Bladder-sparing approaches for muscle invasive bladder cancer: a narrative review of current evidence and future perspectives
Review Article

Bladder-sparing approaches for muscle invasive bladder cancer: a narrative review of current evidence and future perspectives

Xinxiang Fan^, Wang He^, Jian Huang

Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China

Contributions: (I) Conception and design: J Huang; (II) Administrative support: J Huang; (III) Provision of study materials or patients: W He; (IV) Collection and assembly of data: X Fan; (V) Data analysis and interpretation: X Fan; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

^ORCID: Xinxiang Fan, 0000-0001-8495-6488; Wang He, 0000-0002-3765-3991.

Correspondence to: Jian Huang, MD. Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiangxi Road, Guangzhou 510120, China. Email: huangj8@mail.sysu.edu.cn.

Background and Objective: In recent years, the application of less-invasive “bladder-sparing” trimodal therapy (TMT) in selected muscle-invasive bladder cancer (MIBC) patients unfit for or who declined radical cystectomy (RC) has been increasing. This review aims to summarize the current evidence and future perspectives of bladder-sparing therapy for MIBC.

Methods: A non-systematic Medline/PubMed literature search was conducted on July 2022 with the following keywords ‘MIBC’, ‘bladder-sparing’, ‘chemotherapy’, ‘radiotherapy’, ‘trimodal’, ‘multimodal’, and ‘immunotherapy’.

Key Content and Findings: All monotherapies are inferior to RC or combination therapy and should not be routinely used for curative intent. Radiotherapy (RT) alone has been shown to have poorer outcomes when compared to chemoradiotherapy. The ideal selection criteria for TMT include good bladder function and capacity, clinical stage within cT2, complete transurethral resection of bladder tumor (TURBT), no prior history of pelvic RT, no extensive carcinoma in situ (CIS), and absence of hydronephrosis. The emergence of immunotherapy may further increase the effect of bladder-sparing therapy. Novel predictive biomarkers are awaited for more precise patient selection and better oncological outcomes.

Conclusions: TMT is a well-tolerated and offers a curative alternative approach to RC for selected patients with localized MIBC. Appropriate patient selection and a multi-disciplinary approach is crucial in achieving good oncologic control with bladder-sparing therapy.

Keywords: Muscle invasive bladder cancer (MIBC); bladder preservation therapy; bladder-sparing; chemoradiation; trimodal therapy (TMT)


Submitted Feb 13, 2023. Accepted for publication Apr 19, 2023. Published online May 26, 2023.

doi: 10.21037/tau-23-124


Introduction

Bladder cancer is the 7th most common cancer in males, and it ranks the 10th when both genders are included (1). An estimate of 573,278 new bladder cancer patients and 212,536 new bladder cancer-associated deaths occurred in 2020 worldwide according to the recent report of the global cancer burden (2).

The gold standard treatment for muscle invasive bladder cancer (MIBC) is radical cystectomy (RC). However, the 5-year overall survival (OS) after RC is only about 50% (3-6). As RC is associated with nonnegligible morbidity and mortality, clinicians have sought alternative treatments for patients unfit for or refusing RC. Organ-sparing multimodality approaches have been widely used as first-line therapies in multiple malignancies, such as breast cancer (7), cervical cancer (8), and anal cancer (9). Organ preservation could ideally maintain satisfactory function of the native organ following treatment. In recent years, the application of less-invasive “bladder-sparing” trimodal therapy (TMT) has been increasing, which involves maximal transurethral resection of bladder tumor (TURBT), followed by concurrent chemotherapy and RT (10). TMT is a well-tolerated and offers a curative alternative approach to RC for selected localized MIBC patients (11). Several organizations, including the European Association of Urology (EAU) and the American Urological Association (AUA), have updated the guidelines to support the application of chemotherapy combined with RT in selected MIBC patients unfit for or who declined RC (1,12,13). This review aims to summarize the current evidence and future perspectives of bladder-sparing therapy for MIBC. We present this article in accordance with the Narrative Review reporting checklist (available at https://tau.amegroups.com/article/view/10.21037/tau-23-124/rc).


