Vitamin A and Bladder Cancer: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Dasa Zupancic.

Bladder cancer (BC) is the tenth most common cancer worldwide with a high recurrence rate, morbidity and mortality. Therefore, chemoprevention and improved treatment of BC are of paramount importance. Epidemiological studies suggest that adequate vitamin A intake may be associated with reduced BC risk. In addition, retinoids, natural and synthetic derivatives of vitamin A, are intensively studied in cancer research due to their antioxidant properties and their ability to regulate cell growth, differentiation, and apoptosis. Findings from in vivo and in vitro models of BC show great potential for the use of retinoids in the chemoprevention and treatment of BC. However, translation to the clinical practice is limited.

  • bladder cancer
  • vitamin A
  • retinoids

1. Vitamin A and Bladder Cancer

Vitamin A is a generic term for a group of lipophilic isoprenoids consisting of a cyclic group and a linear chain with a hydrophilic polar group that includes the major biologically active forms retinol, retinal, and retinoic acid (RA) [1]. Since vitamin A cannot be synthesized in the human body, it must be obtained from the diet [2]. The importance of vitamin A for human health was already known to the ancient Egyptians around 1500–1800 B.C., although they did not know vitamin A as such. They recommended compressed animal livers for the treatment of night blindness or nyctalopia. Today we know that the liver is the richest source of vitamin A and that night blindness is caused by vitamin A deficiency (VAD) [3,4,5]. Rhodopsin with its covalently bound cofactor retinal is a major light-sensitive receptor protein involved in visual phototransduction and essential for normal vision. But the importance of vitamin A goes beyond visual health. Vitamin A is a regulator of cell growth and differentiation, embryogenesis, reproduction, epithelial cell integrity, and immune function [1,6,7]. In addition, it has antioxidant properties [8] and plays a role in protecting against oxidative stress damage and inflammation [1,9]. Recent data also indicate that vitamin A regulates the interactions between eukaryotic host cells and symbiotic microbes, as well as the complexity of the microbiome. On the other hand, the microbiome regulates vitamin A metabolism in the host [10,11].

Vitamin A is a generic term for a group of lipophilic isoprenoids consisting of a cyclic group and a linear chain with a hydrophilic polar group that includes the major biologically active forms retinol, retinal, and retinoic acid (RA) [1]. Since vitamin A cannot be synthesized in the human body, it must be obtained from the diet [2]. The importance of vitamin A for human health was already known to the ancient Egyptians around 1500–1800 B.C., although they did not know vitamin A as such. They recommended compressed animal livers for the treatment of night blindness or nyctalopia. Today we know that the liver is the richest source of vitamin A and that night blindness is caused by vitamin A deficiency (VAD) [3][4][5]. Rhodopsin with its covalently bound cofactor retinal is a major light-sensitive receptor protein involved in visual phototransduction and essential for normal vision. But the importance of vitamin A goes beyond visual health. Vitamin A is a regulator of cell growth and differentiation, embryogenesis, reproduction, epithelial cell integrity, and immune function [1][6][7]. In addition, it has antioxidant properties [8] and plays a role in protecting against oxidative stress damage and inflammation [1][9]. Recent data also indicate that vitamin A regulates the interactions between eukaryotic host cells and symbiotic microbes, as well as the complexity of the microbiome. On the other hand, the microbiome regulates vitamin A metabolism in the host [10][11].

Vitamin A belongs to the retinoids, a group of over 4000 molecules, which are natural and synthetic compounds that are structurally similar or share functional similarities [2,12,13]. Retinoids are classified into four generations based on the time of introduction and structural features: (i) first generation: retinol, retinaldehyde, all-trans RA (ATRA), tretinoin, isotretinoin; (ii) second generation: etretinate, acitretin; (iii) third generation: adapalene, tazarotene, bexarotene; (iv) fourth generation: seletinoid G [14]. The current use of retinoids in medicine is broad, especially in the field of skin health. For example, they are used for the treatment of various inflammatory and keratinization skin diseases (e.g., psoriasis, pityriasis rubra pilaris, lichen planus), as well as basal cell carcinoma [14]. Moreover, retinoids have been used successfully for the treatment of several other cancers, especially acute promyelocytic leukaemia in adults and neuroblastoma in children [15,16].

Vitamin A belongs to the retinoids, a group of over 4000 molecules, which are natural and synthetic compounds that are structurally similar or share functional similarities [2][12][13]. Retinoids are classified into four generations based on the time of introduction and structural features: (i) first generation: retinol, retinaldehyde, all-trans RA (ATRA), tretinoin, isotretinoin; (ii) second generation: etretinate, acitretin; (iii) third generation: adapalene, tazarotene, bexarotene; (iv) fourth generation: seletinoid G [14]. The current use of retinoids in medicine is broad, especially in the field of skin health. For example, they are used for the treatment of various inflammatory and keratinization skin diseases (e.g., psoriasis, pityriasis rubra pilaris, lichen planus), as well as basal cell carcinoma [14]. Moreover, retinoids have been used successfully for the treatment of several other cancers, especially acute promyelocytic leukaemia in adults and neuroblastoma in children [15][16].

Bladder cancer (BC), which usually arises from the urothelial cells, is one of the ten most common cancers worldwide. As it has a high recurrence rate of 50–70% and represents a huge social and economic burden [17,18,19], new prevention and treatment strategies are needed. Retinoids are among the best-studied chemopreventive agents for various diseases and are used in clinical practice for chemoprevention and treatment of several cancers [15,20]. Meta-analyses of epidemiological studies indicate that high dietary vitamin A intake reduces the risk of BC [21,22]. Several preclinical studies have shown great potential of retinoids for chemoprevention and treatment of BC, however, translation into clinical use remains limited due to application challenges. Nevertheless, novel synthetic retinoids and retinoid delivery systems have been developed, which, together with the discovery of novel therapeutic targets in the retinoid pathway, offer new opportunities for successful translation of retinoid application into the clinical setting.

