy and safety of robotic distal gastrectomy for gastric cancer: A systematic review and meta-analysis. Surg. Endosc. 2022, 36, 2734–2748. [Google Scholar] [CrossRef]
  99. Ju, M.; Wang, S.C.; Syed, S.; Agrawal, D.; Porembka, M.R. Multidisciplinary Teams Improve Gastric Cancer Treatment Efficiency at a Large Safety Net Hospital. Ann. Surg. Oncol. 2020, 27, 645–650. [Google Scholar] [CrossRef]
  100. Cunningham, D.; Allum, W.H.; Stenning, S.P.; Thompson, J.N.; Van de Velde, C.J.; Nicolson, M.; Scarffe, J.H.; Lofts, F.J.; Falk, S.J.; Iveson, T.J.; et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N. Engl. J. Med. 2006, 355, 11–20. [Google Scholar] [CrossRef] [Green Version]
  101. Al-Batran, S.E.; Homann, N.; Pauligk, C.; Goetze, T.O.; Meiler, J.; Kasper, S.; Kopp, H.G.; Mayer, F.; Haag, G.M.; Luley, K.; 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. Lancet 2019, 393, 1948–1957. [Google Scholar] [CrossRef] [PubMed]
  102. Bang, Y.J.; Van Cutsem, E.; Fuchs, C.S.; Ohtsu, A.; Tabernero, J.; Ilson, D.H.; Hyung, W.J.; Strong, V.E.; Goetze, T.O.; Yoshikawa, T.; et al. KEYNOTE-585: Phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol. 2019, 15, 943–952. [Google Scholar] [CrossRef] [PubMed]
  103. Al-Batran, S.-E.; Lorenzen, S.; Thuss-Patience, P.C.; Homann, N.; Schenk, M.; Lindig, U.; Heuer, V.; Kretzschmar, A.; Goekkurt, E.; Haag, G.M.; et al. Surgical and pathological outcome, and pathological regression, in patients receiving perioperative atezolizumab in combination with FLOT chemotherapy versus FLOT alone for resectable esophagogastric adenocarcinoma: Interim results from DANTE, a randomized, multicenter, phase IIb trial of the FLOT-AIO German Gastric Cancer Group and Swiss SAKK. J. Clin. Oncol. 2022, 40, 4003. [Google Scholar] [CrossRef]
  104. Janjigian, Y.Y.; Van Cutsem, E.; Muro, K.; Wainberg, Z.; Al-Batran, S.E.; Hyung, W.J.; Molena, D.; Marcovitz, M.; Ruscica, D.; Robbins, S.H.; et al. MATTERHORN: Phase III study of durvalumab plus FLOT chemotherapy in resectable gastric/gastroesophageal junction cancer. Future Oncol. 2022, 18, 2465–2473. [Google Scholar] [CrossRef] [PubMed]
  105. Rivera, F.; Izquierdo-Manuel, M.; García-Alfonso, P.; Martínez de Castro, E.; Gallego, J.; Limón, M.L.; Alsina, M.; López, L.; Galán, M.; Falcó, E.; et al. Perioperative trastuzumab, capecitabine and oxaliplatin in patients with HER2-positive resectable gastric or gastro-oesophageal junction adenocarcinoma: NEOHX phase II trial. Eur. J. Cancer 2021, 145, 158–167. [Google Scholar] [CrossRef] [PubMed]
  106. Ajani, J.A.; Mansfield, P.F.; Crane, C.H.; Wu, T.T.; Lunagomez, S.; Lynch, P.M.; Janjan, N.; Feig, B.; Faust, J.; Yao, J.C.; et al. Paclitaxel-based chemoradiotherapy in localized gastric carcinoma: Degree of pathologic response and not clinical parameters dictated patient outcome. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2005, 23, 1237–1244. [Google Scholar] [CrossRef]
  107. Ajani, J.A.; Mansfield, P.F.; Janjan, N.; Morris, J.; Pisters, P.W.; Lynch, P.M.; Feig, B.; Myerson, R.; Nivers, R.; Cohen, D.S.; et al. Multi-institutional trial of preoperative chemoradiotherapy in patients with potentially resectable gastric carcinoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2004, 22, 2774–2780. [Google Scholar] [CrossRef] [PubMed]
  108. Lowy, A.M.; Feig, B.W.; Janjan, N.; Rich, T.A.; Pisters, P.W.T.; Ajani, J.A.; Mansfield, P.F. A pilot study of preoperative chemoradiotherapy for resectable gastric cancer. Ann. Surg. Oncol. 2001, 8, 519–524. [Google Scholar] [CrossRef]
  109. Rivera, F.; Galán, M.; Tabernero, J.; Cervantes, A.; Vega-Villegas, M.E.; Gallego, J.; Laquente, B.; Rodríguez, E.; Carrato, A.; Escudero, P.; et al. Phase II trial of preoperative irinotecan-cisplatin followed by concurrent irinotecan-cisplatin and radiotherapy for resectable locally advanced gastric and esophagogastric junction adenocarcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2009, 75, 1430–1436. [Google Scholar] [CrossRef]
  110. Stahl, M.; Walz, M.K.; Riera-Knorrenschild, J.; Stuschke, M.; Sandermann, A.; Bitzer, M.; Wilke, H.; Budach, W. Preoperative chemotherapy versus chemoradiotherapy in locally advanced adenocarcinomas of the oesophagogastric junction (POET): Long-term results of a controlled randomised trial. Eur. J. Cancer 2017, 81, 183–190. [Google Scholar] [CrossRef]
  111. Reynolds, J.V.; Preston, S.R.; O’Neill, B.; Baeksgaard, L.; Griffin, S.M.; Mariette, C.; Cuffe, S.; Cunningham, M.; Crosby, T.; Parker, I.; et al. ICORG 10-14: NEOadjuvant trial in Adenocarcinoma of the oEsophagus and oesophagoGastric junction International Study (Neo-AEGIS). BMC Cancer 2017, 17, 401. [Google Scholar] [CrossRef] [PubMed]
