eISSN: 2084-9869
ISSN: 1233-9687
Polish Journal of Pathology
Current issue Archive Manuscripts accepted About the journal Supplements Editorial board Abstracting and indexing Subscription Contact Instructions for authors Publication charge Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
3/2023
vol. 74
 
Share:
Share:
Original paper

Status of PD-1 and PD-L1 expression in invasive urothelial carcinoma of the bladder with mismatch repair protein deficiency

Nuran Sungu
1
,
Merve M. Kıran
2

1.
Department of Pathology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Turkey
2.
Department of Pathology, Ankara City Hospital, Ankara, Turkey
Pol J Pathol 2023; 74 (3): 161-170
Online publish date: 2023/10/25
Article file
- PJP-02704.pdf  [0.67 MB]
Get citation
 
PlumX metrics:
 

Introduction

Many genetic and epigenetic factors play a role in bladder carcinogenesis. DNA repair mechanism is required to prevent DNA mutations that can be fatal to cells [1]. Mismatch repair (MMR) genes encode a set of DNA repair proteins that cooperatio to recognize and repair DNA mismatches. These proteins are MSH2, MSH6, MLH1 and PMS2. Any defect in these proteins results in an inactive DNA repair process that increases ratios of pathogenic change in genes of the cell growth cycle, leading to defects in tumor suppressor genes and oncogenes, which in turn leads to an increased risk of cancer. DNA repair dysfunction may allow the formation of a high-risk urothelium for malignant transformation in the bladder. Dysfunction of MMR genes may present as absence or reduction of MMR gene expression or microsatellite instability (MSI) phenotype [2].
Lynch syndrome (LS) is an autosomal dominant inherited disorder caused by a germ line pathogenic variant in the DNA mismatch repair (MMR) genes (MLH1, MSH2, MSH6, and PMS2) or deletion of the 3’ end of EPCAM in the upper part of MSH2 [3].
The cancers most commonly related with LS are colon cancer, endometrial cancer, ovarian cancer and malignancies affecting the stomach, small intestine, prostate, breast, brain, and hepato-biliary tract [4-8]. In a review article, it was emphasized that urothelial cancers are the third most common type of cancer in LS-related tumors [2].
Genetic studies on urothelial carcinomas have also indicated that there is a correlation between microsatellite instability (MSI) and urothelial carcinomas. The incidence of MSI in urothelial carcinomas has been reported between 1.1% and 28% in publications [9].
The first test to examine the loss of MMR proteins is immunohistochemistry, which is currently recommended only for colorectal and endometrial carcinomas [10-16]. This technique has also been applied in urothelial carcinomas in recent studies [4].
Urothelial carcinomas with MSI benefit greatly from adjuvant cisplatin-based chemotherapy [17, 18].
Recently, the immune checkpoint inhibitors (ICIs) pembrolizumab and atezolizumab, which target PD-1 or PD-L1, have been approved for patients not eligible for first-line cisplatin-based chemotherapy [19, 20]. The importance of identifying urothelial carcinoma patients with MSI is becoming increasingly evident, as specific treatments are required for better prognosis.
The aim in this study was to assess the prognostic significance of MSI, and PD-1 and PD-L1 expression, which are used as predictive biomarkers in immunotherapy, in patients undergoing radical cystectomy, as well as the presence of PD-1 and PD-L1 expression in patients with MSI.

Material and methods

Study group
This was a retrospective cohort study that included 99 patients with tumors who underwent radical cystectomy.

