Schistosomiasis and bladder cancer: similarities and ...

24 downloads 81 Views 252KB Size Report
Bladder cancer (BC) represents a significant worldwide health problem, with an estimated. 386,300 new cases and 150,200 deaths in 2008 worldwide.
Review

THEMED ARTICLE y Bladder Cancer

Schistosomiasis and bladder cancer: similarities and differences from urothelial cancer Expert Rev. Anticancer Ther. 12(6), 753–763 (2012)

Mohamed S Zaghloul* and Iman Gouda Radiation Oncology and Pathology Departments, Children’s Cancer Hospital and National Cancer Institute, 1 Sekket El Emmam, El Sayeda Zainab, Cairo University, Cairo, Egypt *Author for correspondence: Tel.: +20 225 351 500 (ext. 1192) [email protected]

Through the years, schistosoma-associated bladder cancer was believed to be a unique entity of disease, different from urothelial cancer. As carcinogenesis is a highly complex process resulting from the accumulation of many genetic and epigenetic changes leading to alterations in the cell proliferation and regulation process, confirmation of their minute differences or similarities are extremely difficult. In bladder cancer, many of these carcinogenic cascades were not fully documented in spite of the efforts undertaken. The control of schistosomiasis and the subsequent decrease in the intensity of infestation showed feature changes approaching that of urothelial tumors. However, schistosoma-associated bladder cancer still presents in more advanced stages than schistosoma-non-associated urothelial cancer. Furthermore, many data were collected proving that, upon applying the same treatment protocol and management care, stage-by-stage comparison of the treatment end results were found to be similar in bladder cancer patients with the different etiologies. Keywords : bilharziasis • biology • bladder cancer • chemotherapy • cytogenetics • pathology • radical cystectomy • radiotherapy • schistosomiasis

Bladder cancer (BC) represents a significant worldwide health problem, with an estimated 386,300 new cases and 150,200 deaths in 2008 worldwide. The majority of BC occurs in males, and there is a 14-fold variation in incidence internationally. The highest incidence rates are found in the countries of Europe, North America and Northern Africa [1] . It is estimated that one in every 26 males and one in every 87 females in the USA will develop BC during their life [2] . Smoking and occupational exposures are the major risk factors in Western countries, whereas chronic infection with Schistosoma haematobium (SH) in developing countries, particularly in Africa and the Middle East, accounts for approximately 50% of the total burden. The majority of BCs associated with schistosomiasis are squamous cell carcinomas (SCC). Although the majority of the BC population present with disease confined to the superficial layer of the bladder wall, approximately 20–40% of the patients will present with or subsequently develop invasive cancer. BC is morphologically heterogeneous; more than www.expert-reviews.com

10.1586/ERA.12.49

90% of BC cases are urothelial carcinoma (UC; transitional cell carcinoma [TCC]), whereas primary SCC, adenocarcinoma, small cell carcinoma and other rare tumors are less common [3] . Urothelial cell carcinoma may present mixed with other malignant components. These mixed forms of bladder histologies include squamous differentiation (present in 20–60% of BC cases), adenocarcinoma or glandular differentiation (10%), sarcomatoid (7%), micropapillary (3.7%) and lympho­ epithelioma-like carcinoma. In many developing countries, life expectancy is much lower than in Western countries, which is one of the reasons why overall BC incidence (not agespecific incidence) is lower in these developing countries [2] . BC is associated with substantial morbidity and mortality. History of tobacco smoking not only increases the incidence of BC, but it can also increase tumor grade, size and number [4] . Chronic schistosomal cystitis was related for a long period to the development of BC in areas endemic for schistosomiasis such as Egypt. In these areas, there are many

© 2012 Expert Reviews Ltd

ISSN 1473-7140

753

Review

Zaghloul & Gouda

risk factors, including exposure to schistosomiasis, increased ­smoking rate and exposure to carcinogenic chemicals [5] . Although smoking is recognized as a major risk factor in the etiology of BC, the increasing incidence of BC, despite the reduction in smoking in the USA, suggests that other environmental factors may have a substantial role in the development of this cancer. On the contrary, there was a 5.14% decrease in the death rate due to BC in the USA, upon comparing the death rate in the years 1990 and 2007 [2] , owing to the success in management. Other risk factors, such as positive family history, parents’ consanguinity, exposure to pesticides and chronic cystitis, seem to play roles beside schistosomiasis and smoking in development of this disease in the developing countries such as Egypt [6] . BC formation

Urothelial tumor is characterized by its multifocality. There have been two theories proposed to explain the frequency of this urothelial tumor multifocality. The monoclonal theory suggests that multiple tumors arise from a single transformed cell that proliferates and spreads throughout the urothelium. The second theory (the field-effect theory) suggests independent transforming genetic alterations in the urothelial lining, at different sites, leading to multiple tumors [7] . This may highlight the existence of different histopathologies in the same specimen. Furthermore, many other theories have been proposed to combine the two mechanisms. It is known that the modern carcinogenesis model suggests that malignancy represents clonal expansion of one cancer stem cell that proliferates and can diversify into heterogeneous cancer cell lineages through uneven differentiation at different carcinogenic steps. This asymmetric differentiation means that following cell division, one daughter cell retains the capacity to divide while the other daughter cell possesses genetic plasticity. Through this, phenotypic variation is allowed in the offspring. A specific set of genomic, epigenomic and/or microenvironment niche alterations is essential for continued clonal expansion. As a result, each chromosomal somatic change and its pro/zaxgency possess a unique set of genetic, epigenetic and phenotypic features. Stromal somatic genetic cell alterations assist chromosomal somatic changes in the niche to promote cancer development and progression [8] . With the establishment of different new methods, detection of meaningful chromosomal changes became more apparent and allowed better understanding of the process of evolution of different kinds of cancer. Each new method widened the recognition of karyotypic changes and increased the resolution of cytogenetic details [9] . Schistosomia haematobium

According to the WHO, schistosomiasis infects 200 million people and is endemic in as many as 76  tropical developing countries. Schistosomes are parasitic blood flukes, which have a mammalian host and an intermediate invertebrate host: freshwater snails [10] . There are four human schistosomes: SH, Schistosoma  mansoni, Schistosoma japonicum and Schistosoma mekonji. SH is the one associated with BC. SH, like other 754

schistosomes, is dioecious as the adult female lives in copulo in the gynecophoral canal of the male; and lives in the venules of the human urinary bladder. Eggs laid in the urinary bladder produce irritation and tissue fibrosis that may contribute to the development of human carcinogenicity [11] . All schistosoma infections follow direct contact with fresh water that harbors free-swimming larval forms of the parasite cercariae, which penetrate the skin. The cercariae shed their bifurcated tails, and the resulting schistosomula enter capillaries and lymphatic vessels en route to the lungs. After several days, the worms migrate to the portal venous system, where they mature and unite. Pairs of worms then migrate to the vesical plexus and veins draining the ureters. Egg production starts 4–6 weeks after infection and continues throughout the life of the worm, usually 3–5 years. Eggs pass from the lumen of blood vessels into adjacent tissues, and then they pass through the bladder mucosa to shed in the urine. The life cycle is completed when the eggs hatch, releasing miracidia that, in turn, infect specific freshwater snails (Bulinus species). After two generations – primary and then daughter sporocysts within the snail – cercariae are released [12] . Relation of prevalence of schistosomiasis & BC

