Are primary tumors suitable for biomarker-guided treatment of metastatic urothelial cancer?
Editorial Commentary

Are primary tumors suitable for biomarker-guided treatment of metastatic urothelial cancer?

Mathijs P. Scholtes1 ORCID logo, Geert J. L. H. van Leenders2 ORCID logo, Debbie G. J. Robbrecht3 ORCID logo, Joost L. Boormans1 ORCID logo, Tahlita C. M. Zuiverloon1 ORCID logo

1Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands; 2Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands; 3Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands

Correspondence to: Tahlita C. M. Zuiverloon, MD, PhD. Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands. Email: t.zuiverloon@erasmusmc.nl.

Comment on: Cox A, Klümper N, Stein J, et al. Molecular Urothelial Tumor Cell Subtypes Remain Stable During Metastatic Evolution. Eur Urol 2024;85:328-32.


Keywords: Metastatic urothelial carcinoma (mUC); metastatic evolution; molecular subtypes; biomarkers


Submitted Mar 01, 2024. Accepted for publication Jun 26, 2024. Published online Aug 26, 2024.

doi: 10.21037/tau-24-112


In recent years, the therapeutic armamentarium for patients with metastatic urothelial carcinoma (mUC) has expanded with the emergence of novel therapies like immune checkpoint inhibitors (ICIs), tyrosine kinase inhibitors (TKIs) targeting fibroblast growth factor receptors (FGFRs), and antibody-drug conjugates (ADCs). Although FGFR inhibitors are reserved for patients with FGFR altered urothelial carcinoma (UC), accurate predictive biomarkers for these new drugs that correlate to individual patient responses, are lacking. Biomarker-guided response prediction on primary tumor samples is appealing due to the availability of archived tumor material typically present at the diagnosis of mUC. Additionally, the bladder is easy to access to obtain tumor biopsies necessary for biomarker assessment, whereas fine needle aspiration or biopsy from distant metastatic sites can be technically challenging. However, primary bladder tumors might not be representative of metastatic lesions, as the stability of UC phenotypes during progression to metastatic disease remains elusive.

This poses an important limitation that needs to be addressed before the implementation of biomarker-guided treatment strategies. Biomarkers predictive of response may be present in primary bladder tumors but absent in metastatic lesions, potentially leading to ineffective treatment. The other way around is also possible, as a result of acquired disease features over time. We therefore appreciated reading the study “Molecular Urothelial Tumor Cell Subtypes Remain Stable During Metastatic Evolution” and want to commend the authors for their valuable findings (1). Cox et al. performed immunohistochemistry (IHC)-based molecular subtyping on 138 primary UC and matched mUC samples, and discovered luminal, basal, and neuroendocrine molecular subtypes to remain relatively stable during metastatic evolution (94% concordance between matched pairs). In contrast, transcriptome-based muscle-invasive bladder cancer (MIBC) consensus subtyping on a subset of matched primary (N=20) and metastatic (N=20) samples proved to be highly variable (45% concordance).

Prior genetic studies comparing primary and matched mUC samples, showed early evolutionary branching of metastatic tumor clones, with limited mutations shared between the metastatic lesions and the bulk of the primary tumor (2,3). Noteworthy, potentially actionable gene mutations [FGFR3, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), TSC complex subunit 1 (TSC1), Erb-B2 receptor tyrosine kinase 2 (ERBB2)] have been reported to be discordant in up to 23% of primary and metastatic tumor pairs (4). Intratumoral heterogeneity and sampling bias contribute to this genetic discordance, exemplified by the enrichment of FGFR3 mutations in superficial areas of transurethral resection of bladder tumor (TURBT) samples compared to more invasive regions of the tumor (5). Taken together, evidence suggest that primary tumor samples might not be representative of metastatic lesions. High phenotypic concordance at the protein level observed by Cox et al., however, demonstrated at least partial phenotypic stability during tumor evolution and metastatic dissemination, despite patients undergoing different systemic treatments between sequential samples. The observed similarity between primary tumors and metastatic lesions warrants scientists not to exclude primary tumors from efforts investigating novel biomarkers.

