Next Article in Journal
USP53 Exerts Tumor-Promoting Effects in Triple-Negative Breast Cancer by Deubiquitinating CRKL
Next Article in Special Issue
From Diabetes to Oncology: Glucagon-like Peptide-1 (GLP-1) Receptor Agonist’s Dual Role in Prostate Cancer
Previous Article in Journal
Unlocking Colchicine’s Untapped Potential: A Paradigm Shift in Hepatocellular Carcinoma Prevention
Previous Article in Special Issue
The Influence of the Microbiome on Urological Malignancies: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prognostic Significance of Cyclin D1 Expression in Small Intestinal Adenocarcinoma

1
Department of Pathology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Republic of Korea
2
Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
3
Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(20), 5032; https://doi.org/10.3390/cancers15205032
Submission received: 21 August 2023 / Revised: 8 October 2023 / Accepted: 14 October 2023 / Published: 18 October 2023

Abstract

:

Simple Summary

Small intestinal adenocarcinoma (SIAC) is a rare tumor, with a rising incidence in recent decades. Although the National Comprehensive Cancer Network (NCCN) established the treatment guidelines for SIAC in 2020, there continues to be urgency to search for reliable prognostic factors and therapeutic regimens. Cyclin D1, a critical cyclin-dependent kinase (CDK) 4/6-dependent regulator of G1/S transition, has attracted much interest as a therapeutic target. The cyclin D1 expression in SIACs has not yet been comprehensively studied, owing to the rarity of this tumor. We investigated the clinicopathological and prognostic significance of the cyclin D1 expression in 232 primary SIACs through a multi-institutional study. Cyclin D1 was commonly overexpressed in SIACs, and a high expression of cyclin D1 was identified as a favorable prognostic indicator in SIAC patients. These findings in SIACs may be important to further the understanding of the mechanism of cyclin D1 in carcinogenesis and for applying appropriate patient therapies.

Abstract

Cyclin D1, a critical cyclin-dependent kinase (CDK) 4/6-dependent regulator of G1/S transition, has attracted much interest as a therapeutic target. The cyclin D1 expression in small intestinal adenocarcinomas (SIACs) has not yet been comprehensively studied, owing to the rarity of this tumor. We investigated the clinicopathological and prognostic significance of the cyclin D1 expression in 232 surgically resected primary SIACs through a multi-institutional study. A high expression of cyclin D1 (cyclin D1High) was detected in 145 SIAC cases (63%), which was significantly higher than that in normal small intestinal mucosa (11%). Cyclin D1High was more commonly found in SIACs with a lower T-category and disease stage and KRAS mutation and predicted better patient survival. Multivariate analysis revealed that cyclin D1High, the absence of retroperitoneal seeding and lymphovascular invasion, and the lower N-category were identified as independent prognostic indicators for patients with SIACs. Specifically, cyclin D1High affected patient survival in the lower stage group (stages I and II). In conclusion, cyclin D1 was commonly overexpressed in SIACs, and cyclin D1High acted as a favorable prognostic indicator in patients with SIACs. These findings in SIACs may, thus, be important to further comprehend the mechanism of cyclin D1 in carcinogenesis and to strategize appropriate patient therapies.

1. Introduction

Uncontrolled cell proliferation via dysregulation of the cell cycle is a hallmark of cancer [1]. Thus, the cell cycle regulation mechanism in cancer is a topic of enormous interest. Cyclin D1, which is encoded by CCND1, is a critical regulator of the G1/S transition and promotes cancer progression in a complex with cyclin-dependent kinase (CDK) 4/6 [2]. Cyclin D1 is overexpressed in various types of cancers; however, findings on its prognostic impact remain inconclusive [3]. A recent meta-analysis of 108 original studies revealed that cyclin D1 had varying effects on prognosis, depending on the cancer site [3]. The cyclin D1 overexpression was related to worse survival in patients with head and neck cancers, but not in patients with gastrointestinal (GI) tract, breast, bladder, or lung cancers [3]. Furthermore, an opposite prognostic effect of cyclin D1 was identified between estrogen receptor (ER)-positive and ER-negative breast cancers [4] and between superficial and muscle-invasive bladder cancers [5]. As expected, the cyclin D1 overexpression was not always accompanied by CCND1 amplification, which implied additional mechanisms of cyclin D1 overexpression beyond CCND1 amplification, such as the activation of mitogenic signaling pathways [3,6]. Ishii et al. proposed that cyclin D1 may improve the outcomes by bypassing its conservative oncogene function through the direct inhibition of the oncogenic signal transducer and activator of transcription 3 (STAT3) [7]. Therefore, the contribution of cyclin D1 to carcinogenesis remains unclear.
Small intestinal adenocarcinoma (SIAC) is extremely rare. In the United States, there will be 12,070 new cases of SIAC in 2023, accounting for 0.6% of all new cancer cases [8]. In Korea, the incidence of new cases of SIAC is lower than that in the United States, with an estimated 958 new cases occurring in 2020, which accounted for 0.3% of all new cancer cases [9]. In localized SIACs, surgical resection is the best approach to improve survival [10]. Systemic treatment for SIAC is mainly extrapolated from the management of colorectal carcinoma (CRC) due to the embryonic developmental similarity [11]. Previous studies on SIACs have compared them to CRCs [12] and a few prognostic factors of SIACs have been identified, which include tumor location, microsatellite instability (MSI), and KRAS mutations [13,14,15]. Based on the limited data available from retrospective studies on SIAC and extrapolation from studies on CRC, the National Comprehensive Cancer Network (NCCN) was the first to establish a standard treatment guideline for SIAC in 2020 [16]. The NCCN recommended 5-fluorouracil (5-FU) and leucovorin (LV), 5-FU/LV plus oxaliplatin (FOLFOX), capecitabine plus oxaliplatin (CAPEOX), or capecitabine as adjuvant chemotherapy. However, the efficacy of these combinations in SIAC is variable [16]. Therefore, there continues to be a pressing need for reliable prognostic factors and therapeutic regimens for patients with SIAC.
Previous studies on cyclin D1 in SIAC are extremely rare, although numerous studies have been reported on cyclin D1 expression in CRC [3,17,18]. Furthermore, the CDK4/6 inhibitors—ribociclib, palbociclib, and abemaciclib—have been evaluated in CRC, and their therapeutic effect has been amplified in combination with other drugs [19]. In experimental studies, KRAS-mutated CRCs and RAS- and BRAF-mutant CRCs were found to be sensitive to a combination of palbociclib with MEK inhibitor and a combination of abemaciclib and RAF inhibitor, respectively [20,21]. These promising results from CDK4/6 inhibitors in experimental models were followed by studies for their efficacy in clinical trials. Currently, the combination of abemaciclib with ERK1/2 inhibitor and cetuximab and the combination of CDK4/6 inhibitor with checkpoint inhibitors such as anti-programmed cell death 1 (PD1) therapy is being clinically tested in patients with advanced CRC [19]. Although several studies on cyclin D1 expression have been actively conducted before, considering the application of CDK4/6 inhibitors in CRC, cyclin D1-related studies in SIAC remain limited and rudimentary, owing to the rarity of the disease. To the best of our knowledge, there was only one SIAC study on cyclin D1, which was performed in a small cohort of 24 SIAC cases [22].
Thus, this study aimed to investigate the clinicopathological and prognostic significance of cyclin D1 in SIACs collected through a multi-institutional study. In addition, we compared the cyclin D1 expression in SIACs with that in normal small intestine and evaluated the association of cyclin D1 with KRAS and BRAF mutations.

