Search

Recent Articles

IJMPO—A Journey of a Thousand Miles

Author : Padmaj S. Kulkarni

Coronavirus Disease 2019 Treatment—T-Cells Hold the Key in Severe Cases

Author : Kunal Das, Nitika Agrawal, Mansi Kala, Rakhee Khanduri

Why Is China Importing COVID-19 Vaccine Now?

Author : Purvish M. Parikh

Management of Locally Advanced Unresectable or Metastatic Urothelial Carcinoma: Expert Opinion from an Indian Panel via Delphi Consensus Method

CC BY 4.0 · Indian J Med Paediatr Oncol 2024; 45(05): 365-375

DOI: DOI: 10.1055/s-0042-1760317

Abstract

Introduction Currently, there are no guidelines for the management of locally advanced unresectable or metastatic urothelial carcinoma (mUC) from an Indian perspective. There is a lack of consensus on the utility of treatment options in first-line (1L) and second-line (2L) settings, especially in cisplatin- and platinum-unfit mUC patient subgroups.

Objective This articles aims to develop evidence-based practical consensus recommendations for the management of mUC in Indian settings.

Methods Modified Delphi consensus methodology was considered to arrive at a consensus. An expert scientific committee of 15 medical oncologists from India constituted the panel. Twelve clinically relevant questions were grouped into five categories for presentation and discussion: (1) cisplatin and platinum ineligibility criteria; (2) programmed death ligand 1 and fibroblast growth factor receptor (FGFR) testing in mUC patients; (3) treatment options in 1L settings; (4) role of switch maintenance; and (5) treatment options in 2L. Statements that reached high (≥ 80%) and moderate (60–79%) levels of consensus in the first round (electronic survey) did not undergo the second Delphi round. The questions that received a low level of consensus (< 60>

Results Renal impairment (creatinine clearance [CrCl] < 60 mL/min) and New York Heart Association class 3 heart failure are important assessment criteria for determining cisplatin ineligibility. Patients are unfit for any platinum-based chemotherapy in case of Eastern Cooperative Oncology Group performance status> 3 or severe renal impairment (CrCl < 30 mL/min). Gemcitabine and platinum with cisplatin over carboplatin were preferred in 1L settings. In patients unfit for cisplatin-based regimens, carboplatin–gemcitabine chemotherapy was preferred over immunotherapy (atezolizumab or pembrolizumab). Selected patients who are platinum ineligible may be considered for immunotherapy. Post-induction chemotherapy, those who do not progress may be strongly considered for avelumab maintenance. Experts recommended erdafitinib in FGFR-positive mUC patients in 2L settings. In FGFR-negative patients, immunotherapy (pembrolizumab, nivolumab, or avelumab) may be preferred over chemotherapy (paclitaxel, docetaxel, or vinflunine). Enfortumab vedotin and sacituzumab govitecan may be considered for further lines of therapy.

Conclusion Expert panel consensus will offer expert guidance to oncologists/clinicians on the management of mUC in Indian settings.

Key Points

  • In 1L settings, the experts preferred gemcitabine and platinum with cisplatin over carboplatin in mUC patients.

  • In patients unfit for cisplatin-based regimens, carboplatin–gemcitabine chemotherapy was preferred over immunotherapy (atezolizumab or pembrolizumab). Selected patients who are platinum ineligible (cisplatin and carboplatin) may be considered for immunotherapy (atezolizumab or pembrolizumab) in 1L. Post-induction chemotherapy, those who do not progress should be strongly considered for avelumab switch maintenance.

  • Erdafitinib was recommended in FGFR-positive mUC patients in 2L.

  • In FGFR-negative patients, platinum-based chemotherapy was suggested in 2L for those relapsing late, immunotherapy (pembrolizumab, nivolumab, or avelumab) for those who did not receive targeted immunotherapy in 1L, and single-agent chemotherapy (paclitaxel, docetaxel, or vinflunine) for other mUC patients.

Supplementary Material

Publication History

Article published online:
10 February 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

We recommend

Evaluation and Management of Unresectable Hepatocellular Carcinoma: Multidisciplinary Indian Consensus Statements from a Delphi Panel

Anant Ramaswamy, South Asian Journal of Cancer

Oncology Gold StandardTM practical consensus recommendations 2016 for treatment of advanced clear cell renal cell carcinoma

U Batra, South Asian Journal of Cancer, 2016

Treatment outcomes of metastatic nonclear cell renal cell carcinoma: A single institution retrospective analysis

Vivek Agarwala, South Asian Journal of Cancer, 2018

Real-World Experience with Nivolumab in Metastatic Renal Cell Carcinoma Patients Who Have Progressed on Prior Therapies: A Single-Center Study from India

Amit Rauthan, South Asian Journal of Cancer

Long-Term Results and Prognostic Factors of Pulmonary Metastasectomy in Patients with Metastatic Transitional Cell Carcinoma

Luca Luzzi, The Thoracic and Cardiovascular Surgeon, 2016

Efficacy of Platinum Rechallenge in Metastatic Urothelial Carcinoma After Previous Platinum-Based Chemotherapy for Metastatic Disease

Risa L. Wong, The Oncologist, 2021

The Rapidly Evolving Landscape of First-Line Targeted Therapy in Metastatic Urothelial Cancer: A Systematic Review

Jean-Michel Lavoie, The Oncologist, 2021

Platinum ineligibility and survival outcomes in patients with advanced urothelial carcinoma receiving first-line treatment

Shilpa Gupta, JNCI: Journal of the National Cancer Institute

Outcome and prognostic factors in metastatic urothelial carcinoma patients receiving second-line chemotherapy: an analysis of real-world clinical practice data ...

Ryuji Matsumoto, Japanese Journal of Clinical Oncology, 2018

Erdafitinib for locally advanced or metastatic urothelial carcinoma

Arlene O Siefker-Radtke, American Journal of Health-System Pharmacy-1, 2022


Abstract

Introduction Currently, there are no guidelines for the management of locally advanced unresectable or metastatic urothelial carcinoma (mUC) from an Indian perspective. There is a lack of consensus on the utility of treatment options in first-line (1L) and second-line (2L) settings, especially in cisplatin- and platinum-unfit mUC patient subgroups.

Objective This articles aims to develop evidence-based practical consensus recommendations for the management of mUC in Indian settings.

Methods Modified Delphi consensus methodology was considered to arrive at a consensus. An expert scientific committee of 15 medical oncologists from India constituted the panel. Twelve clinically relevant questions were grouped into five categories for presentation and discussion: (1) cisplatin and platinum ineligibility criteria; (2) programmed death ligand 1 and fibroblast growth factor receptor (FGFR) testing in mUC patients; (3) treatment options in 1L settings; (4) role of switch maintenance; and (5) treatment options in 2L. Statements that reached high (≥ 80%) and moderate (60–79%) levels of consensus in the first round (electronic survey) did not undergo the second Delphi round. The questions that received a low level of consensus (< 60>

Results Renal impairment (creatinine clearance [CrCl] < 60 mL/min) and New York Heart Association class 3 heart failure are important assessment criteria for determining cisplatin ineligibility. Patients are unfit for any platinum-based chemotherapy in case of Eastern Cooperative Oncology Group performance status> 3 or severe renal impairment (CrCl < 30 mL/min). Gemcitabine and platinum with cisplatin over carboplatin were preferred in 1L settings. In patients unfit for cisplatin-based regimens, carboplatin–gemcitabine chemotherapy was preferred over immunotherapy (atezolizumab or pembrolizumab). Selected patients who are platinum ineligible may be considered for immunotherapy. Post-induction chemotherapy, those who do not progress may be strongly considered for avelumab maintenance. Experts recommended erdafitinib in FGFR-positive mUC patients in 2L settings. In FGFR-negative patients, immunotherapy (pembrolizumab, nivolumab, or avelumab) may be preferred over chemotherapy (paclitaxel, docetaxel, or vinflunine). Enfortumab vedotin and sacituzumab govitecan may be considered for further lines of therapy.

Conclusion Expert panel consensus will offer expert guidance to oncologists/clinicians on the management of mUC in Indian settings.

Key Points

  • In 1L settings, the experts preferred gemcitabine and platinum with cisplatin over carboplatin in mUC patients.

