Open access peer-reviewed chapter

Efficacy and Safety of Chagas Disease Drug Therapy and Treatment Perspectives

Written By

Wilton H. Kawaguchi, Leticia Bonancio Cerqueira, Mariana Millan Fachi, Michel L. Campos, Iara J. Messias Reason and Roberto Pontarolo

Submitted: 13 June 2017 Reviewed: 05 February 2018 Published: 13 March 2018

DOI: 10.5772/intechopen.74845

From the Edited Volume

Chagas Disease - Basic Investigations and Challenges

Edited by Veeranoot Nissapatorn and Helieh S. Oz

Chapter metrics overview

1,216 Chapter Downloads

View Full Metrics

Abstract

Chagas disease, also known as American trypanosomiasis, is a neglected disease caused by the protozoan parasite Trypanosoma cruzi. The disease affects about 6–7 million people worldwide, mostly in Latin America. Although Chagas disease was discovered more than 100 years ago, and the first treatments over 40, only 2 drugs were used to treat this pathology, it is still considered one of the neglected diseases. In this chapter, the subjects related to conventional etiological therapies, benznidazole and nifurtimox, such as the drug, the mechanism of action, the therapy schedule for treatment, efficacy and safety and their adverse effects will be discussed. Additionally, it will address alternative therapies of comorbidities related to the progression of Chagas’ disease in patients with chronic disease, such as heart disease and dysfunction of the digestive system. Finally, novel pharmacological strategies and their related compounds will be reviewed accounting for their progression in pharmacological studies and their success rate.

Keywords

  • Chagas disease
  • benznidazole
  • nifurtimox
  • symptomatic treatment
  • new strategies

1. Introduction

Chagas disease or American trypanosomiasis is caused by the protozoan parasite Trypanosoma cruzi (T. cruzi). Endemic largely in Latin American countries, it is transmitted primarily by vectors, the insect vector triatomine, also known as “kissing bug”. It is estimated that 6–7 million people are infected worldwide, and that more than 10,000 people die each year as a result of the disease, with the highest number of cases in Latin American countries [1].

Although Chagas disease has been discovered more than 100 years ago, there are currently only two drugs used to control Chagas disease (CD), benznidazole and nifurtimox [2, 3, 4]. Benznidazole has a similar efficacy profile to nifurtimox. They have high cure rates in the acute phase and at the beginning of the chronic phase, being little or ineffective in the late chronic phase [5]. Benznidazole and nifurtimox belong to the nitro heterocycles class of drugs; however, each presents different mechanisms of action that are considered very aggressive and may cause several adverse effects.

In the chronic phase of CD, the treatment at this stage aims to reduce parasitemia and prevent complications that may lead to progression of visceral lesions. However, the administration of benznidazole and nifurtimox remains controversial [5, 6]. To treat the cardiac and gastrointestinal manifestations attributed to this phase of the disease, it requires specific pharmacological approaches, highlighting the combination of diuretics, angiotensin-converting-enzyme inhibitors or angiotensin receptor blockers, and adrenergic beta blockers. The choice of palliative treatment is in relation to the symptoms of the patient.

Currently, the main treatment for CD is benznidazole and nifurtimox and both compounds are effective in the acute phase to reduce parasitemia and the persistence and the clinical severity of the disease [5]. However, in most countries, benznidazole is the primary choice to begin the treatment, since it has shown less adverse events. The predominant reasons to use nifurtimox are the occurrence of benznidazole resistance or its unavailability [6, 7]. Benznidazole can achieve over 70% of cure in cases of congenital and acute phase, while nifurtimox reaches 80% efficacy. However, these compounds have limited efficacy in the chronic phase, with efficacy rates of only 6–10% [8, 9, 10]. Treatment complications include, need for increasing doses, duration varying according to the stage of infection, and high occurrence of adverse effects caused by the long-term treatment with high doses. In addition, resistance may occur depending on the Trypanosoma. cruzi (T. cruzi) strain [10].

Although the aforementioned therapy is over 40 years old, there are limitations in this first line of treatment for CD. Several studies aimed to bring new options to its arsenal, mainly for the chronic phase. Among them, are drugs used to treat other conditions, such as posaconazole, and new compounds, such as E1224. Each of these has been tested in clinical trials. Other strategies have been suggested as groundbreaking approaches to eradicate, or at least control, CD. These include drugs, such as cruzipain and trypanothione reductase which specifically targets the ergosterol biosynthesis pathway.

