Abstract
Purpose
To investigate the prevalence of deficient DNA mismatch repair (dMMR) status in upper tract urothelial carcinoma (UTUC) and its association with clinicopathological characteristics as well as Nectin-4 immunohistochemical expression.
Methods
We retrospectively identified histologically confirmed UTUC cases treated at Peking University Third Hospital between December 2016 and September 2023. Eligible participants were required to also possess complete clinicopathological records and available formalin-fixed, paraffin-embedded (FFPE) tumor specimens suitable for immunohistochemical evaluation. MMR protein expression was categorized as either dMMR or proficient mismatch repair (pMMR), while Nectin-4 expression was quantitatively assessed using the H-score system. Samples were then classified as negative (H-score 0–14), low (H-score 15–99), medium (H-score 100–199), and high (H-score 200–300). Statistical significance was established at P < 0.05 using two-tailed tests.
Results
A total of 339 patients were deemed eligible, with specimens successfully evaluated. 25 patients (7.4%) demonstrated dMMR status. High Nectin-4 expression was observed in 124 patients (36.7%). A statistically significant association was identified between dMMR status and elevated Nectin-4 expression (P = 0.044). No significant differences were detected between dMMR and pMMR groups regarding clinical parameters, including gender, age, tumor grade, or immunophenotypic characteristics.
Conclusion
Our study revealed that 7.4% of UTUC patients exhibited dMMR status, with heterogeneous Nectin-4 expression observed across the cohort. Notably, we demonstrated a statistically significant correlation between dMMR status and elevated Nectin-4 expression, suggesting potential biological interplay. The combined biomarker profile warrants further investigation as a predictive tool for therapeutic strategies involving antibody–drug conjugates (e.g., enfortumab vedotin) and immune checkpoint inhibitors.
Similar content being viewed by others
Avoid common mistakes on your manuscript.
Introduction
Urothelial carcinoma (UC) represents the predominant histological subtype of malignancies arising from the renal pelvis, ureter, and bladder. UC is anatomically classified into upper urinary tract UC (UTUC) and bladder UC (BUC). While BUC constitutes over 90% of UC cases, UTUC is relatively rare, accounting for 5–10% of diagnoses (Siegel et al. 2024). Up to 65% of UTUC patients present with muscle-invasive disease at diagnosis, compared to 15–25% of BUC patients (Rouprêt et al. 2023). Prognosis remains suboptimal, as approximately 50% of localized UC patients experience disease relapse or recurrence (Bianchi et al. 2023). Despite cisplatin-based chemotherapy remaining the first-line standard for advanced UC, 40% of patients are cisplatin-ineligible due to comorbidities, and most metastatic UC (mUC) cases develop therapeutic resistance (Powles et al. 2020). Immunotherapy has garnered substantial interest in UC, with pivotal trials demonstrating durable responses to immune checkpoint inhibitors (ICIs) as monotherapy or in combination (Necchi et al. 2018; Meeks et al. 2023; Alfred Witjes et al. 2024; Grivas et al. 2023; van der Heijden et al. 2023). Biomarkers of immune evasion and ICI responsiveness are under active investigation to optimize prognostic stratification and therapeutic algorithms (Hui et al. 2023; Liang et al. 2022).
Microsatellite instability (MSI) results from defective DNA mismatch repair (dMMR), manifested by loss of expression in ≥ 1 MMR proteins (MLH1, MSH2, MSH6, PMS2). MSI/dMMR status has been established as a significant biomarker of responses to immunotherapy (Wilbur et al. 2024). The European Association of Urology (EAU) guidelines on UTUC have updated the criteria for selecting UTUC patients for MSI testing (Rouprêt et al. 2023). Reported MSI prevalence varies widely in UTUC (1.6–28.1%) compared to BUC (0.7–1.5%), likely reflecting heterogeneous study populations and testing methodologies (Schneider et al. 2020; Kullmann et al. 2023; Gayhart et al. 2020; Hartmann et al. 2002; Catto et al. 2003; Ericson et al. 2005; Necchi et al. 2021; Audenet et al. 2019; Peak et al. 2023; Gerald et al. 2023).
Nectin-4 has emerged as a highly promising biomarker and therapeutic target in UC. Enfortumab vedotin (EV), an antibody–drug conjugate (ADC) targeting Nectin-4, has demonstrated clinically significant and durable responses (Challita-Eid et al. 2016; Rosenberg et al. 2020). In cisplatin-ineligible patients with locally advanced or metastatic UC, first-line EV combined with pembrolizumab showed enhanced clinical efficacy compared to EV monotherapy (O’Donnell et al. 2023). The combined biomarker profile (dMMR/Nectin-4) warrants further investigation as a predictive tool for therapeutic strategies.
