Next Article in Journal
Endocrine-Disrupting Effects of Bisphenol A on the Cardiovascular System: A Review
Previous Article in Journal
Research Progress of the Endocrine-Disrupting Effects of Disinfection Byproducts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of pH in Regulating Cancer Pyrimidine Synthesis

by
Saad Saeed Alqahtani
1,2,
Tomas Koltai
3,
Muntaser E. Ibrahim
4,
Adil H. H. Bashir
4,
Sari T. S. Alhoufie
5,
Samrein B. M. Ahmed
6,
Daria Di Molfetta
7,
Tiago M. A. Carvalho
7,
Rosa Angela Cardone
7,
Stephan Joel Reshkin
7,
Abdelhameed Hifny
8,
Mohamed E. Ahmed
9 and
Khalid Omer Alfarouk
9,10,11,*
1
Department of Pharmacy Practice, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
2
Pharmacy Practice Research Unit, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
3
Via Pier Capponi 6, 50132 Florence, Italy
4
Department of Molecular Biology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan
5
Medical Laboratories Technology Department, College of Applied Medical Sciences, Taibah University, Medina 42353, Saudi Arabia
6
Department of Biosciences and Chemistry, College of Health, Wellbeing and Life Sciences, Sheffield Hallam University, Sheffield S1 1WB, UK
7
Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 70126 Bari, Italy
8
Faculty of Medicine, Al-Azhar University, Cairo 11651, Egypt
9
Research Center, Zamzam University College, Khartoum 11123, Sudan
10
Alfarouk Biomedical Research LLC, Temple Terrace, FL 33617, USA
11
Hala Alfarouk Cancer Center, Khartoum 11123, Sudan
*
Author to whom correspondence should be addressed.
J. Xenobiot. 2022, 12(3), 158-180; https://doi.org/10.3390/jox12030014
Submission received: 18 April 2022 / Revised: 17 June 2022 / Accepted: 28 June 2022 / Published: 6 July 2022

Abstract

:
Replication is a fundamental aspect of cancer, and replication is about reproducing all the elements and structures that form a cell. Among them are DNA, RNA, enzymes, and coenzymes. All the DNA is doubled during each S (synthesis) cell cycle phase. This means that six billion nucleic acids must be synthesized in each cycle. Tumor growth, proliferation, and mutations all depend on this synthesis. Cancer cells require a constant supply of nucleotides and other macromolecules. For this reason, they must stimulate de novo nucleotide synthesis to support nucleic acid provision. When deregulated, de novo nucleic acid synthesis is controlled by oncogenes and tumor suppressor genes that enable increased synthesis and cell proliferation. Furthermore, cell duplication must be achieved swiftly (in a few hours) and in the midst of a nutrient-depleted and hypoxic environment. This also means that the enzymes participating in nucleic acid synthesis must work efficiently. pH is a critical factor in enzymatic efficiency and speed. This review will show that the enzymatic machinery working in nucleic acid synthesis requires a pH on the alkaline side in most cases. This coincides with many other pro-tumoral factors, such as the glycolytic phenotype, benefiting from an increased intracellular pH. An increased intracellular pH is a perfect milieu for high de novo nucleic acid production through optimal enzymatic performance.

1. Introduction

Folic acid was synthesized in 1937, and it was identified as a necessary factor for bone marrow function. Interestingly, folate-depleted feeding produced bone marrow damage similar to mustard gas. Sidney Farber, a pediatric pathologist working in Boston, used folic acid to treat anemia in children with acute leukemia. The results were devastating. Leukemia lit up and progressed very quickly. Farber concluded that it was necessary to find a substance that could do the opposite: deplete folate. He found the right chemist, Yellapragada Subbarao, who developed aminopterin, a folate antagonist that induced remissions in children with acute leukemia. Thus, in 1948–1949, the first anti-metabolite was found [1,2,3,4,5]. Interestingly, the first antimetabolite directly targeted nucleic acid synthesis. However, Farber and all the oncologists who adopted aminopterin as the gold standard for pediatric leukemia in the 1950s did not know how it worked beyond folate antagonism nor did they know anything about nucleic acid synthesis. It took the discovery of the structure of DNA in 1953 and painstaking biochemical research afterward to find out how nucleic acids are synthesized. This also led to the development of new inhibitors. This short saga about folic acid antagonists shows that a drug can be used successfully even if its mechanism of action is unknown. However, at the same time, ignoring the mechanism can cause unexpected damage. Understanding how nucleic acids are synthesized is necessary if we expect to inhibit their production.
A separate issue was the finding that the intracellular pH (pHi) in tumors is more alkaline than in normal counterparts [6]. In addition, it has been known since the 1970s that most animal eggs become more alkaline immediately before mitosis [7,8,9,10]. In this regard, it was found that cytoplasmic pH controls protein synthesis in fertilized eggs and early embryos [11]. In 2000, Reshkin et al. discovered that one of the first events in the cellular transformation was increased pHi [12].
This review will analyze the enzymes that participate in the synthesis of nucleic acids, particularly pyrimidines, and how intracellular pH interacts with them. Understanding this relationship may help develop new pharmaceuticals and improve the results of those already in use. This analysis will show that an increased intracellular pH seems to be a facilitator, if not a condition, for the synthesis of pyrimidines, thus generating both DNA and RNA.

1.1. Nitrogen Bases Nucleotide Synthesis

Cell division requires adequate nucleotide pools for increased DNA and RNA production in highly proliferative cells. The DNA building blocks consist of pyrimidines, purines, deoxyribose, and phosphate. Two types of nitrogen bases form part of DNA and RNA: purines and pyrimidines (Figure 1).
According to the Chargaff rule (1948), there are as many pyrimidines as purines in DNA [5], the first hint towards the base paring system (a purine always pairs with a pyrimidine and vice versa) that, a few years later, allowed Watson, Crick, Wilkinson, and Franklin to explain the DNA structure. However, in 1953, when this seminal discovery was published, nothing was known about how these nitrogen bases were synthesized. While existing pyrimidines and the re-utilization of pyrimidines from the degradation of some cell structures are sufficient for the resting cell, the situation is completely different in malignancies and highly proliferating cells, where new pyrimidines need to be developed [13].
There are two pathways for nucleotide synthesis [14]:
  • The de novo pathway starts with the precursor molecules, such as amino acids, CO2, NH3, and the sugar ribose-5-phosphate. Usually, proliferating and non-proliferating cells preferentially use the de novo pathway rather than the salvage mechanism [15,16].
  • The salvage pathway is a process of recycling existing nucleotides and bases that originated in the breakdown of existing nucleic acids.
In this review, we focus only on the de novo synthesis of the pyrimidine structure. The de novo synthesis of pyrimidines and purines is similar in all living beings.
A few basic but essential concepts:
In purine and pyrimidine biosynthetic pathways, an amino acid is a precursor of each path:
Glycine for purines.
Aspartate for pyrimidines.
Glutamine is the source of amino groups.
In purine and pyrimidine de novo pathways, many enzymes are organized as large multi-enzyme complexes.
In both purine and pyrimidine de novo synthesis, a negative feedback loop regulates the number of molecules to be synthesized.
The cellular pools of nucleotides are generally very small compared to the amount needed for DNA or RNA synthesis. Thus, nucleotide synthesis is an essential process for cell replication and growth. This becomes even more evident in highly proliferating cells, such as those found in tumors.
Limiting nucleotide synthesis decreases proliferation and growth.
Drugs that can inhibit nucleotide synthesis can impede, delay, or decrease malignant proliferation.
Pyrimidines are mainly produced by tumor cells, but stromal cells, such as macrophages and cancer-associated fibroblasts, can also produce them. In pancreatic cancer, it has been found that pyrimidines produced by macrophages were able to create resistance to gemcitabine treatment [17].
Glutamine transporters that provide glutamine to the cell (glutamine is a nitrogen donor for pyrimidines) alkalinize the intracellular milieu by simultaneously exporting protons [18].

1.2. Pyrimidine De Novo Synthesis

Pyrimidine nucleotides are essential building blocks not only for nucleic acid synthesis but DNA repair and other cell functions as well [19]. Pyrimidine biosynthesis is initiated and regulated by a multienzymatic complex, CAD (CPS-II, aspartate transcarbamoylase, and dihydroorotase), that harbors the enzymes required for the first three steps of biosynthesis. This association of enzymes in one big polypeptide is an unusual finding in superior eukaryotes. This explains the difficulties found in characterizing and defining the properties of each of the enzymatic domains in a separate manner. In addition, CAD self-assembles in hexameres. CAD, which controls the pathway, is in turn, controlled by pro-proliferation and pro-biosynthesis pathways (Figure 2).

2. The Pyrimidine Synthesis Pathway

Figure 3 shows a full view of the de novo synthesis of pyrimidines from their origin in glutamine up to the pyrimidine uridine monophosphate. The diagram shows the origin of the different parts of a uracil molecule, which receives contributions from aspartic acid, glutamine, and CO2 (converted into HCO3− by cytoplasmic carbonic anhydrases). The major amino acid contributors are aspartic acid and glutamine. Each of them also donates an amino group. Glutamine contributes to an NH2 group (see right panel of Figure 3).
CPS II: carbamoyl phosphate synthetase II; ATCase: aspartate transcarbamoylase; OPRT: orotate phosphoribosyltransferase; PRPP: phosphoribosyl diphosphate (PRPP). This whole synthetic process consumes seven ATP molecules.

