IJGII Inernational Journal of Gastrointestinal Intervention

pISSN 2636-0004 eISSN 2636-0012
ESCI
scopus

Article

home All Articles View

Original Article

Int J Gastrointest Interv 2023; 12(2): 69-74

Published online April 30, 2023 https://doi.org/10.18528/ijgii220012

Copyright © International Journal of Gastrointestinal Intervention.

The prognostic value of tumor budding in gastric adenocarcinomas: A cross-sectional study

Tuba Baydas1 , Ganime Coban2 , Mehmet Besiroglu3 , Melih Simsek3,* , Abdallah TM Shbair3 , Atakan Topcu3 , Ayse Irem Yasin3 , Mesut Seker3 , and Haci Mehmet Turk3

1Department of Internal Medicine, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
2Department of Pathology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
3Department of Medical Oncology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey

Correspondence to:*Department of Medical Oncology, Faculty of Medicine, Bezmialem Vakif University, Topkapı Mahallesi, Adnan Menderes, Vatan Bulvarı No:113, Istanbul 34093, Turkey.
E-mail address: mdmelih@gmail.com (M. Simsek).

Received: March 16, 2022; Revised: June 27, 2022; Accepted: June 28, 2022

This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/bync/4.0) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Background: Tumor budding is a negative prognostic factor for many solid malignancies. We investigated the relationship between patients’ clinicopathological characteristics and tumor budding, as well as the effect of budding level on prognosis.
Methods: In this retrospective, cross-sectional, and descriptive study at a tertiary university hospital, 104 patients with gastric adenocarcinoma were divided into two groups according to whether they had a high (≥ 10 buds) or low (< 10 buds) level of budding. The patients were retrospectively evaluated.
Results: Sixty-two patients had a low level of budding and 42 had a high level of budding. The mean time from the first diagnosis to the last followup was significantly longer (P < 0.05) in the low-budding group (27 months) than in the high-budding group (17 months). As the T stage, N stage, grade, and size of the tumor increased, the proportion of patients with high budding was found to be significantly higher (P < 0.05). High tumor budding, an age of 60 or over at diagnosis, a low body mass index, high N stage, presence of lymphovascular invasion, and recurrence were associated with overall survival.
Conclusion: Our study supports that budding is a negative prognostic factor in gastric cancer.

Keywords: Adenocarcinoma, Prognosis, Stomach neoplasms

Gastric cancer is the fourth most common malignancy in the world and accounts for the second-highest number of cancer-related deaths after lung cancer.1 It generally occurs between the fifth and seventh decades of life, and 70% of cases occur in developing countries.2 The prognosis of advanced-stage gastric cancer is poor, and the 5-year overall survival (OS) rate is less than 30%. With developments in new surgical techniques, radiotherapy (RT), and chemotherapy (CT), the 5-year OS rates of early-stage disease have exceeded 95%.3 In addition to TNM staging, new guiding clinical and pathological markers are required to accurately evaluate patients who would benefit from personalized treatment after surgery and to facilitate clinical management.4

Tumor budding (TB), which is thought to constitute the basis for invasion and metastasis, is defined as the presence of single malignant cells or the presence of undifferentiated malignant cell clusters consisting of 2–4 cells, called tumor buds, in the tumor stroma on the invasive surface of the tumor. An important step in the invasion process is the loss of intercellular connections and stromal invasion. Therefore, a close relationship is thought to exist between TB and the epithelial-mesenchymal transition, where adhesion connections between cells disappear, with the loss of membranous E-cadherin and β-catenin expression of epithelial cells.5

TB was first described by Imai in the Japanese medical literature in 1949.6 Subsequent studies have been conducted on the association of TB with the prognosis in many solid malignancies such as breast, gastric, colon, rectal, and cervical cancers. In 1989, Morodomi et al6 found that TB increased the risk of lymphatic invasion and lymph node metastasis in rectal cancers, and following studies showed it to be associated with recurrence and distant metastasis in colorectal cancers.7,8 Currently, high TB in colorectal cancer is accepted as a well-known poor prognostic factor, and routine evaluation of TB has been recommended.9 The International Association for Cancer Control (UICC) has defined TB as an additional independent prognostic factor in colorectal. The UICC emphasized the necessity of studies to determine the most appropriate assessment method before TB is used as a routine marker of metastasis and lymph node invasion risk, especially in T1 tumors.10

The prognostic value of TB in gastric cancer is not well known, and there is a limited number of relevant clinical studies. Therefore, we investigated the relationship between patients’ clinicopathological characteristics and TB, as well as the effect of the budding level on prognosis.