This study aims to define the prevalence of dMMR in UC and correlate MSI status with clinicopathologic features and Nectin-4 expression.
Methods
Patients and materials
A consecutively collected cohort of 339 patients with histologically confirmed UTUC (renal pelvis or ureter) diagnosed between December 2016 and September 2023 at Peking University Third Hospital was retrospectively analyzed. Inclusion criteria: (1) Pathologically confirmed UTUC; (2) Sufficient preserved tumor tissue for immunohistochemistry (IHC); and (3) Complete clinicopathological data. Exclusion criteria: (1) Prior history of BUC.
Immunohistochemistry and evaluation
Formalin-fixed, paraffin-embedded (FFPE) tumor specimens were sectioned into 4-μm slides. After deparaffinization with xylene and rehydration with graded ethanol, endogenous peroxidase activity was blocked with 3% H2O2 for 10 min. Heat-induced epitope retrieval (HIER) was performed using a pressure cooker with EDTA buffer (pH 9.0). Slides were incubated overnight at 4 °C with the following primary antibodies: MSH2 (ZSGB-BIO, ZA-0622, 1:100), MSH6 (ZSGB-BIO, ZA-0541, 1:100), MLH1 (ZSGB-BIO, ZM-0154, 1:25), and PMS2 (ZSGB-BIO, ZA-0542, 1:20), Nectin-4 (Abcam, ab192033, 1:800) at 4 °C overnight. After incubation with secondary antibody (ZSGB-BIO, PV-6000) at 37 °C for 25 min, staining was visualized using 3,3′-diaminobenzidine (DAB; Gene Tech, GK600705) and counterstained with hematoxylin. Slides were scanned at 400 × magnification (Olympus, Japan), and two independent pathologists blinded to clinical data scored staining intensity and distribution.
dMMR: Defined as complete loss of nuclear staining in tumor cells for ≥ 1 MMR protein, with intact staining in internal controls (lymphocytes/stroma).
Nectin-4 expression was quantitatively assessed using the validated histochemical score (H-score) system. This scoring metric was calculated by multiplying the staining intensity (graded 0–3: 0 = negative, 1 = weak, 2 = moderate, 3 = strong) by the corresponding percentage of positively stained tumor cells (0–100%), followed by summation of these products to yield a final H-score range of 0–300. Rigorous validation included human placental tissue as a positive control and parallel sections with primary antibody omission as negative controls. Samples were then classified as negative (H-score 0–14), low (H-score 15–99), medium (H-score 100–199), and high (H-score 200–300), as previously described (Calandrella et al. 2022; Tekin et al. 2023).
Tumors were also categorized into three immune phenotypes as follows: inflamed phenotype, excluded phenotype and desert phenotype (Takahara et al. 2021).
Statistical analysis
Continuous variables are presented as mean ± standard deviation (SD). Categorical variables are summarized as frequencies and percentages. Group comparisons were performed using: Student’s t-test for normally distributed continuous variables, Mann–Whitney U test for non-normally distributed continuous variables, and Chi-square or Fisher’s exact test for categorical variables, as appropriate. Statistical analysis was performed using SPSS version 24.0 (SPSS, Inc.). Significance was defined as P < 0.05.
Results
A total of 339 consecutive, treatment-naïve UTUC tumor samples were included in this study. Clinicopathologic characteristics of the cohort are summarized in Table 1. 25 patients (7.4%) exhibited dMMR. The patterns of MMR proteins IHC expression are shown in Fig. 1.
Immunohistochemical expression of MMR proteins in UTUC specimens. A The positive expression of MSH2. B The negative expression of MSH2. C The positive expression of MSH6. D The negative expression of MSH6. E The positive expression of MLH1. F The negative expression of MLH1. G The positive expression of PMS2. H The negative expression of PMS2. Note: MMR, mismatch repair; UTUC, upper tract urothelial carcinoma
We then analyzed correlation between MMR status and clinicopathologic features of the UTUC patients. Analysis of associations between MMR status and clinicopathologic features is detailed in Table 2. The patterns of Nectin-4 IHC expression are shown in Fig. 2. Notably, dMMR status was significantly associated with medium/high Nectin-4 expression (P = 0.044). However, no significant differences were observed between dMMR and proficient mismatch repair (pMMR) groups in gender distribution, age, BMI (body mass index), tumor grade, or immunophenotypic profiles. MMR protein deficiency patterns are detailed in Table 3. The predominant deficiency pattern was concurrent MSH2/MSH6 loss (56%), followed by combined MLH1/PMS2 deficiency (24%). Furthermore, analysis of MMR protein deficiency correlations with clinicopathological characteristics (Table 4) revealed that MSH6-negative expression was significantly associated with moderate/high Nectin-4 expression (P = 0.013). Comparisons with previous studies are presented in Supplementary Table 1.