  112. Slagter, A.E.; Jansen, E.P.M.; van Laarhoven, H.W.M.; van Sandick, J.W.; van Grieken, N.C.T.; Sikorska, K.; Cats, A.; Muller-Timmermans, P.; Hulshof, M.C.C.M.; Boot, H.; et al. CRITICS-II: A multicentre randomised phase II trial of neo-adjuvant chemotherapy followed by surgery versus neo-adjuvant chemotherapy and subsequent chemoradiotherapy followed by surgery versus neo-adjuvant chemoradiotherapy followed by surgery in resectable gastric cancer. BMC Cancer 2018, 18, 877. [Google Scholar] [CrossRef] [Green Version]
  113. Hoeppner, J.; Lordick, F.; Brunner, T.; Glatz, T.; Bronsert, P.; Röthling, N.; Schmoor, C.; Lorenz, D.; Ell, C.; Hopt, U.T.; et al. ESOPEC: Prospective randomized controlled multicenter phase III trial comparing perioperative chemotherapy (FLOT protocol) to neoadjuvant chemoradiation (CROSS protocol) in patients with adenocarcinoma of the esophagus (NCT02509286). BMC Cancer 2016, 16, 503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Leong, T.; Smithers, B.M.; Haustermans, K.; Michael, M.; Gebski, V.; Miller, D.; Zalcberg, J.; Boussioutas, A.; Findlay, M.; O’Connell, R.L.; et al. TOPGEAR: A Randomized, Phase III Trial of Perioperative ECF Chemotherapy with or Without Preoperative Chemoradiation for Resectable Gastric Cancer: Interim Results from an International, Intergroup Trial of the AGITG, TROG, EORTC and CCTG. Ann. Surg. Oncol. 2017, 24, 2252–2258. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, X.L.; Shi, H.J.; Cui, S.Z.; Tang, Y.Q.; Ba, M.C. Prospective, randomized trial comparing 5-FU/LV with or without oxaliplatin as adjuvant treatment following curative resection of gastric adenocarcinoma. Eur. J. Surg. Oncol. J. Eur. Soc. Surg. Oncol. Br. Assoc. Surg. Oncol. 2011, 37, 466–472. [Google Scholar] [CrossRef] [PubMed]
  116. Sasako, M.; Sakuramoto, S.; Katai, H.; Kinoshita, T.; Furukawa, H.; Yamaguchi, T.; Nashimoto, A.; Fujii, M.; Nakajima, T.; Ohashi, Y. Five-year outcomes of a randomized phase III trial comparing adjuvant chemotherapy with S-1 versus surgery alone in stage II or III gastric cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2011, 29, 4387–4393. [Google Scholar] [CrossRef] [Green Version]
  117. Bang, Y.J.; Kim, Y.W.; Yang, H.K.; Chung, H.C.; Park, Y.K.; Lee, K.H.; Lee, K.W.; Kim, Y.H.; Noh, S.I.; Cho, J.Y.; et al. Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): A phase 3 open-label, randomised controlled trial. Lancet 2012, 379, 315–321. [Google Scholar] [CrossRef]
  118. Noh, S.H.; Park, S.R.; Yang, H.K.; Chung, H.C.; Chung, I.J.; Kim, S.W.; Kim, H.H.; Choi, J.H.; Kim, H.K.; Yu, W.; et al. Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of an open-label, randomised phase 3 trial. Lancet. Oncol. 2014, 15, 1389–1396. [Google Scholar] [CrossRef]
  119. Yoshida, K.; Kodera, Y.; Kochi, M.; Ichikawa, W.; Kakeji, Y.; Sano, T.; Nagao, N.; Takahashi, M.; Takagane, A.; Watanabe, T.; et al. Addition of Docetaxel to Oral Fluoropyrimidine Improves Efficacy in Patients With Stage III Gastric Cancer: Interim Analysis of JACCRO GC-07, a Randomized Controlled Trial. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2019, 37, 1296–1304. [Google Scholar] [CrossRef]
  120. Macdonald, J.S.; Smalley, S.R.; Benedetti, J.; Hundahl, S.A.; Estes, N.C.; Stemmermann, G.N.; Haller, D.G.; Ajani, J.A.; Gunderson, L.L.; Jessup, J.M.; et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N. Engl. J. Med. 2001, 345, 725–730. [Google Scholar] [CrossRef]
  121. Dikken, J.L.; Jansen, E.P.M.; Cats, A.; Bakker, B.; Hartgrink, H.H.; Kranenbarg, E.M.K.; Boot, H.; Putter, H.; Peeters, K.C.M.J.; Van De Velde, C.J.H.; et al. Impact of the extent of surgery and postoperative chemoradiotherapy on recurrence patterns in gastric cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2010, 28, 2430–2436. [Google Scholar] [CrossRef] [PubMed]
  122. Yu, C.; Yu, R.; Zhu, W.; Song, Y.; Li, T. Intensity-modulated radiotherapy combined with chemotherapy for the treatment of gastric cancer patients after standard D1/D2 surgery. J. Cancer Res. Clin. Oncol. 2012, 138, 255–259. [Google Scholar] [CrossRef] [PubMed]
  123. Park, S.H.; Sohn, T.S.; Lee, J.; Lim, D.H.; Hong, M.E.; Kim, K.M.; Sohn, I.; Jung, S.H.; Choi, M.G.; Lee, J.H.; et al. Phase III Trial to Compare Adjuvant Chemotherapy With Capecitabine and Cisplatin Versus Concurrent Chemoradiotherapy in Gastric Cancer: Final Report of the Adjuvant Chemoradiotherapy in Stomach Tumors Trial, Including Survival and Subset Analyses. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2015, 33, 3130–3136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Cats, A.; Jansen, E.P.M.; van Grieken, N.C.T.; Sikorska, K.; Lind, P.; Nordsmark, M.; Meershoek-Klein Kranenbarg, E.; Boot, H.; Trip, A.K.; Swellengrebel, H.A.M.; et al. Chemotherapy versus chemoradiotherapy after surgery and preoperative chemotherapy for resectable gastric cancer (CRITICS): An international, open-label, randomised phase 3 trial. Lancet Oncol. 2018, 19, 616–628. [Google Scholar] [CrossRef]
