Effectiveness and safety of combined therapy versus monotherapy based on immune checkpoint inhibitors and/or targeted drugs as salvage treatment for advanced urothelial carcinoma: a systematic review and meta-analysis
Original Article

Effectiveness and safety of combined therapy versus monotherapy based on immune checkpoint inhibitors and/or targeted drugs as salvage treatment for advanced urothelial carcinoma: a systematic review and meta-analysis

Lichao Wei#, Liang Gao#, Zili Hu, Chuan Liu^

Department of Urology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China

Contributions: (I) Conception and design: L Wei, L Gao; (II) Administrative support: C Liu; (III) Provision of study material or patients: L Wei, L Gao, Z Hu; (IV) Collection and assembly of data: L Wei, L Gao, Z Hu; (V) Data analysis and interpretation: L Wei, L Gao, Z Hu; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

^ORCID: 0000-0002-6362-0261.

Correspondence to: Chuan Liu. Department of Urology, the Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Chongqing 400010, China. Email: liuchuan100@hospital.cqmu.edu.cn.

Background: The standard salvage regimen for the patients with advanced urothelial carcinoma (UC) is uncertain, although lots of novel agents are recommended, including immune checkpoint inhibitors (ICIs) and targeted drugs (TDs). We aimed to compare the effectiveness and safety of combined therapy of novel agents (CNA) and monotherapy of novel agents (MNA) as salvage therapy for advanced UC.

Methods: Studies exploring CNA and/or MNA for advanced UC in second-line setting were searched from PubMed, Embase, Cochrane Library, and Web of Science. The data of objective response rate (ORR), disease control rate (DCR), median progression-free survival (PFS), median overall survival (OS), and grade 3-4 adverse effects rate (grade 3-4 AEs%) were pooled for analyses. Cochrane risk of bias tool was applied for the quality judgment of randomized controlled studies (RCTs).

Results: Forty-one arms from 37 studies including 4,691 patients were included. Significant differences were presented in pooled ORR (22.9% versus 12.2%, OR =1.88, P<0.001) and DCR (62.7% versus 37.5%, OR =2.53, P<0.001) between CNA and MNA groups. The pooled median PFS was 3.66 months in CNA group versus 2.16 months in MNA group (WMD =1.50, P=0.028). No significant difference in pooled median OS was found between two groups (7.93 versus 7.50 months, WMD =0.43, P=0.449). 63.7% versus 25.4% of pooled grade 3-4 AEs% could be seen in CNA and MNA groups (OR =3.52, P<0.001). Additionally, the pooled results of PFS-6m and OS-6m in CNA group demonstrated significant advantages over MNA group (31.5% versus 28.7%, OR =1.31, P=0.049; 66.0% versus 56.7%, OR =1.34, P=0.029, respectively). In the subgroup analysis of CNA, use of ICIs, the positive expression of PD-L1 and ECOG-PS =0 were significantly associated with superior clinical outcomes (P<0.05).

Discussion: For advanced UC patients after first line agents, CNA had potential benefits than MNA in terms of ORR, DCR, median PFS, PFS-6m and OS-6m. However, CNA was associated with a significantly higher grade 3-4 AEs%. Furthermore, potential advantages were presented in CNA patients with ICIs usage, positive PD-L1 expression and ECOG-PS =0.

Keywords: Urothelial carcinoma (UC); salvage therapy; novel agents; combined therapy; monotherapy


Submitted Nov 30, 2020. Accepted for publication Apr 02, 2021.

doi: 10.21037/tcr-20-3354


Introduction

Platinum-based chemotherapy is one of the first-line agents for advanced urothelial carcinoma (UC), with a median overall survival of 13.8 months in gemcitabine plus cisplatin and 15.1 months in high-dose intensity methotrexate, vinblastine, doxorubicin, and cisplatin (1,2). Nevertheless, the standard regimen is still controversial for patients who failed to chemotherapy.

Monotherapy of novel agents (MNA), including immune checkpoint inhibitors (ICIs) and targeted drugs (TDs), was recommended as rational regimens, which demonstrated objective response rate (ORR) of about 20% in some studies (3,4). Furthermore, dozens of trails reported the combination treatment of novel agents (CNA), presenting more satisfactory outcomes than MNA. CheckMate 032 evaluated the locally advanced or metastatic platinum pretreated UC cohort receiving combined inhibition of PD-1 (nivolumab 1 mg/kg) and cytotoxic T-lymphocyte antigen-4 (ipilimumab 3 mg/kg), which showed notable ORR with a complete response of 6.5% and a partial response of 31.5% (5). The anti-tumor activity was also revealed in the combination treatment of tyrosine kinase inhibitor (pazopanib) plus cytotoxic drug (paclitaxel), which presented a remarkable ORR of 54% and median progression-free survival (PFS) of 6.2 months (6). In addition, the combined treatment regimens of everolimus plus pazopanib and pembrolizumab plus docetaxel/gemcitabine were also recommended because of attractive clinical benefits (7,8). Nonetheless, other studies found that MNA might get more beneficial anti-cancer outcomes in median overall survival (OS) and OS-12m than CNA (3,7), which made this issue still obscure. Additionally, possibly more adverse effects in CNA could not be ignored. Grade 3 or 4 adverse events rate (grade 3-4 AEs%) could be observed in approximate 40% patients for CNA, including fatigue, anemia, neutrophil count decrease, etc. (5,8).

Therefore, we designed this systematic review and meta-analysis to compare the effectiveness and safety between CNA and MNA for the management of advanced UC failing to first-line treatment.

We present the following article in accordance with the PRISMA reporting checklist (9) (available at http://dx.doi.org/10.21037/tcr-20-3354).


Methods

This meta-analysis has been registered (registered number: CRD42020199791).

Search strategy

Eligible studies in English language were searched in the PubMed, Cochrane Library, Embase, and Web of Science from the date of inception up to May 1, 2020. The search strategy included the following terms in title/abstract: (‘bladder cancer’ OR ‘urothelial cancer/carcinoma/neoplasm’ OR ‘transitional cell cancer/carcinoma’) AND (‘second line’ OR ‘previously’ OR ‘refractory’ OR ‘resistant’ OR ‘progressive’ OR ‘pretreated’ OR ‘advanced’ OR ‘metastatic’) AND (‘pembrolizumab’ OR ‘nivolumab’ OR ‘durvalumab’ OR ‘avelumab’ OR ‘atezolizumab’ OR ‘ipilimumab’ OR ‘tremelimumab’ OR ‘aflibercept’ OR ‘sunitinib’ OR ‘sorafenib’ OR ‘pazopanib’ OR ‘ramucirumab’ OR ‘icrucumab’ OR ‘vandetanib’ OR ‘lapatinib’ OR ‘everolimus’ OR ‘temsirolimus’ OR ‘apatorsen’ OR ‘cetuximab’ OR ‘SCH66336’). In addition, related references of the acquired literature were reviewed.

