Orginal Research
2023 December
Volume : 11 Issue : 4


Tumor budding in colorectal adenocarcinoma using cytokeratin 20 immunostaining

Chaitra N, Sundar RNS, Kumar HKS, Nataraju G, Esaivani KB

Pdf Page Numbers :- 285-289

Chaitra N1, Naveen Shyam Sundar R2, Sharath Kumar HK3, Nataraju G3 and Esaivani KB3,*

 

1Department of Pathology, Sri Siddhartha Medical College, Tumkur, Karnataka-572107, India

2Department of Community Medicine, Dr. Shushila Nayar School of Public health, Mahatma Gandhi Institute of Medical Sciences, Wardha, Maharashtra 442102, India

3Department of Pathology, Mysore Medical College and Research Institute, Mysore Karnataka 570001, India

 

*Corresponding author: Dr. Esaivani KB, Postgraduate student, Department of Pathology, Mysore Medical College and Research Institute, Mysore Karnataka 570001, India. Email: esaishanu@gmail.com

 

Received 1 July 2023; Revised 21 August 2023; Accepted 31 August 2023; Published 12 September 2023

 

Citation: Chaitra N, Sundar RNS, Kumar HKS, Nataraju G, Esaivani KB. Tumor budding in colorectal adenocarcinoma using cytokeratin 20 immunostaining. J Med Sci Res. 2023; 11(4):285-289. DOI: http://dx.doi.org/10.17727/JMSR.2023/11-53

 

Copyright: © 2023 Chaitra N et al. Published by KIMS Foundation and Research Center. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.



Abstract

Introduction: Colorectal cancer is the third most common cancer worldwide and second most common fatal cancer. Tumor budding is an emerging prognostic indicator in cancers. Though tumor budding can be assessed using Hematoxylin and Eosin stain, sometimes peritumoral inflammation and reactive stromal fibrosis obscures the tumor buds. The present study aimed to assess tumor budding using cytokeratin 20 immunohistochemistry in colorectal cancers.

Methods: This is a cross sectional study which included 30 cases of colorectal carcinomas. cytokeratin 20 stained slides of colorectal carcinoma were assesed for tumor budding using International Tumor Budding Consensus Conference (ITBCC) 2016 consensus criteria and compared with AJCC cancer stage.

Results: Demographic analysis showed peak incidence of colorectal cancer in the age group of 55-64 years (33.3%) and male: female ratio of 1.14:1. Majority of the tumors with score 1(23.3%) showed stage I. While tumors with score 2 had similar incidence of stage I (16.6%) and II (16.6%) and lesser incidence of stage III (3.33%). Most of the tumors with score 3 tumor budding had stage III tumors (26.67%). The p value is < 0.0001, which is statistically significant. Tumor budding signifies the biological and molecular phenomenon of epithelial-mesenchymal transition in the tumor microenvironment. Loss of E-cadherin, alterations in transcription factors including SNAIL, ZEB, TWIST etc and switching of CMS2 to CMS4 are some of the recorded molecular profiles responsible for tumor budding.

Conclusion: This study concludes that the tumor stage increases with tumor budding and thus is a reliable marker in predicting the prognosis. This study also states that immunohistochemical study gives objective scoring of tumor buds in colorectal cancers.

 

Keywords: tumor budding; colorectal cancer; cytokeratin 20; prognostic marker

Full Text

Introduction

Colorectal cancer contributes to a global health problem having a significant disease incidence and mortality. It is the third most common cancer worldwide and accounts to second most common fatal cancer [1]. The occurrence of colon cancer among younger adults has been increased in the recent years [2]. Increase in the modifiable risk factors has contributed to increase in sporadic colon cancers (approximately 75%) [3]. There are various modifiable risk factors having deleterious effect on the incidence of colorectal cancer, including smoking [4], obesity [5], sedentary lifestyle and physical inactivity [6] and poor dietary habits [7].

 

Staging of colorectal cancers using TNM system remains the gold standard for prognostification of these tumors [8]. Furthermore, some of the histopathological features have independent prognostic value in colorectal cancers including histological subtype [9], lymphovascular invasion [10], number of tumor positive lymphnodes collecting at least 12 lymphnodes [11], perineural invasion [12] and tumor budding [13]. Though molecular markers of prognostic significance in colorectal cancers are available [10], the histopathological factors can be considered as cost-effective and widely usable parameters in risk assessment.

