Open access peer-reviewed chapter - ONLINE FIRST

Genetically Modified Organisms in Urological Cancer

Written By

Le Qu, Cheng Chen, Yaoming Li and Xin Pan

Submitted: 19 April 2024 Reviewed: 25 April 2024 Published: 22 May 2024

DOI: 10.5772/intechopen.1005472

Genetically Modified Organisms IntechOpen
Genetically Modified Organisms Edited by Huseyin Tombuloglu

From the Edited Volume

Genetically Modified Organisms [Working Title]

Dr. Huseyin Tombuloglu and Dr. Guzin Tombuloglu

Chapter metrics overview

14 Chapter Downloads

View Full Metrics

Abstract

Genetically modified organisms (GMOs) have become indispensable tools in pre-clinical research for urological cancer. Through genetic engineering techniques, researchers can modify the genetic composition of organisms, enabling the creation of appropriate experimental animal models that provide a deep insight into the mechanisms of tumorigenesis, progression, and potential therapeutic strategies for urological cancer. In this chapter, we provide a comprehensive overview of the current status of research utilizing GMOs in the investigation of prostate cancer, renal cancer, urothelial cancer, and other urological cancers. Topics covered the development of different genetically modified animal models, and the application of these models in urological cancer research. In addition, the limitations of GMOs in cancer research will be discussed.

Keywords

  • genetically modified organisms
  • renal cancer
  • prostate cancer
  • urothelial cancer
  • transgenic animal models

1. Introduction

Urological malignancies, including cancers of the prostate, bladder, kidney, and other organs of the urinary system, pose a rapidly increasing global cancer burden [1]. In 2020, Global Cancer Observatory (GLOBOCAN) data indicated that over 2.5 million individuals were diagnosed with malignant tumors of the urinary system, resulting in nearly 800,000 deaths [2]. Prostate cancer accounts for 29% of all new cancer diagnoses among men, as per the American Cancer Society, with bladder, kidney, and renal pelvis cancers also ranking in the top 10 for incidence. Among women, renal cancer is the 9th most common cancer [3]. Despite advances in diagnostics and treatments, the outlook for patients with advanced urological cancers remains bleak [4, 5, 6, 7, 8, 9], underscoring the critical need for more research into their pathogenesis and treatment options.

The use of genetically modified organisms (GMOs), through transgenic technology, has revolutionized biology and medical research [10, 11]. Genetically modified animal models, in particular, have become indispensable for studying intricate biological processes, including development, aging, and disease progression [12, 13, 14, 15]. These models stand out from other animal models by more accurately mimicking the genetic and pathological aspects of human urological cancers. Notably, some of these models can spontaneously develop metastatic disease, facilitating study of the microenvironment at various stages of tumor metastasis. They also allow for the assessment of therapeutic effects and the validation of drug targets across different stages of the disease [16, 17, 18].

This chapter provides a comprehensive review of the utility of GMOs, particularly genetically modified animal models, in uncovering the pathogenesis of urological cancers and identifying new therapeutic strategies or drug targets.

Advertisement

2. GMOs as essential models in urological cancer research

2.1 Prostate cancer (PCa)

2.1.1 TRAMP model

The transgenic adenocarcinoma of the mouse prostate (TRAMP) model, first reported by N. M. Greenberg in 1995 [19], employs transgenic technology to introduce the androgen-dependent, prostate tissue-specific protein rat probasin (rPB) into fertilized mouse eggs. This process inactivates tumor suppressors p53, retinoblastoma 1 (Rb1), and protein phosphatase 2A (PP2A) in the prostate, leading to PCa development in mice [20, 21]. Characterized by its clinicopathological features and widespread metastasis, the TRAMP model is considered the most detailed PCa model available. By 8 weeks, TRAMP mice show abundant Simian Virus 40 (SV40) large T-antigen markers in the dorsolateral lobes of the prostate, progressing from low-grade to high-grade prostatic intraepithelial neoplasia by 10 weeks. At 12 weeks, distant metastasis begins, with all mice developing metastatic PCa by 28 weeks—a 100% tumor formation rate [22, 23]. Metastases occur in various locations, predominantly in the dorsolateral lobe of the prostate [24]. Due to its extensive metastatic spread [25, 26], the TRAMP model is pivotal for studying PCa’s pathogenesis and metastatic behavior [27].

2.1.2 LADY model

The LPB promoter driving the large T-antigen (LADY) model, using the large PB promoter (LPB) to express SV40 Large T-antigen (Tag) without the small t-antigen, contrasts with the TRAMP model to highlight neuroendocrine differences in metastatic lesions [28, 29]. It demonstrates progression from prostatic intraepithelial neoplasia (PIN) with neuroendocrine features to invasive neuroendocrine carcinoma [30]. Metastatic lesions appear at one site by 6 months and at multiple sites by 9–10 months, yet only 14% develop bone metastases. The model typically shows metastasis to lymph nodes, liver, lung, spleen, kidney, and occasionally bones [29, 30] but exhibits limited PCa cell proliferation and less invasiveness compared to TRAMP. It serves as an effective model for studying early PIN changes and tumor stroma [31, 32], making it suitable for exploring the mechanisms of neuroendocrine PCa (NEPC) [28].

2.1.3 Other SV40 T-antigen

Beyond the commonly used TRAMP and LADY models, other transgenic models have been developed using SV40 large T-antigen and small t-antigen, combined with various promoters. Jeffrey E. Green et al. introduced a C3(1) initiator to express SV40 large T-antigen, creating the first PCa genetically modified animal model [33, 34]. This model causes female mice to develop breast cancer at 6 months and male mice to develop prostate hyperplasia by 8 months, which can progress to PIN and PCa, occasionally metastasizing to the lung and bone [35]. This genetically modified animal model facilitates the study of hormone response components in vivo, as well as the multistage progression from normal tissue to PCa [36, 37].

Jeffrey I. Gordon et al. developed a model for metastatic PCa using the Cryptidin-2 gene promoter to direct expression of SV40 large T-antigen in prostate neuroendocrine cells, leading to neuroendocrine PCa [38]. The model mice developed PIN at 8 weeks, followed by invasive cancer within 2–4 weeks and metastasis to lymph nodes, liver, lung, brain, and bone within 16 weeks [39]. The tumors were androgen-independent, recapitulating multiple histopathological features of human PCa while avoiding androgen dependence on neuroendocrine cancer evolution [40, 41]. Compared to the TRAMP model, this model has a shorter tumor induction period and provides new biomarkers for clinical diagnosis of PCa neuroendocrine differentiation.

Jim W. Xuan et al. utilized the knock-in technique, using the prostate secretory protein 94 amino acid (PSP94) gene promoter/enhancer to target and regulate mouse prostate-specific expression of SV40 large T/small t-antigen, establishing a new PCa model, prostate secretory protein-knock-in mouse adenocarcinoma prostate (PSP-KIMAP) [42, 43]. This model recapitulates aspects of human PCa, with PIN observed around 10 weeks, progressing to invasive adenocarcinoma by 24 weeks and accompanied by distant metastases to lymph nodes, lungs, and liver [40]. Compared to TRAMP and LADY models, PSP-KIMAP more accurately simulates human PCa development, exhibiting more stable phenotypes and precise prostate tissue targeting [30, 44]. While the PSP-KIMAP study is not comprehensive, it is worth acknowledging for replicating PCa and serving as a valuable addition to other models, holding broad application prospects.

2.1.4 Pten

Phosphatase and tensin homolog deleted on chromosome ten (Pten) gene, a significant tumor suppressor, is often deficient in various human cancers [45]. In human PCa, Pten deletion is found in 23% of high-grade PIN (HGPIN), 69% of localized cases [46], and 86% of metastatic castration-resistant PCa (CRPC) [47]. This deficiency underscores Pten’s crucial role in PCa initiation, leading to the development of Pten-deficient mouse models. Shun you Wang et al. created a model with prostate-specific Pten deletion, revealing that heterozygous mice developed PIN at 10 months, while homozygous deletion resulted in invasive adenocarcinoma by 9 weeks, with metastasis by 12 weeks [48]. However, tumors also appeared in non-prostate tissues [49, 50, 51], which could limit the model’s specificity for PCa research.