  • In patients unfit for cisplatin-based regimens, carboplatin–gemcitabine chemotherapy was preferred over immunotherapy (atezolizumab or pembrolizumab). Selected patients who are platinum ineligible (cisplatin and carboplatin) may be considered for immunotherapy (atezolizumab or pembrolizumab) in 1L. Post-induction chemotherapy, those who do not progress should be strongly considered for avelumab switch maintenance.

  • Erdafitinib was recommended in FGFR-positive mUC patients in 2L.

  • In FGFR-negative patients, platinum-based chemotherapy was suggested in 2L for those relapsing late, immunotherapy (pembrolizumab, nivolumab, or avelumab) for those who did not receive targeted immunotherapy in 1L, and single-agent chemotherapy (paclitaxel, docetaxel, or vinflunine) for other mUC patients.


    Introduction

    Carcinoma of the urinary bladder is a common urological malignancy that causes substantial morbidity and mortality. As per the Global Cancer Observatory: CANCER TODAY (GLOBOCAN) statistics, bladder cancer (BC) ranked 10th in incidence, with approximately 573,000 new cases and 213,000 deaths in 2020.[1] Urothelial carcinoma (UC) is the predominant histologic type of BC and accounts for nearly 90% of all BC cases globally.[2] BC has a wide spectrum of disease severity from low-grade non–muscle-invasive BC (NMIBC) to muscle-invasive disease stage and metastatic lesions associated with poor outcomes.[3] The majority of muscle-invasive tumors are high-grade UCs. A high probability of local/systemic recurrences of muscle-invasive tumors within 36 months after local treatment of primary tumor has been observed.[4] BC diagnosed at earlier stages carry a greater chance of 5-year relative survival compared to later disease stages ([Table 1]).[5] Overall, 5-year relative survival rate for patients diagnosed with distant metastatic UC (mUC) is roughly 6%.[5] In India, BC is ranked 17th in incidence and 19th in mortality, with significant heterogeneity in incidence rates across different regions of India.[6] The overall incidence rate of BC in India as per the National Cancer Registry Programme report is 2.25 per 100,000 annually (males: 3.67 and females: 0.83).[7] In India, BC patients are more often diagnosed with locally advanced diseases, resulting in poor outcomes. A study by Prakash et al assessed the stage distribution of patients presenting with BC (N = 419) to a tertiary care cancer center in Mumbai.[8] The median age of patients at diagnosis was 59 (18–88) years.[8] Around 47% of patients had NMIBC, 36% had muscle-invasive and locally advanced disease, and 17% had metastatic disease.[8] The most common sites of distant metastasis were bone, lung, liver, pelvic peritoneum, adrenal glands, and nonregional lymph nodes.[8]

Table 1

Five-year relative survival rates of bladder cancer


The National Comprehensive Cancer Network (NCCN) guidelines recommend either gemcitabine–cisplatin combination chemotherapy or dose-dense methotrexate, vinblastine, doxorubicin, and cisplatin (ddMVAC) with growth factor support for cisplatin-eligible mUC patients in first-line (1L) settings.[3] Subsequent switch maintenance may be considered in patients with a nonprogressive disease on 1L platinum-based chemotherapy.[3] However, a substantial proportion of mUC patients are unfit to receive cisplatin-based chemotherapy in 1L settings due to advanced age, poor performance status, impaired renal function, and presence of multiple comorbidities. Currently, there are no defined criteria to establish cisplatin and platinum ineligibility in India, and it varies among different physicians and institutes. In addition, there is a lack of consensus on the utility of different treatment options in cisplatin- and platinum-unfit patients in 1L settings and treatment options in second-line (2L) settings. In this article, we have attempted to summarize expert opinions and recommendations on (1) cisplatin and platinum ineligibility criteria, (2) programmed death ligand 1 (PD-L1) and fibroblast growth factor receptor (FGFR) testing, (3) treatment options in 1L settings, (4) role of switch maintenance after 1L platinum-containing chemotherapy, and (5) treatment options in 2L settings based on the available efficacy and safety data.

Methodology

Panel Selection

A panel of 15 medical oncologists with significant experience in managing BC patients participated in the development of the consensus article.

Evidence Review

A literature review was carried out based on data from the PubMed database to identify relevant articles between January 2001 and March 2022 using keywords such as “urothelial carcinoma,” “bladder cancer,” “first-line,” “second-line,” “maintenance,” “guidelines,” and “management.” Twelve clinically relevant questions ([Supplementary Material]) belonging to five major domains were drafted.

  • Cisplatin/platinum ineligibility criteria.

  • PD-L1 and FGFR testing.

  • Treatment pattern in 1L settings.

  • Role of switch maintenance.

  • Treatment pattern in 2L settings and subsequent therapy.

An electronic survey link to these questions was sent to all the participants to record their views. Key articles were shortlisted and circulated among the participants before the survey. The “Oxford Centre for Evidence-Based Medicine: Levels of Evidence (LOE)” was used to define the grade or LOE of each recommendation ([Table 2A]).[9]

Stage at diagnosis

5-year relative survival (%)

Stage 0: Noninvasive papillary carcinoma and carcinoma in situ

96

Stage 0–I: Localized (confined to primary sites)

70

Stage III–IV: Regional (spread to regional lymph nodes)

38

Stage IV: Distant (metastasis to lungs, liver, or bones)

6

Table 2

Definitions: (A) Oxford LOE grading system, (B) grades of recommendation, and (C) level of consensus

(A) Oxford Centre for Evidence-Based Medicine: Level of evidence (LOE)

LOE

Therapy/prevention, etiology/harm

Prognosis

1a

Systematic review (with homogeneity) of RCTs

A systematic review (with homogeneity) of inception cohort studies; clinical decision rule validated in different populations

1b

Individual RCT (with narrow CI)

Individual inception cohort study with > 80% follow-up; clinical decision rule validated in a single population

1c

All or none

All or no case series

2a

Systematic review (with homogeneity) of cohort studies

A systematic review (with homogeneity) of either retrospective cohort studies or untreated control groups in RCTs

2b

Individual cohort study (including low-quality

RCTs, < 80% follow-up)

Retrospective cohort study or follow-up of untreated control patients in an RCT; derivation of clinical decision rule or validated on split-sample only

2c

“Outcomes” research and ecological studies

“Outcomes” research

3a

Systematic review (with homogeneity) of case–control studies

3b

Individual case–control study

4

Case series (and poor-quality cohort and case–control studies)

Case series (and poor-quality prognostic cohort studies)

5

Expert opinion without an explicit critical appraisal, or based on physiology, bench research, or “first principles”

Expert opinion without an explicit critical appraisal, or based on physiology, bench research, or “first principles”

(B) Grade of recommendation (GOR)

++

This investigation or therapeutic intervention is highly beneficial for patients, can be recommended without restriction, and should be performed

+

This investigation or therapeutic intervention is of limited benefit for patients and can be performed

+/−

This investigation or therapeutic intervention has not shown benefit for patients and may be performed only in individual cases. According to current knowledge, a general recommendation cannot be given

This investigation or therapeutic intervention can be of disadvantage to patients and might not be performed

−−

This investigation or therapeutic intervention is of clear disadvantage to patients and should be avoided or omitted in any case

(C) Level of consensus (LOC)

High

When ≥ 80% of participants agree/strongly agree or disagree/strongly disagree with a statement

Moderate

When 60–79% of participants agree/strongly agree or disagree/strongly disagree with a statement

Low

When < 60>

Table 3

Cisplatin-/platinum-ineligibility criteria and biomarker testing in mUC patients


Treatment Pattern in 1L Settings

Patients eligible for cisplatin-based chemotherapy: The NCCN guidelines recommend either gemcitabine–cisplatin chemotherapy or ddMVAC with growth factor support for cisplatin-eligible mUC patients in 1L settings.[3]

