ArticlePDF Available

A case of neoadjuvant chemotherapy‐resistant muscle‐invasive bladder cancer that markedly responded to pembrolizumab before conversion radical cystectomy

Wiley
IJU Case Reports
Authors:

Abstract and Figures

Introduction Recently, perioperative use of immune checkpoint inhibitors has improved the prognosis of muscle‐invasive bladder cancer. It is unclear whether radical cystectomy or systemic pembrolizumab is the optimal next treatment in patients with muscle‐invasive bladder cancer and progressive disease in the pelvic lymph node following neoadjuvant chemotherapy (NAC). Case presentation A 62‐year‐old woman with cT3N0M0 bladder cancer and high programmed death‐ligand 1 expression developed solitary obturator lymph node metastasis following 5 cycles of neoadjuvant chemotherapy. Six subsequent cycles of pembrolizumab shrank the lymph node significantly, and conversion radical cystectomy was planned. Pathologically, only carcinoma in situ around the scar of transurethral resection of bladder tumor remained in the primary tumor, and the accumulation of foamy macrophages and fibrosis without viable tumor cells was observed in the dissected lymph node. Eighteen months passed without a recurrence following radical cystectomy. Conclusion Pembrolizumab administration before radical cystectomy achieved a good response in a patient with obturator lymph node metastasis following neoadjuvant chemotherapy.
Content may be subject to copyright.
Case Report
A case of neoadjuvant chemotherapy-resistant muscle-invasive
bladder cancer that markedly responded to pembrolizumab before
conversion radical cystectomy
Ichiro Yonese,
1
Noboru Numao,
1
Kentaro Inamura,
2
Yusuke Yoneoka,
1
Ryo Fujiwara,
1
Yosuke Yasuda,
1
Tomohiko Oguchi,
1
Shinya Yamamoto,
1
Takeshi Yuasa
1
and Junji Yonese
1
Departments of
1
Urology, Cancer Institutes Hospital and
2
Pathology, Cancer Institutes Hospital, Japanese Foundation for Cancer
Research, Tokyo, Japan
Abbreviations & Acronyms
cis = carcinoma in situ
CRP = C-reactive protein
CT = computed tomography
GC = gemcitabine and cisplatin
Hb = hemoglobin
ICI = immune checkpoint inhibitor
LDH = lactate dehydrogenase
LN = lymph node
LNM = lymph node metastasis
MIBC = muscle-invasive bladder
cancer
MRI = magnetic resonance imaging
NAC = neoadjuvant chemotherapy
NLR = neutrophil lymphocyte ratio
pCR = pathological complete response
PD-L1 = programmed death-ligand 1
PLND = pelvic lymph node dissection
PLT = platelet
RARC = robot-assisted radical
cystectomy
RC = radical cystectomy
SII = systemic immune-inammation
index
TURBT = transurethral resection of
bladder tumor
WBC = white blood cell
c-GTP = c-glutamyltranspeptidase
Correspondence: Noboru Numao M.D.,
Department of Urology, Cancer Institute
Hospital, Japanese Foundation for Cancer
Research, 3-8-31 Ariake, Koto-ku, Tokyo
135-0063, Japan. Email: noboru.numao@
jfcr.or.jp
How to cite this article: Yonese I, Numao
N, Inamura K et al. A case of neoadjuvant
chemotherapy-resistant muscle-invasive
bladder cancer that markedly responded to
pembrolizumab before conversion radical
cystectomy. IJU Case Rep. 2023; https://
doi.org/10.1002/iju5.12669.
This is an open access article under the
terms of the Creative Commons Attribution
License, which permits use, distribution
and reproduction in any medium, provided
the original work is properly cited.
Received 30 August 2023; accepted 1
November 2023.
Introduction: Recently, perioperative use of immune checkpoint inhibitors has
improved the prognosis of muscle-invasive bladder cancer. It is unclear whether radical
cystectomy or systemic pembrolizumab is the optimal next treatment in patients with
muscle-invasive bladder cancer and progressive disease in the pelvic lymph node
following neoadjuvant chemotherapy (NAC).
Case presentation: A 62-year-old woman with cT3N0M0 bladder cancer and high
programmed death-ligand 1 expression developed solitary obturator lymph node
metastasis following 5 cycles of neoadjuvant chemotherapy. Six subsequent cycles of
pembrolizumab shrank the lymph node significantly, and conversion radical cystectomy
was planned. Pathologically, only carcinoma in situ around the scar of transurethral
resection of bladder tumor remained in the primary tumor, and the accumulation of
foamy macrophages and fibrosis without viable tumor cells was observed in the
dissected lymph node. Eighteen months passed without a recurrence following radical
cystectomy.
Conclusion: Pembrolizumab administration before radical cystectomy achieved a good
response in a patient with obturator lymph node metastasis following neoadjuvant
chemotherapy.