Researchers have explored the role of interleukin-17 (IL-17) in PCa using these models, shedding light on the tumor microenvironment [52, 53]. They also investigated the interaction between tumor suppressors Rb and Pten using double mutant mice with cyclin-dependent kinase (Cdk) inhibitor p18Ink4c and Pten knockout [54]. Results show that double mutant mice have faster and more extensive tumor growth in the prostate anterior and dorsolateral lobes [54]. Additionally, loss of Nkx3.1, combined with Pten deficiency, significantly increases HGPIN incidence, mirroring early stage human PCa [55, 56]. Similarly, DePinho et al. found that prostate-specific deletion of Pten (Pten pc−/−) and prostate-specific deletion of Smad4 (Smad4 pc−/−) in mouse models exhibited highly invasive characteristics with deep lymphatic and pulmonary metastases [57]. Based on these findings, the Pten pc−/−/Smad4 pc−/− mouse model was constructed and applied to research on combinations of hypoxia-activated prodrug TH-302 and checkpoint blockade [58]. They found that the combination of TH-302 and checkpoint blockade significantly increased the survival of Pten pc−/−/Smad4 pc−/− mice [58]. Moreover, Xin Lu and colleagues also utilized this model to reveal potential pathways for improving immunotherapy in advanced prostate cancer through Pygo2 (Pygopus2 (Pygopus family plant homeo domain (PHD) finger 2))-targeted treatment [59]. Additionally, a model with Pten deletion and speckle-type pox virus and zinc finger protein (Spop) mutation demonstrated the role of Spop mutations in activating phosphatidylinositol-3-kinase/mammalian target of rapamycin (PI3K/mTOR) and androgen receptor (AR) signaling, advancing the understanding of PCa progression [60]. The Pten/Kras (Kirsten rat sarcoma viral oncogene homolog) model reported by David J. Mulholland et al. was found to accelerate prostate cancer progression due to Pten deficiency, with concomitant epithelial-mesenchymal transition (EMT) and extensive metastasis [61].

2.1.5 Myc

Previous studies have suggested that upregulation of Myc may be a critical driving event in the onset and progression of human PCa [62, 63]. Charles L. Sawyers and his team created transgenic mice expressing human c-Myc in the prostate [64], categorized into Hi-Myc and Lo-Myc groups based on androgen sensitivity, with Hi-Myc being androgen-sensitive. The Hi-Myc mice showed accelerated PIN progression compared to the Lo-Myc group [64]. Despite its advantages, the model’s lack of transferability is a significant limitation [64]. Interestingly, concurrent Myc overexpression and Pten deficiency in mice resulted in aggressive adenocarcinomas with distant metastases. The research on this animal model has confirmed that Homeobox protein Hox-B13 (Hoxb13) played a pivotal role in the causation of prostate cancer [65, 66]. Combining Hi-Myc mice with PB-Hepsin mice reduced adenocarcinoma progression from 24 to 12 weeks [67]. However, studies show that mTOR inhibitors are ineffective against Myc overexpression mice, suggesting that they may be contraindicated in Myc overexpression PCa patients [68]. Given Myc’s early amplification in PCa [62, 69], the Myc transgenic model serves as a foundational tool for studying genetic alterations in PCa progression, often used in conjunction with other PCa-related genes.

N-Myc and L-Myc, members of the Myc family, play roles in PCa development and progression. Mycl amplification is observed in precancerous lesions and early tumors [70], whereas N-Myc is associated with aggressive castration-resistant PCa (CRPC) and NEPC [71]. Etienne Dardenne et al. established a transgenic mouse model overexpressing N-Myc and identified it as an oncogenic driver of NEPC [72]. This model suggests that AR signaling is abolished and Polycomb Repressive Complex 2 (Ezh2) signaling is induced [72]. The model helps identify and validate potential therapeutic targets for treating NEPC, such as Ezh2 and Aurora A kinase.

2.1.6 Cdcp1

Abdullah Alajati et al. developed a mouse model of prostate cancer (PCa) that overexpresses CUB domain-containing protein 1 (Cdcp1, 73]. In this model, 50% of mice exhibited prostate hyperplasia within 4–6 months, which progressed to PIN by 7–9 months and to HGPIN by 14 months [73]. The simultaneous overexpression of Cdcp1 and knockdown of Pten markedly accelerated the development of metastatic PCa in these mice. Utilizing this model, the researchers identified promising drug treatment strategies for combating metastatic PCa [73].

2.2 Renal cancer

Research highlights the critical tumor-suppressing role of von Hippel-Lindau (Vhl) in clear cell renal cell carcinoma (ccRCC) development [74], notably at tumorigenesis’s initial stages [75]. While modern sequencing technologies corroborate this [76], studies indicate that Vhl loss alone does not suffice to induce renal cell carcinoma (RCC) [77, 78, 79, 80, 81]. This insight has led researchers to combine Vhl deletion with other oncogenic modifications, aiming to develop an optimal renal cancer model. Following Vhl, Polybromo 1 (Pbrm1) emerges as the second key suppressor gene in renal cancer, with about 40% of ccRCC cases involving Pbrm1 mutations [82, 83, 84]. Interestingly, single knockdown of Pbrm1 by some researchers through knockout technology did not result in the expected renal cancer model. However, discoveries of ccRCC with combined mutations in Vhl and Pbrm1 [83, 85], and metastatic ccRCC with mutations in both Brca1-associated protein 1 (Bap1) and Pbrm1 [85, 86], suggest ccRCC may require simultaneous knockdown of two or more genes.

2.2.1 Vhl/Pbrm1

Amrita M Nargund and colleagues have developed a ccRCC mouse model by knocking out both Vhl and Pbrm1 genes via the Ksp-Cre method [87]. This model not only led to renal cyst formation but also successfully induced ccRCC. Mice deficient in both Vhl and Pbrm1 showed a 30% incidence of preneoplastic polycystic kidney diseases by 6–9 months and a 50% ccRCC incidence by 10 months [87]. Similarly, Yi Feng Gu’s team created a double knockout model using Paired Box 8 (Pax8)-Cre, resulting in about 85% of mice developing extensive tumors by 9 months, escalating to 100% by 13 months [88], with tumors eventually nearly replacing the kidneys by 16 months [88]. The Vhl/Pbrm1 model serves as a valuable tool for exploring the molecular dynamics of Vhl and Pbrm1 mutations and for conducting drug efficacy tests.

2.2.2 Vhl/Bap1

Subsequent research identified the Bap1 gene as significantly associated with ccRCC, ranking it as the third most important ccRCC-associated gene with a mutation rate of approximately 15% [83, 85, 89]. Bap1 mutations are linked to higher-grade ccRCC, contrasting with the lower-grade associations of Pbrm1 mutations. Shan Shan Wang et al. initially attempted a simultaneous Vhl/Bap1 deletion using Six2-Cre, but mice with Bap1 deficiency died within a month of birth [89]. Later, Yi Feng Gu et al. successfully created a Vhl/Bap1 deletion model with Pax8-Cre, which, alongside the Vhl-Pbrm1 model, elucidates the role of these genes in ccRCC formation and tumor grading. Bap1-deficient tumors tend to be of higher-grade compared to Pbrm1-deficient tumors [88, 90].

2.2.3 Vhl/Trp53/Rb1

Sabine Harlander and colleagues developed a new ccRCC mouse model by deleting Vhl, transformation-related protein 53 (Trp53), and Rb1 in renal epithelial cells [91]. This model provides a basis for studying hypoxia-inducible factor-alpha (HIF-α) inhibition as a potential ccRCC treatment and offers a platform for drug screening and testing new therapies [91]. However, such triple gene inactivation is rare in human ccRCC, and the model does not exhibit metastases to lung, liver, bone, or brain, highlighting its limitations.