Discussion
Cisplatin-based chemotherapy continues to serve as the cornerstone of adjuvant therapy for UTUC. However, cisplatin eligibility mandates adequate baseline health status, yet a significant proportion of real-world patients are ineligible. Approximately 50% of patients are ineligible due to post-nephroureterectomy renal impairment, advanced age, or preexisting chronic kidney disease (Huang et al. 2022). Furthermore, among patients receiving cisplatin-based regimens, treatment-related adverse events (TRAEs) are frequently reported. In the POUT trial, Grade ≥ 3 acute treatment-emergent adverse events occurred in 50% of participants, with 33% reporting serious adverse events (Birtle et al. 2020).
The therapeutic landscape for UTUC has shifted toward systemic therapies. Immunotherapy has gained prominence both as an adjuvant treatment for high-risk UTUC and in the second-line management of metastatic UTUC (mUTUC) (Rouprêt et al. 2023). Although PD-L1 expression is a widely used biomarker for predicting response to immune checkpoint inhibitors (ICIs), durable clinical benefit is not guaranteed in PD-L1-high patients, and responses may occur even in PD-L1-negative cases.
The MSI phenotype represents a well-validated biomarker across diverse malignancies (Dudley et al. 2016; Naboush et al. 2017). MSI is strongly associated with response to immunotherapy. Across multiple tumor types, MSI-H tumors demonstrate superior therapeutic efficacy and responsiveness to adjuvant chemotherapy and immunotherapy compared to MSS counterparts (Kullmann et al. 2023; Arabi et al. 2009; Sepulveda et al. 2017; Marcus et al. 2019). MSI-H tumors are characterized by elevated expression of immune checkpoint proteins such as PD-1 and PD-L1, rendering them particularly susceptible to PD-1/PD-L1 blockade (Marcus et al. 2019; Sepulveda et al. 2017). However, research on MSI status in UTUC remains limited. Reported incidence rates of MSI in UTUC vary widely (1.6–28.1%), attributable to heterogeneous study populations and racial disparities. Although pembrolizumab has received tissue-agnostic approval for advanced MSI-H or dMMR malignancies, UTUC remains underrepresented in clinical trials, with urothelial carcinomas constituting only 2.1% of cases. In the phase III KEYNOTE-045 trial comparing pembrolizumab to second-line chemotherapy in recurrent advanced urothelial carcinoma, UTUC cases comprised only 14% (n = 75) of the cohort, and MSI status was not assessed. Comparative analysis with prior investigations is presented in Supplemental Table 1. To our knowledge, this represents the largest cohort to date evaluating MMR status in UTUC patients, including substantial representation from China. This investigation revealed a dMMR prevalence of 7.4% in the UTUC cohort. Reported dMMR prevalence ranges from 2.4 to 26.3% across studies (García-Tello et al. 2014; Ito et al. 2020). Previous investigations (García-Tello et al. 2014; Shang et al. 2022) with relatively large sample size (> 100 cases) primarily employed tissue microarray (TMA) platforms. However, MMR deficiency may be underestimated due to limited tissue sampling of TMA. While interpretation of MMR expression in non-tumorous cores may produce false-positive results with limited samples, our methodology utilized full-tissue sections for IHC analysis, minimizing interpretive errors inherent to restricted sampling. Our results indicated that concurrent MSH2/MSH6 loss was the predominant form of MMR protein loss, consistent with previous studies (Schneider et al. 2020; Gayhart et al. 2020; Calandrella et al. 2022; Shang et al. 2022).
