1,140
Views
1
CrossRef citations to date
0
Altmetric
Articles

A prospective multicenter study of visual response-evaluation by cystoscopy in patients undergoing neoadjuvant chemotherapy for muscle invasive urinary bladder cancer

, , , , , , , , , & ORCID Icon show all
Pages 20-26 | Received 19 Jul 2021, Accepted 11 Oct 2021, Published online: 21 Nov 2021

Abstract

Purpose

To evaluate a method of transurethral visual response-staging in patients with urothelial muscle-invasive urinary bladder cancer (MIBC), undergoing neoadjuvant chemotherapy (NAC) and radical cystectomy (RC).

Methods

A prospective study at four Swedish cystectomy centers, cystoscopy was performed after final NAC-cycle for MIBC. Fifty-six participants underwent cystoscopy for visual staging of the tumor immediately pre-RC. Visual assessments were correlated to pathoanatomical outcomes post-RC.

Results

Seventeen tumors were classified as complete response (CR), i.e. pT0. Twenty-five patients had residual MIBC and 14 had non-muscle invasive residual tumors (NMIBC). Of the 39 patients with residual tumor, 25 were correctly identified visually (64%). Eleven patients were pN+. The diagnostic accuracy of cystoscopy to correctly identify complete response or remaining tumor was 70% (CI = 56–81%) with a sensitivity of 64% (CI = 47–79%), specificity 82% (CI = 57–96%), PPV 89% (CI = 74–96%) and NPV 50% (CI =38–61%). Twenty-eight cystoscopy evaluations showed signs of residual tumors and 3/28 (11%) were false positive. In 4/14 patients assessed having residual NMIBC the estimates were correct, 8/14 had histopathological MIBC and 2/14 had CR. In 11/14 patients (79%), the suggested visual assessment of MIBC was correct, 2/14 had NMIBC and 1/14 had CR. Twenty-eight cystoscopies had negative findings, 14 were false negatives (50%), when cystoscopy falsely predicted pT0. Among them there were eight patients with pTa, pT1 or pTis and six MIBC-tumors. In 17 patients with histopathological pT0, 14 were correctly identified with cystoscopy (82%).

Conclusion

Cystoscopy after the final NAC-cycle cannot robustly differentiate between NAC-responders and non-responders. Visually, negative MIBC-status cannot be determined safely.

Introduction

Bladder cancer is the ninth most common cancer worldwide and ranks 13th in terms of deaths, with smoking as its major risk factor [Citation1,Citation2]. In approximately one-quarter of the patients the cancer is muscle-invasive [Citation3]. The most recent Swedish bladder cancer statistics display an increased incidence but yet improved survival and a stable mortality [Citation4]. The treatment alternatives today depend on the tumor staging, clinical staging, patient age and comorbidity and vary from transurethral resection (TURb) only, radical surgery to local or systemic chemotherapy or a combination of them all [Citation3]. Local control with chemotherapy instillations, immunotherapy with BCG-instillations and repeat TURb are some of the treatment recommendations for non-muscle invasive bladder cancer. The gold standard treatment for muscle-invasive bladder cancer is radical cystectomy (RC) preceded by neoadjuvant chemotherapy (NAC) in fit and eligible patients [Citation3].

The prognosis of urothelial muscle-invasive bladder cancer (MIBC) is relatively poor with a 5-year survival around 50% for all clinical stages T2–T4 following RC [Citation3]. Cisplatin-based neoadjuvant combination NAC significantly increases survival for a chemo-sensitive sub-group of these patients, with an absolute risk reduction (ARR) of 31% for death in completely downstaged patients (pT0N0M0) at 5 years median observation time, compared to cystectomy only patients with complete downstaging [Citation5]. In the original randomized prospective Nordic intention-to-treat-studies, the increased overall survival (OS) in the experimental arm was 8% compared to the control arm [Citation6] and, in a systematic review and meta-analysis of ten randomized trials (n = 2,688), the OS was increased by 5% at median observation time of 5 years [Citation7].