 

Tumor budding is an emerging prognostic indicator, which is defined as single or a cluster of ≤ 4 de-differentiated tumor cells present at the invasive tumor front [14]. Tumor budding can be identified at the tumor center called as intratumoral budding, or at the invasive front which are known as peritumoral budding [15]. Tumor buds are formed due to the Epithelial-Mesenchymal transition in the tumor microenvironment [14]. International Tumor Budding Consensus Conference (ITBCC) 2016 consensus recommends the assessment of peritumoral budding at the invasive front at 20x magnification in a field that has maximum density of buds. These can be further classified as low (0-4 buds/20x), intermediate (5-9 buds/20x) and high (>9 buds/20x) [15]. This stratification of tumor budding is chiefly salient in stage I and stage II colorectal cancers [16-18]. Even though patients with TNM stage I are expected to have best prognosis, some tumors have aggressive course. Thus, patients having increased number of microscopic risk factors for tumor spread (including tumor budding) on biopsy specimen, must be considered for oncological resection [16]. Stage II colorectal cancer patients with high tumor budding have increased risk of metastasis and mortality [17]. Thus, these patients should be considered for adjuvant therapy [16].

 

Though tumor budding can be assessed using Hematoxylin and Eosin stain, in some cases peritumoral inflammation and reactive stromal fibrosis obscures the tumor buds [14, 20]. In addition, the results are subjective and has increased interobserver variability [19]. Thus, the present study aimed to assess the Tumor budding using Cytokeratin 20 immunohistochemistry in colorectal cancers.

 

Materials and methods

This is a cross sectional descriptive study done in the Department of Pathology, Mysore Medical college and research institute, Mysore during the period of 6 months from December 2019 to May 2020. Approval for conducting the study was obtained from the Institutional Ethical committee. The present study included 30 cases of colorectal carcinomas, including resected specimen of colorectal cancers. We have excluded the cases which had extensive necrosis and had undergone prior adjuvant therapy. Routine grossing and histopathological examination of the surgical specimen was done as per the standard protocol. Additional 5 micron thick sections were taken on charged slides and cytokeratin 20 immunostain was done. Cytokeratin 20 stained slides of colorectal carcinoma cases were utilised in the present study to assess tumor budding.

 

A single or group of less than 5 cells stained with cytokeratin 20 were counted as tumor buds. ITBCC 2016 consensus criteria was used to analyse and count the tumor budding. The slides were initially scanned under low power (10x) fields to identify the field with maximum density of buds. Subsequently, the number of buds was counted at 20x objective lens as per the criteria. Three-tiered system was used for stratification of the tumor buds into low/BD1 - 0-4 buds, intermediate/BD2- 5-9 buds and high/BD3- >9 buds per 20x field. Results were tabulated and compared with the stage of the tumor.

 

Statistical analysis

All the data were entered in Microsoft Excel and analyzed using statistical software R version 4.2.2. The qualitative study variables were expressed in frequency with percentages. To find the significant difference Yates Chi-square test is used since the frequencies were ≤ 5. p value of <0.05 is considered statistically significant.

 

Results

The study was conducted in the Department of Pathology of our institute, in which 30 specimen of colorectal cancer have been included. Demographic results were analysed. In the present study, peak incidence of colorectal cancer was in the age group of 55-64 years (33.3%) and least was in younger age group of 35-44 years and older than 75 years (6% each). Gender wise distribution suggests that the males were slightly more affected than females with Male: female ratio of 1.14:1 (Table 1).

 

The H&E stained slides were studied and all the 30 cases (100%) were reported as Adenocarcinoma. Majority of the tumors were of moderately differentiated grade 2 (56.6%), followed by grade 1 (40%) and only 1/30 (3.3%) was graded as poorly differentiated adenocarcinoma. Pathological TNM staging was done according to AJCC 8th edition. Majority of the cases, 12/30 (40%) belonged to stage I followed by stage II and III (30% each) (Table 1).

 

Table 1: Demographic and histomorphological data of tumor budding in colorectal cancer.  