Methods

We performed a non-systematic Medline/PubMed literature search on 20 July 2022 with the following keywords ‘MIBC’, ‘bladder-sparing’, ‘chemotherapy’, ‘radiotherapy’, ‘trimodal’, ‘multimodal’, and ‘immunotherapy’. The search strategy is summarized in Table 1.

Table 1

The search strategy summary

Items Specification
Date of search 20 July 2022
Databases and other sources searched Medline/PubMed
Search terms used ‘MIBC’, ‘bladder-sparing’, ‘chemotherapy’, ‘radiotherapy’, ‘trimodal’, ‘multimodal’, and ‘immunotherapy’
Timeframe January 1987 to July 2022
Inclusion and exclusion criteria Study type: original studies of RCTs, cohort studies, case-control studies, and case series. Language restriction: English
Selection process Selection process was conducted independently by two of the authors (Fan and He). When there was a disagreement, consensus was obtained by the adjudicating of the senior author (Huang)

MeSH, medical subject heading; RCT, randomized controlled trial; MIBC, muscle invasive bladder cancer.


TURBT

A TURBT is usually the first and crucial treatment for bladder cancer. The EAU, AUA, and National Comprehensive Cancer Network (NCCN) guidelines recommend thoracic, abdominal and pelvic CT scan to evaluate the clinical stage MIBC (1,12,13).

However, it is worth noting that all monotherapies are inferior to RC or combination therapy and should not be routinely used for curative purposes (1,12-14). As a therapeutic option, TURBT is only possible when the tumor is confined to the superficial muscle layer and second biopsy reveals a free of residual (invasive) tumor (1,15). It is worth noting that about 20% of MIBC have nodal involvement at final pathology. Therefore, the staging ability and the potential therapeutic effect of an extended pelvic lymph node dissection (ePLND) is missed in a TURBT-only approach. Thus, as a solo modality, TURBT should not be recommended as the standard of care in patients with MIBC.

A prospective study by Solsona et al. reported complete TURBT in 133 highly-selected MIBC patients with no residual tumor in post-treatment biopsy. Recurrent non(N)-MIBC occurred in 30% of patients, whereas disease progression occurred in another 30% of patients. After 5, 10, and 15 years, the results showed that OS was 73.7%, 39.8%, and 24.8%, respectively, and cancer-specific survival (CSS) was 81.9%, 79.5%, and 76.7%, respectively (16). A study by Herr demonstrated similar results with a 76% 10-year CSS in 99 MIBC patients who underwent TURBT alone (17).

The goal of a “maximal TURBT” is to reset all visible tumors without compromising surgical safety. Several studies have investigated the association of a visibly complete TURBT between complete response (CR) rate, OS, and long-term bladder preservation rate after TMT (18,19).

According to Efstathiou et al., although a visibly complete TURBT is preferred, CR was achieved in 57% patients with a visibly incomplete TURBT following the combination therapy of chemoradiation (18). Fluorescence-based cystoscopy may show multifocal lesions which may have otherwise been missed, such as carcinoma in situ (CIS) or smaller Ta/T1 tumors, when focusing on a T2 tumor, thereby reducing recurrence rates (20-23).


Chemotherapy

Chemotherapy alone rarely produces durable complete disease remissions for MIBC. Chemotherapy can be used preoperatively (neoadjuvant chemotherapy, NAC) or postoperatively (adjuvant chemotherapy, AC) prior to RC. In patients who undergo bladder-sparing treatments for MIBC, chemotherapy plays a crucial role by increasing the radio-sensitivity of the bladder tumor. Chemotherapy agents used as radiosensitizers for TMT include gemcitabine, mitomycin C, fluorouracil/cisplatin, etc. Survival outcomes are improved with radio-sensitizing chemotherapy prior to RT when compared to RT alone (24). Compared with RT monotherapy, synchronous chemotherapy with mitomycin C and fluorouracil combined with RT has been shown to significantly improve locoregional tumor control, with no significant increase of adverse events (25). The 2 years’ locoregional disease-free survival (DFS) was 67% in the chemoradiation group and 54% in the RT alone group. The chemoradiation combination therapy was well-tolerated, 80% of patients completed chemotherapy and 95% of patients completed RT (25).