2.2.4 Myc/Vhl/Cdkn2a

Sean T. Bailey and his team created a metastatic renal cancer model by overexpressing Myc and deleting Vhl and cyclin-dependent kinase inhibitor 2A (Cdkn2a), mimicking human ccRCC [92]. Remarkably, about one-third of the mice developed liver metastases [92]. However, the combination of Vhl and Cdkn2a loss, along with Myc activation, is relatively rare in humans, limiting the model’s representativeness [92].

2.2.5 Flcn

The folliculin (Flcn) gene, identified from a Birt-Hogg-Dube syndrome (BHD) patient [93, 94], plays a role in causing BHD syndrome, which includes RCC among other diseases. Initially established whole-body Flcn knockout homozygote mice died at the embryonic stage, whereas heterozygous mice had a very late onset of disease with a low and erratic morbidity rate [95, 96, 97]. Using Ksp-Cre, researchers established an Flcn-deficient model that successfully mimics RCC [98, 99, 100], showing 100% renal cyst incidence and about 70% tumor incidence within 6–7 months [100]. Thus, the Flcn-deficient model can be utilized for molecular mechanism studies and drug testing and screening in renal cancer. Unfortunately, there is a diversity of histologic subtypes of RCC observed in this model, and it is not yet possible to determine whether there are other factors that determine the histologic type of RCC in this model, a question that remains to be investigated.

2.2.6 Other renal cancer models

Zachary S. Morris et al. developed a genetic mouse model with neurofibromatosis type 2 (Nf2) knockout using Villin-Cre [101], observing renal cell hyperplasia and cysts at 15 days, and small renal tumors by 3 months that progressed to invasive renal cancer by 6 to 10 months. This model also exhibited elevated epidermal growth factor receptor (Egfr) expression, indicating that Nf2 inactivation activates the Egfr signaling pathway, promoting renal cancer development [101]. Although Nf2 is not primarily associated with renal cancer, this model presents an early onset opportunity for studying Nf2-related renal cancer pathogenesis and drug screening.

Lorraine J. Gudas et al. reported a mouse model for Vhl renal cancer known as the TRAnsgenic model of Cancer of the Kidney (TRACK). This model is characterized by the specific expression of mutant hypoxia-inducible factor 1-alpha (Hif1a) in the renal proximal tubule cells, closely mirroring many early stage features of ccRCC [102].

Alessia Calcagnì et al. created the first genetic animal model of renal cancer with transcription factor EB (Tfeb) overexpression using cadherin 16 (CDH16)-Cre [103]. This model sheds light on the mechanism of TFE-fusion RCC and suggests a therapeutic approach targeting the Wingless/integrated (WNT) pathway [103]. However, the model mice developed severe renal impairment, an outcome not commonly seen in humans, which may limit the model’s utility for therapeutic exploration.

Qiang Hua Hu et al. produced a Wilms tumor (WT) mouse model by increasing Igf2 expression and eliminating WT1, making it the only WT transgenic mouse model closely resembling human tumors [104]. Visible tumors appeared at 9 weeks, providing a valuable tool for testing therapeutic strategies [105].

2.3 Urothelial cancer (UC)

The uroepithelium, one of the body’s slowest-renewing epithelia, undergoes unique biological transformations. Oncogene inactivation or activation plays a crucial role in bladder tumorigenesis [106]. With gradual clarification of UC-related molecular mechanisms and maturing molecular biology techniques, GMOs have been successfully established, better reproducing human UC biological behavior at the molecular level. GMOs are widely used to study specific gene functions of Hras (Harvey rat sarcoma viral oncogene homolog), P53, Pten, Rb, fibroblast growth factor receptor 3 (Fgfr3), and epidermal growth factor receptor (Egfr) in bladder cancer development [107].

2.3.1 SV40 T-antigen

Zhong Ting Zhang et al. developed a transgenic mouse model in 1999 using the uroplakin II (UpII) promoter to express SV40 T-antigen urothelially [108]. This model showed that both low and high copy number SV40T transgenic mice develop bladder carcinoma in situ (CIS), with high copy number mice progressing to invasive and metastatic transitional cell carcinoma (TCC) [108]. Francisco Ayala de la Peña et al. later created a model using UPK II-Cre that did not progress to invasive tumors [109].

2.3.2 Hras

Hras, the first oncogene identified in human UC [110], led Zhong Ting Zhang et al. in 2001 to establish a mouse model activating Hras in the uroepithelium. This resulted in urothelial hyperplasia and superficial papillary non-invasive bladder tumors [111]. They found that low copy number Hras transgenic mice developed non-invasive lesions, whereas high copy number mice died by 5 months, highlighting the RAS pathway’s role in developing low-grade, non-invasive papillary UC [111].

2.3.3 P53

P53, crucial for uroepithelial cell growth control [112], is frequently mutated or deleted in human UC [113]. Jing Gao and his team have demonstrated that the complete loss of p53 is a prerequisite for the activation of Hras to promote the generation of UC via a model constructed by hybridizing active Hras transgenic mice with p53-deficient mice [114]. In addition, Anna M. Puzio-Kuter et al. have shown that combined deletion of p53 and Pten in bladder epithelial cells leads to invasive cancer in a novel mouse model that provides a validating tool to study mTOR inhibitors for the treatment of invasive UC [115].

2.3.4 Pten

Chao Nan Qian and colleagues created a model by specifically knocking out Pten in kidney epithelial cells using Ksp-Cre [116]. About 57% of these mice developed UC of the renal pelvis by 12 months, with significantly increased phosphorylated mTOR levels, suggesting mTOR inhibitors as effective treatments [116].

2.4 GMOs of other urological cancers

Testicular germ cell tumors (TGCTs), the predominant form of testicular cancers among young men, originate from germ cells [117]. James A. Gill et al. developed TGCT in zebrafish by expressing SV40 large T-antigen under the pufferfish lymphocyte-specific protein tyrosine kinase (Flck) promoter, observing TGCT development after a latency of up to one year. Additionally, overexpression of the stem cell leukemia (Scl) gene in zebrafish testis also led to TGCT [118]. These findings demonstrate the viability of studying TGCT in vivo. Earlier mouse models were limited to benign tumors, not reflecting the malignant TGCT more commonly seen in young men [119, 120, 121]. Amy M. Lyndaker et al. constructed a mouse model of malignant TGCT by localizing Kras activation and Pten inactivation in mouse premeiotic germ cells [122, 123].

Penile cancer, a rare urinary system malignancy, is often preceded by penile intraepithelial neoplasia (PeIN), a precancerous lesion linked to human papillomavirus (HPV) [124, 125]. Beatriz Medeiros Fonseca and colleagues introduced the first mouse model mimicking HPV-related penile cancer [126]. This model was created by treating 10-week-old HPV16 transgenic mice with dimethylbenz(o)anthracene (DMBA) over a 16-week period, leading to the development of HPV-associated penile cancer traits similar to those observed in humans, such as condylomas and PeIN [126]. Their pioneering research has paved the way for further studies into the underlying mechanisms and potential treatment options for penile cancer.

Advertisement

3. Challenges and limitations of GMOs

Advances in genome editing, such as clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) [127], face challenges like off-target effects and exogenous mutations, complicating the maintenance of transgenic models and impacting research. The complexity of gene regulation in urological cancer adds to the difficulty of creating accurate models. These challenges hinder the broader application of transgenic models in urological cancer research. Ethical and safety considerations of GMOs also demand careful attention.

Advertisement

4. Potential development trends of GMOs

Improvements in CRISPR/Cas9 and the advent of new editing techniques like the National Institute for Cancer Epidemiology and Research (NICER) and AsCas12f promise enhanced transgenic model efficiency and accuracy [128, 129]. Future urologic oncology research may benefit from interdisciplinary collaboration and integrating technologies like three-dimensional (3D) printing and artificial intelligence (AI), potentially revolutionizing our understanding of urological tumors.