Considering this the classical treatment of CD, with a focus on the evaluation of efficacy and safety of the current drug therapy, will be properly addressed in this chapter. In addition, a review of new pharmacological therapies will be discussed.

Advertisement

2. Etiological treatment: benznidazole

2.1. Treatment history

Until the publication of the Manual de Doenças Tropicaes e Infectuosas in 1935 by Carlos Chagas and Evandro Chagas (Manual of Tropical and Infectious Diseases), there was no pharmacological treatment available for trypanosomiasis. Drugs with trypanocidal activity have been investigated by a number of researchers; however, without success [11]. In 1936, quinolinic compounds were successfully used to treat an acute case of trypanosomiasis [12]. In the following years, nitrofurazone was administered to treat trypanosomiasis in mice and achieve efficacy rates between 20 and 100%, depending on the therapeutic schedule [13]. These results motivated the experimental trials in humans, in which nitrofurazone demonstrated to be effective against trypanosomes in the circulating blood, cerebrospinal fluids [14] as well as other promising clinical results [15, 16].

Novel studies were performed to identify an alternative therapeutic schedule and main side effects [17, 18]. In 1962, Rio de Janeiro was the host city for the “Meeting of Debates on Chagas Disease”, where the standardization of the methodology and establishment of criteria for the evaluation of attempted pharmacological treatments was discussed. Nifurtimox and benznidazole remain as the only two drugs available for treatment of CD [19].

2.2. The drug

Benznidazole (BNZ) N-benzyl-2-(2-nitro-imidazol-1-yl)acetamide (Figure 1) is a nitro heterocyclic drug. It was nitroimidazolic derivative synthesized in the early 1970s by Wineholt and Liebman and produced by Hoffman-La Roche, Switzerland. In 2003, the rights and manufacturing technology of BNZ was granted to the Laboratório Farmacêutico do Estado de Pernambuco (LAFEPE) – Recife/PE – Brazil [5, 9, 10, 20, 21]. According to the Biopharmaceutical Classification System, BNZ is categorized as class 4, thus exhibiting limited and/or variable absorption due to the characteristics such as low solubility and low permeability. Therefore, there are interests in strategies to improve the absorption rate, hence increasing its bioavailability [22, 23].

Figure 1.

Chemical structure of benznidazole.

BNZ is administered orally in a tablet form of 100 mg for adults and 12.5 mg tablets for children [24]. The drug is fully absorbed in the gastrointestinal tract and the plasma peak concentration was achieved within 2–4 hours. The half-life is 12 hours and its metabolites were eliminated in urine and feces [5, 10, 20].

2.3. Mechanism of action

The mechanism of action of BNZ is not fully understood [9, 25]. However, some reports associate its action with the formation of free radicals and/or electrophilic metabolites. In the nitroimidazole derivatives, the reduction of the nitro group (NO2) in the amino group (NH2) occurs by the action of nitroreductase present in protozoan cells or bacteria. This reaction leads to the formation of radical intermediates, as well as electrophilic metabolites. The process is initiated by the action of the enzyme NADPH cytochrome P450 reductase, which acts on the nitro group (R-NO2) of the molecule nitroimidazole, inducing the production of an intermediate nitro radical (R-NO2) and the formation of hydroxylamine (R-NHOH). These intermediates act on the covalent modification of macromolecules, such as DNA, causing fragmentation in the chain and destabilization of the helix, inhibiting DNA synthesis leading to cell death of protozoa parasitic and/or bacterial. In addition to this alteration in DNA, it involves the modification of other macromolecules, such as lipids and proteins, affecting T. cruzi metabolism. An additional mechanism of action of BNZ is the increase in phagocytosis, leading to lysis and inhibition of T. cruzi growth through an interferon-gamma (IFN-γ)-dependent mechanism and through the enzyme NADH-fumarate reductase, respectively [9, 11, 26, 27, 28].

Concomitantly, the electrophilic metabolites mentioned above due to their high reactivity and low specificity, may present human host action and cytotoxic effects observed during the treatment of patients [27, 28, 29].

