Skip to main content

Bladder cancer: therapeutic challenges and role of 3D cell culture systems in the screening of novel cancer therapeutics

Abstract

Bladder cancer (BC) is the sixth most common worldwide urologic malignancy associated with elevated morbidity and mortality rates if not well treated. The muscle-invasive form of BC develops in about 25% of patients. Moreover, according to estimates, 50% of patients with invasive BC experience fatal metastatic relapses. Currently, resistance to drug-based therapy is the major tumble to BC treatment. The three-dimensional (3D) cell cultures are clearly more relevant not only as a novel evolving gadget in drug screening but also as a bearable therapeutic for different diseases. In this review, various subtypes of BC and mechanisms of drug resistance to the commonly used anticancer therapies are discussed. We also summarize the key lineaments of the latest cell-based assays utilizing 3D cell culture systems and their impact on understanding the pathophysiology of BC. Such knowledge could ultimately help to address the most efficient BC treatment.

Introduction

Bladder cancer (BC) is among the top 10 cancers affecting men [1]. The incidence of BC is higher in men than in women [2, 3]. About 25% of BC cases proceed to the muscle-invasive form. Lethal metastatic relapses are estimated to occur in 50% of patients with invasive malignancies [4]. The rates of metastatic BC increase with age and are seen most frequently in elderly people [5]. The risk factors for BC include smoking, inflammation, exposure to carcinogens, and schistosomiasis [6]. BC typically starts inside the bladder epithelium and travels from there to muscles and other tissues [7]. Over 90% of urothelial carcinomas were reported to originate in the urinary bladder [8]. The most common cause of death in advanced BC patients is associated with metastasis of BC [9]. Hematuria represents the most characteristic symptom of BC and is usually diagnosed by physical inspection of the urine [10]. However, patients can also present with isolated microscopic hematuria which is often detected during routine cystoscopic examinations [11].

Cell culture is an important technique for maintaining cells outside the body. Under optimal conditions, the appearance of cultured cells or organoids can be used to reflect their in vivo behavior. The alignment of cells in the culture vessel significantly affects their structure, and functions, as well as their response to the tested chemical compounds [12]. Three-dimensional (3D) models of cell culture have emerged and are currently representing a useful platform for monitoring cellular organization via comprehensive visualization of the cultured cells [13]. Since the function and morphology of individual cells substantially rely on their interactions with proteins and signaling factors from neighboring cells and their surrounding extracellular matrix, the efforts for culturing cells in 3D systems have progressively evolved [14]. Culturing cells in 3D systems has been suggested to recover some of their natural characteristics that are usually affected during their culture in conventional, two-dimensional (2D), culture systems [15]. Recovering the cellular characteristics of cultured cancer cells could help for increasing both the specificity and sensitivity of cell-based assays used for determining their identities and also for addressing and selecting new drugs for cancer treatment [16].

Classification of bladder cancer (BC)

Understanding the classification of BC is important to establish an appropriate treatment strategy. The World Health Organization (WHO) initially classified BC into three grades: well-differentiated (grade 1 or G1), moderately differentiated (grade 2 or G2), and poorly differentiated (grade 3 or G3) papillary urothelial carcinoma (PUC) [17]. The latter-mentioned grade is the highest grade, in which the poorly differentiated cancer cells grow at faster rates and start to spread to other organs including the regional lymph nodes. Urothelial carcinoma, squamous cell carcinoma, and adenocarcinoma are the three most common microscopic subtypes of BC [7]. Based on the progressive and invasive nature of the continuously proliferating tumorous cells, BC is classified into two main stages: non-muscle invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC) [18, 19]. NMIBC represents roughly 70–85% of BC cases in which patients have tumors restricted to the mucosa-submucosa layers, hence it is superficial BC [20]. It is further classified into Tis, Ta, and T1. In Tis and Ta, cancer is restricted to the urothelial layer, while in T1 it reaches to the underlying connective tissue layer. Similarly, MIBC is further categorized into the degree of cancer invasion into T2 (muscle layer), T3 (perivesical fat and lymph nodes), and T4 (other organs). Various grades and stages of BC are illustrated in Fig. 1.

Fig. 1
figure 1

Grades and stages of bladder cancer

Strategies for treatment and management of bladder cancer (BC)

Proper BC diagnosis is essential for selecting a specific treatment. Urine cytology and cystoscopy are the most widely significant tools for BC diagnosis and follow-up [20]. Although cystoscopy remains as an essential investigative gadget in the disclosure and monitoring of BC, small papillary tumors or carcinoma in situ can be easily omitted by standard white-light cystoscopy (SWLC), which may purpose for early recurrence of the disease. This leads to the development of novel diagnostic technologies such as narrow-band imaging cystoscopy and photodynamic technology [21]. Various molecular urinary tests have been marketed over the years to help in the detection of BC. Although initially hopeful, none of the different technologies has been enough specific or sensitive to prohibit cystoscopic surveillance [22, 23].

Neoteric advances in BC management are increasing. These advances include the use of cystoscopic and fluoroscopic revelation, neoadjuvant chemotherapy, bladder-sparing multimodal therapy, and intravesical therapy [24]. As mentioned earlier, most BC cases do not encompass the bladder muscular wall and are commonly handled with telescopic removal of cancer (transurethral resection of bladder tumor), followed by infiltration of vaccine-based therapy or chemotherapy into the bladder [25].

Resistance of bladder cancer (BC) to chemotherapy

Chemo-resistance is one of the main problems in treatment of various types of cancer as cancer cells become resistant to chemotherapeutic agents [26]. Recurrence of cancer is a serious trouble in patients with BC with increased proliferation rates of drug-resistant cells [27]. The persistence of cancer cells` resistance to chemotherapeutics is a major stumbling to BC treatment [28]. Among the causes that BC is so deadly is its tendency to develop drug resistance typically used as frontline therapies [29]. Some cancers are considered resistant to therapy, either innate drug resistance at the time of drug exposure or acquired drug resistance after an initial response [30]. Although BC is a chemotherapy-sensitive malignancy, nearly most of patients promote disease progression after an initial chemotherapeutic response [31].

Radiotherapy has been suggested as a promising technique for control of muscle-invasive form of BC [32, 33]. Cryotherapy -also called cryoablation or cryosurgery- involves heat extraction from cancer cells via application of extreme cold (< 0° C) which will eventually lead to death of the cancerous cells [34]. Percutaneous cryotherapy revealed a decline in the incidence of complications associated with BC including hematuria and urinary irritations [35].