  125. Park, S.H.; Lim, D.H.; Sohn, T.S.; Lee, J.; Zang, D.Y.; Kim, S.T.; Kang, J.H.; Oh, S.Y.; Hwang, I.G.; Ji, J.H.; et al. A randomized phase III trial comparing adjuvant single-agent S1, S-1 with oxaliplatin, and postoperative chemoradiation with S-1 and oxaliplatin in patients with node-positive gastric cancer after D2 resection: The ARTIST 2 trial. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2021, 32, 368–374. [Google Scholar] [CrossRef] [PubMed]
  126. Yang, T.S.; Wang, X.F.; Fairweather, M.; Sun, Y.H.; Mamon, H.J.; Wang, J.P. The Survival Benefit From the Addition of Radiation to Chemotherapy in Gastric Cancer Patients Following Surgical Resection. Clin. Oncol. 2020, 32, 110–120. [Google Scholar] [CrossRef]
  127. Jeong, O.; Park, Y.K.; Choi, W.Y.; Ryu, S.Y. Prognostic significance of non-curative gastrectomy for incurable gastric carcinoma. Ann. Surg. Oncol. 2014, 21, 2587–2593. [Google Scholar] [CrossRef]
  128. Mariette, C.; Bruyère, E.; Messager, M.; Pichot-Delahaye, V.; Paye, F.; Dumont, F.; Brachet, D.; Piessen, G. Palliative resection for advanced gastric and junctional adenocarcinoma: Which patients will benefit from surgery? Ann. Surg. Oncol. 2013, 20, 1240–1249. [Google Scholar] [CrossRef]
  129. Chang, Y.R.; Han, D.S.; Kong, S.H.; Lee, H.J.; Kim, S.H.; Kim, W.H.; Yang, H.K. The value of palliative gastrectomy in gastric cancer with distant metastasis. Ann. Surg. Oncol. 2012, 19, 1231–1239. [Google Scholar] [CrossRef]
  130. Zhang, J.Z.; Lu, H.S.; Huang, C.M.; Wu, X.Y.; Wang, C.; Guan, G.X.; Zhen, J.W.; Huang, H.G.; Zhang, X.F. Outcome of palliative total gastrectomy for stage IV proximal gastric cancer. Am. J. Surg. 2011, 202, 91–96. [Google Scholar] [CrossRef]
  131. Cheon, S.H.; Rha, S.Y.; Jeung, H.C.; Im, C.K.; Kim, S.H.; Kim, H.R.; Ahn, J.B.; Roh, J.K.; Noh, S.H.; Chung, H.C. Survival benefit of combined curative resection of the stomach (D2 resection) and liver in gastric cancer patients with liver metastases. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2008, 19, 1146–1153. [Google Scholar] [CrossRef] [PubMed]
  132. Linhares, E.; Monteiro, M.; Kesley, R.; Santos, C.E.; Filho, O.S.; Simões, J.H. Major hepatectomy for isolated metastases from gastric adenocarcinoma. HPB Off. J. Int. Hepato Pancreato Biliary Assoc. 2003, 5, 235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Shirabe, K.; Wakiyama, S.; Gion, T.; Watanabe, M.; Miyazaki, M.; Yoshinaga, K.; Tokunaga, M.; Nagaie, T. Hepatic resection for the treatment of liver metastases in gastric carcinoma: Review of the literature. HPB Off. J. Int. Hepato Pancreato Biliary Assoc. 2006, 8, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Okano, K.; Maeba, T.; Ishimura, K.; Karasawa, Y.; Goda, F.; Wakabayashi, H.; Usuki, H.; Maeta, H. Hepatic resection for metastatic tumors from gastric cancer. Ann. Surg. 2002, 235, 86–91. [Google Scholar] [CrossRef] [PubMed]
  135. Koga, R.; Yamamoto, J.; Ohyama, S.; Saiura, A.; Seki, M.; Seto, Y.; Yamaguchi, T. Liver resection for metastatic gastric cancer: Experience with 42 patients including eight long-term survivors. Jpn. J. Clin. Oncol. 2007, 37, 836–842. [Google Scholar] [CrossRef]
  136. Aurello, P.; Petrucciani, N.; Giulitti, D.; Campanella, L.; D’Angelo, F.; Ramacciato, G. Pulmonary metastases from gastric cancer: Is there any indication for lung metastasectomy? A systematic review. Med. Oncol. 2016, 33, 9. [Google Scholar] [CrossRef]
  137. Iijima, Y.; Akiyama, H.; Atari, M.; Fukuhara, M.; Nakajima, Y.; Kinosita, H.; Uramoto, H. Pulmonary Resection for Metastatic Gastric Cancer. Ann. Thorac. Cardiovasc. Surg. Off. J. Assoc. Thorac. Cardiovasc. Surg. Asia 2016, 22, 230–236. [Google Scholar] [CrossRef] [Green Version]
  138. Yoshida, Y.; Imakiire, T.; Yoneda, S.; Obuchi, T.; Inada, K.; Iwasaki, A. Ten cases of resected solitary pulmonary metastases arising from gastric cancer. Asian Cardiovasc. Thorac. Ann. 2014, 22, 578–582. [Google Scholar] [CrossRef]
  139. Oguri, Y.; Okui, M.; Yamamichi, T.; Asakawa, A.; Harada, M.; Horio, H. The impact of pulmonary metastasectomy from gastric cancer. Mol. Clin. Oncol. 2019, 11, 401–404. [Google Scholar] [CrossRef] [Green Version]
  140. Al-Batran, S.E.; Homann, N.; Pauligk, C.; Illerhaus, G.; Martens, U.M.; Stoehlmacher, J.; Schmalenberg, H.; Luley, K.B.; Prasnikar, N.; Egger, M.; et al. Effect of Neoadjuvant Chemotherapy Followed by Surgical Resection on Survival in Patients With Limited Metastatic Gastric or Gastroesophageal Junction Cancer: The AIO-FLOT3 Trial. JAMA Oncol. 2017, 3, 1237–1244. [Google Scholar] [CrossRef]
  141. Al-Batran, S.E.; Goetze, T.O.; Mueller, D.W.; Vogel, A.; Winkler, M.; Lorenzen, S.; Novotny, A.; Pauligk, C.; Homann, N.; Jungbluth, T.; et al. The RENAISSANCE (AIO-FLOT5) trial: Effect of chemotherapy alone vs. chemotherapy followed by surgical resection on survival and quality of life in patients with limited-metastatic adenocarcinoma of the stomach or esophagogastric junction—A phase III trial of the German AIO/CAO-V/CAOGI. BMC Cancer 2017, 17, 893. [Google Scholar] [CrossRef]
