Open access peer-reviewed chapter

Translational Research on Chagas Disease: Focusing on Drug Combination and Repositioning

Written By

Marcos André Vannier-Santos, Ana Márcia Suarez-Fontes, Juliana Almeida-Silva, Alessandra Lifsitch Viçosa, Sandra Aurora Chavez Perez, Alejandro Marcel Hasslocher-Moreno, Gabriel Parreiras Estolano da Silveira, Luciana Fernandes Portela and Roberto Magalhães Saraiva

Submitted: 14 February 2022 Reviewed: 03 March 2022 Published: 24 June 2022

DOI: 10.5772/intechopen.104231

Chapter metrics overview

143 Chapter Downloads

View Full Metrics

Abstract

Chagas disease, caused by the protozoan Trypanosoma cruzi, is a major neglected disease endemic to Latin America, associated to significant morbimortality comprising a remarkable socioeconomic problem mainly for low-income tropical populations. The present chapter focuses translational research on Chagas disease, approaching drug combinations and repositioning, particularly exploiting the parasite oxidative stress by prospecting prooxidant compounds combined with antagonists of antioxidant systems, for developing low-cost and safe therapies for this infection. The pertinent literature on protozoal parasitic diseases is reviewed as well as on repurposing disulfiram aiming the combination with the Chagas disease drug of choice benznidazole. Both disulfiram and its first derivative sodium diethyldithiocarbamate (DETC) are able not only to inhibit p-glycoprotein, possibly reverting resistance phenotypes, but also to reduce toxicity of numerous other drugs, heavy metals, etc. Therefore, this innovation, presently in clinical research, may furnish a novel therapeutic for T. cruzi infections overcoming the adverse effects and refractory cases that impair the effectiveness of Chagas disease treatment.

Keywords

  • drug combination
  • drug repositioning
  • translational medicine
  • Chagas disease
  • oxidative stress
  • Trypanosoma cruzi

1. Introduction

Chagas disease (CD), the parasitic infection caused by the kinetoplastid protozoan Trypanosoma cruzi, is also known as American trypanosomiasis, for the huge endemic areas in South and Central Americas [1], but autochthonous human [2, 3, 4] and domestic/wild animal [5, 6, 7, 8] cases were reported in the United States, and due to migration, it is already considered a public health problem on a global scale reaching different continents [9, 10, 11]. It is noteworthy that climate changes may promote the northward insect vector propagation [12], possibly generating new foci or endemic areas, and suitable climatic conditions may be available in African and Asian nations [13]. Besides the vector bloodmeal, congenital, blood transfusion and organ transplantation [14], CD may be transmitted orally via food and beverages contaminated by triatomine feces such as sugarcane and açai juices [15, 16] and even water, stored in/near domiciles in arid regions [15, 16], as the parasite is able to survive in such media [17].

It is alarming that 6–7 million people are estimated to have CD worldwide, with circa 173,000 new cases/year and over 75 million people are at risk. CD is the parasitic disease of highest mortality in Latin America as 9490 deaths were reported in 2019. Furthermore, the real prevalence is largely unknown as most chronic patients are asymptomatic and even symptomatic patients have poor access to health public system. CD is endemic in 21 countries in Central and Latin America where about 5.7 million people have CD and 25% of the population is at risk [18]. In 2020, it was estimated that there were 3.2 million infected people, which can reach 1.5% of the general population. In addition, about 70 million are at risk of infection [19]. The prevalence of CD is presumably vastly underestimated. In January 2020, a study carried out by the ArtScience Initiative for Health Promotion, carried out by Oswaldo Cruz Foundation (Fiocruz) and collaborating organizations, showed a CD seropositivity of 20% in a tested population of an endemic area [20]. It must be mentioned this study was not designed to access CD prevalence and was biased by the population intention to get diagnosis procedures.

CD represents economic losses in excess of $1.2 billion/year to endemic countries in South America, in addition to more than $7 billion a year at global levels [21], including treatment and loss of productivity. Since no proven effective and approved vaccines are available for this disease, chemotherapy represents the only therapeutic intervention, as well as an important way to control them.

CD etiological treatment is directed according to the phase and clinical presentation of the disease, which is mandatory in the acute phase, congenital cases, or reactivation due to immunosuppression. In the chronic phase, the trypanocidal treatment is indicated in children and adolescents, recent infection, and women of childbearing age [22].

Advertisement

2. Therapeutics

Although CD was discovered and is studied for over a century [14], the etiologic treatment is still based on solely two drugs (Figure 1): the nitrofuran derivative nifurtimox (NFX; Lampit®, Bayer; 5-nitrofuran(3-methyl-4-(5′-nitrofurfurylideneamine)tetrahydro-4H-1,4-tiazine-1,1-dioxide), and the 2-nitromidazole benznidazole (BZ; LAFEPE; N-benzyl-2-nitroimidazole-acetamide) [23]. Both NFX and BZ were shown to produce remarkable ultrastructural alterations in mammal cells and tissues [24, 25], which were apparently more pronounced in NFX-treated animals [26]. Therefore, experimental chemotherapy studies approaching parasites as T. cruzi should preferentially include ultrastructural analysis, in order to offer a subcellular compartmentation understanding to aid the antiparasitic agent mechanism(s) of action elucidation [27, 28] and ultimately leading to the understanding of cell death pathways involved [29].

Figure 1.

Molecular structures of the nitroheterocyclic drugs employed in the treatment of Chagas disease: the 2-nitroimidazole benznidazole (A) and the 5-nitrofuran nifurtimox (B).

The CD therapeutics remain unsatisfactory, as they are associated with adverse effects [30, 31, 32], affecting 84.8 and 95.2% of patients treated with BZ and NFX, respectively [33], which may be severe, leading to the irreversible suspension of therapy in CD, in ≈20% [34, 35], ≈30% [36, 37], 41.5% [38], and up to 50% of the cases [39, 40]. Treatment suspension using NFX was reported in 43.8% of patients [33]. In an early study based on small samples, NFX was reported to be associated to definitive treatment interruption in 75% of patients [38]. Nevertheless, treatment intolerance was reported at similar levels with the use of the two drugs, approached by the same team [34, 35], but adverse effects, including neuropsychiatric events, may be more frequently associated to NFX [33]. In addition, it was reported that among patients who had discontinued BZ treatment and were treated with NFX, 12.3% also developed adverse effects that required definitive discontinuation of therapy [39]. Nevertheless, NFX was reported to be safe as a second-line therapy in patients who discontinued BZ [41].

Most CD patients are not treated because of the insufficient diagnosis and low cure rates observed in chronically infected patients [42], although treatment may diminish the disease progression and cardiovascular events [43, 44]. In addition, the CD treatment accomplishes only a parasitological cure, and a clinical cure is hardly proved [43, 45]. Whereas the bona fide sterile cure or complete clearance of the infection is considered a “prerequisite” for new anti-T. cruzi drug candidates [46], it is usually not achieved in murine model [47, 48] or human infection as immunosuppression often leads to infection reactivation [49]. In this regard, T. cruzi amastigotes may persist in a dormant or quiescent form, which may protect the parasites from antiparasitic agents [50, 51].

As the dormancy state of T. cruzi amastigotes is associated to drug resistance [50, 51], it is desirable to develop drugs able to affect dormant parasites. The mechanisms that allow the establishment of persistence include the capacity to suppress the oxidative burst produced by phagocytes largely depending on iron-containing superoxide dismutases (FeSOD) and trypanothione-acting enzymes [52]. Thus the use of disulfiram (DSF) is of potential relevance since it can diminish glutathione levels [53, 54], and the DETC first derivative of DSF is an SOD inhibitor [55, 56]. Furthermore, DSF could target T. cruzi dormancy. Although the signal transduction pathways involved in this process were not completely elucidated, it is interesting that DSF is able to reverse HIV latency affecting PKC (protein kinase C), AKT (protein kinase B), PI3K (phosphoinositide 3-kinases), NF-kB (nuclear factor kappa-light-chain-enhancer of activated B cells) [57, 58], which also affect T. cruzi infection [59, 60] that leads to the activation of PI3K [61], whereas DSF promotes PI3K inhibition [62].

An important study [63] approached the persistent parasite elimination, but the use of higher BZ doses might pose higher risks for patients. In this regard, the polyamine and thiol synthesis Leishmania are associated to macrophage M2 phenotype, leading to parasite persistence [64].

2.1 Drug resistance

Besides considerable severe adverse effects, one of the greatest problems of CD therapeutics is the selection of resistant parasites, impairing its effectivity, therefore causing refractory cases. BZ and NFX resistance is readily developed in vitro and in vivo [47, 65], in the former case, via different mechanisms that can act in concert [66].

Despite significant time and resources investments by innumerous research institutions over the world, only a few therapeutic candidates advanced the pipeline to treat neglected diseases such as CD [67]. It is alarming that it usually takes over 10 years to develop new drugs, whereas resistant parasites are rapidly selected. Also, there are naturally resistant T. cruzi strains [68, 69, 70] that express a novel ABCG-like transporter [71]. Besides extrusion pumps, T. cruzi resistance may involve SOD and trypanothione (vide infra). Therefore, there is pressing demand for the development of novel effective therapies for CD.

Advertisement

3. Oxidative stress in Chagas disease

Oxidative stress is a central phenomenon involved in aging, cancer, transmissible or infectious diseases, including COVID-19 [72], nontransmissible chronic conditions, such as metabolic diseases, autoimmune and degenerative disorders, inflammation, metal poisoning, etc. [73, 74, 75], produced by the imbalance on the production/uptake of oxidant/antioxidant species [76].

A plethora of antioxidant defenses evolved in order to balance the redox homeostasis [76, 77]. Oxidant species such as superoxide (O2•−) and hydrogen peroxide (H2O2) are detoxified by SOD and catalase, respectively. Most cells rely also on the peptide glutathione (GSH), able to chelate reactive oxidant species (ROS) via cysteine sulfhydryl (SH) group and function as substrate for enzymes including GSH reductase and GSH peroxidase [78].

Although most of these processes are evolutionary conserved, some of the antioxidant defenses pathways differ between mammals and pathogens, therefore comprise potential chemotherapy targets. Contrary to mammals, GSH in trypanosomatid parasites mostly takes part in the adduct with the polyamine spermidine, forming N1,N8-bis(glutathionyl)spermidine (trypanothione, TSH), and therefore its expression depends on the GSH, TSH [79], and polyamine [80] metabolism pathways.

Metabolomics and gene expression studies [81] reveal the participation of both GSH and the spermidine synthesis pathway, indicating the participation of trypanothione, in the regulation of redox metabolism in trypanosomatids. GSH is very relevant not only in oxi-reductive homeostasis, as this molecule is also related to detoxification and resistance to different drugs/xenobiotics in tumor cells [82, 83] binding to drugs that are extruded via multidrug resistance transporters [84]. TSH binding to NFX and BZ is involved in the detoxication of these trypanocides [85, 86]. Therefore, glutathione/trypanothione can promote the action/reverse resistance to different drugs. T. cruzi parasites overexpressing trypanothione synthetase tolerated higher doses of BZ and NFX [87]. Conversely, the GSH biosynthesis inhibition using buthionine sulfoximine increases the efficacy of NFX and BZ upon T. cruzi in vitro [88] and NFX in vivo [89] as well as of stibogluconate on Leishmania (L.) donovani [90].

Interestingly, polyamine play pivotal roles in parasite cells [91, 92], including T. cruzi antioxidant defense [93], and its synthesis and transport pathways provide valuable chemotherapy targets [94, 95], including repositioned drugs [96].

Parasitic diseases such as CD are correlated to oxidative stress [97, 98], associated to triggered chronic inflammatory reactions [99, 100]. Endogenous oxidative stress may be produced by cell organelles, mainly mitochondria [101, 102]. The CD myocarditis is characterized by intense oxidative stress due both to inflammatory response associated to neutrophils and macrophages NADPH oxidase (Nox) activity and the macrophage superoxide produced by Nox2 is required for parasite control in early infection [103]. The mitochondrial ROS produced by cardiomyocytes plays a relevant role in intracellular oxidative stress and inflammation, causing myocardium tissue damage [104, 105, 106]. These events are not independent since mitochondrial ROS may trigger proinflammatory cytokines via NFkB and PARP/PAR pathways [107], and the mitochondrial MnSOD activity may revert much of the inflammatory foci and necrosis [105], and ineffective antioxidant defense is associated to oxidative stress [108]. Exosome or extracellular vesicles liberation may also contribute to inflammation and oxidative stress [107, 109]. The oxidative stress is also involved in neurodegeneration in both cardiac and gastrointestinal tissues [110]. The chronic oxidative stress in the nervous tissue is associated to cognitive deficit, which can be reversed by BZ treatment [111].

Thus, the use of adjuvant antioxidant agents may ameliorate the cardiac pathogenesis [107, 112, 113]. Interestingly, vitamin C, widely considered antioxidant, can at high concentrations also function as a prooxidant, undergoing pH-dependent autoxidation, leading to H2O2 formation [114, 115]. In CD models, ascorbic acid can also reduce parasitemia, promote BZ action, and enhance animal survival in murine infection [116, 117].

ROS production comprises a well-known microbicidal immune effector mechanism [118]; therefore parasite borne antioxidant systems are not only virulence factors [119]. Besides the parasiticidal activity, ROS may function as signaling molecules promoting parasite proliferation. As in the Paracelsus adage, “The dose makes the poison” (Latin: sola dosis facit venenum), ROS in mammalian cells may trigger different responses depending on concentration. Low ROS levels may have signal transduction roles, inducing responses such as activation, proliferation, and differentiation, whereas at higher levels such molecules are generally cytotoxic, leading to cell death [120]. Similarly, in T. cruzi, low ROS levels may signal for parasite invasion of host macrophages [121] and proliferation mainly in the acute phase [122], but high ROS levels culminate in programmed cell death, which may be inhibited by enhanced SOD expression [87]. Interestingly T. cruzi amastigotes undergo stress-induced proliferation [123].