Advertisement

5. Conclusions

In conclusion, these GMOs have greatly aided understanding of urological cancers’ etiology, progression, and metastasis, and are critical for testing novel drug targets, and assessing treatment responses (Table 1). They promote urological cancer research and allow researchers to choose suitable models for deep exploration in their fields.

Cancer typeModelTime of penetrance, PathologyCharacteristicsReferences
Prostate cancerTRAMP10 weeks: LGPIN to HGPIN
12 weeks: distant metastasis begins
28 weeks: 100% metastatic tumor formation
First autochthonous mouse model of PCa[19]
LADY6 months: metastatic tumor at one site
9–10 months: metastatic tumor at multiple sites (14% bone)
Less invasiveness[28]
C3(1)8 months: prostate hyperplasia even PIN and PCaHormone response components in vivo[33]
Cryptidin-28 weeks: PIN
10–12 weeks: invasive cancer
Within 16 weeks: metastasis to lymph nodes, liver, lung, brain, and bone
Shorter tumor induction period[38]
PSP-KIMAP10 weeks: PIN
24 weeks: invasive AD
accompanied by distant metastases to lymph nodes, lungs, and liver
More accurately simulates human PCa[42]
PtenPten+/−
10 months: PIN
Pten−/−
9 weeks: invasive AD
12 weeks: metastasis
Tumors also appeared in non-prostate tissues[48]
Pten/p18Ink4c9 months:
Pten+/− /p18−/−
HGPIN
12 months:
Pten+/− /p18+/−
HGPIN or carcinoma
Faster and more extensive PCa growth[54]
Pten/Nkx3.126–52 weeks: 60% HGPIN
52 weeks: 100% HGPIN
>52 weeks: 84% AD, 25% lymph node metastasis
Mirror early stage human PCa[55, 130]
Pten/Smad47 weeks:
LGPIN
11 weeks: invasive PCa
15 weeks: highly aggressive PCa
32 weeks: 100% lymph node, 12% lung metastasis
Highly invasive[57]
Ptne/Kras10 weeks: PIN
20 weeks: AD
40 weeks: Death
EMT and stem-like features[61]
Hi-Myc>13 weeks: PIN
>26 weeks: AD
Accelerated PIN progression[64]
Hi-Myc/PB-Hepsin4.5 months: invasive ADHigher-grade AD[67]
N-Myc/PtenNEPCAbolish AR signaling[72]
Cdcp14–6 months: prostate hyperplasia
7–9 months: PIN
14 months: HGPIN
SRC/MAPK pathway activation[73]
Renal cancerVhl/Pbrm1Ksp-Cre
6–9 months: 30% preneoplastic polycystic kidney
10 months: 50% ccRCC
Pax8-Cre
9 months: 85% ccRCC
13 months: 100% ccRCC
13 months: tumor nearly replace kidney
Valuable tool for ccRCC research[87]
Vhl/Bap1Six2-Cre: die within a month of birth
Pax-8cre: different grades of ccRCC
Higher-grade tumors[88]
Vhl/Trp53/Rb110 of 25 develop a total of 64 tumorsNo metastasis[91]
Myc/Vhl/Cdkn2aone-third of the mice developed liver metastasesRare in human ccRCC[92]
Flcn6–7 months: 100% renal cyst, 70% tumorDiversity of histologic subtypes[100]
Nf215 days: renal cell hyperplasia and cysts
6–10 months: invasive renal cancer
Nf2-related renal cancer[101]
TRACKccRCCHif1a activation[102]
TfebTFE-fusion RCCFirst genetic animal
models of RCC
[103]
Igf2WTThe only WT transgenic mouse model[104]
Urothelial cancerSV40 T-Antigenlow copy number: CIS
high copy number: metastatic TCC
Develop bladder CIS and TCC[108]
Hraslow copy number: urothelial hyperplasia, superficial papillary non-invasive bladder tumorsRAS pathway activation[111]
P53/PtenInvasive cancerNA[115]
Pten12 months: 57% UCDevelop UC of the renal pelvis[116]
Testicular germ cell tumorsSV40 T-antigen>1 year: TGCTDevelop TGCT in zebrafish[118]
SclTGCTNA[118]
Kras/PtenTGCTNA[122]
Penile cancerHPV1616 weeks: condylomas and PeINFirst mouse model mimicking HPV-related penile cancer[126]

Table 1.

Commonly GMOs in urological cancers.

Abbreviations: PIN prostatic intraepithelial neoplasia; PCa prostate cancer; NEPC: neuroendocrine prostate cancer; LGPIN: low-grade prostatic intraepithelial neoplasia; HGPIN: high-grade prostatic intraepithelial neoplasia; AD: adenocarcinoma; AR: androgen receptor; ccRCC: clear cell renal cell carcinoma; WT: Wilms tumor; CIS: carcinoma in situ; TCC: transitional cell carcinoma; UC: urothelial cancer; TGCT: testicular germ cell tumor; NA: not applicable; PeIN: penile intraepithelial neoplasia.

