Abstract
Urothelial carcinoma of the bladder comprises a spectrum of illnesses ranging from nonmuscle-invasive to muscle-invasive to advanced/metastatic disease. Each of these clinical states is characterized by a unique pathogenesis, prognosis and approach to treatment. However, given the heterogeneity of urothelial carcinoma, differences in biology and outcomes exist not only among these clinical states but also within each state. Personalized medicine, also commonly referred to as individualized or stratified medicine, offers the potential to optimize treatment for a given patient, based on the ability to accurately predict prognosis, response to treatment and tolerability of treatment. This review will discuss recent efforts, current challenges and future opportunities, for the personalized management of urothelial carcinoma.
Keywords: biomarker, bladder cancer, chemotherapy, neoadjuvant therapy, personalized medicine, urothelial carcinoma
Urothelial carcinoma (UC) is a chemosensitive neoplasm. The use of cisplatin-based combination chemotherapy has been shown to improve survival in both the perioperative and advanced disease settings. However, while the advances resulting from combination cisplatin-based chemotherapy in UC are clinically meaningful, the overall impact on this disease has been modest. Current therapies are particularly limited by extreme heterogeneity in patient outcomes and responses to treatment. Only subsets of patients derive benefit from particular systemic treatment regimens, and some patients experience severe treatment-related toxicities despite the lack of any appreciable benefit. The ability to individualize therapy by refining prognosis, and predicting treatment response, may allow more rational deployment of the current anti-cancer armamentarium and optimize patient outcomes. This review will describe the recent attempts to usher in the age of personalized cancer medicine in the care of patient with UC. While there are many opportunities to personalize management of noninvasive UC, given differences in pathogenesis and natural history, the current review will focus on invasive/advanced disease.
The pathogenesis of UC
Localized UCs has historically been referred to as either superficial or invasive, separating cancers which were confined to the either bladder mucosa or submucosa from those that invaded the bladder muscle. Currently, the term non-invasive is preferred instead of superficial in order to better capture the sometimes aggressive features of subsets of these tumors. Multiple molecular pathways have been implicated in the development of UC. However, two major pathways appear to play a role in the development of distinct clinical phenotypes: noninvasive tumors, which tend to recur but not invade the bladder wall, and tumors which invade the bladder wall and metastasize (Figure 1). These former, low-grade and noninvasive tumors, frequently demonstrate constitutive activation of the RAS-MAPK pathway, with activating mutations in HRAS and FGFR3 genes. By contrast, high-grade cancers, both nonmuscle-invasive and muscle-invasive, demonstrate frequent alterations in TP53 and RB genes and pathways [1]. Additional mutations, known to play a role in driving growth and progression of other solid tumors, have recently been identified in small subsets of invasive UC including KRAS and PIK3CA mutations [2]. In addition, loss of the tumor suppressor PTEN has been implicated in the pathogenesis of UC in mouse models [3]. Identification of these altered pathways is not only important in understanding the pathogenesis of UC, but also permits rational approaches to drug development, targeting the relevant pathways in given individuals.
Figure 1. Molecular pathways in the progression of urothelial carcinoma.
CIS: Carcinoma in situ; EGFR: EGF receptor; FGFR: FGF receptor; MUT: Mutation; WT: Wild-type.
Opportunities for personalized medicine in the management of muscle-invasive bladder cancer
Radical cystectomy with pelvic lymph node dissection is the standard approach for the management of patients with muscle-invasive bladder tumors. The probability of cure after surgery for muscle-invasive UC is associated with the pathologic stage, the ‘quality’ of surgery including margin status and extent of lymphadenectomy, and the use of perioperative chemotherapy. In a series of 1054 patients who underwent radical cystectomy at a single institution, the 5-year disease-free survival for patients with node-negative organ-confined disease was >80%. However, patients with lymph node involvement had a 5-year disease-free survival of only 35% [4].
Given the poor outcomes in patients with locally advanced disease, several studies have explored the integration of perioperative chemotherapy. Two large studies have established neoadjuvant cisplatin-based chemotherapy as a standard approach in muscle-invasive bladder cancer. The Southwest Oncology Group 8710 randomized patients with muscle-invasive bladder cancer to radical cystectomy alone (154 patients) compared with three cycles of methotrexate, vinblastine, doxorubicin and cisplatin MVAC followed by radical cystectomy (153 patients) [5]. At a median follow-up of 8.7 years, improvements in median survival (77 compared with 46 months; p = 0.06) and 5-year survival (57% compared with 43%; p = 0.06) favored the use of neoadjuvant arm. A Medical Research Council trial randomized 976 patients with muscle-invasive bladder cancer randomized patients to neoadjuvant cisplatin, methotrexate and vinblastine (CMV) (491 patients) versus local therapy alone (485 patients) [6]. An improvement in survival was observed for patients who received neoadjuvant chemotherapy (HR 0.84; 95% CI: 0.72–0.99; p = 0.037). Trials exploring adjuvant chemotherapy have been less compelling, but have generally been small, underpowered and utilized suboptimal chemotherapy regimens.