Resection materials were re-evaluated under light microscopy by 2 pathologists, 1 of whom was experienced in the field of uropathology, and the diagnosis was confirmed. Construction of the tissue microarray
For the tissue microarray (TMA), the samples were obtained from paraffin blocks with viable tumor areas that were as free of necrosis as possible. For each case, 2 tissue samples with a diameter of 5 mm, one from the tumor center and 1 from the tumor periphery, were taken to represent tumor heterogeneity.
Immunohistochemical analysis procedure
Four-micron thick unstained sections were sequentially cut from the TMA blocks and stained for PD-1 (ab137132, abcam, Cambridge, MA, USA), PD-L1 (ab205921, abcam, Cambridge, MA, USA), MLH-1 (1/100, Cell Marque, Clone G168-728), MSH-2 (1/200, Cell Marque, cloneG219-1129), and MSH-6 (1/200 Thermo Fisher, clone 44). Immunohistochemical expression was performed using a Leica Bond-Max automatic expression device (Leica Microsystems, Wetzlar, Germany).
Analysis of immunohistochemical study results
Two pathologists evaluated all of the stained slides for PD-1, PD-L1, MLH1, MSH2, and MSH6. Membranous staining of PD-1 and PD-L1 was accepted as positive (Fig. 1A-C). For PD-1 and PD-L1, the expression ratios in both the immune and tumor cells was evaluated. The positive cut-off values were accepted as >5% and 10% [19, 21].
MMR protein expression was assessed in tumor nuclei with complete nuclear loss required for a case to be considered as mismatch repair deficiency (d-MMR). Nuclear expression was assessed as proficiency (p-MMR) (Fig. 2). Peritumoral and intra tumoral lymphocytes served as an internal control.
Statistical analyses
The mean ± standard deviation was used to represent descriptive statistics for age variables that fit the normal distribution. The number (n) and percentage (%) were used for the categorical variables (age group, grade, muscle invasion, MLH-1, MSH-2, MSH-6 repair gene losses). Cross-tables were created to compare the categorical variables according to the immune markers. The χ2 test was used to identify different categories. Appropriate chi square values were given according to cell ratios with an expected value < 5 (expected count < 5 cell ratio). Pairwise comparisons with Bonferroni correction were performed to determine the different groups in the muscle invasion variables in which there was a difference as a result of the χ2 test. MS-Excel 2010 and IBM SPSS Statistics for Windows 22.0 (IBM Corp., Armonk, NY, USA) were used for the statistical analyses and calculations. In the statistical decisions, p < 0.05 was accepted as statistically significant.