2.1. The Steps in De Novo Pyrimidine Synthesis

  • Step 1:
In the presence of ATP, L-glutamine and bicarbonate are converted to carbamoyl phosphate (CAP) by carbamoyl phosphate synthetase II (CPS-II) in the cytoplasm (Figure 4).
There are two types of CPS:
(1)
CPS-I is an intra-mitochondrial enzyme;
(2)
CPS-II is cytosolic and is the enzyme that participates in de novo pyrimidine biosynthesis. This is the rate-limiting enzyme in pyrimidine biosynthesis.
CPS-II is upregulated in many cancers [24], and mainly in hepatoma [25,26,27]
The optimal pH for CPS-II activity is 7.4 [28]. Normal cells have an intracellular pH between 7.1 and 7.2. This means that to achieve an efficient function of this rate-limiting enzyme, the cytoplasm requires a higher pH. Interestingly, tumors raise their intracellular pH to 7.4–7.5 [29]. The difference seems small, but it must be remembered that pH is a logarithmic function, thus the difference represents an important increase in proton concentration. When the pH reaches the optimal point for an enzyme, this means that the enzyme is working at its maximum efficiency and speed.
The end product of the events leading to pyrimidine synthesis is uridine monophosphate (UMP), which inhibits CPS-II via a negative feedback loop (not shown in Figure 3).
CPS-II activators. Many activators, such as ATP and phosphoribosyl diphosphate (PRPP), stimulate the expression of this enzyme [30]. mTORC1 is a key modulator that upregulates the whole pathway [31] and, through its downstream protein S6K1, induces the expression of all the enzymes involved in de novo pyrimidine synthesis, including CPS-II [32] and the other two enzymes that participate in steps 2 and 3. Actually, the three enzymes that catalyze the first three steps of pyrimidine synthesis, namely, CPS-II, aspartate transcarbamoylase, and dihydroorotase, form an enzymatic complex that is known by the acronym CAD, as mentioned above.
CPS-II Inhibitors. CPSII is inhibited by acivicin, an analog of glutamine [33]. In this respect, a glutamine-restricted diet might limit pyrimidine biosynthesis, interrupting the nucleic acid synthesis.
  • Step 2:
The carbamoyl phosphate (CAP) interacts with aspartic acid, generating carbamoyl aspartic acid (CAA) through condensation via aspartate transcarbamoylase (ATCase) (Figure 5).
The impact of pH on ATCase (aspartate transcarbamoylase) activity depends on the substrate concentration: the optimal action occurs at an alkaline pH of 8.5 in bacteria [34] and at around 9 in rat liver [35]. Here again, we find that a high intracellular pH favors the synthesizing steps.
There is strong evidence showing that ATCase is upregulated in tumors, and its down/regulation by N-(phosphonacetyl)-l-aspartate (PALA) inhibits tumor growth [36,37,38,39,40]. PALA may have antitumor activity and potentiates the effects of chemotherapeutic agents, e.g., 5-fluorouracil (5-FU) [41]. Moreover, 2-phenyl-1,3-4(H)benzothiazin-4-thione (quinazolinone derivative) is a strong ATCase inhibitor [42]. The pyrimidine pathway might form cytidine triphosphate (CTP); this CTP blocks ATCase, reflecting its negative feedback mechanism. ATP activates ATCase.
  • Step 3:
Carbamoyl aspartic acid (CAA) is converted to dihydroorotic acid (DHOA) via dihydroorotase (EC 3.5.2.3), which is also known as dihydroorotate hydrolase, and carbamoylaspartic dehydrase. Dihydroorotase is a metalloenzyme that catalyzes the reversible conversion of carbamoyl aspartic acid into dihydroorotic acid and closes the ring (Figure 6).
The effect of pH on dihydroorotase in cancer cell lines is a controversial and debated issue. However, the current concept is that dihydroorotase is driven forward by an acidic pH, while the reversible reaction (e.g., the formation of CAA) reaches its optimal efficiency at an alkaline pH [43,44,45,46,47,48].
At this point, the three enzymes forming CAD have catalyzed the ring structure that will form pyrimidines in the next steps from glutamine, CO2, and aspartate. The optimal pH for the first two enzymes is above 7.4, while the third enzyme requires a lower pH (around 7 or lower).
Hypoxia-inducible factor 1 alpha (HIF-1 alpha) and deferoxamine, metal chelator inducing HIF-1α expression, show inhibitory effects on the CAD multienzymatic complex [49]. This seems paradoxical because HIF-1 alpha is a strong tumor driver.
Therefore, although the dihydroorotase appears to be an enzyme that does not enhance the pyrimidine biosynthetic pathway, some data suggest that the same CAD also supports tumorigenesis [50]. However, it was also found that CAD downregulates the Wnt/Beta catenin pathway in colon cancer cells, decreasing migration [51].

2.2. Dihydroorotase Inhibitors

Dihydroorotase seems to interact with the thymidylate synthase inhibitor 5-fluorouracil [52]. However, this needs further research. Plumbagin, a natural product obtained from the carnivorous plant Nepenthes miranda, has shown strong competitive inhibition, causing apoptosis in tumors [52]. Analogs of carbamoyl aspartate have also shown inhibitory effects [53].
  • Step 4:
Dihydroorotic acid (DHOA) is converted to orotic acid (OA) via dihydroorotate dehydrogenase. This is the only redox reaction in the de novo biosynthesis pathway of pyrimidine nucleotides.
There are two dihydroorotate dehydrogenases, namely, class 1 (cytosolic) and class 2 (found in the inner mitochondrial membrane). In different organisms, dihydroorotate dehydrogenases use either NAD+, fumarate, or NADP+ as electron acceptors, yielding NADH, succinate, or NADPH, respectively [54,55,56,57,58]. Class 2 uses quinone as an electron acceptor, yielding hydroquinone [59,60,61,62,63] (Figure 7).
The optimal pH for this is around 8 [64,65]. This is logical because this enzyme carries out its work in the mitochondria [66], which have a higher pH than the cytoplasm.
Leflunomide is a drug used to treat rheumatoid arthritis that has shown significant efficacy against dihydroorotate dehydrogenase [67], and it could be considered as an anticancer drug candidate [68,69,70].
Other inhibitors, such as brequinar sodium, atovaquone, and ML390. have also been identified (See Box 1).
Box 1. Brequinar.
Brequinar sodium (a quinoline-carboxylic acid sodium salt) is a small molecule that acts as a specific inhibitor of dihydroorotate dehydrogenase, the fourth enzyme of the de novo pyrimidine biosynthetic pathway. Brequinar sodium inhibits dihydroorotate dehydrogenase that has ubiquinone as a cofactor but not when NAD is the cofactor [71].
Brequinar has shown a dose-dependent antineoplastic and immunosuppressant activity against different tumor models [72] and went through phase I and II clinical trials during the 1990s [73,74,75,76]. A narrow therapeutic window and high myelotoxicity led this drug to oblivion. In 2018, new studies confirmed that brequinar was not active at lower doses in solid tumors but was quite effective in leukemia [77]. This different behavior in non-solid tumors seems to confirm two concepts:
Solid tumors have a higher level of pyrimidines that require toxic doses of brequinar, while leukemias respond to lower doses due to a decreased pool of uridine;
The pyrimidine synthetic pathway is somehow related to the inhibition of myeloid differentiation because the inhibition of dihydroorotate dehydrogenase overcame a differentiation blockade in acute myeloid leukemia in vivo [78].
Interest in brequinar as a complementary drug for cancer treatment in both non-solid and solid tumors has been revived after 20 years [79,80,81,82,83]. The fact that brequinar has shown the ability to induce ferroptosis and to increase the ferroptotic effect of drugs such as sulfasalazine [84] is particularly interesting.
  • Step 5:
Orotic acid (OA) interacts with phosphoribosyl pyrophosphate to produce orotidine 5′-monophosphate (OMP), also known as orotidylic acid. The enzyme that catalyzes the reaction is called orotate phosphoribosyltransferase (OPRT).
Phosphoribosyl pyrophosphate originates in the first part of the oxidative phase of the pentose phosphate pathway [85]. Therefore, this step of the pentose phosphate pathway (PPP) branches to merge and/or amalgamate with de novo pyrimidine biosynthesis. Interestingly, there is a shared regulation of the glycolytic and PPP pathways by intracellular pH pHi. Indeed, as with glycolysis, the optimal activity of the enzymes driving the PPP occurs at an alkaline pHi, which is compatible with the cytoplasmic pH of cancer cells (Figure 8 and Figure 9).
Importantly, OPRT is an enzyme that contributes to the conversion of 5-FU into fdUMP, which is the active form of 5-FU [86]. Resistance to 5-FU seems to be associated with low OPRT expression [87,88]. OPRT enhances the chemotherapeutic response to 5-fluorouracil (5-FU) [89,90].
OPRT is overexpressed in many tumors, such as bladder cancer [91], and its expression is correlated with recurrence. Further, OPRT is useful as a marker of prognosis and for predicting chemosensitivity [90,92,93].
The optimal pH of the OPRT for the forward reaction is 8, while for the reverse reaction it ranges between 6.5 and 7.5 [94].
OPRT activity inhibitors include xanthosine, uridine 5′-phosphates, cytidine barbiturate, 5-flouro orotate (most effective), and higher concentrations of nucleotides [95,96]. OPRT inhibition also plays a role in other diseases, e.g., tuberculosis, toxoplasmosis, and malaria [97].
  • Step 6:
Orotidine 5′-monophosphate is converted to uridine monophosphate (UMP) by orotidine 5′-phosphate decarboxylase (OMP decarboxylase, ODCase). As a result, OPRT plus OMP decarboxylase form the bifunctional enzyme called uridine monophosphate synthetase (UMPS) (Figure 10) [98].
There is not much information about the expression of ODCase in tumors. However, pyrazofurin and 6-Aza uridine 5′-monophosphate are examples of ODCase inhibitors with antitumor properties [99,100].
The optimal pH is approximately 7.5 [101,102].
  • Step 7 (Uracil Formation):
Two consecutive phosphorylations take place: steps 7.1 and 7.2 (Figure 11).
  • Step 7.1:
UMP is converted to uridine diphosphate (UDP) via cytidine monophosphate kinase (CMP kinase or uridine monophosphate-cytidine monophosphate phosphotransferase). CMP kinase is found in the cytoplasm, nucleus, and mitochondria [103,104,105].
The optimal pH for CMP kinase is 7.4 [106].
When CMP kinase 1 is overexpressed in cancer [107,108,109], it is a powerful indicator of poor prognosis. Importantly, the anticancer drug gemcitabine [108] inhibits CMP kinase [65].
Step 7.1 is a crossroad of de novo pyrimidine biosynthesis, and it can follow two main branches.
  • Step 7.2:
UDP is further phosphorylated to UTP using nucleoside-diphosphate kinase (UDP kinase). UTP can enter additional metabolic pathways, including glycogen, galactose, and glucosamine [110,111]. Moreover, it acts as a substrate (ligand) for purinergic receptors (P2 receptors) [112].
Here, the optimal pH is 8 [113].
NM23 (nucleoside-diphosphate kinase), which is known as a metastasis suppressor [114], produces nucleoside-diphosphate kinase, although in some tumors (e.g., melanoma, breast, and colon), its expression is associated with low metastatic potential. In others (neuroblastoma and osteosarcoma), it is related to a decrease in survival rate [115]. Therefore, its role is unclear. One of the possible explanations is that, while NM23 protein suppresses metastasis, it does not alter the growth of primary tumors [116].
Theophylline (phosphodiesterase inhibitor) inhibits NDK [117].
  • Step 8 (Cytosine Formation):
UTP is converted to cytosine triphosphate via cytosine triphosphate synthase (CTP synthase), also known as UTP: ammonia ligase. Glutamine is also crucial in carrying out this step [118,119]. Therefore, the presence of glutamine is essential for starting de novo pyrimidine biosynthesis and forming cytosine (Figure 12).
The optimal pH of CTP synthase is around 8 [120].
CTP synthase expression is markedly increased in very rapidly growing tumors [121,122]. Moreover, a CTP synthase abnormality may be associated with resistance [123].
Cyclopentenyl cytosine is an inhibitor of CTP synthase [121], while GTP is a CTP synthase activator [120,122,124].
The steps discussed above show the synthesis of the two pyrimidine bases that form the nucleotides for RNA, namely, the uracil and cytosine nitrogen bases and the uridine and cytidine nucleotides, respectively.
  • Step 9 (Thymine Formation):
DNA has thymine instead of uracil. In this step, UDP is reduced to produce dUDP (deoxyuridine diphosphate) via ribonucleotide reductase, which converts NADPH to NADP+ (Figure 13).
  • Step 10:
dUDP is dephosphorylated, yielding dUMP (deoxyuridylic acid or deoxyuridylic acid or deoxyuridylate) (see Figure 14).
Steps 9 and 10 depend on NADPH and its possible cellular level; therefore, this reaction might occur in the cytoplasm rather than the nucleus. Furthermore, it might interact with additional pathways, e.g., the pentose phosphate pathway (PPP) and the methylglyoxal pathway [85,125]. Therefore, it raises more critical questions: Does de novo pyrimidine biosynthesis occur in the nucleus or cytoplasm or even in the mitochondrion? Does the nucleus have its own de novo pyrimidine biosynthesis pathway, while the mitochondrion has a separate biosynthetic pathway?
Because this step supports DNA synthesis, NADPH is crucial for DNA synthesis.
Ribonucleotide reductase (RR), also known as ribonucleoside diphosphate reductase (rNDP), is a ubiquitous cytosolic enzyme with an optimal pH of around 7.5 [126].
RR is expressed in many tumors, e.g., breast, pancreas, lung, adrenal cortex, etc., and it is a prognostic marker and a therapeutic target [127,128,129,130,131,132,133,134].
Some examples of RR inhibitors are (-)-epicatechin 2-aminobenzohydroxamic acid, 5′-O-valproyl-3′-C-methyladenosine, cisplatin, chlorambucil, desferrioxamine, gemcitabine, and hydroxyurea [135,136,137,138,139,140,141]. It has also been shown that H2O2 and glutathione have an inhibitory effect on RR in mice [139,142]. On the other hand, ATP is an RR activator [140,143,144], while p53 also activates RR during the DNA repair process [145].
  • Step 11:
dUMP is converted to dTMP via thymidylate synthase (TS), which requires 5,10-methylenetetrahydrofolate as a co-enzyme. Then, dTMP can be phosphorylated to dTDP and dTTP, which is the molecule used for DNA synthesis (Figure 15) [146].
The optimal pH range in leukemia is 7.0–8.1 [147].
TS is also a prognostic biomarker in tumors and is associated with drug resistance [148,149,150,151,152,153].
Capecitabine and 5 fluorouracil are examples of TS inhibitors [154,155,156,157].