Advertisement

4. Oxidative stress as a source of chemotherapy targets

Numerous therapeutic strategies exploit redox systems [124], including protozoal diseases [125], such as CD [126]. Therefore, antioxidant systems including SOD, trypanothione, and enzymes action on this glutathione-spermidine adduct (N1,N8-bis(glutathionyl)spermidine), such as trypanothione reductase, can comprise important chemotherapy targets [127]. Natural products such as the naphthoquinones

-/β-lapachones [128, 129, 130] and their derivatives [131, 132] have microbicidal activity against T. cruzi, among other pathogens [132]. Interestingly, β-lapachone derivatives were shown to cause mitochondrial dysfunction [131], damage [133], and autophagy, including mitophagy as well as apoptosis and necrosis [134]. In this regard, mitochondria comprise important therapeutical targets for cancer [135], aging [136], cardiovascular diseases [137], and degenerative diseases such as rheumatoid arthritis [138], Alzheimer’s disease [139], Parkinson’s disease [140], etc. Mitochondria are also promising target for antiparasitic [141, 142] and particularly antiprotozoal [143, 144, 145] therapeutic agents, specifically approached in trypanosomatids [146, 147, 148].

Up to 2% of the O2 reaching the mitochondrial matrix is converted to O2•− (superoxide anions) forming H2O2 via SOD [149]. Like mammalian cells, T. cruzi mitochondria are a source of ROS [150] producing superoxide. Therefore, the Mn-SOD is important for controlling oxidative stress in this redox organelle. Contrary to mammals, the trypanosomatid mitochondria present FeSOD [151] that can protect from O2•− produced by macrophages [152].

Because of the prooxidant effects of antiparasitic drugs [126, 153, 154, 155], ROS detoxifying systems may comprise valuable scape mechanisms from pharmaceutical intervention [156] and programmed cell death triggered by mitochondrial O2•− [157].

The prooxidant capacity of both NFX and BZ, particularly in the former, is due to redox cycling with the production of O2•− [126, 158, 159, 160]. Superoxide may be not produced by BZ in the parasite, but in the host cell [161]. Therefore, FeSOD is linked to BZ resistance in T. cruzi [66, 162, 163]. Proteome of BZ-resistant Trypanosoma cruzi revealed enhanced FeSOD activity [164]. BZ resistance was associated to decreased cytosolic SOD but enhanced mitochondrial MnSOD and tryparedoxin-1 [165]. The deletion of the sodb1 gene enhances Trypanosoma brucei susceptibility to BZ and NTX [166]. FeSOD is also implicated in drug resistance in L. (Viannia) braziliensis and L. (Leishmania) infantum [167, 168] Entamoeba histolytica [169]. Tryparedoxin peroxidase is also associated to antimony resistance in L. (V.) braziliensis [170]. In addition, SOD inhibition was reported to decrease parasitemia in T. cruzi murine infection [171].

Sirtuins are a highly conserved family of enzymes that deacetylate lysine residues on histone and non-histone proteins, using NAD+ as a cosubstrate, regulating cellular antioxidant/Redox mechanisms [172, 173]. It is noteworthy that SIRT3, 4, and 5 are found in the mitochondrial matrix [174]. As cardiomyocyte mitochondrial dysfunction plays a central role in chagasic myocarditis (vide supra), the activation of sirtuins such as SIRT1 by agonists including resveratrol may enhance antioxidant defenses [175], and SIRT3 activates MnSOD, scavenging ROS [176]. Nevertheless, the sirtuin TcSir2rp3 was shown to increase T. cruzi resistance to BZ and NTX for overexpressing TcFeSOD-A activities [177].

Selenium and selenium-containing compounds show beneficial effects both in murine [178, 179, 180] and human T. cruzi infection [181, 182], therefore comprise promising coadjuvant therapies for CD [183, 184, 185], although selenium was previously reported to increase tissue parasitism [186].

This activity maybe largely dependent on redox regulation as this inflammatory infection is associated with intense oxidative stress, and selenium may be antioxidant [187] and anti-inflammatory [188], as well as catalyze hydrogen peroxide (H2O2) reduction [189], therefore possibly diminishing the oxidative stress in infected cardiomyocytes, by impairing the Fenton reaction in the presence of iron.

Advertisement

5. Repositioning and combining drugs

The combination of different drugs may pose the advantage of supra-additive effects, which may be synergistic, in parasite models such as T. cruzi [190], Plasmodium falciparum, Trypanosoma rhodesiense [191]. The identification of synergistic combinations is relevant since they tend to present higher selective indices [192193], consequently, avoiding side effects and potentially permitting development of antiparasitic agents used at lower concentrations.

The identification of drug combinations with multiple targets can lead to the use of novel multitarget mechanisms able to cope with the challenge of multigenic diseases [194] and/or chronic infections with complex pathophysiology. It is noteworthy that the pharmaceutical properties of the combination may be absent in the components alone [195], generating the innovative concept or science field termed polypharmacology with numerous applications on drug repurposing [196] and CD [197]. As the philosopher Aristotle (384–322 B.C.) stated: “The whole is greater than the sum of its parts.”1

Furthermore, drug combinations are largely employed for preventing drug resistance [198, 199, 200, 201, 202, 203, 204]. However, this strategy is not constantly successful as the reports of resistance to the sulfadoxine-pyrimethamine combination began in the same year this antimalarial regimen entered the clinic [205]. Similarly, the discovery of artemisinin (ART) costed Youyou Tu over 30 years of hard work [206] and was worthy a Nobel Prize, but P. falciparum resistance to the drug was detected after about 10 years of use [207]. The antimalarial combination therapies based on the use of ART were considered key to the elimination of malaria [208], but in the very same year [209, 210] and even earlier [211], the arteminisin derivatives combination therapy failures were reported. In the case of CD, the problem may be even more upsetting as natural resistance isolates are arising, particularly in the Amazon region (vide supra). Thus, effective strategies to prevent different mechanisms of drug resistance to arise are immediately needed.

Approaching repositioned drugs with available pharmacokinetic and toxicological properties can shorten the long and expensive path between in vitro trials and new drugs. While the period between drug discovery and approval can be 12–16 years at a cost of US$1–2 billion, repositioned drugs can enter the clinic in ½ the time, at circa 1/3 the cost [212], with much higher success rates [213].

Drug repositioning maybe a promising approach in CD [214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227]. Similarly, drug combinations may be instrumental in CD [197, 228, 229, 230, 231, 232, 233], and both strategies may be employed and associated [214, 234, 235, 236]. Furthermore, drug combinations can increase success of drug repositioning [237]. In addition, it was accurately hypothesized that the combined use of repurposed drugs with BZ could be more efficacious than BZ alone [238].

5.1 Repositioning disulfiram

Disulfiram (DS, 1,1′-disulfanediylbis(N,N-diethylmethanethioamide) also termed tetraethylthiuram disulfide; CAS no. 97-77-8; Molecular Formula: C10H20N2S4), a repositioned drug used in alcoholism and marketed as Antabuse® (Figure 2), was approved for medical use over 70 years ago and is widely used since then [239, 240].

Figure 2.

Molecular structures of disulfiram (A) and sodium diethyldithiocarbamate (B).

At the very beginning, the discovery of thiocarbamates and its derivatives was serendipitous and showed clear signs of versatile perspectives that unequivocally culminated in the present promising repurposing strategies for both pharmaceutical and industrial applications [241, 242].

In the 1930s and 1940s, dithiocarbamates such as dimethyldithiocarbamates and diethyldithiocarbamates were used as pesticides against fungal pathogens on different crops [243], besides biocides in household products [244].

The industry plant physician E. E. Williams in 1937 observed that workers using tetramethylthiuram monosulfide and disulfide to facilitate the rubber vulcanization became alcohol-intolerant and quit consuming alcoholic beverages. The DSF-induced alcohol aversion was described in 1948 [245]. At that time, DSF was approached as a vermicide and employed as an ointment to treat scabies.

Afterward, besides alcoholism, DSF started to be studied for heavy metal poisoning, cancer [246, 247, 248, 249], HIV [243, 250], as well as cocaine dependence, pathological gambling, and other psychiatric disorders [239] and other form of addiction, for example, the d-methamphetamine abuse [251]. Further tests are being performed focusing applications such as Alzheimer’s disease [252], Lyme disease and babesiosis [253], tuberculosis [254], non-tuberculous mycobacteria infections [255], giardiasis [256], amoebiasis [257], obesity [258] and to revert drug resistance in different types of cancer [259, 260, 261], tuberculosis [262] bacterial infections [263], mycosis [264], giardiasis [265], etc. The repositioning of low-cost drugs such as DS is considered a “salvation” for global healthcare system [266].

Sodium diethylcarbamodithioate (Figure 2) (DETC also known as sodium (diethylcarbamothioyl)sulfanide; CAS no. 148-18-5; Molecular Formula: C5H11NS2.Na) is the first derivative of DSF, involved in many of the biological activities of the latter.

Seemingly DETC is less toxic than aspirin [243], widely used, and well tolerated in humans [267] for decades being used up to 800 mg/twice/week, with no adverse effects [268]. DETC also known as Imuthiol or Dithiocarb was used as immunomodulator with good results on AIDS patients [269, 270] and was clinically employed in chronic bronchitis, rheumatoid arthritis, tuberculosis, and chronic infection [271].

In a seminal report on its antiparasitic activity, DETC was demonstrated to be leishmanicidal [272]. Afterward, novel delivery systems were developed to optimize the leishmanicidal activity of DETC [273, 274, 275]. In this regard, novel drug delivery systems are also developed for DSF [276]. The data obtained on Leishmania amazonensis motivated us to move to CD, employing the repositioned drug DSF combined to the drug of first choice BZ. Tests on NFX are in progress.

It is worth remembering that CD pathophysiology is associated with oxidative stress (vide supra), and both DSF [277] and DETC [278] can act as antioxidants. In addition, modulation of oxidative stress comprises a valuable tool in heart disease therapeutics [279]. In addition, DSF has antimutagenic properties [280].

5.2 Disulfiram combined to benznidazole in Chagas disease

Both DSF and DETC have antiparasitic activity on T. cruzi [281, 282], but the effectivity was not pronounced.

In our study, the DSF-BZ combination is promising since the antagonism of SOD activity can enhance oxidative stress in cancer cells [249] and T. cruzi [283]. In this regard, the antitumor activity of NTX is enhanced by SOD1 inhibition mediated by tetrathiomolybdate [284]. Both in vitro and in vivo experimental data confirmed the present assumption [Almeida-Silva et al., in press]. The SOD inhibition as well as TSH reaction by DSF/DETC can promote the intracellular accumulation of ROS leading to parasite death (Figure 3).

Figure 3.

Putative mechanisms of action of disulfiram (DSF) or diethyldithiocarbamate (DETC) in combination with trypanocides in T. cruzi infection. Benznidazole (BZ) and nifurtimox are toxic and produce adverse reactions (1), which are ameliorated via detoxification mediated by DSF or DETC (2). The anti-T. cruzi agents trigger the formation of reactive oxygen species (ROS, 3) via nitroanion radicals (RNO2•−) that give rise to superoxide (O2•−), that is detoxified by superoxide dismutase (SOD, 5), generating hydrogen peroxide (H2O2), which in the presence of iron can produce hydroxyl radicals (OH) and hydroxide anions (OH) via Fenton reaction. DETC inhibits SOD (4). ROS may be detoxified by reaction with sulfhydryl or thiol groups of trypanothione (N1,N8-bis(glutathionyl)spermidine, 6), and this adduct can be removed by reaction with thiols of DSF/DETC (7). The BZ molecules in the parasite cytoplasm are extruded from the cell via p-glycoproteins or MDR transporters (8), which are inhibited by DETC (9), presumably reversing resistance phenotypes.

CD etiological therapy is often associated to severe adverse effects caused by the highly toxic drugs (vide supra). In this sense, the present innovation involves the advantage of employing DSF/DETC with cytoprotective properties [243] in different cell types.

DSF/DETC have neuroprotective [285], hepatoprotective [277], and nephroprotective [286] and even radioprotective [287, 288] activity. These protective effects may be beneficial in the treatment of parasitic diseases, because in the treatment of experimental infection by T. rhodesiense, DSF has marked protective activity (disulfiram rescue) against the toxic effects of diaminodichloroplatin and preventing the death of the treated organism [289].

Thus, the development of low-toxicity therapies may be expected, as DSF may have a protective action against the toxic effects of drugs such as cyclophosphamide [290], ifosfamide [291], N-nitrosodimethylamine [292], isoniazid [293] and the toxicity of α-naphthylisothiocyanate [294], acetaminophen [295], pyrrolizidines [296], the lethal effects of hypoxia [297], ischemia [298], as well as lead [299], cadmium [300], mercury, and other heavy metals [301]. Thus, DSF combinations can enable the development of safe medicines. Regarding CD, the cardioprotective and antioxidant activities of DSF/DETC as well as atrial neuroprotection [302] are particularly desirable [303, 304, 305, 306]. In addition, DSF is effective as prophylactics in experimental colitis [307].