Several studies have reported encouraging results of NAC prior to TMT (26,27). Jiang et al. assessed 57 consecutive MIBC who underwent NAC prior to TMT, and the result showed that 2-year OS, disease-specific survival, and bladder-intact DFS were 74%, 88%, and 64%, respectively (26). Hafeez et al. reported that response to NAC is a favorable prognostic factor which can be applied to choose patients for RT, resulting in bladder preservation in more than 80% of selected patients (27). Other studies have investigated adjuvant chemotherapy after TMT, but shown worse completion rates and tolerability compared with NAC, with grade 3–4 toxicity rate as high as 75% (28-30). Therefore, NAC is preferred in TMT.


RT

RT has been used as a treatment for bladder cancer since 1926, but RT alone has been shown to have poorer outcomes when compared to chemoradiotherapy (25). A conventional course of RT usually includes external beam radiotherapy (EBRT) to the bladder and limited pelvic lymph nodes (LNs) with an initial dose of 40–45 Gy, followed by another boost to the whole bladder of 50-54 Gy, and a further tumor boost to a total dose of 60–66 Gy (1). There are 2 RT regimens most commonly used during RT for MIBC: a split course regimen with interval cystoscopy is typically used in the US (18), whereas European centers generally use the continuous course regimen which is now more commonly used (25). If an incomplete response is observed during the split course regimen, patients undergo immediate salvage RC (12).

Controversy exists on the optimal radiation fields, especially on whether treatment should entail partial bladder radiation (tumor) or whole bladder radiation. Partial bladder radiation spares normal tissue with less toxicity, but may miss any occult tumor that has not been diagnosed by cystoscopy or imaging (14). Another controversial topic of RT is the optimal radiation volume to regional LNs. Radiation Therapy Oncology Group (RTOG) protocols in the US include whole pelvic radiation and an additional boost volume to the bladder and gross tumor. Considering the high LNs metastases rate (20–30%) seen in pT2-T3 disease, it seems rational to give radiation to regional LNs (31). Meanwhile, in the BC2001 trial protocol in Europe, radiation was administered to the bladder only, and the results showed that pelvic relapse rate with RT only to the bladder were merely 5.8%, indicating that not all patients need RT to the pelvic LNs (32). Nonetheless, a limitation of this study should be considered as it included a high proportion of low-risk MIBC diseases (84% were T2N0M0) with a low LN metastases rate. Therefore, selective treatment of LNs (when nodes are negative) is optional, and should consider the patient’s comorbidities and the risk of toxicity to adjacent critical structures.


Patient selection

Appropriate patient selection is crucial in achieving good oncologic control with bladder-sparing therapy. The ideal selection criteria for TMT include good bladder function and capacity, clinical stage within cT2, complete TURBT, no prior history of pelvic RT, no extensive CIS, and absence of hydronephrosis (14).

The high-risk factors associated with poor prognosis after TMT include incomplete or inability to achieve maximal TURBT, hydronephrosis, diffuse multifocal tumors, extensive CIS, and cT3-cT4a disease (14). Higher tumor stages appear less responsive to chemotherapy and RT. According to an observational study including 415 MIBC patients treated with bladder-sparing therapy, the response rate decreased as the tumor stage increased (metastases-free survival at 5 years T2, 85%; T3, 63%; and T4, 34%) (19). Multifocal lesions were associated with a greater risk of local recurrence following CR to bladder-sparing treatment. MIBC patients with tumor-related hydronephrosis show reduced CR rates to bladder-sparing therapy and therefore should not be routinely offered (19). Although the above high-risk factors are not absolute contraindications to TMT, the chance of cure is significantly diminished (14).


Oncological outcomes

No prospective randomized trial has successfully compared the oncological outcomes between TMT and RC. A prospective randomized phase III trial to compare TMT versus RC was ultimately closed due to poor recruitment (33).

Several observational studies have compared the oncological outcomes between TMT and RC, with 5-year OS and CSS rates varying between 36% and 74% and 50% and 84%, respectively, and the salvage cystectomy rates have varied between 10% and 30% (25,28,34-36). A meta-analysis of the combined data of 6 RTOG bladder-sparing studies demonstrated 5-year OS and CSS of 57% and 71%, respectively, with salvage cystectomy rates of 21% (37). However, due to the risk of selection bias, misidentification, and misclassification, the clinical significance of these data must be interpreted with caution (11). Clinicopathologic stage discordance and inclusion biases have also limited the validity of direct comparison between TMT and RC (34). Patients treated with TMT were generally older than those who underwent RC, and had more co-morbidities, thus they may have poorer prognosis.