Bladder cancer (BC), which usually arises from the urothelial cells, is one of the ten most common cancers worldwide. As it has a high recurrence rate of 50–70% and represents a huge social and economic burden [17][18][19], new prevention and treatment strategies are needed. Retinoids are among the best-studied chemopreventive agents for various diseases and are used in clinical practice for chemoprevention and treatment of several cancers [15][20]. Meta-analyses of epidemiological studies indicate that high dietary vitamin A intake reduces the risk of BC [21][22]. Several preclinical studies have shown great potential of retinoids for chemoprevention and treatment of BC, however, translation into clinical use remains limited due to application challenges. Nevertheless, novel synthetic retinoids and retinoid delivery systems have been developed, which, together with the discovery of novel therapeutic targets in the retinoid pathway, offer new opportunities for successful translation of retinoid application into the clinical setting.

2. The Role of Dietary Vitamin A in Bladder Cancer: The Epidemiologic Evidence

Vitamin A and retinoids are among the best-studied micronutrients and have great potential for prevention and cancer treatment due to their differentiating, antiproliferative, pro-apoptotic, and antioxidant effects combined with selectivity, high receptor binding affinity, and ability to directly modulate gene expression programs [15,53].

Vitamin A and retinoids are among the best-studied micronutrients and have great potential for prevention and cancer treatment due to their differentiating, antiproliferative, pro-apoptotic, and antioxidant effects combined with selectivity, high receptor binding affinity, and ability to directly modulate gene expression programs [15][23].

An association between VAD and the incidence of cancer was first demonstrated around 1920 in animal studies showing that VAD increased the incidence of spontaneous and carcinogen-induced tumours [54,55,56,57]. In 1979, a retrospective study of human dietary habits and BC showed an increased risk in people with low vitamin A intake [58], implicating vitamin A as a potential agent for BC prevention. Despite the fact that vitamin A is present in a wide variety of foods, many people do not consume this nutrient adequately due to malnutrition or selective diets, leading to VAD. Therefore, the impact of vitamin A intake on BC risk has important public health implications [21,59,60].

An association between VAD and the incidence of cancer was first demonstrated around 1920 in animal studies showing that VAD increased the incidence of spontaneous and carcinogen-induced tumours [24][25][26][27]. In 1979, a retrospective study of human dietary habits and BC showed an increased risk in people with low vitamin A intake [28], implicating vitamin A as a potential agent for BC prevention. Despite the fact that vitamin A is present in a wide variety of foods, many people do not consume this nutrient adequately due to malnutrition or selective diets, leading to VAD. Therefore, the impact of vitamin A intake on BC risk has important public health implications [21][29][30].

Typically, VAD develops in environments of ecological, social and economic deprivation. Recent analysis showed a decline in VAD prevalence primarily due to decrease in East and Southeast Asia, Oceania, Latin America and the Caribbean, while it remains high in South Asia and sub-Saharan Africa [61]. Moreover, we have to point out that Western diets containing mainly processed foods can lead to subclinical VAD, which often goes unnoticed but may be implicated in the development of some cancers [62].

Typically, VAD develops in environments of ecological, social and economic deprivation. Recent analysis showed a decline in VAD prevalence primarily due to decrease in East and Southeast Asia, Oceania, Latin America and the Caribbean, while it remains high in South Asia and sub-Saharan Africa [31]. Moreover, we have to point out that Western diets containing mainly processed foods can lead to subclinical VAD, which often goes unnoticed but may be implicated in the development of some cancers [32].

The highest rates of BC are observed in developed countries in Europe, Northern America, and Western Asia, but also in Syrian, Israeli, Egyptian and Turkish men. Approximately threefold lower rates are seen in Southeast Asia (except Japan) and in Latin America and Northern Africa in both sexes, and the lowest in Sub-Saharan Africa and some Middle Eastern and Central Asian countries [17,19,63,64].

The highest rates of BC are observed in developed countries in Europe, Northern America, and Western Asia, but also in Syrian, Israeli, Egyptian and Turkish men. Approximately threefold lower rates are seen in Southeast Asia (except Japan) and in Latin America and Northern Africa in both sexes, and the lowest in Sub-Saharan Africa and some Middle Eastern and Central Asian countries [17][19][33][34].

Looking at the global distribution of BC incidence and VAD, the association between the two is not immediately apparent. Nevertheless, numerous population-based epidemiological studies investigated the relationship between dietary vitamin A and BC risk, including several meta-analyses [21,22,65,66]. While older studies concluded that dietary retinol and β-carotene play a minimal role in BC [65], more recent studies show a preventive effect of vitamin A on BC. A meta-analysis of 25 studies investigating the quantitative effects of vitamin A on BC revealed that high vitamin A intake and high blood retinol levels were associated with a reduced risk of BC [21]. The most recent meta-analysis of 22 studies conducted in Northern America, Europe, or Japan (19 of which were included in the previous analysis by Tang et al. [21]) indicated that the risk of BC decreased by 76% for every 1 µmol/L increase in circulating concentrations of α-carotene, and by 27% for every 1 µmol/L increase in circulating concentrations of β-carotene. When comparing high and low total dietary carotenoid intake, high intake was associated with a 15% reduced risk of BC in men [22].

Looking at the global distribution of BC incidence and VAD, the association between the two is not immediately apparent. Nevertheless, numerous population-based epidemiological studies investigated the relationship between dietary vitamin A and BC risk, including several meta-analyses [21][22][35][36]. While older studies concluded that dietary retinol and β-carotene play a minimal role in BC [35], more recent studies show a preventive effect of vitamin A on BC. A meta-analysis of 25 studies investigating the quantitative effects of vitamin A on BC revealed that high vitamin A intake and high blood retinol levels were associated with a reduced risk of BC [21]. The most recent meta-analysis of 22 studies conducted in Northern America, Europe, or Japan (19 of which were included in the previous analysis by Tang et al. [21]) indicated that the risk of BC decreased by 76% for every 1 µmol/L increase in circulating concentrations of α-carotene, and by 27% for every 1 µmol/L increase in circulating concentrations of β-carotene. When comparing high and low total dietary carotenoid intake, high intake was associated with a 15% reduced risk of BC in men [22].