As drug resistance limits the successful CD therapy, the T. cruzi PgP expression has a pivotal role [308]. Therefore, it is relevant in the present approach that DSF/DETC inhibit PgP [261, 309, 310], causing the BZ accumulation within the parasite cytoplasm, enhancing trypanocidal activity, potentially reversing resistance phenotypes, such as MDR+ (Figure 3). Interestingly, the ABCC proteins from T. cruzi are involved in thiol transport [311]. In view of the glutathione-drug adduct transport by ABC transporters (vide supra), it is interesting that DSF reduces GSH levels [54] at least in part through the formation of complexes with its different derivatives [312].

DSF [313] affects the redox balance of the cell, to GSH oxidation [314], reducing GSH levels [54] at least in part through the formation of complexes with its different derivatives [312, 315]. DETC can also reduce the GSH/non-protein thiol levels, also leading to the reduction of glutathione peroxidase activities [53, 316].

The combinations tested here may also contribute to resistance reversal, also through DETC-mediated inhibition of Fe-dependent SOD, which is linked to resistance to BZ in T. cruzi [66, 162, 163].

Furthermore, DSF can be used against cancer cells targeting the ubiquitin-proteasome system [317], and the ubiquitin-proteasome pathway is a therapeutic target in T. cruzi [318].

In this way, the strategy based of combinations of the repositioned drugs proposed here can achieve effectiveness, with selectivity and, therefore, safety in the CD treatment and sheds new light on perspectives for new therapeutic strategies.

Advertisement

6. The clinical stage

Translational research in biomedical sciences translates basic research and experimental discoveries into health taking the route from benchtop to bedside. This important field has gained substantial attention and investments in the last two decades [319].

In order to reach a proof of concept on the effectivity of the DSF-BZ combination in human infection, a partnership was established gathering different units of Fiocruz. The present study comprises a translational approach that began with experiments in vitro, on the bench and now reaches the clinical stage at the Evandro Chagas National Institute of Infectious Diseases-Fiocruz, coordinated by the team of the Clinical Research Laboratory of Chagas Disease, with assistance of the Clinical Research platform. Therefore, the phase I/II clinical trial was elaborated (Figure 4) and published recently [320].

Figure 4.

Design of the clinical trial for testing the BZ-DSF combination. Reproduced from Ref. [320] (with permission).

Advertisement

7. Conclusions and future perspectives

The use of DSF/DETC combined to BZ in CD treatment comprises a potential innovative therapeutical tool, possibly overcoming adverse reactions and refractory cases. Since these repositioned drugs exert cytoprotective effects, reducing the adverse reactions of many drugs, safe combinations can be potentially identified, leading to the development of well-tolerated medication. Therefore, therapy interruption can be precluded, consequently increasing patient adherence. In addition, as DSF/DETC can inhibit p-glycoprotein activity as well as reduce GSH levels, two molecules involved in drug extrusion from MDR+ parasites, it is reasonable to suppose the combination could eventually revert/downmodulate natural/acquired resistance phenotypes. Thus, treatment may be effective even in refractory cases. We are now approaching the clinical response of chronic phase CD patients. A possible proof of concept may lead to the development of a safe and effective medication, with profound implications in treatment prognosis, presumably improving the quality of life of the patients.

Advertisement

Acknowledgments

This research was sponsored by grants from the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq grant no. 443886/2018-0 to RMS and CNPq grant no. 314717/2020 to MAVS), Fundação Carlos Chagas Filho de Amparo à Pesquisa do Rio de Janeiro (FAPERJ grant no. 211.167/2019 to RMS; FAPERJ grant no. 260475/2021 and 259286/2021 to MAVS and FAPERJ grant no. 204.388/2021 to AMSF) and Fundação Oswaldo Cruz (Fiocruz grant no. 6221125199 to MAVS).