2. Materials and Methods

2.1. Study Population

A total of 236 surgically resected primary SIACs were collected from the surgical pathology archives of 23 South Korean institutions by the Korean Small Intestinal Cancer Study Group, as previously reported [23]. Carcinomas originating in the duodenum, jejunum, and ileum were included, but tumors extending into the small intestines from the surrounding GI tract organs were excluded. Of the 236 SIAC cases, four cases without available tissue blocks were excluded; hence, the study finally included 232 SIACs. The clinicopathological findings of the patients were updated, including the most recent follow-up data, survival status, and TNM staging in accordance with the 8th American Joint Committee on Cancer (AJCC) staging system [24].

2.2. Immunohistochemical Analysis

For immunohistochemistry (IHC), tissue microarrays (TMAs) were constructed containing three cores of each tumor and one core of possible paired normal mucosa [18]. The TMA sections were immunostained using an anti-cyclin D1 antibody (clone SP4; Cell Marque, Darmstadt, Germany) and the Ventana BenchMark XT immunostainer (Ventana Medical System, Tucson, AZ, USA). The nuclear expression of cyclin D1 was semiquantitatively determined by intensity and percentage, as described previously [18]. The staining intensity was graded as 0 (no staining), 1 (weak staining), 2 (moderate staining), and 3 (strong staining). The percentage of staining was graded on a scale of 0–10, at 10% increments of 0 (no staining), 1 (1–10%), 2 (11–20%), and 3 (21–30%), up to 10 (91–100%). The staining intensity grade (0–3) was multiplied by the percentage grade of the stained cells (0–10) to yield an immunostaining score ranging from 0 to 30. The average score for each case was calculated and selected as the final overall score.

2.3. Molecular Analysis

We obtained information about mutations in codons 12 and 13 of KRAS exon 1 and codon 600 of BRAF exon 15 from elsewhere [15]. Genomic DNA was extracted with the QIAmp DNA Mini Kit (Qiagen, Valencia, CA, USA), and KRAS and BRAF mutations were analyzed by cycle sequencing [15].

2.4. Statistical Analysis

Statistical analyses were performed using Statistics for Windows (version 28.0; IBM, Armonk, NY, USA). The association of the cyclin D1 expression with clinicopathological factors was evaluated using the unpaired Student’s t-test for continuous data and the χ2 and/or Fisher’s exact test for categorical data. The survival curves were plotted using the Kaplan–Meier method, and the associations between the overall survival (OS) rates and various clinicopathological factors were assessed using the log-rank test. The Cox proportional hazards model was used to calculate the significance of any prognostic factors. Receiver operating characteristic (ROC) curves were generated to evaluate the predictive power of the cyclin D1 expression for accurately classifying SIAC cases related to OS. p < 0.05 were considered to indicate statistical significance.

3. Results

3.1. Clinicopathological Characteristics

The baseline clinicopathological characteristics are shown in Table 1. The mean patient age was 59.9 ± 12.8 years (range: 23–84 years), and the male-to-female ratio was 1.8. The tumors, which ranged in size from 0.5 to 16.0 cm (mean size: 4.3 ± 2.5 cm), were located in the duodenum (140 cases, 60.3%), jejunum (58, 25.0%), and ileum (34, 14.7%). An infiltrative growth pattern was observed in 74.5% of the tumors (167/224), followed by polypoid (40/224, 17.9%) and nodular (17/224, 7.6%) patterns. The majority of the tumors were tubular adenocarcinoma (205/232, 88.4%) and of low grade (184/232, 79.3%). Nontubular carcinomas were also detected, which included 12 (5.2%) mucinous carcinomas, six (2.6%) medullar carcinomas, five (2.1%) undifferentiated carcinomas, and four (1.7%) signet ring cell carcinomas. Pancreatic invasion was observed in 87 cases (37.5%). Six cases (2.6%) of other loop invasions and 16 cases (6.9%) of retroperitoneal invasion were seen. Lymphovascular and perineural invasions were evaluated in 231 cases and, respectively, observed in 113 (48.9%) and 76 (32.9%) cases. Resection margins with cancer involvement were noted in nine cases (9/218, 4.1%). According to the AJCC staging scheme, four cases (1.7%) were categorized as Tis, 13 cases (5.6%) as T1, 14 cases (6.1%) as T2, 68 cases (29.3%) as T3, and 133 cases (57.3%) as T4 tumors. The nodal status was investigated in 215 cases. Of these, nodal metastases were detected in 49.3% (106/215) of the tumors, including 54 cases (25.1%) of N1 and 52 cases (24.2%) of N2. Consequently, the tumors were grouped into four cases (4/215, 1.9%) of stage 0, 22 cases (10.2%) of stage I, 83 cases (38.6%) of stage II, and 106 cases (49.3%) of stage III. No case of stage IV was noted in this study. KRAS was mutated in 32.3% (60/186) of the tumors, while BRAF mutations were observed in 1.1% (2/176) of the tumors. Chemotherapy and radiotherapy were performed in 36.1% (82/227) and 11.5% (26/226) of the patients, respectively. The follow-up period after surgical resection was 1.1–168.4 months (mean: 41.5 ± 40.9 months).

3.2. Cyclin D1 Expression

Cyclin D1 immunostaining was interpretable in 189 cases of normal mucosa of the small intestine. Of these, 79 cases (41.8%) expressed cyclin D1 in the transitional zone of the crypt at the lower portion of the gland (Figure 1), which consisted of 47 cases (47/79, 59.5%) with weak staining intensity, 31 (39.2%) with moderate intensity, and 1 (1.3%) with strong intensity. The cyclin D1 staining score in the normal epithelia ranged from 1 to 12, which included 35 cases (35/79, 44.3%) with a score of 1, 18 cases (22.8%) with a score of 2, 11 cases (13.9%) with a score of 4, five cases (6.3%) each with a score of 3 or 8, three cases (3.8%) with a score of 6, and one case (1.3%) each with a score of 5 or 12.
Of the 232 SIAC cases, cyclin D1 was expressed in 220 (94.8%). The cyclin D1 staining scores in SIACs ranged from 0.3 to 30 in the following order: 0 < score ≤ 5 in 95 cases (95/220, 43.2%); 5 < score ≤ 10 in 32 cases (32/220, 14.6%); 10 < score ≤ 15 in 33 cases (33/220, 15.0%); 15 < score ≤ 20 in 28 cases (28/220, 12.7%); 20 < score ≤ 25 in 19 cases (19/220, 8.6%); 25 < score ≤ 30 in 13 cases (13/220, 5.9%). The cyclin D1 staining scores of SIACs were significantly higher than those of the normal mucosa of the small intestine (mean: 9.5 ± 8.4 vs. 1.0 ± 1.9; p < 0.001). Most (210/220, 95.5%) SIACs had a score of ≥1. Based on the ROC curve analysis, a high expression of cyclin D1 (cyclin D1High) was defined as a staining score of >3.5. Cyclin D1High was detected in 145 SIAC cases (62.5%) (Figure 2A–F), which was more often than reported in normal small intestinal mucosa (21/189, 11.1%; p < 0.001). The relationship between cyclin D1High and the clinicopathological factors of SIAC patients is described in Table 1. Cyclin D1High was more commonly found in tumors with a lower T category (p = 0.046) and disease stage (p = 0.003) and KRAS mutations (p = 0.026).