2.4. Therapeutic schedule

The dosage of BNZ in the acute phase of the disease should be provided in two or three administrations daily, during the 60-day period. The dose is established according to the patient’s age and weight. For adults, it is recommended 5 mg/kg/day, whereas for children weighing less than 40 kg, 5–10 mg/kg/day is recommended. Furthermore for children, the therapeutic regimen should be as small as possible to achieve therapeutic adherence. A recommendation for infants is 10 mg/kg/day. In congenital infections, the treatment is indicated for children born to mothers who were serologically positive for CD with the presence of T. cruzi in umbilical cord blood, serum-specific IgM soon after birth, and IgG after 6 months. In the newborn, the recommended etiologic therapy is the administration of BNZ twice a day for 30 days, it is suggested to be provided at 5 mg/kg/day [5, 11, 20, 30, 31, 32].

In the chronic phase, the dosage is 5–7 mg/kg/day divided into two administrations with an interval of 12-hour between doses, and for 30–60 consecutive days [5, 20].

2.5. Efficacy and safety

It is considered as the first choice treatment because it exhibits a better safety and efficacy profile when compared to nifurtimox. Moreover, there is evidence of greater efficacy [7, 28]. It has antiprotozoal and antibacterial performance, acting against the trypomastigote and amastigote forms [5].

In the acute phase, if the treatment is initiated immediately following the confirmation of the presence of T. cruzi in the direct examination of the peripheral blood, the cure percentages are greater than 70–80%, independent of the transmission route [5, 7, 9, 33]. It should be administered to patients in the early chronic phase, children less than 12 years, laboratory accidents and congenital infection. In newborns treated during the first year of life, the chances of cure are greater than 90%; otherwise, in the late chronic phase the patient has lower chances of cure (~10%) [20, 34, 35, 36].

Two placebo-controlled clinical trials were performed in the USA in children between 6 and 12 years old for the evaluation of treatment using BNZ, in which the results were satisfactory. In the first trial, 60% of the children treated had a change from positive to a negative in serology for CD, compared to 14% of them receiving placebo. In the second trial, similar results for children treated with BNZ showed a 55% change from a positive to a negative antibody test, compared to 5% of those receiving placebo [37].

For treatment in cases of organ transplantation, it is essential to know if the donor or recipient presents positive serology and can transmit or reactivate the infection. In cases of absolute requirement for transplantation, the serum-reactive donor in which the recipient is serologically negative for a chagasic infection, the donor should receive BNZ therapy following the traditional dosing regimen, within 60 days prior to transplantation. For the recipient following transplantation, it is recommended to initiate therapy by performing serological tests over time. There is a serum conversion with the treatment for the acute phase. If an acute infection is detected, the treatment need to be started [38, 39, 40].

Immunocompromised patients, carriers of hematological malignancies, users of immunosuppressive drugs, or those co-infected with the acquired immunodeficiency virus may reactivate CD. The treatment is the conventional treatment, lasting 60 days, and depending on the clinical conditions of the patient, it may be increased to 90 days [40, 41, 42].

When accidental infection occurs, the individual who accidentally had contact with contaminated materials; needle puncture, contact in lesions, wounds or mucous membranes, or any other means that indicates the possibility of having been infected by the parasite, is evaluated by serological tests and treatment begins immediately. During the 10–15 days of the treatment period, the serological tests will be repeated [5, 11].

At the beginning of the chronic phase in children, treatment follows the same reasoning for acute phase cases in children younger than or equal to 12 years with positive serology. For adults, therapy is recommended to prevent or reduce the progression of CD in more severe forms, such as cardiomyopathy, and to prevent congenital transmission in pregnant women [5, 11, 20, 43]. Its use in the chronic phase has generated vast discussions. The BENEFIT project – Benznidazole Evaluation for Interrupting Trypanosomiasis, is an international multicenter, randomized, double-blinded, placebo-controlled trial of BNZ for the treatment of patients with mild to moderate Chagas cardiomyopathy. This study has conducted and produced results that indicated that the use of BNZ in the chronic phase significantly reduced the detection of circulating parasites; however, did not attenuate the cardiac clinical progression [20, 33].

Treatment for patients older than 50 years should take into account the risk of toxicity of the drug against the benefits of the therapy individually [6, 20].

2.6. Adverse effects

Therapy with BNZ contains some challenges, such as the large doses administered, time duration varying according to the stage of infection, and high occurrence of adverse effects. These challenges are less frequent when compared with nifurtimox (Table 1).

Adverse effectsBenznidazoleNifurtimox
Anorexia and weight loss5–40%50–75%
Nausea0–5%15–50%
Vomiting0–5%15–26%
Peripheral neuropathy0–30%2–5%
LeukopeniaRare: <1%Rare: <1%

Table 1.