Key words: bladder cancer, conversion surgery, neoadjuvant chemotherapy,
neoadjuvant immunotherapy.
Keynote message
It is unclear whether RC or systemic pembrolizumab is the better next treatment in patients
with MIBC that has progressed in the pelvic LN following NAC. In such cases, pembrolizu-
mab with a view to converting RC could be an option.
Introduction
NAC before RC and PLND demonstrated prolonged survival in localized MIBC
13
and is
recommended as standard treatment.
4
However, in a minority of cases, MIBC showed pelvic
LNM following NAC.
5
It is unclear whether RC or systemic immunotherapy is more effective
for these cases.
Before the advent of ICIs, the prognosis was poor for patients with residual MIBC or
LNM after NAC.
6,7
Recently, perioperative use of ICIs improved the prognosis of MIBC.
8,9
Adjuvant nivolumab was shown to improve the prognosis of residual MIBC or LNM after
NAC in the phase III CheckMate 274 trial.
8
The effectiveness of neoadjuvant immunotherapy
was also reported in the phase II PURE-01 trial.
9
However, we could nd no reports describ-
ing systematic immunotherapy with a view to conversion RC in MIBC patients with pelvic
LNM after NAC. Here, we report a patient with cT3N0M0 bladder cancer developing obtura-
tor LNM following NAC who markedly responded to pembrolizumab before conversion RC
with PLND.
©2023 The Authors. IJU Case Reports published by John Wiley & Sons Australia, Ltd on behalf of Japanese Urological Association. 1
IJU Case Reports (2023) doi: 10.1002/iju5.12669
Case presentation
A 62-year-old woman visited her doctor because of gross
hematuria in 2021. Urine cytology revealed the presence of
atypical cells. Cystoscopy and MRI revealed a nodal tumor
of VI-RADS score 2 in the left lateral wall (Fig. 1). TURBT
revealed pT2 and G3 urothelial carcinoma with cis (Fig. 2a).
Contrast-enhanced CT showed no metastasis or upper urinary
tract tumor. Four cycles of neoadjuvant GC (gemcitabine;
100 mg/m
2
Days 1, 8, 15, and cisplatin; 70 mg/m
2
Day 2)
every 4 weeks were planned and started at 80% dose; the
fourth cycle was reduced to 70% due to myelosuppression.
The patient was referred to us for RARC. We resumed the
fth cycle of GC at a 70% dose because of the waiting
period before RARC. However, after the fth cycle, a preop-
erative CT revealed a solitary left obturator LNM (30 mm)
with uorodeoxyglucose uptake in positron emission
tomography-CT (ycT3N1). We considered this case to be
associated with a high recurrence rate after RC, and the dis-
ease was unable to be resected with curative intent. We
began treatment with pembrolizumab 200 mg every 3 weeks
with a view to conversion RC in case the LNM shrunk.
Three weeks after the rst administration of pembrolizumab,
the LNM (16 mm) had shrank markedly. After 6 cycles, the
LNM had shrunk to an insignicant size, and we planned
conversion RC. NLR and SII (NLR 9PLT count [910
3
/
lL])
10
were decreasing concurrently with shrinkage of the
LNM (Fig. 3). No immune-related adverse effects were
observed.
RARC with PLND and extracorporeal urinary diversion
were performed in 2022. Total operative time and console time
were 447 and 272 min, respectively. The estimated blood loss
during surgery was 200 mL. Pathologically, only the cis
around the scar of TURBT remained in the primary tumor. The
dissected obturator LN showed an accumulation of foamy mac-
rophages and brosis without viable tumor cells (Fig. 2c), sug-
gesting a good therapeutic effect. Immunohistochemical
evaluation using anti- PD-L1 monoclonal antibody (clone 288)
and the TURBT specimen showed PD-L1 expression in the
majority (60%) of tumor cells (Fig. 2b). No systemic therapy
was added following RC. Eighteen months passed without a
recurrence.
Discussion
To the best of our knowledge, there are no reports of preop-
erative pembrolizumab administration for progressive MIBC
following NAC. One report compared pCR to pembrolizumab
for chemotherapy-resistant upper tract urothelial carcinoma.
11
In the present case, the LNM following neoadjuvant GC dis-
appeared with pembrolizumab, which made it possible to per-
form conversion RC.
It is unclear whether RC or systemic pembrolizumab is
better as the next treatment in MIBC patients with pelvic
LNM following NAC. In the former cases, the maximal sur-
vival time of 32 ypN+patients after RC was 3.17 years.
7
Another study reported that 5-year overall survival of patients
with residual cancer (ypT2-4 or ypN+) after NAC was 21%,
whereas that of cases with pCR was 82%.
12
Occult LNM
after RC also showed a poor prognosis.