Transurethral resection of bladder cancer (TURB) is frequently used for management of non-muscle invasive BC with about 50% rate 5-year overall survival [36, 37]. However, this rate declines to 20% by 15 years following tumor resection [36].

The use of multimodal techniques showed better outcomes for treatment of BC than single intervention [38]. For instance, decreased cancer survival was seen in BC patients subjected to cryoablation combined with the chemotherapeutic agent cisplatin compared to those who received cisplatin only [39]. In addition, patients who underwent TURB followed by radiochemotherapy displayed a higher rate of BC remission and longer overall survival than those who received radiotherapy alone [40].

Drug resistance mechanisms in bladder cancer (BC)

Drug resistance in BC comprises numerous mechanisms, such as avoidance of apoptosis by cancer cells via DNA methylation-induced transcriptional repression of genes participating in the apoptotic pathway [27]. Additionally, activation of these genes by epigenetic therapy might expedite the reconquest of chemotherapeutic agents’ sensitivity in BC and could lead to novel therapeutic approaches in BC [31]. Drayton and Catto [41] stated that the mechanisms of drug resistance could be classified into these act to weaken the normal cellular response to drug-induced DNA damage and those act to reduce drug bioavailability within a cell. Reduced influx, increased drug efflux, increased DNA repair, and tolerance to DNA damage appear to be the preponderant mechanisms of drug resistance [28]. The mechanisms related to radio- and chemo-resistance influence many pathways as those involved in DNA damage repair, drug absorption and efflux, cell cycle, and apoptosis [29]. The use of phytochemicals has shown promising effects in mitigation of drug resistance of cell lines and animal models of BC [42, 43], though their implementation in clinical protocols remains under deep investigation [44]. The possible mechanisms of drug resistance in BC are summarized in Fig. 2.

Fig. 2
figure 2

Possible mechanisms of drug resistance in bladder cancer. The diagram was created with BioRender [45]

Drug Resistance regulation by bladder cancer (BC) stem cells

Cancer stem cells (CSCs), also known as tumor-initiating cells (TICs), have been intensively studied in the past decade, with a focus on their origin, possible sources, cellular markers, survival mechanisms, and development of therapeutic strategies targeting them [46, 47]. CSCs have been proposed to play a major role in tumorigenesis, drug resistance, metastasis, and cancer relapse, because of their ability for self-renewal [48]. The subpopulation of CSCs that remains in tumor tissue following chemotherapy is responsible for survival and expansion of tumor cells during recurrence [49, 50].

CSCs can be renitent to DNA damage-induced cell death through different ways. These ways include protection against oxidative DNA damage by enhanced ROS scavenging, promotion of the DNA repair capability through ATM and CHK1/CHK2 phosphorylation, or activation of the anti-apoptotic signaling pathways, such as PI3K/Akt, WNT/b-catenin, and Notch signaling pathways [47, 50] (Fig. 3). For instance, CD44 interacts with a glutamate-cystine transporter and controls the intracellular level of reduced glutathione; hence, the CSCs expressing a high level of CD44 showed an enhanced capacity for GSH synthesis, resulting in stronger defense against ROS [51].

It is widely supposed that CSCs may emerge from normal stem cells that have sustained gene mutations [52]. CSCs can also be constructed from differentiated or progenitor cells that undergo de-differentiation or tumor cells that acquire stem cell properties [53, 54]. It has been noted that BC stem cells (BCSCs) originated from CSCs or from BC non-stem cells (BCNSCs) with clonal identity [55, 56] (Fig. 3).

Fig. 3
figure 3

Different sources of cancer stem cells (CSCs) and their possible role in tumorigenesis and inhibition of DNA damage-induced cell death. The diagram was created with BioRender [45]

Several common markers of BCSCs, including CD44+, BCMab1+, EMA-, and 67LR+, are expressed in the basal cell layer of BC mass that leading to more debates regarding the exporter of BCSCs [57, 58]. Theoretically, if all markers are from a specific cell type in BC, it is supposed that BCSCs may have arisen from mutated normal stem cells. On the other hand, if the markers are expressed on different normal cell types, then the BCSCs may be derived from differentiated or progenitor cells that acquired de-differentiation characteristics due to mutations, thus leading to different BCSCs subgroups [59].

Many types of cancers are associated with autocrine signaling of different cytokines that are capable of activating receptors largely falling under the receptor of tyrosine kinase family [60, 61]. Vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and stem cell factor (SCF) are a few important autocrine players that could mediate the maintenance of BCSCs since their receptors were exposed in BC cell lines [62].

Vascular endothelial growth factor receptor 2 (VEGFR2) is implicated in CSCs that can trigger blebbishield emergency program-mediated sphere formation in RT4 (transitional cell papilloma) bladder cancer cells [63]. cMET and cKit receptors were co-downregulated along with VEGFR2 during blebbishield formation, indicating that these receptors might be activated along with VEGFR2 since receptors usually undergo downregulation after activation [63, 64]. VEGFR2 is usually detected in endothelial cells, and endothelial cells are known to create stem cell niches implicating endothelial cells in tumors as one of the culprits that could transform CCs into CSCs [65]. The presence of cMET, VEGFR2, and cKIT also might help bladder cancer stem cells to mediate metastasis [62].

Three-dimensional (3D) cell culture system for studying drug resistance in bladder cancer (BC)

3D cell culture systems are becoming incrementally popular in contemporaneous cancer studies, tissue engineering, drug discovery, and drug resistance research because of their obvious advantages in providing more predictive data for in vivo tests and more physiologically relevant information [66, 67].

The conventional 2D culture systems involve culturing the targeted cells in a monolayered structure either inside a flat petri dish or a culture flask [68]. The main advantages of these culture systems include low-cost and simple maintenance of the cell culture. On the other hand, 2D culture systems have several disadvantages. The most important disadvantage is that the 2D cultured cells do not mimic the natural morphology and behavior of normal tumor cells. Another disadvantage of the 2D culture systems involves the improper interactions between cultured cells as well as between the cells and their surrounding matrix. Lack of such interactions is suggested to adversely affect the growth, vitality, and differentiation of cancer cells. The latter effects have been linked to abnormal gene expression and lowered drug metabolism and responsiveness [69,70,71]. Indeed, altered morphology and mode of division of cancer cells were noted following their transfer from original tissues into 2D cultures [72, 73]. These alterations in cancer cell morphology affect their secretory and signaling activities [74,75,76,77]. The lack of sufficient contact between cells and extracellular matrix is associated with loss of cell polarity [78]. The latter modifies the cell response to damaging stimuli including apoptosis and other associated phenomena [79, 80].