  142. Terashima, M.; Yoshida, K.; Rha, S.Y.; Bae, J.M.; Li, G.; Katai, H.; Watanabe, M.; Seto, Y.; Yang, H.-K.; Ji, J.; et al. International retrospective cohort study of conversion therapy for stage IV gastric cancer 1 (CONVO-GC-1). J. Clin. Oncol. 2018, 36, 4042. [Google Scholar] [CrossRef]
  143. Riihimäki, M.; Hemminki, A.; Sundquist, K.; Sundquist, J.; Hemminki, K. Metastatic spread in patients with gastric cancer. Oncotarget 2016, 7, 52307–52316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Bonnot, P.E.; Piessen, G.; Kepenekian, V.; Decullier, E.; Pocard, M.; Meunier, B.; Bereder, J.M.; Abboud, K.; Marchal, F.; Quenet, F.; et al. Cytoreductive Surgery With or Without Hyperthermic Intraperitoneal Chemotherapy for Gastric Cancer With Peritoneal Metastases (CYTO-CHIP study): A Propensity Score Analysis. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2019, 37, 2028–2040. [Google Scholar] [CrossRef] [PubMed]
  145. Yang, X.J.; Huang, C.Q.; Suo, T.; Mei, L.J.; Yang, G.L.; Cheng, F.L.; Zhou, Y.F.; Xiong, B.; Yonemura, Y.; Li, Y. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy improves survival of patients with peritoneal carcinomatosis from gastric cancer: Final results of a phase III randomized clinical trial. Ann. Surg. Oncol. 2011, 18, 1575–1581. [Google Scholar] [CrossRef] [Green Version]
  146. Chicago Consensus Working, G. The Chicago Consensus on peritoneal surface malignancies: Management of gastric metastases. Cancer 2020, 126, 2541–2546. [Google Scholar] [CrossRef]
  147. Glehen, O.; Gilly, F.N.; Arvieux, C.; Cotte, E.; Boutitie, F.; Mansvelt, B.; Bereder, J.M.; Lorimier, G.; Quenet, F.; Elias, D. Peritoneal carcinomatosis from gastric cancer: A multi-institutional study of 159 patients treated by cytoreductive surgery combined with perioperative intraperitoneal chemotherapy. Ann. Surg. Oncol. 2010, 17, 2370–2377. [Google Scholar] [CrossRef] [Green Version]
  148. Okabe, H.; Ueda, S.; Obama, K.; Hosogi, H.; Sakai, Y. Induction chemotherapy with S-1 plus cisplatin followed by surgery for treatment of gastric cancer with peritoneal dissemination. Ann. Surg. Oncol. 2009, 16, 3227–3236. [Google Scholar] [CrossRef]
  149. Badgwell, B.; Cormier, J.N.; Krishnan, S.; Yao, J.; Staerkel, G.A.; Lupo, P.J.; Pisters, P.W.T.; Feig, B.; Mansfield, P. Does neoadjuvant treatment for gastric cancer patients with positive peritoneal cytology at staging laparoscopy improve survival? Ann. Surg. Oncol. 2008, 15, 2684–2691. [Google Scholar] [CrossRef]
  150. Badgwell, B.; Blum, M.; Das, P.; Estrella, J.; Wang, X.; Ho, L.; Fournier, K.; Royal, R.; Mansfield, P.; Ajani, J. Phase II Trial of Laparoscopic Hyperthermic Intraperitoneal Chemoperfusion for Peritoneal Carcinomatosis or Positive Peritoneal Cytology in Patients with Gastric Adenocarcinoma. Ann. Surg. Oncol. 2017, 24, 3338–3344. [Google Scholar] [CrossRef]
  151. Takebayashi, K.; Murata, S.; Yamamoto, H.; Ishida, M.; Yamaguchi, T.; Kojima, M.; Shimizu, T.; Shiomi, H.; Sonoda, H.; Naka, S.; et al. Surgery-induced peritoneal cancer cells in patients who have undergone curative gastrectomy for gastric cancer. Ann. Surg. Oncol. 2014, 21, 1991–1997. [Google Scholar] [CrossRef] [PubMed]
  152. Marutsuka, T.; Shimada, S.; Shiomori, K.; Hayashi, N.; Yagi, Y.; Yamane, T.; Ogawa, M. Mechanisms of peritoneal metastasis after operation for non-serosa-invasive gastric carcinoma: An ultrarapid detection system for intraperitoneal free cancer cells and a prophylactic strategy for peritoneal metastasis. Clin Cancer Res 2003, 9, 678–685. [Google Scholar] [PubMed]
  153. Cui, S.-z.; Liang, H.; Li, Y.; Zhou, Y.; Tao, K.; Zhou, Z.; Li, G.; Li, P.; Zhou, B.; Yao, H.; et al. PILGRIM: Phase III clinical trial in evaluating the role of hyperthermic intraperitoneal chemotherapy for locally advanced gastric cancer patients after radical gastrectomy with D2 lymphadenectomy(HIPEC-01). J. Clin. Oncol. 2020, 38, 4538. [Google Scholar] [CrossRef]
  154. Nadiradze, G.; Giger-Pabst, U.; Zieren, J.; Strumberg, D.; Solass, W.; Reymond, M.A. Pressurized Intraperitoneal Aerosol Chemotherapy (PIPAC) with Low-Dose Cisplatin and Doxorubicin in Gastric Peritoneal Metastasis. J. Gastrointest. Surg. Off. J. Soc. Surg. Aliment. Tract 2016, 20, 367–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Alyami, M.; Bonnot, P.E.; Mercier, F.; Laplace, N.; Villeneuve, L.; Passot, G.; Bakrin, N.; Kepenekian, V.; Glehen, O. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) for unresectable peritoneal metastasis from gastric cancer. Eur. J. Surg. Oncol. J. Eur. Soc. Surg. Oncol. Br. Assoc. Surg. Oncol. 2021, 47, 123–127. [Google Scholar] [CrossRef] [PubMed]
  156. Di Giorgio, A.; Schena, C.A.; El Halabieh, M.A.; Abatini, C.; Vita, E.; Strippoli, A.; Inzani, F.; Rodolfino, E.; Romanò, B.; Pacelli, F.; et al. Systemic chemotherapy and pressurized intraperitoneal aerosol chemotherapy (PIPAC): A bidirectional approach for gastric cancer peritoneal metastasis. Surg. Oncol. 2020, 34, 270–275. [Google Scholar] [CrossRef] [PubMed]