3. Discussion

The de novo biosynthesis of pyrimidine is a complex process. Each step of the biosynthetic pathway is influenced by many factors. Part of these factors includes the redox state of the cell NADPH/NADP+, ATP, GTP levels, the optimal pH for the related enzymes, the availability of glutamine and aspartate among others. The synthesis of cytosine and uracil might occur at a higher pH compared to thiamine.
Pyrimidine is not limited to the biosynthesis of nucleic acid. It also plays crucial roles in tumor metabolism. Therefore, can pyrimidine act as an oncometabolite?
Interestingly, the optimal pH of most enzymes participating in this pathway is slightly higher than that of normal cells. An increased intracellular pH is usual in malignancies. Table 1 shows these optimal pH levels and in all cases they are above 7.3, except for thymidylate synthase. The protumoral effects of an alkaline or hyperalkaline intracellular milieu has been extensively investigated and confirmed [6,158,159].
For example, in the case of ribonucleotide reductase (RR), the key enzyme to produce nucleotides for DNA, indirect evidence shows that the optimal pH is between 7.5 and 8. These data come from the fact that at this pH range less iron is needed for the maximum activity of the enzyme [160]. However, with high levels of iron, the optimum pH is in the range of 6.5–7 [161]. RR requires iron and folic acid for its adequate functioning. This case shows the difficulties in correctly determining the optimal pH of many of the enzymes involved in nucleotide synthesis. Furthermore, tumors are high iron consumers.
Hormesis is a biphasic pharmacological dose-dependent response characterized by stimulation by a low dose and inhibition by a higher dose [162,163]. Moreover, hormesis is a redox-dependent process that might support cell death (apoptosis), such as with reactive oxygen species (ROS), an example of a stressful condition; however, the release of ROS at different concentrations supports genetic mutations.
Heat shock proteins (HSPs) are proteins (molecular chaperones) synthesized in response to stressful conditions. They play a crucial role in protein maturation and folding, so they are defense proteins that support cellular survival in stressful conditions. Therefore, it will not be surprising if many tumor cells are found to be HSP-dependent. It has been shown there is an association between HSPs, cancer, and pyrimidine synthesis. Hence, one of the future directions in managing cancer is further studying the de novo pyrimidine biosynthesis–HSP interactions and the potential opportunity that offers a new therapeutic window in managing cancer [164,165].
This review shows another face of pH deregulation and its link with increased proliferation. Hypothetically, we may assume that by lowering intracellular pH the activity of these enzymes may be slowed down, thus decreasing pyrimidine synthesis. Table 1 also shows the possible inhibitors for each enzyme.

4. Conclusions

  • Cancer cells have an increased rate of pyrimidine synthesis, as expected in a cell that must duplicate all its DNA and RNA at an accelerated pace.
  • The enzymes participating in pyrimidine synthesis have an optimal efficiency at a pH that is higher than the intracellular pH of normal cells.
  • This increased intracellular pH is constantly found in malignant cells as part of the pH paradigm.
  • On a theoretical basis, we may assume that lowering the intracellular pH will hamper the efficiency of pyrimidine synthesis and decrease tumor proliferation. There is direct and indirect evidence that intracellular acidification is a valid method for complementing standard treatment schemes.

Author Contributions

Conceptualization, S.S.A., S.J.R., K.O.A. and T.K.; resources, software M.E.I., A.H.H.B. and S.B.M.A.; data curation, D.D.M., T.M.A.C., R.A.C., A.H. and M.E.A.; writing—original draft preparation, S.T.S.A., S.J.R., T.K. and K.O.A.; supervision, K.O.A.; project administration, K.O.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Ministry of Education in Saudi Arabia grant number RUP20-01. TMAC was a fellow of the European Marie Skłodowska-Curie Innovative Training Network (ITN) pH and Ion Transport in Pancreatic Cancer–pHioniC (Grant Agreement number: 813834; H2020-MSCA-ITN-2018) and DDM a fellow of the Programma Operativo Complementare Ricerca e Innovazione, Asse I “Investimenti in Capitale Umano”—Azione I.1 “Dottorati innovativi con caratterizzazione industriale” del PON R&I 2014-2020.

