OVERVIEW
Bladder cancer is a complex and aggressive disease for which treatment strategies have had limited success. Improvements in detection, treatment, and outcomes in bladder cancer will require the integration of multiple new approaches, including genomic profiling, immunotherapeutics, and large randomized clinical trials. New and promising strategies are being tested in all disease states, including nonmuscle-invasive bladder cancer (NMIBC), muscle-invasive bladder cancer (MIBC), and metastatic urothelial carcinoma (UC). Efforts are underway to develop better noninvasive urine biomarkers for use in primary or secondary detection of NMIBC, exploiting our genomic knowledge of mutations in genes such as RAS, FGFR3, PIK3CA, and TP53 and methylation pathways alone or in combination. Recent data from a large, randomized phase III trial of adjuvant cisplatin-based chemotherapy add to our knowledge of the value of perioperative chemotherapy in patients with MIBC. Finally, bladder cancer is one of a growing list of tumor types that respond to immune checkpoint inhibition, opening the potential for new therapeutic strategies for treatment of this complex and aggressive disease.
Cancer is a genetic disease.1 A cancer cell inherits or acquires mutations that enable it to grow efficiently, replicate indefinitely, support angiogenesis, avoid apoptosis, and in some cases, metastasize.2 Molecular profiles obtained by host and tumor DNA sequencing, single nucleotide polymorphism, RNA, and protein microarrays, and methylation screens are helping to pinpoint which mutations drive the cancerous phenotype and which are merely passengers on the malignant journey. Notwithstanding the role of individual genes, aggregate molecular profiles provide patient- and tumor-specific information that details the biologic complexity of a particular cancer and can be exploited for its clinical implications, therapeutic insights, and diagnostic benefit.
DETECTION AND MONITORING OF BLADDER CANCER IN THE GENOMIC ERA
Although the treatment for UC has improved over the last several decades, diagnostic techniques have progressed more slowly. Cystoscopy is still considered the best method for diagnosing UC, but it is invasive, uncomfortable, and can only detect approximately 90% of lesions.3 In addition, when a tumor is discovered and must be biopsied and/or removed, a second procedure is required, transurethral resection of the bladder tumor (TURBT), which requires general anesthesia. Last, the cost of cystoscopy, especially when used to monitor recurrence, is the major reason why per-patient expenses for UC are among the highest for all cancers.4 The major problem associated with NMIBC is that after initial TURBT, 50% to 70% of patients develop multiple recurrences; 10% to 20% of these will progress to MIBC.5 This risk of recurrence and progression calls for life-long surveillance. The current standard procedure is to perform cystoscopy and evaluate urine cytology every 3 to 4 months in the first 2 years, twice per year in years 3 to 4, and yearly thereafter.5
The burden of this follow-up on the patient, as well as the direct and indirect costs for the patient and society in terms of lost wages, have led to extensive efforts to develop noninvasive urine biomarkers for UC. However, to date, none have demonstrated sufficient specificity and sensitivity to monitor the general population or replace cystoscopy and cytology in monitoring for recurrence.6 Urine cytology is particularly insensitive for detecting low-grade tumors. However, advances in genomics have clearly demonstrated that DNA alterations offer great promise for detecting primary or secondary bladder cancer.
NMIBC and MIBC are genetically different.7–10 NMIBC is characterized by a high frequency of mutations in the FGFR3 oncogene, leading to constitutive activation of the RAS/ MAPK pathway. In MIBC, mutations in the TP53 gene prevail. In general, mutations in FGFR3 and TP53 are mutually exclusive, suggesting that NMIBC and MIBC develop along different oncogenetic pathways. However, these mutations often occur simultaneously in stage pT1 tumors that invade the connective tissue layer underlying the urothelium. Recently, somatic mutations in the PIK3CA oncogene, which encodes the catalytic subunit p110α of class-IA PI3 kinase, were described in 13% to 27% of bladder tumors.11 These mutations often coincided with FGFR3 mutations. Mutations in the RAS oncogenes (HRAS, KRAS, and NRAS) have also been found in 13% of bladder tumors and in all stages and grades; they are mutually exclusive with FGFR3 mutations. Given these findings, analyzing urine sediment for genetic mutations may be a promising strategy for noninvasive detection of bladder cancer.
FGFR3
FGFR3 mutations occur in around 50% of both lower and upper urinary tract tumors, clustering in three distinct hotspots in exons 7, 10, and 15.12 The most common mutations in exon 7 and 10 favor ligand-independent dimerization, transactivation, and signaling.13–17 Mutations in exon 15 are rare and induce a conformational change in the kinase domain, resulting in ligand-independent receptor activation and signaling, as well as FGFR3 cellular localization, with aberrant endoplasmic reticulum signaling.18 FGFR3 mutations are thought to occur early during urothelial transformation, as they are reported in over 80% of preneoplastic lesions,13,14 pointing to an overall “benign” effect of FGFR3 mutation in the bladder.17,19 FGFR3-mutant tumors are more chromosomally stable than their wild-type counterparts. A mutually exclusive relationship between FGFR3 mutation and overrepresentation of 8q was observed in NMIBC.20 A recent study found that around 80% of NMIBC and 54% of MIBC have dysregulated FGFR3 with discordant mutation and protein expression patterns, suggesting a key role for FGFR3 in both NMIBC and MIBC, either through mutation, overexpression, or both.21 These discrepancies may reflect differential downstream signaling of wild-type and mutant receptors or the different molecular pathways instigating the development of these tumors. The mechanisms driving FGFR3 overexpression in UC are largely unknown, although a recent study demonstrated the regulation of FGFR3 expression in urothelial cells by two microRNAs (miR-99a/100) that are often downregulated in UC, particularly in low-grade and low-stage tumors.22
FGFR3 mutations were among the first to be used as urine biomarkers of recurrent disease, especially low-grade disease, which is challenging to detect by urine cytology. van Rhijn et al23 reported that combined microsatellite and FGFR3 mutation analysis could detect UC in voided urine. FGFR3 mutations were found in 44% of urothelial tumors (59 tumors), but were absent in 15 G3 tumors. The sensitivity of microsatellites to detect cancer in voided urine was lower for tumors harboring FGFR3 mutations (15 out of 21 tumors; 71%) than for FGFR3 wild-type UC (29 out of 32 tumors; 91%). By including the FGFR3 mutation, the sensitivity of molecular cytology increased from71%to89% and was superior to the sensitivity of morphologic cytology (25%) for every clinical subdivision. These findings highlighted the potential of molecular biology as an adjunct to cystoscopy and cytology in informing follow-up care.