Patients ineligible to receive cisplatin-based chemotherapy: Carboplatin–gemcitabine combination chemotherapy is recommended in cisplatin-ineligible mUC patients in 1L based on the results of phase 1/2 randomized EORTC 30986 trial (overall response rate [ORR]: 41.2%; median OS: 9.3 months) (LOE: 2b).[26] Treatment with an ICI (atezolizumab [PD-L1 inhibitor] or pembrolizumab [PD-1 inhibitor]) could be an alternative option.[3] Currently, the use of PD-L1 testing before 1L therapy is advised in mUC patients who are cisplatin ineligible and have no contraindications to the use of immunotherapy.[3] In phase 2, IMvigor210 cohort study, atezolizumab conferred significant clinical benefits in untreated cisplatin-ineligible mUC.[27] Scoring criteria designated tumors based on tumor-infiltrating immune cells (ICs): (1) IC0 (PD-L1 expression on < 1% of IC), (2) IC1 (PD-L1 expression on ≥ 1% and < 5% of IC), or (3) IC2/3 (PD-L1 expression on ≥ 5% of IC). The study demonstrated favorable durable response rates, survival, and tolerability of atezolizumab in mUC patients in 1L settings.[27] The median OS was 12.3 months in IC2/3 and 19.1 months in IC0/1 group (LOE: 2b).[27] However, in May 2018, the Food and Drug Administration (FDA) issued a safety alert for use of atezolizumab monotherapy in 1L settings due to decreased survival compared to platinum-based chemotherapy in mUC patients who have not received prior therapy and who have low PD-L1 expression.[28] The FDA has restricted the use of atezolizumab in cisplatin-unfit mUC patients with positive PD-L1 status (PD-L1 expression on ≥ 5% of IC) and mUC patients eligible for any platinum-containing chemotherapy regardless of PD-L1 expression in 1L settings.[28] On April 2021, the FDA agreed to continue the accelerated approval of atezolizumab in the frontline treatment of cisplatin-unfit mUC.[29] The efficacy and safety of pembrolizumab were assessed in the phase 2 KEYNOTE-052 trial in 1L settings.[30] [31] The study demonstrated the efficacy of pembrolizumab with acceptable tolerability in cisplatin-unfit patients, most of whom were elderly, had poor performance status, or had serious comorbidities. In patients with positive PD-L1 status defined as a combined positive score (CPS) ≥ 10, the median OS was 18.5 months (95% confidence interval [CI]:12.2–28.5 months) (LOE: 2b).[30] [31] Frail patients and patients with three or more comorbidities are candidates for best supportive care (BSC) alone instead of systemic therapy in 1L (LOE: 2c).[32] [33] [34]

Patients ineligible to receive any platinum-based chemotherapy (cisplatin and carboplatin): On August 2021, the FDA converted the accelerated approval of 1L pembrolizumab in locally advanced or mUC (cisplatin-unfit patients with PD-L1 CPS ≥ 10 or patients who are not eligible for any platinum-containing chemotherapy regardless of PD-L1 status) to a full approval and revised the indication to only cover the treatment of patients who are not eligible for any platinum-containing chemotherapy.[35] Based on the FDA approvals and evidence, the recent 2022 NCCN guideline has recommended: (1) atezolizumab (for patients not eligible for any platinum-containing chemotherapy regardless of PD-L1 expression); or (2) pembrolizumab (for patients who are not eligible for any platinum-containing chemotherapy) (LOE: 2b).[3] [27] [28] [30] [31] [35]

Consensus/recommendations: Experts agreed that they would prefer carboplatin–gemcitabine chemotherapy followed by avelumab maintenance over ICI monotherapy (atezolizumab or pembrolizumab) in mUC patients with positive PD-L1 status unfit for cisplatin-based regimens in 1L settings (GOR: ++; LOC: “high”). Experts preferred ICI monotherapy (atezolizumab or pembrolizumab) over BSC in patients ineligible for any platinum-based chemotherapy (GOR: ++; LOC: “high”). BSC should be strongly preferred over ICI therapy in patients with: (1) poor performance status, (2) multiple uncontrolled comorbidities, and/or (3) poor access to therapies (GOR: ++; LOC: “high”).

Role of ICI–chemotherapy combination therapy: Two trials investigated the relevance of ICI (atezolizumab or pembrolizumab) plus platinum-based chemotherapy combination in 1L settings. The first trial to report was IMvigor130, where atezolizumab plus platinum-based chemotherapy provided PFS benefit (8.2 vs. 6.3 months; p = 0.007); however, OS was not significant after a median follow-up of 11.8 months compared to placebo plus platinum-based chemotherapy.[36] The KEYNOTE 361 study had a similar design and investigated pembrolizumab plus platinum–gemcitabine versus chemotherapy plus placebo in 1L settings.[37] The study revealed no benefit of this combination in terms of PFS or OS.[37]

Consensus/recommendations: Experts agreed that immunotherapy (atezolizumab or pembrolizumab) plus platinumgemcitabine chemotherapy is not suitable in mUC patients in 1L settings (GOR: −; LOC: “high”).

Role of Switch Maintenance in Locally Advanced Unresectable UC or mUC After Platinum-Based Chemotherapy

The JAVELIN Bladder 100 phase 3 RCT explored the impact of switch maintenance with PD-L1 inhibitor avelumab plus BSC versus BSC alone in mUC not progressed with 1L platinum-containing chemotherapy (complete or partial response vs. stable disease).[21] Patients were categorized as having PD-L1-positive status if at least one of the three criteria were met: (1) at least 25% of tumor cells stained for PD-L1, (2) at least 25% of ICs stained for PD-L1 if more than 1% of the tumor area contained ICs, or (3) 100% of ICs stained for PD-L1 if no more than 1% of the tumor area contained ICs.[21] Addition of maintenance avelumab to BSC significantly prolonged median OS (21.4 months; 95% CI: 18.9–26.1) as compared with BSC alone (14.3 months [95% CI: 12.9–17.9]; p = 0.001) (LOE: 1b).[21] With extended follow-up (≥ 38 months), median OS remained significantly longer in the avelumab plus BSC (23.8 months [95% CI: 19.9–28.8]) as compared to BSC alone (15.0 months [95% CI: 13.5–18.2]; p = 0.0036) in unresectable locally advanced UC or mUC without disease progression.[38] Another phase 2 RCT investigated the impact of switch maintenance with PD-1 inhibitor pembrolizumab in mUC patients achieving at least stable disease on 1L platinum-based chemotherapy. In this study, the OS was not significantly different (22 vs. 18.7 months) in patients randomly assigned to maintenance pembrolizumab versus placebo (LOE: 2b).[39]

Consensus/recommendations: Experts strongly recommended avelumab switch maintenance plus BSC in mUC patients with nonprogressive disease after 4 to 6 cycles of 1L platinum-containing chemotherapy (GOR: ++; LOC: “high”). They would not prefer pembrolizumab switch maintenance due to no significant OS benefit in mUC patients (GOR: −; LOC: “high”). They would consider BSC alone in patients with poor performance status and lack of access to immunotherapies (GOR: +/− ; LOC: “high”).

Patient profiles suitable for avelumab switch maintenance: Avelumab switch maintenance plus BSC provided an OS and PFS benefit in patients with PD-L1-positive or PD-L1-negative tumors, with a potentially greater benefit in patients with PD-L1-positive tumors.[21] Avelumab maintenance significantly prolonged OS in the PD-L1-positive mUC patients; OS at 1 year was 79.1% in the avelumab group versus 60.4% in the control group (BSC alone; p < 0.001) (LOE: 1b).[21] With extended follow-up (≥ 38 months), median OS remained significantly longer in the avelumab plus BSC (30.9 months [95% CI: 24.0–39.8]) as compared to BSC alone (18.5 months [95% CI: 14.1–24.2]; p = 0.0064) in unresectable locally advanced UC or mUC patients with PD-L1-positive tumors.[38] The JAVELIN Bladder 100 phase 3 trial was not powered to assess PFS/OS in the PD-L1-negative mUC patients in maintenance settings.[21] In mUC patients with PD-L1-negative tumors, the median OS was 18.8 months (95% CI: 13.3–22.5) in the avelumab plus BSC group versus 13.7 months (95% CI: 10.8–17.8) in the BSC alone (hazard ratio: 0.85; 95% CI: 0.62–1.18) (LOE: 2b).[21] The trial demonstrated OS benefits with avelumab switch maintenance in a range of patient subgroups (categorized by age, ECOG PS 0/1, prior chemotherapy regimen, response to chemotherapy, site of baseline metastasis, CrCl) not progressed with 1L platinum-containing chemotherapy (LOE: 1b).[20] [21]

Consensus/recommendations: Experts agreed that avelumab switch maintenance therapy is beneficial and can be recommended in mUC patients with ECOG 0/1, age < 65 years, regardless of PD-L1 status, CrCl, site of metastasis, and chemotherapy (gemcitabine with cisplatin or carboplatin). The patient profiles that received moderate consensus during the discussion were: (1) stable disease after 1L platinum-containing chemotherapy, (2) visceral metastasis after 1L platinum-containing chemotherapy, and (3) age ≥ 65 years (GOR: ++).