Advertisement

Acknowledgments

This work was supported by the National Natural Science Foundation of China (No. 82173345 and No. 82373154 to Le Qu).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Chunyang L, Xiaoxi Z, Shi Q , Yonghong G, Yonggang Z. Nanomedicine for urologic cancers: Diagnosis and management. Seminars in Cancer Biology. 2022;86(Pt 2):463-475. DOI: 10.1016/j.semcancer.2022.05.014
  2. 2. Hyuna S, Jacques F, Rebecca LS, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2021;71(3):209-249. DOI: 10.3322/caac.21660
  3. 3. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA: A Cancer Journal for Clinicians. 2024;74(1):12-49. DOI: 10.3322/caac.21820
  4. 4. Zhang ZH, Wang Y, Zhang Y, et al. The function and mechanisms of action of circular RNAs in urologic cancer. Molecular Cancer. 2023;22(1):61. DOI: 10.1186/s12943-023-01766-2
  5. 5. Shahneen S, Caroline MM, Edmund C, Himisha B, Robert GB, Scott W. Prostate cancer. The Lancet. 2021;398(10305):1075-1090. DOI: 10.1016/s0140-6736(21)00950-8
  6. 6. Regan MM, Mantia CM, Werner L, et al. Treatment-free survival over extended follow-up of patients with advanced melanoma treated with immune checkpoint inhibitors in CheckMate 067. Journal for Immunotherapy of Cancer. 2021;9(11):e003743-e003743. DOI: 10.1136/jitc-2021-003743
  7. 7. Wei G, Sun H, Dong K, et al. The thermogenic activity of adjacent adipocytes fuels the progression of ccRCC and compromises anti-tumor therapeutic efficacy. Cell Metabolism. 2021;33(10):2021-2039.e8. DOI: 10.1016/j.cmet.2021.08.012
  8. 8. Nadal R, Bellmunt J. Management of metastatic bladder cancer. Cancer Treatment Reviews. 2019;76:10-21. DOI: 10.1016/j.ctrv.2019.04.002
  9. 9. Nair SS, Chakravarty D, Patel V, Bhardwaj N, Tewari AK. Genitourinary cancer neoadjuvant therapies: Current and future approaches. Trends Cancer. 2023;9(12):1041-1057. DOI: 10.1016/j.trecan.2023.07.011
  10. 10. Mauricio R-M, Gabriel RC, Gabriel M-R, Rodrigo APM. From gene targeting to genome editing: Transgenic animals applications and beyond. Anais Da Academia Brasileira De Ciencias. 2015;87(2 suppl):1323-1348. DOI: 10.1590/0001-3765201520140710
  11. 11. Hao Y, Kevin KJ, Anderson DG. Delivery technologies for genome editing. Nature Reviews. Drug Discovery. 2017;16(6):387-399. DOI: 10.1038/nrd.2016.280
  12. 12. Jun-Gyu P, Paula AP, Anwari A, Xavier A, Jordi BT, Luis M-S. Animal models of COVID-19: Transgenic mouse model. Methods in Molecular Biology. 2022:259-289. DOI: 10.1007/978-1-0716-2111-0_16
  13. 13. Laszlo D, Katalin Ö, Orsolya H, et al. Characterization of cardiac effects of OPA1 protein promotion in transgenic animal model. European Heart Journal. 2022;43(Supplement_2). DOI: 10.1093/eurheartj/ehac544.2892
  14. 14. Amélie C, Charbel C, Silvia S, et al. Tu1881 HLA B27 transgenic rat: A new animal model of postsurgical ileitis in inflammatory bowel disease. Gastroenterology. 2016;150(4):S967-S967. DOI: 10.1016/s0016-5085(16)33275-9
  15. 15. Sambhavi B, Amulya V, Anandan B. Genetically engineered and spontaneous animal model: Utilization in preclinical cancer therapy development. In: Handbook of Animal Models and Its Uses in Cancer Research. 2023. pp. 421-431. DOI: 10.1007/978-981-19-1282-5_22-1
  16. 16. Sousa ER, Zoni E, Scarpa M, et al. Cripto as a key factor in aggressive lethal prostate cancer: Generation of a new transgenic mouse model. European Urology. 2023;83:S1667-S1667. DOI: 10.1016/s0302-2838(23)01192-2
  17. 17. Aylin A-D, James LM, Erin M, Justyna G, Han WT, John AC. Abstract 3041: ASPSCR1-TFE3 gene fusion in transgenic mouse as a new pediatric kidney cancer model. Tumor Biology. 2021;81(13_Supplement):3041-3041. DOI: 10.1158/1538-7445.am2021-3041
  18. 18. Aysegul B, Shinjini G, Petros G, et al. A novel orthotopic murine model of neuroendocrine bladder cancer: Insights into the phenotypic plasticity of small cell bladder cancer (SCBC). Journal of Clinical Oncology. 2020;38(6_suppl):571-571. DOI: 10.1200/jco.2020.38.6_suppl.571
  19. 19. Norman MG, Francesco JD, Milton JF, et al. Prostate cancer in a transgenic mouse. Proceedings of the National Academy of Sciences. 1995;92(8):3439-3443. DOI: 10.1073/pnas.92.8.3439
  20. 20. Larissa Akemi K, de Almeida Lamas C, Marostica MR, Cagnon VHA. Transgenic adenocarcinoma of the mouse prostate (TRAMP) model: A good alternative to study PCa progression and chemoprevention approaches. Life Sciences. 2019;217:141-147. DOI: 10.1016/j.lfs.2018.12.002
  21. 21. Irwin HG. How the TRAMP model revolutionized the study of prostate cancer progression. Cancer Research. 2016;76(21):6137-6139. DOI: 10.1158/0008-5472.Can-16-2636
  22. 22. Arthur AH, Foster BA, Allison JP, Greenberg NM, Kwon ED. The TRAMP mouse as a model for prostate cancer. Current Protocols in Immunology. 2001;Chapter 20:20.5.1-20.5.23. DOI: 10.1002/0471142735.im2005s45
  23. 23. Lisa DB-B, Karen EK. Models of neuroendocrine prostate cancer. Endocrine-Related Cancer. 2014;22(1):R33-R49. DOI: 10.1530/erc-14-0393
  24. 24. Chen R, Liang X, Murray MM, et al. A simple quantitative PCR assay to determine TRAMP transgene zygosity. Prostate Cancer and Prostatic Diseases. 2021;24(2):358-361. DOI: 10.1038/s41391-020-00282-4
  25. 25. Jeffrey RG, Roberto B, Ronald AM, et al. Metastatic prostate cancer in a transgenic mouse. Cancer Research. 1996;56(18):4096-4102
  26. 26. Teresa TL, Jonathan AE, Emily AR, Robert HB, Colin CC, William AR. Modeling human prostate cancer progression in vitro. Carcinogenesis. 2018;40(7):893-902. DOI: 10.1093/carcin/bgy185
  27. 27. Jinny S, Robert B, Justin Delos S, et al. Resistance to androgen deprivation leads to altered metabolism in human and murine prostate cancer cell and tumor models. Metabolites. 2021;11(3):139-139. DOI: 10.3390/metabo11030139
  28. 28. Susan K, Patricia S, Yuhao Y, et al. Development, progression, and androgen-dependence of prostate tumors in probasin-large T antigen transgenic mice: A model for prostate cancer. Laboratory Investigation; A Journal of Technical Methods and Pathology. 1998;78(6):i-xv
  29. 29. Olga K, John RC, Janni M, Richard LR, Robert JM, Valeri V. Hepsin promotes prostate cancer progression and metastasis. Cancer Cell. 2004;6(2):185-195. DOI: 10.1016/j.ccr.2004.07.008
  30. 30. Sirin S, Sachin Kumar D, Santanu D, Sachin P, Seema S, Ajay PS. Cellular and molecular progression of prostate cancer: Models for basic and preclinical research. Cancers. 2020;12(9):2651-2651. DOI: 10.3390/cancers12092651
  31. 31. Arriaga JM, Abate-Shen C. Genetically engineered mouse models of prostate cancer in the postgenomic era. Cold Spring Harbor Perspectives in Medicine. 2018;9(2):a030528-a030528. DOI: 10.1101/cshperspect.a030528
  32. 32. Susan K. Survey of genetically engineered mouse models for prostate cancer: Analyzing the molecular basis of prostate cancer development, progression, and metastasis. Journal of Cellular Biochemistry. 2005;94(2):279-297. DOI: 10.1002/jcb.20339
  33. 33. Maroulakou IG, Anver M, Garrett L, Green JE. Prostate and mammary adenocarcinoma in transgenic mice carrying a rat C3(1) simian virus 40 large tumor antigen fusion gene. Proceedings of the National Academy of Sciences of the United States of America. 1994;91(23):11236-11240. DOI: 10.1073/pnas.91.23.11236