  157. Ellebæk, S.B.; Graversen, M.; Detlefsen, S.; Lundell, L.; Fristrup, C.W.; Pfeiffer, P.; Mortensen, M.B. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) of peritoneal metastasis from gastric cancer: A descriptive cohort study. Clin. Exp. Metastasis 2020, 37, 325–332. [Google Scholar] [CrossRef]
  158. Bonnot, P.E.; Rabel, T.; Lintis, A.; Laplace, N.; Bakrin, N.; Kepenekian, V.; Villeneuve, L.; Chauvenet, M.; Bouarioua, N.; Alyami, M.; et al. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) associated to systemic chemotherapy for gastric cancer with diffuse peritoneal metastases in a palliative setting. J. Clin. Oncol. 2020, 38, e16538. [Google Scholar] [CrossRef]
  159. Eveno, C.; Jouvin, I.; Pocard, M. PIPAC EstoK 01: Pressurized IntraPeritoneal Aerosol Chemotherapy with cisplatin and doxorubicin (PIPAC C/D) in gastric peritoneal metastasis: A randomized and multicenter phase II study. Pleura Peritoneum 2018, 3, 20180116. [Google Scholar] [CrossRef]
  160. Kim, G.; Tan, H.L.; Chen, E.; Teo, S.C.; Jang, C.J.M.; Ho, J.; Ang, Y.; Ngoi, N.Y.L.; Chee, C.E.; Lieske, B.; et al. Study protocol: Phase 1 dose escalating study of Pressurized Intra-Peritoneal Aerosol Chemotherapy (PIPAC) with oxaliplatin in peritoneal metastasis. Pleura Peritoneum 2018, 3, 20180118. [Google Scholar] [CrossRef]
  161. Struller, F.; Horvath, P.; Solass, W.; Weinreich, F.J.; Strumberg, D.; Kokkalis, M.K.; Fischer, I.; Meisner, C.; Königsrainer, A.; Reymond, M.A. Pressurized intraperitoneal aerosol chemotherapy with low-dose cisplatin and doxorubicin (PIPAC C/D) in patients with gastric cancer and peritoneal metastasis: A phase II study. Ther. Adv. Med. Oncol. 2019, 11, 1758835919846402. [Google Scholar] [CrossRef] [PubMed]
  162. Thrift, A.P.; El-Serag, H.B. Burden of Gastric Cancer. Clin. Gastroenterol. Hepatol. 2020, 18, 534–542. [Google Scholar] [CrossRef] [PubMed]
  163. Fujitani, K.; Yang, H.K.; Mizusawa, J.; Kim, Y.W.; Terashima, M.; Han, S.U.; Iwasaki, Y.; Hyung, W.J.; Takagane, A.; Park, D.J.; et al. Gastrectomy plus chemotherapy versus chemotherapy alone for advanced gastric cancer with a single non-curable factor (REGATTA): A phase 3, randomised controlled trial. Lancet Oncol. 2016, 17, 309–318. [Google Scholar] [CrossRef]
  164. Choi, Y.B. Laparoscopic gastrojejunostomy for palliation of gastric outlet obstruction in unresectable gastric cancer. Surg. Endosc. Other Interv. Tech. 2002, 16, 1620–1626. [Google Scholar] [CrossRef] [PubMed]
  165. Chen, X.J.; Chen, G.M.; Wei, Y.C.; Yu, H.; Wang, X.C.; Zhao, Z.K.; Luo, T.Q.; Nie, R.C.; Zhou, Z.W. Palliative Gastrectomy versus Gastrojejunostomy for advanced Gastric cancer with outlet obstruction: A propensity score matching analysis. BMC Cancer 2021, 21, 188. [Google Scholar] [CrossRef]
  166. Yuan, S.T.; Wang, F.L.; Liu, N.; Liu, Y.H.; Liu, S.G.; Huang, Y.; Li, Y.Q.; Liu, X.B.; Zhang, Y.; Li, W.H.; et al. Concurrent involved-field radiotherapy and XELOX versus XELOX chemotherapy alone in gastric cancer patients with postoperative locoregional recurrence. Am. J. Clin. Oncol. 2015, 38, 130–134. [Google Scholar] [CrossRef] [PubMed]
  167. Tey, J.; Zheng, H.; Soon, Y.Y.; Leong, C.N.; Koh, W.Y.; Lim, K.; So, J.B.Y.; Shabbir, A.; Tham, I.W.K.; Lu, J. Palliative radiotherapy in symptomatic locally advanced gastric cancer: A phase II trial. Cancer Med. 2019, 8, 1447. [Google Scholar] [CrossRef] [PubMed]
  168. Asakura, H.; Hashimoto, T.; Harada, H.; Mizumoto, M.; Furutani, K.; Hasuike, N.; Matsuoka, M.; Ono, H.; Boku, N.; Nishimura, T. Palliative radiotherapy for bleeding from advanced gastric cancer: Is a schedule of 30 Gy in 10 fractions adequate? J. Cancer Res. Clin. Oncol. 2011, 137, 125–130. [Google Scholar] [CrossRef]
  169. Hashimoto, K.; Mayahara, H.; Takashima, A.; Nakajima, T.E.; Kato, K.; Hamaguchi, T.; Ito, Y.; Yamada, Y.; Kagami, Y.; Itami, J.; et al. Palliative radiation therapy for hemorrhage of unresectable gastric cancer: A single institute experience. J. Cancer Res. Clin. Oncol. 2009, 135, 1117–1123. [Google Scholar] [CrossRef]
  170. Burmeister, B.H.; Denham, J.W.; O’Brien, M.; Jamieson, G.G.; Gill, P.G.; Devitt, P.; Yeoh, E.; Hamilton, C.S.; Ackland, S.P.; Lamb, D.S.; et al. Combined modality therapy for esophageal carcinoma: Preliminary results from a large Australasian multicenter study. Int. J. Radiat. Oncol. Biol. Phys. 1995, 32, 997–1006. [Google Scholar] [CrossRef]
  171. Harvey, J.A.; Bessell, J.R.; Beller, E.; Thomas, J.; Gotley, D.C.; Burmeister, B.H.; Walpole, E.T.; Thomson, D.B.; Martin, I.; Doyle, L.; et al. Chemoradiation therapy is effective for the palliative treatment of malignant dysphagia. Dis. Esophagus Off. J. Int. Soc. Dis. Esophagus 2004, 17, 260–265. [Google Scholar] [CrossRef] [PubMed]