Because a large proportion of patients with muscle-invasive bladder cancer are cured with surgery alone (~50%), and only a subset benefit from the addition of neoadjuvant chemotherapy (approximately 5–10% absolute survival benefit), there is a crucial need for personalized approaches to identify patients who require integration of neoadjuvant therapy and to select patients likely to benefit from integration of particular drugs. A combination of routine clinical and pathologic variables may enhance the ability of individualize risk predictions, such as with the use of bladder cancer risk nomograms [7,101]. However, biomarkers offer the promise of capturing the heterogeneity in prognosis and outcomes not sufficiently captured with routine clinical and pathologic variables alone. Indeed, multiple studies have explored genes and gene panels in an attempt to improve prognostication in patients with clinically localized disease [8,9]. For example, studies have demonstrated that overexpression of four genes (JUN, MAP2K6, STAT3 and ICAM1), molecules associated with cell survival (Bcl-2, caspase-3, p53 and survivin) and insensitivity to antigrowth signals (p53, p21, p16 and pRB) are correlated with poor outcomes following cystectomy [10–12]. Unfortunately, these tools have not yet been extensively analytically or clinically validated, and as a result have not been integrated into routine clinical practice.
A retrospective analysis demonstrated that immunohistochemical detection of p53 (a surrogate for mutant p53 status given the longer half-life than the wild-type protein) significantly correlated with a higher likelihood of recurrence after cystectomy [13]. Additional data from a retrospective trial of adjuvant cisplatin-based chemotherapy suggested that the benefit of adjuvant chemotherapy was restricted to patients with altered p53 status [14]. These data led to the design of a prospective trial that enrolled patients with pT1-T2 N0 bladder cancer status post cystectomy who consented for p53 analysis of their tumor specimen [15]. Patients with 10% nuclear positivity by immunohistochemistry of p53 were randomized to three cycles of adjuvant MVAC, while patients with wild-type p53 were observed. This study was designed to detect a 20% absolute decrease in recurrence rate at 3 years, but closed early when the results of a preplanned interim analysis after randomization of 110 patients suggested that the probability of achieving a significant difference in the time to recurrence in the randomized population was highly unlikely. In addition, the prognostic significance of p53 testing was not validated in this trial, highlighting the importance of prospective validation of retrospective studies.
Personalized medicine in the management of advanced/metastatic UC
The MVAC regimen, developed in the 1980s, represented a significant advance in the management of patients with meta-static bladder cancer [16]. MVAC was subsequently compared with single-agent cisplatin and various multidrug regimens, and based on superiority in randomized trials, was adopted as a treatment standard. However, despite the improvements with MVAC, median overall survival of patients with metastatic bladder cancer remained approximately 12–13 months, and this treatment regimen was associated with multiple toxicities. In an effort to improve the efficacy and tolerability of treatment, a landmark Phase III trial randomized 405 patients with metastatic urothelial carcinoma to gemcitabine plus cisplatin (GC) versus MVAC [17]. This trial demonstrated that the median overall survival was similar on both arms (MVAC: 15.2 months vs GC: 14 months; HR: 1.04; 95% CI: 0.82–1.32; p = 0.75). However, the GC arm was associated with a much more favorable toxicity profile. While the GC regimen represented an advance in terms of tolerability, despite attempts to build on this regimen [18], there have been no improvements in efficacy of treatment for advanced UC since the MVAC regimen was introduced.
Given that multiple chemotherapeutic agents, and ‘targeted’ therapies, have anti-tumor activity only in a subset of patients with bladder cancer, biomarkers predictive of response to therapy represent a major goal for personalized cancer medicine. A limited number of prospective studies in advanced urothelial cancer, thus far, have assigned therapy based on the presence or absence of a particular biomarker.
The EGF family of receptors has been implicated in the pathogenesis of UC, forming the rationale for pursuing therapeutic strategies targeting this pathway in advanced disease [19–21]. A Phase II trial of chemonaive patients with metastatic bladder cancer explored the combination of gemcitabine, carboplatin, paclitaxel, plus the anti-human EGF receptor 2 (HER-2) monoclonal antibody trastu-zumab. For enrollment on this trial, patient’s tumors were required to have HER-2 overexpression by immunohistochemistry, HER2 gene amplification and/or elevated serum HER-2. Notably, the overall response rate with this regimen was 70%. The contribution of trastuzumab to these results is unclear, and this regimen has not been moved forward for more definitive testing.