Results

Clinicopathological characteristics
The ages of the patients ranged between 38 and 95 years and the mean age was 66.9 ±10.7 years. The number of patients over the age of 65 was 56. All of the patients except for 1 were male. Of the 99 urothelial cancer samples included in the study, 14 (14.1%) were low grade, 80 (80.8%) were high grade, and 5 (5.1%) were other types of rare urothelial cancers (plasmacytoid variant etc.). While invasion of the lamina propria was observed in 35 (35.4%) of the samples, 7 (7.1%) of the 64 samples with muscle invasion were superficial muscle invasive and 57 (57.6%) were deep muscle invasive. While 60 (60.6%) of the examined samples had not invasion to perivesical adipose tissue, 39 (39.4%) had invasion to perivesical adipose tissue. Lymph node metastasis was not observed in any of the samples.
Relationship between the MSI status and clinicopathological features
The distribution of MLH1, MSH2, and MSH6 was analyzed in the samples according to MMR status. Loss of MLH1, MSH2, and MSH6 repair genes was observed in 13 (13.1%), 21 (21.2%), and 15 (15.2%) of the samples, respectively. Loss of MLH1-MSH2 was observed in 3 cases and MLH1-MSH2-MSH6 in 1 case. Loss of MSH2 was the most common. The number of samples with missing any repair gene was calculated as 32 (32.3%). In 67 (67.7%) samples, loss of MLH1, MSH2, and MSH6 genes was not observed. When the MSI status was examined according to the presence of muscle invasion of the tumor, muscle invasion was not observed in 12 of the d-MMR tumors, while there was superficial muscle invasion in 6 and deep muscle invasion in 14. It is not significant correlation was observed between the groups with and without muscle invasion (p = 0.320).
When muscle invasion and MSI conditions were compared, a significant correlation was observed between muscle invasion and MSI status (χ2 = 10.301; p = 0.005). A reanalysis was performed to determine whether the relationship was due to superficial or deep muscle invasion. It was determined that 85.7% (6/7) of the cases with d-MMR had superficial muscle invasion, and 75.4% (43/57) of the cases with p-MMR had deep muscle invasion (χ2 = 8.192; p = 0.012). In the analysis performed to determine whether there is a relationship between MSI conditions and lamina propria and superficial muscle invasion, it was determined that d-MMR tumors showed more superficial muscle invasion than p-MMR tumors. There was a relationship between the groups with lamina propria and superficial muscle invasion according to the d-MMR (χ2 = 4.375; p = 0.036). However, when Bonferroni correction was made, it was concluded that there was no difference between these groups (p = 0.036 * 3 = 0.108). There was not correlation between the groups with lamina propria and deep muscle invasion according to the d-MMR (χ2 = 1.011; p = 0.945). When the depth of invasion was evaluated according to MMR proteins, only a significant correlation was found between MLH1 and invasion. Just as in the d-MMR group, the MLH1 group showed superficial muscle invasion compared to the p-MMR group. This was statistically significant (p = 0.002).
It is not significant correlation was observed between MSI status and tumor grade, invasion into perivesical adipose tissue, and age. No significant relationship was found between tumor grade and loss of MLH1, MSH2 and MSH6 (p > 0.05) (Table I).
Relationship between the PD-1 and PD-L1 expression with clinicopathological features
The expression ratio of > 5% with PD-L1 was 38.4% (38) in the tumor cells and 39.4% (39) in the immune cells. The expression ratio of > 10% with PD-L1 was 22.2% (22) in the tumor cells and 24.2% (24) in the immune cells (Table II). PD-1 was not detected in the tumor cells. In the immune cells, while the expression ratio of > 5% was 61.6% (61), the expression ratio of > 10% was 38.4% (38) (Table III).
When the PD-1 and PD-L1 expression ratios were examined to determine if they differed according to the tumor grade, no significant results were obtained for PD-L1 in either the tumor or immune cells. In the immune cells, a significant correlation was observed for both > 5% and > 10% with PD-1 (p > 0.001 and p = 0.012, respectively). The expression ratio was significantly higher in patients with a high grade tumor when compared to those with a low grade (Tables II and III).
When the PD-1 and PD-L1 expression ratio were examined to determine if they differed according to the muscle invasion, the immune cells for PD-1 and the tumor cells for PD-L1 is not significantly correlation detected. A strong association was observed with PD-L1 in immune cells, with only > 5% expression rate (p = 0.012). No expression was observed for PD-L1 in the superficial invasive tumors (Tables II and III).
It is not association was found between PD-1 and PD-L1 and age as well as with invasion to perivesical adipose tissue.
Associations between the expression of PD-1 and PD-L1 in the tumor cells/immune cells with MSI status
According to MMR protein expression status, when the PD-1, PD-L1 expression rates are analyzed, strong relationship were observed between the d-MMR group and > 5% expression ratio of PDL-1 in both tumor and immune cells (p = 0.02 and p = 0.004, respectively). The expression rates were higher in cases p-MMR compared to those with d-MMR.
In addition, the PD-1 and PD-L1 expression status of cases with loss of MLH1, MSH2, and MSH6 were also analyzed. According to the loss of MLH1 and MSH2, there was not significant relationship (p > 0.05) between the PD-L1 expression ratio in the tumor/immune cells (for > 5% and > 10%) (Table II).
When the PD-L1 expression was considered according to loss of MSH6, significant correlation was found between both ratio of > 5% and ratio of > 10% in the tumor cells (p = 0.006 and p = 0.025, respectively). While there was 1 case with > 5% expression in the tumor cells, there were no cases with > 10% expression. Only 1 case was detected in the group with > 5% expression in immune cells, which was statistically significant (p = 0.005). Although 1 case was detected in the group with > 10% expression in immune cells, it is not statistically significant correlation was found (Table II).
According to the loss of MLH1, MSH2 and MSH6, it is not significant correlation was observed between the PD-1 in the tumor/immune cells (for > 5% and > 10%) (p > 0.05) (Table III).
Isolated loss of MSH6 was detected in 7 cases. None of these cases indicated PD-L1 expression in the immune and tumor cells at ratio of > 5% and > 10%. Similarly, there was no expression of > 5% and > 10% for PD-1 in the tumor cells. On the other hand, the number of cases with expression of > 5% and > 10% for PD-1 in the immune cells became prominent, with 3 cases each. Since there were very few cases with isolated MSH6 loss, it was not possible to analyze them statistically.