  172. Tey, J.; Back, M.F.; Shakespeare, T.P.; Mukherjee, R.K.; Lu, J.J.; Lee, K.M.; Wong, L.C.; Leong, C.N.; Zhu, M. The role of palliative radiation therapy in symptomatic locally advanced gastric cancer. Int. J. Radiat. Oncol. Biol. Phys. 2007, 67, 385–388. [Google Scholar] [CrossRef] [PubMed]
  173. Jeurnink, S.M.; van Eijck, C.H.J.; Steyerberg, E.W.; Kuipers, E.J.; Siersema, P.D. Stent versus gastrojejunostomy for the palliation of gastric outlet obstruction: A systematic review. BMC Gastroenterol. 2007, 7, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Wu, K.L.; Tsao, W.L.; Shyu, R.Y. Low-power laser therapy for gastrointestinal neoplasia. J. Gastroenterol. 2000, 35, 518–523. [Google Scholar] [CrossRef] [PubMed]
  175. Freitas, D.; Gouveia, H.; Sofia, C.; Cabral, J.P.; Donato, A. Endoscopic Nd-YAG laser therapy as palliative treatment for esophageal and cardial cancer. Hepatogastroenterology 1995, 42, 633–637. [Google Scholar]
  176. Norberto, L.; Ranzato, R.; Marino, S.; Angriman, I.; Erroi, F.; Donadi, M.; Vella, V.; D’Erminio, A.; D’Amico, D.F. Endoscopic palliation of esophageal and cardial cancer: Neodymium-yttrium aluminum garnet laser therapy. Dis. Esophagus 1999, 12, 294–296. [Google Scholar] [CrossRef] [PubMed]
  177. Barr, H.; Krasner, N. Interstitial laser photocoagulation for treating bleeding gastric cancer. BMJ Br. Med. J. 1989, 299, 659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Mathus-Vliegen, M.H.E.; Tytgat, N.J.G. Laser photocoagulation in the palliation of colorectal malignancies. Cancer 1986, 57, 2212–2216. [Google Scholar] [CrossRef]
  179. Pittayanon, R.; Rerknimitr, R.; Barkun, A. Prognostic factors affecting outcomes in patients with malignant GI bleeding treated with a novel endoscopically delivered hemostatic powder. Gastrointest. Endosc. 2018, 87, 994–1002. [Google Scholar] [CrossRef]
  180. Lei, Z.; Tan, I.B.; Das, K.; Deng, N.; Zouridis, H.; Pattison, S.; Chua, C.; Feng, Z.; Guan, Y.K.; Ooi, C.H.; et al. Identification of molecular subtypes of gastric cancer with different responses to PI3-kinase inhibitors and 5-fluorouracil. Gastroenterology 2013, 145, 554–565. [Google Scholar] [CrossRef]
  181. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Cristescu, R.; Lee, J.; Nebozhyn, M.; Kim, K.M.; Ting, J.C.; Wong, S.S.; Liu, J.; Yue, Y.G.; Wang, J.; Yu, K.; et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat. Med. 2015, 21, 449–456. [Google Scholar] [CrossRef] [PubMed]
  183. Fiorillo, C.; Laterza, V.; Quero, G.; Menghi, R.; Cina, C.; Rosa, F.; Tortorelli, A.P.; Boskoski, I.; Alfieri, S. From biology to surgery: One step beyond histology for tailored surgical treatments of gastric cancer. Surg. Oncol. 2020, 34, 86–95. [Google Scholar] [CrossRef] [PubMed]
Table 1. Recommendations on the extent of resection for GC.
Table 1. Recommendations on the extent of resection for GC.
Cancer Society GuidelinesRecommendation
National Comprehensive Cancer Network [13]The minimum margin length is not specified
European Society of Medical Oncology [12]Subtotal gastrectomy should be performed with a minimum margin length of at least 5 cm for intestinal-type GC and at least 8 cm for diffuse-type GC
Japanese Gastric Cancer Association [11]Subtotal gastrectomy should be performed with a minimum margin length of at least 2 cm for T1 tumors, 3 cm for T2 or deeper tumors with an expansive growth pattern and 5 cm for T2 or deeper tumors with an infiltrative growth pattern
Italian Research Group for Gastric Cancer Guidelines [14]Subtotal gastrectomy should be performed with a minimum margin length of at least 2 cm for T1 tumors, 3 cm for T2 or deeper tumors with an expansive growth pattern and 5 cm for T2 or deeper tumors with an infiltrative growth pattern and diffuse Lauren histotype
Table 2. Japanese Gastric Cancer Association Lymph Node Stations.
Table 2. Japanese Gastric Cancer Association Lymph Node Stations.
StationsAnatomical Location
1–6Perigastric
1: Right of the cardia
2: Left of the cardia
3a: Lesser curvature (branches of the left gastric artery)
3b: Lesser curvature (2nd and distal branches of the right gastric artery)
4sa: Greater curvature (short gastric arteries)
4sb: Greater curvature (left gastroepiploic artery)
4d: Greater curvature (2nd and distal branches of the right gastroepiploic artery)
5: Superior to the pylorus
6: Inferior to the pylorus
7Left gastric artery
8Common hepatic artery
8a: Anterior
8p: Posterior
9Coeliac axis
10Splenic hilum
11Splenic artery
11p: Proximal
11d: Distal
12Hepatoduodenal ligament
12a: Proper hepatic artery
12b: Common bile duct
12p: Portal vein
13Posterior to the pancreas head
14Superior mesenteric vein
15Middle colic vein
16Para-aortic
16a1: Hiatus
16a2: Between celiac artery and left renal vein
16b1: Between left renal vein and inferior mesenteric artery
Sourced from [44].
Table 3. Randomized clinical trials on laparoscopic versus open gastrectomy for gastric cancer.
Table 3. Randomized clinical trials on laparoscopic versus open gastrectomy for gastric cancer.