[Table 4] lists the recommendations for 1L systemic therapy and switch maintenance after 1L platinum-containing chemotherapy.



(A) Cisplatin/platinum ineligibility criteria: Summary of expert recommendations

Expert recommendations on cisplatin-ineligibility criteria

• ECOG PS ≥ 2 (LOE: 1a; GOR: ++; LOC: 66.7%).

• CrCl < 60 mL/min (LOE: 1a; GOR: ++; LOC: 86.7%)

• Grade ≥ 2 peripheral neuropathy (LOE: 2c; GOR: ++; LOC: 60%)

• NYHA class 3 heart failure (LOE: 2c; GOR: ++; LOC: 80%)

• Grade ≥ 2 hearing loss (LOE: 2c; GOR: +; LOC: 80%)

Expert recommendations on platinum (cisplatin and carboplatin)-ineligibility criteria

• ECOG PS > 3 (LOE: 2c; GOR: ++; LOC: 80%)

• CrCl < 30 mL/min (LOE: 2c; GOR: ++; LOC: 80%)

• Grade > 3 peripheral neuropathy (LOE: 2c; GOR: ++; LOC: 60%)

• NYHA class > 3 heart failure (LOE: 2c; GOR: ++; LOC: 60%)

• ECOG PS 2 and CrCl < 30 mL/min are important criteria for determining platinum ineligibility in patients with mUC (LOE: 2c; GOR: ++; LOC: 66.7%)

• Grade ≥ 2 hearing loss (LOE: 2c; GOR: +; LOC: 80%)

(B) Biomarker testing in mUC: Summary of expert recommendations

PD-L1 testing before 1L systemic therapy can be performed in mUC patients who are ineligible to receive cisplatin chemotherapy. PD-L1 testing before 1L systemic therapy is not required for those who are ineligible to receive any platinum-based chemotherapy (LOE: 1a; GOR: +; LOC: 80%)

PD-L1 testing is not required when assessing eligibility for ICI maintenance in patients who have not progressed with platinum-containing chemotherapy (LOE: 2b; GOR: +/− ; LOC: 80%)

PD-L1 testing is not required when assessing eligibility for treatment in 2L settings. According to current knowledge, a general recommendation cannot be given (LOE: 1a; GOR: +/− ; LOC: 80%)

FGFR mutation testing has not shown benefit for mUC patients in 1L settings. A general recommendation regarding FGFR testing before 1L systemic therapy cannot be given (LOE: 2b; GOR: +/− ; LOC: 80%)

It is important to screen mUC patients for FGFR alterations before 2L systemic therapy to plan for optimal treatment (LOE: 1a; GOR: ++; LOC: 80%)

Table 4

First-line systemic therapy and switch maintenance for locally advanced or mUC

Treatment Pattern in 2L and Subsequent Therapy

In the phase 3 KEYNOTE-045 RCT, pembrolizumab conferred significant OS benefits in 2L (10.3 vs. 7.4 months; p = 0.002) as compared to the chemotherapy group (paclitaxel, docetaxel, or vinflunine) in mUC patients who progressed during or after the receipt of platinum chemotherapy (LOE: 1b).[40] Nivolumab, a fully human immunoglobulin G4 PD-1 ICI, demonstrated clinical benefit (ORR was 19·6% [95% CI: 15.0–24.9]) in a phase 2, single-arm study in mUC patients whose disease progressed or recurred despite previous treatment with at least one platinum-based chemotherapy regimen (LOE: 2b).[41] Recently, the efficacy and safety of avelumab in 2L were assessed in phase 1b JAVELIN Solid Tumor study. Avelumab therapy resulted in a median OS of 7.0 months and a 24-month OS rate of 20.1% (LOE: 2b).[42] For management of mUC patients with FGFR alternations, the NCCN guideline recommends erdafitinib, a tyrosine kinase inhibitor of FGFR1–4, in 2L based on the promising result from the phase 2 BLC2001 study.[43] The confirmed response rate to erdafitinib therapy was 40% and the median OS was 13.8 months. Among patients who had undergone prior immunotherapy, the response rate was 59% (LOE: 2b).[43] The indication of atezolizumab was withdrawn by the FDA in March 2021 in mUC patients previously treated with platinum-based chemotherapy based on the results of the phase 3 IMvigor211 trial.[44] The trial failed to meet its primary endpoint of OS benefit in mUC patients with positive PD-L1 status (IC2/3; 11.1 vs. 10.6 months; p = 0.41) as compared to chemotherapy (vinflunine, paclitaxel, or docetaxel).[44] [45] The NCCN guidelines recommend: (1) rechallenge with gemcitabine and cisplatin or carboplatin or MVAC in patients who relapse after a year of last platinum exposure, (2) erdafitinib in patients with FGFR3 or FGFR2 genetic alterations, or (3) ICI therapy (pembrolizumab, nivolumab, or avelumab) in patients who have not received ICI in 1L settings.[3]

Consensus/recommendations: Experts recommended erdafitinib in FGFR-positive mUC patients in 2L settings (GOR: ++; LOC: “high”). Experts agreed that in FGFR-negative patients, ICIs (pembrolizumab, nivolumab, or avelumab) may be preferred over chemotherapy (paclitaxel, docetaxel, or vinflunine). Patient eligibility should be determined before therapy based on the available efficacy and safety data. On the other hand, chemotherapy (paclitaxel, docetaxel, or vinflunine) can be considered in patients who are not eligible for ICI therapy or have poor access to ICI therapy. Experts strongly opined that pembrolizumab can be preferred as it has strong phase 3 clinical evidence with OS benefit (GOR: ++; LOC: “high”) as compared to nivolumab (GOR: +; LOC: “high”).

Patient profiles suitable for ICI in 2L: Experts agreed that ICI therapy (pembrolizumab, nivolumab, or avelumab) is suitable and can be recommended in patients with ECOG status 0/1 (GOR: ++; LOC: “high”). In addition, ICI therapy can be considered in patients with: (1) prior cisplatin chemotherapy, (2) PD-L1 (IC 2/3), and (3) visceral disease (GOR: ++; LOC: “moderate”).

Scope of antibody–drug conjugates (ADCs): In phase 3 EV-301 trial, the efficacy of enfortumab vedotin, a nectin-4-directed ADC, was assessed in patients previously treated with platinum-containing chemotherapy and a PD-1 or PD-L1 inhibitor.[46] The study demonstrated that median OS was significantly longer in the enfortumab vedotin treatment arm (12.88 months [95% CI: 10.58–15.21) as compared to the chemotherapy group (docetaxel, paclitaxel, or vinflunine) (8.97 months [95% CI: 8.05–10.74]; p = 0.001) (LOE: 1b).[46] Another phase 2, open-label (TROPHY-U-01) cohort study investigated the role of TROP-2-directed ADC sacituzumab govitecan in mUC patients who progress on platinum-based combination chemotherapy and ICI therapy.[47] The median OS achieved with sacituzumab govitecan therapy was 10.9 months (95% CI: 9.0–13.8 months) (LOE: 2b).[47]

Consensus/recommendations: Experts agreed that enfortumab vedotin is a suitable treatment option in patients who have previously received platinum-containing chemotherapy and progressed during or after treatment with a PD-1 or PD-L1 inhibitor (GOR: ++; LOC: “moderate”). Currently, enfortumab vedotin is available only on a compassionate basis in India. Sacituzumab govitecan is another treatment option in patients who have previously received platinum-containing chemotherapy and progressed during or after treatment with a PD-1 or PD-L1 inhibitor (GOR: ++; LOC: “low”).

OS improvement from the start of 1L therapy: Experts opined that 1L platinum-based chemotherapy (4–6 cycles) followed by avelumab switch maintenance with BSC is most useful in terms of OS improvement from the start of 1L therapy and can be recommended (GOR: ++; LOC: “high”).

[Table 5] lists the recommendations for 2L systemic therapy for the management of mUC.