13,14
In the PURE-01
study of neoadjuvant pembrolizumab, all MIBC patients
enrolled in the study underwent RC, and 42% of patients
achieved pCR.
9
Considering the poor prognosis of ypN+
cases after NAC and RC, systemic immunotherapy with a
view to converting RC may increase the possibility of cure.
In our patient, after pembrolizumab administration, adjuvant
nivolumab was covered by Japanese insurance. Subgroup
analysis of the CheckMate 274 trial demonstrated the advan-
tage of adjuvant nivolumab in pN+cases.
8
The treatment
sequence combining RC and systemic immunotherapy for
chemotherapy-resistant cases is an issue yet to be resolved. In
the present case, pembrolizumab before RC was effective.
Systemic immunotherapy before RC could lead to further
advancement and a missed opportunity for RC. To estimate
Fig. 1 MRI images of cT3N0M0 bladder cancer
before TURBT: (a) sagittal view of the T2-WI
image; (b) axial view of the T2-WI image; (c) axial
view of the DWI image; (d) axial view of the
contrast-enhanced image.
2©2023 The Authors. IJU Case Reports published by John Wiley & Sons Australia, Ltd on behalf of Japanese Urological Association.
I YONESE ET AL.
the efcacy of ICIs, PD-L1 expression demonstrated predic-
tive value in several clinical trials and meta-analyses.
8,9,15,16
Neoadjuvant pembrolizumab achieved pCR in 54.3% of cases
with high PD-L1 expression, compared with 13.3% of cases
with low PD-L1 expression.
9
In the present case, the primary
tumor showed high PD-L1 expression. Assessment of PD-L1
expression could be useful in determining the treatment of
progressive MIBC after NAC.
Fig. 2 Microscopic examination of the TURBT specimen of the primary tumor showed pT2 and G3 urothelial carcinoma in hematoxylin and eosin staining (a) with
PD-L1 expression in the majority of tumor cells (b). The dissected obturator LN showed an accumulation of foamy macrophages and fibrosis without viable tumor
cells (c).
Fig. 3 Time-course of metastatic obturator LN (a) with prognostic factors associated with metastatic urothelial carcinoma (b, c). NLR and SII represent neutrophil/
lymphocyte and PLT 9neutrophil/lymphocyte, respectively. NLR and SII improved concurrently with the shrinkage of the metastatic LN (b).
©2023 The Authors. IJU Case Reports published by John Wiley & Sons Australia, Ltd on behalf of Japanese Urological Association. 3
Pembrolizumab before radical cystectomy
A recent risk stratication of metastatic urothelial cancer
indicated that NLR, Hb, LDH, and CRP were predictive fac-
tors of response to pembrolizumab.
17,18
SII and c-GTP were
shown to be prognostic factors for urothelial carcinoma.
10,19
In the present case, NLR and SII were improving concur-
rently with the shrinkage of the LNM (Fig. 3). These factors
could be useful in assessing the effect of neoadjuvant therapy
before conversion surgery.
Conclusions
Pembrolizumab administration in advance of conversion RC
achieved a good response in a patient with obturator LNM
after NAC.
Author contributions
Ichiro Yonese: Conceptualization; investigation; visualization;
writing original draft. Noboru Numao: Conceptualization;
investigation; supervision; visualization; writing review and
editing. Kentaro Inamura: Investigation; visualization; writing
review and editing. Yusuke Yoneoka: Writing review and
editing. Ryo Fujiwara: Writing review and editing. Yosuke
Yasuda: Writing review and editing. Tomohiko Oguchi:
Writing review and editing. Shinya Yamamoto: Writing
review and editing. Takeshi Yuasa: Writing review and
editing. Junji Yonese: Writing review and editing.
Conflict of interest
Author T.Y. was supported by grants from MSD.
Approval of the research protocol by
an Institutional Reviewer Board
Not applicable.
Informed consent
Informed consent was obtained from the patient for publica-
tion of this case report.
Registry and the Registration No. of
the study/trial
Not applicable.
References
1 Vale C, Advanced Bladder Cancer Meta-analysis Collaboration. Neoadjuvant
chemotherapy in invasive bladder cancer: a systematic review and meta-
analysis. Lancet 2003; 361: 192734.
2 Grossman HB, Natale RB, Tangen CM et al. Neoadjuvant chemotherapy
plus cystectomy compared with cystectomy alone for locally advanced blad-
der cancer. N. Engl. J. Med. 2003; 349: 85966.
3 Hinata N, Hussein AA, George S et al. Impact of suboptimal neoadjuvant
chemotherapy on peri-operative outcomes and survival after robot-assisted
radical cystectomy: a multicentre multinational study. BJU Int. 2017; 119:
60511.