Acknowledgments

The authors extend their appreciation to the Deputy Research and Innovation Ministry of Education in Saudi Arabia for funding this research work through project Number RUP20-01.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ehrlich, P. Über den jetzigen Stand der Chemotherapie. Ber. Dtsch. Chem. Ges. 1909, 42, 17–47. [Google Scholar] [CrossRef] [Green Version]
  2. Mukherjee, S. Emperor of All Maladies: A Biography of Cancer; Mukherjee, S., Ed.; Scribner: New York, NY, USA, 2010; ISBN1 9781439181713. ISBN2 1439181713. [Google Scholar]
  3. Huggins, C.; Hodges, C.V. Studies on prostatic cancer. I. The effect of castration, of estrogen and androgen injection on serum phosphatases in metastatic carcinoma of the prostate. CA Cancer J. Clin. 1972, 22, 293–297. [Google Scholar] [CrossRef] [PubMed]
  4. Huggins, C.; Scott, W.W.; Hodges, C.V. Studies on Prostatic Cancer. III. The Effects of Fever, of Desoxycorticosterone and of Estrogen on Clinical Patients with Metastatic Carcinoma of the Prostate1,2. J. Urol. 1941, 46, 997–1006. [Google Scholar] [CrossRef]
  5. Farber, S. Some observations on the effect of folic acid antagonists on acute leukemia and other forms of incurable cancer. Blood 1949, 4, 160–167. [Google Scholar] [CrossRef] [Green Version]
  6. Alfarouk, K.O.; Verduzco, D.; Rauch, C.; Muddathir, A.K.; Adil, H.H.B.; Elhassan, G.O.; Ibrahim, M.E.; David Polo Orozco, J.; Cardone, R.A.; Reshkin, S.J.; et al. Glycolysis, tumor metabolism, cancer growth and dissemination. A new pH-based etiopathogenic perspective and therapeutic approach to an old cancer question. Oncoscience 2014, 1, 777–802. [Google Scholar] [CrossRef]
  7. Johnson, J.D.; Epel, D.; Paul, M. Intracellular pH and activation of sea urchin eggs after fertilisation. Nature 1976, 262, 661–664. [Google Scholar] [CrossRef]
  8. Johnson, C.H.; Epel, D. Intracellular pH of sea urchin eggs measured by the dimethyloxazolidinedione (DMO) method. J. Cell Biol. 1981, 89, 284–291. [Google Scholar] [CrossRef] [Green Version]
  9. Dubé, F.; Schmidt, T.; Johnson, C.H.; Epel, D. The hierarchy of requirements for an elevated intracellular pH during early development of sea urchin embryos. Cell 1985, 40, 657–666. [Google Scholar] [CrossRef]
  10. Lopo, A.; Vacquier, V.D. The rise and fall of intracellular pH of sea urchin eggs after fertilisation. Nature 1977, 269, 590–592. [Google Scholar] [CrossRef]
  11. Grainger, J.L.; Winkler, M.M.; Shen, S.S.; Steinhardt, R.A. Intracellular pH controls protein synthesis rate in the sea urchin egg and early embryo. Dev. Biol. 1979, 68, 396–406. [Google Scholar] [CrossRef]
  12. Reshkin, S.J.; Bellizzi, A.; Caldeira, S.; Albarani, V.; Malanchi, I.; Poignee, M.; Alunni-Fabbroni, M.; Casavola, V.; Tommasino, M. Na+/H+ exchanger-dependent intracellular alkalinization is an early event in malignant transformation and plays an essential role in the development of subsequent transformation-associated phenotypes. FASEB J. 2000, 14, 2185–2197. [Google Scholar] [CrossRef] [Green Version]
  13. Fairbanks, L.D.; Bofill, M.; Ruckemann, K.; Simmonds, H.A. Importance of ribonucleotide availability to proliferating T-lymphocytes from healthy humans. Disproportionate expansion of pyrimidine pools and contrasting effects of de novo synthesis inhibitors. J. Biol. Chem. 1995, 270, 29682–29689. [Google Scholar] [CrossRef] [Green Version]
  14. Lehninger, A.L.; Nelson, D.L.; Cox, M.M. Lehninger Principles of Biochemistry; Macmillan Learning: Hong Kong, China, 2005. [Google Scholar]
  15. Tong, X.; Zhao, F.; Thompson, C.B. The molecular determinants of de novo nucleotide biosynthesis in cancer cells. Curr. Opin. Genet. Dev. 2009, 19, 32–37. [Google Scholar] [CrossRef] [Green Version]
  16. Lane, A.N.; Fan, T.W.M. Regulation of mammalian nucleotide metabolism and biosynthesis. Nucleic Acids Res. 2015, 43, 2466–2485. [Google Scholar] [CrossRef] [Green Version]
  17. Halbrook, C.J.; Pontious, C.; Kovalenko, I.; Lapienyte, L.; Dreyer, S.; Lee, H.J.; Thurston, G.; Zhang, Y.; Lazarus, J.; Sajjakulnukit, P.; et al. Macrophage-Released Pyrimidines Inhibit Gemcitabine Therapy in Pancreatic Cancer. Cell Metab. 2019, 29, 1390–1399.e6. [Google Scholar] [CrossRef]
  18. Bhutia, Y.D.; Ganapathy, V. Glutamine transporters in mammalian cells and their functions in physiology and cancer. Biochim. Biophys. Acta 2016, 1863, 2531–2539. [Google Scholar] [CrossRef]
  19. Lallous, N.; Grande-García, A.; Molina, R.; Ramón-Maiques, S. Expression, purification, crystallization and preliminary X-ray diffraction analysis of the dihydroorotase domain of human CAD. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 2012, 68, 1341–1345. [Google Scholar] [CrossRef] [Green Version]
  20. Robitaille, A.M.; Christen, S.; Shimobayashi, M.; Cornu, M.; Fava, L.L.; Moes, S.; Prescianotto-Baschong, C.; Sauer, U.; Jenoe, P.; Hall, M.N. Quantitative phosphoproteomics reveal mTORC1 activates de novo pyrimidine synthesis. Science 2013, 339, 1320–1323. [Google Scholar] [CrossRef]
  21. Graves, L.M.; Guy, H.I.; Kozlowski, P.; Huang, M.; Lazarowski, E.; Pope, R.M.; Collins, M.A.; Dahlstrand, E.N.; Earp, H.S.; Evans, D.R. Regulation of carbamoyl phosphate synthetase by MAP kinase. Nature 2000, 403, 328–332. [Google Scholar] [CrossRef]
  22. Carrey, E.A.; Campbell, D.G.; Hardie, D.G. Phosphorylation and activation of hamster carbamyl phosphate synthetase II by cAMP-dependent protein kinase. A novel mechanism for regulation of pyrimidine nucleotide biosynthesis. EMBO J. 1985, 4, 3735–3742. [Google Scholar] [CrossRef]
  23. Evans, D.R.; Guy, H.I. Mammalian pyrimidine biosynthesis: Fresh insights into an ancient pathway. J. Biol. Chem. 2004, 279, 33035–33038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Aoki, T.; Weber, G. Carbamoyl phosphate synthetase (glutamine-hydrolyzing): Increased activity in cancer cells. Science 1981, 212, 463–464. [Google Scholar] [CrossRef] [PubMed]
  25. Weber, G.; Reardon, M. Regulation of carbamoyl-phosphate synthase II. Adv. Enzym. Regul. 1986, 25, 65–83. [Google Scholar] [CrossRef]
  26. Reardon, M.A.; Weber, G. Increased synthesis of carbamoyl-phosphate synthase II (EC 6.3.5.5) in hepatoma 3924A. Cancer Res. 1986, 46, 3673–3676. [Google Scholar]
  27. Reardon, M.A.; Dixon, J.E.; Weber, G. Increased messenger RNA concentration for carbamoyl-phosphate synthase II in hepatoma 3924A. Biochem. Biophys. Res. Commun. 1987, 147, 494–500. [Google Scholar] [CrossRef]
  28. Makoff, A.J.; Radford, A. Genetics and biochemistry of carbamoyl phosphate biosynthesis and its utilization in the pyrimidine biosynthetic pathway. Microbiol. Rev. 1978, 42, 307–328. [Google Scholar] [CrossRef]
  29. Reshkin, S.J.; Cardone, R.A.; Harguindey, S. Na+-H+ exchanger, pH regulation and cancer. Recent Pat. Anticancer Drug Discov. 2013, 8, 85–99. [Google Scholar] [CrossRef]
  30. Bhagavan, N.V.; Ha, C.-E. Nucleotide Metabolism. In Essentials of Medical Biochemistry; Academic Press: Cambridge, MA, USA, 2015; pp. 465–487. [Google Scholar] [CrossRef]
  31. Valvezan, A.J.; Turner, M.; Belaid, A.; Lam, H.C.; Miller, S.K.; McNamara, M.C.; Baglini, C.; Housden, B.E.; Perrimon, N.; Kwiatkowski, D.J.; et al. mTORC1 Couples Nucleotide Synthesis to Nucleotide Demand Resulting in a Targetable Metabolic Vulnerability. Cancer Cell 2017, 32, 624–638.e5. [Google Scholar] [CrossRef] [Green Version]
  32. Ben-Sahra, I.; Howell, J.J.; Asara, J.M.; Manning, B.D. Stimulation of de novo pyrimidine synthesis by growth signaling through mTOR and S6K1. Science 2013, 339, 1323–1328. [Google Scholar] [CrossRef] [Green Version]
  33. Sebolt, J.S.; Aoki, T.; Eble, J.N.; Glover, J.L.; Weber, G. Inactivation by acivicin of carbamoyl-phosphate synthetase II of human colon carcinoma. Biochem. Pharmacol. 1985, 34, 97–100. [Google Scholar] [CrossRef]
  34. Stieglitz, K.A.; Xia, J.; Kantrowitz, E.R. The First High pH Structure of Escherichia coli Aspartate Transcarbamoylase. Proteins 2009, 74, 318. [Google Scholar] [CrossRef] [Green Version]
  35. Bresnick, E.; Mossé, H. Aspartate carbamoyltransferase from rat liver. Biochem. J. 1966, 101, 63–69. [Google Scholar] [CrossRef] [Green Version]
  36. Johnson, R.K.; Inouye, T.; Goldin, A.; Stark, G.R. Antitumor activity of N-(phosphonacetyl)-L-aspartic acid, a transition-state inhibitor of aspartate transcarbamylase. Cancer Res. 1976, 36, 2720–2725. [Google Scholar]
  37. Lei, Z.; Wang, B.; Lu, Z.; Wang, N.; Tan, H.; Zheng, J.; Jia, Z. New regulatory mechanism-based inhibitors of aspartate transcarbamoylase for potential anticancer drug development. FEBS J. 2020, 287, 3579–3599. [Google Scholar] [CrossRef]
  38. Jayaram, H.N.; Cooney, D.A.; Vistica, D.T.; Kariya, S.; Johnson, R.K. Mechanisms of sensitivity or resistance of murine tumors to N-(phosphonacetyl)-L-aspartate (PALA). Cancer Treat. Rep. 1979, 63, 1291–1302. [Google Scholar]
  39. Ruiz-Ramos, A.; Velázquez-Campoy, A.; Grande-García, A.; Moreno-Morcillo, M.; Ramón-Maiques, S. Structure and Functional Characterization of Human Aspartate Transcarbamoylase, the Target of the Anti-tumoral Drug PALA. Structure 2016, 24, 1081–1094. [Google Scholar] [CrossRef] [Green Version]
  40. Baillon, J.; Guichard, M.; Malaise, E.P.; Hervé, G. Kinetic parameters of aspartate transcarbamylase in human normal and tumoral cell lines. Cancer Res. 1983, 43, 2277–2282. [Google Scholar]
  41. O’Dwyer, P.J. The role of low-dose PALA in biochemical modulation. Pharmacol. Ther. 1990, 48, 371–380. [Google Scholar] [CrossRef]
  42. Balbaa, M.; Abdel-Megeed, M.; Diab, T.; Mansour, H. Inhibition of mammalian aspartate transcarbamylase by quinazolinone derivatives. J. Enzym. Inhib. Med. Chem. 2008, 23, 483–492. [Google Scholar] [CrossRef]
  43. Bidigare, R.R.; Sander, E.G.; Pettigrew, D.W. Evidence for a pH-dependent isomerization of Clostridium oroticum dihydroorotase. Biochim. Biophys. Acta 1985, 831, 159–160. [Google Scholar] [CrossRef]