On the other hand, very high intakes of preformed vitamin A present in animal foods and pharmaceutical supplements can cause acute or chronic toxicity, while very high doses of provitamin A (carotenoids) from plants do not. Acute hypervitaminosis A is a consequence of the ingestion (usually accidental) of more than 300,000 IU of vitamin A as a single dose or several repeated doses over a few days, whereas chronic hypervitaminosis A is a result of continued ingestion of more than 100,000 IU daily for months or years [67]. In addition, a single dose of more than 25,000 IU of vitamin A may be teratogenic if consumed between the 15th and 60th day after conception [68].

On the other hand, very high intakes of preformed vitamin A present in animal foods and pharmaceutical supplements can cause acute or chronic toxicity, while very high doses of provitamin A (carotenoids) from plants do not. Acute hypervitaminosis A is a consequence of the ingestion (usually accidental) of more than 300,000 IU of vitamin A as a single dose or several repeated doses over a few days, whereas chronic hypervitaminosis A is a result of continued ingestion of more than 100,000 IU daily for months or years [37]. In addition, a single dose of more than 25,000 IU of vitamin A may be teratogenic if consumed between the 15th and 60th day after conception [38].

Although the evidence for the correlation between BC aetiology and diet are not yet conclusive, diet is considered one of the modifiable risk factors for BC prevention [69,70]. There is still a large gap to be filled in understanding the molecular mechanisms by which vitamin A affects urothelium and urothelial carcinogenesis. To address this issue, various in vivo and in vitro models mimicking human BC have been widely used.

Although the evidence for the correlation between BC aetiology and diet are not yet conclusive, diet is considered one of the modifiable risk factors for BC prevention [39][40]. There is still a large gap to be filled in understanding the molecular mechanisms by which vitamin A affects urothelium and urothelial carcinogenesis. To address this issue, various in vivo and in vitro models mimicking human BC have been widely used.

3. Experimental Models of Bladder Cancer Play a Key Role in Understanding the Chemopreventive and Therapeutic Effects of Vitamin A and Retinoids

Several retinoids, such as ATRA, 13-cis-RA, and N-(4-hydroxyphenyl)-retinamide (4-HPR, or fenretinide), showed promising chemopreventive effects on BC both in vitro and in vivo (

Table 1

 and 

Table 2, respectively). In vitro studies suggest that retinoids exert their chemopreventive effects on BC through cytostatic, pro-apoptotic, growth inhibitory, cell cycle distribution, and gene expression modulating/regulating functions [71,72,73,74,75,76]. The study by Boström et al. suggested that retinoids may downregulate the expression of matrix metalloproteinases (MMPs), which play an important role in the process of degradation of extracellular matrix essential for tumour growth and invasion [77]. The mechanism of retinoid BC chemoprevention may also include reversion of epithelial-mesenchymal transition, a key process in cancer cell invasion and migration. Wang et al. showed that the synthetic retinoid 4-HPR increased the expression of E-cadherin in invasive BC cell lines and induced the translocation of β-catenin from the nucleus to the cytoplasm, resulting in an altered BC cell morphology that resembles epithelial rather than invasive cancer cells, presumably leading to reduced cell infiltration [78].
 

, respectively). In vitro studies suggest that retinoids exert their chemopreventive effects on BC through cytostatic, pro-apoptotic, growth inhibitory, cell cycle distribution, and gene expression modulating/regulating functions [41][42][43][44][45][46]. The study by Boström et al. suggested that retinoids may downregulate the expression of matrix metalloproteinases (MMPs), which play an important role in the process of degradation of extracellular matrix essential for tumour growth and invasion [47]. The mechanism of retinoid BC chemoprevention may also include reversion of epithelial-mesenchymal transition, a key process in cancer cell invasion and migration. Wang et al. showed that the synthetic retinoid 4-HPR increased the expression of E-cadherin in invasive BC cell lines and induced the translocation of β-catenin from the nucleus to the cytoplasm, resulting in an altered BC cell morphology that resembles epithelial rather than invasive cancer cells, presumably leading to reduced cell infiltration [48].

Table 1.

 In vitro studies investigating the effects of retinoids in human BC cell lines.

