Trimodality therapy in the modern era for management of bladder cancer
Editorial Commentary

Trimodality therapy in the modern era for management of bladder cancer

Shang-Jui Wang

Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA

Correspondence to: Shang-Jui Wang, MD, PhD. Department of Radiation Oncology, The Ohio State University Wexner Medical Center, 410 W 10th Ave., Columbus, OH 43210, USA. Email: Shang-Jui.Wang@osumc.edu.

Keywords: Trimodality therapy (TMT); chemoradiation; bladder cancer; muscle-invasive bladder cancer (MIBC); bladder preservation


Submitted Feb 19, 2024. Accepted for publication Jun 27, 2024. Published online Aug 20, 2024.

doi: 10.21037/tcr-24-262


Treatment decision for muscle-invasive bladder cancer (MIBC) often poses a challenge for physicians and patients. Radical cystectomy (RC) with neoadjuvant chemotherapy remains to be the most widely used curative treatment for MIBC (1,2). However, RC is a major surgery and poses substantial risk of complications and perioperative mortality, especially in older patients with significant comorbidities (3). Bladder-preserving trimodality therapy (TMT) with maximally safe transurethral resection of bladder tumor (TURBT) followed by chemoradiotherapy (CRT), on the other hand, is an effective and less-invasive alternative to RC. Despite this, it is estimated that only ~10–20% of patients receive TMT for definitive treatment of MIBC (4-6). The low rates of TMT utilization are likely multifactorial, which includes the lack of prospective randomized trials to demonstrate equipoise between TMT and RC, misconceptions regarding TMT, and inexperience with this treatment modality. This commentary aims to narrate the establishment of TMT as an effective MIBC therapy and the aspiring role of TMT in the modern era.

The first prospective trial in the 1980s provided key insights into the combined efficacy of CRT for MIBC—improved locoregional control of radiotherapy (RT) with concurrent cisplatin (7). This paved way to several small RTOG trials that evaluated CRT with concurrent cisplatin backbone (8-13). Pooled analyses of the RTOG trials with a total of 468 patients demonstrated clinical complete response (cCR) rate of 69%; 5-year and 10-year overall survival (OS) rates were 57% and 36%, respectively, and 5-year and 10-year disease-specific survival (DSS) rates were 71% and 65%, respectively. Of note, non-muscle-invasive local recurrences post-CRT (31% at 5-year) were more common than muscle-invasive recurrences (13% at 5 years) (14). In addition, two large single-institutional experiences with TMT demonstrated comparable efficacy of bladder preservation therapy to prospective trials. One institutional series of 348 MIBC patients treated with TMT demonstrated 10-year OS and DSS rates of 35% and 59%, respectively, while another series of 415 patients showed 10-year DSS rates of 42% (15,16).

Still, the therapeutic benefit of concurrent chemotherapy with RT needed validation with large phase 3 randomized trial. To this goal, BC2001 trial randomized 360 MIBC patients to RT alone vs. CRT with concurrent mitomycin/5-FU. At 10-year follow-up, CRT improved locoregional control [hazard ratio (HR) =0.61; 95% confidence interval (CI): 0.43–0.86; P=0.004] and invasive locoregional control (HR =0.55; 95% CI: 0.36–0.84; P=0.006) compared to RT alone, which translated to non-statistically-significant improvement in DFS (HR =0.78; 95% CI: 0.60–1.02; P=0.069), metastasis-free survival (HR =0.78; 95% CI: 0.58–1.05; P=0.089), and OS (HR =0.88; 95% CI: 0.69–1.13; P=0.3). Patients who received CRT also had lower rates of needing salvage cystectomy at 5-year compared to RT alone (14% vs. 22%) (17,18). Regarding concurrent chemotherapy options, BC2001 also established the efficacy of mitomycin/5-FU as an appropriate regimen in TMT, especially one with less renal toxicity. Further expanding the radiosensitizer repertoire, RTOG 0712 demonstrated that concurrent gemcitabine is yet another acceptable regimen compared to cisplatin-based chemotherapy (19).

