Amino Acids and Cancer Treatment: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: ,

The antioxidant properties of amino acids and their role in the physicochemical processes accompanying oxidative stress in cancer remain unclear. Cancer cells are known to extensively uptake amino acids, which are used as an energy source, antioxidant precursors that reduce oxidative stress in cancer, and as regulators of inhibiting or inducing tumor cell-associated gene expression. Nine amino acids (Cys, His, Phe, Met, Trp, Tyr, Pro, Arg, Lys) are examined, which play a key role in the non-enzymatic oxidative process in various cancers. Conventionally, these amino acids can be divided into two groups, in one of which the activity increases (Cys, Phe, Met, Pro, Arg, Lys) in cancer, and in the other, it decreases (His, Trp, Tyr). 

  • amino acids
  • cancer
  • oxidative stress
  • non-enzymatic antioxidant system

1. Introduction

Recent research in the field of cancer cell metabolism is significantly changing our understanding of metabolic processes such as the redox mechanism, carbon metabolism, epigenetic regulation, and immune response associated with tumorigenesis and metastasis [1][2]. This includes reconsidering the role of amino acids and changes in their composition during oncological processes. It is known that cancer cells intensively absorb amino acids and use them as a source of energy, precursors of antioxidants that reduce the level of oxidative stress in cancer, and also as regulators of inhibition or induction of gene expression associated with the regulation of tumor cell activity [3].
The pathophysiological process begins with a change in the oxidative capacity of proteins, through modification of the structure and activity of protein binding sites with other molecules, which are elements in the cascade reaction of antioxidant activity [4][5][6]. The choice of the oxidative process path and its activity is determined the features of the reactive type like its the activity and specificity. It is deteremined by the cell type, state of cell differentiation, state of the extracellular environment, level of antioxidants and antioxidant enzymes. The activity of the redox process will depend on the specific type of modified protein, and this, in turn, depends on the amino acid composition of the protein. In addition to the above, there is the concept of protein network formation, which explains the coordinated interaction between proteins and the activation of the redox pathway [7][8].
A number of studies on model systems have established that amino acids exhibit the ability to reduce damaging oxidative effects of various natures at different levels of biological organization [9]. These effects probably happen because of the physicochemical properties of individual amino acids, which are connected to their ability to react with reactive oxygen species. For example, the ability of citrulline and Arg to eliminate the superoxide anion radical has been shown, and leads to normalization of the functioning of the heart muscle when exposed to oxidizing factors [10][11]. It has been established that proline is an effective scavenger of singlet oxygen and prevents cell death under oxidative stress [12]. The ability of His to intercept peroxyl radicals and prevent the carboxylation of proteins and the formation of protein cross-links has been revealed [13]. It was discovered that a number of amino acids prevent the formation of 8-oxoguanine in DNA by protecting guanine from one-electron oxidation to the guanine radical cation [14]. It is still unclear to what extent the change in amino acid composition is tissue-specific, as well as how certain types of cancer are metabolically dependent on a specific amino acid composition [15].

2. Amino Acids as Therapeutic Targets for Cancer Treatment

Scientists around the world are actively exploring the possibility of regulating amino acid metabolism in oncology [16]. In many cases, therapeutic targeting of tumor cell metabolism produces better results with fewer side effects [17]. For example, therapies targeting the regulation of essential amino acids, such as Met, have beneficial effects. This was demonstrated by scientists using dietary Met restriction in mice and rats, which resulted in an increase in the lifespan of mice and rats [18]. It has long been noted that limiting the supply of amino acids leads to a slowdown in tumor growth [18][19].
Thus, the enzyme arginase is attracting increasing interest as a therapeutic target. There are two types of isoforms of this enzyme, arginase 1 (ARG1) and arginase 2 (ARG2) [20]. Abnormal expression of these enzyme isoforms has been reported to frequently occur in cancers such as breast, gastric, colorectal, and liver cancer [21][22][23][24][25]. In addition, signaling pathways associated with arginase, such as arginase/PI3K/RAC-alpha serine/threonine-protein kinase (AKT)/mTOR, arginase/signal transducer and activator of transcription 3 (STAT3), and arginase/mitogen-activated protein kinase (MAPK), can be influenced [26].
