Skip to main content

Causal associations of Sjögren’s syndrome with cancers: a two-sample Mendelian randomization study

Abstract

Background

Several observational studies have explored the associations between Sjögren’s syndrome (SS) and certain cancers. Nevertheless, the causal relationships remain unclear. Mendelian randomization (MR) method was used to investigate the causality between SS and different types of cancers.

Methods

We conducted the two-sample Mendelian randomization with the public genome-wide association studies (GWASs) summary statistics in European population to evaluate the causality between SS and nine types of cancers. The sample size varies from 1080 to 372,373. The inverse variance weighted (IVW) method was used to estimate the causal effects. A Bonferroni-corrected threshold of P < 0.0031 was considered significant, and P value between 0.0031 and 0.05 was considered to be suggestive of an association. Sensitivity analysis was performed to validate the causality. Moreover, additional analysis was used to assess the associations between SS and well-accepted risk factors of cancers.

Results

After correcting the heterogeneity and horizontal pleiotropy, the results indicated that patients with SS were significantly associated with an increased risk of lymphomas (odds ratio [OR] = 1.0010, 95% confidence interval [CI]: 1.0005–1.0015, P = 0.0002) and reduced risks of prostate cancer (OR = 0.9972, 95% CI: 0.9960–0.9985, P = 2.45 × 10−5) and endometrial cancer (OR = 0.9414, 95% CI: 0.9158–0.9676, P = 1.65 × 10−5). Suggestive associations were found in liver and bile duct cancer (OR = 0.9999, 95% CI: 0.9997–1.0000, P = 0.0291) and cancer of urinary tract (OR = 0.9996, 95% CI: 0.9992–1.0000, P = 0.0281). No causal effect of SS on other cancer types was detected. Additional MR analysis indicated that causal effects between SS and cancers were not mediated by the well-accepted risk factors of cancers. No evidence of the causal relationship was observed for cancers on SS.

Conclusions

SS had significant causal relationships with lymphomas, prostate cancer, and endometrial cancer, and suggestive evidence of association was found in liver and bile duct cancer and cancer of urinary tract, indicating that SS may play a vital role in the incidence of these malignancies.

Introduction

Sjögren’s syndrome (SS) is a systemic chronic autoimmune disorder characterized by lymphocytic infiltration of the exocrine glands including salivary and lacrimal glands, which lead to significant loss of the secretory function [1]. SS is among the most common autoimmune diseases, along with systemic lupus erythematosus and progressive systemic sclerosis. Its overall prevalence varies from 0.1 to 4.8% [2]. It is more prevalent in women than in men (average female to male ratio 9:1), and the susceptible population are young women aged 20 s to 30 s and women after the menopause in the mid-50 s. SS can impact multiple organ systems including respiratory system, digestive system, urinary system, circulatory system, and nervous system which lead to different symptoms such as interstitial pneumonitis, tracheobronchial sicca, autoimmune hepatitis, primary biliary cholangitis, interstitial nephritis, pericarditis, pulmonary hypertension, and sensory neuropathies [1]. Clinically, accurate diagnosis and individual treatment for SS are often challenging due to the complexity of etiology and the diversity of manifestation among SS patients. The pathogenesis of SS can be multifactorial. Individuals with genetic predisposition are thought to develop SS through the effect of certain environmental factors; however, the underlying causes and detailed mechanisms remain unclear [3].

Epidemically, patients with Sjögren’s syndrome history seemed to be under higher risks for developing certain cancers. Several studies have investigated the potential connection between SS and different cancers, among which the association between SS and lymphomas was mostly studied. The results indicated that patients with SS tend to have a significantly higher lymphoma risk than healthy individuals [4, 5]. However, the associations between SS and other types of cancers were controversial. A prospective cohort study from Spain showed that SS is associated with the development of thyroid, oral cavity, and gastric cancer [6]. In a retrospective cohort study from Korea, in addition to non-Hodgkin lymphoma, SS patients also have an increased risk of solid cancers including oropharynx, thyroid, and lung cancers [7]. Another retrospective cohort study from England illustrated that patients with SS were not at higher risk for other cancers, such as lung cancer, breast cancer, ovarian cancer, pancreatic cancer, and melanoma, except for lymphoma [8]. Goulabchand et al. [9] found that SS patients were less likely to develop breast cancer and more likely to develop thyroid cancer observed in clinical cohort in France. Brom et al. [10] showed that compared to overall population, women in Argentina with SS were more likely to develop multiple myeloma, breast cancer, and tongue cancer.

The pathophysiological mechanisms about how SS may lead to certain malignancy, that is, whether there are causal effects of SS on cancers remains to be elucidated. Current researches suggests that shared genetic and environmental risk factors or process may contribute to oncogenesis among SS patients. The correlation between DNA methylation and disease progression in SS patients has been studied a lot. Major methylation alterations presenting in B cells and the genetic at-risk loci in SS patients were identified, and the methylation status of B cells was proved to be strongly correlated with disease progression [11]. Imgenberg-Kreuz et al. [12] found prominent hypomethylation of interferon (IFN) regulated genes in whole blood and CD19+ B cells, which resulted in increased expression of these genes. DNA methylation abnormalities are known to cause chromosomal instability, the activation of various oncogenes, or transcriptional inactivation of tumor suppressor genes, thus leading to oncogenesis [13, 14]. Wang et al. [15] identified a small cluster of differently expressed miRNA in SS including miR-146a-5p and miR-30b-5p. miR-146a is significantly upregulated in SS patients and is involved in the upregulation of phagocytic activity and in the reduction of inflammatory cytokine production [16]. miR-146a has been considered as either an oncogene or tumor suppressor gene dependent on the type of cancers [17,18,19]. miR-30b is inversely correlated with the expression of B cell activating factor BAFF in SS, which is associated with B cell tolerance disruption and increased autoantibody production [15]. At the same time, imbalance of miR-30b especially plays an important role in the occurrence and development of tumors [20, 21]. HLA class II genes have been identified as the strongest genetic risk factors for SS [22] and also play an important role in cancers through T cell priming, generation of strong cytotoxicity, and HLA gene methylation [23]. Additionally, chronic inflammation and tissue damage induced by SS may release cytokines such as TNF-α, IFN-γ, IL-6, and chemokines such as CXCL13 and CXCR5 which have been proved to play a role in the development of cancer through multiple mechanisms [24,25,26,27]. In summary, SS may play a vital role in the occurrence and development of cancers, and further studies are needed to explore the specific mechanism.

Mendelian randomization (MR), an epidemiological genetic approach, is applied to estimate the causal effect between exposure and outcome [28]. It uses genetic variants as instrumental variables (IVs). According to Mendel’s second law, genotypes are randomly assigned during gamete formation based on parental genotypes. Consequently, the results of MR will not be influenced by potential confounders or reverse causation [29].

In this study, using two-sample MR analysis based on the published data of genome-wide association studies (GWASs) in European population, we explored the causal effect of SS on nine types of cancers.