In the last 10 years, since the national introduction of NAC in Sweden, an increasing frequency of hospitals in Sweden have started to implement NAC for MIBC-patients. The most common alternatives for chemotherapy are either a combination of methotrexate, vinblastine, doxorubicin and cisplatin (MVAC) given in three-to-four cycles or gemcitabine and cisplatin/carboplatin (GC) for a total of three-to-four cycles [Citation8]. Inclusion criteria for NAC, according to the Swedish guidelines, are biological age ≤ 75 years and intact renal function with GFR > 55–60 plus an acceptable comorbidity. For unfit patients, the standard recommendation is traditional cystectomy as radical treatment, if fit for major surgery. MIBC-patients unfit for any radical surgery are instead offered radical full dose radiation. A small percentage of MIBC-patients who, for medical reasons, are unfit for both surgery or radiation are offered other local options for symptomatic treatment and in some instances a combined oncological approach.

For non-responders to NAC, there will be a cystectomy-delay, in which the patients risk receiving largely ineffective treatment for 2.5–3 months prior to radical surgery. Retrospective studies evaluating the outcomes of NAC in regard of residual bladder cancer have reconfirmed that patients with downstaged tumors following NAC and RC have significantly improved survival projections compared to non-responders [Citation9–11]. These findings confirm the results of the post hoc analysis of the Nordic combined randomized prospective trials [Citation5]. Yet, the urological community still lacks solid and reliable markers and methods to early identify complete responders to NAC. Early and robust identification of responders and non-responders, respectively, could enable alternative treatment options for the latter group at an early point of time, thus, saving non-responders from a treatment with low efficacy and further avoiding the risks of side effects and complications to NAC [Citation12]. Different research groups have therefore attempted to establish methods to predict the effects of NAC, in differentiating responders versus non-responders as early as possible in the clinical process, but yet without robust and validated results translating into clinical practice or recommendations [Citation13–16]. A recently published study evaluated biomarkers from the two randomized prospective cystectomy trials, NCS 1 & 2 [Citation6]. Utilizing immunohistochemistry on pretreatment tumor specimens and clinical data in 250 patients, the study focused on four biomarkers; CCTα, emmprin, survivin, and BCL-2. In conclusion, only CCTα provided some predictive information for NAC-response, which was improved by adding one of the other three markers, preferably emmprin. Yet, due to insufficient statistical power the suggested markers cannot be translated yet to clinical practice, and the study warrants additional trials and further validation [Citation17].

For cystoscopic visual staging of newly diagnosed bladder tumors, accuracy of the method showed, in a single center retrospective study from the UK in 2016, that assessment predictions of MIBC had a sensitivity of 88.9% and a specificity of 91.0%, resulting in a positive predictive value of 78.4% and a negative predictive value of 95.7%. Thus, visual staging is an option up for evaluation [Citation18]. The value of visual staging in newly diagnosed bladder tumors was also shown in a prospective double-blind clinical study from 2017. In 224 enrolled patients, NMIBC and MIBC were predicted accurately in 93.4% and 85.2%, respectively [Citation19]. Yet so far there is only one group that has published a study on visual staging per se, pre-RC and post-NAC, performed by cystoscopic examination following the second cycle of NAC [Citation20]. The study from Mansour et al. [Citation20] showed an OR of 7.79, in the multivariate analysis, in favor of visual staging as a predictor of downstaging. In the study, only 16% of the patients had received the chemotherapy combination MVAC and 84% had received cisplatin-gemcitabine as NAC-treatment. The analysis was performed in a retrospective manner on prospectively collected data. Cystoscopic images were saved from the initial TURb, and later also after the second cycle of NAC. In addition, bimanual palpation was performed in conjunction with the cystoscopies [Citation20]. In a recently published study evaluating clinical restaging and tumor sequencing, 114 patients underwent restaging TUR following NAC prior to RC. In total, 53% had no evidence of disease (pT0) on post-NAC TUR, yet, from these, only 47% were pT0 on final RC pathology. Among patients with no visual evidence of residual disease on white light cystoscopy (n = 88), TUR was performed in 21. Only 81% (n = 17) in that sub-cohort were true pT0. In the seventeen patients, 41% had residual tumor on post-RC pathology, including four with pT2 tumors. Thus, the study highlights the dilemma of using clinical staging for evaluation of NAC-responses in MIBC [Citation21].