Another important disadvantage of 2D culture systems is that the monolayered cells have great access to the culture medium that consists principally of essential nutrients, metabolites, and oxygen. Due to the natural architecture of the tumor cells within the solid tumor mass, the in vivo cancer cells display variable access to oxygen and nutrients [69]. Importantly, the 2D cultures allow the study of cell type only [81], this leads to marked lack of data about tumor microenvironment, which is required in vivo by cancer-initiating cells [82, 83]. Those disadvantages of 2D systems drove scientists and investigators to find alternative culture models able to mimic the natural structure and morphology of tumor cells.

In 3D culture systems, striking parallels between the morphology and behavior of cells expanding in a tumor mass and cells cultivated in a 3D environment have been thoroughly characterized and verified [69, 84]. The idea of 3D spheres is based on the construction of multilayered spheroid structures: the physical and metabolic characteristics of a solid tumor mass are thus mimicked. Around 40 tumor cell lines were morphologically analyzed and cultured in 3D spheroid conditions. These cell lines came from glioblastoma, astrocytoma, Wilms’ tumor, neuroblastoma, head and neck squamous cell carcinoma, melanoma, lung, breast, colon, prostate, ovarian, hepatocellular, and pancreatic cancers. Based on the architecture of spheroids, three distinct groups were identified: (1) tight spheroids, (2) compact aggregates, and (3) loose aggregates [85, 86].

The 3D models provide appropriate cell-cell and cell-environment interactions, which were built in order to get an imitation of tissue structure. As occurs in vivo [87, 88], cells can be stimulated by their immediate surroundings. Additionally, in 3D cultures, the morphology and polarity of the cells are well-preserved and can be changed back to those of cells that were previously cultivated in 2D [89]. Similarities between 3D culture and cells growing in vivo in terms of cellular topology, gene expression, signaling, and metabolism are another significant feature [90,91,92,93,94,95]. Tumor drug resistance appears to be significantly influenced by interactions between cells and the extracellular matrix (ECM). A good technique to replicate the organic structure of a tumor mass is to employ synthetic ECM [96]. In this regard, the use of 3D systems could prevent the over- or underestimation of a particular medicine in the case of drug sensitivity and resistance, as well as its dosage [87, 97].

A negative side of 3D culturing is that single cells must be removed from the spheroid by proteolytic breakdown of single layers, which can take up to a few days [98]. Moreover, data repeatability and worker comfort are frequently more challenging in 3D approaches than in 2D systems [99]. The fact that “spheres” can be constructed from a few cell clusters rather than a single cell is frequently cited as a drawback of 3D structures. However, even structures made from a collection of cells retain a three-dimensional form more accurately than adherent, flat cultures [100].

Tumor masses are made up of tumor cells with a variety of phenotypes rather than being a homogeneous structure. In addition, several cell phenotypes are combined in 2D cultures as well. However, by cultivating a single cell with a single genetic background in a concentrated culture medium, such as soft agar or Matrigel, a homogeneous structure can be produced [101]. Vinci and colleagues’ description of a three-dimensional spheroid-based functional assay for cancer target validation and medication evaluation provided a solution to the problem of low reproducibility in 3D culture. Each well on the 96-well ultra-low attachment plates contained a single spheroid. The resultant spheroids’ sizes were consistent and had a Gaussian (normal) distribution [85, 86].

Detailed protocols for generation of 3D organoids from murine and human BC cells are currently available [102, 103]. The principal steps in generation of these 3D cancer models are simplified in Fig. 4.

Fig. 4
figure 4

Summary of the main steps used for the preparation of three-dimensional (3D) culture models of cancer cells. (A) Dissociated cells are collected from primary tumors or cancer cell lines. (B) Obtained cancer cells are then incubated with an appropriate extracellular matrix, e.g., collagen, to ensure their 3D orientation. (C) A 3D organoid is formed. The diagram was created with BioRender [45]

Due to the numerous issues associated with 2D systems, 3D models would seem to be an excellent substitute that might serve as a bridge between 2D and animal studies [104, 105]. There are benefits and drawbacks to the various technical methods for creating 3D models. The choice of 3D system to be used relies mostly on the type of research being conducted. The fact that using the incorrect model can affect the outcomes must be underlined. It is evident that there is no perfect 3D model. It may be sufficient to employ a 2D culture system in some circumstances, but as automation and costs are reduced, the high cost of the technique and the relative scarcity of the available literature remain as two major obstacles facing the widespread use of 3D culture system in BC modeling (Fig. 5). 3D culture models are believed to be used more frequently in the future.

Fig. 5
figure 5

Major differences between two-dimensional (2D) and three-dimensional (3D) culture systems of cancer cells. (A) Alignments of cells within the culture vessel. (B) Tabulated summary of the advantages and disadvantages of each system. ECM, extracellular matrix

As CSCs are critical for tumorigenesis, metastasis, tumor growth, and recurrence, there is a need to set up a 3D culture system analogous to the conditions of in vivo tumorigenesis [66]. Concerning metastasis and drug resistance, 3D culture models should better enhance the growth of TICs to mimic the actual tumorigenic processes, creating such appropriate conditions will result in the existence of molecular events relevant to metastasis [106].

3D CSCs culture models may also allow for a better assessment of cellular morphology and cellular proliferation rates. CSCs segregated from several types of in vivo tumors that exhibited prevalent lineaments: relative quiescent state, self-renewal capacity, and mutual tumor–stem‐cell morphological properties; their predilection to develop in a spheroid‐like manner [107]. Growing CSCs in 3D cell culture prompts cell proliferation more rapidly at the periphery of the spheroid because of the lack of cellular molecules adherence. Conversely, this is caused by the loss of CSCs polarity during the process of epithelial-mesenchymal transition, an important step for initiation of cancer metastasis [108, 109]. In addition, stem cell markers percentage is generally high in 3D cell culture models [110].

Selection of the most appropriate cancer therapeutic agent requires thorough in vitro analysis and validation before transitioning to clinical trials. 3D CSCs models mimic tumor microenvironments better, elucidate a more factual drug response, exhibit more adequate proliferation rates with more representative cellular morphology, facilitate the formation of ECM and stimulate high expression of ‘stemness-related’ genes [66].