Methods

Study design

This study evaluated whether SS was causally related to cancer risks using two-sample MR analysis based on the summary-level genetic data from previous studies and IEU OpenGWAS database in European population. Instrument variables (IVs) should follow three assumptions: (1) the IVs should be robustly associated with SS; (2) the IVs should not be associated with confounders of the SS-cancers association; (3) the IVs should influence cancers only through SS, not through any other variables (Fig. 1).

Fig. 1
figure 1

Study design of the Mendelian randomization analysis between Sjögren’s syndrome and cancers

The data source of SS

In 2013, the Sjögren’s Genetics Network (SGENE) published the first large-scale genomic study of SS of European descent, which identified seven loci [22]. In 2017, Li et al. [30] identified a new susceptibility locus of SS of European ancestry, OAS1. Carapito et al. [31] also found a new locus of European ancestry, MICA. The Sjögren’s International Collaborative Clinical Alliance (SICCA) reported the first multiethnic GWAS study of SS but did not identify new locus in European population [32]. Lastly, Khatri et al. [33] identified ten novel Sjögren’s genetic susceptibility loci of European ancestry. We combined the outcomes of these studies and obtained seventy-one single-nucleotide polymorphisms (SNPs) from nineteen loci.

The data source of cancers

Summary statistics of multiple cancers in European population were obtained from IEU OpenGWAS database (https://gwas.mrcieu.ac.uk/) and previous studies. The GWAS summary data of overall breast cancer (228,951 participants) and its molecular subtypes including luminal A-like (28,140 participants), luminal B-like/human epidermal growth factor receptor 2 (HER2)-positive-like (22,497 participants), luminal B/HER2-negative-like (22,594 participants), HER2-enriched-like (21,533 participants), and triple-negative (22,821 participants) was extracted from the Breast Cancer Association Consortium (BCAC) [34, 35]. The data of lung cancer was obtained from the International Lung Cancer Consortium (ILCCO), including 27,209 participants were overall lung cancer, 18,336 participants were lung adenocarcinoma, and 18,313 participants were squamous cell lung cancer [36]. The summary statistics of endometrial cancer were obtained from the study of O’Mara et al. [37] (121,885 participants). The GWAS summary data on thyroid cancer was from the study of Köhler et al. [38] (1080 participants). The data of oral cavity and pharyngeal cancer was from Oncoarray oral cavity and oropharyngeal cancer [39]. The summary statistics for prostate cancer (182,625 participants), lymphomas (361,194 participants), liver and bile duct cancer (372,366 participants), and cancer of urinary tract (361,194 participants) were all extracted from UK Biobank [40] (Table 1).

Table 1 Details of cancers and risk factors used in the study

Selection of instrumental variables

The SNPs that were highly associated with exposure (P < 5 × 10−8) and with no linkage disequilibrium (r2 < 0.001 and clump window > 10,000 kb) were used as IVs. For SNPs missed in the outcome data, proxies were identified in high linkage disequilibrium (r2 > 0.8). If no appropriate proxy SNP was available, SNPs were excluded in our analysis. We also harmonized the data so that estimates of SNP-exposure and SNP-outcome were based on the same allele and remove the SNPs which were palindromic with intermediate allele frequencies. To evaluate the correlation strength and avoid bias caused by weak IVs, we calculated the R2 and F-statistic. R2 statistic is used to measure the proportion of variability of exposure phenotype that is explained by instruments. When the F-statistic was greater than 10, it was considered sufficient [41].

The data source of risk factors for cancers

To explore whether the causal effect of SS on cancers was mediated by risk factors of cancers, we conducted the additional MR analysis. We estimated the associations between SS and generally accepted risk factors of cancers including years of education [42, 43], cigarettes consumption, alcohol consumption [44], and body mass index (BMI) [45]. The GWAS summary statistics of years of education were from the Social Science Genetic Association Consortium (SSGAC) [46]. The data of cigarettes and alcohol consumption were retrieved from the GWAS and Sequencing Consortium of Alcohol and Nicotine use (GSCAN) [47], and the data of BMI was retrieved from the Neale Lab (http://www.nealelab.is/uk-biobank) (Table 1). SS was treated as exposure, and these risk factors were taken as outcomes to perform MR analysis. Inverse variance weighted (IVW) method is used to estimate the causality. P < 0.05 was regarded as significant.

Statistical analysis

To estimate the causal effects of SS on different cancers, the IVW method was as the main analysis. When all assumptions are met, IVW method has the highest statistical power [48]. MR-Egger method and weighted median method were used as complementary analyses. MR-Egger method is able to provide a consistent result under the instrument strength independent of direct effect (InSIDE) assumption [49]. Weighted median method can provide a consistent estimate even 50% IVs are invalid [50]. Heterogeneity was evaluated by Cochran’s Q test. The random-effect IVW method was applied in case heterogeneity was observed. MR-Egger regression test was used to assess horizontal pleiotropy. Pleiotropic effects across genetic variants are represented by the intercept [49]. We also used MR pleiotropy residual sum and outlier (MR-PRESSO) test to detect pleiotropic SNPs, correct the horizontal pleiotropy by removing the outliers, and test whether there are significant differences in the causal estimates before and after correcting outliers via distortion test [51]. Additionally, we performed “leave-one-out” analysis to test the stability of our findings.

All statistical analyses were performed in R v4.2.1 with R packages (TwoSampleMR, MR-PRESSO). A threshold of P < 0.0031 (0.05/16 outcomes) using the Bonferroni correction was regarded as significant evidence of an association, and 0.0031 < P < 0.05 was considered as suggestive evidence of an association.

Results

Selection of genetic instrument

After screening for close associations (P < 5 × 10−8) and independence (r2 < 0.001), we obtained twelve SNPs as instrumental variables (Supplementary Table 1). Since the sample size of different cancer types is various, the F-statistics ranged from 302 to 52,234 for different cancers among our study, which suggested the instruments used strongly predicted SS in our study (Table 1). For missed SNPs in outcomes, rs2304256, rs4841465, rs909685, rs1978273, rs4728142, rs12373168, and rs4969331 were chosen as proxies for rs11085725, rs11250098, rs2069235, rs2293765, rs3757387, rs7210219, and rs8071514, but other missed SNPs (rs10774671, rs2431697, rs3135394, and rs7119038) did not find their suitable proxies.

Causal effects between SS and cancers

The main results of the MR estimates between SS and each cancer are presented in Table 2. From the results of IVW method, we detected that genetically predicted SS significantly increased the 0.10% risk of lymphomas (OR = 1.0010, 95% CI: 1.0007–1.0012, P = 2.26 × 10−11) and reduced the 0.28% risk of prostate cancer (OR = 0.9972, 95% CI: 0.9960–0.9985, P = 2.45 × 10−5) and the 5.86% risk of endometrial cancer (OR = 0.9414, 95% CI: 0.9158–0.9676, P = 1.65 × 10−5). There was suggestive evidence of causality between SS and liver and bile duct cancer (OR = 0.9999, 95% CI: 0.9997–1.0000, P = 0.0291), cancer of urinary tract (OR = 0.9996, 95% CI: 0.9993–0.9999, P = 0.0038), and oral cavity and pharyngeal cancer (OR = 1.1068, 95% CI: 1.0068–1.2167, P = 0.0357). For overall breast cancer, overall lung cancer, and thyroid cancer, no significant association was detected. To further explore whether SS is causally related to certain subtypes of breast cancer or lung cancer, subgroup analysis was conducted. The results showed no causal relationship between SS and breast cancer molecular subtypes (luminal A-like, luminal B/HER2-positive-like, luminal B/HER2-negative-like, HER2-enriched-like and triple-negative breast cancer) or lung cancer histological subtypes (lung adenocarcinoma and squamous cell lung cancer). MR-Egger and weighted median methods provided results of the same direction and magnitude with IVW method.

