Novel therapies in urothelial carcinoma: a biomarker-driven approach

Novel therapies in urothelial carcinoma: a biomarker-driven approach

Article type: Review Title: Novel therapies in urothelial carcinoma: a biomarker-driven approach Authors: J. E. Rosenberg G. Iyer Affiliations: Mem...

913KB Sizes 0 Downloads 45 Views

Article type: Review

Title: Novel therapies in urothelial carcinoma: a biomarker-driven approach

Authors: J. E. Rosenberg G. Iyer

Affiliations: Memorial Sloan Kettering Cancer Center, New York, NY, USA

Corresponding Author: Dr. Jonathan E. Rosenberg Memorial Sloan Kettering Cancer Center 1275 York Avenue New York NY 10065 USA Tel.: 001 646 422 4461 Email: [email protected]



Review article word count: max. 4000

© The Author(s) 2018. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: [email protected].

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Abstract Urothelial malignancies, including carcinomas of the bladder, ureters, and renal pelvis comprised approximately 8% of new cancer cases in the United States in 2016. In the metastatic setting, 15% of patients exhibit long-term survival following cisplatin-based chemotherapy and in patients with recurrent disease, response rates to second-line chemotherapy are generally 15-20% with a 3-month progression-free survival. However, recent advances in immunotherapy represent an opportunity to significantly improve patient outcomes. Moreover, the advent of next-generation sequencing (NGS) has resulted in both an improved understanding of the fundamental genetic changes that characterize urothelial carcinoma (UC) and identification of several candidate biomarkers of response to various therapies. Incorporation of prospective genotyping into clinical trials will allow for the identification and enrichment of patients most likely to respond to specific targeted therapies and chemotherapy. Combining different therapeutic classes to enhance outcomes is also an area of active research in UC.

Keywords: Urothelial cancer, urinary bladder, metastasis, predictive biomarkers, gene sequencing

Key message: New developments in biomarker-driven approaches, based on an understanding of the genetic alterations underlying urothelial malignancies, are guiding the development of novel therapies and therapeutic strategies for this disease.

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Introduction In 2017, approximately 150,000 people will be diagnosed with UC in the United States and around 32,000 are likely to die from their disease [ACS 2017]. Urothelial malignancies include carcinomas of the bladder, ureters, and renal pelvis and are more common in men than women (3:1) and in Caucasians than African Americans (2:1) [Hurwitz et al. 2016]. The most common of the urothelial malignancies, accounting for an estimated 54% of cases and 52% of deaths, is urinary bladder cancer [ACS 2017]. Tobacco smoking and occupational exposures (e.g., arylamines) are the leading risk factors for developing urothelial bladder carcinoma in the United States and contribute to an increased risk of recurrence [Wilcox et al. 2016; Hurwitz et al. 2016]. Disease stage is closely related to prognosis. Patients with well-differentiated, non-invasive primary papillary lesions (Ta) without carcinoma in situ have a 95% survival rate, whereas those with higher-grade lesions that have invaded the sub-epithelial lamina propria (T1) have a 10-year survival rate of 50%; muscle-invasive urothelial carcinoma (T2) has a 5-year disease-specific survival rate of 40%–65% [Hurwitz et al. 2016]. However, a 5-year survival rate of only 0%–30% can be expected if lymph nodes are involved (Stage IV) [Hurwitz et al. 2016].

Patients with organ-confined disease are managed with surgical intervention, chemotherapy, radiotherapy, or a combination of these treatment modalities. Transurethral resection of the bladder tumor (TURBT) with or without intravesical therapy is generally recommended in patients with superficial bladder cancer [Bellmunt et al. 2014]. Patients with high-grade, recurrent nonmuscle invasive bladder cancer can be managed with radical cystectomy (RC) prior to the development of muscle invasion [Bellmunt et al. 2014; Hurwitz et al. 2016]. Patients with muscleinvasive disease are optimally managed with neoadjuvant cisplatin-based chemotherapy in eligible patients followed by RC and a bilateral pelvic lymph node dissection, or trimodality therapy consisting of a maximal TURBT followed by chemo-radiotherapy.

The chemotherapeutic management of patients with muscle-invasive and metastatic urothelial carcinoma (UC) has changed little in over 20 years. Platinum-based combinations remain the standard-of-care in perioperative (as neoadjuvant and adjuvant therapy) and first-line metastatic settings [Bellmunt et al. 2014]. Cisplatin-containing combination chemotherapy with either gemcitabine or methotrexate/vinblastine/doxorubicin is recommended for those patients with advanced surgically unresectable or metastatic disease who are able to tolerate cisplatin [Bellmunt et al. 2014]. Despite refinements in treatment schedules and toxicity management, median survival achieved with these treatments is 12-15 months, either in clinical trials [Loehrer et al. 1992; von der

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Masse et al. 2000; von der Masse et al. 2005] or in real-world settings [Robinson et al. 2016]. To address these limitations, intensification strategies including new triplet or quadruplet strategies, or dose-dense therapies, were developed with subsequent improvements in objective response rates but not median overall survival [Galsky et al. 2007; Milowsky et al. 2009; Bellmunt et al. 2012; Necchi et al. 2014].

Advances in the understanding and identification of genetic alterations underlying the spectrum of urothelial malignancies hold out hope for improved outcomes through the development of targeted therapies. Additionally, the recent approval of five immunotherapy agents in the United States has led to a transformation in the management of patients with advanced UC, although only a minority of patients displays responses to this class of agents and predictive biomarkers of sensitivity to immunotherapy have yet to be validated [Rosenberg et al 2016; Sharma et al, 2017]. Despite recent treatment advances, there remains an unmet need for the improved management of patients with UC. This paper provides an overview of the current state-of-the art of therapy for urothelial malignancies, provides insight into the paucity of successful treatments, and offers some perspectives on future directions for additional research, with a focus on the prospective benefits of using biomarker-driven approaches to patient management.

Immunotherapy for metastatic urothelial malignancies: Current state-of-the-art Immunotherapy has fundamentally altered the treatment paradigm for metastatic UC. Until recently, patients with metastatic disease had few treatment options after failure of platinum-based chemotherapy strategies. Single agent chemotherapies in the second-line setting generally display response rates of less than 30%, and more frequently 10-15%. An analysis of studies investigating doublet chemotherapy regimens as salvage therapy after initial platinum-based treatment showed no extended survival benefits compared with single agents [Raggi et al. 2016] and such regimens are frequently limited by toxicity [Oing et al. 2016]. In 2009, vinflunine, a fluorinated Vinca alkaloid, was approved by the European Medicines Agency (EMA) as monotherapy for the treatment of adult patients with advanced or metastatic urothelial carcinoma after failure of a prior platinumcontaining regimen [Javlor SmPC 2009] and is a recommended therapy in treatment guidelines [Bellmunt et al 2014]. This approval is based upon a phase III study of vinflunine plus best supportive care versus best supportive care alone for patients with metastatic UC who had progressed on platinum-based chemotherapy. A 2-month median survival advantage was observed for vinflunine plus best supportive care (6.9 months vs 4.3 months for best supportive care alone, p=0.040) in the

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

eligible patient population. Significant improvements in overall response rate and progression-free survival were also noted with the addition of vinflunine.

In 2016, atezolizumab, a programmed death ligand 1 (PD-L1)-blocking antibody, received accelerated United States Food and Drug Administration (FDA) approval for locally advanced or metastatic UC in patients with disease progression during or following platinum-containing chemotherapy or within 12 months of peri-operative platinum-containing chemotherapy [Tecentriq PI 2016]. This approval was based on a phase 2 study in patients with metastatic UC who had progressed on prior platinum-based chemotherapy (Rosenberg et al, Lancet 2016). In this trial, the overall response rate was 15%, significantly higher than a historical response rate of 10% with chemotherapy. Responses were also organized based upon PD-L1 expression (using the Ventana SP142 assay) on immune infiltrating cells (IC) and were found to be highest in patients with ≥5% infiltration (IC2/3, 26%); notably, responses were also observed in IC0 (8%) and IC1 (10%) patients. At 11.7 months median follow-up, 84% of responders continued to show objective responses, underscoring a durability not usually seen with standard chemotherapy. In April 2017, accelerated approval was also granted for atezolizumab as a first-line treatment for patients with locally advanced or metastatic UC who are ineligible for cisplatin-based chemotherapy or who have disease progression at least 12 months after receiving perioperative chemotherapy [Tecentriq Approval History 2017]. In a phase 2 study of atezolizumab in this patient population, the objective response rate was 23%, including a 9% complete response rate, with a median overall survival of 15.9 months, all significantly higher than observed with carboplatin-based chemotherapy. In both studies, tumor mutation load was significantly higher in responders.