References

  1. 1. Schmunis GA, Yadon ZE. Chagas disease: A Latin American health problem becoming a world health problem. Acta Tropica. 2010;115(1-2):14-21
  2. 2. Montgomery SP, Parise ME, Dotson EM, Bialek SR. What do we know about Chagas disease in the United States? The American Journal of Tropical Medicine and Hygiene. 2016;95(6):1225-1227
  3. 3. Harris N, Woc-Colburn L, Gunter SM, Gorchakov R, Murray KO, Rossmann S, et al. Autochthonous Chagas disease in the Southern United States: A case report of suspected residential and military exposures. Zoonoses and Public Health. 2017;64(6):491-493
  4. 4. Lynn MK, Bossak BH, Sandifer PA, Watson A, Nolan MS. Contemporary autochthonous human Chagas disease in the USA. Acta Tropica. 2020;205:105361
  5. 5. Rowland ME, Maloney J, Cohen S, Yabsley MJ, Huang J, Kranz M, et al. Factors associated with Trypanosoma cruzi exposure among domestic Canines in Tennessee. The Journal of Parasitology. 2010;96(3):547-551
  6. 6. Thompson JM, Habrun CA, Scully CM, Sasaki E, Bauer RW, Jania R, et al. Locally transmitted Trypanosoma cruzi in a Domestic Llama (Lama glama) in a rural area of Greater New Orleans, Louisiana, USA. Vector Borne and Zoonotic Diseases. 2021;21(10):762-768
  7. 7. Busselman RE, Hamer SA. Chagas Disease Ecology in the United States: Recent Advances in Understanding Trypanosoma cruzi Transmission Among Triatomines, Wildlife, and Domestic Animals and a Quantitative Synthesis of Vector-Host Interactions. Annual Review of Animal Biosciences. 2022;10:325-348
  8. 8. Busselman RE, Meyers AC, Zecca IB, Auckland LD, Castro AH, Dowd RE, et al. High incidence of Trypanosoma cruzi infections in dogs directly detected through longitudinal tracking at 10 multi-dog kennels, Texas, USA. PLoS Neglected Tropical Diseases. 2021;15(11):e0009935
  9. 9. Coura JR, Viñas PA. Chagas disease: A new worldwide challenge. Nature. 2010;465(S7301):S6-S7
  10. 10. Franco-Paredes C, Bottazzi ME, Hotez PJ. The unfinished public health agenda of Chagas disease in the era of globalization. PLoS Neglected Tropical Diseases. 2009;3(7):e470
  11. 11. Parker ER, Sethi A. Chagas disease: Coming to a place near you. Dermatologic Clinics. 2011;29(1):53-62
  12. 12. Garza M, Feria Arroyo TP, Casillas EA, Sanchez-Cordero V, Rivaldi C-L, Sarkar S. Projected future distributions of vectors of Trypanosoma cruzi in North America under climate change scenarios. PLoS Neglected Tropical Diseases. 2014;8(5):e2818
  13. 13. Eberhard FE, Cunze S, Kochmann J, Klimpel S. Modelling the climatic suitability of Chagas disease vectors on a global scale. eLife. 2020;6(9):e52072
  14. 14. Telleria J, Tibayrenc M. American Trypanosomiasis: Chagas Disease One Hundred Years of Research. 1st ed. Amsterdam: Elsevier; 2010. 844 p
  15. 15. Dias JP, Bastos C, Araújo E, Mascarenhas AV, Martins Netto E, Grassi F, et al. Acute Chagas disease outbreak associated with oral transmission. Revista da Sociedade Brasileira de Medicina Tropical. 2008;41(3):296-300
  16. 16. de Noya B, González O, Robertson LJ. Trypanosoma cruzi as a Foodborne Pathogen. Cham: Springer International Publishing; 2015
  17. 17. Suárez DC, Rey ÁP, Orduz ML, Prada RL, Tarazona Z. Survival of Trypanosoma cruzi in experimentally contaminated drinks. Biomédica. 2016;32(1):134-148
  18. 18. WHO (World Health Organization). Chagas Disease (American trypanosomiasis). 2018
  19. 19. Carlos Pinto Dias J, Novaes Ramos A, Dias Gontijo E, Luquetti A, Aparecida Shikanai-Yasuda M, Rodrigues Coura J, et al. II Consenso Brasileiro em Doença de Chagas, 2015. Epidemiologia e Serviços de Saúde. 2016;25(21):1-10
  20. 20. Araujo-Jorge TC, Ferreira RR, Rocha RCM, Vieira TM, Costa ND, Santos LL, et al. “Chagas Express XXI”: A new ArtScience social technology for health and science education—A case study in Brazilian endemic areas of Chagas disease with an active search of chronic cases. PLoS Neglected Tropical Diseases. 2021;15(7):e0009534
  21. 21. Lee BY, Bacon KM, Bottazzi ME, Hotez PJ. Global economic burden of Chagas disease: A computational simulation model. The Lancet Infectious Diseases. 2013;13(4):342-348
  22. 22. PAHO (Pan American Health Organization). Chagas disease. 2019
  23. 23. Urbina JA. Specific chemotherapy of Chagas disease: Relevance, current limitations and new approaches. Acta Tropica. 2010;115(1-2):55-68
  24. 24. Bartel LC, Montalto de Mecca M, Fanelli SL, Rodriguez de Castro C, Diaz EG, Castro JA. Early nifurtimox-induced biochemical and ultrastructural alterations in rat heart. Human & Experimental Toxicology. 2007;26(10):781-788
  25. 25. de Mecca MM, Fanelli SL, Bartel LC, de Castro CR, Díaz EG, Castro JA. Nifurtimox nitroreductase activity in different cellular fractions from male rat pancreas. Biochemical and ultrastructural alterations. Life Sciences. 2007;81(2):144-152
  26. 26. Mecca MM de, Bartel LC, Castro CR de, Castro JA. Benznidazole biotransformation in rat heart microsomal fraction without observable ultrastructural alterations: Comparison to Nifurtimox-induced cardiac effects. Memórias do Instituto Oswaldo Cruz 2008;103(6):549-553
  27. 27. Vannier-Santos M, De Castro S. Electron microscopy in antiparasitic chemotherapy: A (close) view to a kill. Current Drug Targets. 2009;10(3):246-260
  28. 28. Vannier-Santos MA, Brunoro GV, Soeiro MN, DeCastro SL, Menna-Barreto FS. Parasite, Compartments, and Molecules: Trick versus Treatment on Chagas Disease. In: De Souza W, editor. Biology of Trypanosoma cruzi. 1st ed. London, UK: IntechOpen; 2019
  29. 29. Menna-Barreto RFS. Cell death pathways in pathogenic trypanosomatids: Lessons of (over)kill. Cell Death & Disease. 2019;10(2):93
  30. 30. Castro JA, Diaz de Toranzo EG. Toxic effects of nifurtimox and benznidazole, two drugs used against American trypanosomiasis (Chagas’ disease). Biomedical and Environmental Sciences. 1988;1(1):19-33
  31. 31. Castro JA, DeMecca MM, Bartel LC. Toxic side effects of drugs used to treat Chagas’ disease (American trypanosomiasis). Human & Experimental Toxicology. 2006;25(8):471-479
  32. 32. Pérez-Molina JA, Crespillo-Andújar C, Bosch-Nicolau P, Molina I. Trypanocidal treatment of Chagas disease. Enfermedades Infecciosas y Microbiología Clínica. 2021;39(9):458-470
  33. 33. Jackson Y, Wyssa B, Chappuis F. Tolerance to nifurtimox and benznidazole in adult patients with chronic Chagas’ disease. The Journal of Antimicrobial Chemotherapy. 2020;75(3):690-696
  34. 34. Olivera MJ, Cucunubá ZM, Álvarez CA, Nicholls RS. Safety profile of nifurtimox and treatment interruption for chronic Chagas disease in Colombian adults. The American Journal of Tropical Medicine and Hygiene. 2015;93(6):1224-1230
  35. 35. Olivera MJ, Cucunubá ZM, Valencia-Hernández CA, Herazo R, Agreda-Rudenko D, Flórez C, et al. Risk factors for treatment interruption and severe adverse effects to benznidazole in adult patients with Chagas disease. PLoS One. 2017;12(9):e0185033
  36. 36. Sperandio da Silva GM, Mediano MFF, da Costa Chambela M, da Silva JA, de Sousa AS, Alvarenga Americano do Brasil PE. A clinical adverse drug reaction prediction model for patients with Chagas disease treated with benznidazole. Antimicrobial Agents and Chemotherapy. 2014;58(11):6371-6377
  37. 37. Hasslocher-Moreno AM, do Brasil PE, de Sousa AS, Xavier SS, Chambela MC, Sperandio da Silva GM. Safety of benznidazole use in the treatment of chronic Chagas’ disease. The Journal of Antimicrobial Chemotherapy. 2012;67(5):1261-1266
  38. 38. Levi GC, Lobo IMF, Kallás EG, Amato NV. Etiological drug treatment of human infection by Trypanosoma cruzi. Revista do Instituto de Medicina Tropical de São Paulo. 1996;38(1):35-38
  39. 39. Crespillo-Andújar C, López-Vélez R, Trigo E, Norman F, Díaz-Menéndez M, Monge-Maillo B, et al. Comparison of the toxicity of two treatment schemes with benznidazole for chronic Chagas disease: A prospective cohort study in two Spanish referral centres. Clinical Microbiology and Infection. 2020;26(3):384
  40. 40. Guggenbühl Noller JM, Froeschl G, Eisermann P, Jochum J, Theuring S, Reiter-Owona I, et al. Describing nearly two decades of Chagas disease in Germany and the lessons learned: A retrospective study on screening, detection, diagnosis, and treatment of Trypanosoma cruzi infection from 2000-2018. BMC Infectious Diseases. 2020;20(1):919
  41. 41. Pérez-Molina JA, Sojo-Dorado J, Norman F, Monge-Maillo B, Díaz-Menéndez M, Albajar-Viñas P, et al. Nifurtimox therapy for Chagas disease does not cause hypersensitivity reactions in patients with such previous adverse reactions during benznidazole treatment. Acta Tropica. 2013;127(2):101-104
  42. 42. Pérez-Molina JA, Molina I. Chagas disease. Lancet. 2018;391(10115):82-94
  43. 43. Viotti R, Vigliano C, Armenti H, Segura E. Treatment of chronic Chagas’ disease with benznidazole: Clinical and serologic evolution of patients with long-term follow-up. American Heart Journal. 1994;127(1):151-162
  44. 44. Hasslocher-Moreno AM, Saraiva RM, Sangenis LHC, Xavier SS, de Sousa AS, Costa AR, et al. Benznidazole decreases the risk of chronic Chagas disease progression and cardiovascular events: A long-term follow up study. eClinicalMedicine. 2021;31:100694
  45. 45. Urbina JA. Parasitological cure of Chagas disease: Is it possible? Is it relevant? Memórias do Instituto Oswaldo Cruz. 1999;94(Suppl. 1):349-355
  46. 46. Cal M, Ioset J-R, Fügi MA, Mäser P, Kaiser M. Assessing anti-T. cruzi candidates in vitro for sterile cidality. International Journal for Parasitology: Drugs and Drug Resistance. 2016;6(3):165-170
  47. 47. Neal RA, van Bueren J. Comparative studies of drug susceptibility of five strains of Trypanosoma cruzi in vivo and in vitro. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1988;82(5):709-714
  48. 48. Lin C, Ferreira de Almeida Fiuza L, Cardoso Santos C, Ferreira Nunes D, Cruz Moreira O, Bouton J, et al. 6-Methyl-7-aryl-7-deazapurine nucleosides as anti-Trypanosoma cruzi agents: Structure-activity relationship and in vivo efficacy. ChemMedChem. 2021;16(14):2231-2253
  49. 49. Pinazo M-J, Espinosa G, Cortes-Lletget C, Posada E, Aldasoro E, Oliveira I, et al. Immunosuppression and Chagas disease: A management challenge. PLoS Neglected Tropical Diseases. 2013;7(1):e1965
  50. 50. Sánchez-Valdéz FJ, Padilla A, Wang W, Orr D, Tarleton RL. Spontaneous dormancy protects Trypanosoma cruzi during extended drug exposure. eLife. 2018;26(7):e34039
  51. 51. Barrett MP, Kyle DE, Sibley LD, Radke JB, Tarleton RL. Protozoan persister-like cells and drug treatment failure. Nature Reviews. Microbiology. 2019;17(10):607-620
  52. 52. Nagajyothi F, Machado FS, Burleigh BA, Jelicks LA, Scherer PE, Mukherjee S, et al. Mechanisms of Trypanosoma cruzi persistence in Chagas disease. Cellular Microbiology. 2012;14(5):634-643
  53. 53. Strömme JH. Effects of diethyldithiocarbamate and disulfiram on glucose metabolism and glutathione content of human erythrocytes. Biochemical Pharmacology. 1963;12(7):705-715
  54. 54. Mittal M, Khan K, Pal S, Porwal K, China SP, Barbhuyan TK, et al. The thiocarbamate disulphide drug, disulfiram induces osteopenia in rats by inhibition of osteoblast function due to suppression of acetaldehyde dehydrogenase activity. Toxicological Sciences. 2014;139(1):257-270
  55. 55. Heikkila RE, Cabbat FS, Cohen G. Inactivation of superoxide dismutase by several thiocarbamic acid derivatives. Experientia. 1978;34(12):1553-1554
  56. 56. Heikkila RE, Cabbat FS, Cohen G. In vivo inhibition of superoxide dismutase in mice by diethyldithiocarbamate. The Journal of Biological Chemistry. 1976;251(7):2182-2185
  57. 57. Doyon G, Zerbato J, Mellors JW, Sluis-Cremer N. Disulfiram reactivates latent HIV-1 expression through depletion of the phosphatase and tensin homolog. AIDS. 2013;27(2):F7-F11
  58. 58. Kim Y, Anderson JL, Lewin SR. Getting the “Kill” into “Shock and Kill”: Strategies to eliminate latent HIV. Cell Host & Microbe. 2018;23(1):14-26
  59. 59. Maeda FY, Cortez C, Yoshida N. Cell signaling during Trypanosoma cruzi invasion. Frontiers in Immunology. 2012;3:361
  60. 60. Burleigh BA, Woolsey AM. Cell signalling and Trypanosoma cruzi invasion. Cellular Microbiology. 2002;4(11):701-711
  61. 61. Chuenkova MV, Furnari FB, Cavenee WK, Pereira MA. Trypanosoma cruzi trans-sialidase: A potent and specific survival factor for human Schwann cells by means of phosphatidylinositol 3-kinase/Akt signaling. Proceedings of the National Academy of Sciences. 2001;98(17):9936-9941
  62. 62. Zhang H, Chen D, Ringler J, Chen W, Cui QC, Ethier SP, et al. Disulfiram treatment facilitates phosphoinositide 3-kinase inhibition in human breast cancer cells in vitro and in vivo. Cancer Research. 2010;70(10):3996-4004
  63. 63. Bustamante JM, Sanchez-Valdez F, Padilla AM, White B, Wang W, Tarleton RL. A modified drug regimen clears active and dormant trypanosomes in mouse models of Chagas disease. Science Translational Medicine. 2020;12(567):eabb7656
  64. 64. Pessenda G, Silva JS. Arginase and its mechanisms in Leishmania persistence. Parasite Immunology. 2020;42(7):e12722
  65. 65. Mejia AM, Hall BS, Taylor MC, Gómez-Palacio A, Wilkinson SR, Triana-Chávez O, et al. Benznidazole-resistance in Trypanosoma cruzi is a readily acquired trait that can arise independently in a single population. The Journal of Infectious Diseases. 2012;206(2):220-228
  66. 66. Campos MCO, Leon LL, Taylor MC, Kelly JM. Benznidazole-resistance in Trypanosoma cruzi: Evidence that distinct mechanisms can act in concert. Molecular and Biochemical Parasitology. 2014;193(1):17-19