For Lys, the enzyme lysine-specific histone demethylase 1A (KDM1A/LSD1) was discovered. It shows activity in relation to the development, metastasis, progression, and therapy resistance of cancer [27].
Pharmacological modification of Cys residues affects the biological activity of this molecule, thereby demonstrating a therapeutic effect [28]. Thus, it is possible to regulate the entry of cysteine into the cell by blocking the xCT/SLC7A11 transporter [29]. SLC7A11 has been found to be widely expressed in a variety of cancers, and has poor survival rates in patients with breast cancer, prostate cancer, and papillary thyroid carcinoma [30]. In prostate cancer patients, a positive association between xCT expression with invasion and metastasis has been widely found, through an influence on the redox status of the tumor microenvironment [31]. It is also possible to influence GSH peroxidase 4 (GPX4), triggering the mechanism of ferroptosis [32][33][34][35][36][37]. A positive effect in cancer therapy was discovered by suppressing the entry of hydrogen sulfide into cancer cells, which, through activation of GLUT, enhances the uptake of glucose and its glycolysis with the subsequent production of ATP, which is spent on maintaining the life of the cancer cell [38]. Hydrogen sulfide is produced by three enzymes, namely cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), and 3-mercaptopyruvate sulfatansferase (3MST). CBS knockdown reduced oxygen consumption and ATP production in colon and ovarian cancer cells [39][40]. Suppression of CSE expression in endothelial cells reduced their ability to induce angiogenesis in breast cancer cells [41]. Cys may also have an antitumor effect through its byproduct, taurine. Cysteine dioxygenases catalyze the oxidation of Cys to cysteine sulfinate. Cysteine sulfinic acid decarboxylase catalyzes the reaction, and carboxyl groups are removed to form hypotaurine, which subsequently generates taurine [42]. Taurine has been shown to have cytoprotective functions and maintain cell homeostasis [43]. Increased taurine content has shown an inhibitory effect on the growth of colon cancer [44], lung cancer [45], HCC [46], melanoma [47], and breast cancer [48]. There are a number of other possibilities for influencing Cys, providing an antitumor effect, for example, through protein kinases and phosphatases [49][50][51][52].
An antitumor effect was demonstrated when His was added to HCC tumor cells. It considerably reduces the release of cytokines in response to liver injury [53]. There is a decrease in the expression of tumor markers that are associated with glycolysis (GLUT1 and HK2), inflammation (pSTAT3), angiogenesis (VEGFB and VEGFC), stem cells (CD133), metastasis (snail/slug), and cell migration [54].
Pro synthesized by proline synthase PYCR1 through cGMP-PKG enhances the hallmarks of stem cell carcinoma. Knockdown of proline synthetase PYCR1 showed a significant reduction in breast cancer growth [55]. Another therapeutic target is the enzyme proline dehydrogenase (PRODH) [56]. On the one hand, increased expression of PRODH induces apoptosis [57][58] of breast cancer cells. On the other hand, it stimulates metastasis to the lung. Although there is currently no clear conclusion on the stimulation and inhibition of this enzyme, since its role in cancer metabolism is twofold, further study of the possibility of using proline dehydrogenase as a therapeutic target is very important [59]. Inhibition of Pro synthesis itself may also reduce the synthesis of collagen, which is essential for the growth and development of cancer cells, and the extracellular matrix [60]. It is necessary to find a balance between reducing and completely suppressing the synthesis of extracellular matrix components, since its complete ablation can free tumor cells from the limiting barrier [61][62][63].
To limit the availability of Trp and prevent its use along the kynurene pathway, it is possible to influence the direct transport of tryptophan into the cancer cell, suppress the activity of Trp 2,3-dioxygenase (TDO2) and indoleamine 2,3-dioxygenase 1 (IDO1), and inhibit N′-formylkynurenine formamidase (FAMID), Kyn aminotransferase 1 (KAT1) and kynureninase (KYNU), nicotinamide phosphoribosyltransferase (NMPRT), nicotinamide mononucleotide adenylyltransferase (NMNAT) aryl hydrocarbon receptor (AhR), interleukin 4-induced protein 1 (IL-4I1), poly (ADP-ribose) polymerase (PARPs), and protein acetylaters (SIRTs). All this is a potential target for depriving the cancer cell of its ability to maintain its vital activity [64].