Study design

This study had a retrospective and cross-sectional design, and it was organized in accordance with the STROBE statement.11

Participants

In our institutional database, the records of patients who were diagnosed with gastric cancer and underwent surgery at our hospital between 2012 and 2020 were retrospectively reviewed. In total, 107 patients aged between 18 and 85 were included.

Patients with gastric lymphomas or neuroendocrine tumors, metastatic disease at diagnosis, mucinous adenocarcinomas, and signet ring cell carcinomas (since they were non-cohesive at diagnosis), and patients who had another primary cancer were not included.

Data collection tools

Patients’ age, sex, blood type, height, and weight were collected, as well as the date of the first diagnosis, tumor size, tumor location, operation type, number of lymph nodes removed, number of lymph nodes involved, surgical margin, lymphovascular invasion (LVI), perineural invasion (PNI), tumor stage, histological grade, whether the patient received adjuvant CT or RT, recurrence status, and the time from diagnosis to recurrence were.

Pathological slides stained with hematoxylin and eosin were retrospectively evaluated with light microscopy to assess TB. The tumor invasion surface was found by ×10 magnification on light microscopy during scoring, and the area where budding was densely observed was called a “hotspot.” In this region, the cell groups separated from the tumor were counted under ×20 magnification of the objective. The number of buds was counted per 0.785 mm2 area, which is the sample area diameter suitable for a lens diameter of 20 mm in accordance with the Colorectal Carcinoma Reporting Protocol that was published in 2016. TB was graded as follows according to the Colorectal Carcinoma Reporting Protocol:

- No tumor budding

- Low grade: 0–4 tumor buds

- Moderate grade: 5–9 tumor buds

- High grade: ≥ 10 tumor buds

Three patients without TB were not included in the analysis. The patients were divided into two groups according to the degree of TB: high (tumor bud count ≥ 10) and low (bud count < 10) (Fig. 1, 2).

Figure 1. High level of tumor budding on invasive tumor surface (H&E stain). (A) High grade tumor budding density (×20 magnification) (asterisk). (B) High grade tumor budding density (×10 magnification) (asterisk).
Figure 2. Low level of tumor budding on invasive tumor surface (H&E stain). (A) Low grade tumor budding density (×10 magnification) (asterisk). (B) Low grade tumor budding density (×20 magnification) (asterisk).

Ethical aspects of the research

Before the study, institutional permission was acquired from the hospital where the data would be collected, as well as ethics committee approval (IRB No: 05.11.2019/21/387). Because of the retrospective nature of this study, the requirement for informed consent was waived. The Helsinki Declaration was followed in the research.

Data analysis

Data were analyzed in SPSS version 26.0 (IBM Corp., Armonk, NY, USA). Patients diagnosed with gastric cancer were staged according to TNM staging system, in which the characteristics of tumor (T), lymph node (N) and metastasis (M) were evaluated according to the recommendations of the American Joint Committee on Cancer.

OS was defined as the time from first diagnosis to the patient’s last visit or death. The TB score was calculated in all patients. The patients were evaluated in terms of OS and recurrence during their follow-up period, and the relationships of the degree of TB with OS, recurrence, and patients’ demographic and histopathological features were investigated.