Our analysis revealed that tumors with dMMR status exhibited significantly elevated Nectin-4 expression compared to pMMR cases. Whereas prior research (Calandrella et al. 2022) was limited to 27 patients, our cohort of 339 cases provides robust validation of the association between dMMR status and enhanced Nectin-4 expression. Notably, we contribute the discovery that MSH6 deficiency exhibited a statistically significant association with elevated Nectin-4 expression levels (P = 0.013), whereas MSH2 deficiency similarly showed elevated expression trends that did not reach statistical significance. Enfortumab vedotin (EV), a novel antibody–drug conjugate (ADC), targets Nectin-4-expressing cells to deliver a microtubule-disrupting agent, thereby inducing apoptosis (Challita-Eid et al. 2016). Given the high prevalence of Nectin-4 expression in UC, EV monotherapy elicited durable responses in metastatic UC (mUC) cohorts in the EV-101 trial (Rosenberg et al. 2020). The EV-301 trial analysis revealed that EV therapy conferred a clinically meaningful overall survival advantage compared to chemotherapy in patients with advanced, previously treated UC. Beyond monotherapy, combination regimens of EV with ICIs (e.g., pembrolizumab) are under investigation in UC. EV plus pembrolizumab yielded superior outcomes to platinum-based chemotherapy in treatment-naïve advanced UC (Powles et al. 2024). Cisplatin-ineligible patients with locally advanced or metastatic UC receiving first-line EV plus pembrolizumab exhibited enhanced clinical benefit compared to EV monotherapy (O’Donnell et al. 2023). Our findings identify a synergistic biomarker profile in dMMR UTUC, characterized by Nectin-4 overexpression and PD-L1 upregulation, which may potentiate EV/ICI combination efficacy. This approach could circumvent cisplatin resistance mechanisms while leveraging the ‘immune-rich’ dMMR microenvironment. To our knowledge, this represents the largest cohort establishing an association between dMMR status and Nectin-4 overexpression in UTUC. We propose a biomarker-driven trial design: dMMR status for ICI cohort selection and Nectin-4 expression to stratify potential EV responders. In dMMR/MSI-H UTUC, Nectin-4 expression screening could optimize EV therapy selection, particularly for cisplatin-ineligible patients. We also advocate for integrating dMMR screening into ongoing ADC trials to validate its predictive role. This dual biomarker strategy may strategically refine therapy selection for EV-based regimens, addressing unmet needs in cisplatin-ineligible UTUC.
Our study has several limitations. First, this was a retrospective, single-center study. Despite representing the largest East Asian cohort to date, prospective multicenter studies are required to validate our findings. Second, a substantial proportion of patients were lost to follow-up, precluding analysis of associations between dMMR status/Nectin-4 expression and clinical outcomes in UTUC. Future studies should integrate longitudinal clinical data to evaluate the prognostic and predictive utility of these biomarkers. Furthermore, our study assessed dMMR/Nectin-4 status in primary tumors only; metastatic lesions may exhibit discordant biomarker profiles. We did not compare dMMR /Nectin-4 expression status in primary lesions and metastatic lesions. The lack of genomic or transcriptomic data precludes exploration of causal links between dMMR and Nectin-4 regulation.
Conclusion
7.4% of UTUC patients exhibited dMMR status and patients showed different levels of Nectin-4 expression. We also indicated a significant correlation between dMMR status and Nectin-4 high expression in UTUC. The observed pattern of combination of these data may be tested as a potential predictive tool for the use of a combination of immune-drug conjugates and immune checkpoint inhibitors. The need for further studies in this area is warranted.
Data availability
The data used and/or analysed during the current study are available from the corresponding author on reasonable request.