Following the approval of atezolizumab, the results of an open-label, multi-center phase III study were reported (IMvigor211) in which patients with progressive metastatic urothelial carcinoma following platinum chemotherapy were randomized to receive either atezolizumab or treating investigator’s choice of paclitaxel, docetaxel, or vinflunine [Powles et al. 2018]. The primary endpoint of overall survival within a predefined patient population with IC2/3 PD-L1 expression was not significantly different between atezolizumab and chemotherapy (median OS 11.1 months vs 10.6 months, respectively, HR 0.87, p=0.41). The objective response rate was also similar in both groups, but the rate of ongoing responses was higher with atezolizumab therapy (62% vs 20% with chemotherapy). Potential reasons for the lack of improvement in OS include the observation that survival with vinflunine was higher than expected within the statistical model for this study and the fact that IC2/3 patients displayed improved survival and response independent of treatment

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

modality. The association of tumor mutation burden with survival was evaluated in an exploratory analysis of the intent to treat patient population. High tumor mutation burden (defined as being higher than the median total mutation burden within assessable patients) was associated with a non-significant improvement in overall survival with atezolizumab therapy (11.3 months vs 8.3 months with chemotherapy, HR 0.68). Comparison of survival between treatment arms within high total mutation burden patients with IC2/3 PD-L1 expression showed a longer median survival with atezolizumab (17.8 months) versus chemotherapy (10.6 months).

Nivolumab, a PD-1 immune checkpoint inhibitor antibody, received accelerated approval from the FDA in February 2017 in patients with locally advanced or metastatic UC whose disease has progressed on platinum chemotherapy [Opdivo PI]. The phase 2 CHECKMATE 275 study explored nivolumab monotherapy in patients with metastatic, pre-treated UC and showed a response rate of 19.6% across all patient subgroups. When segregated by PD-L1 expression status on tumor cells, the response rate was 28.4% in patients with ≥5% expression versus 16.1% in patients with <1% PD-L1 expression. The median overall survival was 8.74 months (11.3 months in patients with ≥1% PD-L1 expression versus 5.95 months in patients with <1% expression). Nivolumab has also been studied in two dose combinations with ipilimumab (an anti-CTLA-4 antibody) in patients with locally advanced or metastatic UC previously treated with platinum-based chemotherapy; interim results showed that high response rates were achieved with combination therapy, but with higher toxicity [Sharma 2016a] rates, which is similar to observations in treatment-naïve melanoma patients [Larkin et al. 2015; Hodi et al. 2016]. Further study is needed to evaluate the clinical utility of this combination.

Durvalumab was investigated in 191 patients with advanced or metastatic UC following progression on prior chemotherapy or who were ineligible or refused chemotherapy in a phase I/II multi-center trial. In the latest evaluation, patients with Stage IV disease (98 PD-L1–positive, 79 PD-L1–negative [1 patient with unknown PD-L1 status]) were treated with intravenous durvalumab 10 mg/kg every 2 weeks for up to 12 months, and 99.5% had received one or more prior therapies. After a median follow-up of 5.8 months, the overall response rate (ORR) was 17.8% (27.6% in the PD-L1–positive subgroup versus 5.1% in the PD-L1–negative subgroup). The initial 20 patients on study were enrolled regardless of PD-L1 expression, while subsequent patients were required to have ≥5% PD-L1 expression (using the Ventana SP263 assay) on tumor cells for enrolment. PD-L1 expression was defined as positive if either ≥25% of tumor cells or ≥25% of immune cells expressed PD-L1. Using this definition, the ORR in PD-L1 positive patients was 46.4% versus 0% in PD-L1 negative patients.

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Avelumab, a fully humanized monoclonal PD-L1 antibody, is in phase 1b investigation in patients with solid tumors (JAVELIN trial program); an analysis of patients with metastatic UC showed promising and durable clinical activity in heavily pre-treated patients [Apolo et al. 2017]. In this study, the unconfirmed ORR was 17.4%, and 25.4% in PD-L1–positive patients (defined using a ≥5% cutoff for tumor cell staining, Dako PD-L1 IHC kit) versus 13.2% in PD-L1–negative patients. Avelumab is also being assessed in a study (JAVELIN Bladder 100) as maintenance treatment for patients with locally advanced/metastatic UC whose disease did not progress after first-line platinum-based treatment [Powles et al. 2016a], with primary outcome results expected in the latter half of 2019.

Pembrolizumab is a humanized monoclonal antibody directed against PD-1. In a phase 3 study of pembrolizumab as second-line therapy in patients with advanced UC that recurred or progressed after platinum-based chemotherapy versus investigator’s choice of docetaxel, paclitaxel, or vinflunine (KEYNOTE 045), pembrolizumab was associated with significantly longer overall survival (10.3 months vs 7.4 months, Hazard Ratio 0.70, p<0.001) [Bellmunt et al. 2017; Bajorin et al. 2017]. Notably, the median duration of response was longer (not reached) in the pembrolizumab treated patients versus 4.4 months with chemotherapy. PD-L1 expression using a 10% or greater combined positive score in tumor and infiltrating immune cells (22C3 pharmDx assay, Dako) was not associated with a statistically significant difference in response rate. This led to full FDA approval for patients previously treated with platinum-based chemotherapy. The phase 2 KEYNOTE 052 study investigated pembrolizumab in cisplatin-ineligible patients; the overall response rate was 24%, including 5% complete responses [Balar et al. 2017]. At an 8-month median follow-up, 74% of patients continued to respond and the median response duration was not reached. The results of this study supported an accelerated FDA approval in cisplatin-ineligible metastatic urothelial carcinoma patients.

The observation that patients without PD-1 or PD-L1 expression may still exhibit responses to immune checkpoint inhibitors underscores the fact that these biomarkers cannot be used in isolation to make treatment decisions. Moreover, several caveats to the application of routine PD-1/ PD-L1 staining exist, including the use of different assays to test for PD-1 and PD-L1 expression, variability in the definitions and cutoffs for positivity (the use of tumor-cell vs immune infiltrating cell surface expression), differences in familiarity with interpreting test results, intratumor heterogeneity of expression, and the impact of prior treatments upon expression levels over time. As discussed below in more detail, several other tumor-specific factors likely contribute to checkpoint blockade sensitivity in UC, including expression subtyping and mutation profile (for example, the presence of

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

DNA damage response gene alterations or FGFR3 alterations). Additionally, host-specific factors, such as T-cell receptor clonality, tumor-infiltrating lymphocytes, and others, also modulates response to checkpoint blockade. A significant challenge will be to understand the relative impact of each of these biomarkers in predicting for checkpoint blockade sensitivity and resistance and implementing testing for these biomarkers in a rapid, prospective fashion prior to initiation of therapy.

Immunotherapies in development A number of other checkpoint inhibitors, interleukins (ILs), and novel immune molecules have been identified as potential immunotherapeutic agents in muscle-invasive and metastatic urothelial cancer (Figure 1).

Novel therapies and therapeutic strategies in urothelial malignancies Identification of prognostic biomarkers (to provide information on overall cancer outcome in patients and/or to facilitate cancer diagnosis) and predictive biomarkers (to inform treatment decisions) are integral to a personalized oncology approach in cancer. A number of strategies have been used to identify clinically useful biomarkers for UC. The Cancer Genome Atlas (TCGA) analyzed 412 muscle-invasive, high-grade bladder cancers using multiple platforms, including whole exome sequencing, RNA sequencing, DNA methylation profiling, and others. The total mutation burden was high, similar to that of melanoma and non-small cell lung cancers; moreover, the most mutations were clonal and were detected within the context of an APOBEC mutation signature. (Figure 2) [Robertson 2017]. RNA expression subtyping delineated 5 distinct molecular subtypes of bladder cancer, including luminal, luminal-papillary, luminal-infiltrated, basal-squamous, and neuronal. The neuronal subtype was associated with the worst survival of the 5 subtypes and was enriched for TP53 and RB1 alterations while the luminal infiltrated subtype has been correlated with response to clinical trials of immune checkpoint blockade in urothelial carcinoma. While several groups have identified similar molecular subtypes based upon expression profiling of urothelial carcinoma, the predictive and prognostic characteristics of these subtypes are as yet unclear and require prospective validation. A novel gene expression algorithm, Co-eXpression ExtrapolatioN (COXEN), is an informatics-based approach to predict sensitivity of independent cell line panels and patient responses to therapeutic agents [Smith et al. 2010; Smith et al. 2011]. The COXEN model combines in vitro and in vivo molecular profiling of cancer cell lines and drug responsiveness using the NCI-60 and BLA-40 cancer cell line panels (Figure 3). For the purposes of prospective validation, the COXEN algorithm is being incorporated into the SWOG S1314 study in

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

which muscle-invasive bladder cancer patients are randomized to two different neoadjuvant chemotherapy regimens. The results of COXEN will be correlated with response to chemotherapy at cystectomy to gauge the capability of the algorithm to predict for chemo-sensitivity. Additional correlative analysis of pre-treatment specimens and post-chemotherapy residual tumors is planned, including whole transcriptome profiling to facilitate correlation of the published molecular subtypes with response and survival.