  34. 34. Ingrid E, Michelle Y, Jennifer W, et al. A low-carbohydrate diet containing soy protein and fish oil reduces breast but not prostate cancer in C3(1)/tag mice. Carcinogenesis. 2021;43(2):115-125. DOI: 10.1093/carcin/bgab106
  35. 35. Nancy EM, Jennifer MT-A, Lei W, Krystle EZ, John WE, Steven KC. Tomatoes, lycopene, and prostate cancer: What have we learned from experimental models? Journal of Nutrition. 2022;152(6):1381-1403. DOI: 10.1093/jn/nxac066
  36. 36. Ioanna GM, Miriam RA, Lisa G, Jeffrey EG. Prostate and mammary adenocarcinoma in transgenic mice carrying a rat C3(1) simian virus 40 large tumor antigen fusion gene. Proceedings of the National Academy of Sciences of the United States of America. 1994;91(23):11236-11240. DOI: 10.1073/pnas.91.23.11236
  37. 37. Wenhao Z, Wytske M v W, Corrina MAdR, et al. Ex vivo treatment of prostate tumor tissue recapitulates in vivo therapy response. The Prostate. 2018;79(4):390-402. DOI: 10.1002/pros.23745
  38. 38. Emily G, Peter AH, Jeffrey IG. A transgenic mouse model of metastatic prostate cancer originating from neuroendocrine cells. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(26):15382-15387. DOI: 10.1073/pnas.95.26.15382
  39. 39. Kosuke O, Kei M, Tomohiro F, et al. Establishment and characterization of a novel treatment-related neuroendocrine prostate cancer cell line KUCaP13. Cancer Science. 2021;112(7):2781-2791. DOI: 10.1111/cas.14935
  40. 40. Sedlack AJH, Saleh-Anaraki K, Kumar S, et al. Preclinical models of neuroendocrine neoplasia. Cancers (Basel). 2022;14:22. DOI: 10.3390/cancers14225646
  41. 41. Tarana A, Peter SN. Endocrine and paracrine characteristics of neuroendocrine prostate cancer. Frontiers in Endocrinology. 2022;13. DOI: 10.3389/fendo.2022.1012005
  42. 42. Wenming D, Manal G, Madeleine M, et al. Knockin of SV40 tag oncogene in a mouse adenocarcinoma of the prostate model demonstrates advantageous features over the transgenic model. Oncogene. 2005;24(9):1510-1524. DOI: 10.1038/sj.onc.1208229
  43. 43. Manal G, Wenming D, Guojun W, et al. A novel knock-in prostate cancer model demonstrates biology similar to that of human prostate cancer and suitable for preclinical studies. Molecular Therapy. 2005;11(3):348-362. DOI: 10.1016/j.ymthe.2004.12.005
  44. 44. Wu G, Yu L-C, Wang L, et al. Application of Gleason analogous grading system and flow cytometry DNA analysis in a novel knock-in mouse prostate cancer model. Postgraduate Medical Journal. 2006;82(963):40-45. DOI: 10.1136/pgmj.2005.038042
  45. 45. Lee YR, Chen M, Pandolfi PP. The functions and regulation of the PTEN tumour suppressor: New modes and prospects. Nature Reviews. Molecular Cell Biology. 2018;19(9):547-562. DOI: 10.1038/s41580-018-0015-0
  46. 46. Maisa Y, Cutz JC, Nuin PA, et al. Interphase FISH analysis of PTEN in histologic sections shows genomic deletions in 68% of primary prostate cancer and 23% of high-grade prostatic intra-epithelial neoplasias. Cancer Genetics and Cytogenetics. 2006;169(2):128-137. DOI: 10.1016/j.cancergencyto.2006.04.003
  47. 47. Holcomb IN, Young JM, Coleman IM, et al. Comparative analyses of chromosome alterations in soft-tissue metastases within and across patients with castration-resistant prostate cancer. Cancer Research. 2009;69(19):7793-7802. DOI: 10.1158/0008-5472.Can-08-3810
  48. 48. Shunyou W, Gao J, Lei Q , et al. Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer Cell. 2003;4(3):209-221. DOI: 10.1016/s1535-6108(03)00215-0
  49. 49. Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP. Pten is essential for embryonic development and tumour suppression. Nature Genetics. 1998;19(4):348-355. DOI: 10.1038/1235
  50. 50. Podsypanina K, Ellenson LH, Nemes A, et al. Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(4):1563-1568. DOI: 10.1073/pnas.96.4.1563
  51. 51. Stambolic V, Tsao MS, Macpherson D, Suzuki A, Chapman WB, Mak TW. High incidence of breast and endometrial neoplasia resembling human Cowden syndrome in pten+/− mice. Cancer Research. 2000;60(13):3605-3611
  52. 52. Qingli L, Lunxu L, Zhang Q , Len S, Ge D, You Z. Interleukin-17 indirectly promotes M2 macrophage differentiation through stimulation of COX-2/PGE2 pathway in the cancer cells. Cancer Research and Treatment. 2014;46(3):297-306. DOI: 10.4143/crt.2014.46.3.297
  53. 53. Qiuyang Z, Liu S, Zhang Q , et al. Interleukin-17 promotes development of castration-resistant prostate cancer potentially through creating an immunotolerant and pro-angiogenic tumor microenvironment. The Prostate. 2014;74(8):869-879. DOI: 10.1002/pros.22805
  54. 54. Feng B, Pei XH, Pandolfi PP, Xiong Y. p18 Ink4c and Pten constrain a positive regulatory loop between cell growth and cell cycle control. Molecular and Cellular Biology. 2006;26(12):4564-4576. DOI: 10.1128/mcb.00266-06
  55. 55. Kim MJ, Cardiff RD, Desai N, et al. Cooperativity of Nkx3.1 and Pten loss of function in a mouse model of prostate carcinogenesis. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(5):2884-2889. DOI: 10.1073/pnas.042688999
  56. 56. Alvaro A, Antonina M, Carolyn Waugh K, et al. ETV4 promotes metastasis in response to activation of PI3-kinase and Ras signaling in a mouse model of advanced prostate cancer. Proceedings of the National Academy of Sciences. 2013;110(37). DOI: 10.1073/pnas.1303558110
  57. 57. Zhihu D, Chang Jiun W, Gerald CC, et al. SMAD4-dependent barrier constrains prostate cancer growth and metastatic progression. Nature. 2011;470(7333):269-273. DOI: 10.1038/nature09677
  58. 58. Jayaprakash P, Ai M, Liu A, et al. Targeted hypoxia reduction restores T cell infiltration and sensitizes prostate cancer to immunotherapy. The Journal of Clinical Investigation. 2018;128(11):5137-5149. DOI: 10.1172/jci96268
  59. 59. Yini Z, Zhao Y, Wen J, et al. Targeting the chromatin effector Pygo2 promotes cytotoxic T cell responses and overcomes immunotherapy resistance in prostate cancer. Science Immunology. 2023;8(81):eade4656. DOI: 10.1126/sciimmunol.ade4656
  60. 60. Mirjam B, Liu D, Brian DR, et al. SPOP mutation drives prostate tumorigenesis in vivo through coordinate regulation of PI3K/mTOR and AR signaling. Cancer Cell. 2017;31(3):436-451. DOI: 10.1016/j.ccell.2017.02.004
  61. 61. David JM, Kobayashi N, Marcus R, et al. Pten loss and RAS/MAPK activation cooperate to promote EMT and metastasis initiated from prostate cancer stem/progenitor cells. Cancer Research. 2012;72(7):1878-1889. DOI: 10.1158/0008-5472.can-11-3132
  62. 62. Bora G, Takashi I, Cheryl MK, et al. Nuclear MYC protein overexpression is an early alteration in human prostate carcinogenesis. Modern Pathology. 2008;21(9):1156-1167. DOI: 10.1038/modpathol.2008.111
  63. 63. Cheryl MK, Charles JB, Chi VD, William GN, Srinivasan Y, Angelo MDM. MYC and prostate cancer. Genes & Cancer. 2010;1(6):617-628. DOI: 10.1177/1947601910379132
  64. 64. Katharine E-Y, Thomas GG, John W, et al. Myc-driven murine prostate cancer shares molecular features with human prostate tumors. Cancer Cell. 2005;8(6):485-485. DOI: 10.1016/s1535-6108(03)00197-1