3.3. Survival Analysis

The survival analysis results are summarized in Table 2. By univariate survival analysis, SIAC patients with cyclin D1High showed significantly longer survival times than those with cyclin D1Low (median, 44.4 months vs. 24.5 months, p = 0.005; Figure 3). In addition, proximal tumor location (p = 0.007), the absence of other loop invasion (p = 0.044) or retroperitoneal seeding (p < 0.001), no lymphovascular (p < 0.001) or perineural invasion (p = 0.004), no radiotherapy (p = 0.005), no nodal metastasis (p < 0.001), and lower T and N categories and disease stage (all p < 0.001) were all related to better OS. In the multivariate analysis, cyclin D1High (p = 0.031), the absence of retroperitoneal seeding (p = 0.007) or lymphovascular invasion (p = 0.010), and lower N category (p = 0.001) were identified as favorable prognostic indicators for patients with SIACs.
Further, the prognostic impact of cyclin D1 expression was investigated with respect to tumor stage (Figure 4A,B). Cyclin D1High significantly predicted the better OS of SIAC patients in the lower stage group. In the lower stage group (stages I and II, n = 105), SIAC patients with cyclin D1High had significantly longer survival times (median, 146.6 months) than those with cyclin D1Low (29.1 months; p < 0.001; Figure 4A). In the higher stage group (stage III, n = 106), no significant difference in OS was identified between the cyclin D1High and cyclin D1Low groups (22.6 months vs. 21.0 months, p = 0.629; Figure 4B).

4. Discussion

Despite our meticulous search for reports of cyclin D1 in SIAC through PubMed, Embase, and Google Scholar, with further searches via manual cross-referencing, we found only one study by Arber et al. conducted in a small cohort of 24 cases [22]. In the SIAC study by Arber and colleagues, they used a non-SP4 clone and found that cyclin D1High was associated with a decrease in 3-year survival rates of patients [22]. Meanwhile, in the present study, we evaluated cyclin D1 expression using SP4 in 232 SIACs and found that cyclin D1High predicted better survival outcomes. As mentioned earlier, SIAC has been studied comparatively with CRC because of its embryonic developmental similarity and anatomical proximity. Given the rarity of SIAC studies on cyclin D1, we alternatively analyzed the present study in comparison with previous CRC studies on cyclin D1 (Supplementary Table S1). Conflicting results have been reported regarding the prognostic effect of cyclin D1 expression in CRCs [18]. The heterogeneity in these results may have stemmed from differences in sample sizes, tissue section type, the clone used, and cut-off values for cyclin D1 expression [18].
To analyze the effect of the clones used, we investigated reports of cyclin D1 IHC in malignancies [25,26,27]. Cyclin D1 IHC has been a key tool in distinguishing mantle cell lymphoma from other small B cell lymphomas but suffers from technical difficulties and ambiguous staining results [25]. Cheuk et al. first demonstrated the superior performance of the newly available rabbit monoclonal antibody SP4 for cyclin D1 IHC in 2004 [25]. In subsequent comparative studies with various anti-cyclin D1 antibodies, SP4 produced the strongest staining with a high sensitivity of 95%, enabling the optimal detection of cyclin D1 expression [26,27]. In CRC, five studies on the prognostic effect of cyclin D1 expression using SP4 were found (Supplementary Table S1) [18,28,29,30,31]. All five studies were conducted with TMAs. Their mean frequency of cyclin D1High was 65.0% (range, 54.0%–78.6%), similar to the 62.5% found in the present study of SIACs. Three of five CRC studies revealed a favorable prognostic role of cyclin D1, and all of these were performed in relatively large CRC cohorts containing >200 cases [18,28,29]. In contrast, the other two studies on <200 CRC cases did not find any prognostic significance of cyclin D1 [30,31]. When searching studies of cyclin D1 expression using non-SP4 clones in CRCs, we found 18 studies (Supplementary Table S1) [32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49]. Most studies (15/18, 83.3%) performed cyclin D1 immunostaining on conventionally sectioned slides [33,34,35,36,37,38,39,40,41,42,43,45,47,48,49]. Nine of 18 studies precisely defined the clones [32,33,34,35,36,37,38,39,40], and two of them on >200 cases consistently showed an association between cyclin D1 and better prognosis [32,33]. To obtain reliable results for the prognostic significance of cyclin D1, we designed a multi-institutional study and selected SP4 for use. We constructed TMAs for IHC because no heterogeneity of cyclin D1 expression was observed in selected SIAC cases. Expectedly, cyclin D1High effectively predicted better prognoses of patients with SIACs. A limitation of this study is that it was conducted in a Korean population with homogeneous ethnic group and there is a lack of comparable studies on cyclin D1 in SIAC. Nevertheless, we assessed cyclin D1 expression in a relatively large cohort of SIAC patients and identified the associations of cyclin D1High with lower T category and disease stage and better survival outcomes. We also found that cyclin D1High significantly predicted the better OS of SIAC patients in the lower stage group. Further studies with a large number of SIAC cases are needed to solidify the prognostic predictive value of cyclin D1.
Various cutoff points for cyclin D1High have been applied in both CRC and SIAC studies, and cyclin D1High has often been interpreted using predetermined cutoffs, such as 5% or 10% cell positivity without relevant information (Supplementary Table S1) [30,33,34,35,39,40,45,46,47,48,49]. The SIAC study by Arber et al. also defined cyclin D1High when staining was present in at least 10% of the tumor, irrespective of the intensity, without any further explanation [22]. Considering the lack of any standardized criteria for cyclin D1 overexpression, we performed ROC analysis to establish the objectivity and optimal decision threshold, which is very necessary for IHC interpretation [50]. When using a staining score cutoff point of >3.5, the area under the curve was 0.610. A substantial accuracy in predicting OS was detected, with a sensitivity of 45.3% and a specificity of 76.2%. Accumulating results from cyclin D1 IHC studies in various carcinomas based on ROC analysis can help establish clear and consistent criteria for assessing cyclin D1 expression in the future. Interestingly, Arber et al. observed the cyclin D1 expression at the base of the crypt of normal epithelia of the small intestine [22]; however, they did not compare the expression of cyclin D1 between the normal mucosa and SIAC. In this study, we observed that cyclin D1 was identically expressed in the transitional zone of the crypt at the lower portion of the normal gland and identified cyclin D1High in 11% (21/189) of the normal small intestinal mucosa. Both the cyclin D1 composite staining score and the frequency of cyclin D1High were consistently higher in SIACs than in normal mucosa of the small intestine.
The amplification of CCND1 as an oncogene is known to promote growth and carcinogenesis by regulating cyclin D1 [3]. In contrast, the overexpression of cyclin D1 is not always accompanied by DNA amplification. CCND1 is amplified in 5–20% of all malignancies, but cyclin D1 is more prevalently overexpressed, with a reported frequency of up to 80% [6,51]. Several unexpected functions of cyclin D1 in addition to the oncogenic effects have been reported [52,53,54]. The upregulation of cyclin D1 in breast cells inhibited growth by a prolonged S-phase via retinoblastoma tumor suppressor protein (pRB) and proliferating cell nuclear antigen (PCNA)-mediated DNA synthesis or repair [52]. In addition, cyclin D1 was involved in programmed cell death [53] and could suppress the proliferation of diploid fibroblasts [54]. Ogino et al. hypothesized that cyclin D1-negative CRCs may bypass cyclin D1 activation and develop more aggressive behavior than cyclin D1-positive tumors by accumulating multiple genetic and epigenetic events during carcinogenesis [29].
Previous in vitro studies have described that mutated KRAS upregulated the cyclin D1 expression through pathways involving RAS-MEK-ERK and PI3K signaling cascades [55,56]. An association between cyclin D1 mRNA expression and KRAS mutation was also reported [56]. We examined CCND1 mutations in a part of our cohort by next-generation sequencing (n = 97) and identified no CCND1 mutation. However, we did identify an association between cyclin D1High and KRAS mutations in this study. These findings support the presence of additional mechanisms of cyclin D1 overexpression beyond CCND1 amplification. Dragnev et al. reported that KRAS-driven lung cancer, which usually responds poorly to the epidermal growth factor receptor (EGFR) inhibitor erlotinib, was particularly dependent on CDK4 and was sensitive to the cyclin D1-degrading combination of bexarotene and erlotinib [57]. In CRCs, KRAS-mutant tumors are particularly sensitive to a combination of mitogen-activated protein kinase (MAPK) and CDK4/6 inhibitors [58]. Therefore, inhibiting the cyclin D1–CDK4/CDK6 pathway may enhance responses to targeted therapy, particularly in KRAS-mutated SIACs. Further studies are, however, warranted to elucidate the role of cyclin D1 in the carcinogenesis of SIAC and the interaction of cyclin D1 with other signaling pathways.
A few studies have noted a relationship between the cyclin D1 expression and the response to cancer therapy [55]. In experimental models, the cyclin D1 overexpression induced radio-resistance [59] and resistance to cytotoxic drugs [60], antiestrogens [61], and an EGFR tyrosine kinase inhibitor [62], as well as inhibitors of BRAF and MEK signaling [63]. An association between the cyclin D1 overexpression and therapeutic response to drugs, such as tamoxifen in breast cancer patients [64] and combined chemotherapy of erlotinib and bexarotene in lung cancer patients, was identified in clinical studies [57]. Cyclin D1 may, thus, help control the therapeutic effect in patients with SIAC.