Next-generation sequencing has enabled a more detailed analysis of the underlying genetic changes characterizing UC and identification of potential new therapeutic targets. Although most tumors harbor potentially tractable genomic alterations that may predict for response to target-selective agents, substantial genomic heterogeneity has been identified in urothelial bladder cancer [Faltas et al 2016]. Additionally, subclonal mutational heterogeneity, which may contribute to chemotherapy resistance [Liu et al. 2017], poses an obstacle to the effective application of targeted agents.

Among the promising therapeutic targets, FGFR3, is of particular interest. It encodes for a transmembrane protein, fibroblast growth factor receptor-3, involved in regulating cell proliferation and angiogenesis; putative downstream signaling pathways include the PI3K-AKT, PKC, and Ras/MAPK pathways (Figure 4). The most common FGFR3 alterations in UC are activating point mutations. Dysregulation of FGFR3 signaling occurs in approximately 80% of non-invasive and 50% of invasive bladder cancers, through mutation, overexpression, or both, and over-expression is also common in tumors with no detectable mutations, including in muscle-invasive tumors [Tomlinson et al. 2007]. FGFR3 alterations have been reported in up to 21% of patients with high-grade and advanced stage bladder UC. Activating FGFR3 fusions have also been described in UC [Glaser et al. 2017] [CGARN 2014]. Urothelial cell lines harboring these fusions are highly sensitive to FGFRselective agents and responses to FGFR-directed inhibitors have been observed in patients with FGFR3 fusion positive UC. Thus, these alterations may serve as predictive biomarkers that could aid patient selection for FGFR-targeted therapy [Williams et al. 2013; Costa et al. 2016].

Although dovitinib, a multi-targeted inhibitor of tyrosine kinases including FGFR3, has not shown significant activity in UC (due in part to challenges with appropriate patient selection using the available sequencing platforms at the time), other agents targeting FGFR3-mutant UC remain under investigation. BGJ398 is an FGFR1–3 inhibitor that showed promising anti-tumor activity in patients with advanced solid tumors, including those with UC [Nogova et al. 2017]. These data led to evaluation of drug efficacy in an extended cohort of patients with advanced/metastatic UC and

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

activating FGFR3 mutations/fusions after platinum-based chemotherapy [Pal et al. 2016]. Seventy-six percent of patients had received 2 or more prior treatments. The ORR was 36%, including 8 partial responses, suggesting clinical benefit with FGFR3 signaling blockade in this patient population. Clinical activity has also been observed with erdafitinib, a pan-FGFR inhibitor, in patients with metastatic UC and this drug recently received FDA breakthrough designation for this disease. In a phase 2 study of erdafitinib in patients with metastatic UC (BLC2001) and pre-specified FGFR alterations, the overall response rate was 42% (3% CR rate) [Siefker-Radtke et al. 2018]. Notably, the response rate was 59% in the subset of patients who had received prior immune checkpoint blockade.

Inhibition of mechanistic target of rapamycin (mTOR) is also a target for therapy in UC under investigation. Everolimus, which blocks mTOR signaling, was tested in two phase 2 studies in patients with advanced UC [Seront et al. 2012; Milowsky et al. 2013]. The study by Milowsky et al did not reach the primary endpoint of an improvement in 2-month progression-free survival compared to historical rates. In the study by Seront et al, a disease control rate of 27% was seen, with 2 partial responses. Everolimus in combination with paclitaxel was not effective as a second-line treatment for UC in the German AUO Trial AB 35/09 [Niegisch et al. 2015], showing a 13% overall response rate. In addition, a regimen combining everolimus with gemcitabine and split-dose cisplatin in advanced UC was not feasible due to dose-limiting hematologic toxicities [Abida et al. 2016]. Despite these outcomes, evidence to support the use of everolimus in specific molecular contexts exists, with some patients harboring specific mutations in the mTOR pathway experiencing exceptional responses to everolimus-containing therapy. The activating mutations mTORE2419K and mTORE2014K were found in a patient tumor that was exquisitely sensitive to mTOR inhibition; the patient had platinum- and taxane-refractory UC and experienced a complete radiologic response that lasted for 14 months on pazopanib plus everolimus [Wagle et al. 2014]. Loss-of-function mutations in TSC1 and NF2, both regulators of mTOR pathway activation, have been correlated with durable everolimus sensitivity in an exceptional responder that has been ongoing for greater than 6 years [Iyer et al. 2012]; additionally, NF2 mutations have been identified as conferring significant response to everolimus-containing regimens in patients with urothelial malignancies [Ali et al. 2015]. Studies to investigate the clinical benefit of everolimus targeted to cancer patients with inactivating TSC1 or TSC2 mutations or activating MTOR mutations (NCT02201212) and NF2 mutations (NCT02352844) are on-going.

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Analysis of TCGA has also shown that epidermal growth factor receptor (EGFR) signaling pathways are upregulated in selected advanced UC tumors, with EGFR amplification in 11% of UC and ERBB2 amplification and mutation in 9% [TCGA 2014]. Lapatinib is a dual tyrosine kinase inhibitor of EGFR and HER2 that showed anti-tumor activity in a phase 2 study in a subset of 34 patients with EGFR/HER2-overexpressing tumors [Wülfing et al. 2009]. Conversely, in a study of 232 patients with metastatic bladder cancer who had clinical benefit from first-line chemotherapy and HER1/HER2positive status confirmed by centralized immunohistochemistry (IHC), no improvement in progression-free survival was observed with maintenance lapatinib compared to placebo [Powles et al. 2015]. Targeting mutant/amplified tumors may still be a useful strategy, however, and negative results may reflect a failure of proper patient selection due to the assay used to determine gene amplification or protein overexpression.

Afatinib, another dual EGFR and HER2 inhibitor, has demonstrated clinical activity in patients with platinum-refractory UC with ERBB2 or ERBB3 genetic alterations detected by quantitative polymerase chain reaction or fluorescent in situ hybridization [Choudhury et al. 2016]. Five of the 6 patients with ERBB2 and/or ERBB3 alterations achieved the primary endpoint of 3-month progression-free survival compared with none of the 15 patients without such alterations (P < 0.001); median time to progression/therapy discontinuation was 6.6 months in patients with ERBB2/ERBB3 alterations versus 1.4 months in those without alterations (P < 0.001). This study suggests that selection by genomic alterations may be more relevant to the development of urothelial carcinoma than protein expression as detected by IHC.

Overall mutational load is also emerging as an additional biomarker of response in advanced UC, at least in patients treated with PD-1-blocking agents (Table 1). The phase 2 studies of atezolizumab in patients with locally advanced and metastatic UC who have progressed following treatment with platinum-based chemotherapy [Rosenberg et al. 2016a] and as first-line treatment in cisplatinineligible patients with locally advanced and metastatic UC [Balar et al. 2017a] showed that mutation load was consistently associated with improved response to atezolizumab therapy.