A Phase II study explored lapatinib, a dual HER-2/EGFR receptor pathway inhibitor, as second-line therapy in patients with metastatic UC. Patients were eligible provided that their tumors had 1+, 2+ or 3+ expression of either EFGR or HER-2 by immunohistochemistry [22]. An objective response to treatment was observed in 1.7% (95% CI: 0.0–9.1%) of patients. However, 18 patients (31%; 95% CI: 19–44%) achieved stable disease. Clinical benefit with this regimen, defined as either an objective response or stable disease, was found to be correlated with EGFR overexpression and to some extent, HER-2 overexpression.
Another Phase II trial exploring lapatinib utilized a novel trial design focused on the putative predictive biomarker, rather than the specific tumor type [23]. Patients eligible for this study could have a wide variety of metastatic solid tumors (including bladder, endometrial, ovarian and gastric cancers) provided tumor tissue tested centrally demonstrated HER2 gene amplification by FISH. Unfortunately, this trial was closed early due to poor accrual. Of the 33 patients with metastatic bladder cancer tested, 12 patients had HER2 amplification by FISH, none of whom achieved an objective response to treatment [24]. These trials, which all utilized inhibitors of the same pathway, but different tests and ‘cutoffs’ for the putative predictive biomarkers, highlight many of the challenges of drug development in the era of targeted therapeutics.
Activating mutations in FGFR3 are present in a large proportion of noninvasive UC. However, recent studies have demonstrated that FGFR3 mutations are also present in 10–20% of muscle-invasive UC specimens, leading to intense interest in exploring FGFR3 inhibition as a therapeutic strategy in UC, in an effort to recreate the successes achieved with targeting oncogenic mutations in other solid tumors [2]. Dovitinib is a small molecule inhibitor of several tyrosine kinase receptors, including the VEGF receptor and FGFR3, which has demonstrated inhibition of tumor growth and proliferation in UC models selected for the presence of FGFR3 activating mutations or protein over-expression. A multicenter, two-stage, open-label Phase II trial is currently evaluating the safety and efficacy of dovitinib in patients with advanced UC who have progressed despite prior systemic therapy [25]. In this trial, 40 patients will be enrolled into two groups of 20, based on the presence or absence of mutation in the FGFR3 gene. The two groups will be recruited and analyzed independently and if at least two patients respond in either group, an additional 20 patients will be enrolled in that group. The results of this important trial, the first to enroll patients with advanced urothelial carcinoma based on the presence of an activating oncogenomic mutation, are anxiously awaited.
Future approaches to personalized medicine in UC
Because UC is a chemosensitive neoplasm, yet only a fraction of patients respond to a given chemotherapeutic regimen, there has been interest in developing tools to allow more rational use of the existing armamentarium of cytotoxics. One such approach has involved evaluating assessment of the levels of DNA-repair genes, or their protein products, in tumors, based on the notion that tumors with higher levels of DNA-repair genes may be more resistant to therapy. Excision repair cross complementing 1 (ERCC-1) is a critical regulator in nucleotide excision repair and its expression has been correlated with outcomes to cisplatin-based chemotherapy in various solid tumors [26–31]. RRM1, the regulatory subunit of ribonucleotide reductase, has been also implicated in carcinogenesis and tumor progression of non-small-cell lung cancer [32], and in addition predicted response to platinums and gemcitabine [33]. In a retrospective analysis, levels of the DNA-repair genes ERCC1, RRM1, BRCA1 and caveolin-1, were evaluated in tumor tissue from 57 patients with bladder cancer treated with cisplatin-based combination chemotherapy [34]. The median survival was significantly higher in patients with low ERCC1 levels (25.4 vs 15.4 months; p = 0.03). However, development of ERCC1 as a widely available predictive biomarker has been hampered by difficulties with assay reproducibility, particularly at the protein level [35].
Given the molecular complexity of solid tumors, reliance on a single gene or protein as a predictive biomarker for response to cytotoxic chemotherapy is unlikely to yield substantial improvements in patient selection. Alternatively, expression profiling of thousands of genes may better capture the heterogeneity of responses to therapy. Indeed, ‘signatures’ of response to platinum-based chemotherapy based on gene expression profiles have been generated and correlated with clinical outcomes in patients with bladder cancer [8,36,37]. This approach is promising, but the predictive signature is limited to the particular treatment regimen evaluated in the study in which the signature was generated. As a result, new signatures must be developed for each new treatment regimen entering clinical use, and these signatures cannot be used to aid in the development of novel drugs that have not yet been explored in human studies.
In an effort to overcome many of the limitations of this conventional approach to treatment-predictive gene expression profile development, a novel bioinformatic approach known as Coexpression Extrapolation or COXEN was developed [38]. COXEN utilizes publicly available data regarding gene-expression profiling and drug sensitivity from the NCI-60 panel of tumor cell lines as a ‘Rosetta Stone’ to predict chemosensitivity of gene-expression profiled bladder cancer samples using a computational algorithm. Perhaps most remarkably, the NCI-60 panel does not contain any bladder cancer cell lines, but the gene expression profiles generated across the cell lines included has still been able to predict responses when applied to human bladder cancer specimens in retrospective studies. The COXEN methodology has also been utilized to successfully predict responses to breast and ovarian cancers in retrospective studies. The COXEN approach can be utilized to predict responses to multiagent regimens, by combining data regarding single agents, and has been utilized successfully to identify novel agents with activity in UC. A study is currently being planned to prospectively evaluate the COXEN approach for selection of second-line chemotherapy for patients with metastatic UC.
Personalized cancer care via profiling of tumor oncogenomics is best exemplified by the emerging treatment approach to advanced non-small-cell lung cancer. Identification of activating mutations EGFR and ALK have changed the treatment paradigm, pairing specific therapies to the presence of these aberrations [39,40]. Several groups evaluated UC samples for particular somatic mutations and these results have been cataloged in various databases including the Catalog of Somatic Mutations in Cancer [102]. However, given the distinct pathways of pathogenesis of UC, and corresponding clinical phenotypes, knowledge of the genomic profiles of non-invasive versus invasive tumors is necessary to identify targets relevant for therapeutic strategies in these particular clinical states. As a result, recent efforts have sought to profile both noninvasive and invasive samples, and samples of varying grades, for a variety of known cancer-related mutations (Table 1). In the most comprehensive analysis published to date, Sjödahl et al. performed mutation analyses of 16 genes (FGFR3, PIK3CA, PIK3R1 PTEN, AKT1, KRAS, HRAS, NRAS, BRAF, ARAF, RAF1, TSC1, TSC2, APC, CTNNB1 and TP53) in 145 cases of UC [41]. This study identified that FGFR3 and PIK3CA mutations were positively associated, and were identified more commonly in noninvasive low-grade tumors. Furthermore, the potential importance of APC signaling was identified as 6% of the investigated tumors either demonstrated inactivating APC or activating CTNNB1 mutations. The mTOR regulatory tuberous sclerosis complex genes (TSC1 and TSC2) were found to be mutated at a combined frequency of 15%. This study highlights the feasibility of potential therapeutic target identification by profiling individual UC tumors for know cancer-related somatic mutations, and future efforts will focus on pairing these aberrations with appropriate therapeutic agents.
Table 1.
Most common somatic mutations identified in urothelial carcinoma.
| Somatic mutation | Frequency | Mechanism of action |
|---|---|---|
| FGFR3 | 40% | FGFR3 binds FGF, with downstream signaling leading to mitogenesis and differentiation |
| TP53 | 30% | Tumor suppressor gene; protein product can activate DNA repair, initiate apoptosis and cause cell growth arrest upon recognizing DNA damage |
| RB1 | 29% | Tumor suppressor gene; prevents the cell from replicating damaged DNA by preventing its progression along the cell cycle through G into S phase |
| PIK3CA | 20% | Encoding the P110a protein, PIK3CA is mutated in several human cancers and believed to be oncogenic |
| CDKN2A | 16% | Also known as p16 gene, it encodes the multiple tumor suppressor-1 protein, which functions as a stabilizer of p53 and sequester MDM-2 |
| RAS (HRAS, NRAS, KRAS) | 13% | As proto-oncogenes in the RAS family, even a single nucleotide change can lead to activating mutations in many tumor types. |
FGFR3: FGF receptor 3.
Data taken from [102].
Iyer et al. recently demonstrated the potential clinical relevance of oncogenic mutations in urothelial cancer [42]. In a study of everolimus as second-line therapy for patients with metastatic urothelial cancers, these investigators had previously demonstrated that a small number of patients achieved tumor regression. Notably, whole-genome sequencing of tumors from patients treated on this study revealed that the presence of TSC1 mutations correlated with response to treatment. This observation paves the way for future studies of whole-genome sequencing in an attempt to identify biomarkers of drug sensitivity in this disease.
The neoadjuvant setting as a paradigm for biomarker development
The traditional paradigm of oncologic drug development often involves first evaluating the efficacy of novel drugs in patients with advanced refractory disease. However, this may not be the optimal setting to detect a signal of drug activity and does not permit access to pre- and post-treatment tissue for biomarker development. Alternatively, the use of ‘window of opportunity’ trials, in which patients scheduled to undergo cystectomy receive a short course of treatment with a novel agent prior to surgery, allowing access to both pre- and post-treatment tissue, may be a more attractive setting for proof of concept and discovery studies. Furthermore, pathologic response rate to neoadjuvant therapy, an intermediate end point associated with improved outcomes in other solid tumors, may be utilized as a signal of activity in studies combining chemotherapy with a novel therapeutic.
Although neoadjuvant cytotoxic chemotherapy is standard for the treatment of muscle-invasive bladder cancer, treatment algorithms have been proposed to integrate ‘window of opportunity’ studies [43]. In one proposed approach, patients are stratified into low- and high-risk groups, distinguished by the presence of lymphovascular invasion, hydronephrosis, advanced clinical stage and variant histology. Patients with low-risk disease are offered enrollment in a single-agent neoadjuvant therapy. Pretreatment tissue (original diagnostic biopsy) and post-treatment tissue (radical cystectomy) are used for pharmacodynamic and molecular profiling. Patients with high-risk disease are offered enrollment in clinical trials that investigate the addition of a novel agent to conventional chemotherapy.
A few studies have recently been completed demonstrating the feasibility of this ‘window of opportunity’ approach. In one such study, 20 patients with muscle-invasive bladder cancer were treated with the EGF receptor inhibitor, erlotinib, 150 mg daily × 4 weeks followed by radical cystectomy [44]. In this trial, 25% achieved a pathologic complete response and 35% of patients were clinically downstaged. The Src inhibitor dasatinib has shown activity in Src-overexpressing human bladder cancer cell lines and the anti-tumor and biologic activity of dasatinib is currently being explored in a ‘window of opportunity’ neoadjuvant trial in patients with UC [45].
Conclusion
An improvement in our understanding of the underlying molecular pathogenesis has led to insights into the observed clinical heterogeneity of this disease. With the increased availability, and decreased cost, of high throughput genomic and proteomic technologies, such heterogeneity may be uncovered in much greater detail and ultimately linked to clinical outcomes. Such advances may not only facilitate more rational use of existing therapeutics, but also lead to the discovery of the next generation of treatments for this disease.
Expert commentary
Current therapies for advanced urothelial carcinoma are limited by heterogeneity in outcomes and short response durations. Improved understanding of disease pathogenesis has led to investigation of altered proteins and pathways, both as biomarkers and therapeutic targets. Despite early promise, outcomes with ‘targeted’ therapeutics in unselected patients with advanced urothelial carcinoma have been disappointing. Continued identification and validation of relevant molecular pathway aberrations, and utilizing these biomarkers to select patients for appropriate therapies, are critical to delivering on the promise of personalized medicine for patients with advanced urothelial carcinoma.
Five-year view
Urothelial carcinoma is heterogeneous in terms of molecular pathogenesis, prognosis and response to antineoplastic agents. Significant progress has been made over the past decade in defining the common molecular aberrations in this disease, particularly with regards to potentially druggable pathways. However, translating these findings into clinically useful predictive biomarkers requires rigorous analytic and clinical validation, which has not routinely occurred with biomarker development. The next 5 years will likely markedly expand our knowledge of the molecular aberrations in individual tumors, with technologies such as next-generation sequencing, and lead to the first examples of truly personalized treatment for advanced urothelial carcinoma.
Key issues.
Current therapies for advanced urothelial carcinoma are limited by extreme heterogeneity in patient outcomes and responses to treatment, thus necessitating the need for a more personalized approach to maximize clinical benefit.
Identification of altered genes and pathways in high-grade urothelial carcinoma (e.g., TP53 and RB genes) has provided further understanding of underlying pathogenesis, and also permits rational approaches to drug development, targeting the relevant pathways in given individuals.
Use of biomarkers offers the promise of capturing disease heterogeneity not sufficiently captured with routine clinical and pathologic variables alone.
A limited number of targets implicated in the pathogenesis of urothelial carcinoma, such as Her2-Neu and FGFR3, have already been explored in advanced urothelial carcinoma in molecularly defined patient populations.
The neoadjuvant setting offers a unique paradigm for drug development, with pre- and post-treatment tissue available, ‘window of activity’ for pharmacodynamic and molecular profiling.
Footnotes
Financial & competing interests disclosure
The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.
No writing assistance was utilized in the production of this manuscript.
References
- 1.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(35):5552–5564. doi: 10.1200/JCO.2006.08.2073. [DOI] [PubMed] [Google Scholar]
- 2.Kompier LC, Lurkin I, van der Aa MN, van Rhijn BW, van der Kwast TH, Zwarthoff EC. FGFR3, HRAS, KRAS, NRAS and PIK3CA mutations in bladder cancer and their potential as biomarkers for surveillance and therapy. PLoS ONE. 2010;5(11):e13821. doi: 10.1371/journal.pone.0013821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Puzio-Kuter AM, Castillo-Martin M, Kinkade CW, et al. Inactivation of p53 and Pten promotes invasive bladder cancer. Genes Dev. 2009;23(6):675–680. doi: 10.1101/gad.1772909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.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(3):666–675. doi: 10.1200/JCO.2001.19.3.666. [DOI] [PubMed] [Google Scholar]
- 5.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(9):859–866. doi: 10.1056/NEJMoa022148. [DOI] [PubMed] [Google Scholar]
- 6.Griffiths G, Hall R, Sylvester R, Raghavan D, Parmar MK 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; Australian Bladder Cancer Study Group; National Cancer Institute of Canada Clinical Trials Group; Finnbladder; Norwegian Bladder Cancer Study Group; Club Urologico Espanol de Tratamiento Oncologico Group. 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(16):2171–2177. doi: 10.1200/JCO.2010.32.3139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Bochner BH, Kattan MW, Vora KC International Bladder Cancer Nomogram Consortium. Postoperative nomogram predicting risk of recurrence after radical cystectomy for bladder cancer. J Clin Oncol. 2006;24(24):3967–3972. doi: 10.1200/JCO.2005.05.3884. [DOI] [PubMed] [Google Scholar]
- 8.Als AB, Dyrskjøt L, von der Maase H, et al. Emmprin and survivin predict response and survival following cisplatin-containing chemotherapy in patients with advanced bladder cancer. Clin Cancer Res. 2007;13(15 Pt 1):4407–4414. doi: 10.1158/1078-0432.CCR-07-0109. [DOI] [PubMed] [Google Scholar]
- 9.Sanchez-Carbayo M, Socci ND, Lozano J, Saint F, Cordon-Cardo C. Defining molecular profiles of poor outcome in patients with invasive bladder cancer using oligonucleotide microarrays. J Clin Oncol. 2006;24(5):778–789. doi: 10.1200/JCO.2005.03.2375. [DOI] [PubMed] [Google Scholar]
- 10.Mitra AP, Pagliarulo V, Yang D, et al. Generation of a concise gene panel for outcome prediction in urinary bladder cancer. J Clin Oncol. 2009;27(24):3929–3937. doi: 10.1200/JCO.2008.18.5744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Karam JA, Lotan Y, Karakiewicz PI, et al. Use of combined apoptosis biomarkers for prediction of bladder cancer recurrence and mortality after radical cystectomy. Lancet Oncol. 2007;8(2):128–136. doi: 10.1016/S1470-2045(07)70002-5. [DOI] [PubMed] [Google Scholar]
- 12.Shariat SF, Tokunaga H, Zhou J, et al. p53, p21, pRB, and p16 expression predict clinical outcome in cystectomy with bladder cancer. J Clin Oncol. 2004;22(6):1014–1024. doi: 10.1200/JCO.2004.03.118. [DOI] [PubMed] [Google Scholar]
- 13.Esrig D, Elmajian D, Groshen S, et al. Accumulation of nuclear p53 and tumor progression in bladder cancer. N Engl J Med. 1994;331(19):1259–1264. doi: 10.1056/NEJM199411103311903. [DOI] [PubMed] [Google Scholar]
- 14.Cote RJ, Esrig D, Groshen S, Jones PA, Skinner DG. p53 and treatment of bladder cancer. Nature. 1997;385(6612):123–125. doi: 10.1038/385123b0. [DOI] [PubMed] [Google Scholar]
- 15.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(25):3443–3449. doi: 10.1200/JCO.2010.34.4028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sternberg CN, Yagoda A, Scher HI, et al. Preliminary results of M-VAC (methotrexate, vinblastine, doxorubicin and cisplatin) for transitional cell carcinoma of the urothelium. J Urol. 1985;133(3):403–407. doi: 10.1016/s0022-5347(17)48996-8. [DOI] [PubMed] [Google Scholar]
- 17.von der Maase H, Hansen SW, Roberts JT, et al. Gemcitabine and cisplatin versus methotrexate, vinblastine, doxorubicin, and cisplatin in advanced or metastatic bladder cancer: results of a large, randomized, multinational, multicenter, Phase III study. J Clin Oncol. 2000;18(17):3068–3077. doi: 10.1200/JCO.2000.18.17.3068. [DOI] [PubMed] [Google Scholar]
- 18.Bellmunt J, Von der Maase H, Mead GM, et al. Randomized phase III study comparing paclitaxel/cisplatin/gemcitabine and gemcitabine/cisplatin in patients with locally advanced or metastatic urothelial cancer without prior systemic therapy; EORTC30987/Intergroup Study. Proc Am Soc Clin Oncol. 2007;25:Abstract 5030. doi: 10.1200/JCO.2011.38.6979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chow NH, Liu HS, Yang HB, Chan SH, Su IJ. Expression patterns of erbB receptor family in normal urothelium and transitional cell carcinoma. An immunohistochemical study. Virchows Arch. 1997;430(6):461–466. doi: 10.1007/s004280050056. [DOI] [PubMed] [Google Scholar]
- 20.Mellon JK, Cook S, Chambers P, Neal DE. Transforming growth factor alpha and epidermal growth factor levels in bladder cancer and their relationship to epidermal growth factor receptor. Br J Cancer. 1996;73(5):654–658. doi: 10.1038/bjc.1996.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Black PC, Dinney CP. Bladder cancer angiogenesis and metastasis–translation from murine model to clinical trial. Cancer Metastasis Rev. 2007;26(3–4):623–634. doi: 10.1007/s10555-007-9084-9. [DOI] [PubMed] [Google Scholar]
- 22.Wülfing C, Machiels JP, Richel DJ, et al. A single-arm, multicenter, open-label Phase 2 study of lapatinib as the second-line treatment of patients with locally advanced or metastatic transitional cell carcinoma. Cancer. 2009;115(13):2881–2890. doi: 10.1002/cncr.24337. [DOI] [PubMed] [Google Scholar]
- 23.Galsky MD, Zaks T, Hassani H, et al. Target-specific randomized discontinuation trial design: a novel approach in molecular therapeutics. Invest New Drugs. 2010;28(2):194–198. doi: 10.1007/s10637-009-9239-3. [DOI] [PubMed] [Google Scholar]
- 24.Galsky MD, Von Hoff DD, Neubauer M, et al. Target-specific, histology-independent, randomized discontinuation study of lapatinib in patients with HER2-amplified solid tumors. Proceedings of: ASCO Annual Meeting; Orlando, FL, USA. 2009; [DOI] [PubMed] [Google Scholar]
- 25.Milowsky GLC, Shi MM, Urbanowitz G, Zhang Y, Sternberg CN. A multicenter, open-label Phase II trial of dovitinib (TKI258) in advanced urothelial carcinoma patients with either mutated or wild-type FGFR3. Proceedings of. ASCO Annual Meeting; Chicago, IL, USA. 2011. [Google Scholar]
- 26.Murray D, Britten RA, Liu D, Tessier A, Hutchison MJ. ERCC1 expression as a molecular marker of cisplatin resistance in human cervical tumor cells. Int J Cancer. 2000;89:453–457. [PubMed] [Google Scholar]
- 27.Dabholkar M, Vionnet J, Bostick-Bruton F, Yu JJ, Reed E. Messenger RNA levels of XPAC and ERCC1 in ovarian cancer tissue correlate with response to platinum-based chemotherapy. J Clin Invest. 1994;94(2):703–708. doi: 10.1172/JCI117388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Rosell R, Lord RVN, Brabender J, et al. Low ERCC1 expression correlates with prolonged survival after cisplatin plus gemcitabine chemotherapy in non-small cell lung cancer. Clin Cancer Res. 2002;8:2286–2291. [PubMed] [Google Scholar]
- 29.Gorlick R, Metzger R, Danenberg KD, et al. Higher levels of thymidylate synthase gene expression are observed in pulmonary as compared with hepatic metastases of colorectal adenocarcinoma. J Clin Oncol. 1998;16(4):1465–1469. doi: 10.1200/JCO.1998.16.4.1465. [DOI] [PubMed] [Google Scholar]
- 30.Lenz HJ, Shirota Y, Stoehlmacher J, et al. ERCC1 and thymidylate synthase mRNA levels predict survival for colorectal cancer patients receiving combination oxaliplatin and fluorouracil chemotherapy. J Clin Oncol. 2001;19:4298–4304. doi: 10.1200/JCO.2001.19.23.4298. [DOI] [PubMed] [Google Scholar]
- 31.Soria JC, Olaussen KA, Dunant A, et al. DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. N Engl J Med. 2006;355:983–991. doi: 10.1056/NEJMoa060570. [DOI] [PubMed] [Google Scholar]
- 32.Zheng Z, Chen T, Li X, Haura E, Sharma A, Bepler G. DNA synthesis and repair genes RRM1 and ERCC1 in lung cancer. N Engl J Med. 2007;356(8):800–808. doi: 10.1056/NEJMoa065411. [DOI] [PubMed] [Google Scholar]
- 33.Reynolds C, Obasaju C, Schell MJ, et al. Randomized Phase III trial of gemcitabine-based chemotherapy with in situ RRM1 and ERCC1 protein levels for response prediction in non-small-cell lung cancer. J Clin Oncol. 2009;27(34):5808–5815. doi: 10.1200/JCO.2009.21.9766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bellmunt J, Paz-Ares L, Cuello M, et al. Spanish Oncology Genitourinary Group. Gene expression of ERCC1 as a novel prognostic marker in advanced bladder cancer patients receiving cisplatin-based chemotherapy. Ann Oncol. 2007;18(3):522–528. doi: 10.1093/annonc/mdl435. [DOI] [PubMed] [Google Scholar]
- 35.Dimou A, Agarwal S, Anagnostou V, et al. Standardization of epidermal growth factor receptor (EGFR) measurement by quantitative immunofluorescence and impact on antibody-based mutation detection in non-small cell lung cancer. Am J Pathol. 2011;179(2):580–589. doi: 10.1016/j.ajpath.2011.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Takata R, Katagiri T, Kanehira M, et al. Validation study of the prediction system for clinical response of M-VAC neoadjuvant chemotherapy. Cancer Sci. 2007;98(1):113–117. doi: 10.1111/j.1349-7006.2006.00366.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Takata R, Katagiri T, Kanehira M, et al. Predicting response to methotrexate, vinblastine, doxorubicin, and cisplatin neoadjuvant chemotherapy for bladder cancers through genome-wide gene expression profiling. Clin Cancer Res. 2005;11(7):2625–2636. doi: 10.1158/1078-0432.CCR-04-1988. [DOI] [PubMed] [Google Scholar]
- 38.Williams PD, Cheon S, Havaleshko DM, et al. Concordant gene expression signatures predict clinical outcomes of cancer patients undergoing systemic therapy. Cancer Res. 2009;69(21):8302–8309. doi: 10.1158/0008-5472.CAN-09-0798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Maemondo M, Inoue A, Kobayashi K, et al. North-East Japan Study Group. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362(25):2380–2388. doi: 10.1056/NEJMoa0909530. [DOI] [PubMed] [Google Scholar]
- 40.Rosell R, Moran T, Queralt C, et al. Spanish Lung Cancer Group. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med. 2009;361(10):958–967. doi: 10.1056/NEJMoa0904554. [DOI] [PubMed] [Google Scholar]
- 41.Sjödahl G, Lauss M, Gudjonsson S, et al. A systematic study of gene mutations in urothelial carcinoma; inactivating mutations in TSC2 and PIK3R1. PLoS ONE. 2011;6(4):e18583. doi: 10.1371/journal.pone.0018583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Iyer G, Hanrahan AJ, Milowsky MI, et al. Genome sequencing identifies a basis for everolimus sensitivity. Science. 2012;338(6104):221. doi: 10.1126/science.1226344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Shah JB, McConkey DJ, Dinney CP. New strategies in muscle-invasive bladder cancer: on the road to personalized medicine. Clin Cancer Res. 2011;17(9):2608–2612. doi: 10.1158/1078-0432.CCR-10-2770. [DOI] [PubMed] [Google Scholar]
- 44.Pruthi RS, Nielsen M, Heathcote S, et al. A Phase II trial of neoadjuvant erlotinib in patients with muscle-invasive bladder cancer undergoing radical cystectomy: clinical and pathological results. BJU Int. 2010;106(3):349–354. doi: 10.1111/j.1464-410X.2009.09101.x. [DOI] [PubMed] [Google Scholar]
- 45.Levitt JM, Yamashita H, Jian W, Lerner SP, Sonpavde G. Dasatinib is preclinically active against Src-overexpressing human transitional cell carcinoma of the urothelium with activated Src signaling. Mol Cancer Ther. 2010;9(5):1128–1135. doi: 10.1158/1535-7163.MCT-10-0096. [DOI] [PubMed] [Google Scholar]
Websites
- 101.Prediction tools. www.nomograms.org.
- 102.Wellcome Trust Sanger Institute. Catalogue of somatic mutations in cancer; prediction tools. www.sanger.ac.uk/genetics/CGP/cosmic.