Table 2 The results of Mendelian randomization analysis for SS on cancers

The scatter plots illustrated the estimated impact of instrumental variables on SS and cancers. The rising lines in the plot demonstrated the positive relationship between SS and cancers (Supplementary Fig. 1).

Sensitivity analysis

To test the reliability of MR analysis results, we conducted sensitivity analysis. The results demonstrated significant heterogeneity in overall breast cancer, luminal A-like breast cancer, squamous cell lung cancer, lymphomas, cancer of urinary tract and oral cavity, and pharyngeal cancer (Q-value < 0.05) (Table 3). Thus, we corrected the heterogeneity with the random-effect IVW method. The causal effect of SS on oral cavity and pharyngeal cancer (OR = 1.1068, 95% CI: 0.9445–1.2970, P = 0.2098) was no longer statistically significant. The causal association between SS and overall breast cancer (OR = 0.9952, 95% CI: 0.9649–1.0264, P = 0.7595), luminal A-like breast cancer (OR = 1.0103, 95% CI: 0.9761–1.0457, P = 0.5597), squamous cell lung cancer (OR = 1.0343, 95% CI: 0.9198–1.1631, P = 0.5731), lymphomas (OR = 1.0010, 95% CI: 1.0005–1.0015, P = 0.0002), cancer of urinary tract (OR = 0.9996, 95% CI: 0.9992–1.0000, P = 0.0281) remained consistent with the previous results.

Table 3 The results of heterogeneity analysis for SS on cancers

Then, the MR-Egger regression and MR-PRESSO methods were used to detect horizontal pleiotropy (Table 4), and the results of MR-Egger regression test were visualized through the funnel plots (Supplementary Fig. 2). Although the MR-Egger regression test showed prostate cancer (P = 0.0338) and endometrial cancer (P = 0.0360) existed in the horizontal pleiotropy, the MR-PRESSO method did not test the horizontal pleiotropy (P = 0.1589; P = 0.1111). From the results of the MR-PRESSO method, overall breast cancer, luminal A-like breast cancer, lymphomas, and oral cavity and pharyngeal cancer were detected in the horizontal pleiotropy (P < 1.00 × 10−4; P = 0.0040; P = 0.0147; P = 0.0179). After removing the outlier SNPs (rs7210219 for overall breast cancer and luminal A-like breast cancer, rs7119038 for lymphomas, rs1978273 and rs3135394 for oral cavity and pharyngeal cancer), the causal effects of SS on overall breast cancer (OR = 0.9867, 95% CI: 0.9686–1.0052, P = 0.1576), luminal A-like breast cancer (OR = 0.9972, 95% CI: 0.9757–1.0190, P = 0.7970), and lymphomas (OR = 1.0009, 95% CI: 1.0006–1.0013, P = 1.15 × 10−6) were still consistent with previous results, and the distortion tests of MR-PRESSO were not statistically significant (P = 0.5067; P = 0.1094; P = 0.5515), the causal effect of SS on oral cavity and pharyngeal cancer was not significant (OR = 1.0309, 95% CI: 0.9059–1.1730, P = 0.6447), and the distortion tests was significant (P = 0.0153). The leave-one-out analysis indicated that no single instrumental variable affected the causal effects of SS on cancers (Supplementary Fig. 3).

Table 4 The results of horizontal pleiotropy analysis for SS on cancers

From the single SNP analysis (Table 5 and Supplementary Fig. 4), we found a potential risk SNP (rs3135394) for endometrial cancer (P = 1.55 × 10−5), prostate cancer (P = 1.28 × 10−6), and lymphomas (P = 4.41 × 10−10) through its impact on SS.

Table 5 The causal effects between single SNP of Sjögren’s syndrome and cancers

Risk factors analysis

To investigate whether the causal effects of SS on cancers were violated through recognized risk factors relating to cancers, we calculated the effects of SS on years of education, cigarettes consumption per day, alcoholic consumption per week, and BMI. The results of IVW method showed that SS was not causally related to these risk factors (all P > 0.05) (Table 6), indicating that the causal effects between SS and cancers were not influenced by these acknowledged risk factors.

Table 6 The results of the risk factors analysis

Causal effects of cancers on SS

From the results of IVW analysis, we found that genetically predicted lymphomas were significantly associated with SS (OR = 2.21 × 1048, 95% CI: 5.03 × 1018–9.75 × 1077, P = 0.0014). The causal relationships between genetically predicted prostate cancer, endometrial cancer, liver and bile duct cancer, cancer of urinary tract, and SS were not detected (Supplementary Table 2). Then, we conducted sensitivity analysis to test the reliability of the results. Heterogeneity was found in lymphomas and liver and bile duct cancer (Supplementary Table 3). MR-Egger regression test did not detect the horizontal pleiotropy. MR-PRESSO test showed the horizontal pleiotropy in lymphomas (Supplement Table 4). After correcting the heterogeneity and removing the outlier SNPs, the causal effect of lymphomas on SS was not significant (OR = 3.07 × 104, 95% CI: 2.10 × 10−48–4.47 × 1056, P = 0.8661), and the causal effect of liver and bile duct cancer was still not significant (OR = 2.68 × 1041, 95% CI: 9.62 × 10−59–7.46 × 10140, P = 0.4142).

Discussion

In this study, we explored the causal associations between SS and nine types of cancers respectively in the European population using the two-sample MR approach. The results demonstrated that SS was significantly associated with increased risks of lymphomas (OR = 1.0010, 95% CI: 1.0005–1.0015, P = 0.0002) and reduced risks of prostate cancer (OR = 0.9972, 95% CI: 0.9960–0.9985, P = 2.45 × 10−5) and endometrial cancer (OR = 0.9414, 95% CI: 0.9158–0.9676, P = 1.65 × 10−5). Suggestive evidence (0.0031 < P < 0.05) between SS and liver and bile duct cancer (OR = 0.9999, 95% CI: 0.9997–1.0000, P = 0.0291) and cancer of urinary tract (OR = 0.9996, 95% CI: 0.9992–1.0000, P = 0.0281) was detected, which indicated that there is a possible causal association between SS and live and bile duct cancer and cancer of urinary tract. The causal association was not observed in other types of cancers including overall breast cancer, overall lung cancer, thyroid cancer, and oral cavity and pharyngeal cancer. Subgroup analysis revealed that SS was not associated with the risk of different molecular subtypes of breast cancer (luminal A-like, luminal B/HER2-positive-like, luminal B/HER2-negative-like, HER2-enriched-like and triple-negative breast cancer) or the histological subtypes of lung cancer (lung adenocarcinoma and squamous cell lung cancer). At the same time, the results of risk factors analysis demonstrated no association between SS and potential risk factors (years of education, cigarettes consumption per day, alcoholic consumption per week and BMI), suggesting that the causal effect of SS on cancers we studied was not mediated by these common risk factors of cancers. We also found that a potential risk SNP (rs3135394) which is in the HLA-DRA promoter region might play an important role that determining the causal associations between SS and prostate cancer, endometrial cancer, and lymphomas. In the reverse direction, we did not find the causal relationship between cancers and SS. As far as we know, this is the first MR study to investigate the causal relationship between SS and cancers.