  65. 65. Gretchen KH, Laura NM, May K, et al. Combined MYC activation and Pten loss are sufficient to create genomic instability and lethal metastatic prostate cancer. Cancer Research. 2016;76(2):283-292. DOI: 10.1158/0008-5472.can-14-3280
  66. 66. McMullin RP, Mutton LN, Bieberich CJ. Hoxb13 regulatory elements mediate transgene expression during prostate organogenesis and carcinogenesis. Developmental Dynamics. 2009;238(3):664-672. DOI: 10.1002/dvdy.21870
  67. 67. Srinivas N, Katharine E-Y, Charles LS, et al. Hepsin cooperates with MYC in the progression of adenocarcinoma in a prostate cancer mouse model. The Prostate. 2009;70(6):591-600. DOI: 10.1002/pros.21093
  68. 68. Clegg NJ, Couto SS, Wongvipat J, et al. MYC cooperates with AKT in prostate tumorigenesis and alters sensitivity to mTOR inhibitors. PLoS One. 2011;6(3):e17449. DOI: 10.1371/journal.pone.0017449
  69. 69. William HF, Andre H, Robert DM, et al. Expression of the c-myc protooncogene in human prostatic carcinoma and benign prostatic hyperplasia. Cancer Research. 1986;46(3):1535-1538
  70. 70. Paul CB, Michael F, Nicholas JH, et al. Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nature Genetics. 2015;47(7):736-745. DOI: 10.1038/ng.3315
  71. 71. Himisha B, Davide P, Juan Miguel M, et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nature Medicine. 2016;22(3):298-305. DOI: 10.1038/nm.4045
  72. 72. Etienne D, Himisha B, Matteo B, et al. N-Myc induces an EZH2-mediated transcriptional program driving neuroendocrine prostate cancer. Cancer Cell. 2016;30(4):563-577. DOI: 10.1016/j.ccell.2016.09.005
  73. 73. Abdullah A, Mariantonietta DA, Martina T, et al. CDCP1 overexpression drives prostate cancer progression and can be targeted in vivo. Journal of Clinical Investigation. 2020;130(5):2435-2450. DOI: 10.1172/jci131133
  74. 74. Kuo CY, Hsieh PC, Chiu V, Lan CC, Lu KC. The von Hippel-Lindau tumor suppressor gene mutations modulate Lipocalin-2 expression in Ferroptotic-inflammatory pathways. Oxidative Medicine and Cellular Longevity. 2023;2023:7736638. DOI: 10.1155/2023/7736638
  75. 75. Marco G, Stuart H, James L, et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nature Genetics. 2014;46(3):225-233. DOI: 10.1038/ng.2891
  76. 76. Yusuke S, Tetsuichi Y, Yuichi S, et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nature Genetics. 2013;45(8):860-867. DOI: 10.1038/ng.2699
  77. 77. Erinn BR, John ET, Volker HH. Renal cyst development in mice with conditional inactivation of the von Hippel-Lindau tumor suppressor. Cancer Research. 2006;66(5):2576-2583. DOI: 10.1158/0008-5472.can-05-3241
  78. 78. Ian JF, Claudio RT, Strahil G, et al. pVHL and PTEN tumour suppressor proteins cooperatively suppress kidney cyst formation. The EMBO Journal. 2008;27(12):1747-1757. DOI: 10.1038/emboj.2008.96
  79. 79. Gunnar S, Bernd K, Johannes S, et al. Hypoxia-inducible transcription factors stabilization in the thick ascending limb protects against ischemic acute kidney injury. Journal of the American Society of Nephrology. 2011;22(11):2004-2015. DOI: 10.1681/asn.2010121249
  80. 80. Ruth S, Thomas H, Maxine T, et al. Renal tubular HIF-2α expression requires VHL inactivation and causes fibrosis and cysts. PLoS One. 2012;7(1):e31034-e31034. DOI: 10.1371/journal.pone.0031034
  81. 81. Tracy LP, Hannah LB, Joel MH, Tien H. Conditional inactivation of the mouse von Hippel–Lindau tumor suppressor gene results in wide-spread hyperplastic, inflammatory and fibrotic lesions in the kidney. Oncogene. 2014;34(20):2631-2639. DOI: 10.1038/onc.2014.197
  82. 82. Rafał P, Sarah MM, Tullio S, Wilhelm K, Holger M, Peter S. Loss of PBRM1 expression is associated with renal cell carcinoma progression. International Journal of Cancer. 2012;132(2). DOI: 10.1002/ijc.27822
  83. 83. Ignacio V, Patrick T, Keiran R, et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. 2011;469(7331):539-542. DOI: 10.1038/nature09639
  84. 84. Xun X, Yong H, Xue Y, et al. Single-cell exome sequencing reveals single-nucleotide mutation characteristics of a kidney tumor. Cell. 2012;148(5):886-895. DOI: 10.1016/j.cell.2012.02.025
  85. 85. Lucy G, Muhammed M, Andrew FS, et al. Clinical and pathological impact of VHL, PBRM1, BAP1, SETD2, KDM6A, and JARID1cin clear cell renal cell carcinoma. Genes, Chromosomes and Cancer. 2013;53(1):38-51. DOI: 10.1002/gcc.22116
  86. 86. Jeanette EE-P, Daniel JS, John CC, et al. BAP1 and PBRM1 in metastatic clear cell renal cell carcinoma: Tumor heterogeneity and concordance with paired primary tumor. BMC Urology. 2017;17(1). DOI: 10.1186/s12894-017-0209-3
  87. 87. Amrita MN, Can GP, Yiyu D, et al. The SWI/SNF protein PBRM1 restrains VHL-loss-driven clear cell renal cell carcinoma. Cell Reports. 2017;18(12):2893-2906. DOI: 10.1016/j.celrep.2017.02.074
  88. 88. Yi G, Shannon MC, Alana C, et al. Modeling renal cell carcinoma in mice: Bap1 and Pbrm1 inactivation drive tumor grade. Cancer Discovery. 2017;7(8):900-917. DOI: 10.1158/2159-8290.cd-17-0292
  89. 89. Shan, Shan W, Yi G, Nicholas CW, et al. Bap1 is essential for kidney function and cooperates with Vhl in renal tumorigenesis. Proceedings of the National Academy of Sciences. 2014;111(46):16538-16543. DOI: 10.1073/pnas.1414789111
  90. 90. James B. Molecular genetics of clear-cell renal cell carcinoma. Journal of Clinical Oncology. 2014;32(18):1968-1976. DOI: 10.1200/jco.2012.45.2003
  91. 91. Sabine H, Désirée S, Nora CT, et al. Combined mutation in Vhl, Trp53 and Rb1 causes clear cell renal cell carcinoma in mice. Nature Medicine. 2017;23(7):869-877. DOI: 10.1038/nm.4343
  92. 92. Sean TB, Aleisha MS, Jordan K, et al. MYC activation cooperates with Vhl and Ink4a/Arf loss to induce clear cell renal cell carcinoma. Nature Communications. 2017;8(1). DOI: 10.1038/ncomms15770
  93. 93. Michael LN, Michelle BW, Jorge RT, et al. Mutations in a novel gene lead to kidney tumors, lung wall defects, and benign tumors of the hair follicle in patients with the Birt-Hogg-Dubé syndrome. Cancer Cell. 2002;2(2):157-164. DOI: 10.1016/s1535-6108(02)00104-6
  94. 94. Birt AR. Hereditary multiple fibrofolliculomas with trichodiscomas and acrochordons. Archives of Dermatology. 1977;113(12):1674-1677. DOI: 10.1001/archderm.113.12.1674
  95. 95. Tiffiney RH, Emmanuelle N, Andres JK-S, et al. The role of the Birt–Hogg–Dubé protein in mTOR activation and renal tumorigenesis. Oncogene. 2009;28(13):1594-1604. DOI: 10.1038/onc.2009.14
  96. 96. Yukiko H, Baba M, Rieko A, et al. Homozygous loss of BHD causes early embryonic lethality and kidney tumor development with activation of mTORC1 and mTORC2. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(44):18722-18727. DOI: 10.1073/pnas.0908853106
  97. 97. Valérie H, Sylvie S, Anders Bondo D, et al. Renal tumour suppressor function of the Birt-Hogg-Dube syndrome gene product folliculin. Journal of Medical Genetics. 2009;47(3):182-189. DOI: 10.1136/jmg.2009.072009
  98. 98. Jindong C, Kunihiko F, David P, et al. Deficiency of FLCN in mouse kidney led to development of polycystic kidneys and renal neoplasia. PLoS One. 2008;3(10):e3581-e3581. DOI: 10.1371/journal.pone.0003581