References

  1. Huang, Z.; Liu, Y.; Qi, G.; Brand, D.; Zheng, S.G. Role of Vitamin A in the Immune System. J. Clin. Med. 2018, 7, 258.
  2. Bushue, N.; Wan, Y.J. Retinoid pathway and cancer therapeutics. Adv. Drug Deliv. Rev. 2010, 62, 1285–1298.
  3. Al Binali, H.A. Night blindness and ancient remedy. Heart Views 2014, 15, 136–139.
  4. Wolf, G. A history of vitamin A and retinoids. FASEB J. 1996, 10, 1102–1107.
  5. Maumenee, A.E. The history of vitamin A and its ophthalmic implications. A personal viewpoint. Arch. Ophthalmol. 1993, 111, 547–550.
  6. Clagett-Dame, M.; Knutson, D. Vitamin A in reproduction and development. Nutrients 2011, 3, 385–428.
  7. Zhu, G. Vitamin A and its Derivatives-Retinoic Acid and Retinoid Pharmacology. Am. J. Biomed. Sci. Res. 2019, 3, 162–177.
  8. Dao, D.Q.; Ngo, T.C.; Thong, N.M.; Nam, P.C. Is Vitamin A an Antioxidant or a Pro-oxidant? J. Phys. Chem. B 2017, 121, 9348–9357.
  9. Siddikuzzaman; Grace, V.M. Antioxidant potential of all-trans retinoic acid (ATRA) and enhanced activity of liposome encapsulated ATRA against inflammation and tumor-directed angiogenesis. Immunopharmacol. Immunotoxicol. 2013, 35, 164–173.
  10. Iyer, N.; Vaishnava, S. Vitamin A at the interface of host-commensal-pathogen interactions. PLoS Pathog. 2019, 15, e1007750.
  11. Cantorna, M.T.; Snyder, L.; Arora, J. Vitamin A and vitamin D regulate the microbial complexity, barrier function, and the mucosal immune responses to ensure intestinal homeostasis. Crit. Rev. Biochem. Mol. Biol. 2019, 54, 184–192.
  12. Polcz, M.E.; Barbul, A. The Role of Vitamin A in Wound Healing. Nutr. Clin. Pract. 2019, 34, 695–700.
  13. Zasada, M.; Budzisz, E. Retinoids: Active molecules influencing skin structure formation in cosmetic and dermatological treatments. Postepy. Dermatol. Alergol. 2019, 36, 392–397.
  14. Dattola, A.; Silvestri, M.; Bennardo, L.; Passante, M.; Scali, E.; Patruno, C.; Nisticò, S.P. Role of Vitamins in Skin Health: A Systematic Review. Curr. Nutr. Rep. 2020, 9, 226–235.
  15. Dobrotkova, V.; Chlapek, P.; Mazanek, P.; Sterba, J.; Veselska, R. Traffic lights for retinoids in oncology: Molecular markers of retinoid resistance and sensitivity and their use in the management of cancer differentiation therapy. BMC Cancer 2018, 18, 1059.
  16. Chlapek, P.; Slavikova, V.; Mazanek, P.; Sterba, J.; Veselska, R. Why Differentiation Therapy Sometimes Fails: Molecular Mechanisms of Resistance to Retinoids. Int. J. Mol. Sci. 2018, 19, 132.
  17. Richters, A.; Aben, K.K.H.; Kiemeney, L. The global burden of urinary bladder cancer: An update. World J. Urol. 2020, 38, 1895–1904.
  18. Sanli, O.; Dobruch, J.; Knowles, M.A.; Burger, M.; Alemozaffar, M.; Nielsen, M.E.; Lotan, Y. Bladder cancer. Nat. Rev. Dis. Primers 2017, 3, 17022.
  19. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424.
  20. Ferreira, R.; Napoli, J.; Enver, T.; Bernardino, L.; Ferreira, L. Advances and challenges in retinoid delivery systems in regenerative and therapeutic medicine. Nat. Commun. 2020, 11, 4265.
  21. Tang, J.E.; Wang, R.J.; Zhong, H.; Yu, B.; Chen, Y. Vitamin A and risk of bladder cancer: A meta-analysis of epidemiological studies. World J. Surg. Oncol. 2014, 12, 130.
  22. Wu, S.; Liu, Y.; Michalek, J.E.; Mesa, R.A.; Parma, D.L.; Rodriguez, R.; Mansour, A.M.; Svatek, R.; Tucker, T.C.; Ramirez, A.G. Carotenoid Intake and Circulating Carotenoids Are Inversely Associated with the Risk of Bladder Cancer: A Dose-Response Meta-analysis. Adv. Nutr. 2020, 11, 630–643.
  23. Uray, I.P.; Dmitrovsky, E.; Brown, P.H. Retinoids and rexinoids in cancer prevention: From laboratory to clinic. Semin. Oncol. 2016, 43, 49–64.
  24. Mori, S. The changes in the paraocular glands which follow the administration of diets low in fat-soluble vitamin A with notes of the effects of the same diets on the salivary glands and the mucosa of the larynx and brachea. John Hopkins Hospital. Bull. 1922, 33, 357–359.
  25. Wolbach, S.B.; Howe, P.R. Tissue changes following deprivation of fat-soluble A vitamin. J. Exp. Med. 1925, 42, 753–777.
  26. Wolbach, S.B.; Howe, P.R. Vitamin A Deficiency in the Guineapig. Arch. Path. Lab. Med. 1928, 5, 239–253.
  27. Fujimaki, Y. Formation of gastric carcinoma in albino rats fed on deficient diets. J. Cancer Res. 1926, 10, 469–477.
  28. Mettlin, C.; Graham, S. Dietary risk factors in human bladder cancer. Am. J. Epidemiol. 1979, 110, 255–263.
  29. Martini, S.; Rizzello, A.; Corsini, I.; Romanin, B.; Fiorentino, M.; Grandi, S.; Bergamaschi, R. Vitamin A Deficiency Due to Selective Eating as a Cause of Blindness in a High-Income Setting. Pediatrics 2018, 141, S439–S444.
  30. Gröber, U. Vitamin A (Retinol): Stiefkind der Ernährungsmedizin. Erfahrungsheilkunde 2020, 69, 334–339.