Future perspectives

In recent years, the emergence of immunotherapy has led to a great innovation in the treatment of malignant tumors (38,39). Immune checkpoint inhibition (ICI) provides enduring response in patients with advanced or metastatic bladder cancer (40,41). Preclinical studies have shown that RT can enhance the effect of ICI by enhancing the systemic antitumor immune response by enhancing the cross-presentation of tumor antigens and the upregulation of PD-L1 expression (42). Several comparative randomized phase II trials evaluating the effect of combining ICI with chemoradiotherapy (iCRT) are still ongoing (43-47). The ICI regimens used include anti-programmed death-1 (PD-1; pembrolizumab or nivolumab) or anti-programmed cell death ligand-1 (PD-L1; atezolizumab or durvalumab) monotherapy, or a combination of anti-PD-1 and anti-CTLA-4 (ipilimumab and nivolumab). Almost all these trials are still in the recruiting phase, with early results showing complete remission in 90% of patients, 1-year DFS, metastasis-free survival (MFS), and OS between 77% and 100%, and immune-mediated toxicity seen in about 10% of patients (43-48). The final results of these trials may reveal the real effect of the immunotherapy combined with radiotherapy (iCRT) treatment modality, and the following limitations should be considered (48). Firstly, several TMT schemes are being combined with ICI, but the best scheme for MIBC remains undetermined. Secondly, a standardized response evaluation after iCRT still needs to be defined. Thirdly, there was a selection bias in the above phase I–II trials in which most included patients had a relatively high performance status. However, in clinical practice, most patients who receive bladder-preservation therapy are usually older, less physically fit, and have more comorbidities. Moreover, the ideal biomarkers of treatment response and immune evasion in patients undergoing iCRT therapy remain to be further developed. Finally, the difficulty in comparing outcomes among studies should be considered. Despite preliminary findings are encouraging, harmonization of terminology and definition of clinical endpoints among trials will be mandatory to correctly assess the potential role of CRT and immunotherapy combination as bladder-sparing solution in routine clinical practice (49).

The current selection criteria for TMT are mainly based on patient’s clinical and pathological factors, but novel biomarkers that can predict response after TMT are receiving increasing attention. Several studies have demonstrated that DNA damage response markers, including MRE11 and ERCC1/2, serve as predictors of CSS in 2 independent TMT cohorts (50,51). In patients with low MRE11 expression, RC was associated with significantly longer CSS compared with RT (51).

In ERCC1 immunohistochemistry, 6 out of 8 positive cases had no complete response to chemoradiotherapy, whereas 12 out of 14 negative cases had complete remission, suggesting ERCC1 expression level may predict the efficacy of chemoradiation therapy for MIBC (50). T-cell activation and interferon-γ signaling signatures appear to be associated with improved CSS after TMT, whereas higher stromal infiltration is associated with poorer outcomes after NAC and RC (52). Although the above biomarkers are promising in predicting response after TMT, further validation is still required in prospective trials.


Conclusions

TMT is a well-tolerated and offers a curative alternative approach to RC for selected patients with localized MIBC. Appropriate patient selection and a multi-disciplinary approach is crucial in achieving good oncologic control with bladder-sparing therapy. Novel predictive biomarkers are anticipated for more precise patient selection and better oncological outcomes.


Acknowledgments

Funding: This study was funded by the National Natural Science Foundation of China (No. 82002682).


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tau.amegroups.com/article/view/10.21037/tau-23-124/rc