Regarding culturing 3D tumorspheres, there are two main techniques: scaffold-free techniques, e.g., the hanging drop and suspension method, and scaffold-based techniques, e.g., scaffolds and hydrogels [111]. Both approaches allow for biochemical communication between TICs and the ECM; this interaction is critical for recreating the tumor-tissue microenvironment (TTM) observed in vivo [111, 112]. Amaral et al. recorded that the forced floating method using ULA 96-well round-bottomed plates was considered more reliable to create RT4 spheroids for drug screening/cytotoxicity assays than the hanging drop method [113].

Studies involving the use of 3D organoids in modeling of BC are progressively increasing. Advantageous effects of the 3D BC culture systems over the traditional BC culture systems include an increased rate of cancer cell proliferation and survival and enhanced sensitivity to chemotherapeutic agents. Summarized findings of studies utilized the 3D culture systems for BC modeling [114,115,116,117,118,119,120,121,122] are listed in Table 1.

Table 1 Summary of studies used three-dimensional (3D) culture models in bladder cancer

Concluding remarks and future perspectives

The present article discussed different types of BC, current practices for its management, and importance of 3D culture systems for screening and evaluation of new cancer therapeutics. Development of appropriate 3D culture models mimicking the in vivo tumorigenesis microenvironment will enable to better addressing of the key steps during cancer formation, growth, and metastasis. Although being more advantageous than 2D culture systems in terms of cell-cell contact and survival, the applicability of 3D culture models is challenged by the fewer number of published studies as well as the cost and complexity of culture conditions. Future studies are still required to overcome these challenges. These studies will definitely help to effectively screen a large number of drugs to be used for the treatment of BC.

Data Availability

All data are available from the corresponding author upon request.

References

  1. Richters A, Aben KKH, Kiemeney L. The global burden of urinary bladder cancer: an update. World J Urol. 2020;38(8):1895–904.

    PubMed  Google Scholar 

  2. Zhang Y. Understanding the gender disparity in bladder cancer risk: the impact of sex hormones and liver on bladder susceptibility to carcinogens. J Environ Sci Health Part C Environ Carcinog Ecotoxicol Reviews. 2013;31(4):287–304.

    CAS  Google Scholar 

  3. Huang CY, Wang SC, Chan L, Hsieh TY, Sung WW, Chen SL. Gender differences in trends of bladder cancer mortality-to-incidence ratios according to health expenditure in 55 countries. PLoS ONE. 2021;16(2):e0244510.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. DeGeorge KC, Holt HR, Hodges SC. Bladder Cancer: diagnosis and treatment. Am Fam Physician. 2017;96(8):507–14.

    PubMed  Google Scholar 

  5. Gunlusoy B, Ceylan Y, Degirmenci T, Aydogdu O, Bozkurt IH, Yonguc T, et al. The potential effect of age on the natural behavior of bladder cancer: does urothelial cell carcinoma progress differently in various age groups? Kaohsiung J Med Sci. 2016;32(5):261–6.

    PubMed  Google Scholar 

  6. Farling KB. Bladder cancer: risk factors, diagnosis, and management. The Nurse Practitioner. 2017;42(3):26–33.

    PubMed  Google Scholar 

  7. Sanli O, Dobruch J, Knowles MA, Burger M, Alemozaffar M, Nielsen ME, et al. Bladder cancer. Nat Reviews Disease Primers. 2017;3:17022.

    PubMed  Google Scholar 

  8. Flaig TW, Spiess PE, Agarwal N, Bangs R, Boorjian SA, Buyyounouski MK, et al. Bladder Cancer, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Cancer Network: JNCCN. 2020;18(3):329–54.

    PubMed  Google Scholar 

  9. Mossanen M. The epidemiology of bladder Cancer. Hematol Oncol Clin N Am. 2021;35(3):445–55.

    Google Scholar 

  10. Lenis AT, Lec PM, Chamie K. Bladder cancer: a review. JAMA. 2020;324(19):1980–91.

    CAS  PubMed  Google Scholar 

  11. Gonzalez AN, Lipsky MJ, Li G, Rutman MP, Cooper KL, Weiner DM, et al. The prevalence of bladder cancer during cystoscopy for asymptomatic microscopic hematuria. Urology. 2019;126:34–8.

    PubMed  Google Scholar 

  12. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19(11):1423–37.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Chaicharoenaudomrung N, Kunhorm P, Noisa P. Three-dimensional cell culture systems as an in vitro platform for cancer and stem cell modeling. World J stem Cells. 2019;11(12):1065.

    PubMed  PubMed Central  Google Scholar 

  14. Abdollahi S. Extracellular vesicles from organoids and 3D culture systems. Biotechnol Bioeng. 2021;118(3):1029–49.

    CAS  PubMed  Google Scholar 

  15. Jensen C, Teng Y. Is it Time to start transitioning from 2D to 3D cell culture? Front Mol Biosci. 2020;7:33.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Pucci C, Martinelli C, Ciofani G. Innovative approaches for cancer treatment: current perspectives and new challenges. Ecancermedicalscience. 2019;13:961.

    PubMed  PubMed Central  Google Scholar 

  17. Epstein JI, Amin MB, Reuter VR, Mostofi FK. The World Health Organization/International Society of Urological Pathology consensus classification of urothelial (transitional cell) neoplasms of the urinary bladder. Bladder Consensus Conference Committee. The American journal of surgical pathology. 1998;22(12):1435-48.

  18. Humphrey PA, Moch H, Cubilla AL, Ulbright TM, Reuter VE. The 2016 WHO classification of Tumours of the urinary system and male genital Organs-Part B: prostate and bladder tumours. Eur Urol. 2016;70(1):106–19.

    PubMed  Google Scholar 

  19. Zhu S, Yu W, Yang X, Wu C, Cheng F. Traditional classification and novel subtyping systems for bladder cancer. Front Oncol. 2020;10:102.

    PubMed  PubMed Central  Google Scholar 

  20. Cheung G, Sahai A, Billia M, Dasgupta P, Khan MS. Recent advances in the diagnosis and treatment of bladder cancer. BMC Med. 2013;11:13.

    PubMed  PubMed Central  Google Scholar 

  21. Jocham D, Stepp H, Waidelich R. Photodynamic diagnosis in urology: state-of-the-art. Eur Urol. 2008;53(6):1138–48.

    CAS  PubMed  Google Scholar 

  22. Yutkin V, Nisman B, Pode D. Can urinary biomarkers replace cystoscopic examination in bladder cancer surveillance? Expert Rev Anticancer Ther. 2010;10(6):787–90.