Descriptive analysis

Adenocarcinoma (N = 30)

Age

 

35-44

2

 

45-54

7

 

55-64

10

 

65-74

9

 

75-84

2

Gender

 

F

14

 

M

16

Tumour grade

 

1

12

 

2

17

 

3

1

Stage

 

I

12

 

II

9

 

III

9

CK 20 score

 

1

7

 

2

11

 

3

12

Tumour budding score

 

1

9

 

2

11

 

3

10

 

The CK 20 immuno-stained slides were examined under 20x field and scored according to three scaled scoring system.

Score 0 = no staining or nonspecific staining of tumor cells.

Score 1+ = weak and incomplete staining of more than 10% of tumor cells.

Score 2+ = moderate and complete staining of more than 10% of tumor cells.

Score 3+ = strong and complete staining of more than 10% of tumor cells.

 

The number of tumor buds were counted using CK20 immunostaining. The tumor buds were grouped into three tiered groups as per ITBCC 2016 consensus criteria. Low tumor budding (score 1) was seen in 9/30 cases (30%) (Figure 1), 11/30 (36.6 %) were scored as intermediate (score 2) (Figure 2) which contributes to the majority of cases, and 10/30(33.3%) showed high tumor budding (Figure 3).

 

Figure 1: Low tumor budding. 20x; IHC-CK20, Shows tumor front.

 

Figure 2: Intermediate tumor budding, 20x; IHC-CK20.

 

Figure 3: High tumor budding, 20x; IHC-CK20.

 

The tumor bud scoring was compared with the known prognostic parameter i.e, tumor staging. Majority of the tumors with score 1 (23.3%) showed stage I tumor. While tumors with score 2 had similar incidence of Stage I (16.6%) and II (16.6%) and lesser incidence of stage III (3.33%). Most of the tumors with score 3 tumor budding had stage III tumors (26.67%). The p value is < 0.0001, which is statistically significant. AJCC and TNM staging of tumors include lymphnodal metastasis also, thus these results imply that the tumor budding score increases with increase in lymph nodal metastasis and in turn results in upstaging of the tumor (Table 2).

 

Table 2: Correlation between tumor budding score with the tumor stage.  

Tumour budding score

Tumor stage

p value

I (%)

II (%)

III (%)

1

23.33

6.67

0.00

<0.0001*

2

16.67

16.67

3.33

3

0.00

6.67

26.67

p value < 0.05 is significant, Chi square test of significance.

 

Discussion

Tumor budding has been a novel prognostic marker used in various organs e.g., breast [21], laryngeal squamous cell carcinoma [22] and in various other solid cancers [23]. Tumor budding signifies the biological and molecular phenomenon of epithelial-mesenchymal transition in the tumor microenvironment [23]. Many scoring systems have been proposed for the analysis of tumor budding, including Choi et al [24], Prall et al [25], Lugli et al [26], Zlobec et al [27] etc.

 

A review done by Lugli et al [21], catalogued various criterias to count the tumor budding, which include the following: (1) ≥10 buds in a single field (20x objective) on H&E (10/24, 42%), (2) ≥5 buds in a single field (20x objective) on H&E (3/24, 13%), (3) 10 HPF method’ using IHC (≥10 HPF, 40x objective) (0/24, 0%), and (4) No cut-off used (prefer continuous scale, reporting buds/HPF) (4/24, 17%).

 

However, the recent ITBCC consensus criteria are been accepted and widely used in reporting of tumor budding scoring.

 

Tumor budding results from Epithelial-mesenchymal transition (EMT) which is a result of various molecular alterations. Loss of E-cadherin, alterations in transcription factors including SNAIL, ZEB, TWIST etc and switching of CMS2 to CMS4 are some of the recorded molecular profiles responsible for tumor budding [30].

 

The results of present study were compared with the studies done by other pioneers. The age incidence of CRC correlated well with Zlobec et al being more prevalent in elderly patients with a median age of 64 years. Also, they documented a significant association between high-grade tumor budding and a lack of objective response in patients with metCRC treated with anti-EGFR therapies [27].

 

Prall et al in their retrospective study identified that majority of patients reported with high tumor budding score had metastatic disease in future compared to those reported as low tumor budding score [25]. In a study conducted by Rieger et al., tumor budding counts showed significant relation with advanced T -stage, presence of nodal metastasis, lymphatic invasion, venous invasion, tumour grade, independent of the location of assessment. This is comparable with our study [28]. Wang et al., in their study related tumor budding with epithelial–mesenchymal transition and appears to be an independent prognostic factor [29]. Our study correlated with the study done by Kumarguru et al., where 60% of patients had high tumor budding score and 40% of patients had low tumor budding score [21].

 

Limitations: This study has been conducted on a small number of samples retrieved at our institution. Comparison with routine hematoxylin and eosin staining could not be done due to extensive stromal reaction and inflammationin few cases. Large sample size and multicentre studies are required to confirm these results.

 

Conclusion

This study concludes that the tumor stage increases with tumor budding score, and thus is a reliable marker in predicting the prognosis. This study also states that immunohistochemical evaluation gives objective scoring of tumor buds compared to routine hematoxylin and eosin staining. Tumor budding is a reliable and cost effective indicator of spread of cancer and can be a futuristic prognostic marker. We recommend the utility of this parameter in routine reporting of colorectal cancers.

 

Acknowledgements

Staff and technician Mrs. Meenakshi for their support during the study.

 

Conflicts of interest

Authors declare no conflicts of interest.

 

References

[1] Hossain MS, Karuniawati H, Jairoun AA, Urbi Z, Ooi DJ, et al. Colorectal cancer: A review of carcinogenesis, global epidemiology, current challenges, risk factors, preventive and treatment strategies. Cancers. 2022; 14:1732.

 

[2] O’Connell JB, Maggard MA, Liu JH, Etzioni DA, Livingston EH, et al. Rates of colon and rectal cancers are increasing in young adults. Am Surg TM. 2003; 69:866–872.

 

[3] GBD 2019 colorectal cancer collaborations. Global, regional, and national burden of colorectal cancer and its risk factors, 1990-2019: A systematic analysis for the global burden of disease study 2019. Lancet Gastroent Hepatol. 2022; 7:627–647.

 

[4] Gram IT, Park SY, Wilkens LR, Haiman CA, Marchand LL. Smoking-related risks of colorectal cancer by anatomical subsite and sex. Am J Epidemiol. 2020; 189:543–553.

 

[5] Ma Y, Yang Y, Wang F, Zhang P, Shi C, el al. Obesity and risk of colorectal cancer: A systematic review of prospective studies. Plos One. 2013; 8:e0053916.

 

[6] Namasivayam V, Lim S. Recent advances in the link between physical activity, sedentary behaviour, physical fitness, and colorectal cancer. F1000Research. 2017; 199:1–8.

 

[7] Fernandez-Villa T, Alvarez-Alvarez L, Rubin-Garcia M, Obon-Santacan M, Moreno V. The role of dietary patterns in colorectal cancer: a 2019 update. Expert Rev Gastroenterol Hepatol. 2020; 14:281–290.

 

[8] Amin MB, Edge S, Greene F, Byrd DR, Brookland RK, et al. AJCC cancer staging manual (8th edition). Springer international publishing: American Joint Commission on Cancer. 2017; 251–394.

 

[9] Wu X, Lin H, Li S. Prognoses of different pathological subtypes of colorectal cancer at different stages: A population-based retrospective cohort study. BMC Gastroenterol. 2019; 19:164–172.

 

[10] Marzouk O, Schofield J. Review of histopathological and molecular prognostic features in colorectal cancer. Cancers. 2011; 3:2767–2810.

 

[11] Fortea-Sanchis C, Martinez-Ramos D, Escrig-Sos J. The lymph node status as a prognostic factor in colon cancer: comparative population study of classifications using the logarithm of the ratio between metastatic and nonmetastatic nodes (LODDS) versus the pN-TNM classification and ganglion ratio systems. BMC Cancer. 2018; 18:1208–1219.

 

[12] Leibig C, Ayala G, Wilks J, Verstovek G, Liu H, et al. Perineural invasion is an independent predictor of outcome in colorectal cancer. J Clin Oncol. 2009; 27:5131–5137.

 

[13] Graham RP, Vierkant RA, Tillman LS, Wang AH, Laird PW, et al. Tumor budding in colorectal carcinoma: Confirmation of prognostic significance and histologic cutoff in a population-based cohort. Am J Surg Pathol. 2015; 39:1340–1346.

 

[14] Koelzer VH, Zlobec I, Lugli A. Tumor budding in colorectal cancer-ready for diagnostic practice?. Hum Pathol. 2016; 47:4–19.

 

[15] Lugli A, Kirsch R, Ajioka Y, Bosman F, Cathomas G, et al. Recommendations for reporting tumor budding in colorectal cancer based on the International Tumor Budding Consensus Conference (ITBCC) 2016. Mod Pathol. 2017; 30:1299–1311.

 

[16] Zlobec I, Berger MD, Lugli A. Tumour budding and its clinical implications in gastrointestinal cancers. Br J Can. 2020. 123:700–708.

 

[17] Garfinkle R, Lee L, Boutros M, Cardin MJ, Spartz A, et al. Tumor budding predicts increased recurrence after curative resection for T2N0 colorectal cancer. J Can Chir. 2019; 62:334–339.

 

[18] Kazuo H, Clayton S, Denise J, Michael T, Mark V. Prognostic value of tumor “budding” in patients with colorectal cancer. ASCRS; Dis Colon Rectum. 1993; 36:627–635.

 

[19] Koezler VH, Zlobec I, Berger MD, Cathomas G, Dawson H, et al. Tumor budding in colorectal cancer revisted: results of a multicenter interobserver study. Virchows Arch. 2015; 466:485–493.

 

[20] Ozturk SK, Haddad TS, Zlobec I, Lugli A, Nagtegaal ID. Attackers and defenders: tumor buds and lymphocytes as morphological biomarkers in colorectal cancer. Diag Histopathol. 2022; 28:480–487.

 

[21] Kumarguru BN, Rangaswamy AS, Shaik S, Karri A, Srinivas VS, et al. Tumor budding in invasive breast cancer- An indispensable budding touchstone. Indian J Pathol Microb. 2020; 63:117–122.

 

[22] Zhang H, Sheng X, Zhang S, Gu X. The prognostic value of tumor budding in laryngeal squamous cell carcinoma. Transl Can Res. 2020; 9:119–127.

 

[23] Lugli A, Zlobec I, Berger MD, Kirsch R, Nagtegaal ID. Tumour budding in solid cancers. Nat Rev Clin Oncol. 2021; 18:101–115.

 

[24] Choi HJ, Park KJ, Shin JS, Roh MS, Kwon HC, et al. Tumor budding as a prognostic marker in stage-III rectal carcinoma. Int J Colorectal Dis 2007; 22:863–868.

 

[25] Prall F, Nizze H, Barten M. Tumour budding as prognostic factor in stage I/II colorectal carcinoma. Histopathology. 2005; 47:17–24.

 

[26] Lugli A, Karamitopoulou E, Panayiotides I, Karakitsos P, Rallis G, et al. CD8+ lymphocytes/tumour-budding index: an independent prognostic factor representing a ‘pro-/anti-tumour’ approach to tumour host interaction in colorectal cancer. Br J Cancer. 2009; 101:1382–1392.

 

[27] Zlobec I, Molinari F, Martin V, Mazzucchelli L, Saletti P, et al. Tumor budding predicts response to anti-EGFR therapies in metastatic colorectal cancer patients. World J Gastroenterol. 2010; 16:4823–4831.

 

[28] Rieger G, Koelzer VH, Dawson HE, Berger MD, Hädrich M, et al. Comprehensive assessment of tumour budding by cytokeratin staining in colorectal cancer. Histopathology. 2017; 70:1044–1051.

 

[29] Wang C, Huang H, Huang Z, Wang A, Chen X, et al. Tumor budding correlates with poor prognosis and epithelial-mesenchymal transition in tongue squamous cell carcinoma. J Oral Pathol Med. 2011; 40:545–551.

 

[30] Smedt LD, Palmans S, Andel D. Expression profiling of budding cells in colorectal cancer reveals an EMT-like phenotype and molecular subtype switching. Br J Cancer. 2017; 116:58–65.

 

Subscription