当前位置: 首页 > 期刊 > 《癌症医师》 > 2006年第6期 > 正文
编号:11121681
The Role of Perioperative Chemotherapy in the Treatment of Urothelial Cancer
http://www.100md.com 《中华首席医学网》
     a Department of Medicine, Division of Hematology/Oncology and b Department of Radiation Oncology, St. Luke’s-Roosevelt Hospital Center, New York, New York, USA; c Department of Medicine, Division of Hematology/Oncology and d Department of Urology, Beth Israel Medical Center, New York, New York, USA; e Continuum Cancer Centers of New York, New York, New York, USA

    Key Words. Urothelial cancer ; Transitional cell carcinoma ; Neoadjuvant ; Adjuvant ; Chemotherapy

    ABSTRACT

    Cancer of the urothelium is the fourth most common malignancy in men in the U.S. and the ninth most common in women. More than 63,000 Americans will be diagnosed with bladder cancer this year (47,010 men and 16,200 women), and more than 13,000 (8,970 men and 4,210 women) can expect to die of their disease. The approximate 5:1 ratio of incidence to mortality roughly parallels the frequency of superficial to invasive disease. Efforts to improve this ratio have generated a potential paradigm shift in the treatment of urothelial cancer, incorporating increasingly active chemotherapy into treatment regimens for high-risk tumors in both the pre-and postoperative settings. This review summarizes the evolution of chemotherapeutic treatment of urothelial cancer and the rationale for its perioperative administration and addresses the future directions of clinical research in this field.

    INTRODUCTION

    Cancer of the urothelium is the fourth most common malignancy in men in the U.S. and the ninth most common in women [1]. More than 63,000 Americans will be diagnosed with bladder cancer this year (47,010 men and 16,200 women), and more than 13,000 (8,970 men and 4,210 women) can expect to die of their disease [1]. The approximate 5:1 ratio of incidence to mortality roughly parallels the frequency of superficial to invasive disease [1]. Efforts to improve this ratio have generated a potential paradigm shift in the treatment of urothelial cancer, incorporating increasingly active chemotherapy into treatment regimens for high-risk tumors in both the pre- and postoperative settings. This review summarizes the evolution of chemotherapeutic treatment of urothelial cancer and the rationale for its perioperative administration and addresses the future directions of clinical research in this field.

    RATIONALE: POOR OUTCOMES WITH LOCAL TREATMENT ALONE

    Tumors that invade the deep muscle layer of the bladder are assigned stage T2, while T3 and T4 lesions invade the perivesical tissue and local structures, respectively (Tables 1 and 2). Standard treatment of muscle-invasive cancers has historically comprised surgical resection via radical cystectomy, achieving 5-year recurrence-free survival rates of 81%, 68%, 47%, and 44% for pathologic T2, T3a, T3b, and T4a tumors, respectively [2]. The 5-year overall survival rate is 78% for organ-confined, node-negative disease but falls to 45% when lymph nodes are involved, though an extended lymphadenectomy for node-positive disease may achieve a slight survival advantage [3]. The 5-year overall survival rate for extravesical, node-negative patients is 47% but likewise falls to 25% in node-positive disease.

    Results for localized radiotherapy (RT) of primary and/or unresectable disease were historically less favorable than those for surgery [4–7]. As a result, efforts were made to improve survival by adding RT before and after surgery. Initial retrospective analyses were encouraging of this strategy, so prospective randomized studies were organized [8]. The National Surgical Adjuvant Bladder Project reported a trial of 475 patients with T2–T4 tumors, randomizing participants to preoperative RT followed by cystectomy versus cystectomy alone [9]. Unfortunately, more than 50% of the enrolled patients were not included in the final analysis because of a failure to complete treatment or study ineligibility. In the remaining 234 patients, the 5-year survival rates were 43% versus 35% favoring the RT arm, but this was not statistically significant. The 8-year survival rates were 19% in both groups, and the pelvic failure rates were similar. In a multi-institutional intergroup phase III study, 140 patients with muscle-invasive bladder cancer, or rapidly recurring superficial high-grade tumors, were randomized to 20 Gy of pelvic radiation followed by cystectomy or cystectomy alone [10]. The 5-year overall survival rate was 43% (95% confidence interval [CI], 30%–56%) in the RT arm and 53% (95% CI, 41%–65%) in the surgery alone arm (p = .23). Although the study was underpowered, with wide confidence intervals, there was not even a trend toward a difference in outcome. A meta-analysis including these and other randomized studies failed to show a benefit for the use of RT prior to cystectomy, and this strategy has since been abandoned [11]. Unfortunately, no phase III randomized study has been performed evaluating RT in the postoperative setting, most likely related to the findings of a Radiation Therapy Oncology Group phase II study revealing a high rate of small bowel obstruction and fistula formation (37% vs. 8%) [12].

    Alternatives to RT have long been sought to improve the results of surgery without affecting the morbidity of the operation. Efforts incorporating chemotherapy into the treatment of high-risk urothelial cancers were undertaken, driven by the finding that cisplatin-based, combination chemotherapy was capable of producing complete responses in an increasing percentage of patients with advanced disease, relative to those seen in patients treated with single-agent regimens.