Among various cancers, the relationship between SS and lymphomas has been the most studied. Our findings remain consistent with previous studies showing that SS patients had an increased risk of lymphomas [52, 53]. Previous studies have shown that in the salivary glands of SS patients, immune complexes stimulate the polyclonal expansion of rheumatoid factor-reactive B cells. B cell activating factor (BAFF) stimulates the proliferation of clonal B cells when it is overproduced [54]. Suppressed IFNα leads to the survival of malignant B cells [55]. The interactions of Fms-like tyrosine kinase 3 (Flt3) and Flt3 ligand contribute to the abnormality of B cell distribution [56]. Chronic antigen stimulation and these factors promote the transformation of polyclonal to monoclonal B cell expansion [4]. The germline abnormalities of A20 (encoded by TNFAIP3) lead to the overactivation of NF-κB pathway in the continuous stimulation of B cells by autoimmunity, and the uncontrolled activation of NF-κB pathway promotes the survival of germline B cells and accumulate oncogenic mutations, which enhance the risk of lymphoma [57]. Moreover, the mutations of tumor suppressor gene p53 lead to the imbalance of cell cycle and uncontrolled cell proliferation, which promotes lymphomas development in patients with SS [58].

For cancers other than lymphomas, the conclusions among studies were inconsistent. A systematic review which included seven studies involving 22,204 SS patients demonstrated that except for lymphoma, the incidence of other malignancies was not related to SS patients [59]. A meta-analysis reviewing fourteen studies involving more than 14,523 patients with SS showed that SS was significantly associated with increased risks of thyroid cancer [60]. Recently, another meta-analysis which included twenty-five studies involving more than 47,607 SS patients found SS was significantly associated with increased risks of solid tumors, including lung, thyroid, non-melanoma skin, kidney/urinary tract, liver, and prostate cancers [61]. Observational studies are easy to be influenced by potential residual bias, insufficient power owing to small sample size, and reverse causality, which may lead to the controversial conclusions. MR designs can simulate randomized controlled trails and avoid reverse causality and confounding bias of observational studies, which are more practical to reveal causal association. Although large-scale randomized controlled trails (RCTs) are advocated for exploring the causal association between certain exposure and diseases, MR could provide reliable evidence for researchers to decide whether the time-consuming and costly RCTs should be further conducted. Therefore, our study used MR method with a large sample size to explore the causal effect of SS on cancers.

Concerning breast cancer, lung cancer, thyroid cancer, and oral cavity and pharyngeal cancer, we obtained the similar results to other studies based on the European population [6, 8, 9, 62]. Causal relationship between SS and these cancers were not proven yet.

Interestingly, we observed significant associations between SS and reduced risks of prostate cancer and endometrial cancer. For prostate cancer, some researches did not observe the difference in the incidence risk of prostate cancer compared with controls [6, 53, 63]. Wang et al. [64] reported that in Taiwanese population, a significant correlation was found between SS and the elevated risk of prostate cancer. However, the conflicting results may be attributed to different genetic and environment background of different ethnic groups [2]. So, further studies based on a larger sample size in European population or comparative analysis among different ethnic groups are needed. For endometrial cancer, our study found a reduced risk of endometrial cancer in patients with SS (OR = 0.9414, 95% CI: 0.9158–0.9676). Two observational studies also suggested reduced incidence of carcinoma of uterine corpus among patient with SS [6, 9], but these differences did not reach statistical significance. Lower level of dehydroepiandrosterone-sulfate pro-hormone is detected in SS patients compared to healthy controls, and the conversion of dehydroepiandrosterone to dihydrotestosterone and estrogen is impaired in patients with SS [65]. This abnormity leads to a lower estrogen exposure in patients with SS compared to their controls [66]. Endometrial cancer has been proved to be driven by estrogen [67], which may explain why SS patients tend to have lower incidence of endometrial cancer. HLA-DRA, a candidate risk gene, might provide a novel point for the causal associations between SS and prostate cancer, endometrial cancer, and lymphomas. HLA-DRA encodes the HLA-DR alpha chain. HLA-DR is known to help presenting tumor-associated antigens (TAA) to T cells, which then triggers series of immune response in the tumor microenvironment [68]. Several studies have explored the association between HLA-DRA and cancers and suggested a role HLA-DRA played during the development of multiple cancers including diffuse large B cell lymphoma [69], endometrial cancer [70], prostate cancer [71], hepatocellular carcinoma [72], cervical cancer [73], bladder cancer [74], colorectal cancer [75], and ovarian cancer [76]. Additionally, HLA-DRA may represent as a predictive marker for cancer risk and prognosis. SNP rs3135394 is located in the HLA-DRA promoter region, which encodes the HLA-DR alpha chain. From the single SNP analysis, we found rs3135394 might play a vital role in the causal relationship between SS and endometrial cancer (P = 1.55 × 10−5), prostate cancer (P = 1.28 × 10−6), and lymphomas (P = 4.41 × 10−10), but further studies on the mechanism of rs3135394 are still warranted.

We also revealed the suggestive evidence of associations between SS and reduced risks of cancer of urinary tract and liver and bile duct cancer. For urinary tract cancer, Theander et al. [53] and Treppo et al. [63] reported that there is no association between SS and kidneys/urinary tract cancer in European population. Zhou et al. [77] illustrated that SS was associated with an increased risk of urinary tract cancer in Chinese population. So, further studies focused on urinary tract cancer and based on a larger sample size in European population, or comparative analysis among different ethnic groups is needed. A few studies showed that SS was correlated with an increased risk of liver cancer [64], but our study suggested lower risk for liver and bile duct cancer among patients with SS. MR approach is based on the GWAS data, of which the sample size is hundreds of thousands of or even millions of genetic variants, much larger than the observational studies we referred to. In addition, the study design and analytical principles of observational studies and MR are different, and the results of observational studies tend to be affected by reverse causality and confounding variables, which may lead to biased associations or conclusions. These aspects may explain the discordance between the observational studies and our MR analysis. Thus, further studies are warranted.