Frequencies of adverse effects associated with benznidazole and nifurtimox.

The adverse effects in patients receiving BNZ may be classified into three groups: manifestations of hypersensitivity (dermatitis with rashes, generalized or peritoneal edema, fever, lymphadenopathy, muscle and joint pain), bone marrow depression (agranulocytosis, neutropenia, and thrombocytopenic purpura), and peripheral polyneuropathy (paresthesias and polyneuritis) [7, 11].

Adverse effects of dermatological cause appear in approximately 30% of the patients being treated with the drug. Rashes occur due to photosensitization, dermatitis is usually mild to moderate and may be treated with topical systemic corticosteroids. However, treatment should be discontinued immediately in cases of severe or exfoliative dermatitis or associated with fever and adenopathy. Bone marrow suppression is rare, but if occurs the treatment should be discontinued immediately. Additional adverse effects include weight loss, nausea and/or vomiting, anorexia, and insomnia [8, 11, 20, 29, 43].

Treatment is contraindicated in pregnancy and in patients with severe renal or hepatic impairment [11, 20, 44].

According to the World Health Organization (WHO), the ideal requirements for treatment are: parasitological cure in both phases (acute and chronic), effective doses in single or few doses, low cost, no side effects or teratogenic effects, without hospitalization, and induction of resistance. Until now, there is no drug for the treatment of CD that meets each of these WHO requirements.

Advertisement

3. Etiological treatment: nifurtimox

3.1. The drug

Nifurtimox (NFX) (Figure 2) belongs to the class of nitrofuran compounds. Her-Linger, Mayer, Petersen and Bock from Bayer™ synthesized it in 1962 in Germany. This was the first drug designed to treat trypanosomiasis, such as sleeping sickness and Chagas diseases [34]. Its production was interrupted in 1980s due to the reduction on world demand [11]. In 2009, the WHO Expert Committee on the Selection and Use of Essential Medicines recommends the inclusion of NFX in the model list of essential medicines (EML) and Bayer resumed the production of NFX [45]. Bayer, through WHO, still provides NFX worldwide in 120 mg and 30 mg tablets under the brand name Lampit [46], in United States and it is redistributed by Center for Disease Control and Prevention (CDC) [47].

Figure 2.

Chemical structure of nifurtimox.

NFX is a 5-nitrofuran (3-methyl-4-(5′-nitrofurfurylideneamine) tetrahydro-4H-1,4-tiazine-1,1-dioxide. NFX possesses an asymmetric center, but it is used as a racemic mixture since pure stereoisomers were not more active or less toxic.

NFX is well absorbed following oral administration. The plasma levels range from 10 to 20 mM and lower concentrations are found in urine and tissues. The therapeutic schedule must vary according to the patient’s age and disease phase [9, 11, 48, 49].

3.2. Mechanism of action

The mechanism of action of NFX is based on cellular damage originating from the production of nitro anion radical through two pathways: through a redox cycle with the formation of O∙ (superoxide anion) and its reduction to the corresponding amine derivative [50, 51, 52].

This mechanism was thoroughly studied by Do Campo and colleagues [51, 53, 54, 55, 56, 57, 58]. The reduction of a nitro group by nitroredutase (NTR) is fundamental for NFX mechanism of action. This enzyme catalyzes the reduction of 2-electrons of the compound, producing a nitrous intermediate, followed by a second 2-electron reduction to generate a hydroxylamine. This derivative can directly lead to cell damage, or generate other cytotoxic agents (Figure 3) [52, 59]. Since the T. cruzi detoxification mechanism is insufficient, it becomes more susceptive to reactive nitro compounds [60, 61, 62]. The presence of the nitro reductase type I in the parasite is the main responsibility for NFX selectivity. This enzyme is absent in mammals, reducing the formation of toxic agents in human.

Figure 3.

Mechanism of NFX based on nitroreductase type 1 action.

3.3. Therapeutic schedule

Several therapeutic schedules were evaluated in the past years. The doses vary between 5 and 30 mg/kg/day, in extended therapies of 30–120 days [63, 64, 65, 66, 67]. Based on these experiments and considering the efficacy/tolerance ratio, the ideal therapeutic schedule of NFX is 8–10 mg/kg/day in adults and 15 mg/kg/day in children, for 60, 90 or 120 days, divided into three daily doses, after meals [1, 68]. It must be highlighted that even at these doses, side effects may potentially be present in adults.