  44. Porter, T.N.; Li, Y.; Raushel, F.M. Mechanism of the dihydroorotase reaction. Biochemistry 2004, 43, 16285–16292. [Google Scholar] [CrossRef] [PubMed]
  45. Del Caño-Ochoa, F.; Grande-García, A.; Reverte-López, M.; D’Abramo, M.; Ramón-Maiques, S. Characterization of the catalytic flexible loop in the dihydroorotase domain of the human multi-enzymatic protein CAD. J. Biol. Chem. 2018, 293, 18903–18913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Simmer, J.P.; Kelly, R.E.; Rinker, A.G.; Zimmermann, B.H.; Scully, J.L.; Kim, H.; Evans, D.R. Mammalian dihydroorotase: Nucleotide sequence, peptide sequences, and evolution of the dihydroorotase domain of the multifunctional protein CAD. Proc. Natl. Acad. Sci. USA 1990, 87, 174–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Christopherson, R.I.; Jones, M.E. The effects of pH and inhibitors upon the catalytic activity of the dihydroorotase of multienzymatic protein pyr1-3 from mouse Ehrlich ascites carcinoma. J. Biol. Chem. 1980, 255, 3358–3370. [Google Scholar] [CrossRef]
  48. Christopherson, R.I.; Jones, M.E. Interconversion of carbamayl-L-aspartate and L-dihydroorotate by dihydroorotase from mouse Ehrlich ascites carcinoma. J. Biol. Chem. 1979, 254, 12506–12512. [Google Scholar] [CrossRef]
  49. Chen, K.F.; Lai, Y.Y.; Sun, H.S.; Tsai, S.J. Transcriptional repression of human cad gene by hypoxia inducible factor-1α. Nucleic Acids Res. 2005, 33, 5190. [Google Scholar] [CrossRef]
  50. Zhao, J.; Tian, M.; Zhang, S.; Delfarah, A.; Gao, R.; Rao, Y.; Savas, A.C.; Lu, A.; Bubb, L.; Lei, X.; et al. Deamidation Shunts RelA from Mediating Inflammation to Aerobic Glycolysis. Cell Metab. 2020, 31, 937–955.e7. [Google Scholar] [CrossRef]
  51. Ding, C.; He, J.; Liao, W.; Chen, J.; Li, J.; He, G.; Jia, Z.; Zhang, Y.; Luo, J.; Dai, X.; et al. Regulation of WNT/β-catenin signaling by carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD) in colorectal cancer cell|Semantic Scholar. Int. J. Clin. Exp. Med. 2017, 10, 16243–16253. [Google Scholar]
  52. Guan, H.H.; Huang, Y.H.; Lin, E.S.; Chen, C.J.; Huang, C.Y. Structural basis for the interaction modes of dihydroorotase with the anticancer drugs 5-fluorouracil and 5-aminouracil. Biochem. Biophys. Res. Commun. 2021, 551, 33–37. [Google Scholar] [CrossRef]
  53. Kinder, D.H.; Frank, S.K.; Ames, M.M. Analogs of carbamyl aspartate as inhibitors of dihydroorotase: Preparation of boronic acid transition-state analogs and a zinc chelator carbamylhomocysteine. J. Med. Chem. 2002, 33, 819–823. [Google Scholar] [CrossRef]
  54. Mohsen, A.W.A.; Rigby, S.E.J.; Jensen, K.F.; Munro, A.W.; Scrutton, N.S. Thermodynamic basis of electron transfer in dihydroorotate dehydrogenase B from Lactococcus lactis: Analysis by potentiometry, EPR spectroscopy, and ENDOR spectroscopy. Biochemistry 2004, 43, 6498–6510. [Google Scholar] [CrossRef]
  55. Rowland, P.; Nørager, S.; Jensen, K.F.; Larsen, S. Structure of dihydroorotate dehydrogenase B: Electron transfer between two flavin groups bridged by an iron-sulphur cluster. Structure 2000, 8, 1227–1238. [Google Scholar] [CrossRef]
  56. Argyrou, A.; Washabaugh, M.W.; Pickart, C.M. Dihydroorotate dehydrogenase from Clostridium oroticum is a class 1B enzyme and utilizes a concerted mechanism of catalysis. Biochemistry 2000, 39, 10373–10384. [Google Scholar] [CrossRef]
  57. Cheleski, J.; Rocha, J.R.; Pinheiro, M.P.; Wiggers, H.J.; Da Silva, A.B.F.; Nonato, M.C.; Montanari, C.A. Novel insights for dihydroorotate dehydrogenase class 1A inhibitors discovery. Eur. J. Med. Chem. 2010, 45, 5899–5909. [Google Scholar] [CrossRef]
  58. Kahler, A.E.; Nielsen, F.S.; Switzer, R.L. Biochemical characterization of the heteromeric Bacillus subtilis dihydroorotate dehydrogenase and its isolated subunits. Arch. Biochem. Biophys. 1999, 371, 191–201. [Google Scholar] [CrossRef]
  59. Fagan, R.L.; Nelson, M.N.; Pagano, P.M.; Palfey, B.A. Mechanism of flavin reduction in class 2 dihydroorotate dehydrogenases. Biochemistry 2006, 45, 14926–14932. [Google Scholar] [CrossRef]
  60. Liu, S.; Neidhardt, E.A.; Grossman, T.H.; Ocain, T.; Clardy, J. Structures of human dihydroorotate dehydrogenase in complex with antiproliferative agents. Structure 2000, 8, 25–33. [Google Scholar] [CrossRef] [Green Version]
  61. Miller, R.W. Dihydroorotate dehydrogenase (Neurospora). Methods Enzymol. 1978, 51, 63–69. [Google Scholar] [CrossRef]
  62. Fagan, R.L.; Palfey, B.A. Roles in binding and chemistry for conserved active site residues in the class 2 dihydroorotate dehydrogenase from Escherichia coli. Biochemistry 2009, 48, 7169–7178. [Google Scholar] [CrossRef] [Green Version]
  63. Zameitat, E.; Pierik, A.J.; Zocher, K.; Löffler, M. Dihydroorotate dehydrogenase from Saccharomyces cerevisiae: Spectroscopic investigations with the recombinant enzyme throw light on catalytic properties and metabolism of fumarate analogues. FEMS Yeast Res. 2007, 7, 897–904. [Google Scholar] [CrossRef] [Green Version]
  64. Krungkrai, J.; Cerami, A.; Henderson, G.B. Purification and characterization of dihydroorotate dehydrogenase from the rodent malaria parasite Plasmodium berghei. Biochemistry 1991, 30, 1934–1939. [Google Scholar] [CrossRef] [PubMed]
  65. Bader, B.; Knecht, W.; Fries, M.; Löffler, M. Expression, purification, and characterization of histidine-tagged rat and human flavoenzyme dihydroorotate dehydrogenase. Protein Expr. Purif. 1998, 13, 414–422. [Google Scholar] [CrossRef] [PubMed]
  66. Rawls, J.; Knecht, W.; Diekert, K.; Lill, R.; Löffler, M. Requirements for the mitochondrial import and localization of dihydroorotate dehydrogenase. Eur. J. Biochem. 2000, 267, 2079–2087. [Google Scholar] [CrossRef] [PubMed]
  67. Leban, J.; Vitt, D. Human dihydroorotate dehydrogenase inhibitors, a novel approach for the treatment of autoimmune and inflammatory diseases. Arzneimittelforschung 2011, 61, 66–72. [Google Scholar] [CrossRef]
  68. Greene, S.; Watanabe, K.; Braatz-Trulson, J.; Lou, L. Inhibition of dihydroorotate dehydrogenase by the immunosuppressive agent leflunomide. Biochem. Pharmacol. 1995, 50, 861–867. [Google Scholar] [CrossRef]
  69. Zhu, S.; Yan, X.; Xiang, Z.; Ding, H.-F.; Cui, H. Leflunomide Reduces Proliferation and Induces Apoptosis in Neuroblastoma Cells In Vitro and In Vivo. PLoS ONE 2013, 8, e71555. [Google Scholar] [CrossRef] [Green Version]
  70. Zhang, C.; Chu, M. Leflunomide: A promising drug with good antitumor potential. Biochem. Biophys. Res. Commun. 2018, 496, 726–730. [Google Scholar] [CrossRef]
  71. Chen, S.F.; Perrella, F.W.; Behrens, D.L.; Papp, L.M. Inhibition of dihydroorotate dehydrogenase activity by brequinar sodium. Cancer Res. 1992, 52, 3521–3527. [Google Scholar]
  72. Arteaga, C.L.; Brown, T.D.; Kuhn, J.G.; Shen, H.S.; O’Rourke, T.J.; Beougher, K.; Brentzel, H.J.; Von Hoff, D.D.; Weiss, G.R. Phase I clinical and pharmacokinetic trial of Brequinar sodium (DuP 785; NSC 368390). Cancer Res. 1989, 49, 4648–4653. [Google Scholar]
  73. Maroun, J.; Ruckdeschel, J.; Natale, R.; Morgan, R.; Dallaire, B.; Sisk, R.; Gyves, J. Multicenter phase II study of brequinar sodium in patients with advanced lung cancer. Cancer Chemother. Pharmacol. 1993, 32, 64–66. [Google Scholar] [CrossRef] [Green Version]
  74. Cody, R.; Stewart, D.; DeForni, M.; Moore, M.; Dallaire, B.; Azarnia, N.; Gyves, J.W. Multicenter phase II study of brequinar sodium in patients with advanced breast cancer. Am. J. Clin. Oncol. 1993, 16, 526–528. [Google Scholar] [CrossRef]
  75. Natale, R.; Wheeler, R.; Moore, M.; Dallaire, B.; Lynch, W.; Carlson, R.; Grillo-lopez, A.; Gyves, L. Multicenter phase II trial of brequinar sodium in patients with advanced melanoma. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 1992, 3, 659–660. [Google Scholar] [CrossRef]
  76. Moore, M.; Maroun, J.; Robert, F.; Natale, R.; Neidhart, J.; Dallaire, B.; Sisk, R.; Gyves, J. Multicenter phase II study of brequinar sodium in patients with advanced gastrointestinal cancer. Investig. New Drugs 1993, 11, 61–65. [Google Scholar] [CrossRef]
  77. Peters, G.J. Re-evaluation of Brequinar sodium, a dihydroorotate dehydrogenase inhibitor. Nucleosides Nucleotides Nucleic Acids 2018, 37, 666–678. [Google Scholar] [CrossRef]
  78. Sykes, D.B.; Kfoury, Y.S.; Mercier, F.E.; Wawer, M.J.; Law, J.M.; Haynes, M.K.; Lewis, T.A.; Schajnovitz, A.; Jain, E.; Lee, D.; et al. Inhibition of Dihydroorotate Dehydrogenase Overcomes Differentiation Blockade in Acute Myeloid Leukemia. Cell 2016, 167, 171–186.e15. [Google Scholar] [CrossRef] [Green Version]
  79. Zheng, H.-C.; Sun, J.-M.; Li, X.-H.; Yang, X.-F.; Zhang, Y.-C.; Xin, Y. Role of PTEN and MMP-7 expression in growth, invasion, metastasis and angiogenesis of gastric carcinoma. Pathol. Int. 2003, 53, 659–666. [Google Scholar] [CrossRef]
  80. Peters, G.J. Antipyrimidine effects of five different pyrimidine de novo synthesis inhibitors in three head and neck cancer cell lines. Nucleosides Nucleotides Nucleic Acids 2018, 37, 329–339. [Google Scholar] [CrossRef]
  81. Cuthbertson, C.R.; Guo, H.; Kyani, A.; Madak, J.T.; Arabzada, Z.; Neamati, N. The Dihydroorotate Dehydrogenase Inhibitor Brequinar Is Synergistic with ENT1/2 Inhibitors. ACS Pharmacol. Transl. Sci. 2020, 3, 1242–1252. [Google Scholar] [CrossRef]
  82. Dorasamy, M.S.; AB, A.; Nellore, K.; Wong, P.F. Synergistic inhibition of melanoma xenografts by Brequinar sodium and Doxorubicin. Biomed. Pharmacother. 2019, 110, 29–36. [Google Scholar] [CrossRef]
  83. Sykes, D.B. The emergence of dihydroorotate dehydrogenase (DHODH) as a therapeutic target in acute myeloid leukemia. Expert Opin. Ther. Targets 2018, 22, 893–898. [Google Scholar] [CrossRef] [Green Version]
  84. Mao, C.; Liu, X.; Zhang, Y.; Lei, G.; Yan, Y.; Lee, H.; Koppula, P.; Wu, S.; Zhuang, L.; Fang, B.; et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 2021, 593, 586–590. [Google Scholar] [CrossRef]