Ferguson first linked schistosomiasis to urinary BC in Egypt in 1911 [13] . The incidence of urinary BC in the Middle East and Africa is greater in areas with high rather than low SH prevalence. The overall prevalence of SH infection in Egypt was 37–48%, which decreased due to the antibilharzial campaign to 3% [14] . Urinary BC previously accounted for approximately 31% of the total incidence of cancers in Egypt, but that subsequently decreased to 12% in recent years. However, it is still the most common type of cancer in males and the second most prevalent in females [15] . There are several factors that may contribute to the oncological potential of schisto­soma infection. Schistosoma ova deposited in the bladder provoke an intense inflammatory reaction, associated with the production of oxygen-derived free radicals, which may induce genetic mutations or promote the production of carcinogenic compounds (such as N-nitrosamines and polycyclic aromatic hydrocarbons) [16,17] , leading to malignant transformation. Shokeir showed that schistosomiasis is often accompanied by chronic bacterial superinfection, which may in itself predispose to squamous cell neoplasia [18] . Bacteria that usually accompany schistosomiasis can promote the formation of N-nitrosation of amines, adding to those from other sources such as the diet. A 54–81% incidence of SCC was found in cases of BCs in endemic areas, compared with 3–10% in Western countries [19] . The International Agency for Research on Cancer found that the intensity of infection was determined by urinary egg counts and confounded by smoking, a recognized cause of BC in nonendemic countries, and the combination was strongly considered. A positive association between BC and SH infection was detected, with odds ratios ranging from 2 to 14. The more heavily infected individuals were, the more likely they were to develop BC, and at a younger age [20] . Expert Rev. Anticancer Ther. 12(6), (2012)

Schistosomiasis & bladder cancer: similarities & differences from urothelial cancer

Pathology of schistosomiasis & schistosomiasisassociated BC

Most of the pathological findings of schistosomiasis are mainly due to inflammatory and immunological responses to egg deposition. Granulomatous areas form around the eggs and induce an exudative cellular response consisting of lymphocytes, polymorpho­nuclear leukocytes and eosinophils. Resultant perioval granulomas, fibrosis and muscular hypertrophy are detected histologically. In the urinary bladder, masses of large granulomatous inflammatory polyp-containing eggs are found at different walls. Polyps may ulcerate and slough, producing hematuria. Hyperplasia of the urothelium was found in 38% of the autopsied SH cases, metaplasia in 31.6% and dysplasia in 27.2%. Schistosomal bladder ulcers and sandy patches, irregularly thickened or atrophic bladder mucosa, fibrosis and granulomas containing calcified or disintegrating eggs were also seen [21] . The inflammatory and fibrotic response to egg deposition could lead to calcification of the urinary bladder, infection and stone disease, and these changes are frequently associated with schistosomaassociated BC (SA-BC) [22] . Moreover, the following sequences of SH-induced events have been suggested: chronic infection leads to schistosoma eggs being trapped in the bladder wall; proliferation of cells in the bladder mucosa results from constant irritation; and frequent inflammation and clones of neoplastic cells develop. These clones were stimulated by N-nitrosamines and other environmental carcinogens such as cigarette smoke and pesticides [23] . The combined urinary retention, from fibrosis and obstruction of the urinary bladder neck, and the prolonged exposure of the bladder mucosa to various exogenous and endogenous carcinogens were suggested as predisposing factors. Schistosomeinduced urinary stasis allows increased absorption of carcinogens, and therefore plays an integral role in carcinogenesis. Recurrent bacterial urinary tract infections are associated with SCC of the urinary bladder, even in the absence of SH infection [24] . Carcinogenesis of chronic schistosomal infection

Carcinogenesis of SH involving an initiating and promoting effect has been described. First, the damage occurs to the DNA template which, unless repaired, leads to irreversible changes in the complementary strand of DNA produced during the S-phase of the cell cycle. Somatic mutation results when the altered strand is used as a template. The promotion phase is followed by the cell proliferation stimulation phase. Efforts have been made to study the specific genes involved in the induction of SA-BC. Cells exposed to SH cell total antigen (warm extract) were found to divide faster than those not exposed to the parasite and died much less. This was probably due to increased levels of BCL2, a protein involved in cancer apoptosis [25] . Mice urothelium exposed to SH total antigen showed dysplasia, low-grade intraurothelial neoplasm and noninvasive malignant flat lesions in 70% of the tested mice. Bladder carcinoma harbors gene mutations that constitutively activate the receptor tyrosine kinase Ras pathway [26] . Botelho et al. suggested that the parasite abstract may possess carcinogenic ability through oncogenic mutation of the KRAS gene [27] . www.expert-reviews.com

Review

Among the most common genetic changes in BC is the loss of heterozygosity on chromosomes 9p and 9q, which is found regardless of tumor grade end stage [28,29] . A prospective study stated that there was no evident line of demarcation between SA-BC and schistosoma non-associated BC (SNA-BC) in terms of loss of heterozygosity of microsatellite markers on chromosome 9. This suggests that data obtained from SA-BC can be extrapolated to BC induced via a schistosomiasis-independent mechanism [30] . BC is a very heterogeneous disease cytogenetically, which suggests that the pathogenesis of the disease may not be consistent for every case. The heterogeneity of cytogenetic findings in BC hints that there may be different ‘fingerprints’ or accumulations of genetic changes that individually lead to BC. The overexpression of the BCL-2 gene in SA-BC patients was found to be up­regulated in squamous but not transitional cell cancers of the urinary bladder. Therefore, this BCL-2 overexpression is consistent with the predominance of SCC in SA-BC. Upregulation of this gene overrides programed cell apoptosis, increasing the risk of genomic instability, and may interact with various proto-oncogenes leading to tumorigenesis. Mutations of TP53 were found in 73% of tumors, BCL-2 expression in 32% and abnormalities of both TP53 and BCL-2 in 13% [31] . Furthermore, COX-2 is overexpressed in SA-BC. The role of COX-2 in the complex multistage process of SA-BC carcinogenesis was proposed: proinflammatory cytokines such as IL-1, TGF-b and TNF-a are generated by activated macrophages in the inflammatory lesions. These growth factors and cytokines are potent inducers of COX-2 production. Additional mutations may be induced: H-RAS, deletion of p16 and p15, increased EGF receptor, c-ERB-2 and TNF-a. Increased prostaglandin production upregulates COX-2, decreases killer T-cell activity and increases BCL-2 and glutathione-S-transferase. These changes increase tumorigenicity by decreasing cell apoptosis and/or creating immuno­suppression. Prostaglandin products of COX-2 cause tumor progression and eventual metastasis by downregulating adhesion molecules, increasing the degradation of the extracellular matrix and increasing angiogenesis [32] . Schistosoma-associated BC

The association between SA-BC and SH was established through many studies, mostly case-controlled ones, and through the intimate correlation of the incidence of BC with the prevalence of SH within different geographic areas. This association was based on the frequent presence of parasitic eggs and egg-induced granulomatous pathology in the examined BC tissue materials. The cellular mechanisms linking SH infestation with BC formation are not yet completely determined. Severe metaplasia in bladder urothelium may represent a precancerous transformation, while in others it may be merely an indicator of prolonged inflammation, which is associated with high cancer risk [33] . It is known that keratinizing metaplasia per se has a strong association with carcinogenesis in patients with chronic irritation due to bladder stones, chronic infection or prolonged catheterization as it has with schistosomiasis. A defined characteristic pathology (i.e., SCC, TCC or adenocarcinoma, rather than mainly transitional) was established by 755