4 Witjes JA, Rink M, Van der Heiden AG et al.EAU Guidelines on Muscle-
Invasive and Metastatic Bladder Cancer EAU Guidelines Ofce, Arnhem,
2023.
5 Zargar H, Espiritu PN, Fairey AS et al. Multicenter assessment of neoadju-
vant chemotherapy for muscle-invasive bladder cancer. Eur. Urol. 2015; 67:
2419.
6 Manoharan M, Katkoori D, Kishore TA, Kava B, Singal R, Soloway MS.
Outcome after radical cystectomy in patients with clinical T2 bladder cancer
in whom neoadjuvant chemotherapy has failed. BJU Int. 2009; 104: 16469.
7 Iyer G, Tully CM, Zabor EC et al. Neoadjuvant gemcitabine-cisplatin plus
radical cystectomy-pelvic lymph node dissection for muscle-invasive bladder
cancer: a 12-year experience. Clin. Genitourin. Cancer 2020; 18: 38794.
8 Bajorin DF, Witjes JA, Gschwend JE et al. Adjuvant nivolumab versus pla-
cebo in muscle-invasive urothelial carcinoma. N. Engl. J. Med. 2021; 384:
210214.
9 Necchi A, Anichini A, Raggi D et al. Pembrolizumab as neoadjuvant therapy
before radical cystectomy in patients with muscle-invasive urothelial bladder
carcinoma (PURE-01): an open-label, single-arm, phase II study. J. Clin.
Oncol. 2018; 36: 335360.
10 Kobayashi S, Ito M, Takemura K, Suzuki H, Yonese I, Koga F. Preoperative
models incorporating the systemic immune-inammation index for predicting
prognosis and muscle invasion in patients with non-metastatic upper tract
urothelial carcinoma. Int. J. Clin. Oncol. 2022; 27: 57484.
11 Ikarashi D, Kitano S, Ishida K et al. Complete pathological response to
neoadjuvant pembrolizumab in a patient with chemoresistant upper urinary
tract urothelial carcinoma: a case report. Front. Oncol. 2020; 10: 564714.
12 Bhindi B, Frank I, Mason RJ et al. Oncologic outcomes for patients with
residual cancer at cystectomy following neoadjuvant chemotherapy: a patho-
logic stage-matched analysis. Eur. Urol. 2017; 72: 6604.
13 Cha EK, Sfakianos JP, Sukhu R, Yee AM, Sjoberg DD, Bochner BH. Poor
prognosis of bladder cancer patients with occult lymph node metastases trea-
ted with neoadjuvant chemotherapy. BJU Int. 2018; 122: 62732.
14 van Hoogstraten LMC, van Gennep EJ, Kiemeney LALM et al. Occult
lymph node metastases in patients without residual muscle-invasive bladder
cancer at radical cystectomy with or without neoadjuvant chemotherapy: a
nationwide study of 5417 patients. World J. Urol. 2022; 40: 1118.
15 Bellmunt J, de Wit R, Vaughn DJ et al. KEYNOTE-045 investigators. Pem-
brolizumab as second-line therapy for advanced urothelial carcinoma. N.
Engl. J. Med. 2017; 376: 101526.
16 Rizzo A, Mollica V, Massari F. Expression of programmed cell death ligand
1 as a predictive biomarker in metastatic urothelial carcinoma patients treated
with rst-line immune checkpoint inhibitors versus chemotherapy: a system-
atic review and meta-analysis. Eur. Urol. Focus 2022; 8: 1529.
17 Kobayashi T, Ito K, Kojima T et al. Risk stratication for the prognosis of
patients with chemoresistant urothelial cancer treated with pembrolizumab.
Cancer Sci. 2021; 112: 76073.
18 Fujiwara M, Fujiwara R, Urasaki T et al. Early serum and hematological
responses to pembrolizumab therapy as predictors of survival in metastatic
urothelial cancer. Anticancer Res 2022; 42: 204551.
19 Takemura K, Fukushima H, Ito M et al. Prognostic signicance of serum
gamma-glutamyltransferase in patients with advanced urothelial carcinoma.
Urol. Oncol. 2019; 37: 10815.
4©2023 The Authors. IJU Case Reports published by John Wiley & Sons Australia, Ltd on behalf of Japanese Urological Association.