    CAS  PubMed  Google Scholar 

  23. Dreyer T, Ernst A, Jensen JB. Optimal intervals for follow-up cystoscopy in non-muscle invasive bladder cancer: a systematic review regarding oncological safety. Scandinavian J Urol. 2022;56(1):39–46.

    Google Scholar 

  24. Racioppi M. Advances in management of bladder Cancer. J Clin Med. 2021;11(1).

  25. Woldu SL, Bagrodia A, Lotan Y. Guideline of guidelines: non-muscle-invasive bladder cancer. BJU Int. 2017;119(3):371–80.

    PubMed  PubMed Central  Google Scholar 

  26. Trédan O, Galmarini CM, Patel K, Tannock IF. Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst. 2007;99(19):1441–54.

    PubMed  Google Scholar 

  27. Roh YG, Mun MH, Jeong MS, Kim WT, Lee SR, Chung JW, et al. Drug resistance of bladder cancer cells through activation of ABCG2 by FOXM1. BMB Rep. 2018;51(2):98–103.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Massari F, Santoni M, Ciccarese C, Brunelli M, Conti A, Santini D, et al. Emerging concepts on drug resistance in bladder cancer: implications for future strategies. Crit Rev Oncol/Hematol. 2015;96(1):81–90.

    PubMed  Google Scholar 

  29. Mari A, D’Andrea D, Abufaraj M, Foerster B, Kimura S, Shariat SF. Genetic determinants for chemo- and radiotherapy resistance in bladder cancer. Translational Androl Urol. 2017;6(6):1081–9.

    Google Scholar 

  30. Elbadawy M, Usui T, Yamawaki H, Sasaki K. Development of an experimental model for analyzing Drug Resistance in Colorectal Cancer. Cancers (Basel). 2018;10(6):164.

    PubMed  Google Scholar 

  31. Ramachandran K, Gordian E, Singal R. 5-azacytidine reverses drug resistance in bladder cancer cells. Anticancer Res. 2011;31(11):3757–66.

    CAS  PubMed  Google Scholar 

  32. Zhang S, Yu YH, Zhang Y, Qu W, Li J. Radiotherapy in muscle-invasive bladder cancer: the latest research progress and clinical application. Am J cancer Res. 2015;5(2):854–68.

    PubMed  PubMed Central  Google Scholar 

  33. Janopaul-Naylor JR, Zhong J, Liu Y, Zhang C, Osunkoya AO, Joshi SS, et al. Bladder preserving chemoradiotherapy compared to surgery for variants of urothelial carcinoma and other tumors types involving the bladder: an analysis of the National Cancer Database. Clin Translational Radiation Oncol. 2021;26:30–4.

    CAS  Google Scholar 

  34. Erinjeri JP, Clark TW. Cryoablation: mechanism of action and devices. J Vascular Interventional Radiology: JVIR. 2010;21(8 Suppl):187–91.

    Google Scholar 

  35. Liang Z, Fei Y, Lizhi N, Jianying Z, Zhikai Z, Jibing C, et al. Percutaneous cryotherapy for metastatic bladder cancer: experience with 23 patients. Cryobiology. 2014;68(1):79–83.

    PubMed  Google Scholar 

  36. Krause FS, Walter B, Ott OJ, Häberle L, Weiss C, Rödel C, et al. 15-year survival rates after transurethral resection and radiochemotherapy or radiation in bladder cancer treatment. Anticancer Res. 2011;31(3):985–90.

    PubMed  Google Scholar 

  37. Oswald D, Pallauf M, Herrmann TRW, Netsch C, Becker B, Lehrich K, et al. [Transurethral resection of bladder tumors (TURBT)]. Der Urologe Ausg A. 2022;61(1):71–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Marta GN, Hanna SA, Gadia R, Correa SF, Silva JL, Carvalho Hde A. The role of radiotherapy in urinary bladder cancer: current status. Int Braz J Urol. 2012;38(2):144–53. discussion 53 – 4.

    PubMed  Google Scholar 

  39. Santucci KL, Baust JM, Snyder KK, Van Buskirk RG, Katz A, Corcoran A et al. Investigation of bladder Cancer cell response to Cryoablation and Adjunctive Cisplatin based Cryo/Chemotherapy. Clinical research (Milpitas, Calif). 2020;6(1).

  40. Rödel C, Dunst J, Grabenbauer GG, Kühn R, Papadopoulos T, Schrott KM et al. Radiotherapy is an effective treatment for high-risk T1-bladder cancer. Strahlentherapie und Onkologie: Organ der Deutschen Rontgengesellschaft []. 2001;177(2):82–8; discussion 9.

  41. Drayton RM, Catto JW. Molecular mechanisms of cisplatin resistance in bladder cancer. Expert Rev Anticancer Ther. 2012;12(2):271–81.

    CAS  PubMed  Google Scholar 

  42. Cho CJ, Yu CP, Wu CL, Ho JY, Yang CW, Yu DS. Decreased drug resistance of bladder cancer using phytochemicals treatment. Kaohsiung J Med Sci. 2021;37(2):128–35.

    CAS  PubMed  Google Scholar 

  43. Cho CJ, Yang CW, Wu CL, Ho JY, Yu CP, Wu ST, et al. The modulation study of multiple drug resistance in bladder cancer by curcumin and resveratrol. Oncol Lett. 2019;18(6):6869–76.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Choudhari AS, Mandave PC, Deshpande M, Ranjekar P, Prakash O. Phytochemicals in Cancer Treatment: from preclinical studies to clinical practice. Front Pharmacol. 2019;10:1614.

    CAS  PubMed  Google Scholar 

  45. BioRender. Available online: https://biorender.com/ accessed on 29 July 2022 [.

  46. Valent P, Bonnet D, De Maria R, Lapidot T, Copland M, Melo JV, et al. Cancer stem cell definitions and terminology: the devil is in the details. Nat Rev Cancer. 2012;12(11):767–75.

    CAS  PubMed  Google Scholar 

  47. Peitzsch C, Kurth I, Kunz-Schughart L, Baumann M, Dubrovska A. Discovery of the cancer stem cell related determinants of radioresistance. Radiotherapy and Oncology: Journal of the European Society for Therapeutic Radiology and Oncology. 2013;108(3):378–87.