  31. Stevens, G.A.; Bennett, J.E.; Hennocq, Q.; Lu, Y.; De-Regil, L.M.; Rogers, L.; Danaei, G.; Li, G.; White, R.A.; Flaxman, S.R.; et al. Trends and mortality effects of vitamin A deficiency in children in 138 low-income and middle-income countries between 1991 and 2013: A pooled analysis of population-based surveys. Lancet Glob. Health 2015, 3, e528–e536.
  32. Surman, S.L.; Penkert, R.R.; Sealy, R.E.; Jones, B.G.; Marion, T.N.; Vogel, P.; Hurwitz, J.L. Consequences of Vitamin A Deficiency: Immunoglobulin Dysregulation, Squamous Cell Metaplasia, Infectious Disease, and Death. Int. J. Mol. Sci. 2020, 21, 5570.
  33. Antoni, S.; Ferlay, J.; Soerjomataram, I.; Znaor, A.; Jemal, A.; Bray, F. Bladder Cancer Incidence and Mortality: A Global Overview and Recent Trends. Eur. Urol. 2017, 71, 96–108.
  34. He, H.; Xie, H.; Chen, Y.; Li, C.; Han, D.; Xu, F.; Lyu, J. Global, regional, and national burdens of bladder cancer in 2017: Estimates from the 2017 global burden of disease study. BMC Public Health 2020, 20, 1693.
  35. Steinmaus, C.M.; Nuñez, S.; Smith, A.H. Diet and bladder cancer: A meta-analysis of six dietary variables. Am. J. Epidemiol. 2000, 151, 693–702.
  36. Luo, X.; Lu, H.; Li, Y.; Wang, S. Carrot intake and incidence of urothelial cancer: A systematic review and meta-analysis. Oncotarget 2017, 8, 77957–77962.
  37. Penniston, K.L.; Tanumihardjo, S.A. The acute and chronic toxic effects of vitamin A. Am. J. Clin. Nutr. 2006, 83, 191–201.
  38. WHO. Guidelines Approved by the Guidelines Review Committee. In Guideline: Vitamin A Supplementation in Pregnant Women; WHO: Geneva, Switzerland, 2011.
  39. Piyathilake, C. Dietary factors associated with bladder cancer. Investig. Clin. Urol. 2016, 57, S14–S25.
  40. Al-Zalabani, A.H.; Stewart, K.F.; Wesselius, A.; Schols, A.M.; Zeegers, M.P. Modifiable risk factors for the prevention of bladder cancer: A systematic review of meta-analyses. Eur. J. Epidemiol. 2016, 31, 811–851.
  41. Clifford, J.L.; Sabichi, A.L.; Zou, C.; Yang, X.; Steele, V.E.; Kelloff, G.J.; Lotan, R.; Lippman, S.M. Effects of novel phenylretinamides on cell growth and apoptosis in bladder cancer. Cancer Epidemiol. Biomark. Prev. 2001, 10, 391–395.
  42. Nutting, C.; Chowaniec, J. Evaluation of the actions and interactions of retinoic acid and epidermal growth factor on transformed urothelial cells in culture: Implications for the use of retinoid therapy in the treatment of bladder cancer patients. Clin. Oncol. 1992, 4, 51–55.
  43. Laaksovirta, S.; Rajala, P.; Nurmi, M.; Tammela, T.; Laato, M. The cytostatic effect of 9-cis-retinoic acid, tretinoin, and isotretinoin on three different human bladder cancer cell lines in vitro. Urol. Res. 1999, 27, 17–22.
  44. Zou, C.; Zhou, J.; Qian, L.; Feugang, J.M.; Liu, J.; Wang, X.; Wu, S.; Ding, H.; Zou, C.; Liebert, M. Comparing the effect of ATRA, 4-HPR, and CD437 in bladder cancer cells. Front. Biosci. 2006, 11, 2007–2016.
  45. Chien, C.-L.; Chen, T.-W.; Lin, Y.-S.; Lu, K.-S. The apoptotic process of human bladder carcinoma T24 cells induced by retinoid. J. Biomed. Sci. 2004, 11, 631–640.
  46. Southgate, J.; Hutton, K.; Thomas, D.; Trejdosiewicz, L.K. Normal human urothelial cells in vitro: Proliferation and induction of stratification. Lab. Investig. 1994, 71, 583.
  47. Boström, P.J.; Ravanti, L.; Reunanen, N.; Aaltonen, V.; Söderström, K.O.; Kähäri, V.M.; Laato, M. Expression of collagenase-3 (matrix metalloproteinase-13) in transitional-cell carcinoma of the urinary bladder. Int. J. Cancer 2000, 88, 417–423.
  48. Wang, E.; Li, J.; Yang, G.; Zhong, S.; Liu, T. Impact of 4HPR on the expression of E-Cad in human bladder transitional epithelial cancer cells T24. Acta Acad. Med. Wuhan 2012, 32, 237–241.
  49. Lin, F.; Kolluri, S.K.; Chen, G.-q.; Zhang, X.-k. Regulation of retinoic acid-induced inhibition of AP-1 activity by orphan receptor chicken ovalbumin upstream promoter-transcription factor. J. Biol. Chem. 2002, 277, 21414–21422.
  50. Costantini, L.; Molinari, R.; Farinon, B.; Lelli, V.; Timperio, A.M.; Merendino, N. Docosahexaenoic Acid Reverted the All-trans Retinoic Acid-Induced Cellular Proliferation of T24 Bladder Cancer Cell Line. J. Clin. Med. 2020, 9, 2494.
  51. Zhiping, W.; Zhihua, Z.; Yinmei, L.; Yirong, C.; Qinxi, L.; Dashan, Q.; Guodong, L.; Liufang, W. Effect of retinoic acid and its complexes with transition metals on human bladder cancer cell line EJ in vitro. Urol. Res. 2000, 28, 191–195.
  52. Zou, C.; Liebert, M.; Zou, C.; Grossman, H.B.; Lotan, R. Identification of effective retinoids for inhibiting growth and inducing apoptosis in bladder cancer cells. J. Urol. 2001, 165, 986–992.