Discussion

Urothelial cancers may be a component of LS [17]. Previous molecular genetic studies have shown a relationship between urothelial carcinomas and MSI. In immunohistochemical studies, while 1.1% to 28% of urothelial carcinomas were found to show MSI, this ratio was reported to be up to 45% in studies using PCR-based methods [2, 9, 22, 23]. Among these, MSH2 loss is the most common [1, 2, 17]. In the current study, it was found that the ratio of MSI was 32% and the loss of MSH2 among the MMR proteins was the most common, in accordance with the literature.
Some studies have shown a correlation between the histologic features of urothelial carcinomas and the presence of MSI [24-26]. In urothelial carcinomas with MSI, these histologic features were found to be high-grade papillary tumors in pTa or pT1 stages, usually without prominent nuclear pleomorphism. In the current study, when d-MMR tumors were compared according to the presence of deep and superficial muscle invasion, it was determined that d-MMR tumors showed more superficial muscle invasion than p-MMR tumors (p = 0.012). The relationship between d-MMR and p-MMR groups according to muscle invasion was statistically significant (p = 0.005). However, no significant result was found between low and high grade.
Approximately 75% of patients with urothelial carcinoma are non-invasive and have a favorable prognosis with transurethral resection, intravesical chemotherapy and immunotherapy. However, the remaining 25% muscle-invasive bladder cancer usually show a poor prognosis despite systemic treatment. In recent years, immune check point inhibitors (ICIs), particularly PD-1, PD-L1, and cytotoxic T lymphocyte-associated antigen 4, have brought a new development in the treatment of urological tumors, especially advanced urothelial cancer. These markers have also been associated with prognosis [27].
The positive cut-off value for PD-1/PD-L1 has not been standardized for ICI treatment. Wang et al. correlated well PD-L1 expression in tumor cells with the pathological grade, clinical stage, recurrence, and postoperative prognosis of bladder cancer in their study including 50 bladder cancers, in which PD-L1 positive expression ratio was >10 [21]. Chen et al. used positive cut-off values of 1%, 5%, 10%, and 50% in tumor cells and 1% in immune cells and applied TMA blocks in a study of 96 cases of invasive urothelial carcinoma and found that a 5% cut-off value for PD-L1 may be a good positive value in PD-1/PD-L1 inhibitor treatment [28]. These two cut-off values were used in the current study. However, as the study was retrospective, further prospective studies involving patients undergoing immunotherapy are needed to confirm these values.
In a comprehensive study including 318 radical cystectomies, Boorjian et al. found that > 5% PD-L1 expression in urothelial tumor cells was significantly related with increasing pathological stage [29]. In this study, there was strong association in the expression ratio of > 5% with PD-L1 in the immune cells (p = 0.012). PD-L1 expression was not observed in the superficial muscle invasive tumors.