    PubMed  Google Scholar 

  48. Phi LTH, Sari IN, Yang YG, Lee SH, Jun N, Kim KS, et al. Cancer Stem cells (CSCs) in Drug Resistance and their therapeutic implications in Cancer Treatment. Stem Cells International. 2018;2018:5416923.

    PubMed  PubMed Central  Google Scholar 

  49. Shibata M, Hoque MO. Targeting Cancer Stem cells: a strategy for effective eradication of Cancer. Cancers. 2019;11(5).

  50. Xia P, Liu DH, Xu ZJ, Ren F. Cancer stem cell markers for urinary carcinoma. Stem Cells International. 2022;2022:3611677.

    PubMed  PubMed Central  Google Scholar 

  51. Ishimoto T, Nagano O, Yae T, Tamada M, Motohara T, Oshima H, et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth. Cancer Cell. 2011;19(3):387–400.

    CAS  PubMed  Google Scholar 

  52. Nimmakayala RK, Batra SK, Ponnusamy MP. Unraveling the journey of cancer stem cells from origin to metastasis. Biochim et Biophys acta Reviews cancer. 2019;1871(1):50–63.

    CAS  Google Scholar 

  53. Bu Y, Cao D. The origin of cancer stem cells. Front Biosci. 2012;4(3):819–30.

    Google Scholar 

  54. Bjerkvig R, Tysnes BB, Aboody KS, Najbauer J, Terzis AJ. Opinion: the origin of the cancer stem cell: current controversies and new insights. Nat Rev Cancer. 2005;5(11):899–904.

    CAS  PubMed  Google Scholar 

  55. Ye R, Xu S, Liu Y, Pang L, Lian X, Zhong Y, et al. Protective effect of Icariin on the development of preimplantation mouse embryos against hydrogen peroxide-induced oxidative injury. Oxidative Med Cell Longev. 2017;2017:2704532.

    Google Scholar 

  56. Zang J, Ye K, Fei Y, Zhang R, Chen H, Zhuang G. Immunotherapy in the treatment of urothelial bladder Cancer: insights from single-cell analysis. Front Oncol. 2021;11:696716.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Yang YM, Chang JW. Bladder cancer initiating cells (BCICs) are among EMA-CD44v6 + subset: novel methods for isolating undetermined cancer stem (initiating) cells. Cancer Invest. 2008;26(7):725–33.

    CAS  PubMed  Google Scholar 

  58. Li C, Yang Z, Du Y, Tang H, Chen J, Hu D, et al. BCMab1, a monoclonal antibody against aberrantly glycosylated integrin α3β1, has potent antitumor activity of bladder cancer in vivo. Clin cancer Research: Official J Am Association Cancer Res. 2014;20(15):4001–13.

    CAS  Google Scholar 

  59. Li Y, Lin K, Yang Z, Han N, Quan X, Guo X, et al. Bladder cancer stem cells: clonal origin and therapeutic perspectives. Oncotarget. 2017;8(39):66668–79.

    PubMed  PubMed Central  Google Scholar 

  60. Hamerlik P, Lathia JD, Rasmussen R, Wu Q, Bartkova J, Lee M, et al. Autocrine VEGF-VEGFR2-Neuropilin-1 signaling promotes glioma stem-like cell viability and tumor growth. J Exp Med. 2012;209(3):507–20.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Nishida S, Hirohashi Y, Torigoe T, Inoue R, Kitamura H, Tanaka T, et al. Prostate cancer stem-like cells/cancer-initiating cells have an autocrine system of hepatocyte growth factor. Cancer Sci. 2013;104(4):431–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Goodwin Jinesh G, Willis DL, Kamat AM. Bladder cancer stem cells: biological and therapeutic perspectives. Curr Stem Cell Res Therapy. 2014;9(2):89–101.

    CAS  Google Scholar 

  63. Jinesh GG, Choi W, Shah JB, Lee EK, Willis DL, Kamat AM. Blebbishields, the emergency program for cancer stem cells: sphere formation and tumorigenesis after apoptosis. Cell Death Differ. 2013;20(3):382–95.

    CAS  PubMed  Google Scholar 

  64. Meyer RD, Srinivasan S, Singh AJ, Mahoney JE, Gharahassanlou KR, Rahimi N. PEST motif serine and tyrosine phosphorylation controls vascular endothelial growth factor receptor 2 stability and downregulation. Mol Cell Biol. 2011;31(10):2010–25.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhu TS, Costello MA, Talsma CE, Flack CG, Crowley JG, Hamm LL, et al. Endothelial cells create a stem cell niche in glioblastoma by providing NOTCH ligands that nurture self-renewal of cancer stem-like cells. Cancer Res. 2011;71(18):6061–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Bielecka ZF, Maliszewska-Olejniczak K, Safir IJ, Szczylik C, Czarnecka AM. Three‐dimensional cell culture model utilization in cancer stem cell research. Biol Rev. 2017;92(3):1505–20.

    PubMed  Google Scholar 

  67. Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol. 2014;12(4):207–18.

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Breslin S, O’Driscoll L. Three-dimensional cell culture: the missing link in drug discovery. Drug Discov Today. 2013;18(5–6):240–9.

    CAS  PubMed  Google Scholar 

  69. Pampaloni F, Reynaud EG, Stelzer EH. The third dimension bridges the gap between cell culture and live tissue. Nat Rev Mol Cell Biol. 2007;8(10):839–45.

    CAS  PubMed  Google Scholar 

  70. Baker BM, Chen CS. Deconstructing the third dimension: how 3D culture microenvironments alter cellular cues. J Cell Sci. 2012;125(Pt 13):3015–24.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Bissell MJ, Rizki A, Mian IS. Tissue architecture: the ultimate regulator of breast epithelial function. Curr Opin Cell Biol. 2003;15(6):753–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. von der Mark K, Gauss V, von der Mark H, Müller P. Relationship between cell shape and type of collagen synthesised as chondrocytes lose their cartilage phenotype in culture. Nature. 1977;267(5611):531–2.

    PubMed  Google Scholar 

  73. Petersen OW, Rønnov-Jessen L, Howlett AR, Bissell MJ. Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. Proc Natl Acad Sci U S A. 1992;89(19):9064–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Mahmud G, Campbell CJ, Bishop KJ, Komarova YA, Chaga O, Soh S, et al. Directing cell motions on micropatterned ratchets. Nat Phys. 2009;5(8):606–12.