Immunosuppressive therapy is the major systemic treatment for SS [78]. However, differences in malignancy rates have been observed between patients with autoimmune diseases who used certain immunosuppressive agents and the general population [79, 80]. It is possible that immunosuppressive agents used for long-term therapy of SS may mediate the causal relationship between SS and cancers. Methotrexate, an immunoregulatory and anti-inflammatory agent, is often used to treat SS [81]. In several retrospective cohort studies, patients with rheumatoid arthritis or psoriatic arthritis who use methotrexate have been linked to an increased risk of skin cancer [82,83,84]. Rituximab is a chimeric anti-CD20 monoclonal antibody, which helps to relieve the sicca symptoms of SS by depleting B cells and impairing the number and function of T cells [85]. A retrospective cohort study in Sweden showed no difference in cancer risk between multiple sclerosis patients using rituximab and the general population [86]. Leflunomide is another immunosuppressive and anti-inflammatory agent use in SS treatment. However, there is currently no evidence of correlation between leflunomide administration and higher cancer incidence. Some studies even suggested anti-cancer potential of leflunomide [87]. It remains unclear whether immunosuppressive agents or antibodies used for SS treatment affects the casual relationship between SS and carcinogenesis.

Several advantages exist in our study. First, it is the first MR study to evaluate the potential causal associations between SS and certain cancers. Our study strictly followed the three major assumptions of MR analysis and could prevent the influence of potential confounders and reverse causality to a certain extent. Second, we included the most comprehensive SS-related SNPs to date, which could better explain genetic variants of SS. At the same time, the sample sizes of nine types of cancers vary from 1080 to 372,373 and are much larger than previous observational studies, which could provide adequate statistical power to assess the causality. Moreover, since we noticed that different molecular or histologic subtypes of cancer may share distinct biological characteristics based on both scientific researches or clinical observations, subgroup analysis were conducted on breast cancer and lung cancer, and the results could help to reach a deeper understanding of the causal relationship between SS and these cancers.

There are also limitations in our study. First, the participants of our study were all of the European descent. Whether conclusions of our study could be applied to other populations or regions is uncertain. So, further studies based on other populations or regions are needed. Second, the influence of potential pleiotropy on MR studies is difficult to be eliminated completely. In this study, we observed the horizontal pleiotropy in prostate cancer and endometrial cancer through MR-Egger regression test, but MR-PRESSO method did not test the horizontal pleiotropy and detect pleiotropic SNPs in them. In addition, though we included the most comprehensive GWASs summary data of SS so far, these SNPs only explained a small part of variants of SS. It is possible that other unknown SS-related SNPs could also play an important role in cancers. Thus, it is necessary to conduct new GWASs to detect novel SS-related SNPs.

Conclusions

This is the first study to revealed the causal effect of Sjögren’s syndrome on cancers using MR analysis. Sjögren’s syndrome was causally associated with risks of some cancers such as lymphomas, prostate cancer, endometrial cancer, liver and bile duct cancer, and urinary tract cancer, indicating that Sjögren’s syndrome may play a vital role in the occurrence of these malignancies. Further large-scale studies are warranted to verify our results and the underlying mechanisms should be further explored.

Availability of data and materials

The data used in this study was obtained from public databases and previous studies. Data sources and handing of these data were described in the Materials and Methods. Further information is available from the corresponding author upon request.

Abbreviations

CI:

Confidence interval

GWASs:

Genome-wide association studies

IVs:

Instrumental variables

IVW:

Inverse variance weighted

MR:

Mendelian randomization

MR-PRESSO:

MR pleiotropy residual sum and outlier

OR:

Odds ratio

SNP:

Single-nucleotide polymorphisms

SS:

Sjögren’s syndrome

References

  1. Fox RI. Sjögren’s syndrome. Lancet. 2005;366:321–31.

    CAS  PubMed  Google Scholar 

  2. Mavragani CP, Moutsopoulos HM. The geoepidemiology of Sjögren’s syndrome. Autoimmun Rev. 2010;9:A305–10.

    PubMed  Google Scholar 

  3. García-Carrasco M, Fuentes-Alexandro S, Escárcega RO, Salgado G, Riebeling C, Cervera R. Pathophysiology of Sjögren’s syndrome. Arch Med Res. 2006;37:921–32.

    PubMed  Google Scholar 

  4. Papageorgiou A, Voulgarelis M, Tzioufas AG. Clinical picture, outcome and predictive factors of lymphoma in Sjӧgren syndrome. Autoimmun Rev. 2015;14:641–9.

    PubMed  Google Scholar 

  5. Chatzis LG, Stergiou IE, Goules AV, Pezoulas V, Tsourouflis G, Fotiadis D, et al. Clinical picture, outcome and predictive factors of lymphoma in primary Sjögren’s syndrome: results from a harmonized dataset (1981–2021). Rheumatology (Oxford). 2022;61:3576–85.

    PubMed  Google Scholar 

  6. Brito-Zerón P, Kostov B, Fraile G, Caravia-Durán D, Maure B, Rascón FJ, et al. Characterization and risk estimate of cancer in patients with primary Sjögren syndrome. J Hematol Oncol. 2017;10:90.

    PubMed  PubMed Central  Google Scholar 

  7. Kang J, Kim H, Kim J, Choi S, Jung SY, Jang EJ, et al. Risk of malignancy in Korean patients with primary Sjögren’s syndrome. Int J Rheum Dis. 2020;23:1240–7.

    CAS  PubMed  Google Scholar 

  8. Lazarus MN, Robinson D, Mak V, Møller H, Isenberg DA. Incidence of cancer in a cohort of patients with primary Sjogren’s syndrome. Rheumatology (Oxford). 2006;45:1012–5.

    CAS  PubMed  Google Scholar 

  9. Goulabchand R, Malafaye N, Jacot W, WitkowskiDurandViel P, Morel J, Lukas C, et al. Cancer incidence in primary Sjögren’s syndrome: data from the French hospitalization database. Autoimmun Rev. 2021;20:102987.

    CAS  PubMed  Google Scholar 

  10. Brom M, Moyano S, Gandino IJ, Scolnik M, Soriano ER. Incidence of cancer in a cohort of patients with primary Sjögren syndrome in Argentina. Rheumatol Int. 2019;39:1697–702.

    CAS  PubMed  Google Scholar 

  11. Miceli-Richard C, Wang-Renault SF, Boudaoud S, Busato F, Lallemand C, Bethune K, et al. Overlap between differentially methylated DNA regions in blood B lymphocytes and genetic at-risk loci in primary Sjögren’s syndrome. Ann Rheum Dis. 2016;75:933–40.

    CAS  PubMed  Google Scholar 

  12. Imgenberg-Kreuz J, Sandling JK, Almlöf JC, Nordlund J, Signér L, Norheim KB, et al. Genome-wide DNA methylation analysis in multiple tissues in primary Sjögren’s syndrome reveals regulatory effects at interferon-induced genes. Ann Rheum Dis. 2016;75:2029–36.

    CAS  PubMed  Google Scholar 

  13. Cheishvili D, Boureau L, Szyf M. DNA demethylation and invasive cancer: implications for therapeutics. Br J Pharmacol. 2015;172:2705–15.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Bhootra S, Jill N, Shanmugam G, Rakshit S, Sarkar K. DNA methylation and cancer: transcriptional regulation, prognostic, and therapeutic perspective. Med Oncol. 2023;40:71.

    CAS  PubMed  Google Scholar 

  15. Wang-Renault SF, Boudaoud S, Nocturne G, Roche E, Sigrist N, Daviaud C, et al. Deregulation of microRNA expression in purified T and B lymphocytes from patients with primary Sjögren’s syndrome. Ann Rheum Dis. 2018;77:133–40.