AuthorsPatientsProceduresResults
Kitano et al., 2002 [59]28 EGCDistal gastrectomyFaster recovery, less pain, and less compromised pulmonary function in the LPS group
Fujii et al., 2003 [60]20 EGCDistal gastrectomyBetter preservation of Th1 immune response in the LPS group
Huscher et al., 2005 [61]59 T1-4 and N0-2 GCDistal gastrectomyNo difference in terms of mean number of resected lymph nodes, mortality, morbidity, five-year OS and DFS. LPS was associated with lower intraoperative blood loss, earlier resumption of oral intake, and earlier hospital discharge.
Hayashi et al., 2005 [62]28 EGCDistal gastrectomyNo difference in terms of oncological radicality. Shorter postoperative epidural anesthesia, lower IL-6 and CRP levels, without major postoperative complications in the LPS group.
Lee et al., 2005 [63]47 EGCDistal gastrectomySimilar oncological outcomes, but fewer pulmonary complications in the LPS group
Kim et al., 2008 [64]164 EGCDistal gastrectomyLPS-related advantages regarding QoL, intraoperative blood loss, analgesic use, and postoperative hospital stay.
Kim et al., 2010 [65]342 EGCDistal gastrectomyNo significant difference in morbidity and mortality rate.
Kim et al., 2013 [66]164 EGCDistal gastrectomySimilar overall postoperative morbidity, QoL, five-year OS and DFS. Mild complications were lower in the LPS group.
Sakuramoto et al., 2013 [67]64 EGCDistal gastrectomyLPS resulted in less postoperative pain with similar short-term outcomes than open surgery.
Takiguchi et al., 2013 [68]40 EGC Distal gastrectomyBenefits related to LPS were faster recovery, less intraoperative blood loss, less postoperative pain, smaller wound size, shorter postoperative hospital stay, and better levels of CRP and SaO2.
Hu et al., 2015 [69]66 stage I-III GCDistal gastrectomyLPS was associated with lower morbidity, less intraoperative blood loss, shorter hospital stay, faster recovery and better humoral and cellular immune response.
Hu et al., 2016 [70] CLASS-01 Trial1056 T2-4a and N0-3 GCDistal gastrectomy with D2 lymphadenectomySimilar node-dissection compliance, morbidity and mortality rate.
Kim et al., 2016 KLASS-01 Trial1416 EGCDistal gastrectomy with D1+ lymphadenectomyLPS resulted in lower wound complication rate, comparable overall morbidity and mortality.
Yamashita et al., 2016 [71]63 EGCDistal gastrectomyLPS was associated with less long-term wound pain.
Katai et al., 2017 [72]921 T1-2 and N0-1 GC non-endoscopically suitableDistal gastrectomyLPS was safe as open surgery presenting similar short-term clinical outcomes, with a significantly higher operative time but smaller blood loss.
Park et al., 2018 [73] COACT 1001 trial204 T2-4a and N0-2 GCDistal gastrectomy with D2 lymphadenectomyNo significant differences in three-year DFS, morbidity and overall lymphadenectomy noncompliance rate, despite the latter being significantly higher for stage III GC in the LPS group.
Shi et al., 2018 [74]328 T2-3 and N0-3 GCProximal, distal and total gastrectomy with D2 lymphadenectomyLPS resulted to be safe and feasible procedure in locally advanced GC compared to open surgery.
Shi et al., 2019 [75]328 T2-3 and N0-3 GCProximal, distal and total gastrectomy with D2 lymphadenectomyNo difference in terms of five-year OS, DFS and recurrence rate.
Wang et al., 2019 [76]446 T2-4a and N0-3Distal gastrectomy with D2 lymphadenectomyNo difference in terms of 30-day morbidity and mortality, three-year DFS and in compliance rate of D2 lymph node dissection.
Li et al., 2019 [77]96 T2-4a and N+ GC after neoadjuvant therapyDistal gastrectomy with D2 lymphadenectomyLPG gastrectomy resulted in a lower overall complication rate, less pain, similar postoperative recovery, better adjuvant chemotherapy completion rate and comparable mortality.
Lee et al., 2019 [78] KLASS-02 Trial1050 T2-4a and N0-1 GCDistal gastrectomy with D2 lymphadenectomyLPS was significantly associated with faster recovery, lower early morbidity rate, postoperative pain and analgesic use, and shorter hospital stay with no difference in terms of mortality and totally retrieved lymph nodes.
Yu et al., 2019 [79] CLASS-01 Trial1056 T2-4a and N0-3 GCDistal gastrectomy with D2 lymphadenectomyNo difference in terms of three-year DFS.
Liu et al., 2020 [80] CLASS-02 trial227 T1-2 and N0-1 (stage I) GCTotal gastrectomy No difference in terms of overall postoperative complication rate and mortality.
Hyung et al., 2020 [81] KLASS-02 Trial1050 T2-4a and N0-1 GCDistal gastrectomy with D2 lymphadenectomyNo difference in terms of three-year relapse-free survival rate.
Van de Veen et al., 2021 [82] LOGICA Trial227 T1-4a and N0-3b GCTotal or distal gastrectomy with D2 lymphadenectomyNo difference in terms of postoperative complications, in-hospital mortality, 30-day readmission rate, R0 resections, median lymph node harvested, one-year OS, and one-year global health-related QoL.
Huang et al., 2022 [83] CLASS-01 Trial1056 T2-4a and N0-3 GCDistal gastrectomy with D2 lymphadenectomySimilar five-year OS.
Son et al., 2022 [84] KLASS-02 Trial1050 T2-4a and N0-1 GCDistal gastrectomy with D2 lymphadenectomyFive-year OS and relapse-free survival rates were not significantly different between LPS and open surgery.
EGC, Early Gastric Cancer; LPS, Laparoscopic; GC, Gastric Cancer; OS, Overall Survival; DFS, Disease-Free Survival; QoL, Quality of Life; IL-6, Interleukin-6; CRP, C-Reactive Protein; CLASS-01, Chinese Laparoscopic Gastrointestinal Surgery Study 01; KLASS-01, Korean Laparoendoscopic Gastrointestinal Surgery Study 01; KLASS-02, Korean Laparoendoscopic Gastrointestinal Surgery Study 02; CLASS-02, Chinese Laparoscopic Gastrointestinal Surgery Study 02; LOGICA, Laparoscopic versus Open Gastrectomy for gastrIc Cancer.