    CAS  Google Scholar 

  75. Kilian KA, Bugarija B, Lahn BT, Mrksich M. Geometric cues for directing the differentiation of mesenchymal stem cells. Proc Natl Acad Sci U S A. 2010;107(11):4872–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Debnath J, Brugge JS. Modelling glandular epithelial cancers in three-dimensional cultures. Nat Rev Cancer. 2005;5(9):675–88.

    CAS  PubMed  Google Scholar 

  77. Nelson CM, Bissell MJ. Of extracellular matrix, scaffolds, and signaling: tissue architecture regulates development, homeostasis, and cancer. Annu Rev Cell Dev Biol. 2006;22:287–309.

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Mseka T, Bamburg JR, Cramer LP. ADF/cofilin family proteins control formation of oriented actin-filament bundles in the cell body to trigger fibroblast polarization. J Cell Sci. 2007;120(Pt 24):4332–44.

    CAS  PubMed  Google Scholar 

  79. Weaver VM, Lelièvre S, Lakins JN, Chrenek MA, Jones JC, Giancotti F, et al. beta4 integrin-dependent formation of polarized three-dimensional architecture confers resistance to apoptosis in normal and malignant mammary epithelium. Cancer Cell. 2002;2(3):205–16.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Meyers J, Craig J, Odde DJ. Potential for control of signaling pathways via cell size and shape. Curr Biol. 2006;16(17):1685–93.

    CAS  PubMed  Google Scholar 

  81. Fischbach C, Chen R, Matsumoto T, Schmelzle T, Brugge JS, Polverini PJ, et al. Engineering tumors with 3D scaffolds. Nat Methods. 2007;4(10):855–60.

    CAS  PubMed  Google Scholar 

  82. Gilbert PM, Havenstrite KL, Magnusson KE, Sacco A, Leonardi NA, Kraft P, et al. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science. 2010;329(5995):1078–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126(4):677–89.

    CAS  PubMed  Google Scholar 

  84. Mazzoleni G, Di Lorenzo D, Steimberg N. Modelling tissues in 3D: the next future of pharmaco-toxicology and food research? Genes Nutr. 2009;4(1):13–22.

    CAS  PubMed  Google Scholar 

  85. Vinci M, Gowan S, Boxall F, Patterson L, Zimmermann M, Court W, et al. Advances in establishment and analysis of three-dimensional tumor spheroid-based functional assays for target validation and drug evaluation. BMC Biol. 2012;10:29.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Vinci M, Box C, Eccles SA. Three-dimensional (3D) tumor spheroid invasion assay. J Vis Exp. 2015;99:e52686.

    Google Scholar 

  87. Cawkill D, Eaglestone SS. Evolution of cell-based reagent provision. Drug Discov Today. 2007;12(19–20):820–5.

    CAS  PubMed  Google Scholar 

  88. Lee J, Cuddihy MJ, Kotov NA. Three-dimensional cell culture matrices: state of the art. Tissue Eng Part B Rev. 2008;14(1):61–86.

    CAS  PubMed  Google Scholar 

  89. Benya PD, Shaffer JD. Dedifferentiated chondrocytes reexpress the differentiated collagen phenotype when cultured in agarose gels. Cell. 1982;30(1):215–24.

    CAS  PubMed  Google Scholar 

  90. Ghosh S, Spagnoli GC, Martin I, Ploegert S, Demougin P, Heberer M, et al. Three-dimensional culture of melanoma cells profoundly affects gene expression profile: a high density oligonucleotide array study. J Cell Physiol. 2005;204(2):522–31.

    CAS  PubMed  Google Scholar 

  91. Berthiaume F, Moghe PV, Toner M, Yarmush ML. Effect of extracellular matrix topology on cell structure, function, and physiological responsiveness: hepatocytes cultured in a sandwich configuration. FASEB J. 1996;10(13):1471–84.

    CAS  PubMed  Google Scholar 

  92. Semino CE, Merok JR, Crane GG, Panagiotakos G, Zhang S. Functional differentiation of hepatocyte-like spheroid structures from putative liver progenitor cells in three-dimensional peptide scaffolds. Differentiation. 2003;71(4–5):262–70.

    CAS  PubMed  Google Scholar 

  93. Powers MJ, Janigian DM, Wack KE, Baker CS, Beer Stolz D, Griffith LG. Functional behavior of primary rat liver cells in a three-dimensional perfused microarray bioreactor. Tissue Eng. 2002;8(3):499–513.

    PubMed  Google Scholar 

  94. Frieboes HB, Zheng X, Sun CH, Tromberg B, Gatenby R, Cristini V. An integrated computational/experimental model of tumor invasion. Cancer Res. 2006;66(3):1597–604.

    CAS  PubMed  Google Scholar 

  95. Marushima H, Shibata S, Asakura T, Matsuura T, Maehashi H, Ishii Y, et al. Three-dimensional culture promotes reconstitution of the tumor-specific hypoxic microenvironment under TGFβ stimulation. Int J Oncol. 2011;39(5):1327–36.

    CAS  PubMed  Google Scholar 

  96. Bulysheva AA, Bowlin GL, Petrova SP, Yeudall WA. Enhanced chemoresistance of squamous carcinoma cells grown in 3D cryogenic electrospun scaffolds. Biomed Mater. 2013;8(5):055009.

    CAS  PubMed  Google Scholar 

  97. Griffith LG, Swartz MA. Capturing complex 3D tissue physiology in vitro. Nat Rev Mol Cell Biol. 2006;7(3):211–24.

    CAS  PubMed  Google Scholar 

  98. Khetan S, Burdick JA. Patterning network structure to spatially control cellular remodeling and stem cell fate within 3-dimensional hydrogels. Biomaterials. 2010;31(32):8228–34.

    CAS  PubMed  Google Scholar 

  99. Hickman JA, Graeser R, de Hoogt R, Vidic S, Brito C, Gutekunst M, et al. Three-dimensional models of cancer for pharmacology and cancer cell biology: capturing tumor complexity in vitro/ex vivo. Biotechnol J. 2014;9(9):1115–28.

    CAS  PubMed  Google Scholar 

  100. Weiswald LB, Bellet D, Dangles-Marie V. Spherical cancer models in tumor biology. Neoplasia. 2015;17(1):1–15.

    PubMed  PubMed Central  Google Scholar 

  101. Li Q, Chen C, Kapadia A, Zhou Q, Harper MK, Schaack J, et al. 3D models of epithelial-mesenchymal transition in breast cancer metastasis: high-throughput screening assay development, validation, and pilot screen. J Biomol Screen. 2011;16(2):141–54.