Peer Review File: Available at https://tau.amegroups.com/article/view/10.21037/tau-23-124/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tau.amegroups.com/article/view/10.21037/tau-23-124/coif). The 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. Cathomas R, Lorch A, Bruins HM, et al. The 2021 Updated European Association of Urology Guidelines on Metastatic Urothelial Carcinoma. Eur Urol 2022;81:95-103. [Crossref] [PubMed]
  2. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209-49. [Crossref] [PubMed]
  3. Bassi P, Ferrante GD, Piazza N, et al. Prognostic factors of outcome after radical cystectomy for bladder cancer: a retrospective study of a homogeneous patient cohort. J Urol 1999;161:1494-7. [Crossref] [PubMed]
  4. Ghoneim MA, el-Mekresh MM, el-Baz MA, et al. Radical cystectomy for carcinoma of the bladder: critical evaluation of the results in 1,026 cases. J Urol 1997;158:393-9. [Crossref] [PubMed]
  5. Stein JP, Lieskovsky G, Cote R, et al. Radical cystectomy in the treatment of invasive bladder cancer: long-term results in 1,054 patients. J Clin Oncol 2001;19:666-75. [Crossref] [PubMed]
  6. Stein JP, Skinner DG. Radical cystectomy for invasive bladder cancer: long-term results of a standard procedure. World J Urol 2006;24:296-304. [Crossref] [PubMed]
  7. Fisher B, Anderson S, Bryant J, et al. Twenty-year follow-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med 2002;347:1233-41. [Crossref] [PubMed]
  8. Landoni F, Colombo A, Milani R, et al. Randomized study between radical surgery and radiotherapy for the treatment of stage IB-IIA cervical cancer: 20-year update. J Gynecol Oncol 2017;28:e34. [Crossref] [PubMed]
  9. Hagen ER, Cleary RK. Organ Preservation in the Treatment of Stage II and III Rectal Cancer. Dis Colon Rectum 2020;63:1185-9. [Crossref] [PubMed]
  10. Williams SB, Shan Y, Jazzar U, et al. Comparing Survival Outcomes and Costs Associated With Radical Cystectomy and Trimodal Therapy for Older Adults With Muscle-Invasive Bladder Cancer. JAMA Surg 2018;153:881-9. [Crossref] [PubMed]
  11. Broughman JR, Vuong W, Mian OY. Current Landscape and Future Directions on Bladder Sparing Approaches to Muscle-Invasive Bladder Cancer. Curr Treat Options Oncol 2020;22:3. [Crossref] [PubMed]
  12. Chang SS, Bochner BH, Chou R, et al. Treatment of Non-Metastatic Muscle-Invasive Bladder Cancer: AUA/ASCO/ASTRO/SUO Guideline. J Urol 2017;198:552-9. [Crossref] [PubMed]
  13. Flaig TW, Spiess PE, Agarwal N, et al. Bladder Cancer, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2020;18:329-54. [Crossref] [PubMed]
  14. Tholomier C, Souhami L, Kassouf W. Bladder-sparing protocols in the treatment of muscle-invasive bladder cancer. Transl Androl Urol 2020;9:2920-37. [Crossref] [PubMed]
  15. Herr HW. Conservative management of muscle-infiltrating bladder cancer: prospective experience. J Urol 1987;138:1162-3. [Crossref] [PubMed]
  16. Solsona E, Iborra I, Collado A, et al. Feasibility of radical transurethral resection as monotherapy for selected patients with muscle invasive bladder cancer. J Urol 2010;184:475-80. [Crossref] [PubMed]
  17. Herr HW. Transurethral resection of muscle-invasive bladder cancer: 10-year outcome. J Clin Oncol 2001;19:89-93. [Crossref] [PubMed]
  18. Efstathiou JA, Spiegel DY, Shipley WU, et al. Long-term outcomes of selective bladder preservation by combined-modality therapy for invasive bladder cancer: the MGH experience. Eur Urol 2012;61:705-11. [Crossref] [PubMed]
  19. Rödel C, Grabenbauer GG, Kühn R, et al. Combined-modality treatment and selective organ preservation in invasive bladder cancer: long-term results. J Clin Oncol 2002;20:3061-71. [Crossref] [PubMed]