Selection criteria

The inclusion criteria were as follows: (I) randomized controlled studies (RCTs) or single-arm studies; (II) patients with advanced UC who were refractory to previous chemotherapy or ICI; (III) patients who were treated by CNA (ICI plus chemotherapy, TD plus chemotherapy, ICI plus TD, dual ICIs, or dual TDs) or MNA (ICI or TD); (IV) studies reporting at least one of outcomes of interest, including ORR, disease control rate (DCR), median PFS, median OS, and grade 3-4 AEs%.

The exclusion criteria were as follows: (I) articles with unrelated topics; (II) papers published as letters, editorials, errata, meta-analyses, reviews, conference abstracts, case-reports, study designs or animal trials; (III) novel agents used as the first choice for advanced UC; (IV) studies with unextractable data, with patient number less than 10, or without full texts.

Data extraction and quality assessment

The data of interest were extracted independently by two investigators. The primary outcomes of interest included ORR, DCR, median PFS, median OS, and grade 3-4 AEs%. ORR was defined as the percentage of patients which had a complete or partial response. DCR was defined as the percentage of patients with a complete response, partial response, or stable disease. In addition, the second outcomes of interest contained PFS-6m, PFS-12m, OS-6m, OS-12m, OS-24m, and any grade AEs%. If disagreements existed, it would be settled by consensus after discussion with a third investigator.

The Cochrane risk of bias tool was applied for methodological quality judgment of RCTs (10). For single-arm studies, no credible tools were found to assess their quality.

Statistical analysis

All analyses were performed using Stata software version 13.0 (StataCorp, College Station, TX, USA) and RevMan software version 5.3 (The Cochrane Collaboration, Software Update, Oxford). For dichotomous variables, rate and standard error (SE) was used to assess the pooled effect sizes (ESs). For continuous variables, the pooled ESs were evaluated through mean and SE. Fisher exact test and Wilcoxon rank sum test was applied for the comparison of pooled outcomes of dichotomous and continuous variables, respectively (11). Two-sided P<0.05 was considered to be statistically significant. Heterogeneity among studies was assessed by the Cochran Q chi-square test and I2 statistics, in which P<0.10 was regarded to be significant. Furthermore, the random-effects models were applied when heterogeneity among studies was significant. Otherwise, the fixed-effects models were used. Certain studies which possibly contributed to a high heterogeneity would be excluded for a sensitivity analysis to lower heterogeneity. Publication bias was evaluated by funnel plots and Egger’s test.

Furthermore, in CNA group, subgroup analyses were carried out according to agent type, programmed death-ligand 1 (PD-L1) expression, gender, age, Eastern Cooperative Oncology Group Performance Status (ECOG-PS), liver metastases, haemoglobin, Bellmunt risk factor (including liver metastases, haemoglobin <10 g/dL, and ECOG-PS score >0), and tumor site.


Results

Study identification and characteristics

A total of 821 studies were obtained after removing duplication from initial literature search, and 37 articles were eligible for final meta-analysis, including 4,691 first-line treatment failed patients with advanced UC of bladder, urethra, ureter, or renal pelvis. The flow chart was exhibited in Figure 1. Twelve RCTs with 16 arms and 25 single-arm studies were included. Of them, 16 arms reported CNA of ICI plus chemotherapy (n=1), TD plus chemotherapy (n=11), ICI plus TD (n=1), dual ICIs (n=2), and dual TDs (n=1). On the other hand, 25 arms reported MNA of ICI (n=14) and TD (n=11).

Figure 1 The flow chart of study identification.

The median sample sizes of studies were 33 (range: 12–263) and 59 (range: 11–997) in CNA and MNA groups, respectively. Overall, in CNA group, the median age was 66 years, with 76% of males. In MNA group, the median age was 67 years, with 75% of males. Additionally, 33 studies only enrolled the patients who progressed after platinum-based chemotherapy, and 4 articles contained a few participants failed from 1st line ICIs. Only one study designed the sub-analysis to research the association of outcomes and CNA in 17 patients after 1st line ICIs (12). The Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 and 1.0 were applied to evaluate ORR and DCR in 28 arms and 13 arms, respectively. The basic characteristics of 41 arms were demonstrated in Table 1.