Review

Zaghloul & Gouda

SA-BC. Cellular and molecular biology may differ from SNA-BC. Few studies have analyzed the cytogenetic and molecular genetic abnormalities in SA-BC and some compared DNA copy number changes in SA-BC and SNA-BC [4,5,34,35] . Demonstrated deletion similarities in schistosoma-associated TCC (SA-TCC) and schistosoma-associated squamous cell carcinoma (SA-SCC) [4] were compared with what has been reported by Kallioniemi et al. on SNA-TCC [5] and Tsutsumi et al. on SNA-SCC [34] . Pools of tissue arising from patients having similar pathological types, were used to more accurately determine the change. These pooled DNAs revealed recurrent primary changes and covered the secondary changes that vary from case to case. The pooled specimens of SA-BC tumors showed no schistosomiasis-specific changes, compared with pools of SNA tumors [36] . The comparison between SA-TCC and SNA-TCC and that between SA-SCC and SNA-SCC gave similar results. Furthermore, the DNA copy number profiles of SA adenocarcinoma BC revealed similarities to that of SA-TCC and SA-SCC [37] . Detailed analysis of individual genes revealed a set of genes with the same copy number changes in all bladder carcinomas, including both SA and SNA tumors. Armengol et al. concluded that there are no major cytogenetic differences among different urinary bladder epithelial tumors, regardless of the suspected predisposed carcinogen [36] . All the detected imbalances in SA-BC have been repeatedly reported in SNA-BC, which suggested that the cytogenetic profiles of chemical- and schistosoma-induced carcinoma are largely similar in the reports of Muscheck et al. [4] and Fadl-Elmula et al. [35] . The decreased intensity of schistosomal infestation in Egypt led to a changing pattern of the clinicoepidemiologic features of SA-BC. The reported clinicoepidemiologic differences between SA-BC and SNA-BC are now continuously decreasing, and the features of SA-BC are slowly approaching that of SNA-BC as reported by Koraitim et al. [38] and Zaghloul et al. [39] . If these changes continue, BC in Egypt is expected to become identical in features to that of Western countries in the near future [15,39] . Clinical presentation

Clinical presentations in SA-BC and SNA-BC are similar, with a few minor differences. Hematuria, dysuria and necroturia are the main symptoms in both situations. However, SA-BC patients have usually experienced these symptoms beforehand as a result of simple schistosomal cystitis. This may be the reason of their relatively late presentation. Table 1 shows the postcystectomy pathological staging in large SA-BC and SNA-BC studies. The early stages (Pa, Pis and P1) were fewer in SA-BC than that in SNA-BC (urothelial and nonurothelial). The pelvic nodal involvement was nearly similar in SA-BC (range: 16.7–25.5%), urothelial SNA-BC (range: 16.3–45%) and nonurothelial SNA-BC (range: 21.8–23%). The clinicopathologic differences between SA-BC and SNA-BC were previously summarized as: late presentation; younger median age; and a higher percentage of SCC [40] . Distant metastases are similar in SA-BC and SNA-BC, including the different histopathological variants. In a study including 357 SA-BC patients, the 5-year incidence of distant metastasis was 23%. The 5-year actuarial incidence of distant metastasis varied according 756

to the tumor cell type. SCC led to a distant metastasis incidence rate of 15% at 5 years, whereas the transitional cell variety led to a distant metastasis incidence of 39. The metastasis rates were 57% for adenocarcinoma. Although this difference was statistically significant on univariate analysis, it was not statistically significant when multivariate analysis was applied [41] . Similarly, the incidence of distant metastasis in urothelial cancer ranged from 12 to 35% [42,43] . Locoregional recurrence with or without distant metastasis was found in 5–31% of the patients treated with radical cystectomy (RC), depending on their clinical and pathological features [44] . Factors associated with local recurrence are generally the same as that determining distant metastasis, namely pathologic tumor stage, grade and regional nodal involvement together with the lymphadenectomy extent. Rates of local recurrence were reported as 6, 18 and 51% in patients with stage pT1, pT2 and pT3, respectively, in a SNA-BC study [44] . The 5-year local recurrence rate increased with higher stage, and it reached 11, 23 and 31% for stages II, III and IV, respectively, as shown in another SNA-BC study [45] . In a third study, the local recurrence rate at 10 years was reported to be 25% for pT3, 51% for pT4 and 49% in nodepositive patients [46] . A fourth study showed that the 10-year recurrence rate was 48.6% [47] . Out of the 444 SNA-BC patients with relapse, 182 (41%) patients experienced local recurrence and 324 (73%) patients developed distant metastasis at the time of first relapse [47] . Furthermore, Gupta et al. reported 145 BC recurrences out of 502 patients (29%) who underwent RC [48] . Of these, 40 (27.6%) patients had local pelvic recurrence while 105 (72.4%) patients developed distant metastasis. Similar results of locoregional recurrences were repeatedly reported in SA-BC [49–53] . Treatment of non-muscle-invasive BC

Treatment of non-muscle-invasive (superficial) BC remains as transurethral resection of the bladder tumor with or without intravesical Bacille Calmette–Guérin or chemotherapy instillation, according to the tumor risk status. Although this treatment type is very popular in urothelial cancer, it is less popular in SA-BC and nonurothelial SNA-BC, probably due to the lower number of noninvasive stages and the prevalence of many precancerous or cancerous lesions in the bladder mucosa. Treatment of muscle-invasive BC Radical cystectomy

Muscle-invasive BC is mostly treated with RC in many parts of the world. The RC procedure includes removal of the bladder, seminal vesicles and prostate together with perivesical fat and peritoneal coverage, in addition to bilateral endopelvic lymphadenectomy (with varying levels of dissection) in male patients. In females, it includes removal of the bladder, its perivesical fat and peritoneal coverage, urethra, uterus, ovary and anterior wall of the vagina (anterior pelvic excentration) [52–54] . A review of recent literature of treatment end-results of different types of BC showed that applying the same treatment yielded nearly the same level of results in the identical pathological stage [51,55] . Similar 5-year overall survival rates were achieved in SA-BC, Expert Rev. Anticancer Ther. 12(6), (2012)

Schistosomiasis & bladder cancer: similarities & differences from urothelial cancer