3.4. Safety and efficacy

Many studies regarding the efficacy of NFX were performed between the 1960 and 1980s, at the beginning of its development and first years of commercialization [5, 11, 34]. Considering these studies, the activity of the NFX against the Trypanosoma sp. is expressive. NFX is capable to achieve efficacy rates of 70–100% in acute parasitemia cases. However, these rates substantially decrease when it comes to adult chronic phase, reaching values of only 7–8% in adults, although in children under 14, it remains around 86% [68, 69].

The primary reason why BNZ is often preferred compared to NFX is due to the presence of adverse effects. NFX frequently produces side effects [5, 11, 34, 70], but the majority are mild and can be managed with dose reduction or temporary suspension of medication [11, 68, 71].

3.5. Adverse effects

The treatment of CD is long and continuous and the presence of adverse effects becomes quite common between 60 and 100% of the patients [68]. The most frequent side effects described in the case of NFX are anorexia, nausea, headache, and amnesia. The possible neurological reactions are restlessness, disorientation, forgetfulness, insomnia, spasms, paresthesias, polyneuritis, and convulsive seizures. Most of the adverse effects (93%) are mild and disappear through dose reduction or after suspending the treatment [68, 70].

Advertisement

4. Symptomatic treatment of chronic phase

CD is classified evolutionary into two phases: an initial acute phase, followed by a chronic phase. Each phase has distinct clinical characteristics, diagnostic parameters, and treatment [72, 73, 74, 75, 76]. The acute phase is characterized by intense parasitism related with nonspecific symptoms, including fever, eyelid edema (denominated Chagoma), edema, and myocarditis. In general, the progression of disease can take years to reach the chronic phase, which presents four clinical forms: indeterminate (without clinical manifestations), cardiac, digestive, and mixed (association of cardiac and digestive) [77, 78, 79, 80, 81].

4.1. Chronic Chagas cardiomyopathy (CCC)

Chronic Chagas cardiomyopathy (CCC) is associated with high rates of morbidity and mortality, and it is categorized into stages of cardiac impairment (A, B, C, and D) according to the manifestations, electrocardiograph, radiological alterations, and changes in ventricular function [72, 74, 75, 82, 83]. Approximately 20–40% of patients with the indeterminate form will develop CCC. Several mechanisms contribute to this including the persistence of the parasite and autoimmunity. Moreover, factors such as dysautonomia (neurogenic mechanisms) and microvascular dysfunctions may potentiate and amplify this damage [74, 75, 77, 79, 80, 81, 84].

The cardiac form of Chagas can occur with or without ventricular failure [25, 82, 84]. Although the most common is the coexistence of arrhythmic manifestations with the congestive form, some patients have only arrhythmias and intraventricular and atrioventricular conductions compromising with normal ventricular function. Management of CCC treatment is based on the following clinical manifestations such as heart failure, cardiac arrhythmia, and thromboembolism [25, 79, 84].

Although, Chagas disease represents an important cause of heart failure (HF), limited studies have established the use of these drugs in Chagas patients. The treatment of CCC aims to reduce symptoms, delay the evolution of ventricular dysfunction, and prolong survival. In the asymptomatic or mild stages of HF, it is intended to delay the evolution of the disease. In the advanced stages, the objective is to improve the quality of life and the survival of patients. The efficacy and tolerability of these drugs in patients with CCC is extrapolated from the results obtained for other etiologies. Therefore, the CCC treatment is suggested to be performed in accordance with the general guidelines for HF treatment and should consist of the combination of three therapeutic classes: angiotensin-converting enzyme inhibitors (ACEI) or angiotensin receptor blockers (ARB), diuretics, and adrenergic beta blockers (BB) [40, 72, 82].

Angiotensin-converting enzyme inhibitors (ACEI) or angiotensin receptor blockers (ARB) have an essential role in adverse cardiac remodeling and ventricular dysfunction progression of HF [40, 72, 82]. In experimental studies reported in the literature, captopril and enalapril demonstrated to diminish myocarditis and fibrosis of CCC [85, 86, 87, 88]. These drugs should be administrated initially in low doses. In spite, some physicians report that CCC does not tolerate high doses of ACEI, according to the degree of tolerability; the dose can be progressively increased [25, 78, 81]. ACEI is recommended to patients with a left ventricular ejection fraction (LVEF) <45% [82, 83]. Regarding ARB, spironolactone and eplerenone have been evaluated in studies and data have shown that these drugs are effective in improving the quality of life and reduction of symptoms associated with CCC [89, 90, 91] and this class is considered as the treatment of choice for Chagas patients with LVEF <45%, or patients with LVEF<35% and New York Heart Association (NYHA) Class III/IV [82, 83]. In addition, in cases that are contraindicated to ACEI and ARB (hyperkalemia and progressive renal failure), the combination of hydralazine and isosorbide dinitrate should be prescribed [25, 73, 79, 83].