RT can be delivered in conventional fractionation (64–64.8 Gy in 1.8–2.0 Gy per fraction) or with hypofractionation (55 Gy in 2.75 Gy per fraction). BC2001 and BCON (another UK bladder trial that randomized patients to RT ± hypoxic sensitizers carbogen/nicotinamide) trials allowed for both fractionations in a non-randomized fashion (17,20). Interestingly, a recent meta-analysis of the two trials showed that patients who received hypofractionation with 55 Gy in 20 fractions was associated with lower risk of invasive locoregional recurrence compared to those that received 64 Gy in 32 fractions (HR =0.71; 95% CI: 0.52–0.96), while both fractionations demonstrated similar toxicity (21).

It is interesting to note that, in the United States, earlier trials and historical paradigm of bladder preservation was done in a split-course fashion, which entails induction CRT to ~40–45 Gy followed by cystoscopic evaluation, and only patients who have achieved cCR will then proceed to consolidative CRT with additional ~20–25 Gy (5). On the other hand, treatment paradigm in Europe favored continuous CRT course, which is followed by cystoscopic surveillance to monitor for local recurrence. Salvage cystectomy is indicated if cCR was not achieved in split-course protocol (early salvage) or at the time of local recurrence with either split-course or continuous regimens (delayed salvage). While unproven, the rationale for split-course is to allow earlier selection of patients with unfavorable disease characteristics (i.e., those who did not achieved complete TURBT prior to induction CRT or those with disease that are less responsive to CRT) for early salvage cystectomy in hopes that this may improve outcome. In contrast, continuous CRT allows for full delivery of treatment to maximize tumor eradication and chance for bladder preservation. Post-operative complication rates from early vs. delayed salvage cystectomy appear to be comparable, although with greater proportion of cardiovascular complications associated with early salvage and more tissue healing complications with delayed salvage (22). In more recent years, paradigm with TMT has largely shifted towards continuous CRT regimens.

Despite having both prospective and retrospective evidence demonstrating favorable efficacy and outcome of TMT, this modality remains to be under-utilized in the real-world setting. One major hurdle is the lack of prospective data comparing TMT to RC, which has long been a standard-of-care treatment for MIBC. The SPARE trial was the only prospective trial randomizing MIBC patients to TMT vs. RC, but trial terminated after only enrollment of 45 patients due to poor accrual, which underscores the inherent challenges of comparing two treatment modalities that do not hold equipoise from the patients’ perspective (most patients would strongly favor one modality over another) (23). Given that there will likely never be a successful randomized control trial comparing TMT to cystectomy for MIBC, we must rely on real-world data on outcomes of TMT vs. surgery to address the comparability of the two modalities. Zlotta et al. recently reported a large multi-institutional retrospective study of 722 patients (n=440 underwent cystectomy, n=282 received TMT) with cT2–T4N0 MIBC who would have been eligible for either treatment modalities (i.e., solitary tumors <7 cm, no bilateral hydronephrosis, or no extensive carcinoma in situ). Patient outcomes by treatment modality were analyzed with propensity score matching (PSM) and inverse probability treatment weighting (IPTW). The study reported no significant differences in 5-year metastasis-free survival between TMT vs. cystectomy with either PSM (74% vs. 74%) or IPTW (75% vs. 74%). There were also no differences in DFS or cancer-specific survival between TMT vs. cystectomy, but interestingly, OS favored TMT. Because the PSM and IPTW analyses accounted for the use of peritreatment chemotherapy (which is best standard of care for patients undergoing cystectomy, but not for TMT), the study also performed a sensitivity analysis of comparing cystectomy patients who received chemotherapy vs. patients who received TMT—again, no differences in survival outcomes were found (24). Similarly, non-metastatic MIBC patients identified from The Netherlands Cancer Registry who received RC (n=1,101) vs. TMT (n=331), with median follow-up of 39 months, demonstrated comparable outcomes between the two treatment cohorts with 2-year DFS of 61.5% vs. 55.3% and 2-year OS of 74% vs. 66% (25). For clinically node-positive MIBC, Swinton et al. analyzed 163 cN+ patients who received definitive treatment with either RT (n=87) or RC (n=76) and showed that there was also no difference in OS or progression-free survival (PFS) for the entire cohort (OS: HR =0.94, 95% CI: 0.63–1.41, P=0.76; PFS: HR =0.74, 95% CI: 0.5–1.08, P=0.12), as well as no difference in outcomes in the subset of RT/RC patients that received chemotherapy (26). These studies, while limited by their retrospective nature, represent the most robust evidence to-date to support bladder-sparing TMT as an appropriate treatment option for the majority of MIBC patients with localized or locoregional disease.