(A) 1L systemic therapy for locally advanced unresectable UC or mUC: Summary of expert recommendations

Treatment of cisplatin-ineligible mUC patients with positive PD-L1 status

Carboplatin–gemcitabine chemotherapy is preferred over ICI monotherapy (atezolizumab or pembrolizumab) in mUC patients with positive PD-L1 status deemed unfit for cisplatin-based therapy in 1L settings (LOE: 2b; GOR: ++; LOC: 80%)

Treatment of mUC patient ineligible for any platinum-based chemotherapy (cisplatin and carboplatin ineligible)

• ICI monotherapy (atezolizumab or pembrolizumab) can be preferred over BSC in patients ineligible for any platinum-based chemotherapy (LOE: 2b; GOR: ++; LOC: 80%)

• BSC is strongly preferred over ICI therapy in patients with: (1) poor performance status; (2) multiple uncontrolled comorbidities; and/or (3) poor access to immunotherapies (LOE: 2c; GOR: ++; LOC: 80%)

Scope of immunotherapy–chemotherapy combination in 1L treatment settings

• Immunotherapy (atezolizumab or pembrolizumab) plus platinum–gemcitabine chemotherapy is not suitable in mUC patients in 1L treatment settings (LOE: 1b; GOR: −; LOC: 80%)

(B) Switch maintenance for locally advanced unresectable UC or mUC patients after 1L platinum-containing chemotherapy: Expert recommendations

Switch maintenance in the general population

• Avelumab switch maintenance plus BSC is strongly recommended in mUC patients with the nonprogressive disease after 4–6 cycles of 1L platinum-containing chemotherapy (LOE: 1b; GOR: ++; LOC: 100%)

• Pembrolizumab switch maintenance is not suitable after 1L platinum-containing chemotherapy due to no

 OS benefit in mUC patients (LOE: 2b; GOR: −; LOC: 80%)

• BSC instead of switch maintenance can be considered in patients with poor performance status and lack of access to immunotherapies (LOE: 2c; GOR: +/− ; LOC: 80%)

Patient profiles suitable for avelumab switch maintenance therapy

PD-L1 status

• Avelumab switch maintenance plus BSC is strongly recommended in PD-L1-positive mUC patients with the nonprogressive disease after 4–6 cycles of 1L platinum-containing chemotherapy (LOE: 1b; GOR: ++; LOC: 86.7%)

• Avelumab switch maintenance plus BSC can be performed in PD-L1-negative mUC patients with the nonprogressive disease after 4–6 cycles of 1L platinum-containing chemotherapy (LOE: 2b; GOR: ++; LOC: 80%)

Prior chemotherapy regimen

• Avelumab switch maintenance therapy is beneficial and can be recommended in mUC patients not progressed on 1L gemcitabine–carboplatin or gemcitabine–cisplatin-based chemotherapy (LOE: 1b; GOR: ++; LOC: 93.3%)

Response to chemotherapy

• Avelumab switch maintenance therapy is beneficial and recommended in mUC patients with partial and complete response after 1L platinum-containing chemotherapy (LOE: 1b; GOR: ++; LOC: 86.7%)

• Avelumab maintenance therapy is also recommended in mUC patients with stable disease after 1L platinum-containing chemotherapy (LOE: 1b; GOR: ++; LOC: 66.7%)

Type of metastases

• Avelumab switch maintenance therapy is beneficial and can be recommended in mUC patients with nonvisceral metastasis after 1L platinum-containing chemotherapy (LOE: 1b; GOR: ++; LOC: 86.7%)

• Avelumab switch maintenance therapy is beneficial in mUC patients with visceral metastasis after 1L platinum-containing chemotherapy (LOE: 1b; GOR: ++; LOC: 73.3%)

ECOG status

Avelumab switch maintenance therapy is beneficial and can be recommended in mUC patients with ECOG status 0/1 (LOE: 1b; GOR: ++; LOC: 93.3%)

CrCl

Avelumab switch maintenance therapy is beneficial and can be recommended in mUC patients regardless of

CrCl (< 60>

Age

• Avelumab switch maintenance therapy is beneficial and can be recommended in mUC patients with age < 65 years (LOE: 1b; GOR: ++; LOC: 100%)

• Avelumab switch maintenance therapy is beneficial and can be recommended in mUC patients with age ≥ 65 years (LOE: 1b; GOR: ++; LOC: 66.7%)

Table 5

Second-line systemic therapy for locally advanced or mUC

Discussion

Clinical and Research Implications

Treatment of UC has evolved over the last few years with improved outcomes across different disease stages. ICI and targeted therapies have emerged as new options for the treatment of persistent diseases. In India, there are no country-specific guidelines or recommendations for the management of locally advanced unresectable or mUC. Furthermore, due to the scarcity of RCTs conducted in India and the lack of local guidelines or recommendations, oncologists rely on data from the Western world. Currently, there are no defined criteria to establish cisplatin and platinum ineligibility in India, and it varies among different physicians and institutes. There is a lack of consensus on the utility of treatment options, especially in cisplatin- and platinum-unfit mUC patient subgroups. To the best of our knowledge, this is the first evidence-based practical consensus document to guide clinicians on the management of mUC in Indian settings. This consensus document will offer expert guidance to Indian oncologists and help achieve consistency in mUC management across various healthcare settings.

Strengths: The members of the panel (in the space of genitourinary oncology) were selected to best represent the breadth of knowledge and clinical expertise in the field from all over India. There was no selection bias during the development of the expert committee. All experts actively participated during the consensus process. The responses of all panelists were generated in the form of graphs (GOR vs. response in percentage) to ensure the protection of participants' data.

Limitation: The patient's voice was not included in the consensus process.

Conclusion

In this article, we have attempted to summarize the Indian consensus on the management of locally advanced unresectable UC or mUC. Patients with treatment-naive mUC should be classified according to cisplatin and platinum eligibility based on clear definitions. In a 1L setting, the experts preferred gemcitabine and platinum with cisplatin over carboplatin. Selected patients who are platinum ineligible may be considered for atezolizumab or pembrolizumab. Post-induction chemotherapy, those who do not progress should be strongly considered for avelumab maintenance. Experts recommended screening mUC patients for FGFR3 alterations or FGFR2/3 fusion before deciding on 2L therapy. Options for 2L therapy include platinum-based chemotherapy for those relapsing late, targeted therapy with erdafitinib for patients with FGFR alterations, ICI (pembrolizumab, nivolumab, or avelumab) for those who have not received ICI in 1L settings, and single-agent chemotherapy (paclitaxel, docetaxel, or vinflunine) for others. Enfortumab vedotin and sacituzumab govitecan should be considered for further lines of therapy.

Conflict of Interest

None declared.

Acknowledgment

We would like to thank BioQuest Solutions Pvt Ltd for their editorial support.

Authors' Contributions

All authors have contributed equally to the concept, design, editing, review, and finalization of manuscript.

2L systemic therapy for locally advanced unresectable UC or mUC: Expert recommendations

• Erdafitinib is recommended in FGFR-positive mUC patients in 2L settings (LOE: 2b; GOR: ++; LOC: 80%)

• In FGFR-negative patients, ICI (pembrolizumab, nivolumab, or avelumab) may be preferred over chemotherapy (paclitaxel, docetaxel, or vinflunine) in 2L settings. Pembrolizumab has strong phase 3 data in terms of OS and can be preferred (LOE: 1b; GOR: ++; LOC: 80%) over nivolumab (LOE: 2b; GOR: +; LOC: 80%) in 2L settings

• Enfortumab vedotin is a suitable treatment option in mUC patients who have previously received platinum-containing chemotherapy and progressed during or after treatment with a PD-1 or PD-L1 inhibitor (LOE: 1b; GOR: ++; LOC: “moderate”). Currently, enfortumab vedotin is available only on a compassionate basis in India

• Sacituzumab govitecan is another treatment option in patients who have previously received platinum-containing chemotherapy and progressed during or after treatment with a PD-1 or PD-L1 inhibitor (LOE: 2b; GOR: ++; LOC: “low”)

Patient profiles suitable for ICI in 2L settings

• ECOG PS status: ICI therapy (pembrolizumab, nivolumab, or avelumab) is suitable and can be recommended in patients with ECOG status 0/1 in 2L settings (GOR: ++; LOC: 86.7%)

• PD-L1 status: ICI therapy (pembrolizumab, nivolumab or avelumab) can be considered in PD-L1 (IC 2/3 [GOR: ++; LOC: 60%] and IC 1 [GOR: +; LOC: 73.3%]) in 2L settings