Bulk RNA sequencing-based consensus cluster (CC) molecular subtypes provide a snapshot of the tumor-microenvironment (TME), and might not be optimal for characterizing metastatic lesions of UC (6,7). Briefly, MIBC consensus subtypes (8) were designed for the classification of primary tumors and lean heavily on stromal content (e.g., luminal infiltrated, stroma-rich). The applicability of CCs to characterize and compare different metastatic lesions with diverse associated TMEs is therefore limited, and site-specific classification systems might be more appropriate (9). Additionally, interpretation of CCs applied to post-treatment samples must be performed with care, as surgical or systemic pre-treatment induces scar-like phenotypes associated with fibroblasts and the stroma-rich subtype (10,11). Subtype switches between TURBT and radical cystectomy (RC) tumor samples observed in earlier studies were often centered around the stroma-rich subtype, as is the case for the majority of discordant samples observed by Cox et al. (8 out of 11). Since the majority of primary tumors analyzed by Cox et al. was derived at RC it is difficult to assess if the observed discordance between primary and metastatic samples truly reflected naturally occurring metastatic tumor evolution or simply highlights treatment effects induced by local surgery. Nonetheless, there is currently no role in routine practice for molecular subtype determination in UC (12).

Biomarkers that are used in current clinical practice are programmed cell death ligand 1 (PD-L1) expression on immune cells (ICs) and the combined positive score (CPS), which are IHC-based companion diagnostics designed to guide ICI in mUC patients (12). A study evaluating PD-L1 expression and CPS scores between matched primary tumors and metastatic lesions suggested that tumor cell PD-L1 expression and CPS positivity tended to decrease during metastatic progression (13). A similar observation was reported in a small study evaluating PD-L1 expression in primary tumors and matched liver metastases (14). While PD-L1 expression remained stable on tumor cells, the presence of tumor infiltrating lymphocytes (TILs) and accompanying PD-L1 expression on TILs was significantly lower in liver metastases than in matched primary tumors (14).

Consistent with these findings, two separate studies reported around 75–80% concordance of PD-L1 expression when comparing primary and matched metastatic samples (15,16). Concordance of PD-L1 expression was significantly lower in patients treated with pre-operative chemotherapy (15), while concordance rates observed between synchronous and metachronous primary and matched metastatic pairs did not significantly differ (16). However, as most samples have low IC PD-L1 positivity and/or CPS, the inherent likelihood of concordance between primary and metastatic samples is relatively high. Consequently, future research should prioritize investigating the predictive value of biomarkers in both primary and metastatic samples to anticipate patient responses to treatment.

With the emergence of ADCs, target proteins like NECTIN-4 [enfortumab vedotin (EV)] and TROP2 [sacituzumab govitecan (SG)] are upcoming markers of particular interest. The phase I EV-101 study, evaluating EV in patients with solid tumors, has been amended during the trial duration because the majority of patients with UC exhibited high levels of NECTIN-4 tumor staining and this eligibility requirement was removed (17). This modification allowed for a broader exploration of EV response, albeit without a specific focus on NECTIN-4 expression. In the EV-201 study, a phase 2 single-arm open label study of EV in either third line cisplatinum-eligible patients or second line in cisplatinum-ineligible UC patients, NECTIN-4 expression was examined via IHC. NECTIN-4 expression was not an eligibility criterion, however, in this study its range of expression was fairly broad (18). Preclinical studies on TROP2 showed high protein expression of TROP2 in UC, and as a result, TROP2 expression was not an inclusion criterion in TROPHY-U-01, the phase 2 single-arm trial evaluating SG in UC patients (19,20).

Disparate expression of NECTIN-4 and TROP2 has been shown in different histological subtypes (21), although data are limited. UCs can have variable and mixed histology in 25–30% of cases (22), comprising a substantial part of the patient population. For molecular subtypes, NECTIN-4 shows a high level of expression mainly in luminal-a UC (9,21), whereas TROP2 mRNA expression is consistently high across molecular subtypes, except for the neuroendocrine subtype (23). A recent study reported decreased NECTIN-4 expression levels in primary versus matched metastatic UC, with 40% of metastatic lesions lacking membranous NECTIN-4 protein expression (24). In contrast, a correlation between TROP2 mRNA levels and TROP2 protein expression (IHC) was observed and, there was no apparent decrease of TROP2 mRNA expression in metastatic samples when compared to the paired primary tumor. Taken together, it is of importance to recognize that expression levels of membranous targets are affected by intra- and inter-patient heterogeneity and disease evaluation overtime.

While ADC effectivity can be affected by several factors, e.g., ADC composition, target expression, intra-tumoral drug penetration, and composition of ICs and stromal cells in the tumor micro-environment, thus so far, no clear predictor of patient response to ADC has been identified, with the relation between target expression and drug efficacy still being unresolved. A relationship between target expression level and degree of ADC activity seems to be logical, although, target protein expression based on IHC may not be the best candidate for patient selection. Membranous NECTIN-4 expression was associated with survival benefit in 47 EV treated patients, with 21 of 47 cases being assessed in metastatic lesions (24). Prospective trials are needed to identify predictive biomarkers for ADC efficacy, including the evaluation in matched primary and metastatic lesions.