I YONESE ET AL.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Purpose Little is known about the prevalence of occult lymph node metastases (LNM) in muscle-invasive bladder cancer (MIBC) patients with pathological downstaging of the primary tumor. We aimed to estimate the prevalence of occult LNM in patients without residual MIBC at radical cystectomy (RC) with or without neoadjuvant chemotherapy (NAC) or neoadjuvant radiotherapy (NAR), and to assess overall survival (OS). Methods Patients with cT2-T4aN0M0 urothelial MIBC who underwent RC plus pelvic lymph node dissection (PLND) with curative intent between January 1995–December 2013 (retrospective Netherlands Cancer Registry (NCR) cohort) and November 2017–October 2019 (prospective NCR-BlaZIB cohort (acronym in Dutch: Bla askankerZorg I n B eeld; in English: Insight into bladder cancer care)) were identified from the nationwide NCR. The prevalence of occult LNM was calculated and OS of patients with <( y )pT2N0 vs. <( y )pT2N+ disease was estimated by the Kaplan–Meier method. Results In total, 4657 patients from the NCR cohort and 760 patients from the NCR-BlaZIB cohort were included. Of 1374 patients downstaged to <( y )pT2, 4.3% ( N = 59) had occult LNM 4.1% ( N = 49) of patients with cT2-disease and 5.6% ( N = 10) with cT3-4a-disease. This was 4.0% ( N = 44) in patients without NAC or NAR, 4.5% ( N = 10) in patients with NAC, and 13.5% ( N = 5) in patients with NAR but number of patients treated with NAR and downstaged disease was small. The prevalence of <( y )pT2N+ disease was 4.2% ( N = 48) in the NCR cohort and 4.6% ( N = 11) in the NCR-BlaZIB cohort. For patients with <( y )pT2N+ and <( y )pT2N0, median OS was 3.5 years (95% CI 2.5–8.9) versus 12.9 years (95% CI 11.7–14.0), respectively. Conclusion Occult LNM were found in 4.3% of patients with cT2-4aN0M0 MIBC with (near-) complete downstaging of the primary tumor following RC plus PLND. This was regardless of NAC or clinical T-stage. Patients with occult LNM showed considerable worse survival. These results can help in counseling patients for bladder-sparing treatments.
Article
Full-text available
The use of immune checkpoint inhibitors to treat urothelial cancer (UC) is increasing rapidly without clear guidance for validated risk stratification. This multicenter retrospective study collected clinicopathological information on 463 patients, and 11 predefined variables were analyzed to develop a multivariate model predicting overall survival (OS). The model was validated using an independent dataset of 292 patients. Patient characteristics and outcomes were well balanced between the discovery and validation cohorts, which had median OS times of 10.2 and 12.5 months, respectively. The final validated multivariate model was defined by risk scores based on the hazard ratios (HRs) of independent prognostic factors including performance status, site of metastasis, hemoglobin levels, and the neutrophil‐to‐lymphocyte ratio. The median OS times (95% confidence intervals [CIs]) for the low‐, intermediate‐, and high‐risk groups (discovery cohort) were not yet reached (NYR) (NYR–19.1), 6.8 months (5.8–8.9), and 2.3 months (1.2–2.6), respectively. The HRs (95% CI) for OS in the low‐ and intermediate‐risk groups versus the high‐risk group were 0.07 (0.04–0.11) and 0.23 (0.15–0.37), respectively. The objective response rates for in the low‐, intermediate‐, and high‐risk groups were 48.3%, 28.8%, and 10.5%, respectively. These differential outcomes were well reproduced in the validation cohort and in patients who received pembrolizumab after perioperative or first‐line chemotherapy (N = 584). In conclusion, the present study developed and validated a simple prognostic model predicting the oncological outcomes of pembrolizumab‐treated patients with chemoresistant UC. The model provides useful information for external validation, patient counseling, and clinical trial design.
Article
Full-text available
Treatment options as second-line therapy for advanced ureteral carcinoma are limited, and patients experiencing recurrence after first-line cisplatin-based chemotherapy have a poor prognosis. Recently, the programmed death-1 (PD-1) inhibitor pembrolizumab provided a better survival benefit with a complete response rate (9.2%) for chemoresistatant urothelial carcinoma. However, the dynamic changes of the cancer microenvironment about the cases of complete response are still unknown. We herein report a case of a 57-year-old man who had been diagnosed with localized, non-muscle-invasive bladder cancer (pT1N0M0, high grade), for which he underwent transurethral resection of the bladder cancer twice. Given that gemcitabine plus carboplatin as first-line neoadjuvant chemotherapy was unable to control left vesico-ureteral junction recurrence with muscle invasion (T3N0M0, high grade), the patient received the PD-1 inhibitor pembrolizumab as second-line neoadjuvant therapy in an attempt to stop tumor growth, which promoted dramatic tumor shrinkage without serious adverse effects and allowed subsequent nephroureterectomy and lymphadenectomy. To the best of our knowledge, this has been the first study to report that pembrolizumab administration before surgery for chemotherapy-resistant ureteral carcinoma promoted a pathological complete response, providing a better understanding of the cancer microenvironment after immunotherapy.