Table 1

Baseline characteristics of 41 arms for meta-analysis

Author Study type Treatment Patients, n Median age, years ORR, %
(95% CI)
DCR, %
(95% CI)
Median PFS, months (95% CI) Median OS, months (95% CI) Grade 3-4 AEs, % (95% CI)
MNA
   Necchi, 2012 (4) P Paz 41 67.0 17.1 (7.2–32.1) 51.2 (35.1–67.1) 2.6 (1.7–3.7) 4.7 (4.2–7.3) 29.2 (NA)
   Sharma, 2019 (5) P Niv 78 65.5 25.6 (16.4–36.8) 52.6 (NA) 2.8 (1.5 to 5.3) 9.9 (7.3–21.1) 26.9 (NA)
   Wong, 2012 (13) P Cet 11 71.0 0.0 (NA) 0.0 (NA) NA 1.9 (1.5–NR) NA
   Fradet, 2019 (14) P Pem 270 67.0 21.1 (16.4–26.5) 38.5 (NA) 2.1 (2.0–2.2) 10.1 (8.0–12.3) 16.5 (NA)
   Yasuoka, 2019 (15) R Pem 40 69.0 20.6 (NA) NA 4.2 10.0 (NA) 10.0 (NA)
   Tamura, 2020 (16) R Pem 41 70.0 14.6 (NA) 56.4 (NA) 2.5 (1.4–6.2) 11.9 (4.9–NA) 4.9 (NA)
   Petrylak, 2018 (17) P Ate 95 66.0 26.3 (NA) 45.3 (NA) 2.7 (1.4–4.3) 10.1 (7.3–17.0) 9.5 (NA)
   Powles, 2018 (18) P Ate 467 67.0 13.4 (10.5–16.9) 33.3 (NA) 2.1 (2.1–2.2) 8.6 (7.8–9.6) 19.8 (NA)
   Pal, 2018 (19) P Ate 214 69.0 14.9 (NA) 49.1 (NA) NA NA 7.9 (NA)
   Sternberg, 2019 (20) P Ate 997 68.0 13.0 (11.0–16.0) 40.0 (37.0–43.0) 2.2 (2.1–2.4) 8.7 (7.8–9.9) 44.9 (NA)
   Rosenberg, 2016 (21) P Ate 310 66.0 15.0 (11.0–19.0) 33.5 (NA) 2.7 (2.1–3.9) 7.9 (6.6–9.3) 16.1 (NA)
   Apolo, 2017 (22) P Ave 44 68.0 18.2 (8.2–32.7) 52.3 (NA) 2.9 (1.5–4.4) 13.7 (8.5–NE) 6.8 (NA)
   Patel, 2018 (23) P Ave 249 68.0 17.0 (11.0–24.0) 39.8 (NA) 1.6 (1.5–2.5) 6.5 (4.8–9.5) 8.4 (NA)
   Ohyama, 2019 (24) P Niv 270 66.0 20.4 (15.7–25.7) 41.5 (NA) 1.9 (1.9–2.3) 8.6 (6.1–11.3) 22.6 (NA)
   Powles, 2017 (25) P Dur 191 67.0 17.8 (12.7–24.0) 36.6 (29.8–43.9) 1.5 (1.4–1.9) 18.2 (8.1–NE) 6.8 (NA)
   Twardowski, 2010 (26) P Afl 22 67.0 4.5 (0.1–22.8) 36.4 (NA) 2.79 (1.7-3.9) NA NA
   Wülfing, 2009 (27) P Lap 59 64.0 2.0 (0.0–9.1) 32.2 (NA) 2.2c (2.0–2.8) 4.5 (3.3–7.6) NA
   Dreicer, 2009 (28) P Sor 22 66.0 0.0 (NA) 13.6 (NA) 2.2b (1.8–3.7) 6.8 (5.7–8.5) NA
   Gallagher, 2010 (29) P Sun 77 NA 5.2 (NA) 28.6 (NA) NA NA 74.0 (NA)
   Sharma, 2020 (30) P Tre 32 66.5 18.8 (7.2–36.4) 28.1 (NA) 2.6 (NA) 10.3 (NA) 28.1 (NA)
   Pili, 2013 (31) P Paz 18 65.6 0.0 (NA) NA 1.9 (NA) NA 38.9 (NA)
   Jones, 2017 (32) P Paz 66 69.0 4.5 (NA) 36.4 3.1a (2.7–4.6) 4.7a (4.2–6.4) 37.9 (NA)
   Seront, 2012 (33) P Eve 37 63.0 5.4 (NA) 27.0 (NA) 2.0 (1.6–2.1) 3.4 (2.7–4.3) NA
   Milowsky, 2013 (34) P Eve 45 66.0 5.4 (NA) 37.8 (NA) 2.6 (1.8–3.5) 8.3 (5.5–12.1) 64.4 (NA)
   Pulido, 2018 (35) P Tem 54 65.0 6.7 (NA) 48.9 (NA) 2.8 (1.8–3.7) 7.2 (5.2–9.5) 52.8 (NA)
CNA
   Sharma, 2019 (5) P N3I1 104 63.0 26.9 (18.7–36.5) 50.0 (NA) 2.6 (1.4– 3.9) 7.4 (5.6–11.0) 30.8 (NA)
   Sharma, 2019 (5) P N1I3 92 64.0 38.0 (28.1–48.8) 63.0 (NA) 4.9 (2.7– 6.6) 15.3 (10.1–27.6) 39.1 (NA)
   Narayanan, 2016 (6) P PP 32 67.0 53.6 (NA) 92.9 (NA) 6.2 (5.6–7.6) 10.0 (5.7–16.0) NA
   Bellmunt, 2018 (7) P EP 19 69.0 21.1 (NA) 63.2 (NA) 3.6 (1.8–5.6) 9.1 (6.2–13.1) 73.7 (NA)
   Parikh, 2018 (8) P PD/PG 12 66.0 41.7 (NA) 58.3 (NA) 4.8 (NA) NA 58.3 (NA)
   Petrylak, 2020 (12) P RD 263 65.0 25.9 (20.6–31.1) 65.4 (59.7–71.1) 4.1 (3.3–4.8) 9.4 (7.9–11.4) 47.7 (NA)
   Wong, 2012 (13) P CP 28 69.0 25.0 (11.0–45.0) NA 4.1 (3.0–6.3) 10.5 (7.6–19.5) NA
   Petrylak, 2016 (36) P RD 46 67.5 24.0 (12.6–38.8) 78.0 (63.6–89.1) 5.4 (3.1–6.9) 10.4 (7.0– 15.1) 82.6 (NA)
   Petrylak, 2016 (36) P ID 49 66.0 12.0 (4.6–24.8) 45.0 (30.7–59.8) 1.6 (1.4– 2.9) 6.7 (4.5–8.5) 83.7 (NA)
   Herbst, 2019 (37) P RP 24 63.0 13.0 (2.7–32.4) 50.0 (29.1–70.9) 1.9 (1.2–2.8) 6.4 (2.5–18.7) NA
   Rosenberg, 2018 (38) P AD 99 68.0 16.1 (11.5–21.9) NA 1.8 (NA) 6.4 (NA) 82.8 (NA)
   Miyata, 2015 (39) P SGP 20 74.0 5.0 (NA) NA NA 7.0 (NA) NA
   Choueiri, 2012 (40) P VD 70 NA 11.4 (NA) 51.4 (NA) 2.6 (NA) 5.9 (NA) 60.0 (NA)
   Niegisch, 2015 (41) P PE 27 63.0 12.5 (NA) 58.3 (NA) 2.9 (1.9–4.2) 5.6 (4.8–10.2) 80.0 (NA)
   Theodore, 2005 (42) P SG 34 63.6 32.3 (17.0–51.0) NA 7.0 (NA) 11.5 (NA) NA
   Shah, 2019 (43) P SV 22 62.5 41.2 (NA) 70.6 (NA) 4.5 (NA) 7.0 (NA) NA

CNA, combined therapy based on novel agents; MNA, monotherapy of novel agents; ORR, objective response rate; DCR, disease control rate; PFS, progression-free survival; OS, overall survival; AEs%, adverse effects rate; CI, confidence interval; P, prospective; R, retrospective; a, 80% CI; b, 90% CI; c, time to progression; NR, not reached; NA, not available; NE, not estimable; Paz, pazopanib; Niv, nivolumab; Cet, cetuximab; Pem, pembrolizumab; Ate, atezolizumab; Ave, avelumab; Dur, durvalumab; Afl, Aflibercept; Lap, lapatinib; Sor, sorafenib; Sun, sunitinib; Tre, tremelimumab; Eve, everolimus; Tem, temsirolimus; N3I1, nivolumab 3 mg/kg, ipilimumab 1 mg/kg; N1I3, nivolumab 1 mg/kg, ipilimumab 3 mg/kg; PP, pazopanib, paclitaxel; EP, everolimus, pazopanib; PD, pembrolizumab, docetaxel; PG, pembrolizumab, gemcitabine; RD, ramucirumab, docetaxel; ID, icrucumab, docetaxel; RP, ramucirumab, pembrolizumab; CP, cetuximab, paclitaxel; AD, apatorsen, docetaxel; SGP, sorafenib, gemcitabine, paclitaxel; VD, vandetanib, docetaxel; PE, paclitaxel, everolimus; SG, SCH66336, gemcitabine; SV, sorafenib, vinflunine.

Quality assessment

Methodological quality was evaluated for RCTs, with low risk of blinding of participants and personnel, blinding of outcome assessment, incomplete outcome data, and selective reporting in a large proportion of articles, but high or unclear risk of random sequence generation, allocation concealment, or other bias in some studies (Figure S1).