Combining various immunotherapies may also yield benefits surpassing those of monotherapy, and several combinations are currently being examined, including combination immune checkpoint blockade plus chemotherapy as well as with novel agents (both targeted therapies and immune modulating agents). A number of trials are examining the utility of combining platinum-based chemotherapy with checkpoint blockade in the first-line setting for metastatic urothelial carcinoma

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

in an attempt to leverage the response rates observed with platinum chemotherapy in this disease with the durability seen with checkpoint blockade. A study of durvalumab as first-line monotherapy or in combination with tremelimumab, an anti-CTLA-4 antibody, in patients with unresectable stage IV urothelial bladder cancer (the phase 3 DANUBE study) is recruiting patients [Powles et al. 2016b], with primary outcome results expected in 2018. The BISCAY trial, an open-label, randomized, multidrug, biomarker-directed, phase 1b study in patients with muscle-invasive bladder cancer who have failed at least one prior platinum regimen, will assess immunotherapy with durvalumab as monotherapy and in combination with select targeted therapies. Treatment assignment is based upon the tumor biomarker profile, and includes: olaparib, a PARP inhibitor; vistusertib, an mTOR inhibitor; AZD4547, an FGFR inhibitor; and AZD1775, a WEE1 inhibitor. The incorporation of prospective molecular analysis of tumors to inform assignment to a specific therapy could optimize the efficacy of small molecule inhibitors and possibly enhance responses to checkpoint blockade through the generation of neoantigens in this setting. Multiple checkpoint inhibitors are currently being investigated in combination with novel immunotherapies to enhance or overcome resistance to PD-1 blockade in patients with advanced solid tumors. Some of these include combinations with GSK3174998, an agonist of OX40, a potent costimulatory tumor necrosis factor receptor expressed on activated CD4+ and CD8+ T cells; MK-4280, targeting the lymphocyte-activation gene 3 (LAG3) receptor; NKTR-214, a CD122 agonist; TSR-022, an anti-T cell immunoglobulin and mucin containing protein-3 (TIM-3) antibody; BMS-98625, an IDO1 inhibitor; and CDX-1127, targeting CD27, a lymphocyte-specific member of the tumor necrosis factor receptor.

Re-conceptualizing targeted therapy Platinum chemotherapy as targeted therapy As discussed above, platinum-based combinations remain the standard-of-care for neoadjuvant chemotherapy (NAC). Cisplatin-based NAC is associated with improved survival, with a 14%–25% relative risk reduction for death from muscle-invasive UC [International collaboration of trialists 1999; Grossman et al. 2003; ABC Collaboration 2005]. The benefits of this approach are most apparent in patients with pathologic down-staging at the time of surgical resection, but the molecular determinants of response to cisplatin are unproven. Identifying extreme responders to platinum chemotherapy would therefore allow targeting of this treatment to those who would benefit most, and spare those who would not from the side effects of treatment.

Of note, somatic genomic alterations in DNA damage repair (DDR)-associated genes have been associated with benefit from both NAC and PD1/PD-L1 blockade. An initial extreme responder

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

analysis of patients with complete responses or residual carcinoma in situ within the bladder following neoadjuvant cisplatin-based chemotherapy and radical cystectomy for muscle-invasive disease versus patients with residual muscle-invasive or higher stage disease identified point mutations within the DNA helicase ERCC2 as strongly associated with response (9 of 25 responders harbored ERCC2 mutations versus 0 of 25 non-responders). These results have since been validated in a separate cohort of patients receiving neoadjuvant chemotherapy for muscle-invasive bladder cancer as part of a clinical trial in which ERCC2 mutations were also associated with improved survival (Liu et al, 2016). A subsequent retrospective analysis of 100 patients with locally advanced or metastatic UC who underwent exon capture sequencing revealed an association between the presence of DDR gene alterations and improved progression-free and overall survival [Teo et al, 2017a]. In this analysis, a higher mutation load was observed in DDR gene altered tumors. Defects in other DDR genes, including RB1, ATM, and FANCC, were detected in patients who responded to neoadjuvant chemotherapy [Plimack 2015]. Emerging evidence also suggests an association between overall response to PD1/PD-L1 blockade and the presence of somatic DDR alterations [Teo et al. 2017b].

A high frequency of patients with germline DDR mutations has also been reported. In a recent study, 12/25 pathogenic or likely pathogenic mutations in 22 patients with urothelial cancer originating from all sites within the urinary tract had heritable DDR gene alterations in CHEK2, BRCA1, BRCA2, ATM, BRIP1, and NBN [Carlo et al. 2017]. The role of these germline alterations will require validation.

Recent research suggests that RNA expression of non-coding microRNAs may contribute to the control of the gene expression patterns and can identify basal and luminal muscle-invasive bladder cancer subtypes. These could act as biomarkers for tumors with higher infiltration rates and provide candidate therapeutic targets [Ochoa et al. 2016]. Molecular subtyping to predict response to NAC in muscle-invasive bladder cancers is also an area which may allow prediction of outcomes with cisplatin-based combination chemotherapy. The benefit of NAC varies between molecular subtypes (as identified by transcriptome-wide microarray analysis), with patients with basal tumors achieving the best outcomes in a recent investigation: luminal/TCGA cluster I tumors could not be improved with NAC while claudin-low tumors appeared resistant to cisplatin-based chemotherapy [Seiler et al. 2017].

Immune-delivery of cytotoxic chemotherapy

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Antibodies can be used for the targeted delivery of chemotherapeutic molecules to tumor cells in the form of antibody-drug conjugates (ADCs); the monoclonal antibody targets a tumor antigen and cleavage of the linker leads to internalization and liberation of the cytotoxic component. Promising results from early phase studies have been seen with some ADCs in advanced UC, which are therefore in further evaluation. Enfortumab vedotin (ASG-22ME) is an ADC comprising a fully human antibody targeting nectin-4 and the potent microtubule-disrupting agent monomethyl auristatin E (MMAE). Moderate-to-strong staining for nectin-4 has been observed in 60% of bladder tumor specimens, with positive pre-clinical results in vitro and in vivo in several cancer models [Challita-Eid et al. 2016]. Initial interim clinical results show encouraging antitumor activity in patients with metastatic UC treated with enfortumab vedotin [Rosenberg et al. 2016b]. ASG-15ME is an ADC comprising an antibody targeting the bladder tumor antigen SLITRK6 and MMAE. It is in phase 1 investigation as monotherapy in patients with metastatic UC, with positive results from an exploratory analysis reported [Petrylak et al. 2016]. In addition, sacituzumab govitecan (IMMU-132), an ADC comprising an anti-TROP-2 antibody and the active metabolite of irinotecan (SN-38). TROP2 is a cell-surface receptor over-expressed by many human tumors, including those of the urinary bladder [Stepan et al. 2011], and in 6 patients with metastatic, platinum-resistant UC, 3 had a clinically significant response to sacituzumab govitecan [Faltas et al. 2016].

Conclusions Currently, platinum-based chemotherapy is standard for first-line treatment of metastatic UC; this has been the case for some time despite its limitations and attempts to re-imagine platinumcontaining management strategies. Although treatment options after failure of platinum-based therapy are improving, they remain suboptimal; new approaches are therefore needed. Novel biomarkers may allow rational patient selection and the range of targeted agents being investigated is promising, with particular interest in those targeting FGFR3. Despite promise in other areas, the validity of EGFR and HER2 alterations as predictive biomarkers for patient selection remains unclear in UC management. Reconceptualizing chemotherapy is also an area in which targeting of patients to maximize outcomes is being evaluated. The variety of novel therapies in development for UC and progress in biomarker-driven approaches is encouraging for enabling future shifts in the treatment paradigm to optimize individual patient management.

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Acknowledgements JER and GI thank Daniel Salamon for his writing assistance. Funding JER and GI are supported in part by P30 CA008748

Disclosures JER: Consultant for Agensys, Seattle Genetics, Roche/Genentech, Sanofi, BMS, Merck, Bayer, Eli Lilly, AstraZeneca, QED Therapeutics. Holds stock in Illumina. GI: None