Statistical analysis

The mean, median, standard deviation, lowest, highest, frequency and ratio values were used to present descriptive statistics. The distribution of variables was measured with Kolmogorov-Smirnov test. The Mann–Whitney U test was used to analyze quantitative independent data. The chi-squared test was used to analyze qualitative independent data, and the Fisher exact test was used when the conditions of the chi-squared test were not met. Cox regression (univariate and multivariate) analysis and the Kaplan-Meier method were used for survival analysis. A P-value of less than 0.05 was considered significant.

Of the 104 patients included, 35 (33.7%) were women, 69 (66.3%) were men, and the mean age at diagnosis was 62 ± 11 years. The mean weight was 69 ± 16 kg, and the mean body mass index (BMI) was 25.3 ± 5.3 kg/m2. The most common blood type was group A (43.2%). The histopathological characteristics of the patients are shown in Table 1. The mean number of lymph nodes removed during surgery was 35 ± 14 (range, 11–81), and the mean tumor size was 5.4 ± 2.5 cm.

Table 1 . Histopathological Characteristics.

CharacteristicValue
Number of dissected lymph nodes32 (11–81)
Size of tumor (cm)5.5 (1–12)
Location of tumor
Cardia24 (23.1)
Antrum34 (32.7)
Corpus33 (31.7)
Incisura angularis3 (2.9)
Diffuse7 (6.7)
Fundus3 (2.9)
Extent of lymph node dissection
D111 (10.6)
D293 (89.4)
Resection type
Subtotal gastrectomy35 (33.7)
Total gastrectomy69 (66.3)
Surgical margins
Positive12 (11.5)
Negative92 (88.5)
T stage
I10 (9.6)
II14 (13.5)
III47 (45.2)
IV33 (31.7)
N stage
028 (26.9)
I15 (14.4)
II27 (26.0)
III34 (32.7)
Grade
I10 (9.6)
II57 (54.8)
III37 (35.6)
Lymphovascular invasion
Present72 (69.2)
Absent32 (30.8)
Perineural invasion
Present66 (63.5)
Absent38 (36.5)

Values are presented as mean (range) or number (%).



The clinical characteristics of the patients are presented in Table 2. Of the deaths, 19 were due to causes other than cancer (e.g., cerebrovascular event, myocardial infarction, and pneumonia). Patients’ age, sex distribution, weight, and BMI at the time of diagnosis did not differ significantly between the groups with low or high budding.

Table 2 . Clinical Characteristics of the Patients.

CharacteristicValue
Total follow-up (mo)23 (1.5–96)
Number of received CT cycles6 (2–8)
Follow up until recurrence (mo)14 (4–49)
Adjuvant CT
Present71 (68.3)
Absent33 (31.7)
Adjuvant CT protocol
Folfox33 (31.7)
FLOT5 (4.8)
Capox12 (11.5)
Cisplatin-Capecitabine9 (8.7)
Other12 (11.5)
Adjuvant RT
Present39 (37.5)
Absent65 (62.5)
Recurrence
Present23 (22.1)
Absent81 (77.9)
Location of recurrence
Lungs3 (2.9)
Brain1 (1.0)
Liver6 (5.8)
Bones1 (1.0)
Local3 (2.9)
Ovaries2 (1.9)
Peritoneum4 (3.8)
Diffuse3 (2.9)
Survival status
Death34 (32.7)
Survival70 (67.3)

Values are presented as mean (range) or number (%).

CT, chemotherapy; RT, radiotherapy; Folfox, folinic acid (leucovorin, FOL), fluorouracil (5-FU, F), oxaliplatin (Eloxatin, OX); FLOT, fluorouracil (5-FU, F), leucovorin (L), oxaliplatin (O), docetaxel (D); Capox, capecitabine and oxaliplatin.



The pathological parameters of the patients in the high-TB and low-TB groups are demonstrated in Table 3. As the tumor size increased, the proportion of patients with high TB was significantly higher (P = 0.018). As the T stage (P = 0.003) and N stage (P = 0.001) of the tumor increased, high TB was observed significantly more frequently. Grade III tumors were more common in the high-TB group (P = 0.03) (Table 3).

Table 3 . Pathological Parameters and Tumor Budding.