Response and survival outcome

A total of 16 trials in CNA group and 25 trials in MNA group reported outcomes of ORR. The pooled ORR in CNA group could be calculated to be 22.9% (95% CI, 17.3–28.5, I2=75.4%, Figure 2A), while 12.2% (95% CI, 9.4–14.9, I2=82.5%, Figure 2B) in MNA group. The difference between two groups was significant (OR =1.88, P<0.001). After excluding 6 studies (5,18,26,28-30) in MNA and 4 studies (5,6,40,41) in CNA because of large heterogeneity for a sensitivity analysis, the pooled ORRs with lower heterogeneity were presented to be 21.9% (95% CI, 18.9–24.9, I2=45.9%) in CNA group and 11.1% (95% CI, 8.8–13.4, I2=63.1%) in MNA group (OR =2.01, P<0.001).

Figure 2 Forest plots of response outcomes of CNA and MNA. (A) Pooled ORR of CNA. (B) Pooled ORR of MNA. (C) Pooled DCR of CNA. (D) Pooled DCR of MNA. CNA, combined therapy of novel agents; MNA, monotherapy of novel agents; ORR, objective response rate; DCR, disease control rate.

The DCR was available for analysis in 12 trials of CNA group and 23 trials of MNA group. The pooled DCR was 62.7% (95% CI, 51.0–71.4, I2=82.6%, Figure 2C) in CNA and 37.5% (95% CI, 33.9–41.2, I2=74.5%, Figure 2D) in MNA, with a significant difference (OR =2.53, P<0.001). The results after omitting 4 studies (5,17,21,29) of MNA and 2 studies (6,37) of CNA for a sensitivity analysis revealed that the modified pooled DCR in CNA was still significantly higher than that in MNA (59.2% versus 36.4%, OR =2.35, P<0.001).

Ten studies in CNA and 20 studies in MNA reported the median PFS. The Pooled data demonstrated that the median PFS in CNA group was significantly longer than that in MNA group [3.66 versus 2.16 months, weighted mean difference (WMD) =1.50, P=0.028, Figure 3A,B]. After omitting 4 studies (14,16,19,30) of MNA and 3 studies (6,13,37) of CNA, the modified result of sensitivity analysis showed that the difference between two groups was still significant (3.76 versus 2.12 months, WMD =1.64, P=0.002). Additionally, there was significant difference between CNA and MNA in PFS-6m (31.5% versus 28.7%, OR =1.31, P=0.049), but not in PFS-12m (14.4% versus 16.7%, OR =0.87, P=0.384).

Figure 3 Forest plots of survival outcomes of CNA and MNA. (A) Pooled median PFS of CNA. (B) Pooled median PFS of MNA. (C) Pooled median OS of CNA. (D) Pooled median OS of MNA. CNA, combined therapy of novel agents; MNA, monotherapy of novel agents; PFS, progression-free survival; OS, overall survival.

The OS was available for analysis from 12 trials in CNA and 15 trials in MNA. However, the pooled median OS was insignificantly different between two groups (WMD =0.43, P=0.449, Figure 3C,D). After omitting 5 studies (4,16,28,30,34) from MNA with high heterogeneity, the difference was insignificant as well (WMD =−0.18, P=0.843). Furthermore, CNA was significantly associated with higher OS-6m (66.0% versus 56.7%, OR =1.34, P=0.029) and lower OS-24m (17.7% versus 28.3%, OR =0.55, P<0.001), but insignificant lower OS-12m (39.5% versus 42.8%, OR =0.94, P=0.47), compared with MNA (Table 2).

Table 2

Summary of the pooled outcomes of effectiveness between CNA and MNA

Groups Cohorts, n Event, % (95% CI) Time, months (95% CI) I2 OR WMD P value
ORR
   CNA 16 22.9 (17.3–28.5) 75.4% 1.88 <0.001
   MNA 25 12.2 (9.4–14.9) 82.5%
DCR
   CNA 12 62.7 (51.0–71.4) 82.6% 2.53 <0.001
   MNA 23 37.5 (33.9–41.2) 74.5%
mPFS
   CNA 10 3.66 (2.61–4.72) 89.3% 1.50 0.028
   MNA 20 2.16 (2.02–2.31) 74.0%
PFS-6m
   CNA 6 35.1 (27.7–35.2) 44.5% 1.31 0.049
   MNA 4 28.7 (20.7–36.7) 71.0%
PFS-12m
   CNA 6 14.4 (8.4–20.3) 74.1% 0.87 0.384
   MNA 5 16.7 (14.2–19.2) 0.0%
mOS
   CNA 12 7.93 (7.08–8.77) 26.1% 0.43 0.449
   MNA 15 7.50 (6.07–8.93) 92.9%
OS-6m
   CNA 3 66.0 (60.7–71.3) 35.4% 1.34 0.029
   MNA 5 56.7 (50.7–62.8) 66.0%
OS-12m
   CNA 9 39.5 (33.9–45.1) 50.0% 0.94 0.470
   MNA 11 42.8 (39.5–46.1) 59.1%
OS-24m
   CNA 3 17.7 (13.7–21.6) 0.0% 0.55 <0.001
   MNA 3 28.3 (24.8–31.8) 0.0%

CI, confidence interval; CNA, combined therapy based on novel agents; MNA, monotherapy of novel agents; ORR, objective response rate; DCR, disease control rate; mPFS, median progression-free survival; mOS, median overall survival; OR, odds ratio; WMD, weighted mean difference.

Toxicity

Ten trials of CNA and 20 trials of MNA reported the grade 3-4 AEs%. The pooled results presented that there was significantly higher grade 3-4 AEs% in CNA than MNA (63.7% versus 25.4%, OR =3.52, P<0.001, Figure 4A,B). The results of sensitivity analysis suggested that omitting any one study did not obviously decrease the heterogeneity. And the most frequent grade 3-4 AEs in CNA were neutropenia (16.6%), leukopenia (10.8%), fatigue (9.8%), anemia (6.5%), diarrhea (4.5%), hypertension (3.3%), stomatitis (3.3%), elevated ALT (3.0%), elevated AST (2.9%), renal failure (2.7%), and rash (2.6%). In addition, similar significance could be found in any grade AEs% between the two groups (87.9% versus 66.9%, OR =2.15, P<0.001) (Table 3).

Figure 4 Forest plots of toxicity of CNA and MNA. (A) Pooled grade 3-4 AEs% of CNA. (B) Pooled grade 3-4 AEs% of MNA. CNA, combined therapy of novel agents; MNA, monotherapy of novel agents; grade 3-4 AEs%, grade 3 or 4 adverse events rate.