HRAS
The HRAS gene, which codes for p21 Ras (or Ras), a small GT-Pase, was the first identified human oncogene. It was found in the T24/EJ urothelial cell line.24–26 In the normal urothelium, normal Ras protein diminishes with differentiation, with highest expression in the basal (progenitor) cells.27 The role of Ras in UC is supported by its ability to transform Simian vacuolating virus 40 (SV40)-immortalized human urothelial cells into invasive transitional-cell carcinomas.28,29 In addition, in elegant transgenic studies, Ras overexpression has been shown to lead to NMIBC.30 Ras interacts with Raf, a serine/threonine kinase, which is activated in tumor cells containing enhanced growth signaling pathways in both NMIBC, MIBC, and metastatic disease with subsequent activation of MAPK.31,32
P53
The p53 tumor suppressor encoded by the TP53 gene located on chromosome 17p13.133 inhibits phase-specific cell cycle progression (G1-S) through transcriptional activation of p21WAF1/CIP1.34 Most UCs exhibit loss of a single 17p allele. Additional mutations in the remaining allele can inactivate TP53, leading to increased nuclear accumulation of the mutant protein, which has a longer half-life than its wild-type counterpart.35 TP53 deletion was correlated with grade and stage of UC.36–41 Invasive carcinoma can also progress from recurrent papillary carcinoma by acquiring additional alterations in TP53, RB1, PTEN, EGFRs, CCND1, MDM2, or E2F.42 In addition, oncogenic HRas has been shown to promote the malignant potency of UC cells that have acquired deficiencies of TP53, RB1, and PTEN.42 Mutations in the TP53 gene that result in a truncated protein (or no protein), homozygous deletion of both alleles of the gene, or gene silencing by methylation of the promoters of both alleles cannot be detected by nuclear accumulation of p53 protein,43,44 thus limiting the sensitivity of immunohistochemistry (IHC) for p53 alterations. Notwithstanding this caveat, overexpression of nuclear p53 protein by IHC has been used as a surrogate marker for detection of mutant p53 in clinical specimens. The expression of p53 has been associated with increased risk of progression of NMIBC or mortality in patients with MIBC, independent of tumor grade, stage, and lymph node status.33,34,45–56 Interestingly, in a recently reported randomized, prospective trial, this was not borne out in patients treated with cystectomy.57 In this and other studies, discordance in the identification of p53 as an independent prognostic marker for UC progression, recurrence, mortality, and response to therapy may be a result of patients’ genetic and epigenetic status, cohort selection, and technical and statistical variations.41,58–60
COMBINING GENOMIC ASSAYS
To develop more sensitive and specific assays, recent studies have simultaneously evaluated RAS, FGFR3, and PIK3CA in UC.61 A study of 257 patients with primary bladder tumors found that 64% (164 out of 257) of tumors contained an FGFR3 mutation, 11% (28) samples were mutant for one of the RAS genes, and 24% (61) harbored a PIK3CA mutation.61 Of the 257 primary tumors, 26% overexpressed p53, which is indicative of missense mutations, as noted above. When RAS, FGFR3, and PIK3CA mutations were calculated with TP53 mutations, only 27 tumors (11%) were wild-type for all examined genes. In 54 patients who developed one or more recurrences, tissue was available from 184 recurrent tumors, including multifocal recurrences. Using the SNaPshot-based mutation assay, investigators examined these tumors for FGFR3, PIK3CA, and RAS mutations. The frequency of p53 overexpression was low (6 out of 54) in the primary tumors of this group of patients, consisting mainly of NMIBC tumors. In patients with a wild-type primary tumor, recurrences were mostly wild-type (49 out of 54), whereas five harbored an FGFR3 mutation. One recurrent tumor contained two different PIK3CA mutations. In recurrences, PIK3CA mutations in addition to an FGFR3 mutation were associated with highergrade tumors compared with recurrences harboring an FGFR3 mutation alone. Importantly, there was 100% consistency in the type of mutation for RAS and PIK3CA among different tumors in the same patient.
Investigators also developed a methylation assay for specific detection of recurrent NMIBC in voided urine.62 Microsatellite analysis was also used to detect loss of heterozygosity in voided urine samples.23 Mutation analysis of FGFR3, PIK3CA, HRAS, KRAS, and NRAS was recently combined with methylation-specific assays to determine whether this combination outperformed either examination alone.63 Results were compared with those of urine cytology in a large, retrospective, longitudinal cohort that was part of the European FP7 UROMOL project. A total of 716 voided urine samples from 136 patients with NMIBC (Ta/T1, G½) were collected at TURBT. Patients with a history of carcinoma in situ were excluded from the analysis. Urine was collected at regular follow-up visits immediately before cystoscopy. During follow-up, 552 histologically proven recurrences were detected, including mainly stage Ta (92%), G½ (82%), and solitary tumors (67%). Sensitivity for detecting a recurrent tumor varied between 66% and 68% for the molecular tests after patient stratification based on tumor DNA analysis. A combination of markers increased sensitivity, but decreased the number of patients eligible for a certain test combination. Combining urine cytology with FGFR3 analysis without stratifying for FGFR3 status of the incident tumor increased sensitivity from 56% to 76%.