Beta-adrenergic blockers (carvedilol, bisoprolol, or metoprolol) are suggested in association with ACEI and ARB due to autonomous nervous system involvement in CCC and the production of antibodies against β-1 adrenergic and M-2 muscarinic receptors [25, 73, 79, 83]. Limited reports address the efficacy of this therapeutic class to treat Chagas patients with ventricular dysfunction [86, 92, 93, 94, 95, 96]. Beta blockers have been avoided because of the presence of frequent bradycardia; therefore these drugs are not indicated for patients with bradycardia ≤50 bpm or AV conduction disorders (PR > 280 ms) [25, 73, 79, 83].

Moreover, digoxin and diuretics are considered a pharmacological option in CCC, justifying the use of digoxin for patients with symptomatic LVEF ≤45%, principally in the presence of atrial fibrillation when the ventricular frequency is increased and diuretics to improve congestive symptoms and signs. For advanced HF stages, the combination of thiazides with loop diuretics has been proven to be more effective [25, 72, 73, 79, 83].

The annual incidence of thromboembolism in Chagas patients is between 1 and 2%, affecting mainly patients with HF. Occasionally, this aggravation is the first manifestation of the CD. Cardiac emboli can reach both the pulmonary and systemic circulation, with the cerebral territory being the most clinically evident [40, 79, 82]. The treatment of thromboembolism is performed based on the established recommendations, alternating according to the extension and compromised organ. For this, a score derived from a prospective cohort study with 1043 patients was recently available to evaluate the risk and to implement preventing thromboembolisms in CCC. Through risk-benefit analysis, warfarin is indicated for patients with 4–5 point. In the case of 3 points, acetylsalicylic acid (AAS) or warfarin could be used. For 2 points, it is suggested to use AAS or no prophylaxis, and 0–1 points do not need prophylaxis [79, 81, 97].

Patients with CCC usually present ventricular arrhythmia. The most frequent ventricular arrhythmias in Chagas patients are ventricular ectopies, isolated or repetitive. The presence of these arrhythmias in asymptomatic patients with preserved ventricular function does not require antiarrhythmic treatment, whereas in symptomatic cases, the antiarrhythmic treatment can be individualized. The goal of pharmacological treatment of arrhythmias is the control of symptoms. Amiodarone is widely used, despite the high incidence of adverse events. At the usual doses of 200–400 mg/day, it can be associated with alternative agents recommended for cardiopathies of other etiologies, such as propafenone, sotalol, and beta blockers, to reduce severe arrhythmic events. However, drugs belonging to class I (sodium-channel blockers) should be avoided, principally in patients with ventricular dysfunction due to proarrhythmic effects, whereas propafenone is contraindicated in patients with left ventricular dysfunction [77, 79, 83, 98]. Bradyarrhythmia is related with sinus node dysfunction or atrioventricular blocks. For the treatment of symptomatic bradyarrhythmias in CCC, a permanent pacemaker implant is usually performed [98].

4.2. Digestive form of CD

The digestive manifestations of the CD correspond to the functional alterations observed in the esophagus and intestine, which result in the formation of mega-esophagus or megacolon, respectively. These deformations are related with the involvement of the enteric nervous system, especially the Auerbachs plexus. Degenerative phenomena in this region are caused by the presence of inflammatory processes associated with autoimmune responses. Therefore, both megacolon and mega-esophagus results in alterations in motility, and consequently, in slow transit and difficulty to empty, followed by increased organ caliber [99, 100, 101, 102].

Mega-esophagus is classified into four groups with the objective of situating the different radiological aspects within the evolutionary spectrum of the affection. In addition, the classification of the mega-esophagus is important for the choice of treatment [99, 100, 101, 103].