Table 3

Summary of the pooled outcomes of safety between CNA and MNA

Groups Cohorts, n Event, % (95% CI) I2 OR P value
Grade 3-4 AEs%
   CNA 10 63.7 (50.0–77.4) 94.1% 3.52 <0.001
   MNA 20 25.4 (18.1–32.7) 97.0%
Any grade AEs%
   CNA 7 87.9 (80.9–94.8) 92.5% 2.15 <0.001
   MNA 15 66.9 (59.2–74.5) 95.9%
Anemia
   CNA 15 6.5 (3.8–9.3) 79.0% 2.72 <0.001
   MNA 15 1.9 (0.9–2.9) 69.7%
Neutropenia
   CNA 11 16.6 (9.6–23.6) 86.2% 22.06 <0.001
   MNA 10 0.5 (0.1–1.0) 0.0%
Leukopenia
   CNA 7 10.8 (3.3–18.4) 87.2% 12.55 <0.001
   MNA 6 0.2 (−0.1–0.5) 31.9%
Thrombocytopenia
   CNA 11 0.7 (0.0–1.3) 47.3% 0.66 0.242
   MNA 8 2.8 (0.5–5.0) 57.3%
Nausea
   CNA 12 1.2 (0.4–2.0) 0.0% 2.48 0.022
   MNA 17 0.3 (0.1–0.4) 0.3%
Vomiting
   CNA 8 1.3 (0.3–2.2) 0.0% 3.16 0.009
   MNA 11 0.4 (0.1–0.7) 0.0%
Decreased appetite
   CNA 5 0.8 (0.1–1.5) 0.0% 1.27 0.76
   MNA 10 0.4 (0.2–0.6) 0.0%
Diarrhea
   CNA 14 4.5 (3.1–5.8) 0.0% 6.28 <0.001
   MNA 22 0.6 (0.3–0.8) 0.0%
Constipation
   CNA 3 0.5 (−0.2–1.3) 3.8% 7.43 0.013
   MNA 7 0.2 (−0.0–0.4) 0.0%
Fatigue
   CNA 15 9.8 (6.1–13.6) 82.5% 4.34 <0.001
   MNA 21 1.7 (0.9–2.5) 68.3%
Neuropathy
   CNA 7 1.6 (−0.2–3.5) 55.0% 23.82 <0.001
   MNA 6 0.2 (−0.0–0.5) 0.0%
Renal failure
   CNA 5 2.7 (−0.4–5.8) 50.3% 2.40 0.068
   MNA 4 0.8 (0.3–1.2) 0.0%
Hypertension
   CNA 10 3.3 (1.9–4.8) 0.0% 2.12 0.008
   MNA 10 1.2 (0.7–1.7) 33.2%
Dyspnoea
   CNA 8 1.4 (0.5–2.3) 22.2% 2.87 0.005
   MNA 6 0.7 (0.3–1.1) 0.0%
Pneumonia
   CNA 7 1.6 (0.5–2.8) 19.4% 2.47 0.018
   MNA 10 1.0 (0.5–1.6) 0.0%
Alopecia
   CNA 4 0.6 (−1.0–2.2) 55.2% 4.45 0.222
   MNA 3 0.2 (−0.3–0.7) 0.0%
Stomatitis
   CNA 2 3.3 (1.2–5.4) 0.0% 4.60 0.006
   MNA 6 0.3 (−0.1–0.7) 0.0%
Hypothyroidism
   CNA 3 0.6 (−0.4–1.5) 0.0% 10.28 0.170
   MNA 9 0.1 (−0.0–0.3) 0.0%
Rash
   CNA 8 2.6 (0.4–4.8) 57.5% 7.45 <0.001
   MNA 16 0.4 (0.2–0.6) 0.0%
Elevated ALT
   CNA 6 3.0 (0.4–5.7) 57.6% 1.31 0.573
   MNA 8 0.8 (0.2–1.4) 42.9%
Elevated AST
   CNA 5 2.9 (0.9–4.9) 0.0% 2.49 0.038
   MNA 8 0.8 (0.2–1.3) 0.0%

CNA, combined therapy based on novel agents; MNA, monotherapy of novel agents; AEs%, adverse effects rate; ALT, alanine aminotransferase; AST, aspartate aminotransferase; OR, odds ratio.

Subgroup analyses in CNA group

In CNA group, the statistical difference could be found between ICIs and TDs in pooled ORR (32.2% versus 21.0%, OR =1.68, P=0.003), but not in pooled median PFS (3.63 versus 3.95 months, WMD =−0.32, P=0.769) and OS (10.24 versus 7.92 months, WMD =2.32, P=0.345).

Furthermore, the ORR of participants with positive PD-L1 expression was significantly higher than those negative (36.9% versus 24.7%, OR =1.97, 95% CI, 1.20–3.24, P=0.008, I2=41.9%, Figure 5A), and the statistical superiority could be also found in DCR (68.6% versus 50.9%, OR =1.81, 95% CI, 1.13–2.91, P=0.014, I2=0.0%, Figure 5B). However, the median PFS and median OS between participants with PD-L1 (+) and PD-L1 (-) were insignificantly different (4.28 versus 3.12 months, WMD =1.05, 95% CI, 0.42–2.51, P=0.162, I2=0.0%; 8.15 versus 6.76 months, WMD =0.81, 95% CI, −3.19–4.81, P=0.69, I2=0.0%). Additionally, the patients of ECOG-PS =0 suggested statistically better ORR than those of ECOG-PS ≥1 (46.7% versus 18.3%, OR =5.55, 95% CI, 1.33–23.22, P=0.019, I2=40.9%, Figure 5C). And the pooled results of other prognostic indicators demonstrated superior clinical outcomes and revealed the trends of significant differences (Tables S1-S3).

Figure 5 Forest plots of the subgroup analyses in CNA group. (A) ORR of PD-L1(+) vs. PD-L1(-). (B) DCR of PD-L1(+) vs. PD-L1(-). (C) ORR of ECOG-PS =0 vs. ECOG-PS ≥1. CNA, combined therapy of novel agents; ORR, objective response rate; DCR, disease control rate; PD-L1, programmed death-ligand 1; ECOG-PS, Eastern Cooperative Oncology Group Performance Status.

Sensitivity analysis and publication bias diagnosis

All sensitivity analyses were presented in Figure S2. No obvious publication bias existed by funnel plot and Egger’s test for most outcomes in CNA and MNA groups. The details of all publication bias were provided in Figure S3.


Discussion

Platinum-based chemotherapy or ICI was the first-line treatment for advanced UC (1,2,44). However, it was still inconclusive for the patients with progressive or relapsed disease after first-line regimens. Although MNA including ICIs and TDs, was proved to be a considerable salvage choice for these patients (4,16), CNA demonstrated better results (8,12). Nevertheless, up to now, there was little study comparing the effectiveness and safety of CNA and MNA as salvage treatment for this condition.