This study highlights the challenge of molecular examination of urine using genomics, and the importance of including all available information (i.e., cytology). However, there is no doubt that next-generation exome sequencing of paired tumor and peripheral blood samples will uncover many more potential biomarkers that could be added to these panels to improve their performance. Such examination was first performed in 2011 in a small set of patients.9 Initial findings from this cohort were examined in light of findings from an additional 88 patients with bladder cancer10 and by the The Cancer Genome Atlas (TCGA) consortium.64 From these contributions, several previously defined mutations were observed (in TP53, RB1, and HRAS), but novel mutations were also noted, the most common of which was in UTX, which was identified in21%of tested individuals. Of note, most of the identified new mutations were related to chromatin remodeling, suggesting a potential new area for bladder cancer research. Mutations in chromatin remodeling genes are commonly found in several other cancer types, suggesting their fundamental contribution to carcinogenesis. Adding to this complexity is a recent study of 537 patients with locally advanced or metastatic UC of the bladder, 74 patients with non-bladder, and 55 patients with nonurothelial bladder cancers profiled using mutation analysis, in situ hybridization, and IHC assays.65 Compared with nonbladder UC, bladder UC exhibited more frequent expression of abnormal protein (and increased amplification) in HER2, androgen receptor, serum protein acidic and rich in cysteine (SPARC), and topoisomerase 1. These findings suggest that bladder UC has higher levels of actionable biomarkers that may have clinical implications for treatment and diagnostic options.
NEOADJUVANT AND ADJUVANT CHEMOTHERAPY IN MUSCLE-INVASIVE BLADDER CANCER
Approximately 25% of patients with bladder cancer present with a tumor invading the muscle layer of the bladder wall (T2 to T4).66 MIBC is associated with a high rate of recurrence and poor overall prognosis, despite aggressive local and systemic therapies. Radical cystectomy is the standard treatment for MIBC, but even with substantial improvements in surgical techniques, mortality remains high because of a high rate of systemic failure. The 5-year mortality rate for patients with MIBC is about 50% to 70%.67,68 MIBC behaves as a systemic disease, and therefore needs systemic therapy early in the disease process to eradicate micrometastases.69
Phase III clinical trials of neoadjuvant cisplatin-based chemotherapy have demonstrated a survival benefit in patients with MIBC,67,70,71 mainly by pathologic down-staging of muscle-invasive tumors (stage T2 to T4a) to nonmuscle-invasive tumors (< T2).67,72–74 In the randomized Southwest Oncology group 8710 trial, neoadjuvant methotrexate, vinblastine, doxorubicin, and cisplatin (MVAC) followed by cystectomy demonstrated a 77-month median survival compared with a 46-month median survival with cystectomy alone. In this study, the likelihood of long-term survival was over 85% at 5 years for patients who achieved pathologic down-staging to pT0 either by TURBT alone before cystectomy or by the addition of neoadjuvant MVAC chemotherapy. However, long-term survival was less than 40% at 5 years in patients with residual muscle-invasive tumors (> pT2) at the time of cystectomy in both treatment arms.67 Patients who do not respond to neoadjuvant chemotherapy have a poor prognosis. However, no data demonstrate a survival benefit for additional chemotherapy in the adjuvant setting after three to four cycles of neoadjuvant cisplatin-based chemotherapy. Large clinical trials are currently in development in this setting.
Data for adjuvant chemotherapy57,75–77 are less compelling than for neoadjuvant chemotherapy. However, some patients benefit from adjuvant chemotherapy, including those who received up-front radical cystectomy and have extensive tumor invasion of the bladder wall or lymph node involvement. European Organisation for Research and Treatment of Cancer (EORTC) 30994 was a phase III trial of adjuvant versus delayed chemotherapy after cystectomy for patients with pT3 to T4 or node-positive disease.77 The study randomly assigned 298 patients to one of three adjuvant cisplatin-based chemotherapy regimens (MVAC, high-dose MVAC, or gemcitabine/cisplatin) or observation and chemotherapy at relapse. After a median follow-up of 7 years, 66 out of 141 patients (47%) in the adjuvant chemotherapy arm had died compared with 82 out of 143 (57%) in the observation arm. No significant improvement in overall survival was noted with adjuvant chemotherapy compared with observation (adjusted hazard ratio [HR] 0.78; 95% CI, 0.56 to 1.08; p = 0.13). However, adjuvant chemotherapy significantly prolonged progression-free survival compared with observation (HR 0.54; 95% CI, 0.4 to 0.73, p < 0.0001), with a 5-year progression-free survival rate of 47.6% (95% CI, 38.8 to 55.9) in the adjuvant chemotherapy arm and 31.8% (95% CI, 24.2 to 39.6) in the observation arm. Although this study did not meet its accrual goal of644 patients and was terminated early, it is the largest randomized adjuvant trial to date. Although the study was limited in power to show a significant improvement in overall survival with adjuvant chemotherapy, it is possible that some subgroups of patients might benefit from adjuvant chemotherapy. Cisplatin-based neoadjuvant chemotherapy remains the standard of care in MIBC. Table 1 summarizes neoadjuvant and adjuvant clinic trials in MIBC.
TABLE 1.