    CAS  PubMed  Google Scholar 

  16. Pauley KM, Stewart CM, Gauna AE, Dupre LC, Kuklani R, Chan AL, et al. Altered miR-146a expression in Sjögren’s syndrome and its functional role in innate immunity. Eur J Immunol. 2011;41:2029–39.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Wilczyński M, Żytko E, Szymańska B, Dzieniecka M, Nowak M, Danielska J, et al. Expression of miR-146a in patients with ovarian cancer and its clinical significance. Oncol Lett. 2017;14:3207–14.

    PubMed  PubMed Central  Google Scholar 

  18. Zhang Z, Zhang Y, Sun XX, Ma X, Chen ZN. microRNA-146a inhibits cancer metastasis by downregulating VEGF through dual pathways in hepatocellular carcinoma. Mol Cancer. 2015;14:5.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Chen J, Jiang Q, Jiang XQ, Li DQ, Jiang XC, Wu XB, et al. miR-146a promoted breast cancer proliferation and invasion by regulating NM23-H1. J Biochem. 2020;167:41–8.

    CAS  PubMed  Google Scholar 

  20. Park SY, Kim H, Yoon S, Bae JA, Choi SY, Jung YD, et al. KITENIN-targeting microRNA-124 suppresses colorectal cancer cell motility and tumorigenesis. Mol Ther. 2014;22:1653–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Qi Z, Zhang B, Zhang J, Hu Q, Xu F, Chen B, et al. MicroRNA-30b inhibits non-small cell lung cancer cell growth by targeting the epidermal growth factor receptor. Neoplasma. 2018;65:192–200.

    CAS  PubMed  Google Scholar 

  22. Lessard CJ, Li H, Adrianto I, Ice JA, Rasmussen A, Grundahl KM, et al. Variants at multiple loci implicated in both innate and adaptive immune responses are associated with Sjögren’s syndrome. Nat Genet. 2013;45:1284–92.

    CAS  PubMed  Google Scholar 

  23. Gong X, Karchin R. Pan-cancer HLA gene-mediated tumor immunogenicity and immune evasion. Mol Cancer Res. 2022;20:1272–83.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Jaiswal M, LaRusso NF, Burgart LJ, Gores GJ. Inflammatory cytokines induce DNA damage and inhibit DNA repair in cholangiocarcinoma cells by a nitric oxide-dependent mechanism. Cancer Res. 2000;60:184–90.

    CAS  PubMed  Google Scholar 

  25. George J, Banik NL, Ray SK. Combination of hTERT knockdown and IFN-gamma treatment inhibited angiogenesis and tumor progression in glioblastoma. Clin Cancer Res. 2009;15:7186–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Taniguchi K, Karin M. IL-6 and related cytokines as the critical lynchpins between inflammation and cancer. Semin Immunol. 2014;26:54–74.

    CAS  PubMed  Google Scholar 

  27. El-Haibi CP, Singh R, Gupta P, Sharma PK, Greenleaf KN, Singh S, et al. Antibody microarray analysis of signaling networks regulated by Cxcl13 and Cxcr5 in prostate cancer. J Proteomics Bioinform. 2012;5:177–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Emdin CA, Khera AV, Kathiresan S. Mendelian randomization. JAMA. 2017;318:1925–6.

    PubMed  Google Scholar 

  29. Boef AG, Dekkers OM, le Cessie S. Mendelian randomization studies: a review of the approaches used and the quality of reporting. Int J Epidemiol. 2015;44:496–511.

    PubMed  Google Scholar 

  30. Li H, Reksten TR, Ice JA, Kelly JA, Adrianto I, Rasmussen A, et al. Identification of a Sjögren’s syndrome susceptibility locus at OAS1 that influences isoform switching, protein expression, and responsiveness to type I interferons. PLoS Genet. 2017;13:e1006820.

    PubMed  PubMed Central  Google Scholar 

  31. Carapito R, Gottenberg JE, Kotova I, Untrau M, Michel S, Naegely L, et al. A new MHC-linked susceptibility locus for primary Sjögren’s syndrome: MICA. Hum Mol Genet. 2017;26:2565–76.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Taylor KE, Wong Q, Levine DM, McHugh C, Laurie C, Doheny K, et al. Genome-wide association analysis reveals genetic heterogeneity of Sjögren’s syndrome according to ancestry. Arthritis Rheumatol. 2017;69:1294–305.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Khatri B, Tessneer KL, Rasmussen A, Aghakhanian F, Reksten TR, Adler A, et al. Genome-wide association study identifies Sjögren’s risk loci with functional implications in immune and glandular cells. Nat Commun. 2022;13:4287.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Michailidou K, Lindström S, Dennis J, Beesley J, Hui S, Kar S, et al. Association analysis identifies 65 new breast cancer risk loci. Nature. 2017;551:92–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Zhang H, Ahearn TU, Lecarpentier J, Barnes D, Beesley J, Qi G, et al. Genome-wide association study identifies 32 novel breast cancer susceptibility loci from overall and subtype-specific analyses. Nat Genet. 2020;52:572–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Wang Y, McKay JD, Rafnar T, Wang Z, Timofeeva MN, Broderick P, et al. Corrigendum: Rare variants of large effect in BRCA2 and CHEK2 affect risk of lung cancer. Nat Genet. 2017;49:651.

    PubMed  Google Scholar 

  37. O’Mara TA, Glubb DM, Amant F, Annibali D, Ashton K, Attia J, et al. Identification of nine new susceptibility loci for endometrial cancer. Nat Commun. 2018;9:3166.

    PubMed  PubMed Central  Google Scholar 

  38. Köhler A, Chen B, Gemignani F, Elisei R, Romei C, Figlioli G, et al. Genome-wide association study on differentiated thyroid cancer. J Clin Endocrinol Metab. 2013;98:E1674–81.

    PubMed  Google Scholar 

  39. Lesseur C, Diergaarde B, Olshan AF, Wünsch-Filho V, Ness AR, Liu G, et al. Genome-wide association analyses identify new susceptibility loci for oral cavity and pharyngeal cancer. Nat Genet. 2016;48:1544–50.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Canela-Xandri O, Rawlik K, Tenesa A. An atlas of genetic associations in UK Biobank. Nat Genet. 2018;50:1593–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Brion MJ, Shakhbazov K, Visscher PM. Calculating statistical power in Mendelian randomization studies. Int J Epidemiol. 2013;42:1497–501.

    PubMed  Google Scholar 

  42. Wang M, Jian Z, Gao X, Yuan C, Jin X, Li H, et al. Causal associations between educational attainment and 14 urological and reproductive health outcomes: a Mendelian randomization study. Front Public Health. 2021;9:742952.

    PubMed  PubMed Central  Google Scholar 

  43. Wang Q, Wang R, Chen C, Feng Y, Ye Z, Zhan M, et al. Educational attainment and endometrial cancer: a Mendelian randomization study. Front Genet. 2022;13:993731.

    PubMed  PubMed Central  Google Scholar 

  44. Larsson SC, Carter P, Kar S, Vithayathil M, Mason AM, Michaëlsson K, et al. Smoking, alcohol consumption, and cancer: a Mendelian randomisation study in UK Biobank and international genetic consortia participants. PLoS Med. 2020;17:e1003178.