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

References Abida W, Milowsky MI, Ostrovnaya I, Gerst SR, Rosenberg JE, Voss MH, Apolo AB, Regazzi AM, McCoy AS, Boyd ME, Bajorin DF. Phase I study of everolimus in combination with gemcitabine and split-dose cisplatin in advanced urothelial carcinoma. Bladder Cancer. 2016 Jan 7;2(1):111-117. Advanced Bladder Cancer (ABC) Meta-analysis Collaboration. Neoadjuvant chemotherapy in invasive bladder cancer: update of a systematic review and meta-analysis of individual patient data advanced bladder cancer (ABC) meta-analysis collaboration. Eur Urol. 2005 Aug;48(2):202-5; discussion 205-6. Ali SM, Miller VA, Ross JS, Pal SK. Exceptional response on addition of everolimus to taxane in urothelial carcinoma bearing an NF2 mutation. Eur Urol. 2015 Jun;67(6):1195-6. American Cancer Society: Cancer Facts and Figures 2017. Atlanta, Ga: American Cancer Society, 2017. Available at: https://old.cancer.org/acs/groups/content/@editorial/documents/document/acspc-048738.pdf. Last accessed XXXX. Apolo AB, Ellerton JA, Infante JR, et al. Updated efficacy and safety of avelumab in metastatic urothelial carcinoma (mUC): Pooled analysis from 2 cohorts of the phase 1b Javelin solid tumor study. J Clin Oncol 35, 2017 (suppl; abstr 4528). Apolo AB, Infante JR, Balmanoukian A, Patel MR, Wang D, Kelly K, Mega AE, Britten CD, Ravaud A, Mita AC, Safran H, Stinchcombe TE, Srdanov M, Gelb AB, Schlichting M, Chin K, Gulley JL. Avelumab, an anti-programmed death-ligand 1 antibody, in patients with refractory metastatic urothelial carcinoma: results from a multicenter, Phase 1b study. J Clin Oncol. 2017 Jul 1;35(19):2117-2124. Balar A, Bellmunt J, O’Donnell PH, et al. Pembrolizumab (pembro) as first-line therapy for advanced/unresectable or metastatic urothelial cancer: Preliminary results from the phase II KEYNOTE-052 study. Presented at: 2016 ESMO Congress; October 7-11, 2016; Copenhagen, Denmark. Abstract LBA32. 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 Nov;18(11):1483-1492. Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt J, Loriot Y, Necchi A, HoffmanCensits J, Perez-Gracia JL, Dawson NA, van der Heijden MS, Dreicer R, Srinivas S, Retz MM, Joseph RW, Drakaki A, Vaishampayan UN, Sridhar SS, Quinn DI, Durán I, Shaffer DR, Eigl BJ, Grivas PD, Yu EY, Li S, Kadel EE 3rd, Boyd Z, Bourgon R, Hegde PS, Mariathasan S, Thåström A, Abidoye OO, Fine GD, Bajorin DF; 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. Lancet. 2017a Jan 7;389(10064):67-76. Balar AV, Castellano D, O'Donnell PH, Grivas P, Vuky J, Powles T, Plimack ER, Hahn NM, de Wit R, Pang L, Savage MJ, Perini RF, Keefe SM, Bajorin D, Bellmunt J. First-line pembrolizumab in cisplatinineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE052): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017b Sep 26. pii: S14702045(17)30616-2. Bajorin DF, De With R, Vaugh DJ, Fradet Y, Lee J-L, Fong L, Vogelzan NJ, Climent MA, Petrylak DP, Choueiri TK, Necchi A, Gerritsen W, Gurney H, Quinn DI, Culine S, Sternberg CN, Mai Y, Puhlmann M, Perini RF, Bellmunt J. Planned survival analysis from KEYNOTE-045: Phase 3, open-label study of pembrolizumab (pembro) versus paclitaxel, docetaxel, or vinflunine in recurrent, advanced urothelial cancer (UC). Presented at: 2017 ASCO Congress; June 1-5, 2017; Chicago. IL, USA. Abstract 4501. Bellmunt J, von der Maase H, Mead GM, Skoneczna I, De Santis M, Daugaard G, Boehle A, Chevreau C, Paz-Ares L, Laufman LR, Winquist E, Raghavan D, Marreaud S, Collette S, Sylvester R, de Wit R.

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Randomized phase III study comparing paclitaxel/cisplatin/gemcitabine and gemcitabine/cisplatin in patients with locally advanced or metastatic urothelial cancer without prior systemic therapy: EORTC Intergroup Study 30987. J Clin Oncol. 2012 Apr 1;30(10):1107-13. Bellmunt J, Orsola A, Leow JJ, Wiegel T, De Santis M, Horwich A; ESMO Guidelines Working Group. Bladder cancer: ESMO Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2014 Sep;25 Suppl 3:iii40-8. Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee JL, Fong L, Vogelzang NJ, Climent MA, Petrylak DP, Choueiri TK, Necchi A, Gerritsen W, Gurney H, Quinn DI, Culine S, Sternberg CN, Mai Y, Poehlein CH, Perini RF, Bajorin DF; KEYNOTE-045 Investigators. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med. 2017 Mar 16;376(11):1015-1026. Carlo MI, Zhang L, Mandelker D, Vijai J, Cipolla CK, Robson ME, Funt S, Hakimi AA, Iyer G, Rosenberg JE, Coleman J, Solit DB, Offit K, Bajorin DF. Cancer predisposing germline mutations in patients (pts) with urothelial cancer (UC) of the renal pelvis (R-P), ureter (U) and bladder (B). J Clin Oncol 35, 2017 (suppl; abstr 4510). Challita-Eid PM, Satpayev D, Yang P, An Z, Morrison K, Shostak Y, Raitano A, Nadell R, Liu W, Lortie DR, Capo L, Verlinsky A, Leavitt M, Malik F, Aviña H, Guevara CI, Dinh N, Karki S, Anand BS, Pereira DS, Joseph IB, Doñate F, Morrison K, Stover DR. Enfortumab vedotin antibody-drug conjugate targeting nectin-4 is a highly potent therapeutic agent in multiple preclinical cancer models. Cancer Res. 2016 May 15;76(10):3003-13. Choudhury NJ, Campanile A, Antic T, Yap KL, Fitzpatrick CA, Wade JL 3rd, Karrison T, Stadler WM, Nakamura Y, O'Donnell PH. Afatinib activity in platinum-refractory metastatic urothelial carcinoma in patients with ERBB alterations. J Clin Oncol. 2016 Jun 20;34(18):2165-71. Costa R, Carneiro BA, Taxter T, Tavora FA, Kalyan A, Pai SA, Chae YK, Giles FJ. FGFR3-TACC3 fusion in solid tumors: mini review. Oncotarget. 2016 Aug 23;7(34):55924-55938. Nogova L, Sequist LV, Perez Garcia JM, Andre F, Delord JP, Hidalgo M, Schellens JH, Cassier PA, Camidge DR, Schuler M, Vaishampayan U, Burris H, Tian GG, Campone M, Wainberg ZA, Lim WT, LoRusso P, Shapiro GI, Parker K, Chen X, Choudhury S, Ringeisen F, Graus-Porta D, Porter D, Isaacs R, Buettner R, Wolf J. Evaluation of BGJ398, a fibroblast growth factor receptor 1-3 kinase inhibitor, in patients with advanced solid tumors harboring genetic alterations in fibroblast growth factor receptors: results of a global phase i, dose-escalation and dose-expansion study. J Clin Oncol. 2017 Jan 10;35(2):157-165. Faltas B, Goldenberg DM, Ocean AJ, Govindan SV, Wilhelm F, Sharkey RM, Hajdenberg J, Hodes G, Nanus DM, Tagawa ST. Sacituzumab govitecan, a novel antibody--drug conjugate, in patients with metastatic platinum-resistant urothelial carcinoma. Clin Genitourin Cancer. 2016 Feb;14(1):e75-9. Galsky MD, Iasonos A, Mironov S, Scattergood J, Boyle MG, Bajorin DF. Phase II trial of dose-dense doxorubicin plus gemcitabine followed by paclitaxel plus carboplatin in patients with advanced urothelial carcinoma and impaired renal function. Cancer. 2007 Feb 1;109(3):549-55. Glaser AP, Fantini D, Shilatifard A, Schaeffer EM, Meeks JJ. The evolving genomic landscape of urothelial carcinoma. Nat Rev Urol. 2017 Feb 7. [Epub ahead of print] Grossman HB, Natale RB, Tangen CM, Speights VO, Vogelzang NJ, Trump DL, deVere White RW, Sarosdy MF, Wood DP Jr, Raghavan D, Crawford ED. Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer. N Engl J Med. 2003 Aug 28;349(9):859-66. Gupta S, Gill D, Poole A, Agarwal N. Systemic immunotherapy for urothelial cancer: current trends and future directions. Cancers (Basel). 2017 Jan 27;9(2). pii: E15.