Tyrosine kinase inhibitors have become very widely used in the treatment of various types of cancer. Medicines containing this enzyme, in addition to having a positive effect in the treatment of oncology, also have a number of serious side effects. This forces scientists and physicians to look for new ways to use this approach [65].
Phe has not yet been presented as a therapeutic target for the supervision of cancer patients. This area requires additional study and understanding of possible control points in the life of a cancer cell.

This entry is adapted from the peer-reviewed paper 10.3390/metabo14010028

References

  1. Isenberg, J.S.; Martin-Manso, G.; Maxhimer, J.B.; Roberts, D.D. Regulation of nitric oxide signalling by thrombospondin 1: Implications for anti-angiogenic therapies. Nat. Rev. Cancer 2009, 9, 182–194.
  2. Fukumura, D.; Kashiwagi, S.; Jain, R.K. The role of nitric oxide in tumour progression. Nat. Rev. Cancer 2006, 6, 521–534.
  3. Wu, G.; Bazer, F.W.; Burghardt, R.C.; Johnson, G.A.; Kim, S.W.; Knabe, D.A.; Li, P.; Li, X.; McKnight, J.R.; Satterfield, M.C.; et al. Proline and hydroxyproline metabolism: Implications for animal and human nutrition. Amino Acids 2011, 40, 1053–1063.
  4. Wensien, M.; von Pappenheim, F.R.; Funk, L.M.; Kloskowski, P.; Curth, U.; Diederichsen, U.; Uranga, J.; Ye, J.; Fang, P.; Pan, K.T.; et al. A lysine-cysteine redox switch with an NOS bridge regulates enzyme function. Nature 2021, 593, 460–464.
  5. Coleman, C.S.; Stanley, B.A.; Pegg, A.E. Effect of mutations at active site residues on the activity of ornithine decarboxylase and its inhibition by active site-directed irreversible inhibitors. J. Biol. Chem. 1993, 268, 24572–24579.
  6. Obin, M.; Shang, F.; Gong, X.; Handelman, G.; Blumberg, J.; Taylor, A. Redox regulation of ubiquitin-conjugating enzymes: Mechanistic insights using the thiol-specific oxidant diamide. FASEB J. 1998, 12, 561–569.
  7. Wall, S.B.; Oh, J.Y.; Diers, A.R.; Landar, A. Oxidative modification of proteins: An emerging mechanism of cell signaling. Front. Physiol. 2012, 14, 369.
  8. Vučetić, M.; Cormerais, Y.; Parks, S.K.; Pouysségur, J. The Central Role of Amino Acids in Cancer Redox Homeostasis: Vulnerability Points of the Cancer Redox Code. Front. Oncol. 2017, 21, 319.
  9. Shtarkman, I.N.; Gudkov, S.V.; Chernikov, A.V.; Bruskov, V.I. Effect of amino acids on X-ray-induced hydrogen peroxide and hydroxyl radical formation in water and 8-oxoguanine in DNA. Biochemistry 2008, 73, 470–478.
  10. Du, J.; Gebicki, J.M. Proteins are major initial cell targets of hydroxyl free radicals. Int. J. Biochem. Cell Biol. 2004, 36, 2334–2343.
  11. Hayashi, T.; Juilet, P.A.R.; Matsui Hirai, H.; Miyazaki, A.; Fukatsu, A.; Funami, J.; Iguchi, A.; Ignarro, L.J. l-citrulline and l-arginine supplementation retards the progression of high-cholesterol-diet-induced atherosclerosis in rabbits. Proc. Natl. Acad. Sci. USA 2005, 102, 13681–13686.
  12. Chen, C.; Dickman, M.B. Proline suppresses apoptosis in the fungal pathogen Colletotrichum trifolii. Proc. Natl. Acad. Sci. USA 2005, 102, 3459–3464.
  13. Dekker, E.A.; Livesey, S.A.; Zhu, S. A re-evaluation of the antioxidant activity of purified carnosine. Biochemistry 2000, 65, 901–906.
  14. Milligan, J.R.; Aguilera, J.A.; Ly, A.; Tran, N.Q.; Hoang, O.; Ward, J.F. Repair of oxidative DNA damage by amino acids. Nucl. Acids Res. 2003, 31, 6258–6263.