Acute side effects of CRT can include transient symptoms of fatigue, cystitis, enteritis and hematological toxicities from concurrent chemotherapy. In some patients, the acute bowel and urinary effects can linger, constituting chronic bowel (diarrhea or looser stool) and urinary (nocturia or increased frequency/urgency) habit changes. Other potential late side effects of CRT can include radiation proctitis, radiation cystitis, and rare instances of small bowel obstruction or contracted bladder requiring surgical intervention. The Erlengen experience reported that 1.6% patients had salvage cystectomy due to having contracted bladder and 1.5% patients with bowel obstruction requiring surgery (16); it is important to note that these patients were treated from 1982–2000 and that modern radiation techniques would likely be associated with even lower rates of these major complications. In more modern prospective trials, BC2001 and BCON reported ~10–20% of grade 3/4 late toxicity with RT or CRT (18,21). Comparing the late toxicity of CRT to RC, the SPARE trial reported more patients who underwent RC (52%) with grade 3/4 toxicity compared to those who received CRT (27%) when excluding erectile dysfunction (23).

Quality-of-life (QoL) after MIBC treatments is an important consideration for patients. In the SPARE trial (randomizing TMT vs. RC) that failed to accrue, many patients who declined randomization selected TMT as their treatment of choice, which underscores the priority for many patients to strive for organ preservation. Moreover, from the limited number of accrued patients, the SPARE trial also showed that there was a greater decline in body image and male sexual function after RC compared to TMT (23). In a recent meta-analysis that strived to examine the QoL after definitive treatment for MIBC, 50 studies were included, although most studies evaluated surgical QoL outcomes. However, the strength of the study lies in pooling the “common denominator” of patient-reported QoL surveys across 18 studies—the European Organization for Research and Treatment of Cancer Core Quality of Life questionnaire-C30 (EORTC QLQ-C30). Although only one of the 18 studies in the pooled analysis evaluated QoL after TMT (without direct comparison with RC), this TMT study showed a higher mean reported values for Global Health Score and Physical Functioning compared to surgical studies, with non-overlapping CIs between TMT and surgical approaches (27). Nevertheless, in the absence of large prospective randomized trials, head-to-head comparison of QoL after TMT vs. RC would not be possible, and this debate shall continue. However, for many patients, the QoL implications and preference for bladder preservation is indisputable. Therefore, given the comparable survival outcomes between TMT and RC as discussed earlier, TMT should be offered to patients who desire bladder preservation in the appropriate clinical settings.

Management of MIBC with variant histologies (other than pure urothelial carcinoma) often poses a dilemma regarding the appropriateness of TMT. This is summarized in a recent review by Brocklehurst et al. (28). In brief, patients with urothelial carcinoma with divergent differentiation (such as squamous differentiation or sarcomatoid features) treated with TMT was found to have comparable outcomes to those with pure urothelial carcinoma in a single-center retrospective study of 303 patients (29). On the other hand, non-urothelial carcinoma histologies (such as squamous cell or adenocarcinoma) are rare and currently lacks robust data to support the use of TMT. The exception to this is small cell carcinoma of the bladder, which follows a similar treatment paradigm as small cell lung cancer, with standard therapy consisting of neoadjuvant platinum/etoposide chemotherapy followed by local therapy with TMT or RC (30).