• First-line chemotherapy: ICI therapy (pembrolizumab, nivolumab, or avelumab) is suitable in patients with prior cisplatin chemotherapy in 2L settings (GOR: ++; LOC: 73.3%)

• Extent of involvement: ICI therapy (pembrolizumab, nivolumab, or avelumab) can be considered in patients with visceral disease in 2L settings (GOR: ++; LOC: 60%)

OS improvement from the start of therapy: Expert recommendations

1L platinum-based chemotherapy (4–6 cycles) followed by avelumab switch maintenance with BSC is most useful in terms of OS improvement from the start of 1L therapy and can be recommended (GOR: ++; LOC: 100%)

Supplementary Material


References

  1. ung H, Ferlay J, Siegel RL. et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71 (03) 209-249
  2. aginala K, Barsouk A, Aluru JS, Rawla P, Padala SA, Barsouk A. Epidemiology of bladder cancer. Med Sci (Basel) 2020; 8 (01) 15
  3. CCN Clinical Practice Guidelines Version 1 (2022) for Bladder Cancer. Accessed April 12, 2022, at:  https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1417
  4. alderrama BP, González-Del-Alba A, Morales-Barrera R. et al. SEOM-SOGUG clinical guideline for localized muscle invasive and advanced bladder cancer (2021). Clin Transl Oncol 2022; 24 (04) 613-624
  5. ational Cancer Institute. Surveillance, Epidemiology, and End Results (SEER) on survival rates for bladder cancer. Accessed April 26, 2022, at:  https://www.cancer.org/cancer/bladder-cancer/detection-diagnosis-staging/survival-rates.html
  6. ishra V, Balasubramaniam G. Urinary bladder cancer and its associated factors – an epidemiological overview. Indian J Med Sci 2021; 73: 239-248
  7. bid A, Sen S, Bandyopadhyay R. Clinicopathological study of urothelial neoplasms in urinary bladder with special reference to expression of Her2/neu and Ki-67 in malignant lesions. Indian J Pathol Oncol 2021; 8: 369-376
  8. rakash G, Pal M, Odaiyappan K. et al. Bladder cancer demographics and outcome data from 2013 at a tertiary cancer hospital in India. Indian J Cancer 2019; 56 (01) 54-58
  9. xford Centre for Evidence-Based Medicine: levels of evidence. Accessed April 29, 2022, at:  https://www.cebm.ox.ac.uk/resources/levels-of-evidence/oxford-centre-for-evidence-based-medicine-levels-of-evidence-march-2009
  10. Nasa P, Jain R, Juneja D. Delphi methodology in healthcare research: how to decide its appropriateness. World J Methodol 2021; 11 (04) 116-129
  11. Scharl A, Thomssen C, Harbeck N, Müller V. AGO recommendations for diagnosis and treatment of patients with early breast cancer: update 2013. Breast Care (Basel) 2013; 8 (03) 174-180
  12. Jünger S, Payne S, Brearley S, Ploenes V, Radbruch L. Consensus building in palliative care: a Europe-wide Delphi study on common understandings and conceptual differences. J Pain Symptom Manage 2012; 44 (02) 192-205
  13. Galsky MD, Hahn NM, Rosenberg J. et al. Treatment of patients with metastatic urothelial cancer “unfit” for cisplatin-based chemotherapy. J Clin Oncol 2011; 29 (17) 2432-2438
  14. Park SB, Krishnan AV, Lin CS, Goldstein D, Friedlander M, Kiernan MC. Mechanisms underlying chemotherapy-induced neurotoxicity and the potential for neuroprotective strategies. Curr Med Chem 2008; 15 (29) 3081-3094
  15. National Cancer Institute. Cancer Therapy Evaluation Program: Common Terminology Criteria for Adverse Events (CTCAE) version 5. Accessed May 05, 2022, at:  https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/ctcae_v5_quick_reference_8.5x1/211.pdf
  16. Raja W, Mir MH, Dar I, Banday MA, Ahmad I. Cisplatin induced paroxysmal supraventricular tachycardia. Indian J Med Paediatr Oncol 2013; 34 (04) 330-332
  17. Gupta S, Bellmunt J, Plimack ER. et al. Defining “platinum-ineligible” patients with metastatic urothelial cancer (mUC). J Clin Oncol 2019; 37: 451
  18. De Santis M, Bellmunt J, Mead G. et al. Randomized phase II/III trial assessing gemcitabine/ carboplatin and methotrexate/carboplatin/vinblastine in patients with advanced urothelial cancer “unfit” for cisplatin-based chemotherapy: phase II–results of EORTC study 30986. J Clin Oncol 2009; 27 (33) 5634-5639
  19. Rouanne M, Radulescu C, Adam J, Allory Y. PD-L1 testing in urothelial bladder cancer: essentials of clinical practice. World J Urol 2021; 39 (05) 1345-1355
  20. Grivas P, Agarwal N, Pal S. et al. Avelumab first-line maintenance in locally advanced or metastatic urothelial carcinoma: applying clinical trial findings to clinical practice. Cancer Treat Rev 2021; 97: 102187
  21. Powles T, Park SH, Voog E. et al. Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. N Engl J Med 2020; 383 (13) 1218-1230
  22. Rui X, Gu TT, Pan HF, Zhang HZ. Evaluation of PD-L1 biomarker for immune checkpoint inhibitor (PD-1/PD-L1 inhibitors) treatments for urothelial carcinoma patients: a meta-analysis. Int Immunopharmacol 2019; 67: 378-385
  23. Casadei C, Dizman N, Schepisi G. et al. Targeted therapies for advanced bladder cancer: new strategies with FGFR inhibitors. Ther Adv Med Oncol 2019; 11: 1758835919890285
  24. Kwon WA, Seo HK. Emerging agents for the treatment of metastatic urothelial cancer. Investig Clin Urol 2021; 62 (03) 243-255
  25. Kacew A, Sweis RF.  FGFR3 alterations in the era of immunotherapy for urothelial bladder cancer. Front Immunol 2020; 11: 575258
  26. De Santis M, Bellmunt J, Mead G. et al. Randomized phase II/III trial assessing gemcitabine/carboplatin and methotrexate/carboplatin/vinblastine in patients with advanced urothelial cancer who are unfit for cisplatin-based chemotherapy: EORTC study 30986. J Clin Oncol 2012; 30 (02) 191-199
  27. Balar AV, Galsky MD, Rosenberg JE. et al; IMvigor210 Study Group. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. [published correction appears in Lancet. 26 August 2017;390(10097):848]. Lancet 2017; 389 (10064): 67-76
  28. FDA alerts health care professionals and oncology clinical investigators about an efficacy issue identified in clinical trials for some patients taking Keytruda (pembrolizumab) or Tecentriq (atezolizumab) as monotherapy to treat urothelial cancer with low expression of PD-L1. Accessed May 18, 2022, at:  https://www.fda.gov/drugs/drug-safety-and-availability/fda-alerts-health-care-professionals-and-oncology-clinical-investigators-about-efficacy-issue
  29. ODAC says ‘yes’ to continued approval of atezolizumab for cisplatin-ineligible locally advanced or metastatic UC. Accessed October 7, 2022, at:  https://www.targetedonc.com/view/odac-says-yes-to-continued-approval-of-atezolizumab-for-cisplatin-ineligible-locally-advanced-metastatic-uc
  30. Balar AV, Castellano D, O'Donnell PH. et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol 2017; 18 (11) 1483-1492
  31. Vuky J, Balar AV, Castellano D. et al. Long-term outcomes in KEYNOTE-052: Phase II study investigating first-line pembrolizumab in cisplatin-ineligible patients with locally advanced or metastatic urothelial cancer. J Clin Oncol 2020; 38 (23) 2658-2666
  32. Hugar LA, Lopa SH, Yabes JG. et al. Palliative care use amongst patients with bladder cancer. BJU Int 2019; 123 (06) 968-975
  33. Hugar LA, Wulff-Burchfield EM, Winzelberg GS, Jacobs BL, Davies BJ. Incorporating palliative care principles to improve patient care and quality of life in urologic oncology. Nat Rev Urol 2021; 18 (10) 623-635
  34. Castagneto B, Zai S, Marenco D. et al. Single-agent gemcitabine in previously untreated elderly patients with advanced bladder carcinoma: response to treatment and correlation with the comprehensive geriatric assessment. Oncology 2004; 67 (01) 27-32
  35. FDA approves updated indication for Merck's KEYTRUDA® (pembrolizumab) for treatment of certain patients with urothelial carcinoma (bladder cancer). Accessed Oct 07, 2022, at:  https://www.merck.com/news/fda-approves-updated-indication-for-mercks-keytruda-pembrolizumab-for-treatment-of-certain-patients-with-urothelial-carcinoma-bladder-cancer/
  36. Galsky MD, Arija JÁA, Bamias A. et al; IMvigor130 Study Group. Atezolizumab with or without chemotherapy in metastatic urothelial cancer (IMvigor130): a multicentre, randomised, placebo-controlled phase 3 trial. Lancet 2020; 395 (10236): 1547-1557
  37. Powles T, Csőszi T, Özgüroğlu M. et al; KEYNOTE-361 Investigators. Pembrolizumab alone or combined with chemotherapy versus chemotherapy as first-line therapy for advanced urothelial carcinoma (KEYNOTE-361): a randomised, open-label, phase 3 trial. Lancet Oncol 2021; 22 (07) 931-945
  38. Pérez-Valderrama B, Powles T, Sridhar SS. et al. Avelumab first-line (1L) maintenance for advanced urothelial carcinoma (UC): long-term follow-up results from the JAVELIN Bladder 100 trial. J Clin Oncol 2022; 40: 4559
  39. Galsky MD, Mortazavi A, Milowsky MI. et al. Randomized double-blind phase II study of maintenance pembrolizumab versus placebo after first-line chemotherapy in patients with metastatic urothelial cancer. J Clin Oncol 2020; 38 (16) 1797-1806
  40. Bellmunt J, de Wit R, Vaughn DJ. et al; KEYNOTE-045 Investigators. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 2017; 376 (11) 1015-1026
  41. Sharma P, Retz M, Siefker-Radtke A. et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol 2017; 18 (03) 312-322
  42. Apolo AB, Ellerton JA, Infante JR. et al. Avelumab as second-line therapy for metastatic, platinum-treated urothelial carcinoma in the phase Ib JAVELIN Solid Tumor study: 2-year updated efficacy and safety analysis. J Immunother Cancer 2020; 8 (02) e001246
  43. Loriot Y, Necchi A, Park SH. et al; BLC2001 Study Group. Erdafitinib in locally advanced or metastatic urothelial carcinoma. N Engl J Med 2019; 381 (04) 338-348
  44. Atezolizumab Indication in US Withdrawn for Previously Treated Metastatic Urothelial Cancer. Accessed May 18, 2022, at:  https://www.cancernetwork.com/view/atezolizumab-indication-in-us-withdrawn-for-previously-treated-metastatic-urothelial-cancer
  45. 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. [published correction appears in Lancet. 2018 Oct 20;392(10156):1402]. Lancet 2018; 391 (10122): 748-757
  46. Powles T, Rosenberg JE, Sonpavde GP. et al. Enfortumab vedotin in previously treated advanced urothelial carcinoma. N Engl J Med 2021; 384 (12) 1125-1135
  47. Tagawa ST, Balar AV, Petrylak DP. et al. TROPHY-U-01: A phase II open-label study of sacituzumab govitecan in patients with metastatic urothelial carcinoma progressing after platinum-based chemotherapy and checkpoint inhibitors. J Clin Oncol 2021; 39 (22) 2474-2485