    PubMed  PubMed Central  Google Scholar 

  45. Ahmed M, Mulugeta A, Lee SH, Mäkinen VP, Boyle T, Hyppönen E. Adiposity and cancer: a Mendelian randomization analysis in the UK biobank. Int J Obes (Lond). 2021;45:2657–65.

    CAS  PubMed  Google Scholar 

  46. Lee JJ, Wedow R, Okbay A, Kong E, Maghzian O, Zacher M, et al. Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nat Genet. 2018;50:1112–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Liu M, Jiang Y, Wedow R, Li Y, Brazel DM, Chen F, et al. Association studies of up to 1.2 million individuals yield new insights into the genetic etiology of tobacco and alcohol use. Nat Genet. 2019;51:237–44.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Burgess S, Butterworth A, Thompson SG. Mendelian randomization analysis with multiple genetic variants using summarized data. Genet Epidemiol. 2013;37:658–65.

    PubMed  PubMed Central  Google Scholar 

  49. Burgess S, Thompson SG. Interpreting findings from Mendelian randomization using the MR-Egger method. Eur J Epidemiol. 2017;32:377–89.

    PubMed  PubMed Central  Google Scholar 

  50. Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent estimation in Mendelian randomization with some invalid instruments using a weighted median estimator. Genet Epidemiol. 2016;40:304–14.

    PubMed  PubMed Central  Google Scholar 

  51. Verbanck M, Chen CY, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet. 2018;50:693–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Kassan SS, Thomas TL, Moutsopoulos HM, Hoover R, Kimberly RP, Budman DR, et al. Increased risk of lymphoma in sicca syndrome. Ann Intern Med. 1978;89:888–92.

    CAS  PubMed  Google Scholar 

  53. Theander E, Henriksson G, Ljungberg O, Mandl T, Manthorpe R, Jacobsson LT. Lymphoma and other malignancies in primary Sjögren’s syndrome: a cohort study on cancer incidence and lymphoma predictors. Ann Rheum Dis. 2006;65:796–803.

    CAS  PubMed  Google Scholar 

  54. Nocturne G, Mariette X. B cells in the pathogenesis of primary Sjögren syndrome. Nat Rev Rheumatol. 2018;14:133–45.

    CAS  PubMed  Google Scholar 

  55. Nezos A, Gravani F, Tassidou A, Kapsogeorgou EK, Voulgarelis M, Koutsilieris M, et al. Type I and II interferon signatures in Sjogren’s syndrome pathogenesis: contributions in distinct clinical phenotypes and Sjogren’s related lymphomagenesis. J Autoimmun. 2015;63:47–58.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Tobón GJ, Renaudineau Y, Hillion S, Cornec D, Devauchelle-Pensec V, Youinou P, et al. The Fms-like tyrosine kinase 3 ligand, a mediator of B cell survival, is also a marker of lymphoma in primary Sjögren’s syndrome. Arthritis Rheum. 2010;62:3447–56.

    PubMed  Google Scholar 

  57. Nocturne G, Boudaoud S, Miceli-Richard C, Viengchareun S, Lazure T, Nititham J, et al. Germline and somatic genetic variations of TNFAIP3 in lymphoma complicating primary Sjogren’s syndrome. Blood. 2013;122:4068–76.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Tapinos NI, Polihronis M, Moutsopoulos HM. Lymphoma development in Sjögren’s syndrome: novel p53 mutations. Arthritis Rheum. 1999;42:1466–72.

    CAS  PubMed  Google Scholar 

  59. Manzo C, Kechida M. Is primary Sjögren’s syndrome a risk factor for malignancies different from lymphomas? What does the literature highlight about it? Reumatologia. 2017;55:136–9.

    PubMed  PubMed Central  Google Scholar 

  60. Liang Y, Yang Z, Qin B, Zhong R. Primary Sjogren’s syndrome and malignancy risk: a systematic review and meta-analysis. Ann Rheum Dis. 2014;73:1151–6.

    PubMed  Google Scholar 

  61. Zhong H, Liu S, Wang Y, Xu D, Li M, Zhao Y, et al. Primary Sjögren’s syndrome is associated with increased risk of malignancies besides lymphoma: a systematic review and meta-analysis. Autoimmun Rev. 2022;21:103084.

    PubMed  Google Scholar 

  62. Gadalla SM, Amr S, Langenberg P, Baumgarten M, Davidson WF, Schairer C, et al. Breast cancer risk in elderly women with systemic autoimmune rheumatic diseases: a population-based case-control study. Br J Cancer. 2009;100:817–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Treppo E, Toffolutti F, Manfrè V, Taborelli M, De Marchi G, De Vita S, et al. Risk of cancer in connective tissue diseases in Northeastern Italy over 15 years. J Clin Med. 2022;11:4272.

    PubMed  PubMed Central  Google Scholar 

  64. Wang LH, Wang WM, Lin CY, Lin SH, Shieh CC. Bidirectional relationship between primary Sjögren syndrome and non-Hodgkin lymphoma: a nationwide Taiwanese population-based study. J Rheumatol. 2020;47:1374–8.

    PubMed  Google Scholar 

  65. Konttinen YT, Stegajev V, Al-Samadi A, Porola P, Hietanen J, Ainola M. Sjögren’s syndome and extragonadal sex steroid formation: a clue to a better disease control? J Steroid Biochem Mol Biol. 2015;145:237–44.

    CAS  PubMed  Google Scholar 

  66. McCoy SS, Sampene E, Baer AN. Association of Sjögren’s syndrome with reduced lifetime sex hormone exposure: a case-control study. Arthritis Care Res (Hoboken). 2020;72:1315–22.

    CAS  PubMed  Google Scholar 

  67. Liang J, Shang Y. Estrogen and cancer. Annu Rev Physiol. 2013;75:225–40.

    CAS  PubMed  Google Scholar 

  68. Seliger B, Kloor M, Ferrone S. HLA class II antigen-processing pathway in tumors: molecular defects and clinical relevance. Oncoimmunology. 2017;6:e1171447.

    PubMed  PubMed Central  Google Scholar 

  69. Bernd HW, Ziepert M, Thorns C, Klapper W, Wacker HH, Hummel M, et al. Loss of HLA-DR expression and immunoblastic morphology predict adverse outcome in diffuse large B-cell lymphoma - analyses of cases from two prospective randomized clinical trials. Haematologica. 2009;94:1569–80.

    PubMed  PubMed Central  Google Scholar 

  70. Lazaris ACh, Chatzigianni EB, Xidias G, Panoskaltsis TA, Thomopoulou GC, Eftychiadis CA, et al. Tissue evaluation of immune markers in endometrial and cervical carcinomas. J Exp Clin Cancer Res. 2004;23:269–75.

    PubMed  Google Scholar 

  71. Vuk-Pavlović S, Bulur PA, Lin Y, Qin R, Szumlanski CL, Zhao X, et al. Immunosuppressive CD14+HLA-DRlow/- monocytes in prostate cancer. Prostate. 2010;70:443–55.