  85. Alfarouk, K.O.; Ahmed, S.B.M.; Elliott, R.L.; Benoit, A.; Alqahtani, S.S.; Ibrahim, M.E.; Bashir, A.H.H.; Alhoufie, S.T.S.; Elhassan, G.O.; Wales, C.C.; et al. The Pentose Phosphate Pathway Dynamics in Cancer and Its Dependency on Intracellular pH. Metabolites 2020, 10, 285. [Google Scholar] [CrossRef]
  86. Okuda, K.; Tatematsu, T.; Yano, M.; Nakamae, K.; Yamada, T.; Kasugai, T.; Nishida, T.; Sano, M.; Moriyama, S.; Haneda, H.; et al. The relationship between the expression of thymidylate synthase, dihydropyrimidine dehydrogenase, orotate phosphoribosyltransferase, excision repair cross-complementation group 1 and class III β-tubulin, and the therapeutic effect of S-1 or carboplatin plus paclitaxel in non-small-cell lung cancer. Mol. Clin. Oncol. 2018, 9, 21–29. [Google Scholar] [CrossRef]
  87. Sakurai, Y.; Sakamoto, K.; Sugimoto, Y.; Yoshida, I.; Masui, T.; Tonomura, S.; Inaba, K.; Shoji, M.; Nakamura, Y.; Uyama, I.; et al. Orotate phosphoribosyltransferase levels measured by a newly established enzyme-linked immunosorbent assay in gastric carcinoma. Cancer Sci. 2006, 97, 492–498. [Google Scholar] [CrossRef]
  88. Choi, I.S.; Lee, H.S.; Lee, K.W.; Kim, H.; Kim, K.H.; Kim, Y.J.; Kim, J.H.; Kim, W.H.; Lee, J.S. Biomarker analysis in patients with advanced gastric cancer treated with S-1 plus cisplatin chemotherapy: Orotate phosphoribosyltransferase expression is associated with treatment outcomes. Med. Oncol. 2011, 28, 991–998. [Google Scholar] [CrossRef]
  89. Ichikawa, W.; Uetake, H.; Shirota, Y.; Yamada, H.; Takahashi, T.; Nihei, Z.; Sugihara, K.; Sasaki, Y.; Hirayama, R. Both gene expression for orotate phosphoribosyltransferase and its ratio to dihydropyrimidine dehydrogenase influence outcome following fluoropyrimidine-based chemotherapy for metastatic colorectal cancer. Br. J. Cancer 2003, 89, 1486–1492. [Google Scholar] [CrossRef] [Green Version]
  90. Sparreboom, A.; Loos, W.J.; de Jonge, M.J.A.; Verweij, J. Clinical trial design: Incorporation of pharmacokinetic, pharmacodynamic, and pharmacogenetic principles. In Anticancer Drug Development; Elsevier: Amsterdam, The Netherlands, 2002; pp. 329–351. [Google Scholar]
  91. Mizutani, Y.; Wada, H.; Fukushima, M.; Yoshida, O.; Nakanishi, H.; Li, Y.N.; Miki, T. Prognostic significance of orotate phosphoribosyltransferase activity in bladder carcinoma. Cancer 2004, 100, 723–731. [Google Scholar] [CrossRef]
  92. Akahoshi, K.; Ban, D.; Kuboki, R.; Oba, A.; Ono, H.; Mitsunori, Y.; Kudo, A.; Tanaka, S.; Tanabe, M. Orotate phosphoribosyltransferase as a predictor of benefit from S-1 adjuvant chemotherapy for cholangiocarcinoma patients. J. Gastroenterol. Hepatol. 2019, 34, 1108–1115. [Google Scholar] [CrossRef]
  93. Yan, D.; Ryu, M.; An, E.; Park, Y.; Na, H.; Ma, J.; Tian, Y.; Cecchi, F.; Hembrough, T.; Kang, Y.; et al. Predicting survival benefit of capecitabine plus cisplatin in patients with metastatic gastric cancer patients using quantitative proteomics. Ann. Oncol. 2018, 29, viii51. [Google Scholar] [CrossRef]
  94. Ashton, R.W.; Strauss, R.S.; Chung, S.H.; Sloan, D.L. Orotate phosphoribosyltransferase from yeast: Studies of the structure of the pyrimidine substrate binding site. Arch. Biochem. Biophys. 1989, 272, 421–432. [Google Scholar] [CrossRef]
  95. Robinson, J.L.; Arson, I. Nucleotide Inhibition of Post-Orotate Pyrimidine Synthesis Pathway Enzymes of Bovine Mammary Tissue. J. Dairy Sci. 1974, 57, 1410–1413. [Google Scholar] [CrossRef]
  96. Traut, T.W.; Jones, M.E. Inhibitors of orotate phosphoribosyl-transferase and orotidine-5′-phosphate decarboxylase from mouse Ehrlich ascites cells: A procedure for analyzing the inhibition of a multi-enzyme complex. Biochem. Pharmacol. 1977, 26, 2291–2296. [Google Scholar] [CrossRef]
  97. Donini, S.; Ferraris, D.M.; Miggiano, R.; Massarotti, A.; Rizzi, M. Structural investigations on orotate phosphoribosyltransferase from Mycobacterium tuberculosis, a key enzyme of the de novo pyrimidine biosynthesis. Sci. Rep. 2017, 7, 1180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Suchi, M.; Mizuno, H.; Kawai, Y.; Tsuboi, T.; Sumi, S.; Okajima, K.; Hodgson, M.E.; Ogawa, H.; Wada, Y. Molecular cloning of the human UMP synthase gene and characterization of point mutations in two hereditary orotic aciduria families. Am. J. Hum. Genet. 1997, 60, 525–539. [Google Scholar]
  99. Gabrielsen, B.; Kirsi, J.J.; Kwong, C.D.; Carter, D.A.; Krauth, C.A.; Hanna, L.K.; Huggins, J.W.; Monath, T.P.; Kefauver, D.F.; Blough, H.A.; et al. In vitro and in vivo antiviral (RNA) evaluation of orotidine 5′-monophosphate decarboxylase inhibitors and analogues including 6-azauridine-5′-(ethyl methoxyalaninyl)phosphate (a 5′-monophosphate prodrug). Antivir. Chem. Chemother. 1994, 5, 209–220. [Google Scholar] [CrossRef]
  100. Kotra, L.; Meza-Avina, M.; Wei, L.; Buhendwa, M.; Poduch, E.; Bello, A.; Pai, E. Inhibition of orotidine 5′-monophosphate decarboxylase and its therapeutic potential. Mini Rev. Med. Chem. 2008, 8, 239–247. [Google Scholar] [CrossRef]
  101. Smiley, J.A.; Paneth, P.; O’Leary, M.H.; Bell, J.B.; Jones, M.E. Investigation of the enzymatic mechanism of yeast orotidine-5′-monophosphate decarboxylase using 13C kinetic isotope effects. Biochemistry 1991, 30, 6216–6223. [Google Scholar] [CrossRef]
  102. Appleby, T.C.; Kinsland, C.; Begley, T.P.; Ealick, S.E. The crystal structure and mechanism of orotidine 5′-monophosphate decarboxylase. Proc. Natl. Acad. Sci. USA 2000, 97, 2005–2010. [Google Scholar] [CrossRef] [Green Version]
  103. Xu, Y.; Johansson, M.; Karlsson, A. Human UMP-CMP kinase 2, a novel nucleoside monophosphate kinase localized in mitochondria. J. Biol. Chem. 2008, 283, 1563–1571. [Google Scholar] [CrossRef] [Green Version]
  104. Liou, J.-Y.; Dutschman, G.E.; Lam, W.; Jiang, Z.; Cheng, Y.-C. Characterization of human UMP/CMP kinase and its phosphorylation of D- and L-form deoxycytidine analogue monophosphates. Cancer Res. 2002, 62, 1624–1631. [Google Scholar]
  105. Van Rompay, A.R.; Johansson, M.; Karlsson, A. Phosphorylation of deoxycytidine analog monophosphates by UMP-CMP kinase: Molecular characterization of the human enzyme. Mol. Pharmacol. 1999, 56, 562–569. [Google Scholar] [CrossRef] [Green Version]
  106. Alexandre, J.A.C.; Roy, B.; Topalis, D.; Périgaud, C.; Deville-Bonne, D. Enantio-selectivity of human nucleoside monophosphate kinases. Nucleosides Nucleotides Nucleic Acids 2007, 26, 1375–1379. [Google Scholar] [CrossRef]
  107. Liu, N.Q.; De Marchi, T.; Timmermans, A.; Trapman-Jansen, A.M.A.C.; Foekens, R.; Look, M.P.; Smid, M.; Van Deurzen, C.H.M.; Span, P.N.; Sweep, F.C.G.J.; et al. Prognostic significance of nuclear expression of UMP-CMP kinase in triple negative breast cancer patients. Sci. Rep. 2016, 6, 32027. [Google Scholar] [CrossRef] [Green Version]
  108. Carvalho, T.M.A.; Di Molfetta, D.; Greco, M.R.; Koltai, T.; Alfarouk, K.O.; Reshkin, S.J.; Cardone, R.A. Tumor Microenvironment Features and Chemoresistance in Pancreatic Ductal Adenocarcinoma: Insights into Targeting Physicochemical Barriers and Metabolism as Therapeutic Approaches. Cancers 2021, 13, 6135. [Google Scholar] [CrossRef]
  109. Chen, S.; Wang, X.; Ye, X.; Ma, D.; Chen, C.; Cai, J.; Fu, Y.; Cheng, X.; Chen, Y.; Gong, X.; et al. Identification of Human UMP/CMP Kinase 1 as Doxorubicin Binding Target Using Protein Microarray. SLAS Discov. 2017, 22, 1007–1015. [Google Scholar] [CrossRef] [Green Version]
  110. Apte, U. Galactosamine. In Encyclopedia of Toxicology; Elsevier: Amsterdam, The Netherlands, 2014; pp. 689–690. ISBN 9780123864543. [Google Scholar]
  111. Preiss, J. Biochemistry and Molecular Biology of Glycogen Synthesis in Bacteria and Mammals and Starch Synthesis in Plants. In Comprehensive Natural Products II; Elsevier: Amsterdam, The Netherlands, 2010; Volume 6, pp. 429–491. ISBN 9780080453828. [Google Scholar]
  112. Hubbard, J.A.; Binder, D.K. Gliotransmitters. In Astrocytes and Epilepsy; Academic Press: Cambridge, MA, USA, 2016; pp. 53–73. [Google Scholar] [CrossRef]
  113. Parks, R.E.E.; Aganwal, R.P.P. Nucleoside Diphosphokinases. Enzymes 1973, 8, 307–333. [Google Scholar] [CrossRef]
  114. Steeg, P.S.; Bevilacqua, G.; Kopper, L.; Thorgeirsson, U.P.; Talmadge, J.E.; Liotta, L.A.; Sobel, M.E. Evidence for a novel gene associated with low tumor metastatic potential. J. Natl. Cancer Inst. 1988, 80, 200–204. [Google Scholar] [CrossRef]
  115. Tee, Y.T.; Chen, G.D.; Lin, L.Y.; Ko, J.L.; Wang, P.H. Nm23-H1: A metastasis-associated gene. Taiwan J. Obstet. Gynecol. 2006, 45, 107–113. [Google Scholar] [CrossRef] [Green Version]
  116. Boissan, M.; Lacombe, M.L. NM23, an example of a metastasis suppressor gene. Bull. Cancer 2012, 99, 431–440. [Google Scholar] [CrossRef]
  117. Lam, S.C.T.; Packham, M.A. Isolation and kinetic studies of nucleoside diphosphokinase from human platelets and effects of cAMP phosphodiesterase inhibitors. Biochem. Pharmacol. 1986, 35, 4449–4455. [Google Scholar] [CrossRef]
  118. Zhu, M.; Sun, W.; Wang, Y.; Meng, J.; Zhang, D.; Guo, T.; Ouyang, P.; Ying, H.; Xie, J. Engineered cytidine triphosphate synthetase with reduced product inhibition. Protein Eng. Des. Sel. 2014, 27, 225–233. [Google Scholar] [CrossRef] [Green Version]
  119. Williams, J.C.; Kizaki, H.; Weber, G.; Morris, H.P. Increased CTP synthetase activity in cancer cells. Nature 1978, 271, 71–73. [Google Scholar] [CrossRef]
  120. Kassel, K.M.; Au, D.R.; Higgins, M.J.; Hines, M.; Graves, L.M. Regulation of Human Cytidine Triphosphate Synthetase 2 by Phosphorylation. J. Biol. Chem. 2010, 285, 33727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Verschuur, A.C.; Van Gennip, A.H.; Leen, R.; Muller, E.J.; Elzinga, L.; Voûte, P.A.; Van Kuilenburg, A.B.P. Cyclopentenyl cytosine inhibits cytidine triphosphate synthetase in paediatric acute non-lymphocytic leukaemia: A promising target for chemotherapy. Eur. J. Cancer 2000, 36, 627–635. [Google Scholar] [CrossRef]