CharacteristicLow budding (n = 62)High budding (n = 42)P-value
Tumor size (cm)4.9 (2.5–7.3)6.1 (3.7–8.5)0.018*
T stage
I9 (14.5)1 (2.4)0.003***
II11(17.7)3 (7.1)
III30 (48.4)17 (40.5)
IV12 (19.4)21 (50.0)
N stage
I25 (40.3)3 (7.1)0.001***
II12 (19.4)3 (7.1)
III11 (17.7)16 (38.1)
IV14 (22.6)20 (47.6)
Grade
I9 (14.5)1 (2.4)0.030***
II36 (58.1)21 (50.0)
III17 (27.4)20 (47.6)
Lymphovascular invasion
Present40 (64.5)32 (76.2)0.206***
Absent22 (35.5)10 (23.8)
Perineural invasion
Present34 (54.8)32 (76.2)0.027***
Absent28 (45.2)10 (23.8)
Location of tumor
Cardia14 (22.6)10 (23.8)0.988**
Antrum19 (30.6)15 (35.7)
Corpus21 (33.9)12 (28.6)
Incisura angularis2 (3.2)1(2.4)
Diffuse4 (6.5)3 (7.1)
Fundus2 (3.2)1 (2.4)

Values are presented as mean (range) or number (%).

*t test; **Fisher exact test; ***Chi-square test (P < 0.05).



In clinical practice, patients with positive surgical margins or with T3-4 tumors and lymph node involvement receive RT. Consistent with the fact that TB increased as the T and N stages increased, high TB was found to be significantly more common in patients who received adjuvant RT (P = 0.03).

In the OS analysis performed in 85 patients, excluding the patients who died due to non-disease-related causes during follow-up (19 patients), the median OS was 58 months (95% confidence interval [CI], 5.9–110.5 months) in the high-TB (Fig. 1), while the median OS could not be calculated in the low-TB group (Fig. 2). OS was significantly longer in the low-TB group than in the high-TB group (P = 0.038). The median disease-free survival (DFS) time predicted in the analysis of 85 patients was not reached in the group with low TB, while the median DFS was 49 months in the group with high TB (95% CI, 21.7–76.3 months). However, the difference did not reach statistical significance due to the small number of patients and the small number of events (P = 0.135).

In the univariate survival analysis, high TB, age at diagnosis over 60 years, low BMI, high N stage, the presence of LVI, and recurrence were found to have significant effects on OS. High TB was identified as an independent risk factor predicting mortality in the multivariate model, and age at diagnosis over 60 years and the presence of LVI had significant associations with OS and recurrence (Fig. 3).

Figure 3. Cumulative survival analysis between the high and low budding groups. CI, confidence interval.

The first step in the development of metastasis is the detachment of tumor cells from the primary lesion and invasion of stromal tissue. TB, which is thought to constitute the basis for invasion and metastasis, has been defined as the presence of single malignant cells on the invasive surface of the tumor or the presence of undifferentiated malignant cell clusters consisting of 2–4 cells, called tumor buds, in the tumor stroma.12 Various methods of evaluating TB have been described. The most common method used in the past was scanning several areas and calculating the average number of buds.13,14 In our study, to count tumor buds, we used an area of 0.785 mm2 in the region where TB was most intense in hematoxylin and eosin-dyed preparations (i.e., the “hotspot”). TB has not yet become a standard prognostic marker in gastric cancer due to variations in definitions and evaluation methods.15

According to the UICC consensus report, TB is considered an independent negative prognostic factor in colorectal cancer. In addition, TB was reported to be associated with higher TNM stage and higher tumor grade, and it was recommended that TB should be routinely taken into consideration for treatment plans of colorectal cancer and should be included in current guidelines.9 TB has also shown associations with a poor prognosis in various epithelial cancer types such as breast cancer, laryngeal squamous cell carcinoma, lung adenocarcinoma, esophageal cancer, pancreatic adenocarcinoma, endometrial carcinoma, and gallbladder cancer.5,1621

In their systematic review, Guo et al20 analyzed seven studies including 2,187 patients. According to this review, high TB is associated with unfavorable prognosis, advanced stage, a more aggressive course, more LVI, and more lymph node involvement.22 In our study, which supports these data, as the tumor size increased, the proportion of high TB was found to be significantly higher (P = 0.018). High TB was also significantly more common in tumors with a higher T stage (P = 0.003), N stage (P < 0.001), and grade (P = 0.03).