  20. Murali-Krishnan S, Pang KH, Greco F, et al. Bladder-sparing treatment in MIBC: where do we stand? Minerva Urol Nefrol 2019;71:101-12. [Crossref] [PubMed]
  21. Inoue K, Fukuhara H, Yamamoto S, et al. Current status of photodynamic technology for urothelial cancer. Cancer Sci 2022;113:392-8. [Crossref] [PubMed]
  22. Abrahimi P, McClure T. Emerging Intraoperative Imaging Technologies in Urologic Oncology. Urol Clin North Am 2022;49:57-63. [Crossref] [PubMed]
  23. Fukuhara H, Yamamoto S, Karashima T, et al. Photodynamic diagnosis and therapy for urothelial carcinoma and prostate cancer: new imaging technology and therapy. Int J Clin Oncol 2021;26:18-25. [Crossref] [PubMed]
  24. Pollack A, Zagars GK, Swanson DA. Muscle-invasive bladder cancer treated with external beam radiotherapy: prognostic factors. Int J Radiat Oncol Biol Phys 1994;30:267-77. [Crossref] [PubMed]
  25. James ND, Hussain SA, Hall E, et al. Radiotherapy with or without chemotherapy in muscle-invasive bladder cancer. N Engl J Med 2012;366:1477-88. [Crossref] [PubMed]
  26. Jiang DM, Jiang H, Chung PWM, et al. Neoadjuvant Chemotherapy Before Bladder-Sparing Chemoradiotherapy in Patients With Nonmetastatic Muscle-Invasive Bladder Cancer. Clin Genitourin Cancer 2019;17:38-45. [Crossref] [PubMed]
  27. Hafeez S, Horwich A, Omar O, et al. Selective organ preservation with neo-adjuvant chemotherapy for the treatment of muscle invasive transitional cell carcinoma of the bladder. Br J Cancer 2015;112:1626-35. [Crossref] [PubMed]
  28. Giacalone NJ, Shipley WU, Clayman RH, et al. Long-term Outcomes After Bladder-preserving Tri-modality Therapy for Patients with Muscle-invasive Bladder Cancer: An Updated Analysis of the Massachusetts General Hospital Experience. Eur Urol 2017;71:952-60. [Crossref] [PubMed]
  29. Mitin T, Hunt D, Shipley WU, et al. Transurethral surgery and twice-daily radiation plus paclitaxel-cisplatin or fluorouracil-cisplatin with selective bladder preservation and adjuvant chemotherapy for patients with muscle invasive bladder cancer (RTOG 0233): a randomised multicentre phase 2 trial. Lancet Oncol 2013;14:863-72. [Crossref] [PubMed]
  30. Kaufman DS, Winter KA, Shipley WU, et al. Phase I-II RTOG study (99-06) of patients with muscle-invasive bladder cancer undergoing transurethral surgery, paclitaxel, cisplatin, and twice-daily radiotherapy followed by selective bladder preservation or radical cystectomy and adjuvant chemotherapy. Urology 2009;73:833-7. [Crossref] [PubMed]
  31. Goldsmith B, Baumann BC, He J, et al. Occult pelvic lymph node involvement in bladder cancer: implications for definitive radiation. Int J Radiat Oncol Biol Phys 2014;88:603-10. [Crossref] [PubMed]
  32. Tunio MA, Hashmi A, Qayyum A, et al. Whole-pelvis or bladder-only chemoradiation for lymph node-negative invasive bladder cancer: single-institution experience. Int J Radiat Oncol Biol Phys 2012;82:e457-62. [Crossref] [PubMed]
  33. Huddart RA, Hall E, Lewis R, et al. Life and death of spare (selective bladder preservation against radical excision): reflections on why the spare trial closed. BJU Int 2010;106:753-5. [Crossref] [PubMed]
  34. Ploussard G, Daneshmand S, Efstathiou JA, et al. Critical analysis of bladder sparing with trimodal therapy in muscle-invasive bladder cancer: a systematic review. Eur Urol 2014;66:120-37. [Crossref] [PubMed]
  35. Hoskin PJ, Rojas AM, Bentzen SM, et al. Radiotherapy with concurrent carbogen and nicotinamide in bladder carcinoma. J Clin Oncol 2010;28:4912-8. [Crossref] [PubMed]
  36. Kulkarni GS, Hermanns T, Wei Y, et al. Propensity Score Analysis of Radical Cystectomy Versus Bladder-Sparing Trimodal Therapy in the Setting of a Multidisciplinary Bladder Cancer Clinic. J Clin Oncol 2017;35:2299-305. [Crossref] [PubMed]