5. Conclusions

Cyclin D1 was commonly overexpressed in SIACs, and cyclin D1High was a favorable prognostic indicator in patients with SIACs. These findings about SIACs may be important to further understand the mechanism of cyclin D1 in carcinogenesis and to strategize appropriate patient therapies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers15205032/s1, Table S1: Previous studies of cyclin D1 expression in SIACs and CRCs.

Author Contributions

Conceptualization and project administration, S.-M.H. and K.-T.J.; funding acquisition, methodology, software, data curation, visualization, writing—original draft preparation, S.-Y.J.; resources, S.-Y.J., S.-M.H. and K.-T.J.; investigation, formal analysis, and validation, S.-Y.J. and S.-M.H.; writing—review and editing and supervision, S.-M.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (No. 2021R1A2C1003898, awarded to S.-Y.J.) and by a Grant of Translational R&D Project through Institute for Bio-Medical Convergence, Incheon St. Mary’s Hospital, The Catholic University of Korea (awarded to S.-Y.J.). The sponsor of the study did not have any role in the study design or collection, analysis, and interpretation of data.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board of Incheon St. Mary’s Hospital (OC14OIMI0133 and 5 January 2015) for studies involving humans.

Informed Consent Statement

The requirement for patient consent was waived because data were retrospectively obtained and anonymized.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to the restriction of the Institutional Review Board of Incheon St. Mary’s Hospital. All other relevant data supporting study findings are within the manuscript.