Review

pure urothelial and combined urothelial and Table 1. Postcystectomy pathological stages and nodal involvement nonurothelial SNA-BC types (Table 2) . The in pure urothelial and mixed urothelial and nonurothelial results were slightly higher in the Stein et al. schistosoma-non-associated and schistosoma-associated bladder (SNA-BC) [54] and Zaghloul studies (SAcancer in large series. BC) [51] . Both studies reported neoadjuvant Patients PTa,is,1 PT2 PT3 PT4 Nodal Ref. or adjuvant radiotherapy and/or chemo- Study (year) (n) (%) (%) (%) (%) involvement therapy as a part of treatment in more than one-third of their patients. Furthermore, SNA-BC (pure urothelial carcinoma) this conclusion applies for the comparison of Bassi et al. (1999) 338 [82] 32.8 19.8 42.0 19.8 NM disease-free survival, overall survival or local [54] 39.9 23.5 23.5 13.0 23.3 control rates for RC, or even in adjuvant and Stein et al. (2001) 1057 [46] 303 36.1 28.6 25.5 9.9 16.3 neoadjuvant radiotherapy types of treatment Cheng et al. (2000) for SA-BC and SNA-BC [51,55] . The treatment end-results of RC were not affected [83] 958 22.0 35.0 31.0 12.0 23.0 by the association with schistosomiasis, nor Shariat et al. (2006) tumor cell type (urothelial or nonurothelial) in most of the recently published literature SNA-BC (urothelial and nonurothelial) (Table 2) [52,54,56] . Favorable end-results were [58] Rogers et al. 955 21.0 33.0 32.0 14.0 23.0 reported for patients with pathologically (2006) organ-confined disease. These results were [84] Lughezzani et al. 12003 13.4 38.9 28.1 19.6 21.8 constant for both SA-BC (ranged from 47 (2010) to 83%), and SNA-BC (ranged from 50 to [59] 1422 14.8 29.0 29.3 26.8 84%). However, the results were signifi- Scosyrev et al. (2009) cantly worse when reporting upon locally advanced tumors (PT3N0M0, PT4aN0M0 SA-BC (urothelial and nonurothelial) [49] or Any N). Again, worse results were experi- El Said et al. 420 1.0 3.8 70.7 24.5 16.7 (1979) enced by both SA-BC and SNA-BC patients [52,54,56,57] . Despite the old belief that aber- Zaghloul (1996) [41] 357 0.0 33.3 47.9 18.8 24.4 rant differentiation leads to worse results, [85] El-Mekresh et al. 185 7.0 25.0 64.0 7.0 16.0 many authors reported similar results of (1998) these variants to UC when comparing stage [50] 192 3.6 28.1 51.6 16.7 25.5 to stage. Rogers et  al. reported a 5-year Zaghloul et al. (1992) progression-free survival rate of 60  ±  2% [53] 2720 10.5 63.9 16.6 9.0 20.4 after RC for UC and 55  ±  11% for SCC Ghoneim et al. (2008) [58] . This difference was statistically insig[39] 5071 1.9 30.1 54.9 13.1 21.9 nificant. Patients with UC or SCC had Zaghloul et al. (2008) statistically significant higher progression[86] free survival rates than non-UC non-SCC Khaled et al. 180 1.2 5.8 14.1 11.5 16.6 (2005) patients, including those with adenocarcinoma. Another study containing a Ali-El-Dein (2009) 180 [87] 10.0 62.8 25.0 2.2 18.3 considerable number of adenocarcinoma NM: Not mentioned; SA-BC: Schistosoma-associated bladder cancer; SNA-BC: Schistosoma-non-associated patients conducted using the Surveillance, bladder cancer. Epidemiology and End Results (SEER) 17 database showed no difference of statistical significance in adeno- treated in The Netherlands during a 12-year period (1995–2006) carcinoma patients who underwent RC than their UC counter- [60] . They concluded that the relative survival of muscle-invasive parts. Another recent study using a similar SEER database dem- adenocarcinoma patients was equal to that of UC patients. For onstrated that SCC was more aggressive than urothelial cancer stage II and III disease, adenocarcinoma patients had even better after adjusting for common prognostic factors, such as stage and outcomes. Muscle-invasive SCC patients showed worse survival grade [59] . Scosyrev et al. concluded that SCC was an indepen- regardless of stage. In SA-BC, Ghoneim et al. demonstrated that dent predictor of mortality among patients with stage III and IV 1345 SCC patients had a 10-year overall survival (OS) rate of disease, and among patients with stage I and II disease who did 53% compared with 48% for pure UC (705 patients) and 51% for not undergo cystectomy as part of their treatment [59] . Therefore, adenocarcinoma (262 patients) [53] . Those patients who had UC squamous histology was not associated with increased mortality with squamous or adenomatous metaplasia (286 patients) showed among patients with stage I and II disease when treated with a lower 10‑year OS of 43%. The lowest 10-year OS was expecystectomy. Moreover, Ploeg et al. studied all invasive BC cases rienced by those patients who had undifferentiated pathology www.expert-reviews.com

757

Review

Zaghloul & Gouda

sterilize microscopic extensions in the perivesical tissues. Only six randomized trials addressed the issue of adding pre­operative radiotherapy to RC in the English litStudy (year) Patients PT1 PT2 PT3 PT4 Nodal Ref. erature. Two of these six studies were on (n) (%) (%) (%) (%) involvement SA-BC patients [61,62] . Only one out of the six trials showed a benefit for adding SNA-BC (pure urothelial carcinoma) [46] pre­operative radiotherapy [61] . Most of the Cheng et al. (2000) 218 50 28 17 11 other five studies showed that the benefi[54] cial effect was restricted to high-stage and Stein et al. (2001) 1054 74 81/68† 47 44 35 [88] high-grade tumors, with no difference in Madersbacher et al. 507 76 62 40 49 26 (2003) early cases. Meta-analysis of these random[57] ized studies showed a corrected odds ratio Takahashi et al. (2004) 466 81 74 47 38 50 of 0.94, indicating no benefit for adding [89] Dhar et al. (2006) 385 63 19 NM 9 preoperative radiotherapy in BC [63] . [90] Ho et al. (2009) 148 77 68 65 11 37 Postoperative radiotherapy (PORT) has [91] the advantage of dealing with microscopic Manoharan et al. 432 79 60 43 17 22 cells that are easier to sterilize. It allows (2009) better identification of the group of patients SNA-BC (urothelial and nonurothelial) that may benefit from this adjuvant ther[92] Nishiyama et al. 1113 82 84/69† 59 43 35 apy. One large prospective randomized (2004) trial proved the benefit of PORT in locally [93] Niu et al. (2008) 356 73/44† 22 0 8 advanced SA-BC. The 5-year disease-free survival rate was 49 and 44% for hyper­ † [48] Gupta et al. (2008) 502 90 78 70/58 46 NM fractionated (HF) and conventional fracSA-BC (urothelial and nonurothelial) tionation (CF) PORT, respectively, com[52] pared with 25% for cystectomy-alone Ghoneim et al. (1997) 1026 73 66 47/31† 19 23 [85] patients [50] . This effect was constant across El-Mekresh et al. 185 83 41 21 all tumor cell types, all muscle-invasive (1998) [86] stages and grades in SA-BC. Similar results Khaled et al. (2005) 180 55 12 6 were replicated in a nonrandomized, pro[50] Zaghloul et al. (1992) 192 100 100/47 40 44 31 spective, controlled Radiation Therapy [94] Zaghloul et al. (2007) 216 100 51 40 30 31 Oncology Group trial on SNA-BC [64] . [53] The results of the two studies were nearly Ghoneim et al. (2008) 2720 82 75/53 40 30 27 identical when compared stage by stage † a/b. NM: Not mentioned; SA-BC: Schistosoma-associated bladder cancer; SNA-BC: Schistosoma-non-associated [40] . The only difference was that the bladder cancer. Radiation Therapy Oncology Group trial (122 patients), with a 10-year OS of 34%. It is clear from this reported a high gastrointestinal tract (GIT) late complication large-number, single-institution study that the OS rates of SCC, rate [64] . Intestinal obstruction was reported in 37% (15 out of UC and adenocarcinoma were similar and had the same profile as 40 patients) after PORT. Nine of these 15 patients required surthat of SNA-BC. They demonstrated that although the univariate gery and three died. On the contrary, Zaghloul et al. reported analysis was significant (undifferentiated carcinoma had much 5 and 18% all grades of late GIT complications for the HF and lower OS), the multivariate analysis proved that tumor cell type CF groups, respectively [50] . Only 4 and 5% of the HF and CF is not an independent working factor determining OS. Many groups, respectively, necessitated surgical interference. Similar authors cautiously concluded that RC treatment end-results low levels of late GIT complications were experienced by other were not affected by tumor histology or etiology but affected by retrospective studies reported on SA-BC and SNA-BC [65–67] . other prognostic factors such as stage, grade, nodal involvement, The differences between late GIT complications were related lympho­vascular invasion, angiogenesis, P53, P21, retinoblastoma to the difference in radiation volume in different studies and genes and other biological factors. These prognostic and predictor not to association with schistosomiasis [55] . factors were shown in many SA-BC and SNA-BC studies with In a prospective, randomized trial, El-Moneim et al. comvarying weight effects [53,55,59,60] . pared preoperative and postoperative radiotherapy in SA-BC [68] . They administered the same dose of 50 Gy in 5 weeks to Preoperative & postoperative radiotherapy both groups. The study reported both similar treatment endThe rationale of preoperative radiotherapy is to prevent intra- results and late complication rates for pre- and post-operative operative seeding of tumor cells in the operative field and to radiotherapy. Table 2. The 5-year survival of each pathological stage in schistosoma-non-associated and schistosoma-associated bladder cancer patients in large series of radical cystectomy.