Currently, treatment of anti-PD-1 have been approved in metastatic and advanced urothelial carcinoma. In a study by Kumar et al.; PD-1/PD-L1 expression was investigated immunohistochemically in 116 patients who underwent transurethral resection of the bladder and were diagnosed with urothelial carcinoma. In the study, a high correlation was found between PD-1 and tumor grade. High expression detected in high grade tumors [30]. In our study; we detected expression rates of > 5% and > 10% with PD-1 in immune cells, significantly higher in high-grade tumors than low-grade tumors (p > 0.001 and p = 0.012, respectively according to the percentage of expression).
Recent studies have shown that patients with multiple tumor types exhibiting MMR protein loss may respond to PD-1/PD-L1 blockade [31, 32]. In the literature, there are very few studies showing PD-1/PD-L1 expression in bladder cancers with loss of MMR. In a study including 201 cases of high-grade muscle invasive bladder urothelial cancer conducted by Hodgson et al., a positive significant correlation was found between PD-L1 positivity and loss of MMR (PD-L1 positive in 3 of 4 cases) [17]. In the current study, negative significant association were obtained between d-MMR and the expression ratio of >5% of PDL-1 in both the tumor and immune cells (p = 0.02 and p = 0.004, respectively). The expression ratio was lower in tumors with loss of MMR. This result may be due only to the absence of high-grade tumors in our study. It may also be due to the fact that the cases in the study in the article included only high-grade invasive tumors and did not include low-grade tumors for comparison.
In addition, MLH1, MSH2 and MSH6 loss states and PD-1, PD-L1 expression ratios also differ. There are very few studies comparing them. In a study of 50 cases, Zavalishina et al. immunohistochemically evaluated the MSI phenotype and its relationship with PD-L1 in T1-T3 urothelial tumors. As a result of their study, while the decreased PMS2 and MLH1 expression was observed in PD-L1 positive cases, significant expression was detected with MSH6 [33]. Moreover, it can be said that there was no MSH6 loss in PD-L1 positive cases. In the present study, > 5% expression with PD-L1 in the immune/tumor cells and 10% expression with PD-L1 in the immune cells was found in only 1 case each with MSH6 loss. In tumor cells, no case with PD-L1 expression at the > 10% cut-off value was observed. This may indicate that MSI patients with loss of MSH6 may have a limitation to utilize the immunotherapy.
In a review article presented by Bellmunt et al., it was reported that the response ratio to immunotherapy in metastatic urothelial carcinomas was 20-30% and this ratio increased up to 39% at higher doses [34]. Since there are very few studies evaluating immunotherapy in urothelial cancer patients with MSI, studies showing response ratio to this treatment could not be found.
In studies involving MMR deficiency patients to whom immunotherapy was applied, MMR protein losses were mostly evaluated as high and low MSI, and MMR proteins were not evaluated separately. This may be attributed to the small number of patients. In one study, it was reported that patients with MSH6 loss would not benefit from immunotherapy. In this study conducted by Liu et al., 15 Chinese families with LS diagnosed clinically according to Amsterdam II criteria were identified. In these patients, immunohistochemical expression was observed to be negative with MSH6 but positive with MLH1, MSH2, and PMS2. While it was thought that these patients would benefit from PD-1 immune checkpoint blockade treatment, on the contrary, it was observed that tumors progressed rapidly after 4 sessions of anti-PD-1 treatment [35]. In the current study, expression was observed in a maximum of 1 case in a patient with loss of MSH6. These results were found to be statistically significant (p < 0.05). This may explain why patients with MSH6 loss could not benefit from ICI treatment. However, comprehensive multicenter studies with multiple cases are required to support this.
The limitations of this study were that it included a small number of patients, it was single centered and retrospective, the MSI status was determined only by immunohistochemistry, and the results could not be correlated with PCR. Therefore, further research is required to interpret and confirm the results.