  67. 67. Francisco AF, Jayawardhana S, Olmo F, Lewis MD, Wilkinson SR, Taylor MC, et al. Challenges in Chagas disease drug development. Molecules. 2020;25(12):2799
  68. 68. Filardi LS, Brener Z. Susceptibility and natural resistance of Trypanosoma cruzi strains to drugs used clinically in Chagas disease. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1987;81(5):755-759
  69. 69. Zingales B. Trypanosoma cruzi genetic diversity: Something new for something known about Chagas disease manifestations, serodiagnosis and drug sensitivity. Acta Tropica. 2018;184:38-52
  70. 70. Teston APM, Monteiro WM, Reis D, Bossolani GDP, Gomes ML, de Araújo SM, et al. In vivo susceptibility to benznidazole of Trypanosoma cruzi strains from the western Brazilian Amazon. Tropical Medicine & International Health. 2013;18(1):85-95
  71. 71. Zingales B, Araujo RGA, Moreno M, Franco J, Aguiar PHN, Nunes SL, et al. A novel ABCG-like transporter of Trypanosoma cruzi is involved in natural resistance to benznidazole. Memórias do Instituto Oswaldo Cruz. 2015;110(3):433-444
  72. 72. Loffredo L, Violi F. COVID-19 and cardiovascular injury: A role for oxidative stress and antioxidant treatment? International Journal of Cardiology. 2020;312:136
  73. 73. Dichi I, Breganó JW, Simão ANC, Cecchini R. Role of Oxidative Stress in Chronic Diseases. 1st ed. Boca Raton: CRC Press; 2014. 720 p
  74. 74. Bondy SC, Campbell A. Inflammation, aging, and oxidative stress. In: Bondy SC, Campbell A, editors. Oxidative Stress in Applied Basic Research and Clinical Practice. Cham: Springer International Publishing; 2016
  75. 75. Armstrong D, Stratton RD. Oxidative Stress and Antioxidant Protection: The Science of Free Radical Biology and Disease. 1st ed. Hoboken, New Jersey: John Wiley & Sons Inc.; 2016. 600 p
  76. 76. Sies H. (Ed.). Oxidative stress: eustress and distress. London: Academic Press; 2020. 829 p
  77. 77. Lushchak V, Semchyshyn H, editors. Oxidative Stress: Molecular Mechanisms and Biological Effects. London, UK: InTechOpen; 2012
  78. 78. Flohé L. Glutathione. Boca Raton: Taylor & Francis; 2018
  79. 79. Piñeyro MD, Arias D, Parodi-Talice A, Guerrero S, Robello C. Trypanothione metabolism as drug target for trypanosomatids. Current Pharmaceutical Design. 2021;27(15):1834-1846
  80. 80. Fairlamb AH, Cerami A. Metabolism and functions of trypanothione in the kinetoplastida. Annual Review of Microbiology. 1992;46(1):695-729
  81. 81. Pountain AW, Weidt SK, Regnault C, Bates PA, Donachie AM, Dickens NJ, et al. Genomic instability at the locus of sterol C24-methyltransferase promotes amphotericin B resistance in Leishmania parasites. PLoS Neglected Tropical Diseases. 2019;13(2):e0007052
  82. 82. Ozols RF, O’Dwyer PJ, Hamilton TC, Young RC. The role of glutathione in drug resistance. Cancer Treatment Reviews. 1990;17:45-50
  83. 83. Schröder CP, Godwin AK, O’Dwyer PJ, Tew KD, Hamilton TC, Ozols RF. Glutathione and drug resistance. Cancer Investigation. 1996;14(2):158-168
  84. 84. Russell TM, Azad MG, Richardson DR. The relationship of glutathione-S-transferase and multi-drug resistance-related protein 1 in nitric oxide (NO) transport and storage. Molecules. 2021;26(19):5784
  85. 85. Moncada C, Repetto Y, Aldunate J, Letelier ME, Morello A. Role of glutathione in the susceptibility of Trypanosoma cruzi to drugs. Comparative Biochemistry and Physiology - Part C. 1989;94(1):87-91
  86. 86. Repetto Y, Opazo E, Maya JD, Agosin M, Morello A. Glutathione and trypanothione in several strains of Trypanosoma cruzi: Effect of drugs. Comparative Biochemistry and Physiology, Part B: Biochemistry & Molecular Biology. 1996;115(2):281-285
  87. 87. Mesías AC, Sasoni N, Arias DG, Pérez Brandán C, Orban OCF, Kunick C, et al. Trypanothione synthetase confers growth, survival advantage and resistance to anti-protozoal drugs in Trypanosoma cruzi. Free Radical Biology & Medicine. 2019;130:23-34
  88. 88. Faúndez M, Pino L, Letelier P, Ortiz C, López R, Seguel C, et al. Buthionine sulfoximine increases the toxicity of nifurtimox and benznidazole to Trypanosoma cruzi. Antimicrobial Agents and Chemotherapy. 2005;49(1):126-130
  89. 89. Faúndez M, López-Muñoz R, Torres G, Morello A, Ferreira J, Kemmerling U, et al. Buthionine sulfoximine has anti-Trypanosoma cruzi activity in a murine model of acute Chagas’ disease and enhances the efficacy of nifurtimox. Antimicrobial Agents and Chemotherapy. 2008;52(5):1837-1839
  90. 90. Carter KC, Sundar S, Spickett C, Pereira OC, Mullen AB. The in vivo susceptibility of Leishmania donovani to sodium stibogluconate is drug specific and can be reversed by inhibiting glutathione biosynthesis. Antimicrobial Agents and Chemotherapy. 2003;47(5):1529-1535
  91. 91. Vannier-Santos MA, Suarez-Fontes AM. Role of polyamines in parasite cell architecture and function. Current Pharmaceutical Design. 2017;23(23):3342-3358
  92. 92. Phillips MA. Polyamines in protozoan pathogens. The Journal of Biological Chemistry. 2018;293(48):18746-18756
  93. 93. Hernández SM, Sánchez MS, de Tarlovsky MNS. Polyamines as a defense mechanism against lipoperoxidation in Trypanosoma cruzi. Acta Tropica. 2006;98(1):94-102
  94. 94. Birkholtz L-M, Williams M, Niemand J, Louw AI, Persson L, Heby O. Polyamine homoeostasis as a drug target in pathogenic protozoa: Peculiarities and possibilities. The Biochemical Journal. 2011;438(2):229-244
  95. 95. Roberts S, Ullman B. Parasite polyamines as pharmaceutical targets. Current Pharmaceutical Design. 2017;23(23):3325-3341
  96. 96. Talevi A, Carrillo C, Comini M. The thiol-polyamine metabolism of Trypanosoma cruzi: Molecular targets and drug repurposing strategies. Current Medicinal Chemistry. 2019;26(36):6614-6635
  97. 97. de Oliveira TB, Pedrosa RC, Filho DW. Oxidative stress in chronic cardiopathy associated with Chagas disease. International Journal of Cardiology. 2007;116(3):357-363
  98. 98. Gupta S, Wen J-J, Garg NJ. Oxidative sress in Chagas disease. Interdisciplinary Perspectives on Infectious Diseases. 2009;2009:1-8
  99. 99. Zacks MA, Wen J-J, Vyatkina G, Bhatia V, Garg N. An overview of chagasic cardiomyopathy: Pathogenic importance of oxidative stress. Anais da Academia Brasileira de Ciências. 2005;77(4):695-715
  100. 100. Wen J, Yachelini PC, Sembaj A, Manzur RE, Garg NJ. Increased oxidative stress is correlated with mitochondrial dysfunction in chagasic patients. Free Radical Biology & Medicine. 2006;41(2):270-276
  101. 101. Turrens JF. Superoxide production by the mitochondrial respiratory chain. Bioscience Reports. 1997;17(1):3-8
  102. 102. Turrens JF. Mitochondrial formation of reactive oxygen species. The Journal of Physiology. 2003;552(2):335-344
  103. 103. Prolo C, Estrada D, Piacenza L, Benítez D, Comini MA, Radi R, et al. Nox2-derived superoxide radical is crucial to control acute Trypanosoma cruzi infection. Redox Biology. 2021;46:102085
  104. 104. Wen J-J, Garg N. Oxidative modification of mitochondrial respiratory complexes in response to the stress of Trypanosoma cruzi infection. Free Radical Biology & Medicine. 2004;37(12):2072-2081
  105. 105. Dhiman M, Wan X, Popov VL, Vargas G, Garg NJ. MnSOD tg mice control myocardial inflammatory and oxidative stress and remodeling responses elicited in chronic Chagas disease. Journal of the American Heart Association. 2013;2(5):e000302
  106. 106. Maldonado E, Rojas DA, Morales S, Miralles V, Solari A. Dual and opposite roles of reactive oxygen species (ROS) in Chagas disease: Beneficial on the pathogen and harmful on the host. Oxidative Medicine and Cellular Longevity. 2020;2020:1-17
  107. 107. Maldonado E, Rojas DA, Urbina F, Solari A. The oxidative stress and chronic inflammatory process in Chagas disease: Role of exosomes and contributing genetic factors. Oxidative Medicine and Cellular Longevity. 2021;2021:1-21
  108. 108. Wen J-J, Vyatkina G, Garg N. Oxidative damage during chagasic cardiomyopathy development: Role of mitochondrial oxidant release and inefficient antioxidant defense. Free Radical Biology & Medicine. 2004;37(11):1821-1833
  109. 109. Dantas-Pereira L, Menna-Barreto R, Lannes-Vieira J. Extracellular vesicles: Potential role in remote signaling and inflammation in Trypanosoma cruzi triggered disease. Frontiers in Cell and Development Biology. 2021;9:798054
  110. 110. Chuenkova MV, Pereiraperrin M. Neurodegeneration and neuroregeneration in Chagas disease. Advances in Parasitology. 2011;76:195-233
  111. 111. Vilar-Pereira G, Castaño Barrios L, Silva AA, Martins Batista A, Resende Pereira I, Cruz Moreira O, et al. Memory impairment in chronic experimental Chagas disease: Benznidazole therapy reversed cognitive deficit in association with reduction of parasite load and oxidative stress in the nervous tissue. PLoS One. 2021;16(1):e0244710
  112. 112. Wen J-J, Garg NJ. Mitochondrial generation of reactive oxygen species is enhanced at the Qo site of the complex III in the myocardium of Trypanosoma cruzi-infected mice: Beneficial effects of an antioxidant. Journal of Bioenergetics and Biomembranes. 2008;40(6):587-598
  113. 113. Sánchez-Villamil JP, Bautista-Niño PK, Serrano NC, Rincon MY, Garg NJ. Potential role of antioxidants as adjunctive therapy in Chagas disease. Oxidative Medicine and Cellular Longevity. 2020;2020:1-13
  114. 114. Du J, Cullen JJ, Buettner GR. Ascorbic acid: Chemistry, biology and the treatment of cancer. Biochimica et Biophysica Acta, Reviews on Cancer. 2012;1826(2):443-457
  115. 115. Mastrangelo D, Pelosi E, Castelli G, Lo-Coco F, Testa U. Mechanisms of anti-cancer effects of ascorbate: Cytotoxic activity and epigenetic modulation. Blood Cells, Molecules & Diseases. 2018;69:57-64
  116. 116. Puente V, Demaria A, Frank FM, Batlle A, Lombardo ME. Anti-parasitic effect of vitamin C alone and in combination with benznidazole against Trypanosoma cruzi. PLoS Neglected Tropical Diseases. 2018;12(9):e0006764
  117. 117. Providello MV, Carneiro ZA, Portapilla GB, do Vale GT, Camargo RS, Tirapelli CR, et al. Benefits of ascorbic acid in association with low-dose benznidazole in treatment of Chagas disease. Antimicrobial Agents and Chemotherapy. 2018;62(9):e00514-18
  118. 118. Piacenza L, Trujillo M, Radi R. Reactive species and pathogen antioxidant networks during phagocytosis. The Journal of Experimental Medicine. 2019;216(3):501-516
  119. 119. Piacenza L, Peluffo G, Alvarez MN, Martínez A, Radi R. Trypanosoma cruzi antioxidant enzymes as virulence factors in Chagas disease. Antioxidants & Redox Signaling. 2013;19(7):723-734
  120. 120. Oyenihi OR, Oyenihi AB, Alabi TD, Tade OG, Adeyanju AA, Oguntibeju OO. Reactive oxygen species: Key players in the anticancer effects of apigenin? Journal of Food Biochemistry. 2022;46(2):e14060
  121. 121. Goes GR, Rocha PS, Diniz ARS, Aguiar PHN, Machado CR, Vieira LQ. Trypanosoma cruzi needs a signal provided by reactive oxygen species to infect macrophages. PLoS Neglected Tropical Diseases. 2016;10(4):e0004555
  122. 122. Paiva CN, Feijó DF, Dutra FF, Carneiro VC, Freitas GB, Alves LS, et al. Oxidative stress fuels Trypanosoma cruzi infection in mice. The Journal of Clinical Investigation. 2012;122(7):2531-2542
  123. 123. Dumoulin PC, Burleigh BA. Stress induced proliferation and cell cycle plasticity of intracellular Trypanosoma cruzi amastigotes. MBio. 2018;9(4):e00673-18
  124. 124. Batinić-Haberle I, Rebouças JS, Spasojević I. Redox-Active Therapeutics. 1st ed. Switzerland: Springer International Publishing; 2016. 1261 p
  125. 125. Turrens JF. Oxidative stress and antioxidant defenses: A target for the treatment of diseases caused by parasitic protozoa. Molecular Aspects of Medicine. 2004;25(1-2):211-220
  126. 126. Docampo R, Moreno SNJ. Free radical metabolites in the mode of action of chemotherapeutic agents and phagocytic cells on Trypanosoma cruzi. Clinical Infectious Diseases. 1984;6(2):223-238
  127. 127. Beltran-Hortelano I, Perez-Silanes S, Galiano S. Trypanothione reductase and superoxide dismutase as current drug targets for Trypanosoma cruzi: An overview of compounds with activity against Chagas disease. Current Medicinal Chemistry. 2017;24(11):1066-1138
  128. 128. Boveris A, Docampo R, Turrens JF, Stoppani AO. Effect of β-lapachone on superoxide anion and hydrogen peroxide production in Trypanosoma cruzi. The Biochemical Journal. 1978;175(2):431-439
  129. 129. Cruz FS, Docampo R, de Souza W. Effect of beta-lapachone on hydrogen peroxide production in Trypanosoma cruzi. Acta Tropica. 1978;35(1):35-40
  130. 130. Docampo R, Cruz FS, Boveris A, Muniz RPA, Esquivel DMS. Lipid peroxidation and the generation of free radicals, superoxide anion, and hydrogen peroxide in β-lapachone-treated Trypanosoma cruzi epimastigotes. Archives of Biochemistry and Biophysics. 1978;186(2):292-297
  131. 131. Bombaça ACS, Viana PG, Santos ACC, Silva TL, Rodrigues ABM, Guimarães ACR, et al. Mitochondrial disfunction and ROS production are essential for anti-Trypanosoma cruzi activity of β-lapachone-derived naphthoimidazoles. Free Radical Biology & Medicine. 2019;130:408-418
  132. 132. Gong Q , Hu J, Wang P, Li X, Zhang X. A comprehensive review on β-lapachone: Mechanisms, structural modifications, and therapeutic potentials. European Journal of Medicinal Chemistry. 2021;210:112962