Ulase et al21 found that TB in intestinal-type gastric cancer was also correlated with histological grade, T and N stage, disease stage, LVI, and PNI.23 In our study, a similar relationship was found between TB and T stage, N stage, grade, and PNI. LVI was seen at a higher rate in the high-TB group (76%) than in the low-TB group, but the difference was not statistically significant (P = 0.206).

Although the 5-year OS rate exceeds 95% in early-stage gastric cancers, some patients experience recurrence after curative treatment. Additional predictive and directive prognostic parameters such as TB are essential, especially in T1N0 and T2N0 tumors, for which the role of adjuvant CT is not clear.24

LVI plays an important role among the known risk factors for lymph node metastasis in early-stage gastric cancers.25,26 In their study on early-stage gastric cancers, Du et al25 found that the association between LVI and TB was significantly higher in patients with lymph node metastasis than in those without lymph node metastasis. The authors reported that LVI and TB were the most important risk factors in submucosal lesions. With the results of more comprehensive studies in the future, additional surgical resection, including lymph node dissection, may be considered if these risk factors are detected after the endoscopic resection of early-stage cancers.27 Similarly, Olsen et al26 found statistically significant associations between TB and recurrence, differentiation, LVI, and the T and N stages of tumors.

Gabbert et al27 concluded that TB was a significant negative prognostic factor for 5-year OS, especially in patients with intestinal-type adenocarcinoma. Likewise, in a study by Kemi et al,28 the 5-year OS rates were found to be significantly lower in the high-TB group (23.0%) than in the low-TB group (41.0%). In our study, the relapse rate and time to recurrence did not differ significantly between the low-TB and high-TB groups (P = 0.732). However, the predicted OS was significantly shorter in the high-TB group than in the low-TB (P = 0.038). Similarly, the predicted DFS of the high-TB group was shorter than that of the low-TB group. However, this difference did not reach statistical significance due to the small number of patients and events (P = 0.135).

In their univariate analysis, Zhang et al29 found that high TB was associated with poor prognosis (e.g., larger tumor size and lymph node involvement). In their multivariate analysis, they found that TB and positive nodal metastases were independent negative prognostic factors in gastric adenocarcinoma.30 In contrast to this study, Ulase et al21 found no effect of TB on OS in multivariate analysis. In the univariate analysis of our study, a high TB score, being over 60 years old, low BMI, high N stage, presence of LVI, and recurrence had significant effects on OS.

TB has a similar mechanism to the epithelial-mesenchymal transition. Loss of expression of E-cadherin and β-catenin, which are known as adhesion molecules, has been observed in cells with TB.15 Therefore, agents targeting the epithelial-mesenchymal transition pathway in patients with high TB may have a place in their treatment in the future.

The limitations of the study

The limitations of our study are the relatively small number of patients and the single-center design. Due to the short follow-up time and few events, a longer follow-up period would be required to obtain more robust findings.

Conclusion

In our study, a high TB rate was found to be associated with tumor size, T stage, N stage, and high tumor grade, supporting data available in the literature. In addition, PNI was found more frequently in the high-TB group. The predicted OS was also significantly shorter in the high-TB group than in the low-TB group. These findings suggest that TB may be a poor prognostic marker in gastric cancer, as well as in colorectal cancer. If more comprehensive studies are conducted and a standard method is established for the detection of TB, we believe that TB will be used to guide treatment decisions and predict patients’ prognoses.

No potential conflict of interest relevant to this article was reported.