The symptoms commonly reported in mega-esophagus are: dysphagia, regurgitation and esophageal pain. Treatment of mega-esophagus included clinical, pharmacological measures, dilatation and surgical procedure to aid in the transit of foods and liquids. The choice of treatment to be applied depends on the following factors: patient agreement, relevance of symptoms, degree of classification, nutritional status, clinical condition, comorbidities, age and available hospital infrastructure [40, 103, 104].

Clinical and pharmacological treatment is indicated for patients in group I, or patients at high risk of being treated with other forms of treatment, or in cases refusing invasive treatment. Patients must frequently drink water during the meals, eat slowly, and give preference to food in a pasty consistency. Hot and cold foods and drink and eating prior to bedtime are not recommended because food may be retained in the esophagus, causing pain or nocturnal regurgitation during sleep. The pharmacological treatment is based on substances that aim to relax the esophageal sphincter; however, the beneficial effect of these drugs is restricted to the period of action, being only a symptomatic treatment. The nitric derivatives (isosorbitol dinitrate) and the calcium channel blockers (nifedipine) are recommended [40, 101, 104, 105].

Alternative treatment is forced dilation of the distal segment of the esophagus and esophagic junction using a pneumatic or hydrostatic balloon or surgery. Surgery is performed for patients classified in group II (according to the intensity of the symptoms), III, and IV, and for patients without adequate response to clinical treatment. Another alternative is the injection of botulinum toxin, which acts to inhibit acetylcholine release [40, 101, 105].

The most frequent symptoms in megacolon consist of intestinal constipations, abdominal distension discomfort, occlusive phenomena associated with fecaloma, and sigmoid volvulus [106, 107]. Megacolon treatment may be clinical or surgical and varies according to patient agreement, level of complications, nutritional status, clinical condition, presence of comorbidity, and age [40]. If the clinical treatment indicates an alteration in the diet, use of laxatives, such as lubrificant laxatives (mineral oils) and emollient laxatives, and intestinal washes with water and glycerin may be used [105].

Advertisement

5. Therapeutic alternatives for Chagas disease treatment

5.1. Clinical trials

As previously described in this chapter, a randomized controlled clinical trial for investigation of BNZ in the treatment of CD was performed only recently: the BENEFIT clinical trial [95]. Therefore, it is not surprising that there is a lack of clinical studies with novel drug candidates for CD, since even the study to evaluate the first choice treatment is very recent.

Posaconazole (POS) (Figure 4A) is a triazole derivative with antifungal activity [108], that has been approved for the treatment of invasive fungal infections in humans [109]. It has shown trypanocidal activity in murine models [110, 111]. In addition, POS treatment led to the cure of the infection in one patient, resulting in parasite levels lower than after treatment with BNZ [112]. Therefore, POS efficacy and safety were compared to BNZ in phase II clinical trial CHAGASAZOL NCT01162967 [4], since it has shown trypanocidal activity in murine models. In this study, 78 patients were randomly selected to receive POS at a dose of 400 mg, POS at a dose of 100 mg, or BNZ at a dose of 150 mg. All drugs were orally administered twice daily for 60 days. During the treatment days, only two individual of POS patients treated with the dose of 100 mg had tested positive for T. cruzi DNA using a RT-PCR assay. The study duration was 1 year, thus the patients were retested following the treatment period. The outcomes have shown a significantly (p-value lower than 0.01) higher incidence of positive T. cruzi DNA results in both POS-treated groups (81–92%) compared to BNZ-treated group, since the last one led to only 38% of patients testing positive for T. cruzi DNA using the RT-PCR assay. While all patients from POS groups completed the study, five patients stopped the BNZ treatment, due to severe skin reactions. The adverse reactions of the current treatment with BNZ are some of the reasons why superior treatments must be pursued. However, BNZ treatment failed in fewer patients than POS [4].

Figure 4.

Structures of the compounds posaconazole (A), fexinidazole (B), ravuconazole (C), and E1224/fosravuconazole (D).

CHAGASAZOL was not the only clinical trial testing the potential application of POS in Chagas Disease treatment. The clinical trial STOP-CHAGAS NCT01377480 [113] investigated if POS or POS plus BNZ were superior to BNZ monotherapy in the reduction of parasites load after 60 days of treatment and 360 days follow up. In this study, 120 subjects were randomly divided into the following groups: POS 400 mg twice a day; BNZ 200 mg and placebo, both twice a day; BNZ 200 mg and POS 400 mg, both twice a day; or placebo 10 mg. Two outcome stages were considered, the persistence of negative RT-PCR in day 180 and the maintenance of this response by the end of the study in day 360. The successful overall outcome was the RT-PCR negative result in both stages. Both groups receiving BNZ achieve 96% subjects with negative RT-PCR at day 360, while placebo group and POS alone group had 16.7% and 23.3% subjects with negative RT-PCR, respectively. Those groups receiving BNZ also had six serious adverse events reported, such as cutaneous reactions, nervous system disorders, and gastrointestinal symptoms. Therefore once again, BNZ monotherapy was superior to POS, and no advantages were observed in the combination therapy.