References
Alfred Witjes J, Max Bruins H, Carrión A, Cathomas R, Compérat E, Efstathiou JA, Fietkau R, Gakis G, Lorch A, Martini A, Mertens LS, Meijer RP, Milowsky MI, Neuzillet Y, Panebianco V, Redlef J, Rink M, Rouanne M, Thalmann GN, Sæbjørnsen S, Veskimäe E, van der Heijden AG (2024) European Association of Urology Guidelines on muscle-invasive and metastatic bladder cancer: summary of the 2023 guidelines. Eur Urol 85:17–31. https://doi.org/10.1016/j.eururo.2023.08.016
Arabi H, Guan H, Kumar S, Cote M, Bandyopadhyay S, Bryant C, Shah J, Abdul-Karim FW, Munkarah AR, Ali-Fehmi R (2009) Impact of microsatellite instability (MSI) on survival in high grade endometrial carcinoma. Gynecol Oncol 113:153–158. https://doi.org/10.1016/j.ygyno.2009.02.004
Audenet F, Isharwal S, Cha EK, Donoghue MTA, Drill EN, Ostrovnaya I, Pietzak EJ, Sfakianos JP, Bagrodia A, Murugan P, Dalbagni G, Donahue TF, Rosenberg JE, Bajorin DF, Arcila ME, Hechtman JF, Berger MF, Taylor BS, Al-Ahmadie H, Iyer G, Bochner BH, Coleman JA, Solit DB (2019) Clonal relatedness and mutational differences between upper tract and bladder urothelial carcinoma. Clin Cancer Res 25:967–976. https://doi.org/10.1158/1078-0432.Ccr-18-2039
Bianchi A, von Deimling M, Pallauf M, Yanagisawa T, Kawada T, Mostafaei H, Quhal F, Laukhtina E, Rajwa P, Majdoub M, Motlagh RS, Pradere B, Karakiewicz PI, Cerruto MA, Antonelli A, Shariat SF (2023) Perspectives on the future of urothelial carcinoma therapy: chemotherapy and beyond. Expert Opin Pharmacother 24:177–195. https://doi.org/10.1080/14656566.2022.2150966
Birtle A, Johnson M, Chester J, Jones R, Dolling D, Bryan RT, Harris C, Winterbottom A, Blacker A, Catto JWF, Chakraborti P, Donovan JL, Elliott PA, French A, Jagdev S, Jenkins B, Keeley FX Jr., Kockelbergh R, Powles T, Wagstaff J, Wilson C, Todd R, Lewis R, Hall E (2020) Adjuvant chemotherapy in upper tract urothelial carcinoma (the POUT trial): a phase 3, open-label, randomised controlled trial. Lancet 395:1268–1277. https://doi.org/10.1016/s0140-6736(20)30415-3
Calandrella ML, Francesconi S, Caprera C, Mosillo C, Caserta C, Giannarelli D, Corsi M, Macrini S, Guida A, Ascani S, Bracarda S (2022) Nectin-4 and DNA mismatch repair proteins expression in upper urinary tract urothelial carcinoma (UTUC) as a model for tumor targeting approaches: an ImGO pilot study. BMC Cancer 22:168. https://doi.org/10.1186/s12885-022-09259-z
Catto JW, Azzouzi AR, Amira N, Rehman I, Feeley KM, Cross SS, Fromont G, Sibony M, Hamdy FC, Cussenot O, Meuth M (2003) Distinct patterns of microsatellite instability are seen in tumours of the urinary tract. Oncogene 22:8699–8706. https://doi.org/10.1038/sj.onc.1206964
Challita-Eid PM, Satpayev D, Yang P, An Z, Morrison K, Shostak Y, Raitano A, Nadell R, Liu W, Lortie DR, Capo L, Verlinsky A, Leavitt M, Malik F, Aviña H, Guevara CI, Dinh N, Karki S, Anand BS, Pereira DS, Joseph IB, Doñate F, Morrison K, Stover DR (2016) Enfortumab vedotin antibody-drug conjugate targeting Nectin-4 is a highly potent therapeutic agent in multiple preclinical cancer models. Cancer Res 76:3003–3013. https://doi.org/10.1158/0008-5472.Can-15-1313
Dudley JC, Lin MT, Le DT, Eshleman JR (2016) Microsatellite instability as a biomarker for PD-1 blockade. Clin Cancer Res 22:813–820. https://doi.org/10.1158/1078-0432.Ccr-15-1678
Ericson KM, Isinger AP, Isfoss BL, Nilbert MC (2005) Low frequency of defective mismatch repair in a population-based series of upper urothelial carcinoma. BMC Cancer 5:23. https://doi.org/10.1186/1471-2407-5-23
García-Tello A, Ramón de Fata F, Andrés G, Ropero S, López JI, Angulo JC (2014) DNA repair genes and prognosis in sporadic forms of urothelial carcinoma of the upper urinary tract. Actas Urol Esp 38:600–607. https://doi.org/10.1016/j.acuro.2014.03.010