  133. 133. Menna-Barreto RFS, Henriques-Pons A, Pinto AV, Morgado-Diaz JA, Soares MJ, De Castro SL. Effect of a β-lapachone-derived naphthoimidazole on Trypanosoma cruzi: Identification of target organelles. The Journal of Antimicrobial Chemotherapy. 2005;56(6):1034-1041
  134. 134. dos Anjos DO, Sobral Alves ES, Gonçalves VT, Fontes SS, Nogueira ML, Suarez-Fontes AM, et al. Effects of a novel β-lapachone derivative on Trypanosoma cruzi: Parasite death involving apoptosis, autophagy and necrosis. International Journal for Parasitology: Drugs and Drug Resistance. 2016;6(3):207-219
  135. 135. Yang Y, Karakhanova S, Hartwig W, D’Haese JG, Philippov PP, Werner J, et al. Mitochondria and mitochondrial ROS in cancer: Novel targets for anticancer therapy. Journal of Cellular Physiology. 2016;231(12):2570-2581
  136. 136. Dhote V, Samundre P, Ganeshpurkar A, Upaganlawar A. Aging of brain related with mitochondrial dysfunctions. Current Drug Targets. 2021;22(14):1668-1687
  137. 137. Dabravolski SA, Nikiforov NG, Starodubova AV, Popkova TV, Orekhov AN. The role of mitochondria-derived peptides in cardiovascular diseases and their potential as therapeutic targets. International Journal of Molecular Sciences. 2021;22(16):8770
  138. 138. Clayton SA, MacDonald L, Kurowska-Stolarska M, Clark AR. Mitochondria as key players in the pathogenesis and treatment of rheumatoid arthritis. Frontiers in Immunology. 2021;12:673916
  139. 139. Sharma VK, Singh TG, Mehta V. Stressed mitochondria: A target to intrude Alzheimer’s disease. Mitochondrion. 2021;59:48-57
  140. 140. Fakhri S, Abdian S, Zarneshan SN, Akkol EK, Farzaei MH, Sobarzo-Sánchez E. Targeting mitochondria by plant secondary metabolites: A promising strategy in combating Parkinson’s disease. International Journal of Molecular Sciences. 2021;22(22):12570
  141. 141. Kita K, Nihei C, Tomitsuka E. Parasite mitochondria as drug target: Diversity and dynamic changes during the life cycle. Current Medicinal Chemistry. 2003;10(23):2535-2548
  142. 142. Monzote L, Gille L. Mitochondria as a promising antiparasitic target. Current Clinical Pharmacology. 2010;5(1):55-66
  143. 143. Mather M, Henry K, Vaidya A. Mitochondrial drug targets in apicomplexan parasites. Current Drug Targets. 2007;8(1):49-60
  144. 144. Sen N, Majumder H. Mitochondrion of protozoan parasite emerges as potent therapeutic target: Exciting drugs are on the horizon. Current Pharmaceutical Design. 2008;14(9):839-846
  145. 145. Goodman CD, Buchanan HD, McFadden GI. Is the mitochondrion a good malaria drug target? Trends in Parasitology. 2017;33(3):185-193
  146. 146. Nihei C, Fukai Y, Kita K. Trypanosome alternative oxidase as a target of chemotherapy. Biochimica et Biophysica Acta, Molecular Basis of Disease. 2002;1587(2-3):234-239
  147. 147. Machado MM. Kinetoplast as a potential chemotherapeutic target of trypanosomatids. Current Pharmaceutical Design. 2008;14(9):847-854
  148. 148. Fidalgo LM, Gille L. Mitochondria and trypanosomatids: Targets and drugs. Pharmaceutical Research. 2011;28(11):2758-2770
  149. 149. Boveris A, Chance B. The mitochondrial generation of hydrogen peroxide. General properties and effect of hyperbaric oxygen. The Biochemical Journal. 1973;134(3):707-716
  150. 150. Menna-Barreto RFS, de Castro SL. The double-edged sword in pathogenic trypanosomatids: The pivotal role of mitochondria in oxidative stress and bioenergetics. BioMed Research International. 2014;2014:1-14
  151. 151. Turrens JF, McCord JM. The iron-containing superoxide dismutases of Trypanosomatidae. Free Radical Biology & Medicine. 2006;40(2):193-195
  152. 152. Martínez A, Prolo C, Estrada D, Rios N, Alvarez MN, Piñeyro MD, et al. Cytosolic Fe-superoxide dismutase safeguards Trypanosoma cruzi from macrophage-derived superoxide radical. Proceedings of the National Academy of Sciences. 2019;116(18):8879-8888
  153. 153. Docampo R, Moreno SN. Free radical metabolism of antiparasitic agents. Federation Proceedings. 1986;45(10):2471-2476
  154. 154. Docampo R. Sensitivity of parasites to free radical damage by antiparasitic drugs. Chemico-Biological Interactions. 1990;73(1):1-27
  155. 155. Perbandt M, Ndjonka D, Liebau E. Protective mechanisms of Helminths against reactive oxygen species are highly promising drug targets. Current Medicinal Chemistry. 2014;21(15):1794-1808
  156. 156. Pal C, Bandyopadhyay U. Redox-active antiparasitic drugs. Antioxidants & Redox Signaling. 2012;17(4):555-582
  157. 157. Piacenza L, Irigoín F, Alvarez MN, Peluffo G, Taylor MC, Kelly JM, et al. Mitochondrial superoxide radicals mediate programmed cell death in Trypanosoma cruzi: Cytoprotective action of mitochondrial iron superoxide dismutase overexpression. The Biochemical Journal. 2007;403(2):323-334
  158. 158. Maya JD, Cassels BK, Iturriaga-Vásquez P, Ferreira J, Faúndez M, Galanti N, et al. Mode of action of natural and synthetic drugs against Trypanosoma cruzi and their interaction with the mammalian host. Comparative Biochemistry and Physiology, Part A: Molecular & Integrative Physiology. 2007;146(4):601-620
  159. 159. Docampo R, Mason RP, Mottley C, Muniz RP. Generation of free radicals induced by nifurtimox in mammalian tissues. The Journal of Biological Chemistry. 1981;256(21):10930-10933
  160. 160. Faúndez M, Rojas M, Bohle P, Reyes C, Letelier ME, Aliaga ME, et al. Pyrogallol red oxidation induced by superoxide radicals: Application to evaluate redox cycling of nitro compounds. Analytical Biochemistry. 2011;419(2):284-291
  161. 161. Moreno SNJ, Docampo R, Mason RP, Leon W, Stoppani AOM. Different behaviors of benznidazole as free radical generator with mammalian and Trypanosoma cruzi microsomal preparations. Archives of Biochemistry and Biophysics. 1982;218(2):585-591
  162. 162. Nogueira FB, Krieger MA, Nirdé P, Goldenberg S, Romanha AJ, Murta SMF. Increased expression of iron-containing superoxide dismutase-A (TcFeSOD-A) enzyme in Trypanosoma cruzi population with in vitro-induced resistance to benznidazole. Acta Tropica. 2006;100(1-2):119-132
  163. 163. Campos MC, Phelan J, Francisco AF, Taylor MC, Lewis MD, Pain A, et al. Genome-wide mutagenesis and multi-drug resistance in American trypanosomes induced by the front-line drug benznidazole. Scientific Reports. 2017;7(1):14407
  164. 164. Andrade HM, Murta SMF, Chapeaurouge A, Perales J, Nirdé P, Romanha AJ. Proteomic analysis of Trypanosoma cruzi resistance to benznidazole. Journal of Proteome Research. 2008;7(6):2357-2367
  165. 165. Quebrada Palacio LP, González MN, Hernandez-Vasquez Y, Perrone AE, Parodi-Talice A, Bua J, et al. Phenotypic diversity and drug susceptibility of Trypanosoma cruzi TcV clinical isolates. PLoS One. 2018;13(9):e0203462
  166. 166. Prathalingham SR, Wilkinson SR, Horn D, Kelly JM. Deletion of the Trypanosoma brucei superoxide dismutase gene sodb1 increases sensitivity to nifurtimox and benznidazole. Antimicrobial Agents and Chemotherapy. 2007;51(2):755-758
  167. 167. Veronica J, Chandrasekaran S, Dayakar A, Devender M, Prajapati VK, Sundar S, et al. Iron superoxide dismutase contributes to miltefosine resistance in Leishmania donovani. The FEBS Journal. 2019;286(17):3488-3503
  168. 168. Santi AMM, Silva PA, Santos IFM, Murta SMF. Downregulation of FeSOD-A expression in Leishmania infantum alters trivalent antimony and miltefosine susceptibility. Parasites & Vectors. 2021;14(1):366
  169. 169. Wassmann C, Hellberg A, Tannich E, Bruchhaus I. Metronidazole resistance in the protozoan parasite Entamoeba histolytica is associated with increased expression of iron-containing superoxide dismutase and peroxiredoxin and decreased expression of ferredoxin 1 and flavin reductase. The Journal of Biological Chemistry. 1999;274(37):26051-26056
  170. 170. Andrade JM, Murta SMF. Functional analysis of cytosolic tryparedoxin peroxidase in antimony-resistant and -susceptible Leishmania braziliensis and Leishmania infantum lines. Parasites & Vectors. 2014;7(1):406
  171. 171. Olmo F, Urbanová K, Rosales MJ, Martín-Escolano R, Sánchez-Moreno M, Marín C. An in vitro iron superoxide dismutase inhibitor decreases the parasitemia levels of Trypanosoma cruzi in BALB/c mouse model during acute phase. International Journal for Parasitology: Drugs and Drug Resistance. 2015;5(3):110-116
  172. 172. Singh CK, Chhabra G, Ndiaye MA, Garcia-Peterson LM, Mack NJ, Ahmad N. The role of sirtuins in antioxidant and redox signaling. Antioxidants & Redox Signaling. 2018;28(8):643-661
  173. 173. Klein MA, Denu JM. Biological and catalytic functions of sirtuin 6 as targets for small-molecule modulators. The Journal of Biological Chemistry. 2020;295(32):11021-11041
  174. 174. Huang J-Y, Hirschey MD, Shimazu T, Ho L, Verdin E. Mitochondrial sirtuins. Biochimica et Biophysica Acta, Proteins and Proteomics. 2010;1804(8):1645-1651
  175. 175. Wan X, Garg NJ. Sirtuin control of mitochondrial dysfunction, oxidative stress, and inflammation in Chagas disease models. Frontiers in Cellular and Infection Microbiology. 2021;11:693051
  176. 176. Chen Y, Zhang J, Lin Y, Lei Q , Guan K, Zhao S, et al. Tumour suppressor SIRT3 deacetylates and activates manganese superoxide dismutase to scavenge ROS. EMBO Reports. 2011;12(6):534-541
  177. 177. dos Santos ML, Santana Nunes V, Gomes AAS, Sousa AC de CN, Fontes MRM, Schenkman S, et al. Mitochondrial sirtuin TcSir2rp3 affects TcSODA activity and oxidative stress response in Trypanosoma cruzi. Frontiers in Cellular and Infection Microbiology. 2021;11:773410
  178. 178. Davis CD, Brooks L, Calisi C, Bennett BJ, McElroy DM. Beneficial effect of selenium supplementation during murine infection with Trypanosoma cruzi. The Journal of Parasitology. 1998;84(6):1274-1277
  179. 179. Gomez RM, Solana ME, Levander OA. Host selenium deficiency increases the severity of chronic inflammatory myopathy in Trypanosoma cruzi-inoculated mice. The Journal of Parasitology. 2002;88(3):541-547
  180. 180. de Souza AP, de Oliveira GM, Vanderpas J, de Castro SL, Rivera MT, Araújo-Jorge TC. Selenium supplementation at low doses contributes to the decrease in heart damage in experimental Trypanosoma cruzi infection. Parasitology Research. 2003;91(1):51-54
  181. 181. Holanda MT, Mediano MFF, Hasslocher-Moreno AM, Gonzaga BMS, Carvalho ACC, Ferreira RR, et al. Effects of selenium treatment on cardiac function in Chagas heart disease: Results from the STCC randomized trial. eClinicalMedicine. 2021;40:101105
  182. 182. Correa-Oliveira R, Araújo-Jorge TC, Rocha MOC, Rivera MT, Nève J, Moreno AHM, et al. Progressive Chagas’ cardiomyopathy is associated with low selenium levels. The American Journal of Tropical Medicine and Hygiene. 2002;66(6):706-712
  183. 183. Alcolea V, Pérez-Silanes S. Selenium as an interesting option for the treatment of Chagas disease: A review. European Journal of Medicinal Chemistry. 2020;206:112673
  184. 184. Maldonado E, Rojas DA, Urbina F, Solari A. The use of antioxidants as potential co-adjuvants to treat chronic Chagas disease. Antioxidants. 2021;10(7):1022
  185. 185. Martín-Escolano R, Etxebeste-Mitxeltorena M, Martín-Escolano J, Plano D, Rosales MJ, Espuelas S, et al. Selenium derivatives as promising therapy for Chagas disease: In vitro and in vivo studies. ACS Infectious Diseases. 2021;7(6):1727-1738
  186. 186. de Freitas MRB, da Costa CMB, Pereira LM, do Prado JC, Sala MA, Abrahão AA. The treatment with selenium increases placental parasitismin pregnant Wistar rats infected with the Y strain of Trypanosoma cruzi. Immunobiology. 2018;223(10):537-543
  187. 187. Kim SJ, Choi MC, Park JM, Chung AS. Antitumor effects of selenium. International Journal of Molecular Sciences. 2021;22(21):11844
  188. 188. Zheng Y, Xie T, Li S, Wang W, Wang Y, Cao Z, et al. Effects of selenium as a dietary source on performance, inflammation, cell damage, and reproduction of livestock induced by heat stress: A review. Frontiers in Immunology. 2022;12:820853
  189. 189. Orian L, Flohé L. Selenium-catalyzed reduction of hydroperoxides in chemistry and biology. Antioxidants. 2021;10(10):1560
  190. 190. Planer JD, Hulverson MA, Arif JA, Ranade RM, Don R, Buckner FS. Synergy testing of FDA-approved drugs identifies potent drug combinations against Trypanosoma cruzi. PLoS Neglected Tropical Diseases. 2014;8(7):e2977
  191. 191. Kaiser M, Mäser P, Tadoori LP, Ioset J-R, Brun R. Antiprotozoal activity profiling of approved drugs: A starting point toward drug repositioning. PLoS One. 2015;10(8):e0135556
  192. 192. Lehár J, Krueger AS, Avery W, Heilbut AM, Johansen LM, Price ER, et al. Synergistic drug combinations tend to improve therapeutically relevant selectivity. Nature Biotechnology. 2009;27(7):659-666
  193. 193. Lehar J, Krueger AS, Zimmermann GR, Borisy AA. Therapeutic selectivity and the multi-node drug target. Discovery Medicine. 2009;8(43):185-190
  194. 194. Zimmermann GR, Lehár J, Keith CT. Multi-target therapeutics: When the whole is greater than the sum of the parts. Drug Discovery Today. 2007;12(1-2):34-42
  195. 195. Borisy AA, Elliott PJ, Hurst NW, Lee MS, Lehar J, Price ER, et al. Systematic discovery of multicomponent therapeutics. Proceedings of the National Academy of Sciences. 2003;100(13):7977-7982
  196. 196. Kabir A, Muth A. Polypharmacology: The science of multi-targeting molecules. Pharmacological Research. 2022;176:106055
  197. 197. Aguilera E, Alvarez G, Cerecetto H, González M. Polypharmacology in the treatment of Chagas disease. Current Medicinal Chemistry. 2019;26(23):4476-4489