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, 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. Erratum in: CA Cancer J Clin. 2020;70:313.
    Pubmed CrossRef
  2. Song Z, Wu Y, Yang J, Yang D, Fang X. Progress in the treatment of advanced gastric cancer. Tumour Biol. 2017;39:1010428317714626.
    Pubmed CrossRef
  3. Ikoma N, Blum M, Chiang YJ, Estrella JS, Roy-Chowdhuri S, Fournier K, et al. Survival rates in T1 and T2 gastric cancer: a Western report. J Surg Oncol. 2016;114:602-6.
    Pubmed CrossRef
  4. Yamaguchi Y, Ishii G, Kojima M, Yoh K, Otsuka H, Otaki Y, et al. Histopathologic features of the tumor budding in adenocarcinoma of the lung: tumor budding as an index to predict the potential aggressiveness. J Thorac Oncol. 2010;5:1361-8.
    Pubmed CrossRef
  5. Imai T. Growth patterns in human carcinoma. Their classification and relation to prognosis. Obstet Gynecol. 1960;16:296-308.
  6. Morodomi T, Isomoto H, Shirouzu K, Kakegawa K, Irie K, Morimatsu M. An index for estimating the probability of lymph node metastasis in rectal cancers. Lymph node metastasis and the histopathology of actively invasive regions of cancer. Cancer. 1989;63:539-43.
    Pubmed CrossRef
  7. Ueno H, Murphy J, Jass JR, Mochizuki H, Talbot IC. Tumour 'budding' as an index to estimate the potential of aggressiveness in rectal cancer. Histopathology. 2002;40:127-32.
    Pubmed CrossRef
  8. Lugli A, Kirsch R, Ajioka Y, Bosman F, Cathomas G, Dawson H, et al. Recommendations for reporting tumor budding in colorectal cancer based on the International Tumor Budding Consensus Conference (ITBCC) 2016. Mod Pathol. 2017;30:1299-311.
    Pubmed CrossRef
  9. Quirke P, Risio M, Lambert R, von Karsa L, Vieth M; International Agency for Research on Cancer. European guidelines for quality assurance in colorectal cancer screening and diagnosis. First Edition--Quality assurance in pathology in colorectal cancer screening and diagnosis. Endoscopy. 2012;44(Suppl 3):SE116-30.
    Pubmed CrossRef
  10. Kazama S, Watanabe T, Ajioka Y, Kanazawa T, Nagawa H. Tumour budding at the deepest invasive margin correlates with lymph node metastasis in submucosal colorectal cancer detected by anticytokeratin antibody CAM5.2. Br J Cancer. 2006;94:293-8.
    Pubmed KoreaMed CrossRef
  11. STROBE Statement. Strengthening the reporting of observational studies in epidemiology. What is strobe? STROBE Website. Available from: http://www.strobe-statement.org/. Published 2019. Accessed September 12, 2019.
  12. Che K, Zhao Y, Qu X, Pang Z, Ni Y, Zhang T, et al. Prognostic significance of tumor budding and single cell invasion in gastric adenocarcinoma. Onco Targets Ther. 2017;10:1039-47.
    Pubmed KoreaMed CrossRef
  13. Tanaka K, Shimura T, Kitajima T, Kondo S, Ide S, Okugawa Y, et al. Tropomyosin-related receptor kinase B at the invasive front and tumour cell dedifferentiation in gastric cancer. Br J Cancer. 2014;110:2923-34.
    Pubmed KoreaMed CrossRef
  14. Liang F, Cao W, Wang Y, Li L, Zhang G, Wang Z. The prognostic value of tumor budding in invasive breast cancer. Pathol Res Pract. 2013;209:269-75.
    Pubmed CrossRef
  15. Koyuncuoglu M, Okyay E, Saatli B, Olgan S, Akin M, Saygili U. Tumor budding and E-Cadherin expression in endometrial carcinoma: are they prognostic factors in endometrial cancer? Gynecol Oncol. 2012;125:208-13.