We intended to evaluate the concept of visual staging as a method for response evaluation to NAC-therapy, in a prospective cohort of MIBC-patients planned for radical cystectomy. The primary aim was to calculate the diagnostic accuracy of cystoscopy to find recurrent tumor and a secondary aim was to assess if visual staging could differentiate between MIBC and NMIBC.

Materials and methods

Design and patients

The study was designed as a prospective pilot investigation.

From May 2015 until April 2021, 56 patients at seven Swedish urological centers were prospectively enrolled, primarily from time of TURb: Norrlands University Hospital, Skellefteå county Hospital, Sundsvall/Härnösand county Hospital, Nyköping county Hospital, Uppsala University Hospital, Norrköping county Hospital and Linköping University Hospital and further cystectomized at four of the mentioned centers. Inclusion criteria were patients with urothelial muscle-invasive cancer, staged cT2-T4aN0M0, receiving NAC prior to RC and giving formal consent to participate. Exclusion criteria were patients with urothelial muscle invasive cancer undergoing RC without NAC or patients undergoing NAC but unwilling to participate.

The mean age was 67 years and the median age 70 years (range = 39–80) at the time of diagnosis. Forty-two males and 14 females were included (). The patients were also included in a larger prospective translational study investigating tumor immunology and tumor biological mechanisms, with focus on primary tumor, sentinel node detection, draining and non-draining regional lymph nodes [Citation22–28].

Table 1. Patient characteristics, staging after TUR-b and cystectomy and details on administered neoadjuvant chemotherapy (NAC).

All included patients had undergone standard clinical practices with an initial transurethral resection of the bladder tumor for primary staging plus CT-evaluations with contrast enhancement.

The TURB-Ts of all included patients were considered as being macroscopically radical and were performed by senior subspecialized bladder cancer urologists in our national research team. After the final NAC-cycle, just prior to the cystectomy, all patients were evaluated with cystoscopy in the OR by a senior specialist in urology at each participating center. The evaluator was always subspecialized in surgical treatment of urinary bladder cancer and a member of the local bladder cancer/cystectomy-team of the center performing RC. The urologists in the national research group for this study have together standardized the evaluation criteria. Following the pre-RC cystoscopy, the urologist evaluator filled a case report form (CRF) indicating if visual residual cancer could be seen or not. If residual cancer was seen, the urologist estimated if the cancer was likely to be muscle invasive (MIBC) or not (NMIBC). Bimanual palpation, considered as being a confounding factor for the visual assessment, was intentionally excluded from the investigation.

Chemotherapy, surgical technique and histopathology

Most of the patients (86%) received MVAC-HD and mainly three cycles of NAC were delivered according to local and regional oncological routines. In Sweden, MVAC-HD is the most preferred treatment combination for NAC by the medical oncologists and cisplatin-gemcitabine is the first choice in only one national center (). All patients underwent radical cystectomy with sentinel node dissection and with the intended extent of including the following stations: Bilateral Obturator fossae, External Iliac artery bilaterally and Common Iliac arteries up to mid-level [Citation22].

Statistical analyses

All data from the CRFs, including clinical information on patients’ age, gender, tumor staging clinically/visually and pathologically, type and number of NAC-cycles and days from last NAC to radical cystectomy were gathered in an SPSS 26 file (SPSS, Chicago, IL). Descriptive statistics were used to present data. Median values with interquartile range (IQR) were reported. The diagnostic accuracy of cystoscopy was assessed with sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV). Point estimates were presented with 95% confidence intervals (CI) based on the clopper Pearson (exact) method.

Results

For baseline characteristics and staging-procedures after TUR-B (cTNM) and after cystectomy (pTNM) see . All patients received neoadjuvant chemotherapy (NAC) and MVAC-HD was the most common regimen (n = 48). Two-to-six NAC cycles were given and 45 patients received three cycles. Post-cystectomy, 12 patients were classified as complete responders (CR), i.e. pT0. Twenty-five patients had residual MIBC locally and 14 had non-muscle invasive residual tumors (NMIBC). Eleven patients had lymph node metastases ().