This meta-analysis demonstrated significant advantages of CNA over MNA in terms of ORR, DCR, PFS, PFS-6m, and OS-6m. The statistical differences remained after sensitivity analyses. However, it could not be ignored that CNA was associated with higher grade 3-4 AEs%. In the subgroup analyses of CNA, ICIs presented better ORR than TDs, and the prognosis was superior in the patients with PD-L1(+) and ECOG-PS =0.

Programmed death 1 (PD-1) receptor was extensively expressed on activated and exhausted T cells, macrophages, dendritic cells, and B cells, resulting in tumor immune escape by the combination with PD-L1 expressing on tumor cells (45). Somatic mutations and increased immunogenicity were previously observed in UC cells, which suggested that UC was a kind of immune-responsive tumors and monoclonal antibody of immune checkpoints could be a probably rational choice for the treatment of UC (46).

On the other hand, a myriad of molecular targets had been also found on the surface of UC cells, demonstrating the potential of TDs. Vascular endothelial growth factor receptor (VEGFR) and epidermal growth factor receptor (EGFR) of UC could induce cell proliferation and migration when binding to VEGF and EGF. Additionally, mammalian target of rapamycin (mTOR) pathway was detected in UC, which was associated with poor prognosis. The inhibitors of VEGFR, EGFR and mTOR pathway presented remarkable anti-cancer efficacy (27,28,33).

Furthermore, it had been proved that chemotherapy-induced immunogenic modulation could enhance the anti-tumor activity cytotoxic T lymphocytes through increasing tumor’s sensitivity (47). Cytotoxic drugs inhibited angiogenesis through increasing microtubule dynamics in endothelial cells, impairing interphase microtubule functions, and degrading heat shock protein 90, etc. (48). Tumor immune microenvironment could be adjusted by the use of ramucirumab, so that ICIs might lead to better outcomes with the adding of antiangiogenic drugs (12). Combined use of immunological agents could stimulate an anti-cancer immunological joint memory, resulting in improved response rate and prolonged duration of response (49). The activation of mTOR had been proved to increase the proliferation of tumor cells and promote their angiogenesis, which demonstrated that antiangiogenic agents could obtain better effects when accompanying with the mTOR inhibitor (7).

Based on these theories, CNA might be superior to MNA in anti-cancer effectiveness. Clinically, adding pemetrexed and carboplatin chemotherapy to gefitinib significantly prolonged PFS and OS compared with gefitinib alone in advanced non-small-cell lung cancer (50). First-line treatment with dabrafenib plus trametinib led to long-term benefit in the patients who had unresectable or metastatic melanoma (51). Additionally, notable response and survival outcomes had also been revealed in urological cancers. The combination of everolimus and bevacizumab had been suggested for the treatment of advanced non-clear cell renal cell carcinoma (52). For patients with metastatic castration-resistant prostate cancer, durvalumab plus olaparib demonstrated satisfactory efficacy of median PFS and PFS-12m (53).

It's worth noting that the pooled grade 3-4 AEs% in CNA group (63.7%) was significantly higher than that in MNA (25.4%), which might limit the application of combined regimens. Therefore, prevention measures were demanded to decrease the treatment-related adverse effects of CNA. Vitamin E and G-CSF were recommended for the prophylaxis of chemotherapy-induced peripheral neuropathy and neutropenia, respectively (54,55). And immunosuppressants were suggested when severe diarrhea and colitis occurred caused by ICIs (56). Though a lot of methods had been applied, the prevention and treatment for adverse effects generated by CNA need to be further explored.

However, several limitations in our study should be concentrated. Firstly, there was the lack of head-to-head RCTs comparing CNA and MNA, which could largely affect the quality of our study. Secondly, the heterogeneous modality of 1st line chemotherapy and ICIs might affect the outcomes despite the particularly small proportion of 1st line ICIs patients. Thirdly, we enrolled advanced UC patients treated with different second-line novel agents, which would possibly lead to a selective bias and contribute to some conflicting results in our study, such as OS-12mo and OS-24mo. Finally, though sensitivity analyses were carried out in our study, heterogeneities among studies could not be eliminated.


Conclusions

In conclusion, CNA showed significantly more effectiveness than MNA for patients with advanced UC failed to the first-line treatment. However, the treatment related toxicity of CNA must be carefully noticed. Particularly, for CNA, the regimens of ICIs could be more suitable than TDs, and the patients with PD-L1 (+) and ECOG-PS =0 would have a superior prognosis. However, our results should be further confirmed because of poor quality, publication bias, and significant heterogeneity among included studies.


Acknowledgments

We thank all the authors of the enrolled published papers for their valued contributions to the field, and Dr. Xi Li from Nuffield Department of Medicine, University of Oxford for his revision of our manuscript.

Funding: None.


Footnote

Reporting Checklist: The authors have completed the PRISMA reporting checklist. Available at http://dx.doi.org/10.21037/tcr-20-3354