    EVOLUTION OF THE TREATMENT OF ADVANCED DISEASE

    Several chemotherapeutic agents with different mechanisms of action were proven active in urothelial cancer in the 1970s and 1980s. Methotrexate, vinblastine, doxorubicin, and cisplatin elicited objective responses in 29%, 18%, 17%, and 30% of patients, respectively, in a series of studies conducted at Memorial Hospital [13–16]. Doublet and triplet combinations induced higher response rates, but few complete responses. Ultimately, the two most potent doublets, cisplatin–doxorubicin and methotrexate–vinblastine, were combined, forming the regimen commonly referred to as M-VAC. In a pilot trial, M-VAC produced a 71% overall response rate, including a 50% complete clinical remission rate, and at least a doubling of the median survival time relative to single-agent cisplatin [17]. Toxicity was significant, however, with myelosuppression resulting in nadir sepsis in 4 of 24 patients, drug-related death in one patient, and varying degrees of anorexia, nausea, emesis, alopecia, and renal dysfunction.

    A series of randomized controlled studies proved that M-VAC was superior to single-agent cisplatin [18] and other cisplatin-based combinations [19] with respect to the overall response rate and complete response rate (13% vs. 3% and 35% vs. 25%, respectively) and overall survival. Unfortunately, only 3.7% of patients treated with M-VAC experienced long-term, disease-free survival, while up to one fourth of patients experienced nadir fever, >50% had at least grade 2 mucositis, 3%–4% experienced a toxic death, and many patients suffered ototoxicity and peripheral neuropathy [20]. One strategy devised to address the shortcomings of M-VAC involved increasing the dose intensity of both cisplatin and doxorubicin while adding a myeloid growth factor [21]. Although the toxicity (including leukopenia, nadir fever, and mucositis) was ameliorated, the complete response rate was improved, and the progression-free survival time was increased, no overall survival difference was noted.

    Recently, newer agents, including gemcitabine, paclitaxel, and docetaxel, were shown to be active in transitional cell carcinoma (TCC). Doublet combinations—including cisplatin and docetaxel [22], carboplatin and paclitaxel [23], and cisplatin and gemcitabine (GC) [24]—have been compared with M-VAC in the phase III setting. While M-VAC was proven to be superior to cisplatin and docetaxel and the randomized trial of M-VAC versus carboplatin and paclitaxel failed to reach accrual goals, GC compares favorably with M-VAC [22, 24–26]. No differences were seen with respect to the median or 5-year overall survival rates, but patients treated with M-VAC suffered more high-grade toxicities, including myelosuppression, nadir fever and sepsis, mucositis, and alopecia. Although this widely cited study was not designed as an equivalency trial, most medical oncologists have adopted the CG as a standard of care in advanced urothelial cancer. Finally, three- and four-drug combinations comprising both targeted and classic chemotherapeutic agents are being investigated [27–29].

    NEOADJUVANT CHEMOTHERAPY

    As chemotherapy in the metastatic setting evolved, prolonging survival and achieving higher complete response rates, it was incorporated into the multimodality treatment of localized but muscle-invasive disease. Neoadjuvant chemotherapy, shown to be effective in rectal cancer [30, 31] and breast cancer [32], was applied to TCC. Advantages of preoperative chemotherapy include better tolerability relative to postoperative treatment and the ability to assess the sensitivity of the primary lesion, shown to be important prognostically. In one trial of postoperative chemotherapy, 30% of patients were unable to complete even one of the three planned cycles of chemotherapy [33]. By administering chemotherapy prior to surgery, a decline in performance status resulting from operative morbidity is not an obstacle to treatment. In a retrospective analysis, Splinter et al. [34] found that 75% of patients who had a major pathologic response (pT2) were alive and disease free.

    Disadvantages of neoadjuvant treatment include the potential for over- or undertreatment based on imprecise clinical staging, or delays in or noncompliance with recommended cystectomy after chemotherapy. In a study by the Italian Bladder Tumor Study Group comparing neoadjuvant chemotherapy followed by cystectomy with cystectomy alone in clinical T2–4, N0, M0 TCC, the accuracy of clinical versus pathological staging in the control arm was only 42.3%, with 22.5% overstaged and 35.2% understaged [35]. While understaged patients could potentially go on to receive adjuvant treatment, overstaged patients may suffer needless drug-related toxicity.

    At least 14 randomized controlled trials have been performed evaluating a neoadjuvant strategy in muscle-invasive TCC, and two meta-analyses including these trials have been published in recent years (Table 3) [36, 37]. Although many studies suffer from insufficient power, limited follow-up time, early closure, and suboptimal chemotherapy, at least two large, well-designed trials have been completed and are incorporated into the most recent meta-analysis [36, 38–40]. A study involving cooperative groups from Europe, Scandinavia, Australia, and Canada, coordinated by the Medical Research Council (MRC) and the European Organization for the Research and Treatment of Cancer (EORTC), randomized 976 patients with high-grade clinical T2, or T3 or T4 TCC of the bladder, without biopsy-proven involved lymph nodes, felt to be reasonable candidates for potentially curative local therapy, to an institutionally chosen local treatment comprising cystectomy, external-beam radiation, or both, with or without preoperative cisplatin, methotrexate, and vinblastine (CMV) every 3 weeks for three cycles [40]. The study was powered to detect a 10% improvement (from 50% to 60%) in the primary outcome, overall survival. The calculated absolute 3-year overall survival difference was 5.5%, with a hazard ratio (HR) of 0.85, favoring the use of neoadjuvant chemotherapy. Although the p-value was .075 at the time of initial publication, an update with a median follow-up of 7 years revealed a statistically significant difference in survival (again 5.5%; p = .048). Further, there was an 18% reduction (p = .019) in the risk for locoregional relapse, metastases, or death (i.e., relapse-free survival). Although this trial is large and adequately powered, has sufficient follow-up, and shows a survival benefit, criticisms have been put forth; the lack of doxorubicin (omitted because of a concern for toxicity associated with radiation), high proportion of good-risk patients (34% had cT2 tumors), 20% rate of noncompletion of chemotherapy in patients assigned to the experimental arm, and control group crossover phenomenon may have underestimated the true effect of preoperative chemotherapy.