Conclusions

This study was conducted to indicate the MSI and PD-1/PD-L1 status in invasive urothelial carcinomas undergoing cystectomy and their relationship with prognostic markers. The PD-1/PD-L1 expression ratio was higher in muscle invasive and high-grade urothelial cancers. This suggests that PD-1 and PD-L1 expression may contribute to the progression and poor prognosis of bladder cancer. Loss of MMR proteins, especially MLH1 loss, was more common in the superficial muscle invasive tumors than in the deep muscle invasive tumors. The histomorphological findings of the MSI tumors indicated that they were more superficial tumors in this study, as reported in some other studies. PD-1/PD-L1 expression ratios were low in the d-MMR invasive urothelial cancers and no expression was observed in patients with MSH6 loss. Our study results suggested that the reason why a group of patients with MMR loss did not benefit from immunotherapy might be due to the loss of MSH6. However, comprehensive studies are required to research the clinical utility of d-MMR as a predictive biomarker of immune therapy response.
The authors declare no conflict of interest.
References
1. Vageli DP, Giannopoulos S, Doukas SG, et al. Mismatch repair hMSH2, hMLH1, hMSH6 and hPMS2 mRNA expression profiles in precancerous and cancerous urothelium. Oncol Lett 2013; 5: 283-294.
2. Lindner AK, Schachtner G, Tulchiner G, et al. Lynch Syndrome: Its Impact on Urothelial Carcinoma. Int J Mol Sci 2021; 22: 531.
3. Ligtenberg MJ, Kuiper RP, Geurts van Kessel A, Hoogerbrugge N. EPCAM deletion carriers constitute a unique subgroup of Lynch syndrome patients. Fam Cancer 2013; 12: 169-174. 
4. Mangold E, Pagenstecher C, Friedl W, et al. Spectrum and frequencies of mutations in MSH2 and MLH1 identified in 1,721 German families suspected of hereditary nonpolyposis colorectal cancer. Int J Cancer 2005; 116: 692-702.
5. Boland CR. Recent discoveries in the molecular genetics of Lynch syndrome. Fam Cancer 2016; 15: 395-403.
6. Modrich, P. Mechanisms in eukaryotic mismatch repair. J Biol Chem 2006; 281: 30305-30309.
7. Bridge G, Rashid S, Martin SA. DNA mismatch repair and oxidative DNA damage: Implications for cancer biology and treatment. Cancers 2014; 6: 1597–1614.
8. Dominguez-Valentin M, Sampson JR, Seppälä TT, et al. Cancer risks by gene, age, and gender in 6350 carriers of pathogenic mismatch repair variants: Findings from the Prospective Lynch Syndrome Database. Genet Med 2020; 22: 15-25.
9. Sobrino-Reig E, Meizoso T, García J, et al. Morphological predictors for microsatellite instability in urothelial carcinoma. Diagn Pathol 2021; 16: 106.
10. Kawakami H, Zaanan A, Sinicrope FA. Microsatellite Instability Testing and Its Role in the Management of Colorectal Cancer. HHS Public Access 2016; 16: 1-14.
11. Chintalacheruvu LM, Shaw T, Buddam A, et al. Major hereditary gastrointestinal cancer syndromes: a narrative review. J Gastrointest Liver Dis 2017; 26: 157-163.
12. Buza N, Ziai J, Hui P. Mismatch repair deficiency testing in clinical practice. Expert Rev Mol Diagn 2016; 16: 591-604.
13. Chen W, Swanson BJ, Frankel WL. Molecular genetics of microsatellite-unstable colorectal cancer for pathologists. Diagn Pathol 2017; 12: 24.
14. Modica I, Soslow RA, Black D, et al. Utility of immunohistochemistry in predicting microsatellite instability in endometrial carcinoma. Am J Surg Pathol 2007; 31: 744-751.
15. Hashmi AA, Mudassir G, Hashmi RN, et al. Microsatellite instability in endometrial carcinoma by immunohistochemistry, association with clinical and histopathologic parameters. Asian Pac J Cancer Prev 2019; 20: 2601-2606.