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Hahn NM, Powles T, Masard C, Arkenau H-T, Friedlander TW, Hoimes CJ, Lee J-Y, Ong M, Sridhar SS< Vogelzang NJ, Fishman NM, Zhang J, Srinivas S, Parikh J, Antal J, Jin X, Ben Y, Gupta AK, O’Donnell PH. Updated efficacy and tolerability of durvalumab in locally advanced or metastatic urothelial carcinoma (UC). J Clin Oncol 35, 2017 (suppl; abstr 4525). Hodi FS, Chesney J, Pavlick AC, Robert C, Grossmann KF, McDermott DF, Linette GP, Meyer N, Giguere JK, Agarwala SS, Shaheen M, Ernstoff MS, Minor DR, Salama AK, Taylor MH, Ott PA, Horak C, Gagnier P, Jiang J, Wolchok JD, Postow MA. Combined nivolumab and ipilimumab versus ipilimumab alone in patients with advanced melanoma: 2-year overall survival outcomes in a multicentre, randomised, controlled, phase 2 trial. Lancet Oncol. 2016 Nov;17(11):1558-1568. Hurwitz M, Spiess PE, Garcia JA, Pisters LL. Urothelial and kidney cancers. Cancer Network. Available at: http://www.cancernetwork.com/cancer-management/urothelial-and-kidney-cancers/page/0/1. Last accessed XXXX. Iyer G, Hanrahan AJ, Milowsky MI, Al-Ahmadie H, Scott SN, Janakiraman M, Pirun M, Sander C, Socci ND, Ostrovnaya I, Viale A, Heguy A, Peng L, Chan TA, Bochner B, Bajorin DF, Berger MF, Taylor BS, Solit DB. Genome sequencing identifies a basis for everolimus sensitivity. Science. 2012 Oct 12;338(6104):221. Iyer G, Al-Ahmadie H, Schultz N, Hanrahan AJ, Ostrovnaya I, Balar AV, Kim PH, Lin O, Weinhold N, Sander C, Zabor EC, Janakiraman M, Garcia-Grossman IR, Heguy A, Viale A, Bochner BH, Reuter VE, Bajorin DF, Milowsky MI, Taylor BS, Solit DB. Prevalence and co-occurrence of actionable genomic alterations in high-grade bladder cancer. J Clin Oncol. 2013 Sep 1;31(25):3133-40. Iyer G, Balar AJ, Milowsky MI, et al. Correlation of DNA damage response (DDR) gene alterations with response to neoadjuvant (neo) dose-dense gemcitabine and cisplatin (ddGC) in urothelial carcinoma (UC). J Clin Oncol 34, 2016 (suppl; abstr 5011). Javlor (vinflunine) SmPC. Available at: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR__Product_Information/human/000983/WC500039604.pdf. Last accessed XXXX. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, Ferrucci PF, Hill A, Wagstaff J, Carlino MS, Haanen JB, Maio M, Marquez-Rodas I, McArthur GA, Ascierto PA, Long GV, Callahan MK, Postow MA, Grossmann K, Sznol M, Dreno B, Bastholt L, Yang A, Rollin LM, Horak C, Hodi FS, Wolchok JD. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015 Jul 2;373(1):23-34. Lee JK, Havaleshko DM, Cho H, Weinstein JN, Kaldjian EP, Karpovich J, Grimshaw A, Theodorescu D. A strategy for predicting the chemosensitivity of human cancers and its application to drug discovery. Proc Natl Acad Sci U S A. 2007 Aug 7;104(32):13086-91. Lerner SP, Kim J, Kwiatkowski DJ, et al. Comprehensive characterization of 412 muscle invasive urothelial carcinomas: Final analysis of The Cancer Genome Atlas (TCGA) project. J Clin Oncol 34, 2016 (suppl 2S; abstr 405). Liu D, Plimack ER, Hoffman-Censits J, Garraway LA, Bellmunt J, Van Allen E, Rosenberg JE. Clinical Validation of chemotherapy response biomarker ERCC2 in muscle-invasive urothelial bladder carcinoma. JAMA Oncol. 2016 Aug 1;2(8):1094-6. Liu D, Abbosh P, Keliher D, Reardon B, Mouw KW, Taylor-Weiner A, Mullane SA, Han G, Teo MY, Kim J, Al-Akhmadie H, Iyer G, Dulaimi E, Chen D, Hoffman-Censits JH, Carter SL, Bellmunt J, Plimack ER, Rosenberg JE, Van Allen EM. Subclonal mutational heterogeneity and survival in cisplatin-resistant muscle-invasive bladder cancer. J Clin Oncol 35, 2017 (suppl; abstr 4512). Loehrer PJ Sr, Einhorn LH, Elson PJ, Crawford ED, Kuebler P, Tannock I, Raghavan D, Stuart-Harris R, Sarosdy MF, Lowe BA, et al. A randomized comparison of cisplatin alone or in combination with

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

methotrexate, vinblastine, and doxorubicin in patients with metastatic urothelial carcinoma: a cooperative group study. J Clin Oncol. 1992 Jul;10(7):1066-73. Milowsky MI, Nanus DM, Maluf FC, Mironov S, Shi W, Iasonos A, Riches J, Regazzi A, Bajorin DF. Final results of sequential doxorubicin plus gemcitabine and ifosfamide, paclitaxel, and cisplatin chemotherapy in patients with metastatic or locally advanced transitional cell carcinoma of the urothelium. J Clin Oncol. 2009 Sep 1;27(25):4062-7. Milowsky MI, Iyer G, Regazzi AM, Al-Ahmadie H, Gerst SR, Ostrovnaya I, Gellert LL, Kaplan R, GarciaGrossman IR, Pendse D, Balar AV, Flaherty AM, Trout A, Solit DB, Bajorin DF. Phase II study of everolimus in metastatic urothelial cancer. BJU Int. 2013 Aug;112(4):462-70. National Center for Biotechnology Information. Gene ID 2661 FGFR3 fibroblast growth factor receptor 3 [ Homo sapiens (human) ]. Available at: https://www.ncbi.nlm.nih.gov/gene/2261. Last accessed XXXX. Necchi A, Mariani L, Giannatempo P, Raggi D, Farè E, Nicolai N, Piva L, Biasoni D, Catanzaro M, Torelli T, Stagni S, Maffezzini M, Pizzocaro G, De Braud FG, Gianni AM, Salvioni R. Long-term efficacy and safety outcomes of modified (simplified) MVAC (methotrexate/vinblastine/doxorubicin/cisplatin) as frontline therapy for unresectable or metastatic urothelial cancer. Clin Genitourin Cancer. 2014 Jun;12(3):203-209. Niegisch G, Retz M, Thalgott M, Balabanov S, Honecker F, Ohlmann CH, Stöckle M, Bögemann M, Vom Dorp F, Gschwend J, Hartmann A, Ohmann C, Albers P. Second-Line Treatment of Advanced Urothelial Cancer with Paclitaxel and Everolimus in a German Phase II Trial (AUO Trial AB 35/09). Oncology. 2015;89(2):70-8. Ochoa AE, Choi W, Su X, Siefker-Radtke A, Czerniak B, Dinney C, McConkey DJ. Specific micro-RNA expression patterns distinguish the basal and luminal subtypes of muscle-invasive bladder cancer. Oncotarget. 2016 Dec 6;7(49):80164-80174. Oing C, Rink M, Oechsle K, Seidel C, von Amsberg G, Bokemeyer C. Second line chemotherapy for advanced and metastatic urothelial carcinoma: vinflunine and beyond-a comprehensive review of the current literature. J Urol. 2016 Feb;195(2):254-63. Opdivo (nivolumab) PI. Available at: http://packageinserts.bms.com/pi/pi_opdivo.pdf. Last accessed XXXX. Pal SK, Rosenberg JE, Keam B, et al. Efficacy of BGJ398, a fibroblast growth factor receptor (FGFR) 1-3 inhibitor, in patients (pts) with previously treated advanced/metastatic urothelial carcinoma (mUC) with FGFR3 alterations. J Clin Oncol 34, 2016 (suppl; abstr 4517). Petrylak DP, Heath EI, Sonpavde G, et al. Anti-tumor activity, safety and pharmacokinetics (PK) of AGS15E (ASG-15ME) in a phase I dose escalation trial in patients (Pts) with metastatic urothelial cancer (mUC). J Clin Oncol 34, 2016 (suppl; abstr 4532). Plimack ER, Dunbrack RL, Brennan TA, Andrake MD, Zhou Y, Serebriiskii IG, Slifker M, Alpaugh K, Dulaimi E, Palma N, Hoffman-Censits J, Bilusic M, Wong YN, Kutikov A, Viterbo R, Greenberg RE, Chen DY, Lallas CD, Trabulsi EJ, Yelensky R, McConkey DJ, Miller VA, Golemis EA, Ross EA. Defects in DNA repair genes predict response to neoadjuvant cisplatin-based chemotherapy in muscle-invasive bladder cancer. Eur Urol. 2015 Dec;68(6):959-67. Plimack ER, Bellmunt J, Gupta S, Berger R, Chow LQ, Juco J, Lunceford J, Saraf S, Perini RF, O'Donnell PH. Safety and activity of pembrolizumab in patients with locally advanced or metastatic urothelial cancer (KEYNOTE-012): a non-randomised, open-label, phase 1b study. Lancet Oncol. 2017 Feb;18(2):212-220. Powles T, Grivas P, Aragon-Ching JB, et al. A multicentre, international, randomised, open-label phase 3 trial of avelumab + best supportive care (BSC) vs BSC alone as maintenance therapy after