In conclusion, TMT is an appropriate and effective treatment for MIBC and should be viewed as a comparable alternative to RC in patients who favor organ preservation. The lack of equipoise between TMT and RC would likely remain for some physicians, citing the absence of level I evidence from randomized clinical trials to support the adoption of TMT as an equal alternative to RC. However, the impracticality of randomizing patients between TMT vs. RC should not deter the ample of real-world evidence that demonstrates favorable therapeutic and QoL outcomes of TMT. It is time that we push TMT into the mainstream and broaden the therapy repertoire for MIBC.


Acknowledgments

Funding: None.


Footnote

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

Peer Review File: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-262/prf

Conflicts of Interest: The author has completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-262/coif). The author has no conflicts of interest to declare.

Ethical Statement: The author is 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. Kamat AM, Hahn NM, Efstathiou JA, et al. Bladder cancer. Lancet 2016;388:2796-810. [Crossref] [PubMed]
  2. Compérat E, Amin MB, Cathomas R, et al. Current best practice for bladder cancer: a narrative review of diagnostics and treatments. Lancet 2022;400:1712-21. [Crossref] [PubMed]
  3. Parekh DJ, Reis IM, Castle EP, et al. Robot-assisted radical cystectomy versus open radical cystectomy in patients with bladder cancer (RAZOR): an open-label, randomised, phase 3, non-inferiority trial. Lancet 2018;391:2525-36. [Crossref] [PubMed]
  4. Fahmy O, Khairul-Asri MG, Schubert T, et al. A systematic review and meta-analysis on the oncological long-term outcomes after trimodality therapy and radical cystectomy with or without neoadjuvant chemotherapy for muscle-invasive bladder cancer. Urol Oncol 2018;36:43-53. [Crossref] [PubMed]
  5. 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]
  6. Vashistha V, Wang H, Mazzone A, et al. Radical Cystectomy Compared to Combined Modality Treatment for Muscle-Invasive Bladder Cancer: A Systematic Review and Meta-Analysis. Int J Radiat Oncol Biol Phys 2017;97:1002-20. [Crossref] [PubMed]
  7. Coppin CM, Gospodarowicz MK, James K, et al. Improved local control of invasive bladder cancer by concurrent cisplatin and preoperative or definitive radiation. The National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 1996;14:2901-7. [Crossref] [PubMed]
  8. Tester W, Caplan R, Heaney J, et al. Neoadjuvant combined modality program with selective organ preservation for invasive bladder cancer: results of Radiation Therapy Oncology Group phase II trial 8802. J Clin Oncol 1996;14:119-26. [Crossref] [PubMed]
  9. Shipley WU, Winter KA, Kaufman DS, et al. Phase III trial of neoadjuvant chemotherapy in patients with invasive bladder cancer treated with selective bladder preservation by combined radiation therapy and chemotherapy: initial results of Radiation Therapy Oncology Group 89-03. J Clin Oncol 1998;16:3576-83. [Crossref] [PubMed]
  10. Kaufman DS, Winter KA, Shipley WU, et al. The initial results in muscle-invading bladder cancer of RTOG 95-06: phase I/II trial of transurethral surgery plus radiation therapy with concurrent cisplatin and 5-fluorouracil followed by selective bladder preservation or cystectomy depending on the initial response. Oncologist 2000;5:471-6. [Crossref] [PubMed]
  11. Hagan MP, Winter KA, Kaufman DS, et al. RTOG 97-06: initial report of a phase I-II trial of selective bladder conservation using TURBT, twice-daily accelerated irradiation sensitized with cisplatin, and adjuvant MCV combination chemotherapy. Int J Radiat Oncol Biol Phys 2003;57:665-72. [Crossref] [PubMed]