Peer Review File: Available at http://dx.doi.org/10.21037/tcr-20-3354

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/tcr-20-3354). 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. von der Maase H, Hansen SW, Roberts JT, et al. Gemcitabine and cisplatin versus methotrexate, vinblastine, doxorubicin, and cisplatin in advanced or metastatic bladder cancer: results of a large, randomized, multinational, multicenter, phase III study. J Clin Oncol 2000;18:3068-77. [Crossref] [PubMed]
  2. Sternberg CN, de Mulder P, Schornagel JH, et al. Seven year update of an EORTC phase III trial of high-dose intensity M-VAC chemotherapy and G-CSF versus classic M-VAC in advanced urothelial tract tumours. Eur J Cancer 2006;42:50-4. [Crossref] [PubMed]
  3. Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med 2017;376:1015-26. [Crossref] [PubMed]
  4. Necchi A, Mariani L, Zaffaroni N, et al. Pazopanib in Advanced and Platinum-Resistant Urothelial Cancer: An Open-Label, Single Group, Phase 2 Trial. Lancet Oncol 2012;13:810-6. [Crossref] [PubMed]
  5. Sharma P, Siefker-Radtke A, de Braud F, et al. Nivolumab Alone and With Ipilimumab in Previously Treated Metastatic Urothelial Carcinoma: CheckMate 032 Nivolumab 1 mg/kg Plus Ipilimumab 3 mg/kg Expansion Cohort Results. J Clin Oncol 2019;37:1608-16. [Crossref] [PubMed]
  6. Narayanan S, Lam A, Vaishampayan U, et al. Phase II Study of Pazopanib and Paclitaxel in Patients With Refractory Urothelial Cancer. Clin Genitourin Cancer 2016;14:432-7. [Crossref] [PubMed]
  7. Bellmunt J, Lalani AA, Jacobus S, et al. Everolimus and pazopanib (E/P) benefit genomically selected patients with metastatic urothelial carcinoma. Br J Cancer 2018;119:707-12. [Crossref] [PubMed]
  8. Parikh M, Pan CX, Beckett LA, et al. Pembrolizumab Combined With Either Docetaxel or Gemcitabine in Patients With Advanced or Metastatic Platinum-Refractory Urothelial Cancer: Results From a Phase I Study. Clin Genitourin Cancer 2018;16:421-8.e1. [Crossref] [PubMed]
  9. Moher D, Liberati A, Tetzlaff J, et al. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med 2009;151:264-9. [Crossref] [PubMed]
  10. Higgins JP, Altman DG, Gøtzsche PC, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ 2011;343:d5928. [Crossref] [PubMed]
  11. Necchi A, Pond GR, Raggi D, et al. Efficacy and Safety of Gemcitabine Plus Either Taxane or Carboplatin in the First-Line Setting of Metastatic Urothelial Carcinoma: A Systematic Review and Meta-Analysis. Clin Genitourin Cancer 2017;15:23-30.e2. [Crossref] [PubMed]
  12. Petrylak DP, de Wit R, Chi KN, et al. Ramucirumab Plus Docetaxel Versus Placebo Plus Docetaxel in Patients With Locally Advanced or Metastatic Urothelial Carcinoma After Platinum-Based Therapy (RANGE): Overall Survival and Updated Results of a Randomised, Double-Blind, Phase 3 Trial. Lancet Oncol 2020;21:105-20. [Crossref] [PubMed]
  13. Wong YN, Litwin S, Vaughn D, et al. Phase II Trial of Cetuximab With or Without Paclitaxel in Patients With Advanced Urothelial Tract Carcinoma. J Clin Oncol 2012;30:3545-51. [Crossref] [PubMed]
  14. Fradet Y, Bellmunt J, Vaughn DJ, et al. Randomized phase III KEYNOTE-045 trial of pembrolizumab versus paclitaxel, docetaxel, or vinflunine in recurrent advanced urothelial cancer: results of >2 years of follow-up. Ann Oncol 2019;30:970-6. [Crossref] [PubMed]
  15. Yasuoka S, Yuasa T, Nishimura N, et al. Initial Experience of Pembrolizumab Therapy in Japanese Patients With Metastatic Urothelial Cancer. Anticancer Res 2019;39:3887-92. [Crossref] [PubMed]
  16. Tamura D, Jinnouchi N, Abe M, et al. Prognostic outcomes and safety in patients treated with pembrolizumab for advanced urothelial carcinoma: experience in real-world clinical practice. Int J Clin Oncol 2020;25:899-905. [Crossref] [PubMed]
  17. Petrylak DP, Powles T, Bellmunt J, et al. Atezolizumab (MPDL3280A) Monotherapy for Patients With Metastatic Urothelial Cancer Long-term Outcomes From a Phase 1 Study. JAMA Oncol 2018;4:537-44. [Crossref] [PubMed]
  18. Powles T, Durán I, van der Heijden MS, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2018;391:748-57. [Crossref] [PubMed]
  19. Pal SK, Hoffman-Censits J, Zheng H, et al. Atezolizumab in Platinum-treated Locally Advanced or Metastatic Urothelial Carcinoma: Clinical Experience from an Expanded Access Study in the United States. Eur Urol 2018;73:800-6. [Crossref] [PubMed]
  20. Sternberg CN, Loriot Y, James N, et al. Primary Results from SAUL, a Multinational Single-arm Safety Study of Atezolizumab Therapy for Locally Advanced or Metastatic Urothelial or Nonurothelial Carcinoma of the Urinary Tract. Eur Urol 2019;76:73-81. [Crossref] [PubMed]
  21. Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 2016;387:1909-20. [Crossref] [PubMed]
  22. Apolo AB, Infante JR, Balmanoukian A, et al. Avelumab, an Anti–Programmed Death-Ligand 1 Antibody, In Patients With Refractory Metastatic Urothelial Carcinoma: Results From a Multicenter, Phase Ib Study. J Clin Oncol 2017;35:2117-24. [Crossref] [PubMed]
  23. Patel MR, Ellerton J, Infante JR, et al. Avelumab in metastatic urothelial carcinoma after platinum failure (JAVELIN Solid Tumor): pooled results from two expansion cohorts of an open-label, phase 1 trial. Lancet Oncol 2018;19:51-64. [Crossref] [PubMed]
  24. Ohyama C, Kojima T, Kondo T, et al. Nivolumab in patients with unresectable locally advanced or metastatic urothelial carcinoma: CheckMate 275 2-year global and Japanese patient population analyses. Int J Clin Oncol 2019;24:1089-98. [Crossref] [PubMed]
  25. Powles T, O'Donnell PH, Massard C, et al. Efficacy and Safety of Durvalumab in Locally Advanced or Metastatic Urothelial Carcinoma Updated Results From a Phase 1/2 Open-label Study. JAMA Oncol 2017;3:e172411. [Crossref] [PubMed]
  26. Twardowski P, Stadler WM, Frankel P, et al. Phase II Study of Aflibercept (VEGF-Trap) in Patients With Recurrent or Metastatic Urothelial Cancer, a California Cancer Consortium Trial. Urology 2010;76:923-6. [Crossref] [PubMed]
  27. Wülfing C, Machiels JP, Richel DJ, et al. A Single-Arm, Multicenter, Open-Label Phase 2 Study of Lapatinib as the Second-Line Treatment of Patients With Locally Advanced or Metastatic Transitional Cell Carcinoma. Cancer 2009;115:2881-90. [Crossref] [PubMed]
  28. Dreicer R, Li H, Stein M, et al. Phase 2 Trial of Sorafenib in Patients With Advanced Urothelial Cancer. Cancer 2009;115:4090-5. [Crossref] [PubMed]