To conclude, consistent protein-expression levels of urothelial differentiation markers observed between primary and metastatic UC suggest that biomarkers can be assessed on (archived) primary tumor material, omitting the need for target metastatic lesion biopsies and associated treatment delay for some patients. Prior to clinical implementation, the predictive value of novel biomarkers should be prospectively evaluated and reported separately for primary tumors and metastatic target lesions. An illustrative example is the THOR trial, wherein cohort I randomized patients with FGFR2 or FGFR3 alterations to either erdafitinib (an FGFR inhibitor) treatment or the investigator’s choice of 3rd line chemotherapy (25). The authors stated that most alterations were evaluated on archived tumor tissue but did not report whether examining fresh metastatic biopsies or archived primary tumor material resulted in different patient outcomes. This overlooks a chance to gain valuable insights into the utilization of archived primary tumor samples for treatment selection of mUC patients whom often receive multiple lines of therapy between initial diagnosis and the start of 3rd line therapy.

With current evidence in mind, it seems that presence of biomarkers in primary tumors often does not reflect their status in metastatic lesions. Consequently, conducting biomarker assessment on primary tumors to guide treatment of mUC patients should be approached with care.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Translational Andrology and Urology. The article has undergone external peer review.

Peer Review File: Available at https://tau.amegroups.com/article/view/10.21037/tau-24-112/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tau.amegroups.com/article/view/10.21037/tau-24-112/coif). D.G.J.R. reports consulting fees (payment to institution) from Janssen, MSD, Merck AG, Pfizer; Payment or honoraria for lectures, presentations, speakers’ bureaus, manuscript writing or educational events for Astellas (payment to institution); and support for attending meetings and/or travel from Janssen. J.L.B. reports consultancy for or speaker’s fee from Janssen, BMS, AstraZeneca, Merck AG/Pfizer, MSD, Bayer, and Ismar Healthcare (all paid to institution); research collaboration with Merck AG/Pfizer, MSD, Janssen, and VitroScan (all paid to institution); Book writing for Astellas; and serving as Member of the EAU Testicular Cancer Guidelines panel. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Cox A, Klümper N, Stein J, et al. Molecular Urothelial Tumor Cell Subtypes Remain Stable During Metastatic Evolution. Eur Urol 2024;85:328-32. [Crossref] [PubMed]
  2. Faltas BM, Prandi D, Tagawa ST, et al. Clonal evolution of chemotherapy-resistant urothelial carcinoma. Nat Genet 2016;48:1490-9. [Crossref] [PubMed]
  3. Winters BR, De Sarkar N, Arora S, et al. Genomic distinctions between metastatic lower and upper tract urothelial carcinoma revealed through rapid autopsy. JCI Insight 2019;5:e128728. [Crossref] [PubMed]
  4. Clinton TN, Chen Z, Wise H, et al. Genomic heterogeneity as a barrier to precision oncology in urothelial cancer. Cell Rep 2022;41:111859. [Crossref] [PubMed]
  5. Pouessel D, Neuzillet Y, Mertens LS, et al. Tumor heterogeneity of fibroblast growth factor receptor 3 (FGFR3) mutations in invasive bladder cancer: implications for perioperative anti-FGFR3 treatment. Ann Oncol 2016;27:1311-6. [Crossref] [PubMed]
  6. Höglund M; Lund Bladder Cancer Group. Re: Alexander Cox, Niklas Klümper, Johannes Stein, et al. Molecular Urothelial Tumor Cell Subtypes Remain Stable During Metastatic Evolution. Eur Urol. In press. 10.1016/j.eururo.2023.03.020. Eur Urol 2023;84:e50.10.1016/j.eururo.2023.03.020
  7. Song Y, Peng Y, Xu T. Letter to the editor for the article “Molecular urothelial tumor cell subtypes remain stable during metastatic evolution”. World J Urol 2023;41:2869-70. [Crossref] [PubMed]