  12. 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]
  13. 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]
  14. 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]
  15. 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]
  16. 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]
  17. 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]
  18. Hall E, Hussain SA, Porta N, et al. Chemoradiotherapy in Muscle-invasive Bladder Cancer: 10-yr Follow-up of the Phase 3 Randomised Controlled BC2001 Trial. Eur Urol 2022;82:273-9. [Crossref] [PubMed]
  19. Coen JJ, Zhang P, Saylor PJ, et al. Bladder Preservation With Twice-a-Day Radiation Plus Fluorouracil/Cisplatin or Once Daily Radiation Plus Gemcitabine for Muscle-Invasive Bladder Cancer: NRG/RTOG 0712-A Randomized Phase II Trial. J Clin Oncol 2019;37:44-51. [Crossref] [PubMed]
  20. Song YP, Mistry H, Irlam J, et al. Long-Term Outcomes of Radical Radiation Therapy with Hypoxia Modification with Biomarker Discovery for Stratification: 10-Year Update of the BCON (Bladder Carbogen Nicotinamide) Phase 3 Randomized Trial (ISRCTN45938399). Int J Radiat Oncol Biol Phys 2021;110:1407-15. [Crossref] [PubMed]
  21. Choudhury A, Porta N, Hall E, et al. Hypofractionated radiotherapy in locally advanced bladder cancer: an individual patient data meta-analysis of the BC2001 and BCON trials. Lancet Oncol 2021;22:246-55. [Crossref] [PubMed]
  22. Eswara JR, Efstathiou JA, Heney NM, et al. Complications and long-term results of salvage cystectomy after failed bladder sparing therapy for muscle invasive bladder cancer. J Urol 2012;187:463-8. [Crossref] [PubMed]
  23. Huddart RA, Birtle A, Maynard L, et al. Clinical and patient-reported outcomes of SPARE - a randomised feasibility study of selective bladder preservation versus radical cystectomy. BJU Int 2017;120:639-50. [Crossref] [PubMed]
  24. Zlotta AR, Ballas LK, Niemierko A, et al. Radical cystectomy versus trimodality therapy for muscle-invasive bladder cancer: a multi-institutional propensity score matched and weighted analysis. Lancet Oncol 2023;24:669-81. [Crossref] [PubMed]
  25. Brück K, Meijer RP, Boormans JL, et al. Disease-Free Survival of Patients With Muscle-Invasive Bladder Cancer Treated With Radical Cystectomy Versus Bladder-Preserving Therapy: A Nationwide Study. Int J Radiat Oncol Biol Phys 2024;118:41-9. [Crossref] [PubMed]
  26. Swinton M, Mariam NBG, Tan JL, et al. Bladder-Sparing Treatment With Radical Dose Radiotherapy Is an Effective Alternative to Radical Cystectomy in Patients With Clinically Node-Positive Nonmetastatic Bladder Cancer. J Clin Oncol 2023;41:4406-15. [Crossref] [PubMed]
  27. Francolini G, Ghoshal A, Caini S, et al. Quality of life after definitive treatment for bladder cancer: A systematic review and meta-analysis. Radiother Oncol 2024;190:110038. [Crossref] [PubMed]
  28. Brocklehurst A, Varughese M, Birtle A. Bladder Preservation for Muscle-Invasive Bladder Cancer With Variant Histology. Semin Radiat Oncol 2023;33:62-9. [Crossref] [PubMed]
  29. Krasnow RE, Drumm M, Roberts HJ, et al. Clinical Outcomes of Patients with Histologic Variants of Urothelial Cancer Treated with Trimodality Bladder-sparing Therapy. Eur Urol 2017;72:54-60. [Crossref] [PubMed]
  30. Holzbeierlein J, Bixler BR, Buckley DI, et al. Treatment of Non-Metastatic Muscle-Invasive Bladder Cancer: AUA/ASCO/SUO Guideline (2017; Amended 2020, 2024). J Urol 2024;212:3-10. [Crossref] [PubMed]
Cite this article as: Wang SJ. Trimodality therapy in the modern era for management of bladder cancer. Transl Cancer Res 2024;13(8):3935-3939. doi: 10.21037/tcr-24-262

Download Citation