758

Expert Rev. Anticancer Ther. 12(6), (2012)

Schistosomiasis & bladder cancer: similarities & differences from urothelial cancer

Neoadjuvant & adjuvant chemotherapy

Neoadjuvant and adjuvant chemotherapy have been utilized in BC, in an attempt to improve the outcomes for patients with high-risk muscle-invasive disease. Several meta-analyses of prospective, randomized trials indicated that patients who underwent neoadjuvant chemotherapy with methotrexate, vinblastine, doxorubicin and cisplatin prior to cystectomy have an approximate 5.0–6.5% survival advantage over those who underwent surgery alone [69,70] . However, some investigators continue to argue that this neoadjuvant advantage is small and chemotherapy may be better targeted to those at highest risk of relapse after surgery. Furthermore, many elderly patients or those who have comorbidities will not tolerate methotrexate, vinblastine, doxorubicin and cisplatin chemotherapy. Therefore, many investigators tried adjuvant chemotherapy in a supposedly more favorable situation. In reality, adjuvant chemotherapy yielded a modest, statistically significant improvement in survival of SNA-BC patients over cystectomy alone [71,72] . The Egyptian Bladder Cancer Cooperative Group compared neoadjuvant chemotherapy using a gemcitabine–cisplatin regimen to cystectomy alone in 109 SA-BC patients in a prospective, controlled, randomized study. The 1-year survival rate was 54% for the cystectomy-alone patients compared with 69% for the neoadjuvant chemotherapy patients [73] . However, this difference did not rank to the level of significance. Bladder preservation trimodality treatment

Since the late 1980s, many centers investigated the bladder preservation strategy as an alternative to RC. The rationale of this strategy depends on three goals: first, eradication of the local disease; second, elimination of potential micrometastasis; and third, maintenance of the best possible quality of life through organ preservation [74] . Several treatment protocols were carried out by different investigators. However, they all comprised three essential procedures, with varying timing and varying minute details. The first main procedure is maximal transurethral resection of the bladder tumor. This is to be followed by the second procedure: neoadjuvant chemotherapy or radiochemotherapy followed by cystoscopic assessment. The

Review

third procedure is either radical radiotherapy or consolidation radiochemotherapy for the complete responders. Cystoscopic assessment will segregate the complete responder (CR) for bladder-conserving management and those showing less than CR to undergo salvage cystectomy [75] . The 5-year OS rates ranged between 39 and 58%, and the 5-year survival with native bladder preservation ranged from 36 to 43% [76–80] . Sabaa et al. reported similar results for UC (SA-BC and SNA-BC) in Egypt using a trimodality treatment [81] . Complete remission was achieved in 79% of cases after initial radiochemotherapy using a gemcitabine–cisplatin regimen. The 5-year OS rate for patients with initial CR was 68%, which is comparable to the results in SNA-BC in Western countries treated with trimodality therapy. Moreover, Sabaa et al. found that the association with schistosomiasis had no significant impact on the results of therapy for the patient [81] . Expert commentary & five-year view

Schistosomiasis that prevails in many countries has a documented association with SA-BC. This disease represented a major health problem in these countries. For a long time, SA-BC was considered a completely different entity, with very few features in common with urothelial cancer. Many investigators considered the state-of-the-art recommendations for urothelial cancer not applicable for SA-BC, and vice versa. The carcinogenic, cytogenetic and epigenetic techniques are sensitive and complex procedures. Therefore extreme caution in performing and interpretation is needed when searching for common criteria that identify certain disease. Minor deviation in the technique may lead to a major defect in the results. Closer and more careful consideration of the biological, genetic, cytogenetic and histopathological characteristics of SA-BC showed great similarity to urothelial cancer in many aspects. With the continuous advancement in healthcare in schistosoma-endemic areas, the management of SA-BC followed, to a great extent, the recommendations for urothelial tumors. The reported treatment end-results for different modalities (surgery, neoadjuvant, adjuvant radiotherapy or chemotherapy and trimodality treatments, among others) showed similar results

Key issues • Muscle-invasive cancer represents 20–40% of urothelial cancer and a higher percentage of nonurothelial variants, particularly in schistosoma-associated bladder cancer (SA-BC). • Bladder cancer is characterized by its multiplicity. Some of the foci may show heterogeneous dysplastic or metaplastic differentiation and may appear as a variant histopathology. • Schistosoma infection produces inflammatory, immunological and fibrotic tissue responses that may be carcinogenic, especially in combination with other factors such as smoking, pesticides and other chemical exposure. • Exposure to Schistosoma haematobium total antigen resulted in the development of bladder cancer in experimental animal urothelium. • Although no entirely specific cytogenetic aberrations have been identified for bladder cancer, various nonrandom deletions, gains of chromosomes, polyploidization and formation of isochromosomes have been observed. These observations depend upon the applied technique. • There are no documented significant differences in clinical presentation or the natural history of SA-BC and schistosoma-non-associated bladder cancer. • The radical cystectomy, adjuvant or neoadjuvant radiotherapy or chemotherapy treatment end-results did not differ in SA-BC from that of schistosoma-non-associated bladder cancer after controlling for stage, grade and nodal involvement.

www.expert-reviews.com

759

Review

Zaghloul & Gouda

to that of urothelial cancer. Similarities between SA-BC and SNA-BC with its urothelial and nonurothelial entities are now evident, although not reaching level  1 evidence-base level. There is no direct randomized trial comparing any of the recommended treatment modalities in SA-BC and urothelial or nonurothelial SNA-BC. The coming years may allow prospective, randomized, multicentric trials comparing SA-BC and SNA-BC that will serve patients in both categories. The application of evidence-base recommendations in the treatment of BC regardless of its etiology may improve the treatment end-results, including quality of life. The availability of large References

2

3

4

5

6

7

Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J. Clin. 61, 69–90 (2011). Siegel R, Ward E, Brawkey O, Jamal A. Cancer statistics 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer death. CA Cancer J. Clin. 61, 212–236 (2011).