  29. Gallagher DJ, Milowsky MI, Gerst SR, et al. Phase II Study of Sunitinib in Patients With Metastatic Urothelial Cancer. J Clin Oncol 2010;28:1373-9. [Crossref] [PubMed]
  30. Sharma P, Sohn J, Shin SJ, et al. Efficacy and Tolerability of Tremelimumab in Locally Advanced or Metastatic Urothelial Carcinoma Patients Who Have Failed First-Line Platinum-Based Chemotherapy. Clin Cancer Res 2020;26:61-70. [Crossref] [PubMed]
  31. Pili R, Qin R, Flynn PJ, et al. A Phase II Safety and Efficacy Study of the Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitor Pazopanib in Patients With Metastatic Urothelial Cancer. Clin Genitourin Cancer 2013;11:477-83. [Crossref] [PubMed]
  32. Jones RJ, Hussain SA, Protheroe AS, et al. Randomized Phase II Study Investigating Pazopanib Versus Weekly Paclitaxel in Relapsed or Progressive Urothelial Cancer. J Clin Oncol 2017;35:1770-7. [Crossref] [PubMed]
  33. Seront E, Rottey S, Sautois B, et al. Phase II Study of Everolimus in Patients With Locally Advanced or Metastatic Transitional Cell Carcinoma of the Urothelial Tract: Clinical Activity, Molecular Response, and Biomarkers. Ann Oncol 2012;23:2663-70. [Crossref] [PubMed]
  34. Milowsky MI, Iyer G, Regazzi AM, et al. Phase II Study of Everolimus in Metastatic Urothelial Cancer. BJU Int 2013;112:462-70. [Crossref] [PubMed]
  35. Pulido M, Roubaud G, Cazeau AL, et al. Safety and Efficacy of Temsirolimus as Second Line Treatment for Patients With Recurrent Bladder Cancer. BMC Cancer 2018;18:194. [Crossref] [PubMed]
  36. Petrylak DP, Tagawa ST, Kohli M, et al. Docetaxel As Monotherapy or Combined With Ramucirumab or Icrucumab in Second-Line Treatment for Locally Advanced or Metastatic Urothelial Carcinoma: An Open-Label, Three-Arm, Randomized Controlled Phase II Trial. J Clin Oncol 2016;34:1500-9. [Crossref] [PubMed]
  37. Herbst RS, Arkenau HT, Santana-Davila R, et al. Ramucirumab plus pembrolizumab in patients with previously treated advanced non-small-cell lung cancer, gastro-oesophageal cancer, or urothelial carcinomas (JVDF): a multicohort, non-randomised, open-label, phase 1a/b trial. Lancet Oncol 2019;20:1109-23. [Crossref] [PubMed]
  38. Rosenberg JE, Hahn NM, Regan MM, et al. Apatorsen Plus Docetaxel Versus Docetaxel Alone in Platinum-Resistant Metastatic Urothelial Carcinoma (Borealis-2). Br J Cancer 2018;118:1434-41. [Crossref] [PubMed]
  39. Miyata Y, Asai A, Mitsunari K, et al. Safety and efficacy of combination therapy with low-dose gemcitabine, paclitaxel, and sorafenib in patients with cisplatin-resistant urothelial cancer. Med Oncol 2015;32:235. [Crossref] [PubMed]
  40. Choueiri TK, Ross RW, Jacobus S, et al. Double-Blind, Randomized Trial of Docetaxel Plus Vandetanib Versus Docetaxel Plus Placebo in Platinum-Pretreated Metastatic Urothelial Cancer. J Clin Oncol 2012;30:507-12. [Crossref] [PubMed]
  41. Niegisch G, Retz M, Thalgott M, et al. Second-Line Treatment of Advanced Urothelial Cancer with Paclitaxel and Everolimus in a German Phase II Trial (AUO Trial AB 35/09). Oncology 2015;89:70-8. [Crossref] [PubMed]
  42. Theodore C, Geoffrois L, Vermorken JB, et al. Multicentre EORTC study 16997: Feasibility and phase II trial of farnesyl transferase inhibitor & gemcitabine combination in salvage treatment of advanced urothelial tract cancers. Eur J Cancer 2005;41:1150-7. [Crossref] [PubMed]
  43. Shah CH, Pappot H, Agerbæk M, et al. Safety and Activity of Sorafenib in Addition to Vinflunine in Post-Platinum Metastatic Urothelial Carcinoma (Vinsor): Phase I Trial. Oncologist 2019;24:745-e213. [Crossref] [PubMed]
  44. Balar AV, Galsky MD, Rosenberg JE, et al. Atezolizumab as First-Line Treatment in Cisplatin-Ineligible Patients With Locally Advanced and Metastatic Urothelial Carcinoma: A Single-Arm, Multicentre, Phase 2 Trial. Lancet 2017;389:67-76. [Crossref] [PubMed]
  45. Boussiotis VA. Molecular and Biochemical Aspects of the PD-1 Checkpoint Pathway. N Engl J Med 2016;375:1767-78. [Crossref] [PubMed]
  46. Lawrence MS, Stojanov P, Polak P, et al. Mutational Heterogeneity in Cancer and the Search for New Cancer-Associated Genes. Nature 2013;499:214-8. [Crossref] [PubMed]
  47. Hodge JW, Garnett CT, Farsaci B, et al. Chemotherapy-induced Immunogenic Modulation of Tumor Cells Enhances Killing by Cytotoxic T Lymphocytes and Is Distinct From Immunogenic Cell Death. Int J Cancer 2013;133:624-36. [Crossref] [PubMed]
  48. Bocci G, Di Paolo A, Danesi R. The Pharmacological Bases of the Antiangiogenic Activity of Paclitaxel. Angiogenesis 2013;16:481-92. [Crossref] [PubMed]
  49. Mahoney KM, Rennert PD, Freeman GJ. Combination Cancer Immunotherapy and New Immunomodulatory Targets. Nat Rev Drug Discov 2015;14:561-84. [Crossref] [PubMed]
  50. Noronha V, Patil VM, Joshi A, et al. Gefitinib Versus Gefitinib Plus Pemetrexed and Carboplatin Chemotherapy in EGFR-Mutated Lung Cancer. J Clin Oncol 2020;38:124-36. [Crossref] [PubMed]
  51. Robert C, Grob JJ, Stroyakovskiy D, et al. Five-Year Outcomes with Dabrafenib plus Trametinib in Metastatic Melanoma. N Engl J Med 2019;381:626-36. [Crossref] [PubMed]
  52. Voss MH, Molina AM, Chen YB, et al. Phase II Trial and Correlative Genomic Analysis of Everolimus Plus Bevacizumab in Advanced Non-Clear Cell Renal Cell Carcinoma. J Clin Oncol 2016;34:3846-53. [Crossref] [PubMed]
  53. Karzai F, VanderWeele D, Madan RA, et al. Activity of durvalumab plus olaparib in metastatic castration-resistant prostate cancer in men with and without DNA damage repair mutations. J Immunother Cancer 2018;6:141. [Crossref] [PubMed]
  54. Eum S, Choi HD, Chang MJ, et al. Protective Effects of Vitamin E on Chemotherapy-Induced Peripheral Neuropathy: A Meta-Analysis of Randomized Controlled Trials. Int J Vitam Nutr Res 2013;83:101-11. [Crossref] [PubMed]
  55. Shikata H, Yakushijin Y, Yamanouchi J, et al. Analysis of Chemotherapy-Induced Neutropenia and Optimal Timing for Prophylactic Use of G-CSF in B-cell non-Hodgkin Lymphoma Patients Treated With R-CHOP. Int J Clin Oncol 2014;19:178-85. [Crossref] [PubMed]
  56. Tian Y, Abu-Sbeih H, Wang Y. Checkpoint Inhibitors-Induced Colitis. Adv Exp Med Biol 2018;995:151-7. [Crossref] [PubMed]
Cite this article as: Wei L, Gao L, Hu Z, Liu C. Effectiveness and safety of combined therapy versus monotherapy based on immune checkpoint inhibitors and/or targeted drugs as salvage treatment for advanced urothelial carcinoma: a systematic review and meta-analysis. Transl Cancer Res 2021;10(5):2091-2107. doi: 10.21037/tcr-20-3354

Download Citation