    Similarly, an American Intergroup study (INT 0080) was designed to test the efficacy of neoadjuvant chemotherapy in muscle-invasive bladder cancer [38]. Patients with T2–T4a, N0, M0 TCC who were to undergo radical cystectomy were randomized to surgery alone or three cycles of M-VAC prior to surgery. Patients were stratified by age (<65 years vs. 65 years) and stage (T2 vs. T3). The study was powered to detect a 50% difference in the primary end point of median overall survival. Accrual was slow, requiring 11 years to enroll 317 patients (or two patients per month from across the U.S.). With a median follow-up of 8.4 years, the median survival time was longer (77 months vs. 46 months) in the chemotherapy arm (p = .06; p = .05 in an unstratified analysis) and the 5-year overall survival rate was greater (57% vs. 43%; p = .06). At the time of surgery, more patients in the experimental arm were found to have no residual disease in the cystectomy specimen, relative to patients in the control arm who had undergone cystoscopic biopsies (38% vs. 15%; p < .001), which was found to be a positive prognostic factor in both groups. Fever, infection, mucositis, and treatment-related death were less frequent with the use of M-VAC in the neoadjuvant setting than in the treatment of advanced disease. Importantly, preoperative chemotherapy did not prevent patients from undergoing potentially curative surgery and did not increase the frequency or severity of operative morbidity. Criticisms have included the long accrual time, suggestive of selection bias, the use of a one-sided p-value (the two-sided p-value was calculated at .088 in one editorial [41]), and the use of medians read from the survival curves as opposed to HRs, which better estimate the whole curves and not just a point in time.

    In order to address the variability and methodological flaws in the literature on neoadjuvant chemotherapy in invasive TCC, Winquist et al. [36] compiled data from 11 of 16 identified randomized, controlled trials, representing 2,605 patients, and performed a meta-analysis of overall survival. The MRC/EORTC and the INT 0080 trials were both included. The pooled HR for death was 0.90 (95% CI, 0.82–0.99; p = .02), a 10% reduction in the risk for death. Focusing on the eight trials that incorporated cisplatin-based combination chemotherapy, the pooled HR was 0.87 (95% CI, 0.78–0.96; p = .006), consistent with an absolute survival benefit of 6.5% (50%–56.5%). An earlier meta-analysis did not include the results of INT 0080 but did confirm a 13% reduction in the risk for death (p = .016) and a 5% absolute survival benefit at 5 years for the use of preoperative cisplatin-based combination chemotherapy [37].

    In summary, on the basis of two randomized, controlled trials from the MRC/EORTC and an American Intergroup collaboration, as well as subsequent meta-analyses, the standard of care for clinically staged high-risk muscle-invasive bladder TCC is shifting from radical cystectomy alone to a multimodality regimen employing neoadjuvant cisplatin-based combination chemotherapy. Preoperative treatment is tolerable and does not exacerbate operative morbidity or lead to delays in resection. Unfortunately, far too few patients who would otherwise qualify for neoadjuvant chemotherapy are referred for such treatment by their surgeons. The challenge remains to determine the optimal chemotherapeutic regimen and to reduce barriers to referral by identifying reliable predictors of benefit.

    ADJUVANT CHEMOTHERAPY

    An alternative approach to improving survival in muscle-invasive urothelial cancer, and one that may be more acceptable to the surgical community, is the administration of chemotherapy following radical cystectomy. The benefits of chemotherapy in this setting include: (a) the availability of accurate pathological staging prior to systemic therapy, precluding overtreatment and allowing the identification of patients at high risk for micrometastatic disease, and (b) the minimization of potential delays in definitive local therapy, especially in patients whose cancer may not be chemosensitive and may progress during treatment. This approach does, however, suffer from two major disadvantages: (a) the inability to assess pathological response of primary tumors, and, most important, (b) delays in or intolerance of systemic treatment as a result of postoperative morbidity.

    The benefit of adjuvant chemotherapy was first demonstrated by Logothetis et al. [42] in a retrospective analysis of postoperative cyclophosphamide, doxorubicin, and cisplatin (CISCA) in patients with pathological findings associated with a high risk for recurrence, including the presence of nodal metastasis, extravesicular involvement, or lymphovascular or pelvic visceral invasion. At a median follow-up of 118 weeks, the disease-free survival rates were 70% versus 37% (p = .00012), favoring patients who received adjuvant chemotherapy. Despite the inherent selection bias in this study, the provocative results ultimately led to clinical trials evaluating the role of adjuvant chemotherapy in a prospective fashion.

    Review of existing literature reveals five such trials, all suboptimal in that they were underpowered [43], often enrolling patients with good prognoses [33, 44], not fully analyzed in an intention-to-treat fashion [45], not randomized [46], or employed substandard chemotherapy regimens (Table 4) [45, 47]. Freiha and colleagues at Stanford University randomized 55 patients with pT3–4 or node-positive disease to observation or adjuvant chemotherapy with CMV [43]. With a median follow-up of more than 5 years, CMV resulted in a longer freedom from progression time, a median of 37 months versus 12 months (p = .01), but no difference in overall survival was seen (p = .32). Of note, 20% of the patients in the observation arm were salvaged with CMV at the time of recurrence. The Italian Uro-Oncologic Cooperative group treated 125 patients, with a clinical tumor deemed stage T2, >3 cm, T3, or T4, on a protocol that randomized node-negative patients after radical cystectomy to adjuvant treatment with methotrexate and cisplatin or observation, while all node-positive patients received methotrexate and cisplatin [46]. No survival benefit was seen for treated patients relative to observed patients in either the whole study population or in the randomized node-negative subgroup. Studer et al. [47] randomized 77 patients with invasive, nonmetastatic bladder cancer, without clinical N2 or N3 disease, to adjuvant cisplatin alone or observation. After a median follow-up of almost 6 years, no statistically significant difference in the 5-year overall survival rate was noted (57% vs. 54%; p = .65).

    In contrast, Skinner et al. [45], randomized 91 patients with pathological T3, T4, or node-positive disease to either four cycles of chemotherapy or observation following radical cystectomy. A greater 3-year disease-free survival rate (70% vs. 46%; p = .0010) and median survival time (4.3 years vs. 2.4 years; p = .0062) were observed. In an update published at a later date, a strong trend toward an overall survival benefit for chemotherapy became apparent (p = .056) with an additional 15 months of follow-up. Several criticisms of that trial exist. First, the 91 enrolled patients fell short of the initially planned goal (75 patients per arm). Second, a heterogeneous group of chemotherapy agents was used early in the course of the study, based on an unproven clonogenic assay meant to assess tumor chemo-sensitivity. In fact, only 5 of the first 17 patients in the treatment arm received the defined chemotherapeutic intervention, CISCA. Third, Kaplan-Meier survival curves were evaluated using the Wilcoxon test, which is more sensitive for early differences, perhaps artificially assigning a difference that may not exist with extended follow-up. Although no data are provided, the authors do note that the use of the log-rank test made no difference.

    A similar study was conducted by Stockle et al. [33, 44] at the University of Mainz in Germany. Forty-nine of an originally planned 100 pT3b, pT4a, or pN+ patients were randomized to either three cycles of M-VAC or M-VEC (methotrexate, vinblastine, epirubicin, and cyclophosphamide), depending on treatment site, or observation, after radical cystectomy. In an interim analysis, a significantly higher 3-year relapse-free survival rate (p = .0012) in favor of adjuvant treatment necessitated closure to accrual. Notably, patients with lymph node involvement seemed to benefit the most. No analysis of overall survival by the intention-to-treat principle has been presented to date.

    This body of evidence is significantly less convincing than that for neoadjuvant chemotherapy and remains controversial. Nevertheless, efforts to optimize adjuvant treatments are ongoing. A German phase III trial comparing two adjuvant chemotherapy regimens was recently reported. Patients with pT3a–4a and/or N+ TCC of the bladder were randomized to either three cycles of cisplatin and methotrexate (CM) or three cycles of a standard regimen, M-VEC, after radical cystectomy [48]. Although the 5-year progression-free survival rates were similar (46.3% vs. 48.8%) in this noninferiority trial, the degree of efficacy a clinician must be willing to concede (represented by the upper limit of the 95% CI of the HR for survival) in exchange for reduced toxicity, was 48%. While patients receiving CM experienced less grade 3 and 4 leukopenia (7% vs. 22.2%; p < .0001), there was no difference in the incidence of clinically significant toxicities, including neutropenic fever, infection, and treatment-related death, calling into question the reason for accepting such reduced efficacy.

    NEOADJUVANT VERSUS ADJUVANT THERAPY

    Although each of these potentially curative strategies has advantages and disadvantages, no randomized comparison has been undertaken to date (Table 5). The group from M.D Anderson Cancer Center has, however, compared pre- and postoperative treatment with postoperative treatment alone in patients with high-risk, resectable urothelial cancer (clinical T2 with lymphovascular invasion, T3, or T4a, N0, M0) [49]. One hundred forty patients were randomly assigned to either cystectomy followed by five cycles of adjuvant M-VAC or to two preoperative and three postoperative cycles of M-VAC. The study was powered to detect a 20% difference in survival between the arms (60% to 80%). In the final analysis, there was no significant difference between the groups with respect to overall survival, time to progression, or cause-specific survival. Further studies, directly comparing patients treated with neoadjuvant or adjuvant chemotherapy, are needed to clarify the optimal timing of such treatment.

    BARRIERS TO THE INCORPORATION OF CHEMOTHERAPY INTO THE MANAGEMENT OF LOCALIZED DISEASE

    Despite the cited large-scale randomized trials and meta-analyses showing evidence of a survival benefit for neoadjuvant chemotherapy, it is not widely practiced. Clearly, oncologic urologic surgeons are more concerned by the potential delays in surgery [50] and lack of pathologic staging mandated by the use of neoadjuvant chemotherapy than they are impressed by its efficacy. The 11 years required to accrue patients to the U.S. intergroup trial is evidence to this fact. In that vein, efforts to confirm the benefit of postoperative therapy are ongoing (Table 6).

    Table 6. Phase III trials of perioperative chemotherapy currently open to accrual

    Another barrier to the standard incorporation of perioperative chemotherapy in the management of bladder cancer is the perceived toxicity of cisplatin-based combination regimens. The widespread adoption of GC in the treatment of metastatic disease based on a lower incidence of toxicity has prompted practicing oncologists to apply this regimen in the perioperative setting as a result. In order to confirm the efficacy of GC in this setting, investigators have shown its tolerability in the phase II setting [51] and, in Italy, have initiated a randomized, controlled, postoperative trial with observation as the reference arm [52]. Firm evidence of the efficacy of GC when used with curative intent is preferable to extrapolation.

    Other efforts have focused on developing the combination of carboplatin and paclitaxel as adjuvant therapy [25]. Both carboplatin and paclitaxel have shown single-agent activity in phase II studies [53, 54] but as a combination have not been proven to be superior to M-VAC [23]. Alternatively, high-dose M-VAC with growth factor support has been postulated to be a reasonable area of investigation in the neoadjuvant setting in light of a shorter cycle length (14 days), less toxicity, and higher response rates relative to M-VAC, although no difference in survival was seen in patients with advanced disease [21].

    The EORTC is conducting a well-designed, large-scale study of the value of adjuvant modern chemotherapy, relative to treatment with that same chemotherapy at the time of relapse (Table 6). At least one North American cooperative group (the Cancer and Leukemia Group B), already convinced of the value of adjuvant therapy, abandoned observation as the control arm in a prospective study in an attempt to optimize postoperative treatment. Unfortunately, that trial recently closed as a result of slow accrual, calling into question the ability to study perioperative chemotherapy in the treatment of urothelial cancer in U.S.

    PROGNOSTIC MARKERS

    Prognostic markers in the premolecular era were based solely on clinical and pathological parameters. A retrospective analysis of the long-term outcomes of >1,000 patients at the University of Southern California between 1971 and 1997 found pathologic determinants of survival, stratifying patients into prognostic categories [2]. Patients with organ-confined (pT2 and pT3a), node-negative lesions had 89% and 78% 5-year recurrence-free survival rates, respectively. Patients with extravesical (pT3b and pT4), node-negative lesions had a statistically significant higher risk for recurrence (p < .001), with 5-year recurrence-free survival rates of 62% and 50%, respectively.

    Patients could also be stratified by the presence of lymph node involvement. The 5-year recurrence-free survival rate for patients with lymph node metastasis was 35%, significantly lower than that for patients without node involvement (p < .001). These prognostic factors have been corroborated in a number of studies [55–57]. The presence of either perineural or angiolymphatic invasion are independent predictors of prognosis in some studies [57–60] but not in others [55, 61]. Extracapsular extension of pelvic lymph node metastasis was recently shown to be a marker of a group with a particularly poor prognosis [62]. Unlike for superficial tumors [63], tumor grade has never been shown to predict outcome in the invasive setting [55].

    Surgical factors, including the extent and quality of lymph node dissection, have also been shown to influence survival. In single-institution studies, the number of examined lymph nodes has consistently been correlated with outcomes [64, 65]. Importantly, in the context of a large, multi-institutional trial of neoadjuvant chemotherapy, survival varied both with the extent of lymph node dissection and with the presence or absence of positive margins [66].

    FUTURE DIRECTIONS

    It is important that future multimodality trials incorporate these known surgical and pathologic prognostic factors as stratification variables, to ensure adequate randomization, and by mandating high-quality and extensive lymph node dissections.

    Studies incorporating biological markers of prognosis as stratification and randomization variables have already been initiated. TP53 mutation and resultant p53 nuclear overexpression correlate with progression of superficial TCC to invasive stages [67, 68]. Although the data are somewhat variable, p53 overexpression is also thought to confer a negative prognosis in patients treated with M-VAC chemotherapy in the neoadjuvant and advanced settings, possibly related to resistance [69, 70]. The Southwest Oncology Group and the National Cancer Institute of Canada currently are enrolling patients with organ-confined TCC of the bladder in a marker-dependent, risk-directed trial, randomizing patients with TP53 mutations to M-VAC versus observation; those patients without mutations are observed [71].

    Other biologic predictors of resistance or sensitivity to TCC-directed therapy include P-glycoprotein, glutathione, metalloproteinase, the nucleotide excision-repair (NER) system, BRCA-1, and RRM-1, and their clinical utility is being evaluated [72]. DNA microarray and proteomic analyses are active areas of research. In addition to improving our understanding of urothelial cancer development and progression, it is hoped that they might illuminate new therapeutic targets.

    CONCLUSIONS

    The outcome of local therapy in muscle-invasive, extravesical, or lymph node-involved urothelial cancer is poor. Attempts to improve the cure fraction have included neoadjuvant and adjuvant chemotherapy strategies. Preoperatively, cisplatin-based combinations were proven to be effective in two large, randomized clinical trials of M-VAC and CMV. Additional validation for this approach derives from a meta-analysis including these and other trials. Still, today the neoadjuvant approach is not practiced as commonly as one might expect. Obstacles to the employment of neoadjuvant chemotherapy include the inaccuracy of clinical staging techniques, the necessary delay in definitive local therapy, and concerns regarding the generalizability of existing data [73].

    Data for the use of adjuvant chemotherapy are significantly less compelling, and it remains controversial, although most medical oncologists feel it is reasonable to offer such treatment to patients found to have pathologic evidence of extravesical invasion or lymph node involvement. Certainly, one might extrapolate from other tumors for which neoadjuvant and adjuvant chemotherapy were equally effective [32], but in the attempt to cure, explicit data in urothelial cancer are desirable. The ongoing EORTC study of early versus delayed chemotherapy in pT3–4 and/or lymph node-positive disease may confirm the value of adjuvant chemotherapy.

    Investigation of the clinical utility of biologic markers is expanding as new markers are discovered, and exciting techniques, including quantitative real-time polymerase chain reaction and DNA microarray and proteomic analysis, are applied. It is important to continue to support the clinical trials that will clarify the efficacy of adjuvant chemotherapy, perhaps elucidate the optimal timing of chemotherapy (i.e., neoadjuvant vs. adjuvant), and incorporate these new biologic markers. Finally, it is important to remember that a multidisciplinary team composed of medical oncologists, surgical oncologists, and others is required to obtain the best results for patients.

    DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST

    The authors indicate no potential canflicts of interest.

    REFERENCES

    Jemal A, Murray T, Ward E et al. Cancer Statistics, 2005. CA Cancer J Clin 2005;55:10–30.

    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.

    Stein JP, Skinner DG. The role of lymphadenectomy in high-grade invasive bladder cancer. Urol Clin North Am 2005;32:187–197.

    Herr H, Shipley W, Bajorin D. Cancer of the bladder. In: DeVita V Jr, Hellman S, Rosenberg S, eds. Cancer: Principles and Practice of Oncology, Sixth Edition. Philadelphia: Lippincott, Williams, and Wilkins, 2001:1396–1415.

    Shelley MD, Barber J, Wilt T et al. Surgery versus radiotherapy for muscle invasive bladder cancer. Cochrane Database Syst Rev 2002;(1): CD002079.

    Jenkins BJ, Caulfield MJ, Fowler CG et al. Reappraisal of the role of radical radiotherapy and salvage cystectomy in the treatment of invasive (T2/T3) bladder cancer. Br J Urol 1988;62:343–346.

    Duncan W, Quilty PM. The results of a series of 963 patients with transitional cell carcinoma of the urinary bladder primarily treated by radical megavoltage X-ray therapy. Radiother Oncol 1986;7:299–310.

    Parsons JT, Million RR. Planned preoperative irradiation in the management of clinical stage B2-3 (T3) bladder carcinoma. Int J Radiat Oncol Biol Phys 1988;14:797–810.

    Slack NH, Bross ID, Prout GR Jr. Five-year follow-up results of a collaborative study of therapies for carcinoma of the bladder. J Surg Oncol 1977;9:393–405.

    Smith JA Jr, Crawford ED, Paradelo JC et al. Treatment of advanced bladder cancer with combined preoperative irradiation and radical cystectomy versus radical cystectomy alone: a phase III intergroup study. J Urol 1997;157:805–807; discussion 807–808.

    Huncharek M, Muscat JF, Geschwind J. Planned preoperative radiation therapy in muscle invasive bladder cancer; results of a meta-analysis. Anticancer Res 1998;18:1931–1934.

    Reisinger SA, Mohiuddin M, Mulholland SG. Combined pre- and postoperative adjuvant radiation therapy for bladder cancer--a ten year experience. Int J Radiat Oncol Biol Phys 1992;24:463–468.

    Natale RB, Yagoda A, Watson RC et al. Methotrexate: an active drug in bladder cancer. Cancer 1981;47:1246–1250.

    Blumenreich MS, Yagoda A, Natale RB et al. Phase II trial of vinblastine sulfate for metastatic urothelial tract tumors. Cancer 1982;50:435–438.

    Yagoda A, Watson RC, Whitmore WF et al. Adriamycin in advanced urinary tract cancer: experience in 42 patients and review of the literature. Cancer 1977;39:279–285.

    Yagoda A, Watson RC, Gonzalez-Vitale JC et al. Cis-dichlorodiamminepla tinum(II) in advanced bladder cancer. Cancer Treat Rep 1976;60:917–923.

    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;133:403–407.

    Loehrer PJ Sr, Einhorn LH, Elson PJ et al. A randomized comparison of cisplatin alone or in combination with methotrexate, vinblastine, and doxorubicin in patients with metastatic urothelial carcinoma: a cooperative group study. J Clin Oncol 1992;10:1066–1073.

    Logothetis CJ, Dexeus FH, Finn L et al. A prospective randomized trial comparing MVAC and CISCA chemotherapy for patients with metastatic urothelial tumors. J Clin Oncol 1990;8:1050–1055.

    Saxman SB, Propert KJ, Einhorn LH et al. Long-term follow-up of a phase III intergroup study of cisplatin alone or in combination with methotrexate, vinblastine, and doxorubicin in patients with metastatic urothelial carcinoma: a cooperative group study. J Clin Oncol 1997;15:2564–2569.

    Sternberg CN, de Mulder PH, Schornagel JH et al. Randomized phase III trial of high-dose-intensity methotrexate, vinblastine, doxorubicin, and cisplatin (MVAC) chemotherapy and recombinant human granulocyte colony-stimulating factor versus classic MVAC in advanced urothelial tract tumors: European Organization for Research and Treatment of Cancer Protocol No. 30924. J Clin Oncol 2001;19:2638–2646.

    Bamias A, Aravantinos G, Deliveliotis C et al. Docetaxel and cisplatin with granulocyte colony-stimulating factor (G-CSF) versus MVAC with G-CSF in advanced urothelial carcinoma: a multicenter, randomized, phase III study from the Hellenic Cooperative Oncology Group. J Clin Oncol 2004;22:220–228.

    Dreicer R, Manola J, Roth BJ et al. Phase III trial of methotrexate, vinblastine, doxorubicin, and cisplatin versus carboplatin and paclitaxel in patients with advanced carcinoma of the urothelium. Cancer 2004;100:1639–1645.

    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;18:3068–3077.

    Bamias A, Deliveliotis C, Aravantinos G et al. Adjuvant chemotherapy with paclitaxel and carboplatin in patients with advanced bladder cancer: a study by the Hellenic Cooperative Oncology Group. J Urol 2004;171:1467–1470.

    von der Maase H, Sengelov L, Roberts J et al. 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; 23:4602–4608.

    Bellmunt J, Guillem V, Paz-Ares L et al. Phase I–II study of paclitaxel, cisplatin, and gemcitabine in advanced transitional-cell carcinoma of the urothelium. Spanish Oncology Genitourinary Group. J Clin Oncol 2000;18:3247–3255.

    Philips G, Halabi S, Sanford B et al. Phase II trial of cisplatin (C), fixed-dose rate gemcitabine (G) and gefitinib for advanced transitional cell carcinoma (TCC) of the urothelial tract: preliminary results of CALGB 90102. J Clin Oncol 2004;22(14 suppl):4540.

    Hussain M, Petrylak D, Dunn R et al. Trastuzumab (T), paclitaxel (P), carboplatin (C), and gemcitabine (G) in advanced HER2-positive urothelial carcinoma: results of a multi-center phase II NCI trial. Presented at the 2005 Annual Meeting of the American Society of Clinical Oncology, Orlando, FL, 2005, May 13–17, 2005.

    Rich TA, Skibber JM, Ajani JA et al. Preoperative infusional chemoradiation therapy for stage T3 rectal cancer. Int J Radiat Oncol Biol Phys 1995;32:1025–1029.

    Sauer R, Becker H, Hohenberger W et al. Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med 2004;351:1731–1740.

    Fisher B, Bryant J, Wolmark N et al. Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. J Clin Oncol 1998;16:2672–2685.

    Stockle M, Meyenburg W, Wellek S et al. Advanced bladder cancer (stages pT3b, pT4a, pN1, and pN2): improved survival after radical cystectomy and 3 adjuvant cycles of chemotherapy. Results of a controlled prospective study. J Urol 1992;148:302–306; discussion 306–307.

    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.

    Cortesi E. Neoadjuvant treatment for locally advanced bladder cancer: a randomized prospective clinical trial. Proc Am Soc Clin Oncol 1995;14:237..

    Winquist E, Kirchner TS, Segal R et al. Neoadjuvant chemotherapy for transitional cell carcinoma of the bladder: a systematic review and meta-analysis. J Urol 2004;171:561–569.

    Advanced Bladder Cancer Meta-analysis Collaboration. Neoadjuvant chemotherapy in invasive bladder cancer: a systematic review and meta-analysis. Lancet 2003;361:1927–1934.

    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.

    Hall R. Updated results of a randomised controlled trial of neoadjuvant cisplatin (C), methotrexate (M), and vinblastine (V) chemotherapy for muscle-invasive bladder cancer. Proc Am Soc Clin Oncol 2002;21:710a.

    Neoadjuvant cisplatin, methotrexate, and vinblastine chemotherapy for muscle-invasive bladder cancer: a randomised controlled trial. Lancet 1999;354:533–540.

    Sternberg CN, Parmar MK. Neoadjuvant chemotherapy is not (yet) standard treatment for muscle-invasive bladder cancer. J Clin Oncol 2001;19(18 suppl):21s–6s.

    Logothetis CJ, Johnson DE, Chong C et al. Adjuvant cyclophosphamide, doxorubicin, and cisplatin chemotherapy for bladder cancer: an update. J Clin Oncol 1988;6:1590–1596.

    Freiha F, Reese J, Torti FM. A randomized trial of radical cystectomy versus radical cystectomy plus cisplatin, vinblastine, and methotrexate chemotherapy for muscle-invasive bladder cancer. J Urol 1996;155:495–499; discussion 499–500.

    Stockle M, Meyenburg W, Wellek S et al. Adjuvant polychemotherapy of nonorgan-confined bladder cancer after radical cystectomy revisited: long-term results of a controlled prospective study and further clinical experience. J Urol 1995;153:47–52.

    Skinner DG, Daniels JR, Russell CA et al. The role of adjuvant chemotherapy following cystectomy for invasive bladder cancer: a prospective comparative trial. J Urol 1991;145:459–464; discussion 464–467.

    Bono AV, Benvenuti C, Reali L et al. Adjuvant chemotherapy in advanced bladder cancer. Italian Uro-Oncologic Cooperative Group. Prog Clin Biol Res 1989;303:533–540.

    Studer UE, Bacchi M, Biedermann C et al. Adjuvant cisplatin chemotherapy following cystectomy for bladder cancer: results of a prospective randomized trial. J Urol 1994;152:81–84.

    Lehmann J, Retz M, Wiemers C et al. Adjuvant cisplatin plus methotrexate versus methotrexate, vinblastine, epirubicin, and cisplatin in locally advanced bladder cancer: results of a randomized, multicenter, phase III trial (AOU-AB 05/95). J Clin Oncol 2005;23:4963–4974.

    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.

    Lee CT, Madii R, Daignault S et al. Cystectomy delay more than 3 months from initial bladder cancer diagnosis results in decreased disease specific and overall survival. J Urol 2006;175:1262–1267; discussion 1267.

    Meliani E, Lapini A, Serni S et al. Gemcitabine plus cisplatin in adjuvant regimen for bladder cancer. Toxicity evaluation. Urol Int 2003;71: 37–40.

    Clinical Trials.gov. Cisplatin and Gemcitabine Compared With Observation in Treating Patients Who Have Undergone Surgery for Bladder Cancer. Available at http://www.clinicaltrials.gov/ct/show/NCT00054626?order=4. Accessed August 5, 2005.

    Roth BJ, Dreicer R, Einhorn LH et al. Significant activity of paclitaxel in advanced transitional-cell carcinoma of the urothelium: a phase II trial of the Eastern Cooperative Oncology Group. J Clin Oncol 1994;12: 2264–2270.

    Mottet-Auselo N, Bons-Rosset F, Costa P et al. Carboplatin and urothelial tumors. Oncology 1993;50(suppl 2):28–36.

    Jimenez RE, Gheiler E, Oskanian P et al. Grading the invasive component of urothelial carcinoma of the bladder and its relationship with progression-free survival. Am J Surg Pathol 2000;24:980–987.

    Novara G, Ficarra V, Alrabi N et al. Prognostic factors in a recent series of patients treated with radical cystectomy for bladder cancer. Urol Int 2005;75:10–16.

    Knap MM, Lundbeck F, Overgaard J. Prognostic factors, pattern of recurrence and survival in a Danish bladder cancer cohort treated with radical cystectomy. Acta Oncologia 2003;42:160–168.

    Leissner J, Koeppen C, Wolf HK. Prognostic significance of vascular and perineural invasion in urothelial bladder cancer treated with radical cystectomy. J Urol 2003;169:955–960.

    Thieblemont C, Fendler JP, Trillet-Lenoir V et al. [Prognostic factors of survival in infiltrating urothelial bladder carcinoma. A retrospective study of 158 patients treated by radical cystectomy.] Bull Cancer 1996;83:139–146. French.

    Lotan Y, Gupta A, Shariat SF et al. Lymphovascular invasion is independently associated with overall survival, cause-specific survival, and local and distant recurrence in patients with negative lymph nodes at radical cystectomy. J Clin Oncol 2005;23:6533–6539.

    Nangia AK, Konety BR, Wachtel S et al. Perineural invasion in transitional cell carcinoma and the effect on prognosis following radical cystectomy. Urology 1997;49:968–972.

    Fleischmann A, Thalmann G, Markwalder R et al. Extracapsular extension of pelvic lymph node metastases from urothelial carcinoma of the bladder is an independent prognostic factor. J Clin Oncol 2005;23: 2358–2365.

    Jordan AM, Weingarten J, Murphy WM. Transitional cell neoplasms of the urinary bladder. Can biologic potential be predicted from histologic grading? Cancer 1987;60:2766–2774.

    Herr HW, Bochner BH, Dalbagni G et al. Impact of the number of lymph nodes retrieved on outcome in patients with muscle invasive bladder cancer. J Urol 2002;167:1295–1298.

    Leissner J, Hohenfellner R, Thuroff JW et al. Lymphadenectomy in patients with transitional cell carcinoma of the urinary bladder; significance for staging and prognosis. BJU Int 2000;85:817–823.

    Herr HW, Faulkner JR, Grossman HB et al. Surgical factors influence bladder cancer outcomes: a cooperative group report. J Clin Oncol 2004;22:2781–2789.

    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.

    Serth J, Kuczyk M, Bokemeyer C et al. p53 immunohistochemistry as an independent prognostic factor for superficial transitional cell carcinoma of the bladder. Br J Cancer 1995;71:201–205.

    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.

    Jankevicius F, Goebell P, Kushima M et al. p21 and p53 immunostaining and survival following systemic chemotherapy for urothelial cancer. Urol Int 2002;69:174–180.

    ClinicalTrials.gov. Phase III Randomized Study of Methotrexate, Vinblastine, Doxorubicin, and Cisplatin Versus Observation Alone Based on p53 Gene Status in Patients With Organ Confined Transitional Cell Carcinoma of the Bladder Who Have Undergone Radical Cystectomy and Bilateral Pelvic Lymphadenectomy. Available at http://www.clinicaltrials.gov/ct/show/NCT00005047?order=1. Accessed August 5, 2005.

    Bajorin D, Bochner B, Sandler H et al. Changing paradigms in advanced bladder cancer. In: Perry MC, ed. American Society of Clinical Oncology: Educational Book 2005. Alexandria, VA: Lisa Greaves, 2005:330–350.

    Madersbacher S, Thalmann GN, Fritsch JC et al. Is eligibility for a chemotherapy protocol a good prognostic factor for invasive bladder cancer after radical cystectomy? J Clin Oncol 2004;22:4103–4108.

    Greene FL, Page DL, Fleming ID et al. AJCC Cancer Staging Manual, Sixth Edition. New York: Springer 2002.

    Wallace DM, Raghavan D, Kelly KA et al. Neo-adjuvant (pre-emptive) cisplatin therapy in invasive transitional cell carcinoma of the bladder. Br J Urol 1991;67:608–615.

    Font A, Abad A, Saladie J et al. Improved survival with induction chemotherapy in bladder cancer: preliminary results of a randomized trial. Ann Oncol 1994;5:71.

    Martinez-Pineiro JA, Gonzalez-Martin M, Arocena F et al. Neoadjuvant cisplatin chemotherapy before radical cystectomy in invasive transitional cell carcinoma of the bladder: a prospective randomized phase III study. J Urol 1995;153:964–973.

    Malmstrom PU, Rintala E, Wahlqvist R et al. Five-year followup of a prospective trial of radical cystectomy and neoadjuvant chemotherapy: Nordic Cystectomy Trial I. The Nordic Cooperative Bladder Cancer Study Group. J Urol 1996;155:1903–1906.

    Bassi P, Pappagallo G, Sperandio P et al. Neoadjuvant M-VAC chemotherapy of invasive bladder cancer: results of a multicenter phase III trial. J Urol 1999;161(4 suppl):264.

    Sherif A, Rintala E, Mestad O et al. Neoadjuvant cisplatin-methotrexate chemotherapy for invasive bladder cancer--Nordic Cystectomy Trial 2. Scand J Urol Nephrol 2002;36:419–425.

    Sengelov L, von der Maase H, Lundbeck F et al. Neoadjuvant chemotherapy with cisplatin and methotrexate in patients with muscle-invasive bladder tumours. Acta Oncol 2002;41:447–456.(Seth M. Cohena,e, Anupama)