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.

Sandberg AA, Meloni-Ehrig AM. Cytogenetics and genetics of human cancer: methods and accomplishments. Cancer Genet. Cytogenet. 203(2), 102–126 (2010).

10

Kuper H, Adami HO, Trichopoulos D. Infections as a major preventable cause of human cancer. J. Intern. Med. 248, 171–183 (2000).

11

Fried B, Reddy A, Mayer D. Helminths in human carcinogenesis. Cancer Lett. 305(2), 239–249 (2011).

19

Tricker AR, Mostafa MH, Spiegelhalder B, Preussmann R. Urinary excretion of nitrate, nitrite and N-nitroso compounds in schistosomiasis and bilharzial bladder cancer patients. Carcinogenesis 10, 547–552 (1989).

20

WHO International Agency for Research on Cancer. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: Schistosomes, Liver Flukes and Helicobacter pylori. WHO International Agency for Research on Cancer, Lyon, France, 9–175 (1994).

12

Lopez-Beltran A, Cheng L. Histologic variants of urothelial carcinoma: differential diagnosis and clinical implications. Hum. Pathol. 37, 1371–1388 (2006).

Ross AGP, Bartly PB, Sleigh AC et al. Schistosomiasis. N. Engl. J. Med. 346(16), 1212–1220 (2002).

21

13

22

Muscheck M, Abol-Enein H, Chew K et al. Comparison of genetic changes in schistosome-related transitional and squamous bladder cancers using comparative genomic hybridization. Carcinogenesis 21, 1721–1726 (2000).

Ferguson AR. Associated bilharziasis and primary malignant disease of the urinary bladder with observations on a series of forty cases. J. Pathol. Bacteriol. 16, 76–94 (1911).

Smith JH, Christie JD. The pathobiology of Schistosoma haematobium infection in humans. Hum. Pathol. 17, 333–345 (1986).

•• First report to etiologically correlate schistosomiasis and bladder cancer (BC).

El-Bolkainy MN, Mokhtar NM, Ghonim MA, Hussein MH. The impact of schistosomiasis on the pathology of bladder carcinoma. Cancer 48, 2643–2648 (1981).

23

Abdel MM, Hassan A, El-Sewedy S. Human bladder cancer, schistosomiasis, N-nitroso compounds and their precursors. Int. J. Cancer 88, 682–683 (2000).

24

Gentile JM. Schistosome related cancers: a possible role for genotoxins. Environ. Mutagen. 7, 775–785 (1985).

25

Botelho M, Ferreia AC, Olivieira MJ, Domingues A, Machado JC, de Costa JM. Schistosoma haematobium total antigen and decreased apoptosis of normal epithelial cells. Int. J. Parasitol. 39, 1083–1091 (2009).

26

Marletta MA. Mammalian synthesis of nitrite, nitrate, nitric oxide, and N-nitrosating agents. Chem. Res. Toxicol. 1, 249–257 (1988).

Wu XR. Urothelial tumorigenesis: a tale of divergent pathway. Nat. Rev. Cancer 5, 713–725 (2005).

27

Shokeir AA. Squamous cell carcinoma of the bladder: pathology, diagnosis and treatment. BJU Int. 93, 216–220 (2004).

Botelho MC, Machado JC, deCosta JM. Schistosoma haematobium and bladder cancer. Virulence 1(2), 84–87 (2010).

•• Experimental proof of the capability of Schistosoma haematobium to act as a direct carcinogenic factor in murine urothelium.

Kallioniemi A, Kallioniemi OP, Sudar D et al. Comparative genomic hybridization for molecular cytogenetic ana­lysis of solid tumors. Science 258, 818–821 (1992). Zarzour AH, Selim M, Abd-Elsayed AA, Hameed DA, Abdelaziz MA. Muscle invasive bladder cancer in Upper Egypt: the shift in risk factors and tumor characteristics. BMC Cancer 8, 250–255 (2008). Cheng L, Davidson D, Mac Lennan GT et al. The origin of urothelial cancer. Expert Rev. Anticancer Ther. 10(6), 865–880 (2010).

•• Extensive review dealing with all theories of urothelium carcinogenesis and their evidence. 8

Financial & competing interests disclosure

9

Papers of special note have been highlighted as: •• of considerable interest 1

numbers of patients will allow the determination of a recommended risk-adapted therapy that minimizes the under- or over-treatment for such patients.

Cheng L, Zhang D. Molecular Genetic Pathology. Humana Press/Springer, NY, USA (2008).

760

14

Ministry of Health and Population, Egypt, Department of Endemic Diseases. Prevalence of Schistosomiasis in Egypt Over Time (2004).

15

Gouda I, Mokhtar N, Bilal D, El-Bolkainy T, El-Bolkainy NM. Bilharziasis and bladder cancer: a time trend ana­lysis of 9843 patients. J. Egypt. Natl Canc. Inst. 19(2), 158–162 (2007).

16

17

18

Rosin MP, Saad el Din Zaki S, Ward AJ, Anwar WA. Involvement of inflammatory reactions and elevated cell proliferation in the development of bladder cancer in schistosomiasis patients. Mutat. Res. 305, 83–92 (1994).

Expert Rev. Anticancer Ther. 12(6), (2012)

Schistosomiasis & bladder cancer: similarities & differences from urothelial cancer

28

Jacobs BL, Lee CT, Montie JE. Bladder cancer in 2010: how far have we come? CA Cancer J. Clin. 60(4), 244–272 (2010).

29

McConkey DJ, Lee S, Choi W et al. Molecular genetics of bladder cancer: emerging mechanisms of tumor initiation and progression. Urol. Oncol. 28(4), 429–440 (2010).

30

31

32

33

34

35

36

Abdel Wahab AH, Abo-Zeid HI, ElHusseini MI, Ismail M, El-Khor AM. Role of loss of heterozygosity on chromosomes 8 and 9 in the development and progression of cancer bladder. J. Egypt. Natl Canc. Inst. 17(4), 260–269 (2005). Chaudhary KS, Lu KS, Abel PD et al. Expression of BCL-2 and p53 oncoproteins in schistosomiasis-associated transitional and squamous cell carcinoma of the urinary bladder. Br. J. Urol. 79, 78–84 (1997). El-Sheikh SS, Madaan S, Alhasso A, Abel P, Stamp G, Lalani EN. Cyclooxygenase-2: a possible target in schistosoma associated bladder cancer. Br. J. Urol. 88, 921–927 (2001). Hodder SL, Mahmoud AA, Sorenson K et al. Predisposition to urinary tract epithelial metaplasia in Schistosoma haematobium infection. Am. J. Trop. Med. Hyg. 63, 133–138 (2000). Tsutsumi M, Tsai YC, Gonzalgo ML, Nichols PW, Jones PA. Early acquisition of homozygous deletions of p16/p19 during squamous cell carcinogenesis and genetic mosaicism in bladder cancer. Oncogene 17, 3021–3027 (1998). Fadl-Elmula I, Kytola S, Leithy ME et al. Chromosomal aberrations in benign and malignant bilharzia-associated bladder lesions analyzed by comparative genomic hybridization. BMC Cancer 2, 5 (2002). Armengol G, Eissa S, Lozano JJ et al. Genomic imbalances in schistosomaassociated and non schistosomaassociated bladder carcinoma: an array comparative genomic hybridization ana­lysis. Cancer Genet. Cytogenet. 177, 16–19 (2007).