Acknowledgments

We would like to thank the members of the Korean Small Intestinal Cancer Study Group for providing samples and data for this study: Hee-Kyung Chang, Kosin University College of Medicine, Pusan; Young Kyung Bae, Yeungnam University College of Medicine, Daegu; Ghil Suk Yoon, Kyungpook National University School of Medicine, Dague; Joon Mee Kim, Inha University College of Medicine, Incheon; Gwang Il Kim, CHA Bundang Medical Center, CHA University, Seongnam; Hee Kyung Kim, Soonchunhyang University Bucheon Hospital, Bucheon; Kyu Yun Jang, Chonbuk National University Medical School, Jeonju; Dae Woon Eom, University of Ulsan College of Medicine, Gangneung Asan Hospital, Gangneung; Soo Jin Jung, Inje University College of Medicine, Busan; Gyeong Hoon Kang, Seoul National University College of Medicine, Seoul; Ji Shin Lee, Chonnam National University Medical School, Gwangju; Soon Won Hong, Yonsei University College of Medicine, Seoul; Han-Ik Bae, Kyungpook National University School of Medicine, Dague; Hyun-Jung Kim, Inje University Sanggye Paik Hospital, Seoul; Jinwon Seo, Hallym University Sacred Heart Hospital, Anyang; and Eun Kyoung Kwak, Catholic University of Daegu, Daegu, Korea.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  2. Li, Z.; Liu, J.; Zhang, X.; Fang, L.; Zhang, C.; Zhang, Z.; Yan, L.; Tang, Y.; Fan, Y. Prognostic significance of cyclin D1 expression in renal cell carcinoma: A systematic review and meta-analysis. Pathol. Oncol. Res. 2020, 26, 1401–1409. [Google Scholar] [CrossRef] [PubMed]
  3. Moradi Binabaj, M.; Bahrami, A.; Khazaei, M.; Ryzhikov, M.; Ferns, G.A.; Avan, A.; Mahdi Hassanian, S. The prognostic value of cyclin D1 expression in the survival of cancer patients: A meta-analysis. Gene 2020, 728, 144283. [Google Scholar] [CrossRef] [PubMed]
  4. Ahlin, C.; Lundgren, C.; Embretsén-Varro, E.; Jirström, K.; Blomqvist, C.; Fjällskog, M.L. High expression of cyclin D1 is associated to high proliferation rate and increased risk of mortality in women with ER-positive but not in ER-negative breast cancers. Breast Cancer Res. Treat. 2017, 164, 667–678. [Google Scholar] [CrossRef]
  5. Levidou, G.; Saetta, A.A.; Karlou, M.; Thymara, I.; Pratsinis, H.; Pavlopoulos, P.; Isaiadis, D.; Diamantopoulou, K.; Patsouris, E.; Korkolopoulou, P. D-type cyclins in superficial and muscle-invasive bladder urothelial carcinoma: Correlation with clinicopathological data and prognostic significance. J. Cancer Res. Clin. Oncol. 2010, 136, 1563–1571. [Google Scholar] [CrossRef] [PubMed]
  6. Luangdilok, S.; Wanchaijiraboon, P.; Chantranuwatana, P.; Teerapakpinyo, C.; Shuangshoti, S.; Sriuranpong, V. Cyclin D1 expression as a potential prognostic factor in advanced KRAS-mutant non-small cell lung cancer. Transl. Lung Cancer Res. 2019, 8, 959–966. [Google Scholar] [CrossRef]
  7. Ishii, Y.; Pirkmaier, A.; Alvarez, J.V.; Frank, D.A.; Keselman, I.; Logothetis, D.; Mandeli, J.; O’Connell, M.J.; Waxman, S.; Germain, D. Cyclin D1 overexpression and response to bortezomib treatment in a breast cancer model. J. Natl. Cancer Inst. 2006, 98, 1238–1247. [Google Scholar] [CrossRef]
  8. Cancer Stat Facts: Small Intestine Cancer. Available online: https://seer.cancer.gov/statfacts/html/smint.html (accessed on 6 July 2023).
  9. Annual Report of Cancer Statistics in Korea in 2020. Available online: http://ncc.re.kr/cancerStatsList.ncc?searchKey=total&searchValue=&pageNum=1 (accessed on 27 September 2023).
  10. Klimstra, D.; Nagteggal, I.; Rugge, M.; Salto-Tellez, M. Tumours of the small intestine and ampulla. In WHO Classification of Tumours: Digestive System Tumours, 5th ed.; Carneiro, F., Ochiai, A., Chan, J., Oliva, E., Cheung, N.-Y., Rous, B., Cree, I., Singh, R., Eds.; IARC: Lyon, France, 2019; pp. 111–134. [Google Scholar]
  11. Symons, R.; Daly, D.; Gandy, R.; Goldstein, D.; Aghmesheh, M. Progress in the treatment of small intestine cancer. Curr. Treat. Options Oncol. 2023, 24, 241–261. [Google Scholar] [CrossRef]
  12. Young, J.I.; Mongoue-Tchokote, S.; Wieghard, N.; Mori, M.; Vaccaro, G.M.; Sheppard, B.C.; Tsikitis, V.L. Treatment and survival of small-bowel adenocarcinoma in the United States: A comparison with colon cancer. Dis. Colon Rectum 2016, 59, 306–315. [Google Scholar] [CrossRef]
  13. Jun, S.Y.; Park, E.S.; Lee, J.J.; Chang, H.K.; Jung, E.S.; Oh, Y.H.; Hong, S.M. Prognostic significance of stromal and intraepithelial tumor-infiltrating lymphocytes in small intestinal adenocarcinoma. Am. J. Clin. Pathol. 2020, 153, 105–118. [Google Scholar] [CrossRef]
  14. Jun, S.Y.; Lee, E.J.; Hong, S.M.; Jung, E.S.; Chung, J.Y. Tumor microenvironmental prognostic risk in primary operable small intestinal adenocarcinoma. Am. J. Surg. Pathol. 2021, 45, 917–929. [Google Scholar] [CrossRef]
  15. Jun, S.Y.; Kim, M.; Gu, M.J.; Bae, Y.K.; Chang, H.K.; Jung, E.S.; Jang, K.T.; Kim, J.; Yu, E.; Eom, D.W.; et al. Clinicopathologic and prognostic associations of KRAS and BRAF mutations in small intestinal adenocarcinoma. Mod. Pathol. 2016, 29, 402–415. [Google Scholar] [CrossRef] [PubMed]
  16. Benson, A.B.; Venook, A.P.; Al-Hawary, M.M.; Arain, M.A.; Chen, Y.J.; Ciombor, K.K.; Cohen, S.A.; Cooper, H.S.; Deming, D.A.; Garrido-Laguna, I.; et al. Small bowel adenocarcinoma, version 1.2020, NCCN clinical practice guidelines in oncology. J. Natl. Compr. Canc. Netw. 2019, 17, 1109–1133. [Google Scholar] [CrossRef] [PubMed]
  17. Li, Y.; Wei, J.; Xu, C.; Zhao, Z.; You, T. Prognostic significance of cyclin D1 expression in colorectal cancer: A meta-analysis of observational studies. PLoS ONE 2014, 9, e94508. [Google Scholar] [CrossRef] [PubMed]
  18. Jun, S.Y.; Kim, J.; Yoon, N.; Maeng, L.S.; Byun, J.H. Prognostic potential of cyclin D1 expression in colorectal cancer. J. Clin. Med. 2023, 12, 572. [Google Scholar] [CrossRef] [PubMed]
  19. Thoma, O.M.; Neurath, M.F.; Waldner, M.J. Cyclin-dependent kinase inhibitors and their therapeutic potential in colorectal cancer treatment. Front. Pharmacol. 2021, 12, 757120. [Google Scholar] [CrossRef]
  20. Lee, M.S.; Helms, T.L.; Feng, N.; Gay, J.; Chang, Q.E.; Tian, F.; Wu, J.Y.; Toniatti, C.; Heffernan, T.P.; Powis, G.; et al. Efficacy of the combination of MEK and CDK4/6 inhibitors in vitro and in vivo in KRAS mutant colorectal cancer models. Oncotarget 2016, 7, 39595–39608. [Google Scholar] [CrossRef]
  21. Chen, S.H.; Gong, X.; Zhang, Y.; Van Horn, R.D.; Yin, T.; Huber, L.; Burke, T.F.; Manro, J.; Iversen, P.W.; Wu, W.; et al. RAF inhibitor LY3009120 sensitizes RAS or BRAF mutant cancer to CDK4/6 inhibition by abemaciclib via superior inhibition of phospho-RB and suppression of cyclin D1. Oncogene 2018, 37, 821–832. [Google Scholar] [CrossRef]