16. Clarke BA, Cooper K. Identifying Lynch syndrome in patients with endometrial carcinoma shortcomings of morphologic and clinical schemas. Adv Anat Pathol 2012; 19: 231-238.
17. Hodgson A, Vesprini D, Liu SK, et al. Correlation of mismatch repair protein deficiency, PD-L1 and CD8 expression in high-grade urothelial carcinoma of the bladder. J Clin Pathol 2020; 73: 519-522.
18. Laguna MP. Re: hereditary-like urothelial carcinomas of the upper urinary tract benefit more from adjuvant cisplatin-based chemotherapy after radical nephroureterectomy than do sporadic tumours. J Urol 2015; 193: 72.
19. Schulz GB, Todorova R, Braunschweig T, et al. PD-L1 expression in bladder cancer: Which scoring algorithm in what tissue? Urol Oncol 2021; 39: 734.e1-734.e10.
20. Zhou TC, Sankin AI, Porcelli SA, et al. A review of the PD-1/PD-L1 checkpoint in bladder cancer: From mediator of immune escape to target for treatment. Urol Oncol 2017; 35: 14-20.
21. Wang Y, Zhuang Q, Zhou S, et al. Costimulatory molecule B7-H1 on the immune escape of bladder cancer and its clinical significance. J Huazhong Univ Sci Technolog Med Sci 2009; 29: 77-79.
22. Giedl J, Schneckenpointner R, Filbeck T, et al. Low frequency of HNPCC-associated microsatellite instability and aberrant MMR protein expression in early-onset bladder cancer. Am J Clin Pathol 2014; 142: 634-639.
23. Vaish M, Mandhani A, Mittal RD, et al. Microsatellite instability as prognostic marker in bladder tumors: a clinical significance. BMC Urol 2005; 5: 2.
24. Joost P, Therkildsen C, Dominguez-Valentin M, et al. Urinary tract Cancer in lynch syndrome; increased risk in carriers of MSH2 mutations. Urology 2015; 86: 1212-1217.
25. Harper HL, McKenney JK, Heald B, et al. Upper tract urothelial carcinomas: frequency of association with mismatch repair protein loss and lynch syndrome. Mod Pathol 2017; 30: 146-156.
26. Ju JY, Mills AM, Mahadevan MS, et al. Universal Lynch Syndrome Screening Should be Performed in All Upper Tract Urothelial Carcinomas. Am J Surg Pathol 2018; 42: 1549-1555.
27. Ma J, Zhou Q, Xu W, et al. Urine PD-L1 is a tumor tissue candidate substitute and is associated with poor survival in muscle-invasive bladder cancer patients. Int Immunopharmacol 2023; 114: 109535.
28. Chen X, Chen H, Lin R, et al. Correlation between PD-L1 expression of the tumour cells and lymphocytes infiltration in the invasive front of urothelial carcinoma. J Clin Pathol 2022; doi: 10.1136/jclinpath-2021-207795.
29. Boorjian SA, Sheinin Y, Crispen PL, et al. T-cell coregulatory molecule expression in urothelial cell carcinoma: clinicopathologic correlations and association with survival. Clin Cancer Res 2008; 14: 4800-4808.
30. Kumar U, Anthony ML, Sahai R, et al. Immunoexpression of PD-L1 and PD-1 and Its Clinicopathological Correlation in Urothelial Carcinomas. J Lab Physicians 2021; 14: 197-201.
31. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015; 372: 2509-2520.
32. Kim ST, Klempner SJ, Park SH, et al. Correlating programmed death ligand 1 (PD-L1) expression, mismatch repair deficiency, and outcomes across tumor types: implications for immunotherapy. Oncotarget 2017; 8: 77415-77423.
33. Zavalishina LE, Andreeva YY, Olyushina EM, et al. Immunohistochemical study of the MSI phenotype of urothelial bladder cancer. Arkh Patol 2020; 82: 5-14.
34. Bellmunt J, Powles T, Vogelzang NJ. A review on the evolution of PD-1/PD-L1 immunotherapy for bladder cancer: The future is now. Cancer Treat Rev 2017; 54: 58-67.
35. Liub Y, Wangc M, Cheng Q, et al. A novel heterozygous large deletion of MSH6 gene in a Chinese family with Lynch syndrome. Gene 2019; 704: 103-112.
Copyright: © 2023 Polish Association of Pathologists and the Polish Branch of the International Academy of Pathology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.