Table 4. Key trials on adjuvant chemotherapy and chemoradiotherapy for gastric cancer.
Table 4. Key trials on adjuvant chemotherapy and chemoradiotherapy for gastric cancer.
AuthorsPatientsGroupsResults
Adjuvant Chemotherapy
Zhang et al., 2011 [115] (China) 80 resected (R0) GC38: 5-FU/LV;
42: 5-FU/LV + oxaliplatin
Improved RFS and OS for FOLFOX regime:
3-year RFS: 30 vs. 16 months, p < 0.05;
3-year OS: 36 vs. 28 months, p < 0.05
Sasako et al., 2011 [116] (Japan) ACTS-GC trial1034 Stage II–III resected (D2) GC515: S-1;
519: observation
Improved OS for adjuvant S-1:
5-year OS 71.7% vs. 61.1% (HR 0.669; 95% CI, 0.540–0.828).
Bang et al., 2012–2014 [117,118] (China, Taiwan, South Korea) CLASSIC trial1035 Stage II-IIIB resected (D2) GC520: CAPOX;
515: observation
Improved DFS for adjuvant CAPOX:
3-year DFS 74% vs. 59% (HR 0.56, 95% CI 0.44–0.72; p < 0.0001);
5-year DFS 68% vs. 53% (HR 0.66, 95% CI 0.51–0.85; p = 0.0015)
Improved OS for adjuvant CAPOX:
78% vs. 69%
Yoshida et al., 2019 [119] (Japan) Interim analysis of JACCRO GC-07 trial 913 Stage III resected (R0) GC454: S-1 + docetaxel;
459: S-1
Improved ReFS for S-1 plus docetaxel:
3-year ReFS 66% vs. 50% (HR 0.632; 99.99% CI, 0.400 to 0.998; p < 0.001)
Adjuvant chemotherapy plus chemoradiotherapy
Macdonald et al., 2001 [120] (US) INT 0116 556 resected gastric/GEJ cancer281: CRT + 5-FU/LV;
275: observation
Improved OS with CRT:
36 vs. 27 months (HR 1.35; 95% CI 1.09–1.66; p < 0.001)
Dikken et al., 2010 [121] (The Netherlands) 91 resected GC5: CRT + LV
39: CRT + X
47: CRT + XP
694: surgery only (from the DGCT trial)
Fewer local recurrences after CRT:
2% vs. 8%; p = 0.001
Yu et al., 2012 [122] (China) 68 T3/T4 and/or N+ resected GC34: 5-FU/LV + CRT;
34: 5-FU/LV
Improved OS and DFS for CRT:
3-year OS 67.7%, vs. 44.1 (p < 0.05);
3-year DFS 55.8 vs. 29.4% (p < 0.05)
Park et al., 2015 [123] (South Korea) ARTIST trial 458 resected (D2) GC230: XP + CRT;
228: XP
DFS not different:
HR 0.74; 95% CI 0.52 to 1.05; p = 0.0922
OS not different:
HR 1.130; 95% CI 0.775 to 1.647; p = 0.5272)
Cats et al., 2018 [124] (The Netherlands) CRITICS 788 Stage IB-IVA resectable gastric/GEJ cancer395: ECX/EOX + surgery + CRT;
393: ECX/EOX + surgery + ECX/EOX
OS not different:
37 vs. 43 months (HR 1.01; 95% CI 0.84–1.22; p = 0.90)
Park et al., 2021 [125] (South Korea) ARTIST-II546 Stage II-III pN > 0 resected (D2) GC183: SOX + CRT;
181: SOX;
182: S-1
DFS not different between SOX and SOX + CRT;
3-year DFS: 72.8% SOX + CRT vs. 74.3% SOX vs. 64.8% S-1 (HR 0.97; 0.66–1.42; p = 0.879)
GC, Gastric Cancer; CLASSIC, Capecitabine and Oxaliplatin Adjuvant Study in Stomach Cancer; CAPOX, capecitabine and oxaliplatin; LV, Leucovorin; FOLFOX, Folinic acid, fluorouracil and oxaliplatin; DFS, Disease free survival; HR, Hazard ratio; OS, Overall survival; RFS, Recurrence-free survival; ACTS-GC, Adjuvant Chemotherapy Trial of TS-1 for Gastric Cancer; 5-FU, 5-Fluorouracil; JACCRO, Japan Clinical Cancer Research Organization; ReFS, Relapse-free survival; GEJ, Gastroesophageal junction; CRT, Chemioradiotherapy; ARTIST, Adjuvant Chemoradiation Therapy in Stomach Cancer; XP, Capecitabine and cisplatin; SOX, S-1 and oxaliplatin; CRITICS, Chemoradiotherapy After Induction Chemotherapy in Cancer of the Stomach; ECX, epirubicin, cisplatin, and capecitabine; EOX, epirubicin, oxaliplatin, and capecitabine; DGCT, Dutch Gastric Cancer Group Trial; T, tumor stage; N, nodal stage from TNM staging system, as per the American Joint Committee on Cancer, 8th Edition.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rosa, F.; Schena, C.A.; Laterza, V.; Quero, G.; Fiorillo, C.; Strippoli, A.; Pozzo, C.; Papa, V.; Alfieri, S. The Role of Surgery in the Management of Gastric Cancer: State of the Art. Cancers 2022, 14, 5542. https://doi.org/10.3390/cancers14225542

AMA Style

Rosa F, Schena CA, Laterza V, Quero G, Fiorillo C, Strippoli A, Pozzo C, Papa V, Alfieri S. The Role of Surgery in the Management of Gastric Cancer: State of the Art. Cancers. 2022; 14(22):5542. https://doi.org/10.3390/cancers14225542

Chicago/Turabian Style

Rosa, Fausto, Carlo Alberto Schena, Vito Laterza, Giuseppe Quero, Claudio Fiorillo, Antonia Strippoli, Carmelo Pozzo, Valerio Papa, and Sergio Alfieri. 2022. "The Role of Surgery in the Management of Gastric Cancer: State of the Art" Cancers 14, no. 22: 5542. https://doi.org/10.3390/cancers14225542

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop