Clinicopathological Significance of SMAD4 Expression in Breast Cancer

Article information

J Breast Dis. 2019;7(2):52-58
Publication date (electronic) : 2019 December 31
doi : https://doi.org/10.14449/jbd.2019.7.2.52
1Department of Surgery, College of Medicine, Hanyang University, Seoul, Korea
2Department of Pathology, College of Medicine, Hanyang University, Seoul, Korea
Correspondence: Min Sung Chung, M.D., Ph.D. Department of Surgery, Hanyang University Medical Center, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea Tel: +82-2-2290-8460, Fax:+82-2-2281-0224, E-mail: bovie@hanyang.ac.kr
Received 2019 August 5; Revised 2019 September 25; Accepted 2019 October 23.

Abstract

Purpose

SMAD4 is a member of the SMAD family and acts as a central mediator of transforming growth factor beta signaling. Little is known about SMAD4 expression and its prognostic significance in breast cancer. We evaluated the clinicopathological and prognostic significance of SMAD4 expression in breast cancer.

Methods

Two hundred and fifty-five patients with invasive ductal carcinoma of the breast from 2000 to 2008 were retrospectively analyzed. We investigated SMAD4 expression using a tissue microarray-based immunohistochemical assay and evaluated the association between SMAD4 and prognosis of breast cancer.

Results

High SMAD4 expression was positively associated with early stage (p=0.009), estrogen receptor positivity (p=0.026), and human epidermal growth factor receptor 2 negativity (p=0.001). A significant difference in overall survival (OS) was associated with high SMAD4 expression in patients with T1 stage tumors (hazard ratio: 0.459, p=0.024).

Conclusion

High SMAD4 expression was correlated with several favorable prognostic factors and was associated with favorable OS in T1 stage breast cancer. SMAD4 in breast cancer has potential prognostic significance, and further investigations and understanding about SMAD4 expression are needed.

INTRODUCTION

Breast cancer is among the most common malignancies and a leading cause of death in women [1]. However, advances in surgical techniques and systemic therapies have improved the survival of breast cancer patients in recent decades [2]. Breast cancer is a heterogeneous disease with a variety of pathologic and molecular features. The prognosis of breast cancer and therapeutic decision-making are known to depend on classic immunohistochemistry (IHC) markers, namely estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Recently, genomic assays have been used to provide information and to help in decision-making for adjuvant treatment [3]. Differential expression of various proteins has been researched to develop more efficient options for diagnosis and treatment.

SMAD4 is a major downstream mediator in the signaling of the transforming growth factor beta (TGF-β) pathway. The TGF-β signaling process is initiated when TGF-β binds to the homodimers of the TGF-β type II receptor (TβRII) on the cell surface. TβRII recruits and activates the TGF-β type I receptor (TβRI). The activated TβRI phosphorylates SMAD2 or SMAD3, which heterodimerizes with SMAD4. These complexes translocate into the nucleus where they bind to DNA and regulate TGF-β dependent gene expression [4]. TGF-β is crucial in supporting tissue homeostasis through its ability to regulate cell proliferation, differentiation, migration, apoptosis, and development. The loss of SMAD4 expression results in the deprivation of a major factor of cell growth inhibition, contributing to carcinogenesis. SMAD4 has been previously identified as a possible tumor suppressor since SMAD4 mutations have been reported with high frequency in solid tumors [5,6]. The SMAD4 expression level has been reported to correlate with prognosis in many types of cancers, including colon [7], pancreatic [8], and esophageal [9] cancers. However, very few studies on SMAD4 expression and prognosis in breast cancer have been performed, and the results have been conflicting [10-13]. In the present study, we investigated SMAD4 expression using a tissue microarray (TMA)-based IHC assay and evaluated its clinicopathological and prognostic significance in breast cancer.

METHODS

Patients and tissue samples

Four hundred and seventy-one patients treated for invasive ductal carcinoma (IDC) of breast at Hanyang University Medical Center between December 2000 and December 2008 were considered. The present study was approved by the Institutional Review Board of Hanyang University Medical Center (No. 2019-08-029-004). Patients with incomplete clinical data sets, patients without sufficient archived tissues, and patients with stage IV cancers were excluded. Two hundred and fifty-five patients were selected for the study after exclusions. We examined patients’ age at diagnosis, tumor size, lymph node status, and pathological findings according to the American Joint Committee on Cancer (AJCC, 7th edition) classification for stage, tumor size, lymph node metastasis stage, and hormone receptor status. The hormone receptor (ER and PR) statuses were assessed using IHC and were scored using the Allred score [14]. Patients with a score >1 were defined as ER/PR positive. Testing for HER2 was performed using either IHC staining or fluorescent in situ hybridization (FISH). HER2 expression was classified into levels 0 to 3+ according to the guidelines by American Society of Clinical Oncology/College of American Pathologists [15]. A score of 3+ was given to specimens showing uniform and intense membrane staining in >30% of invasive tumor cells, and these specimens were rated as HER2 positive. The cases with a score of 2+ that were equivocal for HER2 protein expression were evaluated by FISH analysis to measure HER2 amplification using an original paraffin block.

TMA construction

Slides stained with hematoxylin and eosin were used to define the most morphologically representative, well fixed, and non-necrotic areas. Single tissue cores (2 mm in diameter) were sampled from each paraffin block and were assembled into a recipient paraffin block using a TMA instrument (AccuMax Array, ISU ABXIS, Seoul, Korea).

IHC staining and interpretation of SMAD4 expression

Multiple 4-μm sections were cut using Leica microtome (Leica Biosystems, Wetzlar, Germany) and transferred to adhesive-coated slides. One section was routinely deparaffinized with standard xylene and hydrated using graded ethanol in water. It was stained with hematoxylin and eosin and covered with a coverslip. For IHC, the TMA slides were dewaxed by heating at 55°C for 30 minutes and by three 5-minute xylene washes.

Primary monoclonal mouse anti-Smad4 antibody (Santa Cruz Biotechnology, Santa Cruz, USA) was diluted (1:200) in goat serum and incubated at room temperature for 1 hour. After three 2-minute washes with phosphate-buffered saline, the sections were incubated with a biotinylated goat anti-mouse secondary antibody for 30 minutes (DAKO, Carpinteria, USA). The slides were then dehydrated following standard procedure and sealed with coverslips. Negative controls were performed by omitting Smad4 antibody during the primary antibody incubation.

To analyze SMAD4 expression in breast cancer, we used the IHC assay with the application of immunoreactivity score (IRS) criteria [16]. The extensional standards were the “fraction of positive cells” (0: <5%, 1: 6%−25%, 2: 26%−50%, 3: 51%−75%, and 4: >75%) and the “staining intensity score” (0: colorless, 1: pallide-flavens, 2: yellow, and 3: brown). The IRS was calculated by multiplying the “staining intensity score” and the “fraction of positive cells” [17]. The staining score was stratified as absent (−, score 0), weak positive (+, score 1−4), moderate positive (++, score 5−8), and strong positive (+++, score 9−12) (Figure 1). To evaluate SMAD4 expression, specimens with absent and weak positive (0 ≤IRS ≤4) scores were classified into the low SMAD4 expression (SMAD4 low) group. Specimens with moderate and strong positive scores (5 ≤IRS ≤12) were classified into the high SMAD4 expression (SMAD4 high) group (Figure 1). The interpretation of IHC staining in this study was performed by a single pathologist (SS Paik).

Figure 1.

Representative sections of the immunohistochemistry of SMAD4 in breast cancer tissue (magnification, × 200). (A) Negative SMAD4 staining. (B) Weak SMAD4 staining. (C) Intermediate SMAD4 staining. (D) Strong SMAD4 staining.

Statistical analysis

Fisher’s exact test, chi-squared test, and logistic regression analysis were used to evaluate the correlations between the clinicopathological features and SMAD4 expression. In the survival analyses, the plots were generated using the Kaplan-Meier curve and were compared using the log-rank test. A multivariate analysis was performed to identify the independent prognostic markers for disease-free survival (DFS) and overall survival (OS) using the Cox multistep regression model. Statistical analyses were performed using SAS version 9.4 (SAS Institute Inc., Cary, USA) and R package version 3.5.1 (RStudio, Boston, USA).

RESULTS

The median follow-up period was 66.3 months (range 3.9 to 134.2 months) and the median age was 49 years (range 27 to 79 years). Among the 255 cases, 239 (93.7%) patients had primary tumor size of less than 2 cm and 16 (6.3%) patients had primary tumor size of more than 2 cm. Regional lymph node metastasis was present in 131 cases (51.4%). According to the AJCC classification scheme, 77 (30.2%) patients had stage I cancer, while 178 (69.8%) had stage II and stage III cancer. ER expression was positive in 142 cases (55.7%) and PR expression was positive in 134 cases (52.5%). Out of all the cases, 67 (26.3%) were positive for HER2 expression on IHC analysis and/or positive for HER2 gene amplification detected by the FISH analysis. One hundred sixty-six specimens (65.1%) were categorized into the SMAD4 low group and 89 specimens (34.9%) were categorized into the SMAD4 high group (Table 1, Figure 1).

Association between SMAD4 expression and clinicopathological parameters in breast cancer patients (n=255)

Relationship between SMAD4 expression and clinicopathological parameters

High SMAD4 expression was positively associated with good clinical phenotypes of breast cancer such as low early AJCC stage (p= 0.009), ER positivity (p=0.026), or HER2 negativity (p=0.001). No significant correlation was detected between SMAD4 expression and the other clinicopathological parameters such as histological grade, age, lymph node metastasis, PR status, and triple-negative breast cancer (TNBC) (Table 1).

Comparison between survival outcome and SMAD4 expression

During the follow-up period, there were 64 (25.1%) recurrences among the 255 patients. Out of these, 47 cases were SMAD4 low and 17 were SMAD4 high (73.4% and 26.6%, respectively). In overall survival, 55 events (21.6%) occurred. Out of these, 42 patients were SMAD4 low and 13 were SMAD4 high (76.4% and 23.6%, respectively). No difference was observed in DFS between the SMAD4 low group and the SMAD4 high group (p=0.115), but the SMAD4 high group showed significantly better OS (p=0.045) than the SMAD4 low group (Figure 2A, 2B). In the subgroup analysis, high SMAD4 expression was correlated with better OS in T1 stage tumor (size ≤2 cm) (p=0.034). However, it did not show significant correlation with DFS (Figure 2C, 2D). In the multivariate analysis, a significant association was observed between OS and SMAD4. High SMAD4 expression was observed to be an independent prognostic factor for better OS among patients with T1 stage tumor in the multivariate analysis (hazard ratio: 0.459, p=0.024) (Table 2).

Figure 2.

Kaplan-Meier curves for disease-free survival (DFS) and overall survival (OS) for breast cancer according to SMAD4 expression. (A) All patients –DFS. (B) All patient – OS. (C) T1 staged tumor (≤2 cm) – DFS. (D) T1 staged tumor (≤2 cm) – OS.

Univariate and multivariate analysis of various prognostic parameters for survival in cancer patients with primary tumor size ≤ 2 cm

DISCUSSION

SMAD4 is a key mediator of the TGF-β pathway. In the present study, the expression of SMAD4 was investigated using IHC analysis in 255 cases of IDC. The present data showed that high SMAD4 expression was associated with favorable clinicopathological parameters (stage, ER positivity, and HER2 negativity) and OS. There was a significant association between SMAD4 expression and OS in patients with T1 stage tumors (tumor size ≤2 cm).

SMAD4 expression was found to be lower in breast tumor cells when compared with normal breast epithelium [11]. Two previous reports applied the IRS criteria with respect to the expression of SMAD4 in breast cancer. Lui et al. [13] reported that 43.1% of breast cancer cases were SMAD4 low (0 ≤IRS ≤4) and 56.9% were SMAD4 high (5 ≤IRS ≤2 ), whereas Stuelten et al. [11] reported the figures of 36.3% and 64%, respectively. Our results are closely analogous to those of these studies, with 34.9% SMAD4 low cases and 65.1% SMAD4 high cases. There are inconsistent reports on the correlation between SMAD4 expression and prognostic markers in breast cancer. In previous studies, SMAD4 showed no significant correlation with tumor size, metastases, nodal status, histological grade, histological type, or estrogen receptor expression [11]. However, Lui et al. [13] reported that SMAD4 expression was negatively associated with histological grade.

The role of SMAD4 as a tumor suppressor is consistent with the observation that high expression of this protein is associated with a favorable prognosis. Recent studies have indicated that inactivation of SMAD4 is related to progression of disease in various cancers [18-21]. In colon cancer, SMAD4 inactivation promotes malignancy and drug resistance [22]. In pancreatic cancer, low expression of SMAD4 is associated with malignant progression [23]. Similarly, in non-small-cell lung carcinoma, SMAD4 expression was higher in the normal broncho-tracheal epithelium, but lower in the tumor tissues and closely correlated with lymph node metastasis [24]. Thus, low expression of SMAD4 shows an unfavorable outcome in other types of cancers. Our study demonstrated similar results in breast cancer.

Very few studies have reported the correlation between SMAD4 and prognosis in breast cancer. Lui et al. [13] reported that SMAD4 expression appeared to be decreased in breast cancer when compared with normal tissues and reduced SMAD4 expression levels tended to exhibit more poorly differentiated tumors, a higher risk of recurrence, and shorter OS. Kruijt et al. [12] also reported that low expression of SMAD4 had an unfavorable prognosis regarding progression-free survival. These results are in agreement with the present study. However, one study suggested that a SMAD4-negative tumor showed marginally better overall 5-year survival than a SMAD4-positive tumor, though this difference was not statistically significant [11]. Another study demonstrated that loss of SMAD4 was correlated with a decrease in axillary lymph node metastasis [25]. Since there are conflicting reports in terms of prognosis, recent studies have attempted to determine whether SMAD4 as a prognostic factor combines with other genes or protein expressions [10,12].

TGF-β is considered to play a dual role in cancer development as it displays both tumorigenic and tumor-suppressive effects. TGF-β has been reported to act as a tumor suppressor by inhibiting the cell proliferation of breast cancer cell lines in early stage [26,27]. In contrast, in later stages of cancer, TGF-β has direct pro-tumorigenic effects through the stimulation of invasion, the migration of tumor cells, and the activation of the tumor stroma [28,29]. Positive association of high expression of SMAD4 in T1 stage early breast cancer with better survival outcome in the present study could be explained by the fact that SMAD4 is a key mediator of TGF-β pathway and TGF-β acts as a tumor suppressor in the early stage.

The present study has several limitations. No stage IV patients were included in the TMA data. The IHC analysis using TMA may not reflect the intratumoral heterogeneity. Moreover, the proportion of small-sized tumors was unintentionally higher than the large-sized tumors in our data. However, our results could provide data for SMAD4 expression and prognosis in the early stage breast cancer.

In conclusion, high SMAD4 expression was correlated with several favorable prognostic factors including tumor size, regional lymph node metastasis, AJCC stage, ER positivity, and HER2 negativity. Additionally, high SMAD4 expression was associated with favorable OS in T1 stage breast cancer. Therefore, SMAD4 in breast cancer has potential prognostic significance and further investigation and understanding are needed to elucidate it.

Notes

The authors declare that they have no competing interests.

References

1. Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A, et al. Colorectal cancer statistics, 2017. CA Cancer J Clin 2017;67:177–93.
2. Guo F, Kuo YF, Shih YCT, Giordano SH, Berenson AB. Trends in breast cancer mortality by stage at diagnosis among young women in the United States. Cancer 2018;124:3500–9.
3. Fayanju OM, Park KU, Lucci A. Molecular genomic testing for breast cancer: utility for surgeons. Ann Surg Oncol 2018;25:512–9.
4. Heldin CH, Miyazono K, ten Dijke. TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature 1997;390:465–71.
5. Miyaki M, Iijima T, Konishi M, Sakai K, Ishii A, Yasuno M, et al. Higher frequency of Smad4 gene mutation in human colorectal cancer with distant metastasis. Oncogene 1999;18:3098–103.
6. Shi Y, Hata A, Lo RS, Massague J, Pavletich NP. A structural basis for mutational inactivation of the tumour suppressor Smad4. Nature 1997;388:87–93.
7. Yan P, Klingbiel D, Saridaki Z, Ceppa P, Curto M, McKee TA, et al. Reduced expression of SMAD4 is associated with poor survival in colon cancer. Clin Cancer Res 2016;22:3037–4.
8. Singh P, Srinivasan R, Wig JD. SMAD4 genetic alterations predict a worse prognosis in patients with pancreatic ductal adenocarcinoma. Pancreas 2012;41:541–6.
9. Singhi AD, Foxwell TJ, Nason K, Cressman KL, McGrath KM, Sun W, et al. Smad4 loss in esophageal adenocarcinoma is associated with an increased propensity for disease recurrence and poor survival. Am J Surg Pathol 2015;39:487–95.
10. Min KW, Kim DH, Do SI, Chae SW, Kim K, Sohn JH, et al. Expression pattern of Smad4/GATA3 as a predictor of survival in invasive ductal carcinoma of the breast. Pathobiology 2017;84:130–8.
11. Stuelten CH, Buck MB, Dippon J, Roberts AB, Fritz P, Knabbe C. Smad4-expression is decreased in breast cancer tissues: a retrospective study. BMC Cancer 2006;6:25.
12. De Kruijf, Dekker TJ, Hawinkels LJ, Putter H, Smit VT, Kroep JR, et al. The prognostic role of TGF-beta signaling pathway in breast cancer patients. Ann Oncol 2013;24:384–90.
13. Liu N, Yu C, Shi Y, Jiang J, Liu Y. SMAD4 expression in breast ductal carcinoma correlates with prognosis. Oncol Lett 2015;10:1709–15.
14. Harvey JM, Clark GM, Osborne CK, Allred DC. Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol 1999;17:1474–81.
15. Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, et al. American Society of Clinical Oncology/College of american pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med 2007;131:18–43.
16. Brown RS, Wahl RL. Overexpression of Glut-1 glucose transporter in human breast cancer. An immunohistochemical study. Cancer 1993;72:2979–85.
17. Remmele W, Stegner HE. Recommendation for uniform definition of an immunoreactive score (IRS) for immunohistochemical estrogen receptor detection (ER-ICA) in breast cancer tissue. Pathologe 1987;8:138–40.
18. Wilentz RE, Su GH, Dai JL, Sparks AB, Argani P, Sohn TA, et al. Immunohistochemical labeling for dpc4 mirrors genetic status in pancreatic adenocarcinomas : a new marker of DPC4 inactivation. Am J Pathol 2000;156:37–43.
19. Wilentz RE, Iacobuzio-Donahue CA, Argani P, McCarthy DM, Parsons JL, Yeo CJ, et al. Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia: evidence that DPC4 inactivation occurs late in neoplastic progression. Cancer Res 2000;60:2002–6.
20. Natsugoe S, Xiangming C, Matsumoto M, Okumura H, Nakashima S, Sakita H, et al. Smad4 and transforming growth factor beta1 expression in patients with squamous cell carcinoma of the esophagus. Clin Cancer Res 2002;8:1838–42.
21. Cardillo MR, Lazzereschi D, Gandini O, Di Silverio, Colletta G. Transforming growth factor-beta pathway in human renal cell carcinoma and surrounding normal-appearing renal parenchyma. Anal Quant Cytol Histol 2001;23:109–17.
22. Papageorgis P, Cheng K, Ozturk S, Gong Y, Lambert AW, Abdolmaleky HM, et al. Smad4 inactivation promotes malignancy and drug resistance of colon cancer. Cancer Res 2011;71:998–1008.
23. Blackford A, Serrano OK, Wolfgang CL, Parmigiani G, Jones S, Zhang X, et al. SMAD4 gene mutations are associated with poor prognosis in pancreatic cancer. Clin Cancer Res 2009;15:4674–9.
24. Ke Z, Zhang X, Ma L, Wang L. Deleted in pancreatic carcinoma locus 4/Smad4 participates in the regulation of apoptosis by affecting the Bcl-2/Bax balance in non-small cell lung cancer. Hum Pathol 2008;39:1438–45.
25. Xie W, Mertens JC, Reiss DJ, Rimm DL, Camp RL, Haffty BG, et al. Alterations of Smad signaling in human breast carcinoma are associated with poor outcome: a tissue microarray study. Cancer Res 2002;62:497–505.
26. Zugmaier G, Ennis BW, Deschauer B, Katz D, Knabbe C, Wilding G, et al. Transforming growth factors type beta 1 and beta 2 are equipotent growth inhibitors of human breast cancer cell lines. J Cell Physiol 1989;141:353–61.
27. Gobbi H, Dupont WD, Simpson JF, Plummer WD, Schuyler PA, Olson SJ, et al. Transforming growth factor-β and breast cancer risk in women with mammary epithelial hyperplasia. J Natl Cancer Inst 1999;91:2096–101.
28. Wiercinska E, Naber HP, Pardali E, van der, van Dam, ten Dijke. The TGF-beta/Smad pathway induces breast cancer cell invasion through the up-regulation of matrix metalloproteinase 2 and 9 in a spheroid invasion model system. Breast Cancer Res Treat 2011;128:657–66.
29. Ronnov-Jessen L, Petersen OW. Induction of alpha-smooth muscle actin by transforming growth factor-beta 1 in quiescent human breast gland fibroblasts. Lab Invest 1993;68:696–707.

Article information Continued

Figure 1.

Representative sections of the immunohistochemistry of SMAD4 in breast cancer tissue (magnification, × 200). (A) Negative SMAD4 staining. (B) Weak SMAD4 staining. (C) Intermediate SMAD4 staining. (D) Strong SMAD4 staining.

Figure 2.

Kaplan-Meier curves for disease-free survival (DFS) and overall survival (OS) for breast cancer according to SMAD4 expression. (A) All patients –DFS. (B) All patient – OS. (C) T1 staged tumor (≤2 cm) – DFS. (D) T1 staged tumor (≤2 cm) – OS.

Table 1.

Association between SMAD4 expression and clinicopathological parameters in breast cancer patients (n=255)

Characteristic No. (%) SMAD4
p-value
Low expression (n=166) High expression (n=89)
Age(yr)
 ≤ 50 139 (54.5) 91 (54.8) 48 (53.9) 0.892
 > 50 116 (45.5) 75 (45.2) 41 (46.1)
Histologic grade
 Grade 1 38 (14.9) 23 (13.9) 15 (16.9) 0.749
 Grade 2 and 3 217 (85.1) 143 (86.1) 74 (83.1)
Primary tumor size
 ≤ 2 cm 239 (93.7) 152 (91.6) 87 (97.8) 0.052
 > 2 cm 16 (6.3) 14 (8.4) 2 (2.2)
Lymph node metastasis
 Negative 124 (48.6) 74 (44.6) 50 (56.2) 0.077
 Positive 131 (51.4) 92 (55.4) 39 (43.8)
Stage
 Stage I 77 (30.2) 41 (24.7) 36 (40.5) 0.009
 Stage II & III 178 (69.8) 125 (75.3) 53 (59.5)
Lymphovascular invasion
 Negative 111 (43.5) 66 (39.8) 45 (50.6) 0.097
 Positive 144 (56.5) 100 (60.2) 44 (49.4)
Perineural invasion
 Negative 194 (76.1) 128 (77.1) 66 (74.2) 0.599
 Positive 61 (23.9) 38 (22.9) 23 (25.8)
ER expression
 Negative 113 (44.3) 82 (49.4) 31 (34.8) 0.026
 Positive 142 (55.7) 84 (50.6) 58 (65.2)
PR expression
 Negative 121 (47.5) 86 (51.8) 35 (39.3) 0.057
 Positive 134 (52.5) 80 (48.2) 54 (60.7)
HER2 amplification
 Negative 188 (73.7) 111 (66.9) 77 (86.5) 0.001
 Positive 67 (26.3) 55 (33.1) 12 (13.5)
TNBC
 Non-TNBC 201 (78.8) 131 (78.9) 70 (78.7) 0.961
 TNBC 54 (21.2) 35 (21.1) 19 (21.3)

ER=estrogen receptor; PR=progesterone receptor; HER2=human epidermal growth factor receptor 2; TNBC=triple negative breast cancer.

Table 2.

Univariate and multivariate analysis of various prognostic parameters for survival in cancer patients with primary tumor size ≤ 2 cm

Characteristic Disease free survival
Overall survival
Univariate
Multivariate
Univariate
Multivariate
HR p-value HR 95% CI p-value HR p-value HR 95% CI p-value
Age (≤ 50 vs. > 50) 1.554 0.101 2.042 0.017 2.158 1.196–3.893 0.011
Histologic grade (G1 vs. G2 & G3) 3.410 0.002 2.995 1.346–6.665 0.007 1.645 0.149
Lymph node metastasis (absent vs. present) 2.295 0.004 1.472 0.191
Lymphovascular invasion (absent vs. present) 1.781 0.044 1.274 0.415
Perineural invasion (absent vs. present) 1.811 0.034 1.749 1.003–3.048 0.049 1.760 0.063 1.918 1.054–3.489 0.033
SMAD4 expression (low vs. high) 0.598 0.089 0.653 0.358–1.191 0.165 0.488 0.038 0.459 0.233–0.903 0.024
ER expression (negative vs. positive) 0.670 0.135 0.537 0.035
PR expression (negative vs. positive) 0.563 0.035 0.504 0.288–0.882 0.016 0.540 0.038
HER2 amplification (absent vs. present) 0.809 0.514 0.585 0.299–1.145 0.118 0.940 0.853
Non TNBC vs. TNBC 1.481 0.204 1.999 0.030 2.298 1.223–4.317 0.010

HR=hazard ratio; CI=confidence interval;ER=estrogen receptor; PR=progesterone receptor; HER2=human epidermal growth factor receptor 2; TNBC=triple negative breast cancer.