  198. 198. Mitchison DA. Prevention of drug resistance by combined drug treatment of tuberculosis. In: Coates, A, editor. Antibiotic Resistance. Handbook of Experimental Pharmacology. Springer, Berlin, Heidelberg. 2012;211:87-98
  199. 199. Worthington RJ, Melander C. Combination approaches to combat multidrug-resistant bacteria. Trends in Biotechnology. 2013;31(3):177-184
  200. 200. Hill JA, Cowen LE. Using combination therapy to thwart drug resistance. Future Microbiology. 2015;10(11):1719-1726
  201. 201. Yadav B, Wennerberg K, Aittokallio T, Tang J. Searching for drug synergy in complex dose–response landscapes using an interaction potency model. Computational and Structural Biotechnology Journal. 2015;13:504-513
  202. 202. Singh N, Yeh PJ. Suppressive drug combinations and their potential to combat antibiotic resistance. Journal of Antibiotics (Tokyo). 2017;70(11):1033-1042
  203. 203. Wani M, Ahmad A. Combination Therapy Against Multidrug Resistance. 1st ed. London: Elsevier, Academic Press; 2020. 260 p
  204. 204. Liu Y, Tong Z, Shi J, Li R, Upton M, Wang Z. Drug repurposing for next-generation combination therapies against multidrug-resistant bacteria. Theranostics. 2021;11(10):4910-4928
  205. 205. Wongsrichanalai C, Pickard AL, Wernsdorfer WH, Meshnick SR. Epidemiology of drug-resistant malaria. The Lancet Infectious Diseases. 2002;2(4):209-218
  206. 206. Tu Y. From Artemisia annua L. to Artemisinins the Discovery and Development of Artemisinins and Antimalarial Agents. 1st ed. London: Agents Academic Press; 2017. 468 p
  207. 207. Ashley EA, Dhorda M, Fairhurst RM, Amaratunga C, Lim P, Suon S, et al. Spread of Artemisinin resistance in Plasmodium falciparum malaria. The New England Journal of Medicine. 2014;371(5):411-423
  208. 208. Eastman RT, Fidock DA. Artemisinin-based combination therapies: A vital tool in efforts to eliminate malaria. Nature Reviews. Microbiology. 2009;7(12):864-874
  209. 209. Rogers WO, Sem R, Tero T, Chim P, Lim P, Muth S, et al. Failure of artesunate-mefloquine combination therapy for uncomplicated Plasmodium falciparum malaria in Southern Cambodia. Malaria Journal. 2009;8(1):10
  210. 210. Lim P, Alker AP, Khim N, Shah NK, Incardona S, Doung S, et al. Pfmdr1 copy number and arteminisin derivatives combination therapy failure in falciparum malaria in Cambodia. Malaria Journal. 2009;8(1):11
  211. 211. Alker AP, Lim P, Sem R, Shah NK, Yi P, Bouth DM, et al. Pfmdr1 and in vivo resistance to artesunate-mefloquine in falciparum malaria on the Cambodian-Thai border. The American Journal of Tropical Medicine and Hygiene. 2007;76(4):641-647
  212. 212. Nosengo N. Can you teach old drugs new tricks? Nature. 2016;534(7607):314-316
  213. 213. Xue H, Li J, Xie H, Wang Y. Review of drug repositioning approaches and resources. International Journal of Biological Sciences. 2018;14(10):1232-1244
  214. 214. Simões-Silva MR, De Araújo JS, Peres RB, Da Silva PB, Batista MM, De Azevedo LD, et al. Repurposing strategies for Chagas disease therapy: The effect of imatinib and derivatives against Trypanosoma cruzi. Parasitology. 2019;146(8):1006-1012
  215. 215. Juárez-Saldivar A, Schroeder M, Salentin S, Haupt VJ, Saavedra E, Vázquez C, et al. Computational drug repositioning for Chagas disease using protein-ligand interaction profiling. International Journal of Molecular Sciences. 2020;21(12):4270
  216. 216. Trindade JDS, Freire-de-Lima CG, Côrte-Real S, Decote-Ricardo D, Freire de Lima ME. Drug repurposing for Chagas disease: In vitro assessment of nimesulide against Trypanosoma cruzi and insights on its mechanisms of action. PLoS One. 2021;16(10):e0258292
  217. 217. Martinez-Peinado N, Cortes-Serra N, Sherman J, Rodriguez A, Bustamante JM, Gascon J, et al. Identification of Trypanosoma cruzi growth inhibitors with activity in vivo within a collection of licensed drugs. Microorganisms. 2021;9(2):406
  218. 218. Rivero CV, Martínez SJ, Novick P, Cueto JA, Salassa BN, Vanrell MC, et al. Repurposing Carvedilol as a novel inhibitor of the Trypanosoma cruzi autophagy flux that affects parasite replication and survival. Frontiers in Cellular and Infection Microbiology. 2021;11:657257
  219. 219. Valsecchi WM, Delfino JM, Santos J, Fernández Villamil SH. Zoledronate repositioning as a potential trypanocidal drug. Trypanosoma cruzi HPRT an alternative target to be considered. Biochemical Pharmacology. 2021;188:114524
  220. 220. Bellera CL, Alberca LN, Sbaraglini ML, Talevi A. In silico drug repositioning for Chagas disease. Current Medicinal Chemistry. 2020;27(5):662-675
  221. 221. Reigada C, Sayé M, Phanstiel O, Valera-Vera E, Miranda MR, Pereira CA. Identification of Trypanosoma cruzi polyamine transport inhibitors by computational drug repurposing. Frontiers in Medicine. 2019;6:256
  222. 222. Valera-Vera EA, Sayé M, Reigada C, Miranda MR, Pereira CA. In silico repositioning of etidronate as a potential inhibitor of the Trypanosoma cruzi enolase. Journal of Molecular Graphics & Modelling. 2020;95:107506
  223. 223. Miranda MR, Sayé MM. Chagas disease treatment: From new therapeutic targets to drug discovery and repositioning. Current Medicinal Chemistry. 2019;26(36):6517-6518
  224. 224. Santos SS, de Araújo RV, Giarolla J, Seoud O El, Ferreira EI. Searching for drugs for Chagas disease, leishmaniasis and schistosomiasis: A review. International Journal of Antimicrobial Agents 2020;55(4):105906.
  225. 225. Adasme MF, Bolz SN, Adelmann L, Salentin S, Haupt VJ, Moreno-Rodríguez A, et al. Repositioned drugs for Chagas disease unveiled via structure-based drug repositioning. International Journal of Molecular Sciences. 2020;21(22):8809
  226. 226. Martínez-Flórez A, Galizzi M, Izquierdo L, Bustamante JM, Rodriguez A, Rodriguez F, et al. Repurposing bioenergetic modulators against protozoan parasites responsible for tropical diseases. International Journal for Parasitology: Drugs and Drug Resistance. 2020;14:17-27
  227. 227. Benaim G, Paniz-Mondolfi AE, Sordillo EM. The rationale for use of amiodarone and its derivatives for the treatment of Chagas’ disease and Leishmaniasis. Current Pharmaceutical Design. 2021;27(15):1825-1833
  228. 228. Zuma AA, de Souza W. Chagas disease chemotherapy: What do we know so far? Current Pharmaceutical Design. 2021;27(38):3963-3995
  229. 229. Torrico F, Gascón J, Barreira F, Blum B, Almeida IC, Alonso-Vega C, et al. New regimens of benznidazole monotherapy and in combination with fosravuconazole for treatment of Chagas disease (BENDITA): A phase 2, double-blind, randomised trial. The Lancet Infectious Diseases. 2021;21(8):1129-1140
  230. 230. Mazzeti AL, Gonçalves KR, Mota SLA, Pereira DE, Diniz L, Bahia MT. Combination therapy using nitro compounds improves the efficacy of experimental Chagas disease treatment. Parasitology. 2021;148(11):1320-1327
  231. 231. Mazzeti AL, Capelari-Oliveira P, Bahia MT, Mosqueira VCF. Review on experimental treatment strategies against Trypanosoma cruzi. Journal of Experimental Pharmacology. 2021;13:409-432
  232. 232. Peerzada MN, Gaur A, Azam A. Advances in drug discovery against neglected tropical diseases: Human African and American Trypanosomiasis. Current Medicinal Chemistry. 2021;28(36):7544-7582
  233. 233. Ferraz LRM, Silva LC, de Souza ML, Alves LP, de Sales V, Barbosa NG, et al. Drug associations as alternative and complementary therapy for neglected tropical diseases. Acta Tropica. 2022;225:106210
  234. 234. Araujo-Lima CF, Peres RB, Silva PB, Batista MM, Aiub CAF, Felzenszwalb I, et al. Repurposing strategy of atorvastatin against Trypanosoma cruzi: In vitro monotherapy and combined therapy with benznidazole exhibit synergistic trypanocidal activity. Antimicrobial Agents and Chemotherapy. 2018;62(9):e00979-18
  235. 235. Sass G, Madigan RT, Joubert L-M, Bozzi A, Sayed N, Wu JC, et al. A combination of itraconazole and amiodarone is highly effective against Trypanosoma cruzi infection of human stem cell-derived cardiomyocytes. The American Journal of Tropical Medicine and Hygiene. 2019;101(2):383-391
  236. 236. Simões-Silva MR, De Araújo JS, Oliveira GM, Demarque KC, Peres RB, D’Almeida-Melo I, et al. Drug repurposing strategy against Trypanosoma cruzi infection: In vitro and in vivo assessment of the activity of metronidazole in mono- and combined therapy. Biochemical Pharmacology. 2017;145:46-53
  237. 237. Sun W, Sanderson PE, Zheng W. Drug combination therapy increases successful drug repositioning. Drug Discovery Today. 2016;21(7):1189-1195
  238. 238. Pandey RP, Nascimento MS, Moore CE, Raj VS, Kalil J, Cunha-Neto E. New approaches for the treatment of Chagas disease. Current Drug Targets. 2021;22(7):835-841
  239. 239. Sousa A. Disulfiram: Its Use in Alcohol Dependence and Other Disorders. 1st ed. Singapore: Springer; 2019. 133 p
  240. 240. Suh JJ, Pettinati HM, Kampman KM, O’Brien CP. The status of disulfiram. Journal of Clinical Psychopharmacology. 2006;26(3):290-302
  241. 241. Bala V, Gupta G, Sharma V. Chemical and medicinal versatility of dithiocarbamates: An overview. Mini Reviews in Medicinal Chemistry. 2014;14(12):1021-1032
  242. 242. Shinde SD, Sakla AP, Shankaraiah N. An insight into medicinal attributes of dithiocarbamates: Bird’s eye view. Bioorganic Chemistry. 2020;105:104346
  243. 243. Gessner PK, Gessner T. Disulfiram and Its Metabolite, Diethyldithiocarbamate. Dordrecht: Springer Netherlands; 1992
  244. 244. Rath NC, Rasaputra KS, Liyanage R, Huff GR and Huff WE. Dithiocarbamate Toxicity - An Appraisal. In: Stoytcheva M, editor. Pesticides in the Modern World - Effects of Pesticides Exposure. London, UK: InTechOpen; 2011. pp. 323-340
  245. 245. Hald J, Jacobsen E. A drug sensitising the organism to ethyl alcohol. Lancet. 1948;252(6539):1001-1004
  246. 246. Lu C, Li X, Ren Y, Zhang X. Disulfiram: A novel repurposed drug for cancer therapy. Cancer Chemotherapy and Pharmacology. 2021;87(2):159-172
  247. 247. Jia Y, Huang T. Overview of Antabuse® (Disulfiram) in radiation and cancer biology. Cancer Management and Research. 2021;13:4095-4101
  248. 248. Meraz-Torres F, Plöger S, Garbe C, Niessner H, Sinnberg T. Disulfiram as a therapeutic agent for metastatic malignant melanoma—Old myth or new logos? Cancers (Basel). 2020;12(12):3538
  249. 249. Jiao Y, Hannafon BN, Ding W-Q. Disulfiram’s anticancer activity: Evidence and mechanisms. Anti-Cancer Agents in Medicinal Chemistry. 2016;16(11):1378-1384
  250. 250. Spivak AM, Planelles V. Novel latency reversal agents for HIV-1 cure. Annual Review of Medicine. 2018;69(1):421-436
  251. 251. de Moura FB, Kohut SJ, Bergman J. Limited modulation of the abuse-related behavioral effects of d-methamphetamine by disulfiram. Experimental and Clinical Psychopharmacology. 2018;26(5):497-502
  252. 252. Reinhardt S, Stoye N, Luderer M, Kiefer F, Schmitt U, Lieb K, et al. Identification of disulfiram as a secretase-modulating compound with beneficial effects on Alzheimer’s disease hallmarks. Scientific Reports. 2018;8(1):1329
  253. 253. Gao J, Gong Z, Montesano D, Glazer E, Liegner K. “Repurposing” disulfiram in the treatment of lyme disease and babesiosis: Retrospective review of first 3 years’ experience in one medical practice. Antibiotics. 2020;9(12):868
  254. 254. Zitko J, Doležal M. Old drugs and new targets as an outlook for the treatment of tuberculosis. Current Medicinal Chemistry. 2019;25(38):5142-5167
  255. 255. Das S, Garg T, Chopra S, Dasgupta A. Repurposing disulfiram to target infections caused by non-tuberculous mycobacteria. The Journal of Antimicrobial Chemotherapy. 2019;74(5):1317-1322
  256. 256. Castillo-Villanueva A, Rufino-González Y, Méndez S-T, Torres-Arroyo A, Ponce-Macotela M, Martínez-Gordillo MN, et al. Disulfiram as a novel inactivator of Giardia lamblia triosephosphate isomerase with antigiardial potential. International Journal for Parasitology: Drugs and Drug Resistance. 2017;7(3):425-432
  257. 257. Shirley D-A, Sharma I, Warren CA, Moonah S. Drug repurposing of the alcohol abuse medication disulfiram as an anti-parasitic agent. Frontiers in Cellular and Infection Microbiology. 2021;11:633194
  258. 258. Omran Z, Sheikh R, Baothman OA, Zamzami MA, Alarjah M. Repurposing disulfiram as an anti-obesity drug: Treating and preventing obesity in high-fat-fed rats. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy. 2020;13:1473-1480
  259. 259. Kannappan V, Ali M, Small B, Rajendran G, Elzhenni S, Taj H, et al. Recent advances in repurposing disulfiram and disulfiram derivatives as copper-dependent anticancer agents. Frontiers in Molecular Biosciences. 2021;8:741316
  260. 260. Fong W, To KK. Drug repurposing to overcome resistance to various therapies for colorectal cancer. Cellular and Molecular Life Sciences. 2019;76(17):3383-3406
  261. 261. Ekinci E, Rohondia S, Khan R, Dou QP. Repurposing disulfiram as an anti-cancer agent: Updated review on literature and patents. Recent Patents on Anti-Cancer Drug Discovery. 2019;14(2):113-132
  262. 262. Horita Y, Takii T, Yagi T, Ogawa K, Fujiwara N, Inagaki E, et al. Antitubercular activity of disulfiram, an antialcoholism drug, against multidrug- and extensively drug-resistant mycobacterium tuberculosis isolates. Antimicrobial Agents and Chemotherapy. 2012;56(8):4140-4145
  263. 263. Long TE. Repurposing thiram and disulfiram as antibacterial agents for multidrug-resistant Staphylococcus aureus infections. Antimicrobial Agents and Chemotherapy. 2017;61(9):e00898-17