    Pubmed CrossRef
  16. O'Connor K, Li-Chang HH, Kalloger SE, Peixoto RD, Webber DL, Owen DA, et al. Tumor budding is an independent adverse prognostic factor in pancreatic ductal adenocarcinoma. Am J Surg Pathol. 2015;39:472-8.
    Pubmed CrossRef
  17. Almangush A, Salo T, Hagström J, Leivo I. Tumour budding in head and neck squamous cell carcinoma - a systematic review. Histopathology. 2014;65:587-94.
    Pubmed CrossRef
  18. Brown M, Sillah K, Griffiths EA, Swindell R, West CM, Page RD, et al. Tumour budding and a low host inflammatory response are associated with a poor prognosis in oesophageal and gastro-oesophageal junction cancers. Histopathology. 2010;56:893-9.
    Pubmed CrossRef
  19. Kai K, Kohya N, Kitahara K, Masuda M, Miyoshi A, Ide T, et al. Tumor budding and dedifferentiation in gallbladder carcinoma: potential for the prognostic factors in T2 lesions. Virchows Arch. 2011;459:449-56.
    Pubmed CrossRef
  20. Guo YX, Zhang ZZ, Zhao G, Zhao EH. Prognostic and pathological impact of tumor budding in gastric cancer: a systematic review and meta-analysis. World J Gastrointest Oncol. 2019;11:898-908.
    Pubmed KoreaMed CrossRef
  21. Ulase D, Heckl S, Behrens HM, Krüger S, Röcken C. Prognostic significance of tumour budding assessed in gastric carcinoma according to the criteria of the International Tumour Budding Consensus Conference. Histopathology. 2020;76:433-46.
    Pubmed CrossRef
  22. Koelzer VH, Langer R, Zlobec I, Lugli A. Tumor budding in upper gastrointestinal carcinomas. Front Oncol. 2014;4:216.
    CrossRef
  23. Fang C, Shi J, Sun Q, Gold JS, Xu GF, Liu WJ, et al. Risk factors of lymph node metastasis in early gastric carcinomas diagnosed by WHO criteria in 379 Chinese patients. J Dig Dis. 2016;17:526-37.
    Pubmed CrossRef
  24. Gotoda T, Yanagisawa A, Sasako M, Ono H, Nakanishi Y, Shimoda T, Kato Y. Incidence of lymph node metastasis from early gastric cancer: estimation with a large number of cases at two large centers. Gastric Cancer. 2000;3:219-25.
    Pubmed CrossRef
  25. Du M, Chen L, Cheng Y, Wang Y, Fan X, Zhang Y, et al. Tumor budding and other risk factors of lymph node metastasis in submucosal early gastric carcinoma: a multicenter clinicopathologic study in 621 radical gastrectomies of Chinese patients. Am J Surg Pathol. 2019;43:1074-82.
    Pubmed CrossRef
  26. Olsen S, Jin L, Fields RC, Yan Y, Nalbantoglu I. Tumor budding in intestinal-type gastric adenocarcinoma is associated with nodal metastasis and recurrence. Hum Pathol. 2017;68:26-33.
    Pubmed CrossRef
  27. Gabbert HE, Meier S, Gerharz CD, Hommel G. Tumor-cell dissociation at the invasion front: a new prognostic parameter in gastric cancer patients. Int J Cancer. 1992;50:202-7.
    Pubmed CrossRef
  28. Kemi N, Eskuri M, Ikäläinen J, Karttunen TJ, Kauppila JH. Tumor budding and prognosis in gastric adenocarcinoma. Am J Surg Pathol. 2019;43:229-34.
    Pubmed CrossRef
  29. Zhang N, Wang D, Duan Y, Ayarick VA, Cao M, Wang Y, et al. The special immune microenvironment of tumor budding and its impact on prognosis in gastric adenocarcinoma. Pathol Res Pract. 2020;216:152926.
    Pubmed CrossRef
  30. Grigore AD, Jolly MK, Jia D, Farach-Carson MC, Levine H. Tumor budding: the name is EMT. Partial EMT. J Clin Med. 2016;5:51.
    Pubmed KoreaMed CrossRef