Gayhart MG, Johnson N, Paul A, Quillin JM, Hampton LJ, Idowu MO, Smith SC (2020) Universal mismatch repair protein screening in upper tract urothelial carcinoma. Am J Clin Pathol 154:792–801. https://doi.org/10.1093/ajcp/aqaa100
Gerald T, Margulis V, Meng X, Bagrodia A, Cole S, Qin Q, Call SG, Mauer E, Lotan Y, Woldu SL (2023) Actionable genomic landscapes from a real-world cohort of urothelial carcinoma patients. Urol Oncol 41:148.e17–48.e24. https://doi.org/10.1016/j.urolonc.2022.12.008
Grivas P, Park SH, Voog E, Caserta C, Gurney H, Bellmunt J, Kalofonos H, Ullén A, Loriot Y, Sridhar SS, Yamamoto Y, Petrylak DP, Sternberg CN, Gupta S, Huang B, Costa N, Laliberte RJ, di Pietro A, Valderrama BP, Powles T (2023) Avelumab first-line maintenance therapy for advanced urothelial carcinoma: comprehensive clinical subgroup analyses from the JAVELIN bladder 100 phase 3 trial. Eur Urol 84:95–108. https://doi.org/10.1016/j.eururo.2023.03.030
Hartmann A, Zanardo L, Bocker-Edmonston T, Blaszyk H, Dietmaier W, Stoehr R, Cheville JC, Junker K, Wieland W, Knuechel R, Rueschoff J, Hofstaedter F, Fishel R (2002) Frequent microsatellite instability in sporadic tumors of the upper urinary tract. Cancer Res 62:6796–6802
Huang J, Su R, Chen Z, Jiang S, Chen M, Yuan Y, Hu H, Fu C, Huang Z, Wang Z, Zheng B, Li C, Wang Z, Bao Y, Cai M, Guo J, Wei Q, Xue W (2022) The efficacy and safety of first-line treatment in cisplatin-ineligible advanced upper tract urothelial carcinoma patients: a comparison of PD-1 inhibitor and carboplatin plus gemcitabine chemotherapy. Oncoimmunology 11:2124691. https://doi.org/10.1080/2162402x.2022.2124691
Hui G, Stefanoudakis D, Zektser Y, Isaacs DJ, Hannigan C, Pantuck AJ, Drakaki A (2023) Do cancer genetics impact treatment decision making? Immunotherapy and beyond in the management of advanced and metastatic urothelial carcinoma. Curr Oncol 30:7398–7411. https://doi.org/10.3390/curroncol30080536
Ito T, Kono K, Eguchi H, Okazaki Y, Yamamoto G, Tachikawa T, Akagi K, Okada Y, Kawakami S, Morozumi M, Tamaru JI, Ishida H (2020) Prevalence of Lynch syndrome among patients with upper urinary tract carcinoma in a Japanese hospital-based population. Jpn J Clin Oncol 50:80–88. https://doi.org/10.1093/jjco/hyz140
Kullmann F, Strissel PL, Strick R, Stoehr R, Eckstein M, Bertz S, Wullich B, Sikic D, Wach S, Taubert H, Olbert P, Heers H, Lara MF, Macias ML, Matas-Rico E, Lozano MJ, Prieto D, Hierro I, van Doeveren T, Bieche I, Masliah-Planchon J, Beaurepere R, Boormans JL, Allory Y, Herrera-Imbroda B, Hartmann A, Weyerer V (2023) Frequency of microsatellite instability (MSI) in upper tract urothelial carcinoma: comparison of the Bethesda panel and the Idylla MSI assay in a consecutively collected, multi-institutional cohort. J Clin Pathol 76:126–132. https://doi.org/10.1136/jclinpath-2021-207855
Liang Y, Tan Y, Guan B, Guo B, Xia M, Li J, Shi Y, Yu Z, Zhang Q, Liu D, Yang X, Hao J, Gong Y, Shakeel M, Zhou L, Ci W, Li X (2022) Single-cell atlases link macrophages and CD8(+) T-cell subpopulations to disease progression and immunotherapy response in urothelial carcinoma. Theranostics 12:7745–7759. https://doi.org/10.7150/thno.77281
Marcus L, Lemery SJ, Keegan P, Pazdur R (2019) FDA approval summary: pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin Cancer Res 25:3753–3758. https://doi.org/10.1158/1078-0432.Ccr-18-4070
Meeks JJ, Black PC, Galsky M, Grivas P, Hahn NM, Hussain SA, Milowsky MI, Steinberg GD, Svatek RS, Rosenberg JE (2023) Checkpoint inhibitors in urothelial carcinoma-future directions and biomarker selection. Eur Urol 84:473–483. https://doi.org/10.1016/j.eururo.2023.05.011
Naboush A, Roman CA, Shapira I (2017) Immune checkpoint inhibitors in malignancies with mismatch repair deficiency: a review of the state of the current knowledge. J Investig Med 65:754–758. https://doi.org/10.1136/jim-2016-000342