  264. 264. Sauna ZE, Shukla S, Ambudkar SV. Disulfiram, an old drug with new potential therapeutic uses for human cancers and fungal infections. Molecular BioSystems. 2005;1(2):127
  265. 265. Argüello-García R, Leitsch D, Skinner-Adams T, Ortega-Pierres MG. Drug resistance in Giardia: Mechanisms and alternative treatments for Giardiasis. Advances in Parasitology. 2020;107:201-282
  266. 266. Cvek B. Nonprofit drugs as the salvation of the world’s healthcare systems: The case of Antabuse (disulfiram). Drug Discovery Today. 2012;17(9-10):409-412
  267. 267. Jørgensen CH, Pedersen B, Tønnesen H. The efficacy of disulfiram for the treatment of alcohol use disorder. Alcoholism, Clinical and Experimental Research. 2011;35(10):1749-1758
  268. 268. Sinclair JMA, Chambers SE, Shiles CJ, Baldwin DS. Safety and tolerability of pharmacological treatment of alcohol dependence: Comprehensive review of evidence. Drug Safety. 2016;39(7):627-645
  269. 269. Reisinger EC, Kern P, Dietrich M, Ernst M, Flad HD, Bock P, et al. Inhibition of HIV progression by dithiocarb. Lancet. 1990;335(8691):679-682
  270. 270. Pompidou A, Delsaux MC, Telvi L, Mace B, Coutance F, Falkenrodt A, et al. Isoprinosine and Imuthiol, two potentially active compounds in patients with AIDS-related complex symptoms. Cancer Research. 1985;45(9):4671-4673
  271. 271. Lemarie E, Musset M, Charbonnier C, Renoux M, Renoux G. Clinical characterization of imuthiol. Methods and Findings in Experimental and Clinical Pharmacology. 1986;8(1):51-54
  272. 272. Khouri R, Novais F, Santana G, de Oliveira CI, Vannier dos Santos MA, Barral A, et al. DETC induces leishmania parasite killing in human in vitro and murine in vivo models: A promising therapeutic alternative in leishmaniasis. PLoS One. 2010;5(12):e14394
  273. 273. Assolini JP, Tomiotto-Pellissier F, da Silva Bortoleti BT, Gonçalves MD, Sahd CS, Carloto ACM, et al. Diethyldithiocarbamate encapsulation reduces toxicity and promotes leishmanicidal effect through apoptosis-like mechanism in promastigote and ROS production by macrophage. Journal of Drug Targeting. 2020;28(10):1110-1123
  274. 274. Mazur KL, Feuser PE, Valério A, Poester Cordeiro A, de Oliveira CI, Assolini JP, et al. Diethyldithiocarbamate loaded in beeswax-copaiba oil nanoparticles obtained by solventless double emulsion technique promote promastigote death in vitro. Colloids and Surfaces. B, Biointerfaces. 2019;176:507-512
  275. 275. Celes FS, Trovatti E, Khouri R, Van Weyenbergh J, Ribeiro SJL, Borges VM, et al. DETC-based bacterial cellulose bio-curatives for topical treatment of cutaneous leishmaniasis. Scientific Reports. 2016;6(1):38330
  276. 276. McMahon A, Chen W, Li F. Old wine in new bottles: Advanced drug delivery systems for disulfiram-based cancer therapy. Journal of Controlled Release. 2020;319:352-359
  277. 277. Kyle ME, Serroni A, Farber JL. The inhibition of lipid peroxidation by disulfiram prevents the killing of cultured hepatocytes by allyl alcohol, tert-butyl hydroperoxide, hydrogen peroxide and diethyl maleate. Chemico-Biological Interactions. 1989;72(3):269-275
  278. 278. Liu T, Wang P, Cong M, Zhao X, Zhang D, Xu H, et al. Diethyldithiocarbamate, an anti-abuse drug, alleviates steatohepatitis and fibrosis in rodents through modulating lipid metabolism and oxidative stress. British Journal of Pharmacology. 2018;175(24):4480-4495
  279. 279. Sundar S, Chakravarty J, Meena LP. Leishmaniasis: Treatment, drug resistance and emerging therapies. Expert Opinion on Orphan Drugs. 2019;7(1):1-10
  280. 280. Calle LM, Sullivan PD. Screening of antioxidants and other compounds for antimutagenic properties towards benzo[a]pyrene-induced mutagenicity in strain TA98 of Salmonella typhimurium. Mutation Research. 1982;101(2):99-114
  281. 281. Lane JE, Ribeiro-Rodrigues R, Suarez CC, Bogitsh BJ, Jones MM, Singh PK, et al. In vitro trypanocidal activity of tetraethylthiuram disulfide and sodium diethylamine-N-carbodithioate on Trypanosoma cruzi. The American Journal of Tropical Medicine and Hygiene. 1996;55(3):263-266
  282. 282. de Freitas Oliveira JW, Torres TM, Moreno CJG, Amorim-Carmo B, Damasceno IZ, Soares AKMC, et al. Insights of antiparasitic activity of sodium diethyldithiocarbamate against different strains of Trypanosoma cruzi. Scientific Reports. 2021;11(1):11200
  283. 283. Giulivi C, Turrens JF, Boveris A. Chemiluminescence enhancement by trypanocidal drugs and by inhibitors of antioxidant enzymes in Trypanosoma cruzi. Molecular and Biochemical Parasitology. 1988;30(3):243-251
  284. 284. Koto KS, Lescault P, Brard L, Kim K, Singh RK, Bond J, et al. Antitumor activity of nifurtimox is enhanced with tetrathiomolybdate in medulloblastoma. International Journal of Oncology. 2011;38(5):1329-1341
  285. 285. Prentice H, Modi JP, Wu J-Y. Mechanisms of neuronal protection against excitotoxicity, endoplasmic reticulum stress, and mitochondrial dysfunction in stroke and neurodegenerative diseases. Oxidative Medicine and Cellular Longevity. 2015;2015:1-7
  286. 286. Masuda Y, Nakayama N. Protective action of diethyldithiocarbamate and carbon disulfide against renal injury induced by chloroform in mice. Biochemical Pharmacology. 1983;32(21):3127-3135
  287. 287. Gandhi NM, Gopalaswamy UV, Nair CKK. Radiation protection by disulfiram: Protection of membrane and DNA in vitro and in vivo against γ-radiation. Journal of Radiation Research. 2003;44(3):255-259
  288. 288. Gandhi NM, Nair CKK. Radiation protection by diethyldithiocarbamate: Protection of membrane and DNA in vitro and in vivo against γ-radiation. Journal of Radiation Research. 2004;45(2):175-180
  289. 289. Wysor MS, Zwelling LA, Sanders JE, Grenan MM. Cure of mice infected with Trypanosoma rhodesiense by cis-diamminedichloroplatinum (ii) and disulfiram rescue. Science 80. 1982;217(4558):454-456
  290. 290. Ishikawa M, Aoki T, Yomogida S, Takayanagi Y, Sasaki K. Drug interaction effects on antitumour drugs (XV): Disulfiram as protective agent against cyclophosphamide-induced urotoxicity without compromising antitumour activity in mice. Pharmacology & Toxicology. 1994;74(6):255-261
  291. 291. Ishikawa M, Takayanagi Y, Sasaki K. Inhibition of ifosfamide-induced urotoxicity by disulfiram in mice. Japanese Journal of Pharmacology. 1989;49(1):147-150
  292. 292. Brambilla G, Carlo P, Finollo R. Effect of ten thiocompounds on rat liver DNA damage induced by a small dose of N-nitrosodimethylamine. Archives of Toxicology. 1992;66(4):286-290
  293. 293. Burman WJ, Terra M, Breese P, Cohn D, Reves R. Lack of toxicity from concomitant directly observed disulfiram and isoniazid-containing therapy for active tuberculosis. The International Journal of Tuberculosis and Lung Disease. 2002;6(9):839-842
  294. 294. Traiger GJ, Vyas KP, Hanzlik RP. Effect of thiocarbonyl compounds on α-naphthylisothiocyanate-induced hepatotoxicity and the urinary excretion of [35S]α-naphthylisothiocyanate in the rat. Toxicology and Applied Pharmacology. 1984;72(3):504-512
  295. 295. Hazai E, Vereczkey L, Monostory K. Reduction of toxic metabolite formation of acetaminophen. Biochemical and Biophysical Research Communications. 2002;291(4):1089-1094
  296. 296. Kim HL, Jones LP. Protective effects of butylated hydroxyanisole, ethoxyquin, and disulfiram on acute pyrrolizidine alkaloids poisoning in mice. Research Communications in Chemical Pathology and Pharmacology. 1982;36(2):341-344
  297. 297. Masukawa T, Nakanishi K. Protection by disulfiram and diethyldithiocarbamate against hypoxia-induced lethality in mice. Japanese Journal of Pharmacology. 1993;63(3):279-284
  298. 298. Jennische E, Hansson H. Disulfiram is protective against postischemic cell death in the liver. Acta Physiologica Scandinavica. 1984;122(2):199-201
  299. 299. Llobet JM, Domingo JL, Paternain JL, Corbella J. Treatment of acute lead intoxication. A quantitative comparison of a number of chelating agents. Archives of Environmental Contamination and Toxicology. 1990;19(2):185-189
  300. 300. Hiroshige Ono Takayuki Funakoshi H. Comparative effects of disulfiram and diethyldithiocarbamate against testicular toxicity in rats caused by acute exposure to cadmium. Journal of Toxicology and Environmental Health. 1997;50(4):389-400
  301. 301. Pages A, Casaş JS, Sanchez A, Sordo J, Bravo J, Gayoso M. Dithiocarbamates in heavy metal poisoning: Complexes of N,N-di(2-hidroxyethyl)dithiocarbamate with Zn(II), Cd(II), Hg(II), CH3Hg(II), and C6H5Hg(II). Journal of Inorganic Biochemistry. 1985;25(1):35-42
  302. 302. Cohen G, Heikkila RE, Allis B, Cabbat F, Dembiec D, MacNamee D, et al. Destruction of sympathetic nerve terminals by 6-hydroxydopamine: Protection by 1-phenyl-3-(2-thiazolyl)-2-thiourea, diethyldithiocarbamate, methimazole, cysteamine, ethanol and n-butanol. The Journal of Pharmacology and Experimental Therapeutics. 1976;199(2):336-352
  303. 303. Sonawane VK, Mahajan UB, Shinde SD, Chatterjee S, Chaudhari SS, Bhangale HA, et al. A chemosensitizer drug: Disulfiram prevents doxorubicin-induced cardiac dysfunction and oxidative stress in rats. Cardiovascular Toxicology. 2018;18(5):459-470
  304. 304. Kis A, Yellon DM, Baxter GF. Role of nuclear factor-κB activation in acute ischaemia-reperfusion injury in myocardium. British Journal of Pharmacology. 2003;138(5):894-900
  305. 305. McPherson BC, Yao Z. Signal transduction of opioid-induced cardioprotection in ischemia-reperfusion. Anesthesiology. 2001;94(6):1082-1088
  306. 306. Wei S, Xiao Z, Huang J, Peng Z, Zhang B, Li W. Disulfiram inhibits oxidative stress and NLRP3 inflammasome activation to prevent LPS-induced cardiac injury. International Immunopharmacology. 2022;105:108545
  307. 307. Köksoy FN, Köse H, Soybír GR, Yalçin O, Cokneşelí B. The prophylactic effects of superoxide dismutase, catalase, desferrioxamine, verapamil and disulfiram in experimental colitis. Journal of the Royal College of Surgeons of Edinburgh. 1997;42(1):27-30
  308. 308. Campos MCO, Castro-Pinto DB, Ribeiro GA, Berredo-Pinho MM, Gomes LHF, da Silva Bellieny MS, et al. P-glycoprotein efflux pump plays an important role in Trypanosoma cruzi drug resistance. Parasitology Research. 2013;112(6):2341-2351
  309. 309. Loo TW, Bartlett MC, Clarke DM. Disulfiram metabolites permanently inactivate the human multidrug resistance P-glycoprotein. Molecular Pharmaceutics. 2004;1(6):426-433
  310. 310. Sauna ZE, Peng X-H, Nandigama K, Tekle S, Ambudkar SV. The molecular basis of the action of disulfiram as a modulator of the multidrug resistance-linked ATP binding cassette transporters MDR1 (ABCB1) and MRP1 (ABCC1). Molecular Pharmacology. 2004;65(3):675-684
  311. 311. da Costa KM, Valente RC, Salustiano EJ, Gentile LB, Freire-de-Lima L, Mendonça-Previato L, et al. Functional characterization of ABCC proteins from Trypanosoma cruzi and their involvement with thiol transport. Frontiers in Microbiology. 2018;9:205
  312. 312. Jin L, Davis MR, Hu P, Baillie TA. Identification of novel glutathione conjugates of disulfiram and diethyldithiocarbamate in rat bile by liquid chromatography-tandem mass spectrometry. Evidence for metabolic activation of disulfiram in vivo. Chemical Research in Toxicology. 1994;7(4):526-533
  313. 313. Giustarini D, Galvagni F, Tesei A, Farolfi A, Zanoni M, Pignatta S, et al. Glutathione, glutathione disulfide, and S-glutathionylated proteins in cell cultures. Free Radical Biology & Medicine. 2015;89:972-981
  314. 314. Arnelle DR, Day BJ, Stamler JS. Diethyl dithiocarbamate-induced decomposition of S-Nitrosothiols. Nitric Oxide. 1997;1(1):56-64
  315. 315. Johnston CD. The in vitro reaction between tetraethylthiuram disulfide (antabuse) and glutathione. Archives of Biochemistry and Biophysics. 1953;44(1):249-251
  316. 316. Goldstein BD, Rozen MG, Quintavalla JC, Amoruso MA. Decrease in mouse lung and liver glutathione peroxidase activity and potentiation of the lethal effects of ozone and paraquat by the superoxide dismutase inhibitor diethyldithio-carbamate. Biochemical Pharmacology. 1979;28(1):27-30
  317. 317. Kona R, Buac D, Burger M. Disulfiram, and disulfiram derivatives as novel potential anticancer drugs targeting the ubiquitin proteasome system in both preclinical and clinical studies. Current Cancer Drug Targets. 2011;11(3):338-346
  318. 318. Gupta I, Aggarwal S, Singh K, Yadav A, Khan S. Ubiquitin proteasome pathway proteins as potential drug targets in parasite Trypanosoma cruzi. Scientific Reports. 2018;8(1):8399
  319. 319. Banerjee ER. Perspectives in Translational Research in Life Sciences and Biomedicine. Springer Singapore: Singapore; 2016
  320. 320. Saraiva RM, Portela LF, Silveira GPE da, da Gomes NL, Pinto DP, de Silva AC, et al. Disulfiram repurposing in the combined chemotherapy of Chagas disease. Medicine: Case Reports and Study Protocols. 2021;2(7):e0110

Notes

  • “Since that which is compounded out of something so that the whole is one, not like a heap (…), then, is something-not only its elements (…) but also something else (…)” ‘Metaphysics’ Book VII by Aristotle, Translated by W. D. Ross, often misquoted or mistranslated.

Written By

Marcos André Vannier-Santos, Ana Márcia Suarez-Fontes, Juliana Almeida-Silva, Alessandra Lifsitch Viçosa, Sandra Aurora Chavez Perez, Alejandro Marcel Hasslocher-Moreno, Gabriel Parreiras Estolano da Silveira, Luciana Fernandes Portela and Roberto Magalhães Saraiva

Submitted: 14 February 2022 Reviewed: 03 March 2022 Published: 24 June 2022