  15. Lieu, E.L.; Nguyen, T.; Rhyne, S.; Kim, J. Amino acids in cancer. Exp. Mol. Med. 2020, 52, 15–30.
  16. Vettore, L.; Westbrook, R.L.; Tennant, D.A. New aspects of amino acid metabolism in cancer. Br. J. Cancer 2020, 122, 150–156.
  17. Lukey, M.J.; Katt, W.P.; Cerione, R.A. Targeting amino acid metabolism for cancer therapy. Drug Discov. Today 2017, 22, 796–804.
  18. Miller, R.A.; Buehner, G.; Chang, Y.; Harper, J.M.; Sigler, R.; Smith-Wheelock, M. Methionine-deficient diet extends mouse lifespan, slows immune and lens aging, alters glucose, T4, IGF-I and insulin levels, and increases hepatocyte MIF levels and stress resistance. Aging Cell 2005, 4, 119–125.
  19. Choi, B.H.; Coloff, J.L. The diverse functions of non-essential amino acids in cancer. Cancers 2019, 11, 675.
  20. Sedillo, J.C.; Cryns, V.L. Targeting the methionine addiction of cancer. Am. J. Cancer Res. 2022, 12, 2249–2276.
  21. Roci, I.; Watrous, J.D.; Lagerborg, K.A.; Lafranchi, L.; Lindqvist, A.; Jain, M.; Nilsson, R. Mapping metabolic events in the cancer cell cycle reveals arginine catabolism in the committed SG2M phase. Cell Rep. 2019, 26, 1691–1700.
  22. Wu, C.W.; Chi, C.W.; Lin, E.C.; Lui, W.Y.; P’Eng, F.K.; Wang, S.R. Serum arginase level in patients with gastric cancer. J. Clin. Gastroenterol. 1994, 18, 84–85.
  23. Leu, S.Y.; Wang, S.R. Clinical significance of arginase in colorectal cancer. Cancer 1992, 70, 733–736.
  24. Sovova, V.; Sloncova, E.; Fric, P. Differences of alkaline phosphatase and arginase activities in human colorectal carcinoma cell lines. Folia Biol. 1997, 43, 101–104.
  25. Chrzanowska, A.; Grabon, W.; Mielczarek-Puta, M.; Baranczyk-Kuzma, A. Significance of arginase determination in body fluids of patients with hepatocellular carcinoma and liver cirrhosis before and after surgical treatment. Clin. Biochem. 2014, 47, 1056–1059.
  26. Niu, F.; Yu, Y.; Li, Z.; Ren, Y.; Li, Z.; Ye, Q.; Liu, P.; Ji, C.; Qian, L.; Xiong, Y. Arginase: An emerging and promising therapeutic target for cancer treatment. Biomed. Pharmacother. 2022, 149, 112840.
  27. Johnson, J.D.; Alejo, S.; Jayamohan, S.; Sareddy, G.R. Lysine-specific demethylase 1 as a therapeutic cancer target: Observations from preclinical study. Expert Opin. Ther. Targets 2023, 24, 1–12.
  28. Yuta, K.; Yukio, N. Design of cysteine-based self-assembling polymer drugs for anticancer chemotherapy. Colloid. Surf. B 2022, 220, 112909.
  29. Chevallier, V.; Zoller, M.; Kochanowski, N.; Andersen, M.R.; Workman, C.T.; Malphettes, L. Use of novel cystine analogs to decrease oxidative stress and control product quality. J. Biotechnol. 2021, 327, 1–8.
  30. Lin, W.; Wang, C.; Liu, G.; Bi, C.; Wang, X.; Zhou, Q.; Jin, H. SLC7A11/xCT in cancer: Biological functions and therapeutic implications. Am. J. Cancer Res. 2020, 10, 3106–3126.
  31. Zhong, W.; Weiss, H.L.; Jayswal, R.D.; Hensley, P.J.; Downes, L.M.; St. Clair, D.K.; Chaiswing, L. Extracellular redox state shift: A novel approach to target prostate cancer invasion. Free Radic. Biol. Med. 2018, 117, 99–109.
  32. Hanigan, M.H.; Ricketts, W.A. Extracellular glutathione is a source of cysteine for cells that express gamma-glutamyl transpeptidase. Biochemistry 1993, 32, 6302–6306.