  53. Lubet, R.A.; Clapper, M.L.; McCormick, D.L.; Pereira, M.A.; Chang, W.; Steele, V.E.; Fischer, S.M.; Juliana, M.M.; Grubbs, C.J. Chemopreventive efficacy of Targretin in rodent models of urinary bladder, colon/intestine, head and neck and mammary cancers. Oncol. Rep. 2012, 27, 1400–1406.
  54. Murasaki, G.i.; Miyata, Y.; Babaya, K.; Arai, M.; Fukushima, S.; Ito, N. Inhibitory effect of an aromatic retinoic acid analog on urinary bladder carcinogenesis in rats treated with N-butyl-N-(4-hydroxybutyl) nitrosamine. GANN 1980, 71, 333–340.
  55. Fujita, J.; Tokuda, H.; Ito, Y.; Yoshida, O. Therapeutic effect of a retinoid (Ro 10-9359) on rats with bladder tumours induced by N-butyl-N-(4-hydroxybutyl)-nitrosamine upon administration alone or in combination with mitomycin C. Urol. Res. 1983, 11, 227–230.
  56. Zupančič, D.; Korać-Prlić, J.; Kreft, M.E.; Franković, L.; Vilović, K.; Jeruc, J.; Romih, R.; Terzić, J. Vitamin A Rich Diet Diminishes Early Urothelial Carcinogenesis by Altering Retinoic Acid Signaling. Cancers 2020, 12, 1712.
  57. Dawson, W.; Miller, W.; Liles, W. Retinyl acetate prophylaxis in cancer of the urinary bladder. Investig. Urol. 1979, 16, 376–377.
  58. Hemstreet III, G.P.; Rao, J.Y.; Hurst, R.E.; Bonner, R.B.; Jones, P.L.; Vaidya, A.M.; Fradet, Y.; Moon, R.C.; Kelloff, G.J. Intermediate endpoint biomarkers for chemoprevention. J. Cell Biochem. 1992, 50, 93–110.
  59. Qian, L.; Ding, H.; Zhou, J.; Wang, X.; Shao, P.; Wu, S.; Yang, J.; Feugang, J.M.; Zou, C. Intravesical N-(4-hydroxyphenyl) retinamide and adriamycin induces apoptosis in bladder cancer. Front. Biosci. 2006, 11, 2045–2051.
  60. Grubbs, C.J.; Moon, R.C.; Squire, R.A.; Farrow, G.M.; Stinson, S.F.; Goodman, D.G.; Brown, C.C.; Sporn, M.B. 13-cis-Retinoic acid: Inhibition of bladder carcinogenesis induced in rats by N-butyl-N-(4-hydroxybutyl) nitrosamine. Science 1977, 198, 743–744.
  61. Becci, P.J.; Thompson, H.J.; Grubbs, C.J.; Brown, C.C.; Moon, R.C. Effect of delay in administration of 13-cis-retinoic acid on the inhibition of urinary bladder carcinogenesis in the rat. Cancer Res. 1979, 39, 3141–3144.
  62. Becci, P.J.; Thompson, H.J.; Grubbs, C.J.; Squire, R.A.; Brown, C.C.; Sporn, M.B.; Moon, R.C. Inhibitory effect of 13-cis-retinoic acid on urinary bladder carcinogenesis induced in C57BL/6 mice by N-butyl-N-(4-hydroxybutyl) nitrosamine. Cancer Res. 1978, 38, 4463–4466.
  63. Becci, P.J.; Thompson, H.J.; Strum, J.M.; Brown, C.C.; Sporn, M.B.; Moon, R.C. N-butyl-N-(4-hydroxybutyl) nitrosamine-induced urinary bladder cancer in C57BL/6× DBA/2 F1 mice as a useful model for study of chemoprevention of cancer with retinoids. Cancer Res. 1981, 41, 927–932.
  64. Sporn, M.B.; Squire, R.A.; Brown, C.C.; Smith, J.M.; Wenk, M.L.; Springer, S. 13-cis-retinoic acid: Inhibition of bladder carcinogenesis in the rat. Science 1977, 195, 487–489.
  65. Squire, R.A.; Sporn, M.B.; Brown, C.C.; Smith, J.M.; Wenk, M.L.; Springer, S. Histopathological evaluation of the inhibition of rat bladder carcinogenesis by 13-cis-retinoic acid. Cancer Res. 1977, 37, 2930–2936.
  66. Tannenbaum, M.; Tannenbaum, S.; Richelo, B.; Trown, P. Effects of 13-cis and all-trans-retinoic acid on the development of bladder cancer in rats: An ultrastructural study. Scan. Electron. Microsc. 1979, 3, 673–678.
  67. Thompson, H.J.; Becci, P.J.; Grubbs, C.J.; Shealy, Y.F.; Stanek, E.J.; Brown, C.C.; Sporn, M.B.; Moon, R.C. Inhibition of urinary bladder cancer by N-(ethyl)-all-trans-retinamide and N-(2-hydroxyethyl)-all-trans-retinamide in rats and mice. Cancer Res. 1981, 41, 933–936.
  68. Croft, W.A.; Croft, M.A.; Paulus, K.P.; Williams, J.H.; Wang, C.Y.; Lower, G.M., Jr. Synthetic retinamides: Effect on urinary bladder carcinogenesis by FANFT in Fischer rats. Carcinogenesis 1981, 2, 515–517.
  69. Croft, W.A.; Croft, M.A.; Paulus, K.P.; Williams, J.H.; Wang, C.Y.; Lower, G.M., Jr. 13-cis-retinoic acid: Effect on urinary bladder carcinogenesis by N-[4-(5-nitro-2-furyl)-2-thiazolyl]-formamide in Fischer rats. Cancer Lett. 1981, 12, 355–360.
  70. Kobayashi, T.; Owczarek, T.B.; McKiernan, J.M.; Abate-Shen, C. Modelling bladder cancer in mice: Opportunities and challenges. Nar. Rev. Cancer 2015, 15, 42–54.
  71. John, B.A.; Said, N. Insights from animal models of bladder cancer: Recent advances, challenges, and opportunities. Oncotarget 2017, 8, 57766.
  72. He, Z.; Kosinska, W.; Zhao, Z.-L.; Wu, X.-R.; Guttenplan, J.B. Tissue-specific mutagenesis by N-butyl-N-(4-hydroxybutyl)nitrosamine as the basis for urothelial carcinogenesis. Mutat. Res. 2012, 742, 92–95.
  73. Nagao, M.; Suzuki, E.; Yasuo, K.; Yahagi, T.; Seino, Y. Mutagenicity of N-butyl-N-(4-hydroxybutyl)nitrosamine, a bladder carcinogen, and related compounds. Cancer Res. 1977, 37, 399–407.