    CAS  PubMed  Google Scholar 

  102. Wang Y, Day ML, Simeone DM, Palmbos PL. 3-D Cell Culture System for studying Invasion and evaluating therapeutics in bladder Cancer. J Vis Exp. 2018(139).

  103. Kim Y, Lee J, Kim S, Shin K. Culture, Manipulation, and Orthotopic transplantation of mouse bladder Tumor Organoids. J Vis Exp. 2020(155).

  104. Yamada KM, Cukierman E. Modeling tissue morphogenesis and cancer in 3D. Cell. 2007;130(4):601–10.

    CAS  PubMed  Google Scholar 

  105. Hamburger AW, Salmon SE. Primary bioassay of human tumor stem cells. Science. 1977;197(4302):461–3.

    CAS  PubMed  Google Scholar 

  106. Oskarsson T, Batlle E, Massagué J. Metastatic stem cells: sources, niches, and vital pathways. Cell Stem Cell. 2014;14(3):306–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Qureshi-Baig K, Ullmann P, Haan S, Letellier E. Tumor-initiating cells: a criTICal review of isolation approaches and new challenges in targeting strategies. Mol Cancer. 2017;16(1):40.

    PubMed  PubMed Central  Google Scholar 

  108. Feder-Mengus C, Ghosh S, Reschner A, Martin I, Spagnoli GC. New dimensions in tumor immunology: what does 3D culture reveal? Trends Mol Med. 2008;14(8):333–40.

    CAS  PubMed  Google Scholar 

  109. Ribatti D, Tamma R, Annese T. Epithelial-mesenchymal transition in Cancer: a historical overview. Translational Oncol. 2020;13(6):100773.

    Google Scholar 

  110. Maliszewska-Olejniczak K, Brodaczewska KK, Bielecka ZF, Czarnecka AM. Three-dimensional cell culture model utilization in Renal Carcinoma Cancer Stem Cell Research. Methods in Molecular Biology (Clifton NJ). 2018;1817:47–66.

    CAS  Google Scholar 

  111. Zhang C, Yang Z, Dong DL, Jang TS, Knowles JC, Kim HW, et al. 3D culture technologies of cancer stem cells: promising ex vivo tumor models. J Tissue Eng. 2020;11:2041731420933407.

    PubMed  PubMed Central  Google Scholar 

  112. Lau EY-T, Ho NP-Y, Lee TK-W. Cancer stem cells and their microenvironment: biology and therapeutic implications. Stem cells international. 2017;2017.

  113. Amaral RLF, Miranda M, Marcato PD, Swiech K. Comparative analysis of 3D bladder tumor spheroids obtained by forced floating and hanging Drop methods for drug screening. Front Physiol. 2017;8:605.

    PubMed  PubMed Central  Google Scholar 

  114. Kim MJ, Chi BH, Yoo JJ, Ju YM, Whang YM, Chang IH. Structure establishment of three-dimensional (3D) cell culture printing model for bladder cancer. PLoS ONE. 2019;14(10):e0223689.

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Kloskowski T, Szeliski K, Fekner Z, Rasmus M, Dąbrowski P, Wolska A, et al. Ciprofloxacin and levofloxacin as potential drugs in Genitourinary Cancer Treatment-The Effect of dose-response on 2D and 3D cell cultures. Int J Mol Sci. 2021;22(21):11970.

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Torab P, Yan Y, Yamashita H, Warrick JI, Raman JD, DeGraff DJ, et al. Three-dimensional microtumors for probing heterogeneity of invasive bladder Cancer. Anal Chem. 2020;92(13):8768–75.

    CAS  PubMed  Google Scholar 

  117. Yoshida T, Sopko NA, Kates M, Liu X, Joice G, McConkey DJ, et al. Three-dimensional organoid culture reveals involvement of Wnt/β-catenin pathway in proliferation of bladder cancer cells. Oncotarget. 2018;9(13):11060–70.

    PubMed  PubMed Central  Google Scholar 

  118. Miyake M, Hori S, Morizawa Y, Tatsumi Y, Nakai Y, Anai S, et al. CXCL1-Mediated Interaction of Cancer cells with Tumor-Associated Macrophages and Cancer-Associated fibroblasts promotes Tumor Progression in human bladder Cancer. Neoplasia. 2016;18(10):636–46.

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Yoshida T, Sopko NA, Kates M, Liu X, Joice G, McConkey DJ, et al. Impact of spheroid culture on molecular and functional characteristics of bladder cancer cell lines. Oncol Lett. 2019;18(5):4923–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Burgués JP, Gómez L, Pontones JL, Vera CD, Jiménez-Cruz JF, Ozonas M. A chemosensitivity test for superficial bladder cancer based on three-dimensional culture of tumour spheroids. Eur Urol. 2007;51(4):962–9. discussion 9–70.

    PubMed  Google Scholar 

  121. Wang P, Zhou R, Thomas P, Zhao L, Zhou R, Mandal S et al. Epithelial-to-mesenchymal transition enhances Cancer Cell Sensitivity to Cytotoxic Effects of Cold Atmospheric Plasmas in breast and bladder Cancer Systems. Cancers (Basel). 2021;13(12).

  122. Miranda MA, Marcato PD, Mondal A, Chowdhury N, Gebeyehu A, Surapaneni SK, et al. Cytotoxic and chemosensitizing effects of glycoalkaloidic extract on 2D and 3D models using RT4 and patient derived xenografts bladder cancer cells. Mater Sci Eng C Mater Biol Appl. 2021;119:111460.

    CAS  PubMed  Google Scholar 

Download references

Funding

This work received no external funding.

Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB).

Author information

Authors and Affiliations

Authors

Contributions

S.F. and A.A. wrote the main manuscript text. A.K. and A.A. prepared Figs. 1, 2, 3, 4 and 5. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Sameh M. Farouk or Ahmed M. Abdellatif.

Ethics declarations

Ethical approval

Not applicable.

Competing interests

There is no potential conflict of interest to declare.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Farouk, S.M., Khafaga, A.F. & Abdellatif, A.M. Bladder cancer: therapeutic challenges and role of 3D cell culture systems in the screening of novel cancer therapeutics. Cancer Cell Int 23, 251 (2023). https://doi.org/10.1186/s12935-023-03069-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-023-03069-4

Keywords