    PubMed  PubMed Central  Google Scholar 

  72. Matoba K, Iizuka N, Gondo T, Ishihara T, Yamada-Okabe H, Tamesa T, et al. Tumor HLA-DR expression linked to early intrahepatic recurrence of hepatocellular carcinoma. Int J Cancer. 2005;115:231–40.

    CAS  PubMed  Google Scholar 

  73. Balakrishnan CK, Tye GJ, Balasubramaniam SD, Kaur G. CD74 and HLA-DRA in cervical carcinogenesis: potential targets for antitumour therapy. Medicina (Kaunas). 2022;58:190.

    PubMed  Google Scholar 

  74. Piao XM, Kang HW, Jeong P, Byun YJ, Lee HY, Kim K, et al. A prognostic immune predictor, HLA-DRA, plays diverse roles in non-muscle invasive and muscle invasive bladder cancer. Urol Oncol. 2021;39(237):e21-237.e29.

    Google Scholar 

  75. Matsushita K, Takenouchi T, Shimada H, Tomonaga T, Hayashi H, Shioya A, et al. Strong HLA-DR antigen expression on cancer cells relates to better prognosis of colorectal cancer patients: possible involvement of c-myc suppression by interferon-gamma in situ. Cancer Sci. 2006;97:57–63.

    CAS  PubMed  Google Scholar 

  76. Rangel LB, Agarwal R, Sherman-Baust CA, Mello-Coelho Vd, Pizer ES, Ji H, et al. Anomalous expression of the HLA-DR alpha and beta chains in ovarian and other cancers. Cancer Biol Ther. 2004;3:1021–7.

    CAS  PubMed  Google Scholar 

  77. Zhou Z, Liu H, Yang Y, Zhou J, Zhao L, Chen H, et al. The five major autoimmune diseases increase the risk of cancer: epidemiological data from a large-scale cohort study in China. Cancer Commun (Lond). 2022;42:435–46.

    PubMed  Google Scholar 

  78. Brito-Zerón P, Baldini C, Bootsma H, Bowman SJ, Jonsson R, Mariette X, et al. Sjögren syndrome. Nat Rev Dis Primers. 2016;2(1):16047.

    PubMed  Google Scholar 

  79. Lebrun C, Debouverie M, Vermersch P, Clavelou P, Rumbach L, de Seze J, et al. Cancer risk and impact of disease-modifying treatments in patients with multiple sclerosis. Mult Scler. 2008;14(3):399–405.

    PubMed  Google Scholar 

  80. Yates WB, Vajdic CM, Na R, McCluskey PJ, Wakefield D. Malignancy risk in patients with inflammatory eye disease treated with systemic immunosuppressive therapy: a tertiary referral cohort study. Ophthalmology. 2015;122(2):265–73.

    PubMed  Google Scholar 

  81. Linardaki G, Moutsopoulos HM. The uncertain role of immunosuppressive agents in Sjögren’s syndrome. Cleve Clin J Med. 1997;64(10):523–6.

    CAS  PubMed  Google Scholar 

  82. Kreher MA, Konda S, Noland MMB, Longo MI, Valdes-Rodriguez R. Risk of melanoma and nonmelanoma skin cancer with immunosuppressants, part II: methotrexate, alkylating agents, biologics, and small molecule inhibitors. J Am Acad Dermatol. 2023;88(3):534–42.

    CAS  PubMed  Google Scholar 

  83. Lange E, Blizzard L, Venn A, Francis H, Jones G. Disease-modifying anti-rheumatic drugs and non-melanoma skin cancer in inflammatory arthritis patients: a retrospective cohort study. Rheumatology (Oxford). 2016;55(9):1594–600.

    CAS  PubMed  Google Scholar 

  84. Scott FI, Mamtani R, Brensinger CM, Haynes K, Chiesa-Fuxench ZC, Zhang J, et al. Risk of Nonmelanoma skin cancer associated with the use of immunosuppressant and biologic agents in patients with a history of autoimmune disease and nonmelanoma skin cancer. JAMA Dermatol. 2016;152(2):164–72.

    PubMed  PubMed Central  Google Scholar 

  85. Chen YH, Wang XY, Jin X, Yang Z, Xu J. Rituximab therapy for primary Sjögren’s syndrome. Front Pharmacol. 2021;12: 731122.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Alping P, Askling J, Burman J, Fink K, Fogdell-Hahn A, Gunnarsson M, et al. Cancer risk for fingolimod, natalizumab, and rituximab in multiple sclerosis patients. Ann Neurol. 2020;87(5):688–99.

    CAS  PubMed  Google Scholar 

  87. Zhang C, Chu M. Leflunomide: a promising drug with good antitumor potential. Biochem Biophys Res Commun. 2018;496(2):726–30.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We appreciate a lot for the investigators who shared the GWAS data. We also appreciate the funding of the National Natural Science Foundation of China (Grant Number: 82003047), the Fundamental Research Funds for the Central Universities (Grant Number: xzy012020061), the research fund from the Second Affiliated Hospital of Xi’an Jiaotong University (Grant Number: 2020YJ(ZYTS)381), and Shaanxi Key Laboratory of Traditional Chinese Medicine (Grant Number: 2022-ZXY-SYS-002).

Funding

This work was supported by the National Natural Science Foundation of China (Grant Number: 82003047), the Fundamental Research Funds for the Central Universities (Grant Number: xzy012020061), the research fund from the Second Affiliated Hospital of Xi’an Jiaotong University (Grant Number: 2020YJ(ZYTS)381), and Shaanxi Key Laboratory of Traditional Chinese Medicine (Grant Number: 2022-ZXY-SYS-002).

Author information

Authors and Affiliations

Authors

Contributions

YJ, XM, SZ, JL designed the study. YJ, PY, WW, CF, CL, YZ acquired and analyzed the data. YJ, XM, XL, XW, YC, HW drafted the article and figures. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Shuqun Zhang or Xingcong Ma.

Ethics declarations

Ethics approval and consent to participate

All the studies used to compliment the current study were approved by relevant ethics committees. All participants involved provided a written informed consent.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Supplementary Table 1.

Details of SNPs selected as instrumental variables for Mendelian randomization analysis. Supplementary Table 2. The results of the causal effects of cancers on Sjögren’s syndrome. Supplementary Table 3. The results of heterogeneity analysis in inverse direction analysis. Supplementary Table 4. The results of horizontal pleiotropy analysis in inverse direction analysis. Supplementary Fig. 1. The scatter plots of the effects of genetic instruments on Sjögren’s syndrome against their effects on cancers. Supplementary Fig. 2. The funnel plots of the causal effect of Sjögren’s syndrome on cancers. Supplementary Fig. 3. Leave-one-out analysis of the causal effect of Sjögren’s syndrome on cancers. Supplementary Fig. 4. The forest plots of the causal effect of Sjögren’s syndrome on cancers

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jia, Y., Yao, P., Li, J. et al. Causal associations of Sjögren’s syndrome with cancers: a two-sample Mendelian randomization study. Arthritis Res Ther 25, 171 (2023). https://doi.org/10.1186/s13075-023-03157-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13075-023-03157-w

Keywords