  74. Ariel, I.; Ayesh, S.; Gofrit, O.; Ayesh, B.; Abdul-Ghani, R.; Pizov, G.; Smith, Y.; Sidi, A.A.; Birman, T.; Schneider, T.; et al. Gene expression in the bladder carcinoma rat model. Mol. Carcinog. 2004, 41, 69–76.
  75. Kates, M.; Nirschl, T.; Sopko, N.A.; Matsui, H.; Kochel, C.M.; Reis, L.O.; Netto, G.J.; Hoque, M.O.; Hahn, N.M.; McConkey, D.J. Intravesical BCG induces CD4+ T-cell expansion in an immune competent model of bladder cancer. Cancer Immunol. Res. 2017, 5, 594–603.
  76. Chopra, B.; Hinley, J.; Oleksiewicz, M.B.; Southgate, J. Trans-species comparison of PPAR and RXR expression by rat and human urothelial tissues. Toxicol. Pathol. 2008, 36, 485–495.
  77. Sanz, M.A.; Fenaux, P.; Tallman, M.S.; Estey, E.H.; Löwenberg, B.; Naoe, T.; Lengfelder, E.; Döhner, H.; Burnett, A.K.; Chen, S.-J. Management of acute promyelocytic leukemia: Updated recommendations from an expert panel of the European LeukemiaNet. Blood 2019, 133, 1630–1643.
  78. Villablanca, J.G.; Khan, A.A.; Avramis, V.I.; Seeger, R.C.; Matthay, K.K.; Ramsay, N.; Reynolds, C.P. Phase I trial of 13-cis-retinoic acid in children with neuroblastoma following bone marrow transplantation. J. Clin. Oncol. 1995, 13, 894–901.
  79. Decensi, A.; Bruno, S.; Costantini, M.; Torrisi, R.; Curotto, A.; Gatteschi, B.; Nicolò, G.; Polizzi, A.; Perloff, M.; Malone, W.F.; et al. Phase IIa study of fenretinide in superficial bladder cancer, using DNA flow cytometry as an intermediate end point. J. Natl. Cancer Inst. 1994, 86, 138–140.
  80. Decensi, A.; Torrisi, R.; Bruno, S.; Costantini, M.; Curotto, A.; Nicolò, G.; Malcangi, B.; Baglietto, L.; Bruttini, G.P.; Gatteschi, B.; et al. Randomized trial of fenretinide in superficial bladder cancer using DNA flow cytometry as an intermediate end point. Cancer Epidemiol. Biomarkers Prev. 2000, 9, 1071–1078.
  81. Puntoni, M.; Petrera, M.; Campora, S.; Garrone, E.; Defferrari, C.; Torrisi, R.; Johansson, H.; Bruno, S.; Curotto, A.; DeCensi, A. Prognostic Significance of VEGF after Twenty-Year Follow-up in a Randomized Trial of Fenretinide in Non–Muscle-Invasive Bladder Cancer. Cancer Prev. Res. 2016, 9, 437–444.
  82. Torrisi, R.; Mezzetti, M.; Johansson, H.; Barreca, A.; Pigatto, F.; Robertson, C.; Decensi, A. Time course of fenretinide-induced modulation of circulating insulin-like growth factor (IGF)-i, IGF-II and IGFBP-3 in a bladder cancer chemoprevention trial. Int. J. Cancer. 2000, 87, 601–605.
  83. Sabichi, A.L.; Lerner, S.P.; Atkinson, E.N.; Grossman, H.B.; Caraway, N.P.; Dinney, C.P.; Penson, D.F.; Matin, S.; Kamat, A.; Pisters, L.L. Phase III Prevention Trial of Fenretinide in Patients with Resected Non–Muscle-Invasive Bladder Cancer. Clin. Cancer Res. 2008, 14, 224–229.
  84. Alfthan, O.; Tarkkanen, J.; Gröhn, P.; Heinonen, E.; Pyrhönen, S. Tigason®(etretinate) in prevention of recurrence of superficial bladder tumors. Eur. Urol. 1983, 9, 6–9.
  85. Studer, U.E.; Jenzer, S.; Biederman, C.; Chollet, D.; Rainer, K.; von Toggenburg, H.; Vonbank, F. Adjuvant treatment with a vitamin A analogue (etretinate) after transurethral resection of superficial bladder tumors. Eur. Urol. 1995, 28, 284–290.
  86. Pedersen, H.; Wolf, H.; Jensen, S.K.; Lund, F.; Hansen, E.; Olsen, P.R.; Sørensen, B.L. Administration of a retinoid as prophylaxis of recurrent non-invasive bladder tumors. Scand. J. Urol. Nephrol. 1984, 18, 121–123.
  87. Prout, G.R., Jr.; Barton, B.A.; Kontz, W., Jr.; Loening, S.; Flangan, M.; Branen, G. 13-cis-Retinoic acid in chemopreventiion of superficial bladder cancer. J. Cell Biochem. 1992, 50, 148–152.
  88. Hameed, D.A.; el-Metwally, T.H. The effectiveness of retinoic acid treatment in bladder cancer: Impact on recurrence, survival and TGFalpha and VEGF as end-point biomarkers. Cancer Biol. Ther. 2008, 7, 92–100.
  89. Pili, R.; Salumbides, B.; Zhao, M.; Altiok, S.; Qian, D.; Zwiebel, J.; Carducci, M.A.; Rudek, M.A. Phase I study of the histone deacetylase inhibitor entinostat in combination with 13-cis retinoic acid in patients with solid tumours. Br. J. Cancer 2012, 106, 77–84.
  90. Sabichi, A.L.; Lerner, S.P.; Grossman, H.B.; Lippman, S.M. Retinoids in the chemoprevention of bladder cancer. Curr. Opin. Oncol 1998, 10, 479–484.
  91. Lu, C.S.; Shieh, G.S.; Wang, C.T.; Su, B.H.; Su, Y.C.; Chen, Y.C.; Su, W.C.; Wu, P.; Yang, W.H.; Shiau, A.L.; et al. Chemotherapeutics-induced Oct4 expression contributes to drug resistance and tumor recurrence in bladder cancer. Oncotarget 2016, 8, 30844–30858.
  92. Ozgun, G.; Senturk, S.; Erkek-Ozhan, S. Retinoic acid signaling and bladder cancer: Epigenetic deregulation, therapy and beyond. Int. J. Cancer 2020.
  93. Chen, J.; Cao, X.; An, Q.; Zhang, Y.; Li, K.; Yao, W.; Shi, F.; Pan, Y.; Jia, Q.; Zhou, W.; et al. Inhibition of cancer stem cell like cells by a synthetic retinoid. Nat. Commun. 2018, 9, 1406.