Necchi A, Anichini A, Raggi D, Briganti A, Massa S, Lucianò R, Colecchia M, Giannatempo P, Mortarini R, Bianchi M, Farè E, Monopoli F, Colombo R, Gallina A, Salonia A, Messina A, Ali SM, Madison R, Ross JS, Chung JH, Salvioni R, Mariani L, Montorsi F (2018) Pembrolizumab as neoadjuvant therapy before radical cystectomy in patients with muscle-invasive urothelial bladder carcinoma (PURE-01): an open-label, single-arm, phase ii study. J Clin Oncol 36:3353–3360. https://doi.org/10.1200/jco.18.01148
Necchi A, Madison R, Pal SK, Ross JS, Agarwal N, Sonpavde G, Joshi M, Yin M, Miller VA, Grivas P, Chung JH, Ali SM (2021) Comprehensive genomic profiling of upper-tract and bladder urothelial carcinoma. Eur Urol Focus 7:1339–1346. https://doi.org/10.1016/j.euf.2020.08.001
O’Donnell PH, Milowsky MI, Petrylak DP, Hoimes CJ, Flaig TW, Mar N, Moon HH, Friedlander TW, McKay RR, Bilen MA, Srinivas S, Burgess EF, Ramamurthy C, George S, Geynisman DM, Bracarda S, Borchiellini D, Geoffrois L, Maroto Rey JP, Ferrario C, Carret AS, Yu Y, Guseva M, Homet Moreno B, Rosenberg JE (2023) Enfortumab vedotin with or without pembrolizumab in cisplatin-ineligible patients with previously untreated locally advanced or metastatic urothelial cancer. J Clin Oncol 41:4107–4117. https://doi.org/10.1200/jco.22.02887
Peak T, Spiess PE, Li R, Grivas P, Necchi A, Pavlick D, Huang RSP, Lin D, Danziger N, Jacob JM, Bratslavsky G, Ross JS (2023) Comparative genomic landscape of urothelial carcinoma of the bladder among patients of East and South Asian genomic ancestry. Oncologist 28:e910–e920. https://doi.org/10.1093/oncolo/oyad120
Powles T, van der Heijden MS, Castellano D, Galsky MD, Loriot Y, Petrylak DP, Ogawa O, Park SH, Lee JL, De Giorgi U, Bögemann M, Bamias A, Eigl BJ, Gurney H, Mukherjee SD, Fradet Y, Skoneczna I, Tsiatas M, Novikov A, Suárez C, Fay AP, Duran I, Necchi A, Wildsmith S, He P, Angra N, Gupta AK, Levin W, Bellmunt J (2020) Durvalumab alone and durvalumab plus tremelimumab versus chemotherapy in previously untreated patients with unresectable, locally advanced or metastatic urothelial carcinoma (DANUBE): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol 21:1574–1588. https://doi.org/10.1016/s1470-2045(20)30541-6
Powles T, Valderrama BP, Gupta S, Bedke J, Kikuchi E, Hoffman-Censits J, Iyer G, Vulsteke C, Park SH, Shin SJ, Castellano D, Fornarini G, Li JR, Gümüş M, Mar N, Loriot Y, Fléchon A, Duran I, Drakaki A, Narayanan S, Yu X, Gorla S, Homet Moreno B, van der Heijden MS (2024) Enfortumab vedotin and pembrolizumab in untreated advanced urothelial cancer. N Engl J Med 390:875–888. https://doi.org/10.1056/NEJMoa2312117
Rosenberg J, Sridhar SS, Zhang J, Smith D, Ruether D, Flaig TW, Baranda J, Lang J, Plimack ER, Sangha R, Heath EI, Merchan J, Quinn DI, Srinivas S, Milowsky M, Wu C, Gartner EM, Zuo P, Melhem-Bertrandt A, Petrylak DP (2020) EV-101: a phase I study of single-agent enfortumab vedotin in patients with Nectin-4-positive solid tumors, including metastatic urothelial carcinoma. J Clin Oncol 38:1041–1049. https://doi.org/10.1200/jco.19.02044
Rouprêt M, Seisen T, Birtle AJ, Capoun O, Compérat EM, Dominguez-Escrig JL, Gürses Andersson I, Liedberg F, Mariappan P, Hugh Mostafid A, Pradere B, van Rhijn BWG, Shariat SF, Rai BP, Soria F, Soukup V, Wood RG, Xylinas EN, Masson-Lecomte A, Gontero P (2023) European Association of Urology Guidelines on upper urinary tract urothelial carcinoma: 2023 update. Eur Urol 84:49–64. https://doi.org/10.1016/j.eururo.2023.03.013
Schneider B, Glass Ä, Jagdmann S, Hühns M, Claus J, Zettl H, Dräger DL, Maruschke M, Hakenberg OW, Erbersdobler A, Zimpfer A (2020) Loss of Mismatch-repair protein expression and microsatellite instability in upper tract urothelial carcinoma and clinicopathologic implications. Clin Genitourin Cancer 18:e563–e572. https://doi.org/10.1016/j.clgc.2020.03.006