  33. Meister, A. Glutathione, ascorbate, and cellular protection. Cancer Res. 1994, 54, 1969–1975.
  34. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 49, 1060–1072.
  35. Brigelius-Flohe, R.; Maiorino, M. Glutathione peroxidases. Biochim. Biophys. Acta 2013, 1830, 3289–3303.
  36. Yang, W.S.; SriRamaratnam, R.; Welsch, M.E.; Shimada, K.; Skouta, R.; Viswanathan, V.S.; Cheah, J.H.; Clemons, P.A.; Shamji, A.F.; Clish, C.B.; et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 2014, 156, 317–331.
  37. Fujii, J.; Homma, T.; Kobayashi, S. Ferroptosis caused by cysteine insufficiency and oxidative insult. Free Radic. Res. 2020, 54, 969–980.
  38. Hellmich, M.R.; Coletta, C.; Chao, C.; Szabo, C. The therapeutic potential of cystathionine beta synthetase/hydrogen sulfide inhibition in cancer. Antioxid. Redox Signal 2015, 22, 424–448.
  39. Bhattacharyya, S.; Saha, S.; Giri, K.; Lanza, I.R.; Nair, K.S.; Jennings, N.B.; Rodriguez-Aguayo, C.; Lopez-Berestein, G.; Basal, E.; Weaver, A.L.; et al. Cystathionine beta-synthase (CBS) contributes to advanced ovarian cancer progression and drug resistance. PLoS ONE 2013, 8, e79167.
  40. Szabo, C.; Coletta, C.; Chao, C.; Modis, K.; Szczesny, B.; Papapetropoulos, A.; Hellmich, M.R. Tumor-derived hydrogen sulfide, produced by cystathionine-β-synthase, stimulates bioenergetics, cell proliferation, and angiogenesis in colon cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 12474–12479.
  41. Pupo, E.; Pla, A.F.; Avanzato, D.; Moccia, F.; Cruz, J.E.; Tanzi, F.; Merlino, A.; Mancardi, D.; Munaron, L. Hydrogen sulfide promotes calcium signals and migration in tumor-derived endothelial cells. Free Radic. Biol. Med. 2011, 51, 1765–1773.
  42. Stipanuk, M.H.; Jurkowska, H.; Roman, H.B.; Niewiadomski, J.; Hirschberger, L.L. Insights into taurine synthesis and function based on studies with cysteine dioxygenase (CDO1) knockout mice. Adv. Exp. Med. Biol. 2015, 803, 29–39.
  43. Marcinkiewicz, J.; Kontny, E. Taurine and inflammatory diseases. Amino Acids 2014, 46, 7–20.
  44. Liu, Z.; Xia, Y.; Zhang, X.; Liu, L.; Tu, S.; Zhu, W.; Yu, L.; Wan, H.; Yu, B.; Wan, F. Roles of the MST1-JNK signaling pathway in apoptosis of colorectal cancer cells induced by taurine. Libyan J. Med. 2018, 13, 1500346.
  45. Tu, S.; Zhang, X.-L.; Wan, H.-F.; Xia, Y.-Q.; Liu, Z.-Q.; Yang, X.-H.; Wan, F.-S. Effect of taurine on cell proliferation and apoptosis human lung cancer A549 cells. Oncol. Lett. 2018, 15, 5473–5480.
  46. Park, S.H.; Lee, H.; Park, K.K.; Kim, H.W.; Park, T. Taurine-responsive genes related to signal transduction as identified by cDNA microarray analyses of HepG2 cells. J. Med. Food. 2006, 9, 33–41.
  47. Yu, J.; Kim, A.K. Effect of taurine on antioxidant enzyme system in B16F10 melanoma cells. Adv. Exp. Med. Biol. 2009, 643, 491–499.
  48. Choi, E.J.; Tang, Y.; Lee, C.B.; Cheong, S.H.; Sung, S.H.; Oh, M.R.; Jang, S.Y.; Park, P.J.; Kim, E.K. Effect of taurine on in vitro migration of MCF-7 and MDA-MB-231 human breast carcinoma cells. Adv. Exp. Med. Biol. 2015, 803, 191–201.
  49. Lennicke, C.; Cocheme, H.M. Redox metabolism: ROS as specific molecular regulators of cell signaling and function. Mol. Cell 2021, 81, 3691–3707.