The analysis of diagnostic accuracy of cystoscopy to correctly identify complete response or remaining tumor was 70% (CI = 56–81%) with a sensitivity of 64% (CI = 47–79%), specificity 82% (CI = 57–96%), PPV 89% (CI = 74–96%) and NPV 50% (CI = 38–61%). In 28/56 patients (50%) the visual staging was pT0 and in the remaining half the visual staging was any residual cancer. Three of the 28 cystoscopies with residual cancer were false positive (11%) ( and ). The visual staging of pT0 was false in 14/28 patients (50%). The false predictions of pT0 (n = 14) were distributed over true MIBC-patients as 6/14 and over true NMIBC-patients as 8/14. Of the 17 patients with actual pT0, 14 were correctly identified with a negative cystoscopy (82%) and of the 39 patients with actual residual tumor, 25 were correctly identified with a positive cystoscopy (64%). For further individual level data on cystoscopy findings, T- and N-stages ().

Table 2. Visual cystoscopy assessments over dichotomous pathoanatomical assessments of NAC-responses post-RC.

Table 3. Cystoscopy assessment after the final treatment with neoadjuvant chemotherapy (NAC) and assessment of NAC-response groups after cystectomy of the 56 study cases.

Table 4. Visual staging and response to NAC in each participating patient.

In 28/56 patients (50%) there were evaluations performed regarding the visual staging of actually residual tumor. In 3/14 patients (21%) with visual staging being residual MIBC, the estimates were incorrect; two had NMIBC and one had CR. In 4/14 patients with visual staging being residual NMIBC, the estimates were correct, 8/14 had MIBC and 2/14 had CR ().

Table 5. Results of the visual estimates of residual cancer, distributed over visual MIBC resp. visual NMIBC.

Discussion

The optimal long-term survival treatment for MIBC is yet to be found. Even though NAC has been a successful addition, in terms of improved OS, for a substantial number of patients since the introduction, yet there is a significant proportion of non-responders [Citation5–6].

One major problem with NAC is to early differentiate responders to treatment from non-responders. Patients with non-responding tumors risk undergoing substantial delays in treatment with an increased risk of dissemination in the course of time.

The Mansour trial suggested that, after the second NAC cycle, cystoscopy could be performed routinely for identifying responders versus non-responders to NAC [Citation20]. The investigators based their conclusions on registering that there were a significantly higher percentage of visually observed pT0 stages among responders to cisplatin-gemcitabine chemotherapy compared to what was found in non-responders; 36.5% versus 2.5%. This correlated with significantly better survival rates in the responder group.

In our study the patients had mainly received HD-MVAC chemotherapy instead of cisplatin-gemcitabine as in the Mansour-study. Mansour et al. performed the cystoscopy routinely after the second cycle of NAC, while the patients in our study were examined after the last cycle of NAC, mainly after the third cycle. Our study was performed prospectively and the possible confounder of bimanual palpation was excluded in conjunction with the cystoscopy. In contrast to the Mansour study, the same surgeon had not performed the cystoscopy on all the patients in our study. Although the visual evaluators were always subspecialized in surgical treatment of urinary bladder cancer, and members of the local bladder cancer-teams, there is of course a risk of interindividual bias. In our complete cohort of 56 patients, 14 patients were false negative at cystoscopy with 43% (6/14) turning out to have MIBC. Thus, if visual evaluation, as a clinical marker of response/non-response, would have been utilized for therapy choices in a real clinical setting, more than a third of the patients with FN cystoscopy would have been considered as successful NAC responders – yet being complete non-responders with remaining MIBC. Further, in as much as 8/14 patients had estimated visual NMIBC, the final histopathology post-RC revealed MIBC (non-response).

The strength of the study is the prospective design in a multicenter setting. However, the number of patients were low and larger sufficiently powered cohorts are needed to verify the results.