  122. Richards, N.G.J.; Humkey, R.N.; Li, K.; Meyer, M.E.; Córdova de Sintjago, T.C. Tunnels and Intermediates in the Glutamine-Dependent Amidotransferases. In Comprehensive Natural Products II Chemistry and Biology; Elsevier: Amsterdam, The Netherlands, 2010; Volume 8, pp. 161–230. [Google Scholar] [CrossRef]
  123. Endrizzi, J.A.; Kim, H.; Anderson, P.M.; Baldwin, E.P. Mechanisms of Product Feedback Regulation and Drug Resistance in Cytidine Triphosphate Synthetases from the Structure of a CTP-Inhibited Complex. Biochemistry 2005, 44, 13491. [Google Scholar] [CrossRef] [Green Version]
  124. Higgins, M.J.; Loiselle, D.; Haystead, T.A.; Graves, L.M. Human cytidine triphosphate synthetase 1 interacting proteins. Nucleosides. Nucleotides Nucleic Acids 2008, 27, 850–857. [Google Scholar] [CrossRef]
  125. Alfarouk, K.O.; Alqahtani, S.S.; Alshahrani, S.; Morgenstern, J.; Supuran, C.T.; Reshkin, S.J. The possible role of methylglyoxal metabolism in cancer. J. Enzym. Inhib. Med. Chem. 2021, 36, 2010–2015. [Google Scholar] [CrossRef]
  126. Eklund, H.; Uhlin, U.; Färnegårdh, M.; Logan, D.T.; Nordlund, P. Structure and function of the radical enzyme ribonucleotide reductase. Prog. Biophys. Mol. Biol. 2001, 77, 177–268. [Google Scholar] [CrossRef]
  127. Aye, Y.; Li, M.; Long, M.J.C.; Weiss, R.S. Ribonucleotide reductase and cancer: Biological mechanisms and targeted therapies. Oncogene 2014, 34, 2011–2021. [Google Scholar] [CrossRef]
  128. Su, Y.F.; Wu, T.F.; Ko, J.L.; Tsai, H.T.; Tee, Y.T.; Chien, M.H.; Chou, C.H.; Lin, W.L.; Low, H.Y.; Chou, M.Y.; et al. The Expression of Ribonucleotide Reductase M2 in the Carcinogenesis of Uterine Cervix and Its Relationship with Clinicopathological Characteristics and Prognosis of Cancer Patients. PLoS ONE 2014, 9, e91644. [Google Scholar] [CrossRef]
  129. Zhang, H.; Liu, X.; Warden, C.D.; Huang, Y.; Loera, S.; Xue, L.; Zhang, S.; Chu, P.; Zheng, S.; Yen, Y. Prognostic and therapeutic significance of ribonucleotide reductase small subunit M2 in estrogen-negative breast cancers. BMC Cancer 2014, 14, 664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Nuno, M.F.; Cerqueira, S.A.; Ramos, M.J. Ribonucleotide Reductase: A Critical Enzyme for Cancer Chemotherapy and Antiviral Agents. Recent Pat. Anticancer Drug Discov. 2007, 2, 11–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Morikawa, T.; Hino, R.; Uozaki, H.; Maeda, D.; Ushiku, T.; Shinozaki, A.; Sakatani, T.; Fukayama, M. Expression of ribonucleotide reductase M2 subunit in gastric cancer and effects of RRM2 inhibition in vitro. Hum. Pathol. 2010, 41, 1742–1748. [Google Scholar] [CrossRef] [PubMed]
  132. Xie, H.; Jiang, W.; Jiang, J.; Wang, Y.; Kim, R.; Liu, X.; Liu, X. Predictive and prognostic roles of ribonucleotide reductase M1 in resectable pancreatic adenocarcinoma. Cancer 2013, 119, 173–181. [Google Scholar] [CrossRef]
  133. Volante, M.; Terzolo, M.; Fassnacht, M.; Rapa, I.; Germano, A.; Sbiera, S.; Daffara, F.; Sperone, P.; Scagliotti, G.; Allolio, B.; et al. Ribonucleotide reductase large subunit (RRM1) gene expression may predict efficacy of adjuvant mitotane in adrenocortical cancer. Clin. Cancer Res. 2012, 18, 3452–3461. [Google Scholar] [CrossRef] [Green Version]
  134. Souglakos, J.; Boukovinas, I.; Taron, M.; Mendez, P.; Mavroudis, D.; Tripaki, M.; Hatzidaki, D.; Koutsopoulos, A.; Stathopoulos, E.; Georgoulias, V.; et al. Ribonucleotide reductase subunits M1 and M2 mRNA expression levels and clinical outcome of lung adenocarcinoma patients treated with docetaxel/gemcitabine. Br. J. Cancer 2008, 98, 1710–1715. [Google Scholar] [CrossRef] [Green Version]
  135. Schroeder, P.; Voevodskaya, N.; Klotz, L.O.; Brenneisen, P.; Gräslund, A.; Sies, H. Loss of the tyrosyl radical in mouse ribonucleotide reductase by (-)-epicatechin. Biochem. Biophys. Res. Commun. 2005, 326, 614–617. [Google Scholar] [CrossRef]
  136. Elford, H.L.; Van’t Riet, B.; Wampler, G.L.; Lin, A.L.; Elford, R.M. Regulation of ribonucleotide reductase in mammalian cells by chemotherapeutic agents. Adv. Enzym. Regul. 1980, 19, 151–168. [Google Scholar] [CrossRef]
  137. Holmgren, A.; Sengupta, R. The use of thiols by ribonucleotide reductase. Free Radic. Biol. Med. 2010, 49, 1617–1628. [Google Scholar] [CrossRef]
  138. Petrelli, R.; Meli, M.; Vita, P.; Torquati, I.; Ferro, A.; Vodnala, M.; D’Alessandro, N.; Tolomeo, M.; Del Bello, F.; Kusumanchi, P.; et al. From the covalent linkage of drugs to novel inhibitors of ribonucleotide reductase: Synthesis and biological evaluation of valproic esters of 3′-C-methyladenosine. Bioorg. Med. Chem. Lett. 2014, 24, 5304–5309. [Google Scholar] [CrossRef]
  139. Lammers, M.; Follmann, H. The ribonucleotide reductases—A unique group of metalloenzymes essential for cell proliferation. In Inorganic Elements in Biochemistry; Springer: Berlin/Heidelberg, Germany, 1983; pp. 27–91. [Google Scholar]
  140. Zhu, L.; Zhou, B.; Chen, X.; Jiang, H.; Shao, J.; Yen, Y. Inhibitory mechanisms of heterocyclic carboxaldehyde thiosemicabazones for two forms of human ribonucleotide reductase. Biochem. Pharmacol. 2009, 78, 1178–1185. [Google Scholar] [CrossRef]
  141. Sigmond, J.; Kamphuis, J.A.E.; Laan, A.C.; Hoebe, E.K.; Bergman, A.M.; Peters, G.J. The synergistic interaction of gemcitabine and cytosine arabinoside with the ribonucleotide reductase inhibitor triapine is schedule dependent. Biochem. Pharmacol. 2007, 73, 1548–1557. [Google Scholar] [CrossRef]
  142. Sato, A.; Bacon, P.E.; Cory, J.G. Studies on the differential mechanisms of inhibition of ribonucleotide reductase by specific inhibitors of the non-heme iron subunit. Adv. Enzym. Regul. 1984, 22, 231–241. [Google Scholar] [CrossRef]
  143. Krishnan, K.; Prathiba, K.; Jayaprakash, V.; Basu, A.; Mishra, N.; Zhou, B.; Hu, S.; Yen, Y. Synthesis and ribonucleotide reductase inhibitory activity of thiosemicarbazones. Bioorg. Med. Chem. Lett. 2008, 18, 6248–6250. [Google Scholar] [CrossRef]
  144. Holmgren, A. Regulation of ribonucleotide reductase. Curr. Top. Cell. Regul. 1981, 19, 47–76. [Google Scholar] [CrossRef]
  145. Smith, P.; Zhou, B.; Ho, N.; Yuan, Y.C.; Su, L.; Tsai, S.C.; Yen, Y. 2.6 A X-ray crystal structure of human p53R2, a p53-inducible ribonucleotide reductase. Biochemistry 2009, 48, 11134–11141. [Google Scholar] [CrossRef] [Green Version]
  146. Gropper, S.S.; Smith, J.L.; Carr, T.P. Advanced Nutrition and Human Metabolism, 8th ed.; Cengage Learning: Boston, MA, USA, 2021; ISBN1 0357450108. ISBN2 9780357450109. [Google Scholar]
  147. Lockshin, A.; Moran, R.G.; Danenberg, P.V. Thymidylate synthetase purified to homogeneity from human leukemic cells. Proc. Natl. Acad. Sci. USA 1979, 76, 750–754. [Google Scholar] [CrossRef] [Green Version]
  148. Mirza, A.; Brown, M.; McNulty, C.; Valentine, J.; Annesley, A.; Galloway, S.; Welch, I.; West, C.M.; Pritchard, S. A pilot study to investigate the role of thymidylate synthase as a marker of prognosis for neoadjuvant chemotherapy in gastric and gastro-oesophageal junction adenocarcinoma. Gastroenterol. Res. Pract. 2013, 2013, 502153. [Google Scholar] [CrossRef] [Green Version]
  149. Squires, M.H.; Fisher, S.B.; Fisher, K.E.; Patel, S.H.; Kooby, D.A.; El-Rayes, B.F.; Staley, C.A.; Farris, A.B.; Maithel, S.K. Differential expression and prognostic value of ERCC1 and thymidylate synthase in resected gastric adenocarcinoma. Cancer 2013, 119, 3242–3250. [Google Scholar] [CrossRef] [Green Version]
  150. Houghton, P.J.; Houghton, J.A.; Germain, G.; Torrance, P.M. Development and characterization of a human colon adenocarcinoma xenograft deficient in thymidine salvage. Cancer Res. 1987, 47, 2117–2122. [Google Scholar]
  151. Yoo, B.K.; Emdad, L.; Gredler, R.; Fuller, C.; Dumur, C.I.; Jones, K.H.; Jackson-Cook, C.; Su, Z.Z.; Chen, D.; Saxena, U.H.; et al. Transcription factor Late SV40 Factor (LSF) functions as an oncogene in hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 2010, 107, 8357–8362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Calascibetta, A.; Martorana, A.; Cabibi, D.; Aragona, F.; Sanguedolce, R. Relationship between thymidylate synthase and p53 and response to FEC versus taxane adjuvant chemotherapy for breast carcinoma. J. Chemother. 2011, 23, 354–357. [Google Scholar] [CrossRef] [PubMed]
  153. Pestalozzi, B.C.; Peterson, H.F.; Gelber, R.D.; Goldhirsch, A.; Gusterson, B.A.; Trihia, H.; Lindtner, J.; Cortés-Funes, H.; Simmoncini, E.; Byrne, M.J.; et al. Prognostic importance of thymidylate synthase expression in early breast cancer. J. Clin. Oncol. 1997, 15, 1923–1931. [Google Scholar] [CrossRef] [Green Version]
  154. Rustum, Y.M. Thymidylate synthase: A critical target in cancer therapy? Front. Biosci. 2004, 9, 2467–2473. [Google Scholar] [CrossRef] [PubMed]
  155. Jason, T.L.H.; Berg, R.W.; Vincent, M.D.; Koropatnick, J. Antisense targeting of thymidylate synthase (TS) mRNA increases TS gene transcription and TS protein: Effects on human tumor cell sensitivity to TS enzyme-inhibiting drugs. Gene Expr. 2007, 13, 227–239. [Google Scholar] [CrossRef] [Green Version]
  156. Giovannetti, E.; Backus, H.H.J.; Wouters, D.; Ferreira, C.G.; Van Houten, V.M.M.; Brakenhoff, R.H.; Poupon, M.F.; Azzarello, A.; Pinedo, H.M.; Peters, G.J. Changes in the status of p53 affect drug sensitivity to thymidylate synthase (TS) inhibitors by altering TS levels. Br. J. Cancer 2007, 96, 769–775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Luo, Y.; Walla, M.; Wyatt, M.D. Uracil incorporation into genomic DNA does not predict toxicity caused by chemotherapeutic inhibition of thymidylate synthase. DNA Repair 2008, 7, 162–169. [Google Scholar] [CrossRef] [Green Version]