  22. Arber, N.; Hibshoosh, H.; Yasui, W.; Neugut, A.I.; Hibshoosh, A.; Yao, Y.; Sgambato, A.; Yamamoto, H.; Shapira, I.; Rosenman, D.; et al. Abnormalities in the expression of cell cycle-related proteins in tumors of the small bowel. Cancer Epidemiol Biomark. Prev. 1999, 8, 1101–1105. [Google Scholar]
  23. Jun, S.Y.; Chung, J.Y.; Yoon, N.; Jung, E.S.; Oh, Y.H.; Hong, S.M. Tumor budding and poorly differentiated clusters in small intestinal adenocarcinoma. Cancers 2020, 12, 2199. [Google Scholar] [CrossRef]
  24. Edge, S.B.; Greene, F.L.; Schilsky, R.L.; Gaspar, L.E.; Washington, M.K.; Sullivan, D.C.; Brookland, R.K.; Brierley, J.D.; Balch, C.M.; Campton, C.C.; et al. AJCC Cancer Staging Manual, 8th ed.; Springer Nature: Cham, Switzerland, 2017. [Google Scholar]
  25. Cheuk, W.; Wong, K.O.; Wong, C.S.; Chan, J.K. Consistent immunostaining for cyclin D1 can be achieved on a routine basis using a newly available rabbit monoclonal antibody. Am. J. Surg. Pathol. 2004, 28, 801–807. [Google Scholar] [CrossRef] [PubMed]
  26. Pruneri, G.; Valentini, S.; Bertolini, F.; Del Curto, B.; Maiorano, E.; Viale, G. SP4, a novel anti-cyclin D1 rabbit monoclonal antibody, is a highly sensitive probe for identifying mantle cell lymphomas bearing the t(11;14)(q13;q32) translocation. Appl. Immunohistochem. Mol. Morphol. 2005, 13, 318–322. [Google Scholar] [CrossRef] [PubMed]
  27. Torlakovic, E.; Nielsen, S.; Vyberg, M. Antibody selection in immunohistochemical detection of cyclin D1 in mantle cell lymphoma. Am. J. Clin. Pathol. 2005, 124, 782–789. [Google Scholar] [CrossRef] [PubMed]
  28. Jang, K.Y.; Kim, Y.N.; Bae, J.S.; Chung, M.J.; Moon, W.S.; Kang, M.J.; Lee, D.G.; Park, H.S. Expression of cyclin D1 is associated with β-catenin expression and correlates with good prognosis in colorectal adenocarcinoma. Transl. Oncol. 2012, 5, 370–378. [Google Scholar] [CrossRef]
  29. Ogino, S.; Nosho, K.; Irahara, N.; Kure, S.; Shima, K.; Baba, Y.; Toyoda, S.; Chen, L.; Giovannucci, E.L.; Meyerhardt, J.A.; et al. A cohort study of cyclin D1 expression and prognosis in 602 colon cancer cases. Clin. Cancer Res. 2009, 15, 4431–4438. [Google Scholar] [CrossRef]
  30. Seicean, R.; Crisan, D.; Boers, J.E.; Mocan, T.; Seicean, A.; Funariu, G.; Ciuce, C. The prognostic role of apoptosis mediators in rectal adenocarcinoma. Hepato-Gastroenterol. 2011, 58, 1490–1494. [Google Scholar] [CrossRef]
  31. Stor, Z.; Frković, G.S.; Bracko, M.; Repse, S. Prognostic value of clinical, pathological and immunohistochemical markers in stage II colon cancer patients. Acta Chir. Lugosl. 2008, 55, 39–44. [Google Scholar] [CrossRef]
  32. Hilska, M.; Collan, Y.U.; Laine, J.V.O.; Kossi, J.; Hirsimaki, P.; Laato, M.; Roberts, P.J. The significance of tumor markers for proliferation and apoptosis in predicting survival in colorectal cancer. Dis. Colon Rectum 2005, 48, 2197–2208. [Google Scholar] [CrossRef]
  33. McKay, J.A.; Douglas, J.J.; Ross, V.G.; Curran, S.; Loane, J.F.; Ahmed, F.Y.; Cassidy, J.; McLeod, H.L.; Murray, G.I. Analysis of key cell-cycle checkpoint proteins in colorectal tumours. J. Pathol. 2002, 196, 386–393. [Google Scholar] [CrossRef]
  34. Von Stockmar-Von Wangenheim, C.A.; Monig, S.P.; Schneider, P.M.; Landsberg, S.; Drebber, U.; Holscher, A.H.; Dienes, H.P.; Baldus, S.E. p16, cyclin D1 and Rb expression in colorectal carcinomas: Correlations with clinico-pathological parameters and prognosis. Mol. Med. Rep. 2008, 1, 27–32. [Google Scholar]
  35. Lyall, M.S.; Dundas, S.R.; Curran, S.; Murray, G.I. Profiling markers of prognosis in colorectal cancer. Clin. Cancer Res. 2006, 12, 1184–1191. [Google Scholar] [CrossRef] [PubMed]
  36. McKay, J.A.; Douglas, J.J.; Ross, V.G.; Curran, S.; Murray, G.I.; Cassidy, J.; McLeod, H.L. Cyclin D1 protein expression and gene polymorphism in colorectal cancer. Aberdeen Colorectal Initiative. Int. J. Cancer 2000, 88, 77–81. [Google Scholar] [CrossRef] [PubMed]
  37. Bhatavdekar, J.M.; Patel, D.D.; Chikhlikar, P.R.; Shah, N.G.; Vora, H.H.; Ghosh, N.; Trivedi, T.I. Molecular markers are predictors of recurrence and survival in patients with Dukes B and Dukes C colorectal adenocarcinoma. Dis. Colon Rectum 2001, 44, 523–533. [Google Scholar] [CrossRef]
  38. Bahnassy, A.A.; Zekri, A.R.; El-Houssini, S.; El-Shehaby, A.M.; Mahmoud, M.R.; Abdallah, S.; El-Serafi, M. Cyclin A and cyclin D1 as significant prognostic markers in colorectal cancer patients. BMC Gastroentero. 2004, 4, 22. [Google Scholar] [CrossRef] [PubMed]
  39. Moore, H.G.; Shia, J.; Klimstra, D.S.; Ruo, L.; Mazumdar, M.; Schwartz, G.K.; Minsky, B.D.; Saltz, L.; Guillem, J.G. Expression of p27 in residual rectal cancer after preoperative chemoradiation predicts long-term outcome. Ann. Surg. Oncol. 2004, 11, 955–961. [Google Scholar] [CrossRef] [PubMed]
  40. Holland, T.A.; Elder, J.; McCloud, J.M.; Hall, C.; Deakin, M.; Fryer, A.A.; Elder, J.B.; Hoban, P.R. Subcellular localisation of cyclin D1 protein in colorectal tumours is associated with p21(WAF1/CIP1) expression and correlates with patient survival. Int. J. Cancer 2001, 95, 302–306. [Google Scholar] [CrossRef] [PubMed]
  41. Maeda, K.; Chung, Y.; Kang, S.; Ogawa, M.; Onoda, N.; Nishiguchi, Y.; Ikehara, T.; Nakata, B.; Okuno, M.; Sowa, M. Cyclin D1 overexpression and prognosis in colorectal adenocarcinoma. Oncology 1998, 55, 145–151. [Google Scholar] [CrossRef]
  42. Maeda, K.; Chung, Y.S.; Kang, S.M.; Ogawa, M.; Onoda, N.; Nakata, B.; Nishiguchi, Y.; Ikehara, T.; Okuno, M.; Sowa, M. Overexpression of cyclin D1 and p53 associated with disease recurrence in colorectal adenocarcinoma. Int. J. Cancer 1997, 74, 310–315. [Google Scholar] [CrossRef]
  43. Tsai, H.L.; Yeh, Y.S.; Chang, Y.T.; Yang, I.P.; Lin, C.H.; Kuo, C.H.; Juo, S.H.; Wang, J.Y. Co-existence of cyclin D1 and vascular endothelial growth factor protein expression is a poor prognostic factor for UICC stage I-III colorectal cancer patients after curative resection. J. Surg. Oncol. 2013, 107, 148–154. [Google Scholar] [CrossRef]
  44. Belt, E.J.; Brosens, R.P.; Delis-van Diemen, P.M.; Bril, H.; Tijssen, M.; van Essen, D.F.; Heymans, M.W.; Belien, J.A.; Stockmann, H.B.; Meijer, S.; et al. Cell cycle proteins predict recurrence in stage II and III colon cancer. Ann. Surg. Oncol. 2012, 19 (Suppl. 3), S682–S692. [Google Scholar] [CrossRef]