  8. Kamoun A, de Reyniès A, Allory Y, et al. A Consensus Molecular Classification of Muscle-invasive Bladder Cancer. Eur Urol 2020;77:420-33. [Crossref] [PubMed]
  9. Nakauma-González JA, Rijnders M, van Riet J, et al. Comprehensive Molecular Characterization Reveals Genomic and Transcriptomic Subtypes of Metastatic Urothelial Carcinoma. Eur Urol 2022;81:331-6. [Crossref] [PubMed]
  10. Necchi A, de Jong JJ, Raggi D, et al. Molecular Characterization of Residual Bladder Cancer after Neoadjuvant Pembrolizumab. Eur Urol 2021;80:149-59. [Crossref] [PubMed]
  11. Seiler R, Gibb EA, Wang NQ, et al. Divergent Biological Response to Neoadjuvant Chemotherapy in Muscle-invasive Bladder Cancer. Clin Cancer Res 2019;25:5082-93. [Crossref] [PubMed]
  12. Powles T, Bellmunt J, Comperat E, et al. Bladder cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2022;33:244-58. [Crossref] [PubMed]
  13. 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. [Crossref] [PubMed]
  14. Eckstein M, Sikic D, Strissel PL, et al. Evolution of PD-1 and PD-L1 Gene and Protein Expression in Primary Tumors and Corresponding Liver Metastases of Metastatic Bladder Cancer. Eur Urol 2018;74:527-9. [Crossref] [PubMed]
  15. de Jong JJ, Stoop H, Nieboer D, et al. Concordance of PD-L1 expression in matched urothelial bladder cancer specimens. Histopathology 2018;73:983-9. [Crossref] [PubMed]
  16. Erlmeier F, Klümper N, Landgraf L, et al. Spatial Immunephenotypes of Distant Metastases but not Matched Primary Urothelial Carcinomas Predict Response to Immune Checkpoint Inhibition. Eur Urol 2023;83:133-42. [Crossref] [PubMed]
  17. Rosenberg J, Sridhar SS, Zhang J, et al. 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 2020;38:1041-9. [Crossref] [PubMed]
  18. Rosenberg JE, O’Donnell PH, Balar AV, et al. Pivotal Trial of Enfortumab Vedotin in Urothelial Carcinoma After Platinum and Anti-Programmed Death 1/Programmed Death Ligand 1 Therapy. J Clin Oncol 2019;37:2592-600. [Crossref] [PubMed]
  19. Tagawa ST, Balar AV, Petrylak DP, et al. TROPHY-U-01: A Phase II Open-Label Study of Sacituzumab Govitecan in Patients With Metastatic Urothelial Carcinoma Progressing After Platinum-Based Chemotherapy and Checkpoint Inhibitors. J Clin Oncol 2021;39:2474-85. [Crossref] [PubMed]
  20. Abbas M, Heitplatz B, Bernemann C, et al. Immunohistochemical expression of TROP 2 (TACSTD2) on the urothelial carcinoma of the urinary bladder and other types of cancer. Oncol Lett 2023;26:527. [Crossref] [PubMed]
  21. Bahlinger V, Branz A, Strissel PL, et al. Associations of TACSTD2/TROP2 and NECTIN-4/NECTIN-4 with molecular subtypes, PD-L1 expression, and FGFR3 mutational status in two advanced urothelial bladder cancer cohorts. Histopathology 2024;84:863-76. [Crossref] [PubMed]
  22. Black AJ, Black PC. Variant histology in bladder cancer: diagnostic and clinical implications. Transl Cancer Res 2020;9:6565-75. [Crossref] [PubMed]
  23. Chou J, Trepka K, Sjöström M, et al. TROP2 Expression Across Molecular Subtypes of Urothelial Carcinoma and Enfortumab Vedotin-resistant Cells. Eur Urol Oncol 2022;5:714-8. [Crossref] [PubMed]
  24. Klümper N, Ralser DJ, Ellinger J, et al. Membranous NECTIN-4 Expression Frequently Decreases during Metastatic Spread of Urothelial Carcinoma and Is Associated with Enfortumab Vedotin Resistance. Clin Cancer Res 2023;29:1496-505. [Crossref] [PubMed]
  25. Loriot Y, Matsubara N, Park SH, et al. Erdafitinib or Chemotherapy in Advanced or Metastatic Urothelial Carcinoma. N Engl J Med 2023;389:1961-71. [Crossref] [PubMed]
Cite this article as: Scholtes MP, van Leenders GJLH, Robbrecht DGJ, Boormans JL, Zuiverloon TCM. Are primary tumors suitable for biomarker-guided treatment of metastatic urothelial cancer? Transl Androl Urol 2024;13(8):1341-1345. doi: 10.21037/tau-24-112

Download Citation