  158. Cardone, R.A.R.; Alfarouk, K.O.K.; Elliott, R.L.R.; Alqahtani, S.S.S.; Ahmed, S.S.B.M.; Aljarbou, A.A.N.; Greco, M.; Cannone, S.; Reshkin, S.S.J. The Role of Sodium Hydrogen Exchanger 1 in Dysregulation of Proton Dynamics and Reprogramming of Cancer Metabolism as a Sequela. Int. J. Mol. Sci. 2019, 20, 3694. [Google Scholar] [CrossRef] [Green Version]
  159. Schwartz, L.; Seyfried, T.; Alfarouk, K.O.; Da Veiga Moreira, J.; Fais, S. Out of Warburg effect: An effective cancer treatment targeting the tumor specific metabolism and dysregulated pH. Semin. Cancer Biol. 2017, 43, 134–138. [Google Scholar] [CrossRef]
  160. Moore, E.C. The Effects of Ferrous Ion and Dithioerythritol on Inhibition by Hydroxyurea of Ribonucleotide Reductase1|Cancer Research|American Association for Cancer Research. Cancer Res. 1969, 29, 291–295. [Google Scholar]
  161. Moore, E.C.; Reichard, P. Enzymatic synthesis of deoxyribonucleotides. vi. the cytidine diphosphate reductase system from novikoff hepatoma. J. Biol. Chem. 1964, 239, 3453–3456. [Google Scholar] [CrossRef]
  162. Trovato Salinaro, A.; Pennisi, M.; Di Paola, R.; Scuto, M.; Crupi, R.; Cambria, M.T.; Ontario, M.L.; Tomasello, M.; Uva, M.; Maiolino, L.; et al. Neuroinflammation and Neurohormesis in the Pathogenesis of Alzheimer’s Disease and Alzheimer-Linked Pathologies: Modulation by Nutritional Mushrooms. Immun. Ageing 2018, 15, 8. [Google Scholar] [CrossRef] [Green Version]
  163. Calabrese, E. Hormesis: Path and Progression to Significance. Int. J. Mol. Sci. 2018, 19, 2871. [Google Scholar] [CrossRef] [Green Version]
  164. Zhang, Y.; Ahn, Y.H.; Benjamin, I.J.; Honda, T.; Hicks, R.J.; Calabrese, V.; Cole, P.A.; Dinkova-Kostova, A.T. HSF1-Dependent Upregulation of Hsp70 by Sulfhydryl-Reactive Inducers of the KEAP1/NRF2/ARE Pathway. Chem. Biol. 2011, 18, 1355–1361. [Google Scholar] [CrossRef] [Green Version]
  165. Zuehlke, A.D.; Wren, N.; Tenge VJohnson, J.L. Interaction of heat shock protein 90 and the co-chaperone Cpr6 with Ura2, a bifunctional enzyme required for pyrimidine biosynthesis. J. Biol. Chem. 2013, 288, 27406–27414. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Chemical structure of nitrogen bases.
Figure 1. Chemical structure of nitrogen bases.
Jox 12 00014 g001
Figure 2. Signaling pathways that control CAD activity. This diagram is based on references [20,21,22,23]. CAD initiates de novo pyrimidine synthesis. CAD is activated by the activation of growth factors binding growth factor receptors and triggering the MAP kinases pathway. This activation takes place at the beginning of the S phase in the cell cycle. After the S phase is over, CAD is deactivated by protein kinase A (PKA) phosphorylation. The lower panel shows that phosphorylation of the Thr456 residue activates the enzyme, while phosphorylation of Ser1406 acts in the opposite way. The diagram also shows the three enzymes that form CAD. In this regard, the activation and deactivation residues form part of the first enzyme that initiates synthesis, namely, CPS (carbamoyl phosphate synthetase). DHO: dihydroorotase; ATC: aspartate transcarbamoylase.
Figure 2. Signaling pathways that control CAD activity. This diagram is based on references [20,21,22,23]. CAD initiates de novo pyrimidine synthesis. CAD is activated by the activation of growth factors binding growth factor receptors and triggering the MAP kinases pathway. This activation takes place at the beginning of the S phase in the cell cycle. After the S phase is over, CAD is deactivated by protein kinase A (PKA) phosphorylation. The lower panel shows that phosphorylation of the Thr456 residue activates the enzyme, while phosphorylation of Ser1406 acts in the opposite way. The diagram also shows the three enzymes that form CAD. In this regard, the activation and deactivation residues form part of the first enzyme that initiates synthesis, namely, CPS (carbamoyl phosphate synthetase). DHO: dihydroorotase; ATC: aspartate transcarbamoylase.
Jox 12 00014 g002
Figure 3. All the steps of de novo pyrimidines synthesis (left panel). The participating enzymes are in red frames. The right panel shows the origin of the different parts that form the pyrimidine structure. Uracil is being used as an example.
Figure 3. All the steps of de novo pyrimidines synthesis (left panel). The participating enzymes are in red frames. The right panel shows the origin of the different parts that form the pyrimidine structure. Uracil is being used as an example.
Jox 12 00014 g003aJox 12 00014 g003b
Figure 4. Reaction catalyzed by carbamoylphosphate synthase II.
Figure 4. Reaction catalyzed by carbamoylphosphate synthase II.
Jox 12 00014 g004
Figure 5. Condensation between aspartic acid and carbamoylphosphate, generating carbamoyl aspartate through the enzymatic action of ATCase (aspartate transcarbamoylase). The structure of uracil is shown in the lower panel for a comparative view of how this structure is being built.
Figure 5. Condensation between aspartic acid and carbamoylphosphate, generating carbamoyl aspartate through the enzymatic action of ATCase (aspartate transcarbamoylase). The structure of uracil is shown in the lower panel for a comparative view of how this structure is being built.
Jox 12 00014 g005
Figure 6. Third step. Conversion of carbamoylaspartate into dihydroorotate through the enzymatic action of dihydroorotase.
Figure 6. Third step. Conversion of carbamoylaspartate into dihydroorotate through the enzymatic action of dihydroorotase.
Jox 12 00014 g006
Figure 7. Step 4. Chemical reaction catalyzed by dihydroorotate dehydrogenase. The pyrimidine ring is initially formed as orotate. Then, in the next step (step 5), it is attached to ribose phosphate (which is generated in the pentose phosphate pathway) and finally converted to the pyrimidine nucleotides that will be used for DNA and RNA synthesis.
Figure 7. Step 4. Chemical reaction catalyzed by dihydroorotate dehydrogenase. The pyrimidine ring is initially formed as orotate. Then, in the next step (step 5), it is attached to ribose phosphate (which is generated in the pentose phosphate pathway) and finally converted to the pyrimidine nucleotides that will be used for DNA and RNA synthesis.
Jox 12 00014 g007
Figure 8. The origin of the sugar molecule of ribonucleotides.
Figure 8. The origin of the sugar molecule of ribonucleotides.
Jox 12 00014 g008
Figure 9. Binding of orotic acid with PPRP generating OMP through the enzymatic activity of OPRT.
Figure 9. Binding of orotic acid with PPRP generating OMP through the enzymatic activity of OPRT.
Jox 12 00014 g009
Figure 10. Decarboxylation of orotidylate to form UMP (uridine monophosphate, step 6).
Figure 10. Decarboxylation of orotidylate to form UMP (uridine monophosphate, step 6).
Jox 12 00014 g010
Figure 11. Phosphorylation of UMP generating UDP and phosphorylation of UDP generating UTP.
Figure 11. Phosphorylation of UMP generating UDP and phosphorylation of UDP generating UTP.
Jox 12 00014 g011
Figure 12. CTP synthesis.
Figure 12. CTP synthesis.
Jox 12 00014 g012
Figure 13. Step 9: The sugar, ribose, is reduced to deoxyribose, forming dUDP.
Figure 13. Step 9: The sugar, ribose, is reduced to deoxyribose, forming dUDP.
Jox 12 00014 g013
Figure 14. Dephosphorylation of dUTP yielding dUMP.
Figure 14. Dephosphorylation of dUTP yielding dUMP.
Jox 12 00014 g014
Figure 15. Thymidilate synthase as the enzyme and methylentetrahydrofolate as cofactor add a methyl group on the nitrogen base. This converts dUMP to dTMP. Two further phosphorylations generate dTTP.
Figure 15. Thymidilate synthase as the enzyme and methylentetrahydrofolate as cofactor add a methyl group on the nitrogen base. This converts dUMP to dTMP. Two further phosphorylations generate dTTP.
Jox 12 00014 g015
Table 1. The key enzymes that participate in the de novo biosynthesis of pyrimidine.
Table 1. The key enzymes that participate in the de novo biosynthesis of pyrimidine.
EnzymeOptimal pHPossible Inhibitor
Phosphate synthetase II7.4Acivicin [33]
Aspartate transcarbamoylase (ATCase)pH in bacteria (8.5)N-(phosphonacetyl)-l-aspartate (PALA) [38]
Dihydroorotate dehydrogenase8Leflunomide [68,69,70]
Orotate phosphoribosyltransferaseThe forward reaction is 8, the reverse one is 6.5–7.5Include xanthosine, uridine 5′-phosphates, cytidine barbiturate, 5-flouro orotate [95,96]
Orotidine 5′-phosphate decarboxylaseapprox. 7.5Pyrazofurin and 6-aza uridine 5′-monophosphate [99,100]
Cytidine monophosphate kinase7.4Gemcitabine [65]
Nucleoside-diphosphate kinase8Theophylline [117]
Cytosine triphosphate synthase8Cyclopentenyl cytosine [121]
Ribonucleotide reductase enzyme7.5–8Cisplatin, chlorambucil, desferrioxamine, gemcitabine, and hydroxyurea [135,136,137,138,139,140,141]
Tymidylate synthase7.0 and 8.1Capecitabine and 5 fluorouracil [154,155,156,157]
Ribonucleotide reductase7.5 to 8 with a low iron levelGemcitabine and iron chelators [160]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Alqahtani, S.S.; Koltai, T.; Ibrahim, M.E.; Bashir, A.H.H.; Alhoufie, S.T.S.; Ahmed, S.B.M.; Molfetta, D.D.; Carvalho, T.M.A.; Cardone, R.A.; Reshkin, S.J.; et al. Role of pH in Regulating Cancer Pyrimidine Synthesis. J. Xenobiot. 2022, 12, 158-180. https://doi.org/10.3390/jox12030014

AMA Style

Alqahtani SS, Koltai T, Ibrahim ME, Bashir AHH, Alhoufie STS, Ahmed SBM, Molfetta DD, Carvalho TMA, Cardone RA, Reshkin SJ, et al. Role of pH in Regulating Cancer Pyrimidine Synthesis. Journal of Xenobiotics. 2022; 12(3):158-180. https://doi.org/10.3390/jox12030014

Chicago/Turabian Style

Alqahtani, Saad Saeed, Tomas Koltai, Muntaser E. Ibrahim, Adil H. H. Bashir, Sari T. S. Alhoufie, Samrein B. M. Ahmed, Daria Di Molfetta, Tiago M. A. Carvalho, Rosa Angela Cardone, Stephan Joel Reshkin, and et al. 2022. "Role of pH in Regulating Cancer Pyrimidine Synthesis" Journal of Xenobiotics 12, no. 3: 158-180. https://doi.org/10.3390/jox12030014

Article Metrics

Back to TopTop