Stadler (p53) | Cognetti | Paz-Ares | Sternberg | Grossman | MRC/EORTC | |
---|---|---|---|---|---|---|
Chemotherapy | Adjuvant MVAC × 3 | Adjuvant GC × 4 | Adjuvant PGC × 4 | Adjuvant ddMVAC/ GC/MVAC × 4 | Neoadjuvant MVAC | Neoadjuvant CMV |
Patients | T1 and T2 negative LN | T2G3, T3 to T4, N0–2 | T3 to T4, N0 to N2 | T3 to T4 and/or pT × N1 to N3 | T2 to T4aN0 | T2 to T4aN0 |
Design | ||||||
α error | 5% | 5% | 5% | 5% | 5% | 5% |
Power | 90% | 80% | 80% | 80% | 80% | 90% |
Endpoint | Recurrence | OS | OS | OS | OS | OS |
0.5 to 0.3 at 3 years (20%) | 50% to > 60% at 2 years (10%) | 50% to > 65% at 2 years (15%) | 35% to > 42% at 5 years (7%) | 35% to > 42% median OS (50%) | 50% to > 60% at 2 years (10%) | |
Hazard Ratio | 0.52 | 0.75 | 0.77 | 0.826 | ||
Planned Sample Size | 190 | 610 | 340 | 660 (originally 1,344) | 298 | 915 |
Results | ||||||
Patients randomized | 114 (499 tested and 272 +p53) | 192 | 142 | 284 | 307 | 976 |
Years to Accrue | 9 | 6 | 7 | 6 | 11 | 6 |
5-Year Recurrence (Observation vs Chemotherapy) | TTR, 0.20; p = 0.62; HR, 0.78 | DFS, 42.3% vs. 37.2%; p = 0.70; HR, 1.08; all, 40% | 3 years 44% vs. 73%; p < 0.0001; HR, 0.36; all, 54% | PFS, 31.8% vs. 47.6%; p = < 0.0001; HR, 0.54 | 5-year DFS, 32% vs. 39%; 10-year DFS, 20% vs. 27% p = 0.008; HR, 0.82 | |
5-Year OS (Observation vs. Chemotherapy) | 85% (both arms) | 53.7% vs. 43.4%; p = 0.24; HR, 1.29; all, 48.5% | 31% vs. 60%; p < 0.0009; HR, 0.44; all, 49% | 47.7% vs. 53.6%; p = 0.13; HR, 0.78; all, 38.6% | 43% vs. 57%; p = 0.06 | 5-year OS, 43% vs. 49%; 10-year OS, 30% vs. 36%; p = 0.037; HR, 0.84 |
Median Follow-up | 5.4 years | 35 months | 30 months | 7 years | 8.7 years | 8 years |
Abbreviations: CMV, cispiatin/methotrexate/vinbiastine; dd, dose-dense; DFS, disease-free survival; EORTC, European Organisation for Research and Treatment of Cancer; GC, gemcitabine/cisplatin; HR, hazard ratio; MVAC, methotrexate/vinblastine/doxorubicin/cisplatin; OS, overall survival; PFS, progression-free survival; PGC, paclitaxel/gemcitabine/cisplatin; TTR, time to progression.
IMMUNE CHECKPOINT INHIBITION IN SOLID TUMORS
Immune checkpoint inhibition for cancer treatment is an area of growing research. Immune checkpoint pathways regulate T-cell activation to escape antitumor immunity. Immune checkpoint molecules involved in this mechanism include CTLA-4, programmed cell death 1 (PD-1) and its ligands PD-L1 and PD-L2, T-cell immunoglobulin mucin-3, and lymphocyte activation gene-3.78 Ipilimumab, a monoclonal antibody targeting CTLA-4, a potent immune checkpoint molecule expressed on T cells, demonstrated a survival benefit in a phase III study of patients with metastatic melanoma.79 PD-1 is an immune inhibitory receptor expressed on several immune-cell subsets, particularly cytotoxic T cells.80 PD-1 interacts with PD-L1 (B7- H1, CD274), which is expressed on tumor cells and immune cells, including T cells.81,82 Recent studies have demonstrated that upregulation of PD-L1 is an important mechanism of immune escape in NMIBC.83–86 Overexpression of PD-L1 in UC correlates with high-grade disease and worse clinical outcome. Anti-PD-1 and anti-PD-L1 have an improved toxicity profile compared with historic data from anti-CTLA-4 clinical trials.87,88 In September 2014, the U.S. Food and Drug Administration (FDA) granted accelerated approval of pembrolizumab for the treatment of unresectable or metastatic melanoma, and in December 2014 granted accelerated approval to nivolumab for unresectable or metastatic melanoma refractory to standard therapy.
TREATMENT OF METASTATIC BLADDER CANCER
The treatment options for metastatic UC are very limited; however, progress has been made in treating metastatic transitional carcinoma of the urothelial tract with combination chemotherapy. The median survival of 15 to 18 months with either MVAC or gemcitabine/cisplatin is substantially better than the 6 to 9 months with single-agent chemotherapy. In fact, 5% of patients have a complete, sometimes durable, remission.
CLINICAL STUDIES OF PD-1/PD-L1 INHIBITORS IN UROTHELIAL CARCINOMA
Two clinical trials of checkpoint inhibitors have reported preliminary efficacy in advanced/refractory metastatic UC. Remarkable efficacy and safety was seen in a phase I expansion cohort of 67 patients with heavily pretreated metastatic bladder cancer. Patients received 15 mg/kg of MPDL3280A, a human monoclonal antibody to PD-L1 containing an engineered Fcdomain, later revised to a flat dose of 1,200 mg intravenously every 3 weeks.89 Response rates were reported by PD-L1 positivity status, defined as 5% or higher of tumor-infiltrating immune cells staining for PD-L1 by IHC.1 In this study, 27% of tumors were IHC 2- or 3-positive, as defined by expression of PD-L1 on tumor-infiltrating immune cells. The overall response rate for all patients by Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 was 26%, and was even more remarkable (43%) among patients with PD-L1+ tumor-infiltrating cells. Even among patients whose tumor infiltrating immune cells were PD-L1‒,theresponseratewas11%asmeasured by RECIST v1.1. The median time to first response was 42 days (range, 38 to 85 days). Based on these results, MPDL3280A received breakthrough designation by the FDA in June 2014.
A phase I trial of pembrolizumab/MK-3475, a PD-1 inhibitor, studied 33 patients with advanced UC expressing PD-L1 in at least 1% of tumor cells by IHC. Patients received 10 mg/kg of pembrolizumab every 2 weeks. A response was seen in 7 out of 29 (24%) evaluable patients, and 64% of patients experienced a decrease in target lesions.90 With a median follow-up of 11 months, six patients have ongoing responses (median duration 16 to 40 weeks; median not reached).
Multiple PD-1/PDL-1 agents are currently being tested alone or in combination in advanced/refractory UC. Many more trials are in development in earlier disease states, testing agents such as MPDL3280A (NCT02302807) in the first-line setting in cisplatin-ineligible patients with metastatic bladder cancer, nivolumab in the maintenance setting after first-line cisplatin-based chemotherapy, and pembrolizumab in patients with NMIBC (NCT02324582).
KEY POINTS.
Detection of mutations in genes such as RAS, FGFR3, PIK3CA, and TP53, and methylation pathways in urine sediment are promising noninvasive strategies for diagnosis of bladder cancer.
The EORTC randomized phase III trial did not show a substantial improvement in overall survival with adjuvant cisplatin-based chemotherapy. Although the trial was limited in power, there was a 22% decrease in the risk of death (p = 0.13 trend) and a 55% decrease in the risk of recurrence.
Cisplatin-based neoadjuvant chemotherapy remains the standard of care in muscle-invasive bladder cancer.
Immune checkpoint inhibitors have shown efficacy in pretreated patients with metastatic bladder cancer, offering new hope for potential therapeutic strategies in this disease.
Multiple clinical trials are ongoing and in development in all stages of urothelial carcinoma, testing checkpoint inhibitors alone or in combination with other active agents that enhance the immune system.
ACKNOWLEDGMENT
The authors thank Bonnie L. Casey for editorial assistance in the preparation of this manuscript.
Footnotes
Disclosures of Potential Conflicts of Interest
Relationships are considered self-held and compensated unless otherwise noted. Relationships marked “L” indicate leadership positions. Relationships marked “I” are those held by an immediate family member; those marked “B” are held by the author and an immediate family member. Institutional relationships are marked “Inst.” Relationships marked “U” are uncompensated.
Employment: Nicholas J. Vogelzang, US Oncology. Leadership Position: None. Stock or Other Ownership Interests: Dan Theodorescu, Aurora Oncology, Keygenomics, Myriad. Nicholas J. Vogelzang, Caris Life Sciences. Honoraria: Nicholas J. Vogelzang, Abbvie, Bavarian Nordic, DAVAOncology, Endocyte, Mannkind, UpToDate. Consulting or Advisory Role: Nicholas J. Vogelzang, Amgen, AVEO, BIND Biosciences, Janssen Biotech, Inc. Speakers’ Bureau: Nicholas J. Vogelzang, Bayer, Caris MPI, Dendreon, GlaxoSmithKline, Medivation, Millennium Takeda, Sanofi. Research Funding: Nicholas J. Vogelzang, Endocyte (Inst), Exelixis (Inst), GlaxoSmithKline (Inst), PAREXEL International (Inst), Progenics (Inst), US Oncology (Inst), Viamet Pharmaceuticals (Inst). Patents, Royalties, or Other Intellectual Property: None. Expert Testimony: None. Travel, Accommodations, Expenses: Nicholas J. Vogelzang, Bayer/Onyx, Celgene, Dendreon, Exelixis, Genentech/Roche, Novartis, Pfizer, US Oncology. Other Relationships: None.
References
- 1.Stratton MR, Campbell PJ, Futreal PA The cancer genome. Nature. 2009;458:719–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hanahan D, Weinberg RA The hallmarks of cancer. Cell. 2000;100: 57–70. [DOI] [PubMed] [Google Scholar]
- 3.Grossman HB, Messing E, Soloway M, et al. Detection of bladder cancer using a point-of-care proteomic assay. JAMA. 2005;293:810–816. [DOI] [PubMed] [Google Scholar]
- 4.Botteman MF, Pashos CL, Redaelli A, et al. The health economics of bladder cancer: a comprehensive review of the published literature. Pharmacoeconomics. 2003;21:1315–1330. [DOI] [PubMed] [Google Scholar]
- 5.Theodorescu D, Wittke S, Ross MM, et al. Discovery and validation of new protein biomarkers for urothelial cancer: a prospective analysis. Lancet Oncol. 2006;7:230–240. [DOI] [PubMed] [Google Scholar]
- 6.van Rhijn BW, van der Poel HG, van der Kwast TH Urine markers for bladder cancer surveillance: a systematic review. Eur Urol. 2005;47:736–748. [DOI] [PubMed] [Google Scholar]
- 7.Ehdaie B, Theodorescu D. Molecular markers in transitional cell carcinoma of the bladder: new insights into mechanisms and prognosis. Indian J Urol. 2008;24:61–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Theodorescu D. Molecular pathogenesis of urothelial bladder cancer. Histol Histopathol. 2003;18:259–274. [DOI] [PubMed] [Google Scholar]
- 9.Gui Y, Guo G, Huang Y, et al. Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat Genet. 2011;43:875–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Guo G, Sun X, Chen C, et al. Whole-genome and whole-exome sequencing of bladder cancer identifies frequent alterations in genes involved in sister chromatid cohesion and segregation. Nat Genet. 2013;45:1459–1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Korkolopoulou P, Levidou G, Trigka EA, et al. A comprehensive immunohistochemical and molecular approach to the PI3K/AKT/mTOR (phosphoinositide 3-kinase/v-akt murine thymoma viral oncogene/ mammalian target of rapamycin) pathway in bladder urothelial carcinoma. BJUInt. 2012;110:E1237–E1248. [DOI] [PubMed] [Google Scholar]
- 12.di Martino E, Tomlinson DC, Knowles MA A decade of FGF receptor research in bladder cancer: past, present, and future challenges. Adv Urol. 2012;2012;429213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bernard-Pierrot I, Brams A, Dunois-Lardé C, et al. Oncogenic properties of the mutated forms of fibroblast growth factor receptor 3b. Carcinogenesis. 2006;27:740–747. [DOI] [PubMed] [Google Scholar]
- 14.Chaffer CL, Dopheide B, Savagner P, et al. Aberrant fibroblast growth factor receptor signaling in bladder and other cancers. Differentiation. 2007;75:831–842. [DOI] [PubMed] [Google Scholar]
- 15.Hart KC, Robertson SC, Kanemitsu MY, et al. Transformation and Stat activation by derivatives of FGFR1, FGFR3, and FGFR4. Oncogene. 2000;19:3309–3320. [DOI] [PubMed] [Google Scholar]
- 16.Knowles MA Role of FGFR3 in urothelial cell carcinoma: biomarker and potential therapeutic target. World J Urol. 2007;25:581–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.van Rhijn BW, Montironi R, Zwarthoff EC, et al. Frequent FGFR3 mutations in urothelial papilloma. J Pathol. 2002;198:245–251. [DOI] [PubMed] [Google Scholar]
- 18.Lievens PM, Roncador A, Liboi E. K644E/M FGFR3 mutants activate Erk½ from the endoplasmic reticulum through FRS2 alpha and PLC gamma-independent pathways. J MolBiol. 2006;357:783–792. [DOI] [PubMed] [Google Scholar]
- 19.van Rhijn BW, van der Kwast TH, Vis AN, et al. FGFR3 and P53 characterize alternative genetic pathways in the pathogenesis of urothelial cell carcinoma. CancerRes. 2004;64:1911–1914. [DOI] [PubMed] [Google Scholar]
- 20.Hurst CD, Platt FM, Taylor CF, et al. Novel tumor subgroups of urothelial carcinoma of the bladder defined by integrated genomic analysis. Clin CancerRes. 2012;18:5865–5877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Tomlinson DC, Baldo O, Harnden P, et al. FGFR3 protein expression and its relationship to mutation status and prognostic variables in bladder cancer. J Pathol. 2007;213:91–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Catto JW, Miah S, Owen HC, et al. Distinct microRNA alterations characterize high- and low-grade bladder cancer. Cancer Res. 2009;69:8472–8481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.van Rhijn BW, Lurkin I, Chopin DK, et al. Combined microsatellite and FGFR3 mutation analysis enables a highly sensitive detection of urothelial cell carcinoma in voided urine. Clin Cancer Res. 2003;9:257–263. [PubMed] [Google Scholar]
- 24.Taparowsky E, Suard Y, Fasano O, et al. Activation of the T24 bladder carcinoma transforming gene is linked to a single amino acid change. Nature. 1982;300:762–765. [DOI] [PubMed] [Google Scholar]
- 25.Parada LF, Tabin CJ, Shih C, et al. Human EJ bladder carcinoma oncogene is homologue of Harvey sarcoma virus ras gene. Nature. 1982;297: 474–478. [DOI] [PubMed] [Google Scholar]
- 26.Kawano H, Komaba S, Yamasaki T, et al. New potential therapy for or thotopic bladder carcinoma by combining HVJ envelope with doxorubicin. Cancer Chemother Pharmacol. 2008;61:973–978. [DOI] [PubMed] [Google Scholar]
- 27.Oxford G, Theodorescu D. The role of Ras superfamily proteins in bladder cancer progression. J Urol. 2003;170:1987–1993. [DOI] [PubMed] [Google Scholar]
- 28.Christian BJ, Kao CH, Wu SQ, et al. EJ/ras neoplastic transformation of simian virus 40-immortalized human uroepithelial cells: a rare event. Cancer Res. 1990;50:4779–4786. [PubMed] [Google Scholar]
- 29.Pratt CI, Kao CH, Wu SQ, et al. Neoplastic progression by EJ/ras at different steps of transformation in vitro of human uroepithelial cells. Cancer Res. 1992;52:688–695. [PubMed] [Google Scholar]
- 30.Wu X, Pandolfi PP Mouse models for multistep tumorigenesis. Trends CellBiol. 2001;11:S2–9. [DOI] [PubMed] [Google Scholar]
- 31.Fondrevelle ME, Kantelip B, Reiter RE, et al. The expression of Twist has an impact on survival in human bladder cancer and is influenced by the smoking status. Urol Oncol. 2009;27:268–276. [DOI] [PubMed] [Google Scholar]
- 32.Wallerand H, Reiter RR, Ravaud A. Molecular targeting in the treatment of either advanced or metastatic bladder cancer or both according to the signalling pathways. Curr Opin Urol. 2008;18:524–532. [DOI] [PubMed] [Google Scholar]
- 33.Mitra AP, Datar RH, Cote RJ Molecular pathways in invasive bladder cancer: new insights into mechanisms, progression, and target identification. J Clin Oncol. 2006;24:5552–5564. [DOI] [PubMed] [Google Scholar]
- 34.Mitra AP, Birkhahn M, Cote RJ p53 and retinoblastoma pathways in bladder cancer. World J Urol. 2007;25:563–571. [DOI] [PubMed] [Google Scholar]
- 35.Iggo R, Gatter K, Bartek J, et al. Increased expression of mutant forms of p53 oncogene in primary lung cancer. Lancet. 1990;335:675–679. [DOI] [PubMed] [Google Scholar]
- 36.Fujimoto K, Yamada Y, Okajima E, et al. Frequent association of p53 gene mutation in invasive bladder cancer. Cancer Res. 1992;52:1393–1398. [PubMed] [Google Scholar]
- 37.Soini Y, Turpeenniemi-Hujanen T, Kamel D, et al. p53 immunohisto-chemistry in transitional cell carcinoma and dysplasia of the urinary bladder correlates with disease progression. Br J Cancer. 1993;68:1029–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Moch H, Sauter G, Moore D, et al. p53 and erbB-2 protein overexpression are associated with early invasion and metastasis in bladder cancer. Virchows Arch A Pathol Anat Histopathol. 1993;423:329–334. [DOI] [PubMed] [Google Scholar]
- 39.Matsuyama H, Pan Y, Mahdy EA, et al. p53 deletion as a genetic marker in urothelial tumor by fluorescence in situ hybridization. Cancer Res. 1994;54:6057–6060. [PubMed] [Google Scholar]
- 40.Yamamoto S, Masui T, Murai T, et al. Frequent mutations of the p53 gene and infrequent H- and K-ras mutations in urinary bladder carcinomas of NON/Shi mice treated with N-butyl-N-(4-hydroxybutyl) nitrosamine. Carcinogenesis. 1995;16:2363–2368. [DOI] [PubMed] [Google Scholar]
- 41.Wang HC, Choudhary S. Reactive oxygen species-mediated therapeutic control of bladder cancer. NatRev Urol. 2011;8:608–616. [DOI] [PubMed] [Google Scholar]
- 42.Choudhary S, Wang HC Proapoptotic ability of oncogenic H-Ras to facilitate apoptosis induced by histone deacetylase inhibitors in human cancer cells. Mol Cancer Ther. 2007;6:1099–1111. [DOI] [PubMed] [Google Scholar]
- 43.Chaffer CL, Thomas DM, Thompson EW, et al. PPARgamma-independent induction of growth arrest and apoptosis in prostate and bladder carcinoma. BMC Cancer. 2006;6:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Thomas CY, Theodorescu D. Molecular markers of prognosis and novel therapeutic strategies for urothelial cell carcinomas. World J Urol. 2006; 24:565–578. [DOI] [PubMed] [Google Scholar]
- 45.Esrig D, Spruck CH 3rd, Nichols PW, et al. p53 nuclear protein accumulation correlates with mutations in the p53 gene, tumor grade, and stage inbladder cancer. Am J Pathol. 1993;143:1389–1397. [PMC free article] [PubMed] [Google Scholar]
- 46.Esrig D, Elmajian D, Groshen S, et al. Accumulation of nuclear p53 and tumor progression in bladder cancer. N Engl J Med. 1994;331:1259–1264. [DOI] [PubMed] [Google Scholar]
- 47.Spruck CH 3rd, Ohneseit PF, Gonzalez-Zulueta M, et al. Two molecular pathways to transitional cell carcinoma of the bladder. Cancer Res. 1994; 54:784–788. [PubMed] [Google Scholar]
- 48.Stavropoulos NE, Filliadis I, Ioachim E, et al. CD44 standard form expression as a predictor of progression in high risk superficial bladder tumors. Int Urol Nephrol. 2001;33:479–483. [DOI] [PubMed] [Google Scholar]
- 49.Stavropoulos NE, Ioachim E, Charchanti A, et al. Tumor markers in stage P1 bladder cancer. AnticancerRes. 2001;21:1495–1498. [PubMed] [Google Scholar]
- 50.Ioachim E, Charchanti A, Stavropoulos NE, et al. Immunohistochemical expression of retinoblastoma gene product (Rb), p53 protein, MDM2, c-erbB-2, HLA-DRand proliferation indices in human urinary bladder carcinoma. Histol Histopathol. 2000;15:721–727. [DOI] [PubMed] [Google Scholar]
- 51.Lianes P, Orlow I, Zhang ZF, et al. Altered patterns of MDM2 and TP53 expression in human bladder cancer. J Natl Cancer Inst. 1994;86:1325–1330. [DOI] [PubMed] [Google Scholar]
- 52.Sarkis AS, Dalbagni G, Cordon-Cardo C, et al. Nuclear overexpression of p53 protein in transitional cell bladder carcinoma: a marker for disease progression. J Natl Cancer Inst. 1993;85:53–59. [DOI] [PubMed] [Google Scholar]
- 53.Sarkis AS, Bajorin DF, Reuter VE, et al. Prognostic value of p53 nuclear overexpression in patients with invasive bladder cancer treated with neoadjuvant MVAC. J Clin Oncol. 1995;13:1384–1390. [DOI] [PubMed] [Google Scholar]
- 54.Stavropoulos NE, Filiadis I, Ioachim E, et al. Prognostic significance of p53, bcl-2 and Ki-67 in high risk superficial bladder cancer. Anticancer Res. 2002;22:3759–3764. [PubMed] [Google Scholar]
- 55.Peyromaure M, Ravery V. Prognostic value of p53 overexpression in bladder tumors treated with Bacillus Calmette-Guerin. Expert Rev Anticancer Ther. 2002;2:667–670. [DOI] [PubMed] [Google Scholar]
- 56.Moonen P, Witjes JA Risk stratification of Ta, Tis, T1 cancer In Lerner SP, Schoenberg M, Sternberg C (eds). Textbook of Bladder Cancer. Oxon, UK: Taylor and Francis; 2006;281–286. [Google Scholar]
- 57.Stadler WM, Lerner SP, Groshen S, et al. Phase III study of molecularly targeted adjuvant therapy in locally advanced urothelial cancer of the bladder based on p53 status. J Clin Oncol. 2011;29:3443–3449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Malats N, Bustos A, Nascimento CM, et al. P53 as a prognostic marker for bladder cancer: a meta-analysis and review. Lancet Oncol. 2005;6: 678–686. [DOI] [PubMed] [Google Scholar]
- 59.Ecke TH, Sachs MD, Lenk SV, et al. TP53 gene mutations as an independent marker for urinary bladder cancer progression. Int J Mol Med. 2008;21:655–661. [PubMed] [Google Scholar]
- 60.Hutterer GC, Karakiewicz PI, Zippe C, et al. Urinary cytology and nuclear matrix protein 22 in the detection of bladder cancer recurrence other than transitional cell carcinoma. BJU Int. 2008;101:561–565. [DOI] [PubMed] [Google Scholar]
- 61.Kompier LC, Lurkin I, van der Aa MN, et al. FGFR3, HRAS, KRAS, NRAS and PIK3CA mutations in bladder cancer and their potential as biomarkers for surveillance and therapy. PLoS One. 2010;5:e13821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Beukers W, Hercegovac A, Vermeij M, et al. Hypermethylation of the polycomb group target gene PCDH7 in bladder tumors from patients of all ages. JUrol. 2013;190:311–316. [DOI] [PubMed] [Google Scholar]
- 63.Zuiverloon TC, Beukers W, van der Keur KA, et al. Combinations of urinary biomarkers for surveillance ofpatients with incident nonmuscle invasive bladder cancer: the European FP7 UROMOL project. J Urol. 2013;189:1945–1951. [DOI] [PubMed] [Google Scholar]
- 64.Cancer Genome Atlas Research Network. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature. 2014;507:315–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Millis SZ, Bryant D, Basu G, et al. Molecular profiling of infiltrating urothelial carcinoma of bladder and nonbladder origin. Clin Genitourin Cancer. 2015;13:e37–49. [DOI] [PubMed] [Google Scholar]
- 66.Raghavan D. Chemotherapy and cystectomy for invasive transitional cell carcinoma of bladder. UrolOncol. 2003;21:468–474. [DOI] [PubMed] [Google Scholar]
- 67.Grossman HB, Natale RB, Tangen CM, et al. Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer. N Engl J Med. 2003;349:859–866. [DOI] [PubMed] [Google Scholar]
- 68.Stein JP, Lieskovsky G, Cote R, et al. Radical cystectomy in the treatment of invasive bladder cancer: long-term results in 1,054 patients. J Clin Oncol. 2001;19:666–675. [DOI] [PubMed] [Google Scholar]
- 69.Apolo AB, Grossman HB, Bajorin D, et al. Practical use of perioperative chemotherapy for muscle-invasive bladder cancer: summary ofsession at the Society of Urologic Oncology annual meeting. Urol Oncol. 2012; 30:772–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Neoadjuvant cisplatin, methotrexate, and vinblastine chemotherapy for muscle-invasive bladder cancer: a randomised controlled trial. International collaboration of trialists. Lancet. 1999;354:533–540. [PubMed] [Google Scholar]
- 71.International Collaboration of Trialists, Medical Research Council Advanced Bladder Cancer Working Party (now the National Cancer Research Institute Bladder Cancer Clinical Studies Group), European Organisation for Research and Treatment of Cancer Genito-Urinary Tract Cancer Group, et al. International phase III trial assessing neoadjuvant cisplatin, methotrexate, and vinblastine chemotherapy for muscle-invasive bladder cancer: long-term results of the BA06 30894 trial. J Clin Oncol. 2011;29:2171–2177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Splinter TA, Scher HI, Denis L, et al. The prognostic value of the pathological response to combination chemotherapy before cystectomy in patients with invasive bladder cancer. European Organization for Research on Treatment of Cancer-Genitourinary Group. J Urol. 1992;147: 606–608. [DOI] [PubMed] [Google Scholar]
- 73.Schultz PK, Herr HW, Zhang ZF, et al. Neoadjuvant chemotherapy for invasive bladder cancer: prognostic factors for survival of patients treated with M-VACwith 5-year follow-up. J Clin Oncol. 1994;12:1394–1401. [DOI] [PubMed] [Google Scholar]
- 74.Millikan R, Dinney C, Swanson D, et al. Integrated therapy for locally advanced bladder cancer: final report of a randomized trial of cystectomy plus adjuvant M-VAC versus cystectomy with both preoperative and postoperative M-VAC. J Clin Oncol. 2001;19:4005–4013. [DOI] [PubMed] [Google Scholar]
- 75.Cognetti F, Ruggeri EM, Felici A, et al. Adjuvant chemotherapy with cisplatin and gemcitabine versus chemotherapy at relapse in patients with muscle-invasive bladder cancer submitted to radical cystectomy: an Italian, multicenter, randomized phase III trial. Ann Oncol. 2012;23: 695–700. [DOI] [PubMed] [Google Scholar]
- 76.Paz-Ares L, Solsona E, Esteban E, et al. Randomized phase III trial comparing adjuvant paclitaxel/gemcitabine/cisplatin (PGC) to observation in patients with resected invasive bladder cancer: results of the Spanish Oncology Genitourinary Group (SOGUG) 99/01 study. J Clin Oncol. 2010;28 (suppl; abstr LBA4518). [Google Scholar]
- 77.Sternberg CN, Skoneczna I, Kerst JM, et al. Immediate versus deferred chemotherapy after radical cystectomy in patients with pT3-pT4 or N+M0 urothelial carcinoma of the bladder (EORTC 30994): an intergroup, open-label, randomised phase 3 trial. Lancet Oncol. 2015;16:76–86. [DOI] [PubMed] [Google Scholar]
- 78.Pardoll DM The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363: 711–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Keir ME, Butte MJ, Freeman GJ, et al. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Sharpe AH, Wherry EJ, Ahmed R, et al. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8:239–245. [DOI] [PubMed] [Google Scholar]
- 82.Francisco LM, Sage PT, Sharpe AH The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. 2010;236:219–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Nakanishi J, Wada Y, Matsumoto K, et al. Overexpression of B7-H1 (PD-L1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer Immunol Immunother. 2007;56:1173–1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Inman BA, Sebo TJ, Frigola X, et al. PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: associations with localized stage progression. Cancer. 2007;109:1499–1505. [DOI] [PubMed] [Google Scholar]
- 85.Wang Y, Zhuang Q, Zhou S, et al. Costimulatory molecule B7-H1 on the immune escape of bladder cancer and its clinical significance. J Hua-zhong Univ Sci Technolog Med Sci. 2009;29:77–79. [DOI] [PubMed] [Google Scholar]
- 86.Xylinas E, Robinson BD, Kluth LA, et al. Association of T-cell co-regulatory protein expression with clinical outcomes following radical cystectomy for urothelial carcinoma of the bladder. Eur J Surg Oncol. 2014;40:121–127. [DOI] [PubMed] [Google Scholar]
- 87.Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366: 2443–2454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012; 366:2455–2465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Powles T, Eder JP, Fine GD, et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature. 2014;515: 558–562. [DOI] [PubMed] [Google Scholar]
- 90.Plimack ER, Gupta S, Bellmunt J, et al. A phase 1b study of pembrolizumab (Pembro; MK-3475) in patients (Pts) with advanced urothelial tract cancer. Ann Oncol. 2014;25:1–41 (suppl; abstr LBA23). [Google Scholar]