•• Very important study that proves that there is no difference between schistosoma-associated BC and schistosoma-non-associated BC using pools of tissues arising from patients having the same pathology to detect recurrent genomic abnormalities. 37

Vauhkonen H, Bohling T, Eissa S, Shoman S, Knuutila S. Can bladder

www.expert-reviews.com

adenocarcinomas be distinguished from schistosomiasis associated bladder cancer by using comparative genomic hybridization ana­lysis? Cancer Genet. Cytogenet. 177, 153–157 (2007). 38

Koraitim MM, Metwalli NE, Atta MA, El-Sadr AA. Changing age incidence and pathological types of schistosomaassociated bladder carcinoma. J. Urol. 154, 1714–1716 (1995).

39

Zaghloul MS, Nouh A, Moneer M, El-Baradie M, Nazmy M, Younis A. Time-trend in epidemiological and pathological features of schistosomaassociated bladder cancer. J. Egypt. Natl Canc. Inst. 20(2), 168–174 (2008).

40

Zaghloul MS. Radiation as adjunctive therapy to cystectomy for bladder cancer. Is there a difference for bilharzial association? Int. J. Radiat. Oncol. Biol. Phys. 28, 783 (1994).

41

Zaghloul MS. Distant metastasis from bilharzial bladder cancer. Cancer 77, 743–749 (1996).

Review

A review of 420 cases of radical cystectomy. Jpn J. Clin. Oncol. 9, 117–122 (1979). 50

Zaghloul MS, Awwad HK, Omar S et al. Postoperative radiotherapy of carcinoma in bilharzial bladder. Improved disease-free survival through improving local control. Int. J. Radiat. Oncol. Biol. Phys. 22, 511–517 (1992).

•• The first prospective randomized trial that proved the beneficial effect of adding adjuvant radiotherapy to cystectomy to improve disease-free survival. 51

Zaghloul MS. Adjuvant radiation therapy for locally advanced bladder cancer. US Oncol. Dis. 2, 86–89 (2006).

52

Ghoneim MA, El-Mekresh MM, El-Baz MA, El-Attar IA, Ashamallah A. Radical cystectomy for carcinoma of the bladder: critical evaluation of the results in 1,026 cases. J. Urol. 158, 393–399 (1997).

53

Ghoneim MA, Abdel-Latif M, el-Mekresh M et al. Radical cystectomy for carcinoma of the bladder: 2,720 consecutive cases 5 years later. J. Urol. 180(1), 121–127 (2008).

42

Miller LS. Bladder cancer. Superiority of preoperative irradiation and cystectomy in clinical stages B2 and C. Cancer 39, 973–980 (1977).

43

Werf-Messing BV, Friedell GH, Menon RS, Raigopa SM. Carcinoma of the urinary bladder T3 Nx MO treated by preoperative irradiation followed by simple cystectomy. Int. J. Radiat. Oncol. Biol. Phys. 8, 1849–1855 (1982).

54

Greven KM, Spera JA, Solin LW. Local recurrence after cystectomy alone for bladder carcinoma. Cancer 69, 2767–2770 (1992).

Stein JP, Lieskovsky G, Cote R et al. Radical cystectomy in treatment of invasive bladder cancer: long-term results in 1,054 patients. J. Clin. Oncol. 19, 666–675 (2001).

55

Zaghloul MS. Adjuvant and neoadjuvant radiotherapy for bladder cancer: revisited. Future Oncol. 6(7), 1177–1191 (2010).

56

Gschwend JE, Dahm P, Fair WR. Disease specific survival as endpoint of outcome for bladder cancer patients following radical cystectomy. Eur. Urol. 41(4), 440–448 (2002).

57

Takahashi A, Tsukamoto T, Tobisu K et al. Radical cystectomy for invasive bladder cancer, results of multi-institutional pooled ana­lysis. Jpn J. Clin. Oncol. 34, 14–19 (2004).

58

Rogers CG, Palapattu GS, Shariat SF et al. Clinical outcomes following radical cystectomy for primary nontransitional cell carcinoma for the bladder compared with transitional cell carcinoma of the bladder. J. Urol. 175, 2048–2053 (2006).

59

Scosyrev E, Yao J, Messing E. Urothelial carcinoma versus squamous cell carcinoma of bladder: is survival different with stage adjustment? Urology 73, 822–827 (2009).

44

45

Visser O, Nieuwenhuijzen JA, Horenblas S. Local recurrence after cystectomy and survival of patients with bladder cancer: a population based study in greater Amsterdam. J. Urol. 174, 97–102 (2005).

46

Cheng L, Weaver AL, Leibovich BC et al. Predicting the survival of bladder carcinoma patients treated with radical cystectomy. Cancer 88, 2326–2332 (2000).

47

Volkmer BG, Kuefer R, Bartsch GC Jr et al. Oncological followup after radical cystectomy for bladder cancer – is there any benefit? J. Urol. 181(4), 1587–1593 (2009).

48

Gupta NP, Kolla SB, Seth A et al. Radical cystectomy for bladder cancer: a single center experience. Indian J. Urol. 24(1), 54–59 (2008).

49

El-Said A, Omar S, Ibrahim AS et al. Bilharzial bladder cancer in Egypt.

•• Large single-institution prospective study proving there is no effect of tumor type on survival in multivariate ana­lysis.

761

Review

Zaghloul & Gouda

•• Important study that proved that there was no difference in survival between urothelial and squamous pathology after adjustment for stage. 60

61

Ploeg M, Aben KK, Hulsbergen-van de Kaa CA et al. Clinical epidemiology of nonurothelial bladder cancer: ana­lysis of The Netherlands Cancer Registry. J. Urol. 183(3), 915–920 (2010). Awwad HK, Baki HA, El Bolkainy et al. Preoperative irradiation of T3 carcinoma in bilharzial bladder. Int. J. Radiat. Oncol. Biol. Phys. 5, 787–794 (1979).

•• Only prospective randomized trial that proved the value of adding preoperative radiotherapy to surgery in BC. 62

63

64

65

Ghoneim MA, Ashamalla AG, Awwad HK, Whitmore WF Jr. Randomized trial of cystectomy with or without preoperative radiotherapy for carcinoma of the bilharzial bladder. J. Urol. 134, 266–268 (1985). Huncharek M, Muscat J, Geschwind JF. Planned preoperative radiation therapy in muscle invasive bladder cancer. Results of metaana­lysis. Anticancer Res. 18, 1931–1934 (1998). Reisinger S, Mohiuddin M, Mulholland S. Combined pre- and post-operative adjuvant radiation therapy for bladder cancer – a ten year experience. Int. J. Radiat. Oncol. Biol. Phys. 24, 463–468 (1992). Cozzarini C, Pelegrini D, Fallini M et al. Reappraisal of the role of adjuvant radiotherapy in muscle-invasive transitional cell carcinoma of the bladder. Int. J. Radiat. Oncol. Biol. Phys. 45, 221 (1999).