  45. Mao, Y.; Li, Z.; Lou, C.; Zhang, Y. Expression of phosphorylated Stat5 predicts expression of cyclin D1 and correlates with poor prognosis of colonic adenocarcinoma. Int. J. Colorectal Dis. 2011, 26, 29–35. [Google Scholar] [CrossRef]
  46. Fang, Y.J.; Lu, Z.H.; Wang, G.Q.; Pan, Z.Z.; Zhou, Z.W.; Yun, J.P.; Zhang, M.F.; Wan, D.S. Elevated expressions of MMP7, TROP2, and survivin are associated with survival, disease recurrence, and liver metastasis of colon cancer. Int. J. Color. Dis. 2009, 24, 875–884. [Google Scholar] [CrossRef]
  47. Kouraklis, G.; Theocharis, S.; Vamvakas, P.; Vagianos, C.; Glinavou, A.; Giaginis, C.; Sioka, C. Cyclin D1 and Rb protein expression and their correlation with prognosis in patients with colon cancer. World J. Surg. Oncol. 2006, 4, 5. [Google Scholar] [CrossRef]
  48. Bondi, J.; Husdal, A.; Bukholm, G.; Nesland, J.M.; Bakka, A.; Bukholm, I.R. Expression and gene amplification of primary (A, B1, D1, D3, and E) and secondary (C and H) cyclins in colon adenocarcinomas and correlation with patient outcome. J. Clin. Pathol. 2005, 58, 509–514. [Google Scholar] [CrossRef] [PubMed]
  49. Bondi, J.; Bukholm, G.; Nesland, J.M.; Bukholm, I.R. Expression of non-membranous beta-catenin and gamma-catenin, c-Myc and cyclin D1 in relation to patient outcome in human colon adenocarcinomas. APMIS 2004, 112, 49–56. [Google Scholar] [CrossRef] [PubMed]
  50. Shukla, A.; Sachan, R.; Maurya, M.; Patel, M.L.; Sankhwar, P. ROC-analysis derived immunohistochemical p53 cut-off scores as an adjunct to routine histopathology for better diagnostic compartmentalisation of cervical lesions. Int. J. Appl. Basic Med. Res. 2022, 12, 177–184. [Google Scholar] [CrossRef]
  51. Reis-Filho, J.S.; Savage, K.; Lambros, M.B.; James, M.; Steele, D.; Jones, R.L.; Dowsett, M. Cyclin D1 protein overexpression and CCND1 amplification in breast carcinomas: An immunohistochemical and chromogenic in situ hybridisation analysis. Mod. Pathol. 2006, 19, 999–1009. [Google Scholar] [CrossRef] [PubMed]
  52. Han, E.K.; Sgambato, A.; Jiang, W.; Zhang, Y.J.; Santella, R.M.; Doki, Y.; Cacace, A.M.; Schieren, I.; Weinstein, I.B. Stable overexpression of cyclin D1 in a human mammary epithelial cell line prolongs the S-phase and inhibits growth. Oncogene 1995, 10, 953–961. [Google Scholar]
  53. Freeman, R.S.; Estus, S.; Johnson, E.M., Jr. Analysis of cell cycle-related gene expression in postmitotic neurons: Selective induction of cyclin D1 during programmed cell death. Neuron 1994, 12, 343–355. [Google Scholar] [CrossRef]
  54. Atadja, P.; Wong, H.; Veillete, C.; Riabowol, K. Overexpression of cyclin D1 blocks proliferation of normal diploid fibroblasts. Exp. Cell Res. 1995, 217, 205–216. [Google Scholar] [CrossRef]
  55. Musgrove, E.A.; Caldon, C.E.; Barraclough, J.; Stone, A.; Sutherland, R.L. Cyclin D as a therapeutic target in cancer. Nat. Rev. Cancer 2011, 11, 558–572. [Google Scholar] [CrossRef]
  56. Dragoj, M.; Milosevic, Z.; Bankovic, J.; Dinic, J.; Pesic, M.; Tanic, N.; Stankovic, T. Association of CCND1 overexpression with KRAS and PTEN alterations in specific subtypes of non-small cell lung carcinoma and its influence on patients’ outcome. Tumor Biol. 2015, 36, 8773–8780. [Google Scholar] [CrossRef] [PubMed]
  57. Dragnev, K.H.; Ma, T.; Cyrus, J.; Galimberti, F.; Memoli, V.; Busch, A.M.; Tsongalis, G.J.; Seltzer, M.; Johnstone, D.; Erkmen, C.P.; et al. Bexarotene plus erlotinib suppress lung carcinogenesis independent of KRAS mutations in two clinical trials and transgenic models. Cancer Prev. Res. 2011, 4, 818–828. [Google Scholar] [CrossRef] [PubMed]
  58. Ziemke, E.K.; Dosch, J.S.; Maust, J.D.; Shettigar, A.; Sen, A.; Welling, T.H.; Hardiman, K.M.; Sebolt-Leopold, J.S. Sensitivity of KRAS-mutant colorectal cancers to combination therapy that cotargets MEK and CDK4/6. Clin. Cancer Res. 2016, 22, 405–414. [Google Scholar] [CrossRef] [PubMed]
  59. Shimura, T.; Kakuda, S.; Ochiai, Y.; Nakagawa, H.; Kuwahara, Y.; Takai, Y.; Kobayashi, J.; Komatsu, K.; Fukumoto, M. Acquired radioresistance of human tumor cells by DNA-PK/AKT/GSK3beta-mediated cyclin D1 overexpression. Oncogene 2010, 29, 4826–4837. [Google Scholar] [CrossRef]
  60. Biliran, H., Jr.; Wang, Y.; Banerjee, S.; Xu, H.; Heng, H.; Thakur, A.; Bollig, A.; Sarkar, F.H.; Liao, J.D. Overexpression of cyclin D1 promotes tumor cell growth and confers resistance to cisplatin-mediated apoptosis in an elastase-myc transgene-expressing pancreatic tumor cell line. Clin. Cancer Res. 2005, 11, 6075–6086. [Google Scholar] [CrossRef]
  61. Musgrove, E.A.; Sutherland, R.L. Biological determinants of endocrine resistance in breast cancer. Nat. Rev. Cancer 2009, 9, 631–643. [Google Scholar] [CrossRef]
  62. Kalish, L.H.; Kwong, R.A.; Cole, I.E.; Gallagher, R.M.; Sutherland, R.L.; Musgrove, E.A. Deregulated cyclin D1 expression is associated with decreased efficacy of the selective epidermal growth factor receptor tyrosine kinase inhibitor gefitinib in head and neck squamous cell carcinoma cell lines. Clin. Cancer Res. 2004, 10, 7764–7774. [Google Scholar] [CrossRef]
  63. Smalley, K.S.; Lioni, M.; Dalla Palma, M.; Xiao, M.; Desai, B.; Egyhazi, S.; Hansson, J.; Wu, H.; King, A.J.; Van Belle, P.; et al. Increased cyclin D1 expression can mediate BRAF inhibitor resistance in BRAF V600E-mutated melanomas. Mol. Cancer Ther. 2008, 7, 2876–2883. [Google Scholar] [CrossRef]
  64. Rudas, M.; Lehnert, M.; Huynh, A.; Jakesz, R.; Singer, C.; Lax, S.; Schippinger, W.; Dietze, O.; Greil, R.; Stiglbauer, W.; et al. Cyclin D1 expression in breast cancer patients receiving adjuvant tamoxifen-based therapy. Clin. Cancer Res. 2008, 14, 1767–1774. [Google Scholar] [CrossRef]
Figure 1. Cyclin D1 expression in normal mucosa of the small intestine. The cyclin D1 was expressed in the transitional zone of the crypt at the lower portion of the gland of normal small intestinal mucosa (original magnification ×200).
Figure 1. Cyclin D1 expression in normal mucosa of the small intestine. The cyclin D1 was expressed in the transitional zone of the crypt at the lower portion of the gland of normal small intestinal mucosa (original magnification ×200).
Cancers 15 05032 g001
Figure 2. Representative images of cyclin D1 expression in SIAC. Cyclin D1Low showed (A) a score of 0, (B) a score of 1, and (C) a score of 2. Cyclin D1High had (D) a score of 14, (E) a score of 18, and (F) a score of 30 (original magnification ×100).
Figure 2. Representative images of cyclin D1 expression in SIAC. Cyclin D1Low showed (A) a score of 0, (B) a score of 1, and (C) a score of 2. Cyclin D1High had (D) a score of 14, (E) a score of 18, and (F) a score of 30 (original magnification ×100).