  94. Ye, L.; Song, Q. Promising potency of retinoic acid-poly(ethylene glycol)-thiol gold nanoparticle conjugates for cervical cancer treatment. Int. J. Clin. Exp. Med. 2015, 8, 10501–10507.
  95. Orienti, I.; Francescangeli, F.; De Angelis, M.L.; Fecchi, K.; Bongiorno-Borbone, L.; Signore, M.; Peschiaroli, A.; Boe, A.; Bruselles, A.; Costantino, A.; et al. A new bioavailable fenretinide formulation with antiproliferative, antimetabolic, and cytotoxic effects on solid tumors. Cell Death Dis. 2019, 10, 529.
  96. Thomas, J.S.; El-Khoueiry, A.B.; Maurer, B.J.; Groshen, S.; Pinski, J.K.; Cobos, E.; Gandara, D.R.; Lenz, H.J.; Kang, M.H.; Reynolds, C.P.; et al. A phase I study of intravenous fenretinide (4-HPR) for patients with malignant solid tumors. Cancer Chemother. Pharmacol. 2021, 87, 525–532.
  97. Orienti, I.; Salvati, V.; Sette, G.; Zucchetti, M.; Bongiorno-Borbone, L.; Peschiaroli, A.; Zolla, L.; Francescangeli, F.; Ferrari, M.; Matteo, C.; et al. A novel oral micellar fenretinide formulation with enhanced bioavailability and antitumour activity against multiple tumours from cancer stem cells. J. Exp. Clin. Cancer Res. 2019, 38, 373.
  98. Sun, R.; Liu, Y.; Li, S.Y.; Shen, S.; Du, X.J.; Xu, C.F.; Cao, Z.T.; Bao, Y.; Zhu, Y.H.; Li, Y.P.; et al. Co-delivery of all-trans-retinoic acid and doxorubicin for cancer therapy with synergistic inhibition of cancer stem cells. Biomaterials 2015, 37, 405–414.
  99. Kong, M.; Tang, J.; Qiao, Q.; Wu, T.; Qi, Y.; Tan, S.; Gao, X.; Zhang, Z. Biodegradable Hollow Mesoporous Silica Nanoparticles for Regulating Tumor Microenvironment and Enhancing Antitumor Efficiency. Theranostics 2017, 7, 3276–3292.
  100. Du, Y.; Bao, C.; Huang, J.; Jiang, P.; Jiao, L.; Ren, F.; Li, Y. Improved stability, epithelial permeability and cellular antioxidant activity of β-carotene via encapsulation by self-assembled α-lactalbumin micelles. Food Chem. 2019, 271, 707–714.
  101. Hidalgo, T.; Giménez-Marqués, M.; Bellido, E.; Avila, J.; Asensio, M.C.; Salles, F.; Lozano, M.V.; Guillevic, M.; Simón-Vázquez, R.; González-Fernández, A.; et al. Chitosan-coated mesoporous MIL-100(Fe) nanoparticles as improved bio-compatible oral nanocarriers. Sci. Rep. 2017, 7, 43099.
  102. Maghsoudi, S.; Taghavi Shahraki, B.; Rabiee, N.; Fatahi, Y.; Bagherzadeh, M.; Dinarvand, R.; Ahmadi, S.; Rabiee, M.; Tahriri, M.; Hamblin, M.R.; et al. The colorful world of carotenoids: A profound insight on therapeutics and recent trends in nano delivery systems. Crit. Rev. Food Sci. Nutr. 2021, 1–40.
  103. Pereira, A.R.; Mendes, T.F.; Ministro, A.; Teixeira, M.; Filipe, M.; Santos, J.M.; Barcia, R.N.; Goyri-O’Neill, J.; Pinto, F.; Cruz, P.E.; et al. Therapeutic angiogenesis induced by human umbilical cord tissue-derived mesenchymal stromal cells in a murine model of hindlimb ischemia. Stem Cell Res. Ther. 2016, 7, 145.
  104. Cao, D.; Qi, Z.; Pang, Y.; Li, H.; Xie, H.; Wu, J.; Huang, Y.; Zhu, Y.; Shen, Y.; Zhu, Y. Retinoic Acid–Related Orphan Receptor C Regulates Proliferation, Glycolysis, and Chemoresistance via the PD-L1/ITGB6/STAT3 Signaling Axis in Bladder Cancer. Cancer Res. 2019, 79, 2604–2618.
  105. Toki, K.; Enokida, H.; Kawakami, K.; Chiyomaru, T.; Tatarano, S.; Yoshino, H.; Uchida, Y.; Kawahara, K.; Nishiyama, K.; Seki, N.; et al. CpG hypermethylation of cellular retinol-binding protein 1 contributes to cell proliferation and migration in bladder cancer. Int. J. Oncol. 2010, 37, 1379–1388.
  106. Doldo, E.; Costanza, G.; Ferlosio, A.; Passeri, D.; Bernardini, S.; Scioli, M.G.; Mazzaglia, D.; Agostinelli, S.; Del Bufalo, D.; Czernobilsky, B.; et al. CRBP-1 expression in ovarian cancer: A potential therapeutic target. Anticancer Res. 2014, 34, 3303–3312.
  107. Yokoi, K.; Yamashita, K.; Ishii, S.; Tanaka, T.; Nishizawa, N.; Tsutsui, A.; Miura, H.; Katoh, H.; Yamanashi, T.; Naito, M. Comprehensive molecular exploration identified promoter DNA methylation of the CRBP1 gene as a determinant of radiation sensitivity in rectal cancer. Br. J. Cancer 2017, 116, 1046–1056.
  108. Namekawa, T.; Ikeda, K.; Horie-Inoue, K.; Suzuki, T.; Okamoto, K.; Ichikawa, T.; Yano, A.; Kawakami, S.; Inoue, S. ALDH1A1 in patient-derived bladder cancer spheroids activates retinoic acid signaling leading to TUBB3 overexpression and tumor progression. Int. J. Cancer 2020, 146, 1099–1113.
  109. Liu, H.J.; Lam, H.C.; Baglini, C.V.; Nijmeh, J.; Cottrill, A.A.; Chan, S.Y.; Henske, E.P. Rapamycin-upregulated miR-29b promotes mTORC1-hyperactive cell growth in TSC2-deficient cells by downregulating tumor suppressor retinoic acid receptor β (RARβ). Oncogene 2019, 38, 7367–7383.
More
Video Production Service