Address for correspondence

Senthil J. Rajappa, DM
Department of Medical Oncology, Basavatarakam Indo American Cancer Hospital and RI
Banjara Hills, Hyderabad 500034, Telangana
India   


Publication History

Article published online:
10 February 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

We recommend

Evaluation and Management of Unresectable Hepatocellular Carcinoma: Multidisciplinary Indian Consensus Statements from a Delphi Panel

Anant Ramaswamy, South Asian Journal of Cancer

Oncology Gold StandardTM practical consensus recommendations 2016 for treatment of advanced clear cell renal cell carcinoma

U Batra, South Asian Journal of Cancer, 2016

Treatment outcomes of metastatic nonclear cell renal cell carcinoma: A single institution retrospective analysis

Vivek Agarwala, South Asian Journal of Cancer, 2018

Real-World Experience with Nivolumab in Metastatic Renal Cell Carcinoma Patients Who Have Progressed on Prior Therapies: A Single-Center Study from India

Amit Rauthan, South Asian Journal of Cancer

Long-Term Results and Prognostic Factors of Pulmonary Metastasectomy in Patients with Metastatic Transitional Cell Carcinoma

Luca Luzzi, The Thoracic and Cardiovascular Surgeon, 2016

Efficacy of Platinum Rechallenge in Metastatic Urothelial Carcinoma After Previous Platinum-Based Chemotherapy for Metastatic Disease

Risa L. Wong, The Oncologist, 2021

The Rapidly Evolving Landscape of First-Line Targeted Therapy in Metastatic Urothelial Cancer: A Systematic Review

Jean-Michel Lavoie, The Oncologist, 2021

Platinum ineligibility and survival outcomes in patients with advanced urothelial carcinoma receiving first-line treatment

Shilpa Gupta, JNCI: Journal of the National Cancer Institute

Outcome and prognostic factors in metastatic urothelial carcinoma patients receiving second-line chemotherapy: an analysis of real-world clinical practice data ...