  99. 99. Mingsong W, Shuhui S, Yan L, et al. Flcn-deficient renal cells are tumorigenic and sensitive to mTOR suppression. Oncotarget. 2015;6(32):32761-32773. DOI: 10.18632/oncotarget.5018
  100. 100. Jindong C, Dachuan H, Isabelle R, et al. Disruption of tubular Flcn expression as a mouse model for renal tumor induction. Kidney International. 2015;88(5):1057-1069. DOI: 10.1038/ki.2015.177
  101. 101. Zachary SM, Andrea IM. Aberrant epithelial morphology and persistent epidermal growth factor receptor signaling in a mouse model of renal carcinoma. Proceedings of the National Academy of Sciences. 2009;106(24):9767-9772. DOI: 10.1073/pnas.0902031106
  102. 102. Leiping F, Gang W, Maria MS, David MN, Lorraine JG. Generation of a mouse model of Von Hippel–Lindau kidney disease leading to renal cancers by expression of a constitutively active mutant of HIF1α. Cancer Research. 2011;71(21):6848-6856. DOI: 10.1158/0008-5472.can-11-1745
  103. 103. Alessia C, Lotte K, Eric HJV, et al. Modelling TFE renal cell carcinoma in mice reveals a critical role of WNT signaling. eLife. 2016;5. DOI: 10.7554/elife.17047
  104. 104. Qianghua H, Fei G, Weihua T, et al. Wt1 ablation and Igf2 upregulation in mice result in Wilms tumors with elevated ERK1/2 phosphorylation. Journal of Clinical Investigation. 2011;121(1):174-183. DOI: 10.1172/jci43772
  105. 105. Leo GF, Hung-Chieh Y, Suren S, et al. Monitoring therapy with MEK inhibitor U0126 in a novel Wilms tumor model in Wt1 knockout Igf2 transgenic mice using 18F-FDG PET with dual-contrast enhanced CT and MRI: Early metabolic response without inhibition of tumor growth. Molecular Imaging and Biology. 2012;15(2):175-185. DOI: 10.1007/s11307-012-0588-5
  106. 106. Wu XR. Biology of urothelial tumorigenesis: Insights from genetically engineered mice. Cancer Metastasis Reviews. 2009;28(3-4):281-290. DOI: 10.1007/s10555-009-9189-4
  107. 107. Regina A-R, Aura C, Rosário P-L, Paula AO. In vitro and in vivo experimental models as tools to investigate the efficacy of antineoplastic drugs on urinary bladder cancer. Anticancer Research. 2013;33(4):1273-1296
  108. 108. Zhong-Ting Z, Joanne P, Ellen S, Tung-Tien S, Xue-Ru W. Urothelium-specific expression of an oncogene in transgenic mice induced the formation of carcinoma in situ and invasive transitional cell carcinoma. Cancer Research. 1999;59(14):3512-3517
  109. 109. Ayala de la Peña F, Kanasaki K, Kanasaki M, Tangirala N, Maeda G, Kalluri R. Loss of p53 and acquisition of angiogenic microRNA profile are insufficient to facilitate progression of bladder urothelial carcinoma in situ to invasive carcinoma. The Journal of Biological Chemistry. 2011;286(23):20778-20787. DOI: 10.1074/jbc.M110.198069
  110. 110. Reddy EP, Roberta KR, Eugenio S, Mariano B. A point mutation is responsible for the acquisition of transforming properties by the T24 human bladder carcinoma oncogene. Nature. 1982;300(5888):149-152. DOI: 10.1038/300149a0
  111. 111. Zhong, Ting Z, Joanne P, Hong H, et al. Role of Ha-ras activation in superficial papillary pathway of urothelial tumor formation. Oncogene. 2001;20(16):1973-1980. DOI: 10.1038/sj.onc.1204315
  112. 112. Arnold JL. p53, the cellular gatekeeper for growth and division. Cell. 1997;88(3):323-331. DOI: 10.1016/s0092-8674(00)81871-1
  113. 113. Xue-Ru W. Urothelial tumorigenesis: A tale of divergent pathways. Nature Reviews Cancer. 2005;5(9):713-725. DOI: 10.1038/nrc1697
  114. 114. Jing G, Hong H, Joanne P, et al. p53 deficiency provokes urothelial proliferation and synergizes with activated Ha-ras in promoting urothelial tumorigenesis. Oncogene. 2004;23(3):687-696. DOI: 10.1038/sj.onc.1207169
  115. 115. Anna MP-K, Mireia C-M, Carolyn Waugh K, et al. Inactivation of p53 and Pten promotes invasive bladder cancer. Genes & Development. 2009;23(6):675-680. DOI: 10.1101/gad.1772909
  116. 116. Chao-Nan Q , Kyle AF, Jared K, et al. Activation of the PI3K/AKT pathway induces urothelial carcinoma of the renal pelvis: Identification in human tumors and confirmation in animal models. Cancer Research. 2009;69(21):8256-8264. DOI: 10.1158/0008-5472.can-09-1689
  117. 117. Kenneth AI. Germ cell neoplasms of the testis: Update for 2022. Seminars in Diagnostic Pathology. 2023;40(1):2-21. DOI: 10.1053/j.semdp.2022.07.001
  118. 118. Gill JA, Lowe L, Nguyen J, et al. Enforced expression of simian virus 40 large T-antigen leads to testicular germ cell tumors in zebrafish. Zebrafish. 2010;7(4):333-341. DOI: 10.1089/zeb.2010.0663
  119. 119. Leroy CS, Little CC. Spontaneous testicular teratomas in an inbred strain of mice. Proceedings of the National Academy of Sciences of the United States of America. 1954;40(11):1080-1087. DOI: 10.1073/pnas.40.11.1080
  120. 120. Leroy CS. Studies on transplantable testicular teratomas of strain 129 Mice2. Journal of the National Cancer Institute. 1958;20(6):1257-1275. DOI: 10.1093/jnci/20.6.1257
  121. 121. Leroy CS. A new inbred subline of mice (129/terSv) with a high incidence of spontaneous congenital testicular Teratomas2. Journal of the National Cancer Institute. 1973;50(1):235-242. DOI: 10.1093/jnci/50.1.235
  122. 122. Timothy MP, Amy ML, Claire A, et al. Chemotherapy-induced depletion of OCT4-positive cancer stem cells in a mouse model of malignant testicular cancer. Cell Reports. 2017;21(7):1896-1909. DOI: 10.1016/j.celrep.2017.10.078
  123. 123. Lyndaker AM, Pierpont TM, Loehr AR, Weiss RS. A genetically engineered mouse model of malignant testicular germ cell tumors. Methods in Molecular Biology. 2021;2195:147-165. DOI: 10.1007/978-1-0716-0860-9_11
  124. 124. Antoin D, Timothy AM. Updates on the epidemiology and risk factors for penile cancer. Translational Andrology and Urology. 2017;6(5):785-790. DOI: 10.21037/tau.2017.05.19
  125. 125. Catherine, de M, Martyn P, Jerome V, Silvia F. Worldwide burden of cancer attributable to HPV by site, country and HPV type. International Journal of Cancer. 2017;141(4):664-670. DOI: 10.1002/ijc.30716
  126. 126. Beatriz M-F, Verónica FM, Diogo E, et al. HPV16induces penile intraepithelial neoplasia and squamous cell carcinoma in transgenic mice: First mouse model for HPV-related penile cancer. The Journal of Pathology. 2020;251(4):411-419. DOI: 10.1002/path.5475
  127. 127. Martin AN, Frederick MA. Introduction to gene editing and manipulation using CRISPR/Cas9 technology. Current Protocols in Molecular Biology. 2016;115(1). DOI: 10.1002/cpmb.14
  128. 128. Hino T, Omura SN, Nakagawa R, et al. An AsCas12f-based compact genome-editing tool derived by deep mutational scanning and structural analysis. Cell. 2023;186(22):4920-4935 e23. DOI: 10.1016/j.cell.2023.08.031
  129. 129. Akiko T, Hiroyuki S, Tomoo O, et al. Inducing multiple nicks promotes interhomolog homologous recombination to correct heterozygous mutations in somatic cells. Nature Communications. 2023;14(1). DOI: 10.1038/s41467-023-41048-5
  130. 130. Minjung K, Rajula B-G, Whitney B-P, et al. Nkx3.1 mutant mice recapitulate early stages of prostate carcinogenesis. Cancer Research. 2002;62(11):2999-3004

Written By

Le Qu, Cheng Chen, Yaoming Li and Xin Pan

Submitted: 19 April 2024 Reviewed: 25 April 2024 Published: 22 May 2024