Article
Full-text available
Purpose: To determine the activity of pembrolizumab as neoadjuvant immunotherapy before radical cystectomy (RC) for muscle-invasive bladder carcinoma (MIBC) for which standard cisplatin-based chemotherapy is poorly used. Patients and methods: In the PURE-01 study, patients had a predominant urothelial carcinoma histology and clinical (c)T≤3bN0 stage tumor. They received three cycles of pembrolizumab 200 mg every 3 weeks before RC. The primary end point in the intention-to-treat population was pathologic complete response (pT0). Biomarker analyses included programmed death-ligand 1 (PD-L1) expression using the combined positive score (CPS; Dako 22C3 pharmDx assay), genomic sequencing (FoundationONE assay), and an immune gene expression assay. Results: Fifty patients were enrolled from February 2017 to March 2018. Twenty-seven patients (54%) had cT3 tumor, 21 (42%) cT2 tumor, and two (4%) cT2-3N1 tumor. One patient (2%) experienced a grade 3 transaminase increase and discontinued pembrolizumab. All patients underwent RC; there were 21 patients with pT0 (42%; 95% CI, 28.2% to 56.8%). As a secondary end point, downstaging to pT<2 was achieved in 27 patients (54%; 95% CI, 39.3% to 68.2%). In 54.3% of patients with PD-L1 CPS ≥ 10% (n = 35), RC indicated pT0, whereas RC indicated pT0 in only 13.3% of those with CPS < 10% (n = 15). A significant nonlinear association between tumor mutation burden (TMB) and pT0 was observed, with a cutoff at 15 mutations/Mb. Expression of several genes in pretherapy lesions was significantly different between pT0 and non-pT0 cohorts. Significant post-therapy changes in the TMB and evidence of adaptive mechanisms of immune resistance were observed in residual tumors. Conclusion: Neoadjuvant pembrolizumab resulted in 42% of patients with pT0 and was safely administered in patients with MIBC. This study indicates that pembrolizumab could be a worthwhile neoadjuvant therapy for the treatment of MIBC when limited to patients with PD-L1-positive or high-TMB tumors.
Article
Background: In order to search for predictive biomarkers of efficacy of pembrolizumab therapy for metastatic urothelial cancer (UC), we investigated the relationship between treatment outcomes and early neutrophil-lymphocyte ratio (NLR), lactate dehydrogenase (LDH), and C-reactive protein (CRP) responses. Patients and methods: Medical records of 101 patients with metastatic UC who started pembrolizumab as a second-line or later treatment were reviewed. NLR, LDH, and CRP were recorded after 3 weeks of therapy. In addition, we investigated whether these factors had an association with prolonged progression-free (PFS) or overall (OS) survival. Results: The objective response rate, median PFS, and median OS were 25.7%, 6.3 months, and 15.2 months, respectively. PFS and OS were significantly shorter in patients with NLR>3, LDH>upper limit of normal (ULN), and CRP>0.5 mg/dl after 3 weeks of pembrolizumab treatment (p<0.05). A predictive model comprising these factors (favorable risk group: 0 risk factors; intermediate-risk group: 1-2 risk factors; poor-risk group: 3 risk factors) revealed distinct PFS and OS curves (p<0.001). In the favorable risk group, 12-month OS was 79.6%; in the poor-risk group, it was 12.8%. Harrell's C-indices for NLR >3, LDH >ULN, CRP >0.5 mg/dl, and all three combined for predicting OS were 0.656, 0.625, 0.633 and 0.678, respectively. Early responses were also non-significantly associated with ORR (p=0.37). Conclusion: Pembrolizumab treatment outcomes are associated with early NLR, LDH, and CRP responses in metastatic urothelial cancer.
Article
Objective To develop preoperative models as a guide to indications for neoadjuvant chemotherapy (NAC) and regional lymph node dissection (LND) before and at radical nephroureterectomy (RNU), respectively, in patients with non-metastatic upper tract urothelial carcinoma (UTUC) by incorporating the systemic immune-inflammation index (SII).Methods This retrospective study enrolled 103 consecutive patients with UTUC undergoing RNU. The SII was calculated as neutrophils × platelets / lymphocytes. Multivariable Cox proportional hazard model was used to develop preoperative models for cancer-specific survival (CSS) and overall survival (OS). A model for predicting muscle invasion was developed using logistic regression analysis. Harrell's concordance-index (c-index) or the area under the receiver operating characteristic curve (AUC) was used to evaluate the accuracy of the models.ResultsDuring follow-up (median: 41 months), 26 and three patients died of UTUC and other causes, respectively. Performance status > 0, clinical tumor (cT) stage ≥ 3, and SII > 520 were independent adverse prognosticators for CSS, and one point was assigned to each prognosticator. Risk score models comprising the sum of the points stratified patients into three risk groups (0, 1, and 2–3; P < 0.001 for CSS and OS) with respective c-indices of 0.843 and 0.820. SII > 677 and ≥ cT3 were independently associated with muscle invasion. A model based on these variables predicted muscle invasion with AUC of 0.804.Conclusion Preoperative SII is significantly associated with worse survival outcomes and muscle invasion in patients with non-metastatic UTUC. Our preoperative predictive models may serve as a guide to indications for NAC and LND.