Ryuji Matsumoto, Japanese Journal of Clinical Oncology, 2018

Erdafitinib for locally advanced or metastatic urothelial carcinoma

Arlene O Siefker-Radtke, American Journal of Health-System Pharmacy-1, 2022

References

  1. ung H, Ferlay J, Siegel RL. et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71 (03) 209-249
  2. aginala K, Barsouk A, Aluru JS, Rawla P, Padala SA, Barsouk A. Epidemiology of bladder cancer. Med Sci (Basel) 2020; 8 (01) 15
  3. CCN Clinical Practice Guidelines Version 1 (2022) for Bladder Cancer. Accessed April 12, 2022, at:  https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1417
  4. alderrama BP, González-Del-Alba A, Morales-Barrera R. et al. SEOM-SOGUG clinical guideline for localized muscle invasive and advanced bladder cancer (2021). Clin Transl Oncol 2022; 24 (04) 613-624
  5. ational Cancer Institute. Surveillance, Epidemiology, and End Results (SEER) on survival rates for bladder cancer. Accessed April 26, 2022, at:  https://www.cancer.org/cancer/bladder-cancer/detection-diagnosis-staging/survival-rates.html
  6. ishra V, Balasubramaniam G. Urinary bladder cancer and its associated factors – an epidemiological overview. Indian J Med Sci 2021; 73: 239-248
  7. bid A, Sen S, Bandyopadhyay R. Clinicopathological study of urothelial neoplasms in urinary bladder with special reference to expression of Her2/neu and Ki-67 in malignant lesions. Indian J Pathol Oncol 2021; 8: 369-376
  8. rakash G, Pal M, Odaiyappan K. et al. Bladder cancer demographics and outcome data from 2013 at a tertiary cancer hospital in India. Indian J Cancer 2019; 56 (01) 54-58
  9. xford Centre for Evidence-Based Medicine: levels of evidence. Accessed April 29, 2022, at:  https://www.cebm.ox.ac.uk/resources/levels-of-evidence/oxford-centre-for-evidence-based-medicine-levels-of-evidence-march-2009
  10. Nasa P, Jain R, Juneja D. Delphi methodology in healthcare research: how to decide its appropriateness. World J Methodol 2021; 11 (04) 116-129
  11. Scharl A, Thomssen C, Harbeck N, Müller V. AGO recommendations for diagnosis and treatment of patients with early breast cancer: update 2013. Breast Care (Basel) 2013; 8 (03) 174-180
  12. Jünger S, Payne S, Brearley S, Ploenes V, Radbruch L. Consensus building in palliative care: a Europe-wide Delphi study on common understandings and conceptual differences. J Pain Symptom Manage 2012; 44 (02) 192-205
  13. Galsky MD, Hahn NM, Rosenberg J. et al. Treatment of patients with metastatic urothelial cancer “unfit” for cisplatin-based chemotherapy. J Clin Oncol 2011; 29 (17) 2432-2438
  14. Park SB, Krishnan AV, Lin CS, Goldstein D, Friedlander M, Kiernan MC. Mechanisms underlying chemotherapy-induced neurotoxicity and the potential for neuroprotective strategies. Curr Med Chem 2008; 15 (29) 3081-3094
  15. National Cancer Institute. Cancer Therapy Evaluation Program: Common Terminology Criteria for Adverse Events (CTCAE) version 5. Accessed May 05, 2022, at:  https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/ctcae_v5_quick_reference_8.5x1/211.pdf
  16. Raja W, Mir MH, Dar I, Banday MA, Ahmad I. Cisplatin induced paroxysmal supraventricular tachycardia. Indian J Med Paediatr Oncol 2013; 34 (04) 330-332
  17. Gupta S, Bellmunt J, Plimack ER. et al. Defining “platinum-ineligible” patients with metastatic urothelial cancer (mUC). J Clin Oncol 2019; 37: 451
  18. De Santis M, Bellmunt J, Mead G. et al. Randomized phase II/III trial assessing gemcitabine/ carboplatin and methotrexate/carboplatin/vinblastine in patients with advanced urothelial cancer “unfit” for cisplatin-based chemotherapy: phase II–results of EORTC study 30986. J Clin Oncol 2009; 27 (33) 5634-5639
  19. Rouanne M, Radulescu C, Adam J, Allory Y. PD-L1 testing in urothelial bladder cancer: essentials of clinical practice. World J Urol 2021; 39 (05) 1345-1355
  20. Grivas P, Agarwal N, Pal S. et al. Avelumab first-line maintenance in locally advanced or metastatic urothelial carcinoma: applying clinical trial findings to clinical practice. Cancer Treat Rev 2021; 97: 102187
  21. Powles T, Park SH, Voog E. et al. Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. N Engl J Med 2020; 383 (13) 1218-1230
  22. Rui X, Gu TT, Pan HF, Zhang HZ. Evaluation of PD-L1 biomarker for immune checkpoint inhibitor (PD-1/PD-L1 inhibitors) treatments for urothelial carcinoma patients: a meta-analysis. Int Immunopharmacol 2019; 67: 378-385
  23. Casadei C, Dizman N, Schepisi G. et al. Targeted therapies for advanced bladder cancer: new strategies with FGFR inhibitors. Ther Adv Med Oncol 2019; 11: 1758835919890285
  24. Kwon WA, Seo HK. Emerging agents for the treatment of metastatic urothelial cancer. Investig Clin Urol 2021; 62 (03) 243-255
  25. Kacew A, Sweis RF.  FGFR3 alterations in the era of immunotherapy for urothelial bladder cancer. Front Immunol 2020; 11: 575258
  26. De Santis M, Bellmunt J, Mead G. et al. Randomized phase II/III trial assessing gemcitabine/carboplatin and methotrexate/carboplatin/vinblastine in patients with advanced urothelial cancer who are unfit for cisplatin-based chemotherapy: EORTC study 30986. J Clin Oncol 2012; 30 (02) 191-199
  27. Balar AV, Galsky MD, Rosenberg JE. et al; IMvigor210 Study Group. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. [published correction appears in Lancet. 26 August 2017;390(10097):848]. Lancet 2017; 389 (10064): 67-76
  28. FDA alerts health care professionals and oncology clinical investigators about an efficacy issue identified in clinical trials for some patients taking Keytruda (pembrolizumab) or Tecentriq (atezolizumab) as monotherapy to treat urothelial cancer with low expression of PD-L1. Accessed May 18, 2022, at:  https://www.fda.gov/drugs/drug-safety-and-availability/fda-alerts-health-care-professionals-and-oncology-clinical-investigators-about-efficacy-issue
  29. ODAC says ‘yes’ to continued approval of atezolizumab for cisplatin-ineligible locally advanced or metastatic UC. Accessed October 7, 2022, at:  https://www.targetedonc.com/view/odac-says-yes-to-continued-approval-of-atezolizumab-for-cisplatin-ineligible-locally-advanced-metastatic-uc
  30. Balar AV, Castellano D, O'Donnell PH. et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol 2017; 18 (11) 1483-1492
  31. Vuky J, Balar AV, Castellano D. et al. Long-term outcomes in KEYNOTE-052: Phase II study investigating first-line pembrolizumab in cisplatin-ineligible patients with locally advanced or metastatic urothelial cancer. J Clin Oncol 2020; 38 (23) 2658-2666
  32. Hugar LA, Lopa SH, Yabes JG. et al. Palliative care use amongst patients with bladder cancer. BJU Int 2019; 123 (06) 968-975
  33. Hugar LA, Wulff-Burchfield EM, Winzelberg GS, Jacobs BL, Davies BJ. Incorporating palliative care principles to improve patient care and quality of life in urologic oncology. Nat Rev Urol 2021; 18 (10) 623-635
  34. Castagneto B, Zai S, Marenco D. et al. Single-agent gemcitabine in previously untreated elderly patients with advanced bladder carcinoma: response to treatment and correlation with the comprehensive geriatric assessment. Oncology 2004; 67 (01) 27-32
  35. FDA approves updated indication for Merck's KEYTRUDA® (pembrolizumab) for treatment of certain patients with urothelial carcinoma (bladder cancer). Accessed Oct 07, 2022, at:  https://www.merck.com/news/fda-approves-updated-indication-for-mercks-keytruda-pembrolizumab-for-treatment-of-certain-patients-with-urothelial-carcinoma-bladder-cancer/
  36. Galsky MD, Arija JÁA, Bamias A. et al; IMvigor130 Study Group. Atezolizumab with or without chemotherapy in metastatic urothelial cancer (IMvigor130): a multicentre, randomised, placebo-controlled phase 3 trial. Lancet 2020; 395 (10236): 1547-1557
  37. Powles T, Csőszi T, Özgüroğlu M. et al; KEYNOTE-361 Investigators. Pembrolizumab alone or combined with chemotherapy versus chemotherapy as first-line therapy for advanced urothelial carcinoma (KEYNOTE-361): a randomised, open-label, phase 3 trial. Lancet Oncol 2021; 22 (07) 931-945
  38. Pérez-Valderrama B, Powles T, Sridhar SS. et al. Avelumab first-line (1L) maintenance for advanced urothelial carcinoma (UC): long-term follow-up results from the JAVELIN Bladder 100 trial. J Clin Oncol 2022; 40: 4559
  39. Galsky MD, Mortazavi A, Milowsky MI. et al. Randomized double-blind phase II study of maintenance pembrolizumab versus placebo after first-line chemotherapy in patients with metastatic urothelial cancer. J Clin Oncol 2020; 38 (16) 1797-1806
  40. Bellmunt J, de Wit R, Vaughn DJ. et al; KEYNOTE-045 Investigators. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 2017; 376 (11) 1015-1026
  41. Sharma P, Retz M, Siefker-Radtke A. et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol 2017; 18 (03) 312-322
  42. Apolo AB, Ellerton JA, Infante JR. et al. Avelumab as second-line therapy for metastatic, platinum-treated urothelial carcinoma in the phase Ib JAVELIN Solid Tumor study: 2-year updated efficacy and safety analysis. J Immunother Cancer 2020; 8 (02) e001246
  43. Loriot Y, Necchi A, Park SH. et al; BLC2001 Study Group. Erdafitinib in locally advanced or metastatic urothelial carcinoma. N Engl J Med 2019; 381 (04) 338-348
  44. Atezolizumab Indication in US Withdrawn for Previously Treated Metastatic Urothelial Cancer. Accessed May 18, 2022, at:  https://www.cancernetwork.com/view/atezolizumab-indication-in-us-withdrawn-for-previously-treated-metastatic-urothelial-cancer
  45. 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. [published correction appears in Lancet. 2018 Oct 20;392(10156):1402]. Lancet 2018; 391 (10122): 748-757
  46. Powles T, Rosenberg JE, Sonpavde GP. et al. Enfortumab vedotin in previously treated advanced urothelial carcinoma. N Engl J Med 2021; 384 (12) 1125-1135
  47. Tagawa ST, Balar AV, Petrylak DP. et al. TROPHY-U-01: A phase II open-label study of sacituzumab govitecan in patients with metastatic urothelial carcinoma progressing after platinum-based chemotherapy and checkpoint inhibitors. J Clin Oncol 2021; 39 (22) 2474-2485