Sepulveda AR, Hamilton SR, Allegra CJ, Grody W, Cushman-Vokoun AM, Funkhouser WK, Kopetz SE, Lieu C, Lindor NM, Minsky BD, Monzon FA, Sargent DJ, Singh VM, Willis J, Clark J, Colasacco C, Rumble RB, Temple-Smolkin R, Ventura CB, Nowak JA (2017) Molecular biomarkers for the evaluation of colorectal cancer. Am J Clin Pathol 147:221–260. https://doi.org/10.1093/ajcp/aqw209
Shang Z, Jin S, Wang W, Wei Y, Gu C, Yang C, Zhu Y, Zhu Y, Shen Y, Wu J, Ye D (2022) Clinicopathological characteristics and loss of mismatch repair protein expression in Chinese upper tract urothelial carcinomas. Front Oncol 12:1012168. https://doi.org/10.3389/fonc.2022.1012168
Siegel RL, Giaquinto AN, Jemal A (2024) Cancer statistics, 2024. CA Cancer J Clin 74:12–49. https://doi.org/10.3322/caac.21820
Takahara T, Murase Y, Tsuzuki T (2021) Urothelial carcinoma: variant histology, molecular subtyping, and immunophenotyping significant for treatment outcomes. Pathology 53:56–66. https://doi.org/10.1016/j.pathol.2020.09.004
Tekin B, Cheville JC, Herrera Hernandez L, Negron V, Smith CY, Jenkins SM, Dasari S, Enninga EAL, Norgan AP, Menon S, Cubilla AL, Whaley RD, Jimenez RE, Thompson RH, Leibovich BC, Karnes RJ, Boorjian SA, Pagliaro LC, Erickson LA, Guo R, Gupta S (2023) Assessment of PD-L1, TROP2, and nectin-4 expression in penile squamous cell carcinoma. Hum Pathol 142:42–50. https://doi.org/10.1016/j.humpath.2023.10.003
van der Heijden MS, Sonpavde G, Powles T, Necchi A, Burotto M, Schenker M, Sade JP, Bamias A, Beuzeboc P, Bedke J, Oldenburg J, Chatta G, Ürün Y, Ye D, He Z, Valderrama BP, Ku JH, Tomita Y, Filian J, Wang L, Purcea D, Patel MY, Nasroulah F, Galsky MD (2023) Nivolumab plus gemcitabine-cisplatin in advanced urothelial carcinoma. N Engl J Med 389:1778–1789. https://doi.org/10.1056/NEJMoa2309863
Wilbur HC, Le DT, Agarwal P (2024) Immunotherapy of MSI cancer: facts and hopes. Clin Cancer Res 30:1438–1447. https://doi.org/10.1158/1078-0432.Ccr-21-1935
Funding
This work was supported by CSCO Clinical Oncology Research Foundation of Beijing (Y-tongshu2021/ms-0072).
Author information
Authors and Affiliations
Contributions
All authors contributed to the study conception and design. Study design was performed by Yichang Hao and Shudong Zhang. Material preparation and data collection were performed by Peichen Duan, Le Yu, Shaohui Deng and Peng Hong. Immunohistochemistry and analysis were performed by Peichen Duan, Le Yu and Min Lu. The first draft of the manuscript was written by Peichen Duan and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.
Corresponding authors
Ethics declarations
Conflict of interest
The authors declare no competing interests.
Ethics approval and consent to participate
All patients have been given informed consent and that the study protocol was approved by the institute’s committee on human research. This study protocol was reviewed and approved by Peking University Third Hospital Medical Science Research Ethics Committee (M2023800). This study protocol was performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki and its later amendments.
Consent for publication
Human research participants have provided informed consent for publication of the images in Figs. 1 and 2.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.
About this article
Cite this article
Duan, P., Yu, L., Hao, Y. et al. Deficient DNA mismatch repair and Nectin-4 expression in upper tract urothelial carcinoma (UTUC). J Cancer Res Clin Oncol 151, 280 (2025). https://doi.org/10.1007/s00432-025-06312-9
Received:
Accepted:
Published:
DOI: https://doi.org/10.1007/s00432-025-06312-9