Article
Background The role of adjuvant treatment in high-risk muscle-invasive urothelial carcinoma after radical surgery is not clear. Methods Download a PDF of the Research Summary. In a phase 3, multicenter, double-blind, randomized, controlled trial, we assigned patients with muscle-invasive urothelial carcinoma who had undergone radical surgery to receive, in a 1:1 ratio, either nivolumab (240 mg intravenously) or placebo every 2 weeks for up to 1 year. Neoadjuvant cisplatin-based chemotherapy before trial entry was allowed. The primary end points were disease-free survival among all the patients (intention-to-treat population) and among patients with a tumor programmed death ligand 1 (PD-L1) expression level of 1% or more. Survival free from recurrence outside the urothelial tract was a secondary end point. Results A total of 353 patients were assigned to receive nivolumab and 356 to receive placebo. The median disease-free survival in the intention-to-treat population was 20.8 months (95% confidence interval [CI], 16.5 to 27.6) with nivolumab and 10.8 months (95% CI, 8.3 to 13.9) with placebo. The percentage of patients who were alive and disease-free at 6 months was 74.9% with nivolumab and 60.3% with placebo (hazard ratio for disease recurrence or death, 0.70; 98.22% CI, 0.55 to 0.90; P<0.001). Among patients with a PD-L1 expression level of 1% or more, the percentage of patients was 74.5% and 55.7%, respectively (hazard ratio, 0.55; 98.72% CI, 0.35 to 0.85; P<0.001). The median survival free from recurrence outside the urothelial tract in the intention-to-treat population was 22.9 months (95% CI, 19.2 to 33.4) with nivolumab and 13.7 months (95% CI, 8.4 to 20.3) with placebo. The percentage of patients who were alive and free from recurrence outside the urothelial tract at 6 months was 77.0% with nivolumab and 62.7% with placebo (hazard ratio for recurrence outside the urothelial tract or death, 0.72; 95% CI, 0.59 to 0.89). Among patients with a PD-L1 expression level of 1% or more, the percentage of patients was 75.3% and 56.7%, respectively (hazard ratio, 0.55; 95% CI, 0.39 to 0.79). Treatment-related adverse events of grade 3 or higher occurred in 17.9% of the nivolumab group and 7.2% of the placebo group. Two treatment-related deaths due to pneumonitis were noted in the nivolumab group. Conclusions In this trial involving patients with high-risk muscle-invasive urothelial carcinoma who had undergone radical surgery, disease-free survival was longer with adjuvant nivolumab than with placebo in the intention-to-treat population and among patients with a PD-L1 expression level of 1% or more. (Funded by Bristol Myers Squibb and Ono Pharmaceutical; CheckMate 274 ClinicalTrials.gov number, NCT02632409.) QUICK TAKE VIDEO SUMMARY Nivolumab vs. Placebo in Muscle-Invasive Urothelial Carcinoma 02:22
Article
Context Immune checkpoint inhibitors (ICIs) have reported durable responses in selected groups of patients with metastatic urothelial carcinoma (mUC). However, identification of biomarkers predictive of response to ICIs remains an unmet need in mUC management, and the role of programmed cell death ligand 1 (PD-L1) expression remains controversial. Objective In this systematic review and meta-analysis, we aimed at assessing the predictive value of PD-L1 in mUC patients receiving first-line ICIs as monotherapy or in combination with other anticancer agents across randomized controlled clinical trials (RCTs). Evidence acquisition We retrieved all the relevant RCTs through PubMed/Medline, Cochrane library, and EMBASE; proceedings of the main international oncological meetings were also searched for relevant abstracts. Eligible studies included RCTs evaluating ICIs versus chemotherapy in PD-L1–positive mUCs; the primary endpoint was overall survival (OS). Evidence synthesis Three phase III trials matched our eligibility criteria; a total of 2237 PD-L1–positive mUC patients (ICIs: 1125; chemotherapy: 1112) were included in the analysis. Compared with chemotherapy, ICIs were associated with higher OS in PD-L1–positive patients (hazard ratio [HR] 0.86, 95% confidence interval [CI] 0.78–0.96; p = 0.007); conversely, no differences were observed in the PD-L1–negative patient population (HR 1.03, 95% CI 0.89–1.19; p < 0.001). Conclusions Compared with standard chemotherapy, our results suggested a survival benefit in PD-L1–positive mUC patients receiving ICIs; conversely, no benefit was observed in patients with PD-L1–negative disease. Although this systematic review and meta-analysis should be interpreted with caution due to several issues, our results highlight the need for reliable predictive biomarkers of response to ICIs, providing a benchmark for future studies in this setting. Patient summary Despite immune checkpoint inhibitors (ICIs) having revolutionized the treatment landscape of metastatic urothelial carcinoma (mUC), an important percentage of patients do not experience clinical benefit or durable responses. Identification of reliable biomarkers of response to ICIs remains a mandatory need in mUC management, and further efforts are needed to standardize the assessment of programmed cell death ligand 1 in urothelial carcinoma.