We conclude that cystoscopy after the final NAC-cycle could not robustly differentiate between NAC-responders and non-responders in this prospective pilot study. Visually, negative MIBC-status could not be determined safely and to utilize visual staging as part of response/non-response evaluations in patients with MIBC undergoing NAC seems to be of no clinical and predictive value.

Ethical approval

The study was approved by the Regional Ethics Board in Stockholm, dnr: 2007/71-31/2. The study conforms to the provisions of the Declaration of Helsinki (as revised in Fortaleza, Brazil, October 2013).

Informed consent

All included patients gave their oral and written consent to participate.

Consent for publication

All included patients gave their oral and written consent for the study to be published.

Author contributions

Protocol/project development: Amir Sherif, Danna Asad, Firas Aljabery. Data collection or management: Amir Sherif, Danna Asad, Firas Aljabery, Mohammad Hagsheno, Markus Johansson, Ylva Huge, Hans Netterling. Data analysis: Johan Svensson, Johan Styrke, Danna Asad, Firas Aljabery. Manuscript writing/editing: Amir Sherif, Danna Asad, Johan Styrke, Firas Aljabery, Mohammad Hagsheno, Markus Johansson, Ylva Huge, Hans Netterling, Johan Svensson, Funding: Amir Sherif. Supervision: Amir Sherif, Firas Aljabery, Johan Styrke.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

On reasonable request, the corresponding author can make available all codified data from the data base used for this study.

Additional information

Funding

This work was supported by the Swedish Research Council funding for clinical research in medicine (ALF) in Västerbotten, VLL [Grant No. Bas-ALF/VLL RV-848051], Sweden and the Cancer Research Foundation in Norrland, Umeå, Sweden [Grant No. CFF LP 13-2000].