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

first-line platinum-based chemotherapy in patients with advanced urothelial cancer (JAVELIN bladder 100). Poster presented at ESMO 2016a. Abstract #842. Powles T, Duran I, Van der Heijden MS, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2018 391(10122): 748-757. Powles T, Galsky MD, Castellano D, et al. A phase 3 study of first-line durvalumab (MEDI4736) ± tremelimumab versus standard of care (SoC) chemotherapy (CT) in patients (pts) with unresectable Stage IV urothelial bladder cancer (UBC): DANUBE. J Clin Oncol 34, 2016b (suppl; abstr TPS4574). Powles T, Huddart RA, Elliott T, et al. A phase II/III, double-blind, randomized trial comparing maintenance lapatinib versus placebo after first line chemotherapy in HER1/2 positive metastatic bladder cancer patients. J Clin Oncol 33, 2015 (suppl; abstr 4505). Powles T, O'Donnell PH, Massard C, Arkenau HT, Friedlander TW, Hoimes CJ, Lee JL, Ong M, Sridhar SS, Vogelzang NJ, Fishman MN, Zhang J, Srinivas S, Parikh J, Antal J, Jin X, Gupta AK, Ben Y, Hahn NM. Efficacy and safety of durvalumab in locally advanced or metastatic urothelial carcinoma: updated results from a Phase 1/2 open-label study. JAMA Oncol. 2017 Sep 14;3(9):e172411. Raggi D, Miceli R, Sonpavde G, Giannatempo P, Mariani L, Galsky MD, Bellmunt J, Necchi A. Secondline single-agent versus doublet chemotherapy as salvage therapy for metastatic urothelial cancer: a systematic review and meta-analysis. Ann Oncol. 2016 Jan;27(1):49-61. Robinson AG, Wei X, Vera-Badillo FE, Mackillop WJ, Booth CM. Palliative chemotherapy for bladder cancer: treatment delivery and outcomes in the general population. Clin Genitourin Cancer. 2016 Dec 29. pii: S1558-7673(16)30371-8 Roche press release. FDA grants Roche’s cancer immunotherapy TECENTRIQ(atezolizumab) Priority Review in additional type of advanced bladder cancer. January 9 2017. Available at: http://www.roche.com/media/store/releases/med-cor-2017-01-09.htm. Last accessed XXXX. Rosenberg JE, Heath E, Perez R, et al. Interim analysis of a phase I dose escalation trial of ASG-22CE (ASG-22ME; enfortumab vedotin), an antibody drug conjugate (ADC), in patients (Pts) with metastatic urothelial cancer (mUC). Poster presented at ESMO 2016. Abstract #788 Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, Dawson N, O'Donnell PH, Balmanoukian A, Loriot Y, Srinivas S, Retz MM, Grivas P, Joseph RW, Galsky MD, Fleming MT, Petrylak DP, Perez-Gracia JL, Burris HA, Castellano D, Canil C, Bellmunt J, Bajorin D, Nickles D, Bourgon R, Frampton GM, Cui N, Mariathasan S, Abidoye O, Fine GD, Dreicer R. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet. 2016a May 7;387(10031):1909-20. Ross JS, Wang K, Khaira D, Ali SM, Fisher HA, Mian B, Nazeer T, Elvin JA, Palma N, Yelensky R, Lipson D, Miller VA, Stephens PJ, Subbiah V, Pal SK. Comprehensive genomic profiling of 295 cases of clinically advanced urothelial carcinoma of the urinary bladder reveals a high frequency of clinically relevant genomic alterations. Cancer. 2016 Mar 1;122(5):702-11. Siefker-Radtke AO, Necchi A, et al. First results from the primary analysis population of the phase 2 study of erdafitinib (ERDA; JNJ-42756493) in patients (pts) with metastatic or unresectable urothelial carcinoma (mUC) and FGFR alterations (FGFRalt). 2018 J Clin Oncol 36, (suppl; abstr 4503) Seiler R, Winters B, Douglas J, et al. Muscle-invasive bladder cancer: Molecular subtypes and response to neoadjuvant chemotherapy. J Clin Oncol 35, 2017 (suppl 6S; abstract 281). Seront E, Rottey S, Sautois B, Kerger J, D'Hondt LA, Verschaeve V, Canon JL, Dopchie C, Vandenbulcke JM, Whenham N, Goeminne JC, Clausse M, Verhoeven D, Glorieux P, Branders S, Dupont P, Schoonjans J, Feron O, Machiels JP. Phase II study of everolimus in patients with locally

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

advanced or metastatic transitional cell carcinoma of the urothelial tract: clinical activity, molecular response, and biomarkers. Ann Oncol. 2012 Oct;23(10):2663-70. Sharma P, Callahan MK, Calvo E, et al. Efficacy and safety of nivolumab plus ipilimumab in metastatic urothelial carcinoma: first results from the phase I/II CheckMate 032 study. Presented at: 2016 SITC Annual Meeting; November 9-13, 2016a; National Harbor, MD. Abstract #O3. Sharma P, Callahan MK, Bono P, Kim J, Spiliopoulou P, Calvo E, Pillai RN, Ott PA, de Braud F, Morse M, Le DT, Jaeger D, Chan E, Harbison C, Lin CS, Tschaika M, Azrilevich A, Rosenberg JE. Nivolumab monotherapy in recurrent metastatic urothelial carcinoma (CheckMate 032): a multicentre, openlabel, two-stage, multi-arm, phase 1/2 trial. Lancet Oncol. 2016b Nov;17(11):1590-1598. Sharma P, Retz M, Siefker-Radtke A, Baron A, Necchi A, Bedke J, Plimack ER, Vaena D, Grimm MO, Bracarda S, Arranz JÁ, Pal S, Ohyama C, Saci A, Qu X, Lambert A, Krishnan S, Azrilevich A, Galsky MD. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017 Mar;18(3):312-322. Smith SC, Baras AS, Lee JK, Theodorescu D. The COXEN principle: translating signatures of in vitro chemosensitivity into tools for clinical outcome prediction and drug discovery in cancer. Cancer Res. 2010 Mar 1;70(5):1753-8. Smith SC, Havaleshko DM, Moon K, Baras AS, Lee J, Bekiranov S, Burke DJ, Theodorescu D. Use of yeast chemigenomics and COXEN informatics in preclinical evaluation of anticancer agents. Neoplasia. 2011 Jan;13(1):72-80. Stepan LP, Trueblood ES, Hale K, Babcook J, Borges L, Sutherland CL. Expression of Trop2 cell surface glycoprotein in normal and tumor tissues: potential implications as a cancer therapeutic target. J Histochem Cytochem. 2011 Jul;59(7):701-10. Tabernero J, Bahleda R, Dienstmann R, Infante JR, Mita A, Italiano A, Calvo E, Moreno V, Adamo B, Gazzah A, Zhong B, Platero SJ, Smit JW, Stuyckens K, Chatterjee-Kishore M, Rodon J, Peddareddigari V, Luo FR, Soria JC. Phase I dose-escalation study of JNJ-42756493, an oral pan-fibroblast growth factor receptor inhibitor, in patients with advanced solid tumors. J Clin Oncol. 2015 Oct 20;33(30):3401-8. TECENTRIQ® (atezolizumab) PI. Available at: https://www.gene.com/download/pdf/tecentriq_prescribing.pdf. Last accessed XXXX. The Cancer Genome Atlas Research Network. comprehensive molecular characterization of urothelial bladder carcinoma. Nature. 2014 Mar 20; 507(7492): 315-22. Tecentriq Approval History. Available at: https://www.drugs.com/history/tecentriq.html. Last accessed XXXX. Teo MY, Bambury R, Zabor EC, Jordan EJ, Al-Ahmadie HA, Boyd M, Bouvier N, Mullane S, Cha EK, Roper N, Ostrovnaya I, Hyman DM, Bochner BH, Arcila ME, Solit DB, Berger MF, Bajorin DF, Bellmunt J1, Iyer G, Rosenberg JE. DNA damage response and repair gene alterations are associated with improved survival in patients with platinum-treated advanced urothelial carcinoma. Clin Cancer Res. 2017a Jan 30. pii. Teo MY, Seier K, Ostrovnaya I, Regazzi AM, Kani BE, Moran MM, Cipolla CK, Bluth MJ, Chaim J, AlAhmadie H, Solit DB, Funt S, Wolchok JD, Iyer G, Charen AS, Bajorin DF, Rosenberg JE, Callahan MK. DNA damage repair and response (DDR) gene alterations (alt) and response to PD1/PDL1 blockade in platinum-treated metastatic urothelial carcinoma (mUC). J Clin Oncol 35, 2017 (suppl; abstr 4509). Tomlinson DC, Baldo O, Hamden P, Knowles MA. FGFR3 protein expression and its relationship to mutation status and prognostic variables in bladder cancer. J Pathol. 2007;213(1):91-8.