Cancers 15 05032 g002
Figure 3. Univariate survival analysis of cyclin D1High in SIAC patients. The patients with cyclin D1High had significantly longer survival times than those with cyclin D1Low (p = 0.005).
Figure 3. Univariate survival analysis of cyclin D1High in SIAC patients. The patients with cyclin D1High had significantly longer survival times than those with cyclin D1Low (p = 0.005).
Cancers 15 05032 g003
Figure 4. Prognostic impact of cyclin D1 expression in SIAC depending on tumor stage. (A) Cyclin D1High significantly predicted the longer OS of patients in the lower stage group (stages I and II, n = 105; p < 0.001), while (B) no significant difference in survival was identified between the cyclin D1High and cyclin D1Low groups in the higher stage group (stage III, n = 106; p = 0.629).
Figure 4. Prognostic impact of cyclin D1 expression in SIAC depending on tumor stage. (A) Cyclin D1High significantly predicted the longer OS of patients in the lower stage group (stages I and II, n = 105; p < 0.001), while (B) no significant difference in survival was identified between the cyclin D1High and cyclin D1Low groups in the higher stage group (stage III, n = 106; p = 0.629).
Cancers 15 05032 g004
Table 1. Association between cyclin D1 expression and clinicopathological factors in SIAC.
Table 1. Association between cyclin D1 expression and clinicopathological factors in SIAC.
Characteristics, n (%)TotalCyclin D1LowCyclin D1Highp
No. of patients 87 (37.5)145 (62.5)
Age (y)≤5052 (22.4)25 (48.1)27 (51.9)0.074
>50180 (77.6)62 (34.4)118 (65.6)
SexMale148 (63.8)58 (39.2)90 (60.8)0.481
Female84 (36.2)29 (34.5)55 (65.5)
Tumor size (cm, mean ± SD) 4.3 ± 2.44.4 ± 2.60.200
Growth pattern (n = 224) aPolypoid40 (17.9)11 (27.5)29 (72.5)0.164
Nodular17 (7.6)9 (52.9)8 (47.1)
Infiltrative167 (74.5)66 (39.5)101 (60.5)
Tumor locationDuodenum140 (60.3)55 (39.3)85 (60.7)0.289
Jejunum58 (25.0)17 (29.3)41 (70.7)
Ileum34 (14.7)15 (44.1)19 (55.9)
Histologic typeTubular205 (88.4)74 (36.1)131 (63.9)0.489
Mucinous12 (5.2)6 (50.0)6 (50.0)
Signet ring cell4 (1.7)3 (75.0)1 (25.0)
Medullary6 (2.6)2 (33.3)4 (66.7)
Undifferentiated5 (2.1)2 (40.0)3 (60.0)
DifferentiationLow grade184 (79.3)64 (34.8)120 (65.2)0.094
High grade48 (20.7)23 (47.9)25 (52.1)
Pancreatic invasionPresent87 (37.5)37 (42.5)50 (57.5)0.220
Other loop invasionPresent6 (2.6)2 (33.3)4 (66.7)1.000
Retroperitoneal seedingPresent16 (6.9)9 (56.2)7 (43.8)0.108
Lymphovascular invasion (n = 231) aAbsent118 (51.1)38 (32.2)80 (67.8)0.106
Present113 (48.9)48 (42.5)65 (57.5)
Perineural invasion (n = 231) aAbsent155 (67.1)53 (34.2)102 (65.8)0.173
Present76 (32.9)33 (43.4)43 (56.6)
Margin status (n = 218) aNo involvement 209 (95.9)78 (37.3)131 (62.7)0.491
Involved by cancer9 (4.1)2 (22.2)7 (77.8)
Chemotherapy (n = 227) aAbsent145 (63.9)49 (33.8)96 (66.2)0.183
Present82 (36.1)35 (42.7)47 (57.3)
Radiotherapy (n = 226) aAbsent200 (88.5)71 (35.5)129 (64.5)0.150
Present26 (11.5)13 (50.0)13 (50.0)
Nodal metastasis (n = 215) aAbsent109 (50.7)40 (36.7)69 (53.3)0.764
Present106 (49.3)41 (38.7)65 (61.3)
T categoryTis4 (1.7)04 (100)0.046 b
T113 (5.6)1 (7.7)12 (92.3)
T214 (6.1)4 (28.6)10 (71.4)
T368 (29.3)25 (36.8)43 (63.2)
T4133 (57.3)57 (42.9)76 (57.1)
N category (n = 215) aN0109 (50.7)40 (36.7)69 (63.3)0.865
N154 (25.1)22 (40.7)32 (59.3)
N252 (24.2)19 (36.5)33 (63.5)
Stage grouping (n = 215) aStage 04 (1.9)04 (100)0.003 b
Stage I22 (10.2)2 (9.1)20 (90.9)
Stage II83 (38.6)38 (45.8)45 (54.2)
Stage III106 (49.3)41 (38.7)65 (61.3)
KRAS (n = 186) aAbsent126 (67.7)55 (43.7)71 (56.3)0.026 b
Present60 (32.3)16 (26.7)44 (73.3)
BRAF (n = 176) aAbsent174 (98.9)66 (37.9)108 (62.1)
Present2 (1.1)1 (50.0)1 (50.0)
Low expression of cyclin D1 (cyclin D1Low); high expression of cyclin D1 (cyclin D1High); standard deviation (SD); a only calculated using cases with available information; b significant at p < 0.05.
Table 2. Association between clinicopathological factors and OS in SIAC.
Table 2. Association between clinicopathological factors and OS in SIAC.
CharacteristicsUnivariateMultivariate
Median (mo)pHR (95% CI)p
Cyclin D1 expressionCyclin D1Low24.50.005 a0.68 (0.47–0.96)0.031 a
Cyclin D1High44.4
Age (y)≤5039.90.143
>5030.0
SexMale30.00.873
Female31.4
Tumor size (cm) 1.00 (0.94–1.07) b0.970
Growth pattern (n = 224) cPolypoid48.50.408
Nodular36.2
Infiltrative26.6
Tumor locationProximal (duodenum)41.70.007 a1.34 (0.92–1.94)0.125
Distal (jejunum and ileum)22.5
Histologic typeTubular36.20.578
Nontubular28.2
DifferentiationLow grade36.20.397
High grade29.1
Pancreatic invasionAbsent36.20.931
Present31.1
Other loop invasionAbsent36.20.044 a0.98 (0.22–4.27)0.976
Present5.1
Retroperitoneal seedingAbsent37.4<0.001 a2.53 (1.28–4.98)0.007 a
Present14.0
Lymphovascular invasion (n = 231) cAbsent66.6<0.001 a1.68 (1.33–2.50)0.010 a
Present17.8
Perineural invasion (n = 231) cAbsent47.60.004 a 0.329
Present18.7
Margin status (n = 218) cNo involvement 36.20.636
Involved by cancer15.9
Chemotherapy (n = 227) cAbsent37.40.314
Present29.1
Radiotherapy (n = 226) cAbsent39.70.005 a1.34 (0.82–2.21)0.247
Present22.0
Nodal metastasis (n = 215) cAbsent133.7<0.001 a
Present21.6
T category (n = 228) cT1-T2d<0.001 a1.30 (0.57–2.96)0.528
T3-T426.3
N category (n = 215) cN0133.7<0.001 a 0.001 a
N128.2 1.68 (1.07–2.63)0.024 a
N217.8 2.41 (1.51–3.83)<0.001 a
Stage grouping (n = 211) cStage Id<0.001 a
Stage II60.4
Stage III21.6
KRAS (n = 186) cAbsent39.70.098
Present21.0
BRAF (n = 176) cAbsent30.00.682
Present22.6
Hazard ratio (HR); confidence interval (CI); a significant at p < 0.05.; b displayed as HR with 95% CI; c only calculated using cases with available information.; d could not be calculated because >50% of the patients were alive.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jun, S.-Y.; Hong, S.-M.; Jang, K.-T. Prognostic Significance of Cyclin D1 Expression in Small Intestinal Adenocarcinoma. Cancers 2023, 15, 5032. https://doi.org/10.3390/cancers15205032

AMA Style

Jun S-Y, Hong S-M, Jang K-T. Prognostic Significance of Cyclin D1 Expression in Small Intestinal Adenocarcinoma. Cancers. 2023; 15(20):5032. https://doi.org/10.3390/cancers15205032

Chicago/Turabian Style

Jun, Sun-Young, Seung-Mo Hong, and Kee-Taek Jang. 2023. "Prognostic Significance of Cyclin D1 Expression in Small Intestinal Adenocarcinoma" Cancers 15, no. 20: 5032. https://doi.org/10.3390/cancers15205032

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