References

  • Burger M, Catto JW, Dalbagni G, et al. Epidemiology and risk factors of urothelial bladder cancer. Eur Urol. 2013;63(2):234–241.
  • Antoni S, Ferlay J, Soerjomataram I, et al. Bladder cancer incidence and mortality: a global overview and recent trends. Eur Urol. 2017;71(1):96–108.
  • Witjes JA, Compérat E, Cowan NC, et al. Updated 2016 EAU guidelines on muscle-invasive and metastatic bladder cancer. Eur Urol. 2017;71(3):462–475.
  • Malmström P-U, Gårdmark T, Sherif A, et al. Incidence, survival and mortality trends of bladder cancer in Sweden 1997–2016. Scand J Urol. 2019;53(4):193–199.
  • Rosenblatt R, Sherif A, Rintala E, et al. Pathologic downstaging is a surrogate marker for efficacy and increased survival following neoadjuvant chemotherapy and radical cystectomy for muscle-invasive urothelial bladder cancer. Eur Urol. 2012;61(6):1229–1238.
  • Sherif A, Holmberg L, Rintala E, et al. Neoadjuvant cisplatinum based combination chemotherapy in patients with invasive bladder cancer: a combined analysis of two Nordic studies. Eur Urol. 2004;45(3):297–303.
  • Advanced Bladder Cancer (ABC) Meta-analysis Collaboration. Neoadjuvant chemotherapy in invasive bladder cancer: update of a systematic review and Meta-analysis of individual patient data advanced bladder cancer (ABC) Meta-analysis collaboration. Eur Urol. 2005;48(2):202–205. Augdiscussion 205-6.
  • Jerlström T, Chen R, Liedberg F, et al. No increased risk of short-term complications after radical cystectomy for muscle-invasive bladder cancer among patients treated with preoperative chemotherapy: a nation-wide register-based study. World J Urol. 2020;38(2):381–388.
  • Bhindi B, Frank I, Mason RJ, et al. Oncologic outcomes for patients with residual cancer at cystectomy following neoadjuvant chemotherapy: a pathologic stage-matched analysis. Eur Urol. 2017;72(5):660–664.
  • Lavery HJ, Stensland KD, Niegisch G, et al. Pathological T0 following radical cystectomy with or without neoadjuvant chemotherapy: a useful surrogate. J Urol. 2014;191(4):898–906.
  • Martini A, Jia R, Ferket BS, et al. Tumor downstaging as an intermediate endpoint to assess the activity of neoadjuvant systemic therapy in patients with muscle-invasive bladder cancer. Cancer. 2019;125(18):3155–3163.
  • Salminen A, Montoya Perez I, Klén R, et al. Adverse events during neoadjuvant chemotherapy for muscle invasive bladder cancer. BLC. 2019;5(4):273–279.
  • Seiler R, Al Deen Ashab H, Erho N, et al. Impact of molecular subtypes in muscle-invasive bladder cancer on predicting response and survival after neoadjuvant chemotherapy. Eur Urol. 2017;72(4):544–554.
  • Pietzak EJ, Zabor EC, Bagrodia A, et al. Genomic differences between “primary” and “secondary” muscle-invasive bladder cancer as a basis for disparate outcomes to cisplatin-based neoadjuvant chemotherapy. Eur Urol. 2019;75(2):231–239.
  • Font A, Domènech M, Benítez R, et al. Immunohistochemistry-Based taxonomical classification of bladder cancer predicts response to neoadjuvant chemotherapy. Cancers (Basel). 2020;12(7):1784.
  • Vollmer T, Schlickeiser S, Amini L, et al. The intratumoral CXCR3 chemokine system is predictive of chemotherapy response in human bladder cancer. Sci Transl Med. 2021;13(576):eabb3735.
  • Türker P, Wernroth M-L, Malmström P-U, et al. Combination of biomarkers for neoadjuvant systemic chemotherapy before cystectomy in patients with urinary bladder cancer. Transl Res. 2021;235:77–84.
  • During VA, Sole GM, Jha AK, et al. Prediction of histological stage based on cystoscopic appearances of newly diagnosed bladder tumours. Ann R Coll Surg Engl. 2016;98(8):547–551.
  • Mariappan P, Lavin V, Phua CQ, et al. Predicting grade and stage at cystoscopy in newly presenting bladder cancers-a prospective double-blind clinical study. Urology. 2017;109:134–139.
  • Mansour AM, Soloway MS, Eldefrawy A, et al. Prognostic significance of cystoscopy findings following neoadjuvant chemotherapy for muscle-invasive bladder cancer. Can J Urol. 2015;22(2):7690–7697.
  • Becker REN, Meyer AR, Aaron Brant A, et al. Clinical restaging and tumor sequencing are inaccurate indicators of response to neoadjuvant chemotherapy for muscle-invasive bladder cancer. Eur Urol. 2021;79(3):364–371.
  • Alvaeus J, Rosenblatt R, Johansson M, et al. Fewer tumour draining sentinel nodes in patients with progressing muscle invasive bladder cancer, after neoadjuvant chemotherapy and radical cystectomy. World J Urol. 2020;38(9):2207–2213.
  • Zirakzadeh AA, Kinn J, Krantz D, et al. Doxorubicin enhances the capacity of B cells to activate T cells in urothelial urinary bladder cancer. Clin Immunol. 2017;176:63–70.
  • Winerdal ME, Krantz D, Hartana CA, et al. Urinary bladder cancer tregs suppress MMP2 and potentially regulate invasiveness. Cancer Immunol Res. 2018;6(5):528–538.
  • Hartana CA, Ahlén Bergman E, Zirakzadeh AA, et al. Urothelial bladder cancer may suppress perforin expression in CD8+ T cells by an ICAM-1/TGFβ2 mediated pathway. PLoS One. 2018;13(7):e0200079.
  • Hartana CA, Ahlén Bergman E, Broomé A, et al. Tissue-resident memory T cells are epigenetically cytotoxic with signs of exhaustion in human urinary bladder cancer. Clin Exp Immunol. 2018;194(1):39–53.
  • Ahlén Bergman E, Hartana CA, Johansson M, et al. Increased CD4+ T cell lineage commitment determined by CpG methylation correlates with better prognosis in urinary bladder cancer patients. Clin Epigenetics. 2018;10(1):102.
  • Krantz D, Hartana CA, Winerdal ME, et al. Neoadjuvant chemotherapy reinforces antitumour T cell response in urothelial urinary bladder Cancer. Eur Urol. 2018;74(6):688–692.