  37. Mak RH, Hunt D, Shipley WU, et al. Long-term outcomes in patients with muscle-invasive bladder cancer after selective bladder-preserving combined-modality therapy: a pooled analysis of Radiation Therapy Oncology Group protocols 8802, 8903, 9506, 9706, 9906, and 0233. J Clin Oncol 2014;32:3801-9. [Crossref] [PubMed]
  38. Larkin HD. Finding Ways to Improve Patients' Cancer Immunotherapy Response. JAMA 2022;328:518. [Crossref] [PubMed]
  39. Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711-23. [Crossref] [PubMed]
  40. Powles T, Park SH, Voog E, et al. Avelumab Maintenance Therapy for Advanced or Metastatic Urothelial Carcinoma. N Engl J Med 2020;383:1218-30. [Crossref] [PubMed]
  41. Balar AV, Castellano D, O'Donnell PH, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol 2017;18:1483-92. [Crossref] [PubMed]
  42. Sharabi AB, Nirschl CJ, Kochel CM, et al. Stereotactic Radiation Therapy Augments Antigen-Specific PD-1-Mediated Antitumor Immune Responses via Cross-Presentation of Tumor Antigen. Cancer Immunol Res 2015;3:345-55. [Crossref] [PubMed]
  43. Marcq G, Souhami L, Cury FL, et al. Phase 1 Trial of Atezolizumab Plus Trimodal Therapy in Patients With Localized Muscle-Invasive Bladder Cancer. Int J Radiat Oncol Biol Phys 2021;110:738-41. [Crossref] [PubMed]
  44. Balar AV, Milowsky MI, O’Donnell PH, et al. Pembrolizumab (pembro) in combination with gemcitabine (Gem) and concurrent hypofractionated radiation therapy (RT) as bladder sparing treatment for muscle-invasive urothelial cancer of the bladder (MIBC): A multicenter phase 2 trial. J Clin Oncol 2021;39:abstr 4504.
  45. Weickhardt AJ, Foroudi F, Lawrentschuk N, et al. Pembrolizumab with chemoradiotherapy as treatment for muscle invasive bladder cancer: A planned interim analysis of safety and efficacy of the PCR-MIB phase II clinical trial (ANZUP 1502). J Clin Oncol 2020;38:abstr 485.
  46. Gupta S, Maughan BL, Dechet CB, et al. NEXT: A phase II, open-label study of nivolumab adjuvant to chemoradiation in patients (pts) with localized muscle invasive bladder cancer. J Clin Oncol 2020;38:abstr TPS605.
  47. Balar AV, James ND, Shariat SF, et al. Phase III study of pembrolizumab (pembro) plus chemoradiotherapy (CRT) versus CRT alone for patients (pts) with muscle-invasive bladder cancer (MIBC): KEYNOTE-992. J Clin Oncol 2020;38:abstr TPS5093.
  48. van Hattum JW, de Ruiter BM, Oddens JR, et al. Bladder-Sparing Chemoradiotherapy Combined with Immune Checkpoint Inhibition for Locally Advanced Urothelial Bladder Cancer-A Review. Cancers (Basel) 2021;14:38. [Crossref] [PubMed]
  49. Basile G, Bandini M, Raggi D, et al. Bladder-sparing combination treatments for muscle-invasive bladder cancer: A plea for standardized assessment and definition of clinical trials endpoints. Urol Oncol 2022;40:37-44. [Crossref] [PubMed]
  50. Kawashima A, Nakayama M, Kakuta Y, et al. Excision repair cross-complementing group 1 may predict the efficacy of chemoradiation therapy for muscle-invasive bladder cancer. Clin Cancer Res 2011;17:2561-9. [Crossref] [PubMed]
  51. Choudhury A, Nelson LD, Teo MT, et al. MRE11 expression is predictive of cause-specific survival following radical radiotherapy for muscle-invasive bladder cancer. Cancer Res 2010;70:7017-26. [Crossref] [PubMed]
  52. Efstathiou JA, Mouw KW, Gibb EA, et al. Impact of Immune and Stromal Infiltration on Outcomes Following Bladder-Sparing Trimodality Therapy for Muscle-Invasive Bladder Cancer. Eur Urol 2019;76:59-68. [Crossref] [PubMed]

(English Language Editor: J. Jones)

Cite this article as: Fan X, He W, Huang J. Bladder-sparing approaches for muscle invasive bladder cancer: a narrative review of current evidence and future perspectives. Transl Androl Urol 2023;12(5):802-808. doi: 10.21037/tau-23-124

Download Citation