66

Zaghloul MS, Mohran TZ, Saber RA, Agha N. Postoperative radiotherapy in bladder cancer. J. Egypt. Natl Cancer Inst. 14, 161–168 (2002).

67

Zaghloul MS, Nouh A, Nazmy M et al. Long-term results of primary adenocarcinoma of the urinary bladder: a report on 192 patients. Urol. Oncol. 24, 13–20 (2006).

68

69

El-Moneim HA, El-Baradie MM, Younis A, Ragab Y, Labib A, El-Attar I. A prospective randomized trial for postoperative vs. preoperative adjuvant radiotherapy for muscle-invasive bladder cancer. Urol. Oncol. doi:10.1016/j. urolonc.2011.01.008 (2011) (Epub ahead of print). Vaughn DJ, Malkowicz SB. Neoadjuvant chemotherapy in patients with invasive bladder cancer. Urol. Clin. North Am. 32, 231–237 (2005).

762

70

71

Winquist E, Kichner TS, Segal R et al. Neoadjuvant chemotherapy for transitional cell carcinoma of the bladder: a systematic review and meta-ana­lysis. J. Urol. 171, 561–569 (2004). Vale CL. Advanced bladder cancer meta-ana­lysis collaboration. Adjuvant chemotherapy for invasive bladder cancer. Cochrane Database Syst. Rev. CD006018 (2006).

organ-sparing treatment of bladder cancer. Int. J. Radiat. Oncol. Biol. Phys. 40(1), 121–127 (1998). 81

Sabaa MA, El-Gamal OM, Abo-Elenen M, Khanam A. Combined modality treatment with bladder preservation for muscle invasive bladder cancer. Urol. Oncol. 28, 14–20 (2010).

82

Bassi P, Ferrante GD, Piazza N et al. Prognostic factors of outcome after radical cystectomy for bladder cancer, a retrospective study of homogenous patient cohort. J. Urol. 161, 1494–1497 (1999).

72

Ruggeri EM, Giannarelli D, Bria E et al. Adjuvant chemotherapy in muscle-invasive bladder cancer: a pooled ana­lysis from Phase III studies. Cancer 106, 783–788 (2006).

83

73

Khaled H, Zaghloul M, Ghoneim M et al. Gemcitabine and cisplatin as neoadjuvant chemotherapy for invasive bladder cancer: effect on bladder preservation and survival. Proc. Am. Soc. Clin. Oncol. 22, 411 (2003) (Abstract 1652).

Shariat SF, Karakiewicz PI, Palapattu GS. Outcomes of radical cystectomy for transitional cell carcinoma of the bladder: a contemporary series from the Bladder Cancer Research Consortium. J. Urol. 176(6), 2414–2422 (2006).

84

74

Rödel C. Current status of radiation therapy and combined-modality treatment for bladder cancer. Strahlenther Onkol. 180(11), 701–709 (2004).

75

Zaghloul MS, Mousa AG. Trimodality treatment for bladder cancer: does modern radiotherapy improve the end results? Expert Rev. Anticancer Ther. 10(12), 1933–1944 (2010).

Lughezzani G, Sun M, Jeldres C et al. Adenocarcinoma versus urothelial carcinoma of the urinary bladder: comparison between pathologic stage at radical cystectomy and cancerspecific mortality. Urology 75(2), 376–381 (2010).

85

Kachnic LA, Kaufman DS, Heney NM et al. Bladder preservation by combined modality therapy for invasive bladder cancer. J. Clin. Oncol. 15(3), 1022–1029 (1997).

El-Mekresh MM, el-Baz MA, Abol-Enein H, Ghoneim MA. Primary adenocarcinoma of the urinary bladder: a report of 185 cases. Br. J. Urol. 82(2), 206–212 (1998).

86

Khaled H, El-Hattab O, Moneim DA, Kassem HA, Morsi A, Sherif G. A prognostic index (bladder prognostic index) for bilharzial-related invasive bladder cancer. Urol. Oncol. 23, 254–260 (2005).

87

Ali-El-Dein B. Oncological outcome after radical cystectomy and orthotopic bladder substitution in women. Eur. J. Surg. Oncol. 35(3), 320–325 (2009).

88

Madersbacher S, Hochreiter W, Burkhard F et al. Radical cystectomy for bladder cancer today: a homogeneous series without neoadjuvant therapy. J. Clin. Oncol. 21, 690–669 (2003).

89

Tester W, Porter A, Asbell S et al. Combined modality program with possible organ preservation for invasive bladder carcinoma: results of RTOG protocol 85-12. Int. J. Radiat. Oncol. Biol. Phys. 25(5), 783–790 (1993).

Dhar NB, Campbell SC, Zippe CD et al. Outcomes in patients with urothelial carcinoma of the bladder with limited pelvic lymph node dissection. BJU Int. 98(6), 1172–1175 (2006).

90

Sauer R, Birkenhake S, Kühn R, Wittekind C, Schrott KM, Martus P. Efficacy of radiochemotherapy with platin derivatives compared with radiotherapy alone in

Ho CH, Huang CY, Lin WC et al. Radical cystectomy in the treatment of bladder cancer: oncological outcome and survival predictors. J. Formos. Med. Assoc. 108(11), 872–878 (2009).

91

Manoharan M, Ayyathurai R, Soloway MS. Radical cystectomy for urothelial

76

77

78

79

80

Shipley WU, Winter KA, Kaufman DS et al. Phase III trial of neoadjuvant chemotherapy in patients with invasive bladder cancer treated with selective bladder preservation by combined radiation therapy and chemotherapy: initial results of Radiation Therapy Oncology Group 89-03. J. Clin. Oncol. 16(11), 3576–3583 (1998). Arias F, Domínguez MA, Martínez E et al. Chemoradiotherapy for muscle invading bladder carcinoma. Final report of a single institutional organ-sparing program. Int. J. Radiat. Oncol. Biol. Phys. 47(2), 373–378 (2000).

Expert Rev. Anticancer Ther. 12(6), (2012)

Schistosomiasis & bladder cancer: similarities & differences from urothelial cancer

including 1131 patients treated during 1990–2000 in Japan. Eur. Urol. 45, 176–181 (2004).

carcinoma of the bladder: an ana­lysis of perioperative and survival outcome. BJU Int. 104(9), 1227–1232 (2009). 92

Nishiyama H, Habuchi T, Watanabe J et al. Clinical outcome of a large-scale multi-institutional retrospective study for locally advanced bladder cancer: a survey

www.expert-reviews.com

93

Niu HT, Xu T, Zhang YB et al. Outcomes for a large series of radical cystectomies for bladder cancer. Eur. J. Surg. Oncol. 34(8), 911–915 (2008).

94

Review

Zaghloul MS, El Baradie Nouh MA, Abdel-Fatah S, Taher A, Shalaan M. Prognostic index for primary adenocarcinoma of the urinary bladder. Gulf J. Oncol. 1(2), 47–54 (2007).

763