  50. Turdo, A.; D’Accardo, C.; Glaviano, A.; Porcelli, G.; Colarossi, C.; Colarossi, L.; Mare, M.; Faldetta, N.; Modica, C.; Pistone, G.; et al. Targeting phosphatases and kinases: How to checkmate cancer. Front. Cell Dev. Biol. 2021, 9, 690306.
  51. Neel, B.G.; Tonks, N.K. Protein tyrosine phosphatases in signal transduction. Curr. Opin. Cell Biol. 1997, 9, 193–204.
  52. Zhang, Z.Y. Protein tyrosine phosphatases: Structure and function, substrate specificity, and inhibitor development. Annu. Rev. Pharmacol. Toxicol. 2002, 42, 209–234.
  53. Yan, S.L.; Wu, S.T.; Yin, M.C.; Chen, H.T.; Chen, H.C. Protective effects from carnosine and histidine on acetaminophen-induced liver injury. J. Food Sci. 2009, 74, 259–265.
  54. Park, Y.; Han, Y.; Kim, D.; Cho, S.; Kim, W.; Hwang, H.; Lee, H.W.; Han, D.H.; Kim, K.S.; Yun, M.; et al. Impact of Exogenous Treatment with Histidine on Hepatocellular Carcinoma Cells. Cancers 2022, 14, 1205.
  55. Bai, C.; Bin, H.; Yanping, H.; Cenxin, W.; Huandong, L.; Jinxin, L.; Keyu, S.; Xiaoyu, Z.; Yuanyuan, L.; Zhuoran, Z.; et al. Pyrroline-5-carboxylate reductase 1 reprograms proline metabolism to drive breast cancer stemness under psychological stress. Cell Death Dis. 2023, 14, 682.
  56. Amaravadi, R.K.; Kimmelman, A.C.; Debnath, J. Targeting autophagy in cancer: Recent advances and future directions. Cancer Discov. 2019, 9, 1167–1181.
  57. Maxwell, S.A.; Davis, G.E. Differential gene expression in p53-mediated apoptosis-resistant vs. apoptosis-sensitive tumor cell lines. Proc. Natl. Acad. Sci. USA 2000, 97, 13009–13014.
  58. Raimondi, I.; Ciribilli, Y.; Monti, P.; Bisio, A.; Pollegioni, L.; Fronza, G.; Inga, A.; Campomenosi, P. P53 Family Members Modulate the Expression of PRODH, but Not PRODH2, via Intronic p53 Response Elements. PLoS ONE 2013, 8, e69152.
  59. Xu, X.; Zhang, G.; Chen, Y.; Xu, W.; Liu, Y.; Ji, G.; Xu, H. Can proline dehydrogenase-a key enzyme involved in proline metabolism-be a novel target for cancer therapy? Front. Oncol. 2023, 13, 1254439.
  60. Kay, E.J.; Zanivan, S.; Rufini, A. Proline metabolism shapes the tumor microenvironment: From collagen deposition to immune evasion. Curr. Opin. Biotechnol. 2023, 84, 103011.
  61. Chen, Y.; Kim, J.; Yang, S.; Wang, H.; Wu, C.J.; Sugimoto, H.; LeBleu, V.S.; Kalluri, R. Type I collagen deletion in αSMA(+) myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell 2021, 39, 548–565.
  62. Provenzano, P.P.; Inman, D.R.; Eliceiri, K.W.; Knittel, J.G.; Yan, L.; Rueden, C.T.; White, J.G.; Keely, P.J. Collagen density promotes mammary tumor initiation and progression. BMC Med. 2008, 6, 11.
  63. Cox, T.R. The matrix in cancer. Nat. Rev. Cancer 2021, 11, 217–238.
  64. Badawy, A.A. Tryptophan metabolism and disposition in cancer biology and immunotherapy. Biosci. Rep. 2022, 42, BSR20221682.
  65. Shyam Sunder, S.; Sharma, U.C.; Pokharel, S. Adverse effects of tyrosine kinase inhibitors in cancer therapy: Pathophysiology, mechanisms and clinical management. Signal Transduct. Target. Ther. 2023, 8, 262.
More
This entry is offline, you can click here to edit this entry!
Video Production Service