Article
Introduction To determine drug delivery/toxicity, and pathological/surgical outcomes of muscle-invasive bladder cancer (MIBC) patients receiving neoadjuvant gemcitabine-cisplatin (GC) plus radical cystectomy-pelvic lymph node dissection (RC-PLND). Patients and Methods Chemotherapy and surgical/pathologic outcomes were retrospectively analyzed with 5-year survival follow-up at a referral center. Post-neoadjuvant chemotherapy (NAC) pathologic endpoints included complete response (pT0N0), residual non-MIBC (pTa/Tis/T1N0) and ≥MIBC (≥pT2 and/or N+). Associations of pathologic/surgical findings with overall survival (OS), disease-free survival (DFS), and surgical management with RC-PLND were analyzed (Cox regression). Results Clinical T2a-T4aN0M0 MIBC patients (154) from 1/2000-10/2012 received GC plus RC-PLND. Patients (117, 76%) received GCx4 and 136 (88%) GCx3. Five-year OS was 61% (95% CI 53-71). Median number of resected lymph nodes (LN) was 19. Down-staging was observed as follows: pT0N0: 21%; pTa/Tis/T1N0: 25%, with similar 5-yr OS (85% and 89%, respectively). Five-year OS for pT2 residual disease was 87% (95% CI, 78%-98%) vs. 38% (95% CI, 27%-53%); p<0.001. Post-NAC stage ≥pT2 (HR 6.79; 95% CI 2.63-17.53; p<0.001), positive LN (HR 3.64; 95% CI 1.84-7.19; p<0.001) and positive margins (HR 4.15; 95% CI 1.68-10.25; p=0.002) were associated with increased risk of all-cause death (multivariable analysis). A hazard ratio of 0.97 (95% CI: 0.94-1.00) was observed for each additional node removed, but this effect was not statistically significant (p=0.056). Conclusions Neoadjuvant GC achieves meaningful pathologic responses. Patients with ≥pT2 residual disease, positive margins, or positive LN post-chemotherapy have inferior survival.
Article
Objectives Serum γ-glutamyltransferase (GGT) is reportedly associated with prognosis in patients with various malignancies. However, the prognostic role of GGT is unknown among patients with advanced urothelial carcinoma (aUC). This study was designed to examine the prognostic role of serum GGT in patients with aUC. Materials and methods Charts of 125 consecutive aUC patients (inoperable cT4 and/or metastasis to lymph nodes/distant organs) managed at a single cancer center between 2004 and 2016 were retrospectively reviewed. Variables collected included age, sex, body mass index, Karnofsky performance status, primary site, clinical tumor stage, lymph node/visceral metastasis, hepatic comorbidities, the presence of curative treatment before the diagnosis of aUC, white blood cell count, neutrophil-to-lymphocyte ratio, hemoglobin, albumin, lactate dehydrogenase, alkaline phosphatase, GGT, C-reactive protein, and treatments given after the diagnosis of aUC. Associations of variables with overall survival (OS) were analyzed using the Cox proportional hazard model. Results Serum GGT was elevated (≥60 U/l) at the diagnosis of aUC in 16 patients (13%). During follow-up period (median 12.1 months), 101 patients died (2-year OS rate, 32%). Patients with elevated GGT at the diagnosis of aUC had a significantly poorer prognosis than those with normal GGT with respective 2-year OS rates of 0% and 37% (P < 0.001). On multivariate analysis, elevated GGT was a significant and independent risk factor for shorter OS (hazard ratio, HR = 2.97; P < 0.001) as were poorer Karnofsky performance status (HR = 3.47; P < 0.001), elevated lactate dehydrogenase (HR = 1.86; P = 0.033), advanced age (HR = 1.82; P = 0.013), elevated neutrophil-to-lymphocyte ratio (HR = 1.80; P = 0.015), elevated C-reactive protein (HR = 1.73; P = 0.018), the absence of systemic chemotherapy (HR = 1.71; P = 0.035), and primary site of upper urinary tract (HR = 1.71; P = 0.014) in descending order by HR. The prognostic significance of elevated GGT was also observed in a subset of 101 patients who had been diagnosed with aUC at their first presentation. Conclusion The present study for the first time demonstrated that elevated serum GGT was an independent adverse prognostic factor in aUC patients.