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Touat M, Ileana E, Postel-Vinay S, André F, Soria JC. Targeting FGFR signaling in cancer. Clin Cancer Res. 2015 Jun 15;21(12):2684-94. von der Maase H, Hansen SW, Roberts JT, Dogliotti L, Oliver T, Moore MJ, Bodrogi I, Albers P, Knuth A, Lippert CM, Kerbrat P, Sanchez Rovira P, Wersall P, Cleall SP, Roychowdhury DF, Tomlin I, Visseren-Grul CM, Conte PF. Gemcitabine and cisplatin versus methotrexate, vinblastine, doxorubicin, and cisplatin in advanced or metastatic bladder cancer: results of a large, randomized, multinational, multicenter, phase III study. J Clin Oncol. 2000 Sep;18(17):3068-77. von der Maase H, Sengelov L, Roberts JT, Ricci S, Dogliotti L, Oliver T, Moore MJ, Zimmermann A, Arning M. Long-term survival results of a randomized trial comparing gemcitabine plus cisplatin, with methotrexate, vinblastine, doxorubicin, plus cisplatin in patients with bladder cancer. J Clin Oncol. 2005 Jul 20;23(21):4602-8. Wagle N, Grabiner BC, Van Allen EM, Hodis E, Jacobus S, Supko JG, Stewart M, Choueiri TK, Gandhi L, Cleary JM, Elfiky AA, Taplin ME, Stack EC, Signoretti S, Loda M, Shapiro GI, Sabatini DM, Lander ES, Gabriel SB, Kantoff PW, Garraway LA, Rosenberg JE. Activating mTOR mutations in a patient with an extraordinary response on a phase I trial of everolimus and pazopanib. Cancer Discov. 2014 May;4(5):546-53. doi: 10.1158/2159-8290.CD-13-0353. Epub 2014 Mar 13. Wilcox AN, Silverman DT, Friesen MC, Locke SJ, Russ DE, Hyun N, Colt JS, Figueroa JD, Rothman N, Moore LE, Koutros S. Smoking status, usual adult occupation, and risk of recurrent urothelial bladder carcinoma: data from The Cancer Genome Atlas (TCGA) Project. Cancer Causes Control. 2016 Dec;27(12):1429-1435. Williams SV, Hurst CD, Knowles MA. Oncogenic FGFR3 gene fusions in bladder cancer. Hum Mol Genet. 2013 Feb 15;22(4):795-803. Wülfing C, Machiels JP, Richel DJ, Grimm MO, Treiber U, De Groot MR, Beuzeboc P, Parikh R, Pétavy F, El-Hariry IA. A single-arm, multicenter, open-label phase 2 study of lapatinib as the second-line treatment of patients with locally advanced or metastatic transitional cell carcinoma. Cancer. 2009 Jul 1;115(13):2881-90.

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Suggested figures and table [Copyright may be required for use] Figure 1. Immunotherapeutic targets in urothelial malignancies [Gupta et al. 2017]

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Figure 2. Characterization of muscle-invasive bladder cancer [Robertson et al. 2017]

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Figure 3. Application and performance characteristics of the COXEN algorithm for prediction of drug sensitivity in the BLA-40 human urothelial cancer cell lines [Lee et al. 2007]

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Figure 4 FGFR pathway and dysregulation in cancer [Touat et al. 2015]

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Annals of Oncology Review Article – Second Draft

Table 1. Biomarkers of interest in clinical studies of approved agents

Page 27 of 27

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Annals of Oncology Review Article – Second Draft

Tablew 1. Biomarkers of interest in clinical studies of approved agents

Atezolizumab

Study

Biomarkers

Rosenberg et al.

• • • • • • •

PD-L1 status Gene expression Mutation detection Mutation load PD-L1 status Gene expression Mutation load

• • •

Elevated PD-L1 expression did not correlate with objective response rate Response higher in TCGA luminal cluster II subtype Mutation load significantly increased in responders

• • •

Objective responses occurred across all PD-L1 subgroups Responses more frequent in TCGA luminal II subtype Mutation load associated with overall survival



PD-L1 expression

• •

Responses in patients with PD-L1–positive and PD-L1–negative tumours at all prespecified PD-L1 expression-level thresholds Trends to higher response rate and longer survival with PD-L1–positive tumors

2016a

Balar 2017

Avelumab

Apolo 2017

Key outcome

Durvalumab

Powles 2017



PD-L1 expression



Durable response observed regardless of PD-L1 expression

Pembrolizumab

Plimack 2017



PD-L1 status



PD-L1 positivity seemed to be crucial for detecting responders

Bellmunt 2017



PD-L1 status



Benefit independent of PD-L1 expression

Balar 2017b



PD-L1 expression



Responses were observed across all categories of PD-L1 expression

Sharma 2016b



PD-L1 expression



PD-L1 expression did not correlate with objective responses

Sharma 2017

• •

PD-L1 expression Gene expression



Objective responses across PD-L1 subgroups compared favorably with the 10% historical objective response High interferon-γ gene expression and urothelial carcinoma molecular subtype were associated with response CXCL9, CXCL10, CD8, and 12-chemokine signature were highly enriched in responders

Nivolumab

• •

Page 1 of 2

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Annals of Oncology Review Article – Second Draft

Page 2 of 2

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Suggested figures [Copyright may be required for use] Figure 1. Immunotherapeutic targets in urothelial malignancies [Gupta et al. 2017]

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Figure 2. Characterization of muscle-invasive bladder cancer [Robertson et al. 2017]

Alteration landscape for 412 primary tumors. Top to bottom: synonymous and non-synonymous somatic mutation rates, with one ultra-mutated sample with a POLE signature. Mutational signature (MSig) cluster, APOBEC mutation load, and neoantigen load by quartile. Normalized activity of 4 mutational signatures. Combined tumor stage (T1,2 versus T3,4) and node status, papillary histology, gender, and squamous histology. Somatic mutations for significantly mutated genes (SMGs) with frequency R7%. Copy number alterations for selected genes and FGFR3 and PPARG gene fusions.

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Figure 3. Application and performance characteristics of the COXEN algorithm for prediction of drug sensitivity in the BLA-40 human urothelial cancer cell lines [Lee et al. 2007]

Step 1. Experimentally determine the drug's pattern of activity in cells of set 1. Step 2. Experimentally measure molecular characteristics of the cells in set 1. Step 3. Select a subset of those molecular characteristics that most accurately predicts the drug's activity in cell set 1 (“chemosensitivity signature” selection). Step 4. Experimentally measure the same molecular characteristics of the cells in set 2. Step 5. Among the molecular characteristics selected in step 3, identify a subset that shows a strong pattern of co-expression extrapolation between cell sets 1 and 2. Step 6. Use a multivariate algorithm to predict the drug's activity in set 2 cells on the basis of the drug's activity pattern in set 1 and the molecular characteristics of set 2 selected in step 5. The output of the multivariate analysis is a COXEN score.

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018

Figure 4 FGFR pathway and dysregulation in cancer [Touat et al. 2015]

Downloaded from https://academic.oup.com/annonc/advance-article-abstract/doi/10.1093/annonc/mdy254/5055049 by guest on 19 July 2018