Ravuconazole (Figure 4C) is another triazole compound that has shown potent and specific anti-T. cruzi action in vitro, but with limited in vivo activity, probably as a consequence of its poor pharmacokinetic properties [114]. Therefore, E1224 (Figure 4D) was developed as a prodrug of ravuconazole. A phase II Clinical Trial was performed in 2013 to evaluate the prodrug efficacy and safety compared to BNZ, as a randomized double-blinded placebo-controlled proof-of-concept study [115]. The study was performed in 224 patients with chronic indeterminate CD. Two groups received E1224 for 60 days at doses of 200 mg (low dose) and 400 mg (high dose), one group took E1224 for 30 days at a dose of 400 mg (short dose), and one group received BNZ at dose of 5 mg/kg/day. All treatments were able to eliminate blood-circulating parasites during the treatment period. E1224 had a parasite clearance rate of 90, 89, and 76% for the low dose group, short dose group, and high dose group, respectively. However, at the 12-month evaluation clearance of all patients that received E1224 was significantly low, and only the high dose group (29%) led to results significantly better than placebo. Alternatively, the BNZ clearance was 91% at 60 days after treatment and it dropped to 82% at 12 months, also being significant versus placebo. In summary, the drug candidate E1224 was as effective as BNZ at clearing the parasite following the treatment course, but the sustained efficacy 1 year following treatment was very low. Further, BNZ led to significant side effects. Therefore E1224, now called fosravuconazole, will be clinically tested in association to BNZ in order to improve treatment tolerability issues by combining them in the disease therapy.

A search for CD studies of new treatments on the clinical trials website (clinicaltrials.gov) provides one more study, with fexinidazole (Figure 4B). This compound is an old nitroimidazole that has shown promising results in preclinical models of CD [116]. According to Drug for Neglected Disease initiative (DNDi), this clinical study NCT02498782 was stopped due to safety and tolerability issues that occurred at the highest tested dose. High efficacy was observed at the lowest dose with acceptable safety and tolerability. Based on this, DNDi claims to be planning a new study in the near future [117].

5.2. Preclinical studies

Considering the lack of new and safer treatment options for CD, it is very encouraging to observe the efforts of many research groups investigating suitable candidates, which may improve the quality of life of many Chagas disease patients. Among the proposed targets for new alternative therapies examples are, ergosterol synthesis, cruzipain, trypanothione reductase, and type I nitroreductase. Studies addressing these targets in several phases of development will be discussed in this text. However, only a few compounds have demonstrated in vivo activity and favorable disposition, while none of the published preclinical candidates seems to be in the boundary of the clinical studies.

5.2.1. CYP51

The sterol 14α-demethylase (CYP51) is an important enzyme for T. cruzi survival [118], which works as a target of broad-spectrum antifungal agents, such as POS and ravuconazole, or its prodrug fosravuconazole [119]. Since they were clinically tested, as previously mentioned, they are the most close that a new treatment has come to the patients, even though the clinical studies outcomes were not the best [113]. Albaconazole was considered promising after in vitro and in vivo studies, but its effect was not satisfactory against a resistant strain [120, 121].

Several others inhibitors of sterol biosynthesis have been tested in non-clinical phases. TAK-187 is another antifungal agent [122] tested for CD due to its good drug-like properties and mechanism of action aiming at CYP51. It has shown potent anti-T. cruzi activity in vitro and in vivo [122], while preventing Trypanosoma cruzi-induced cardiac damage [123]. Recently, a new formulation of TAK-187 was patented [124], indicating that a clinical study is potentially possible.

Dialkyl imidazoles have shown high potency (EC50 in the 0.4–10 nM range) against T. cruzi in vitro, leading two of them to be tested in vivo [125]. These derivatives were able to reduce the parasitemia to undetectable levels in a mouse model of acute CD. According to the authors, these compounds are less expensive to be produced than POS. Carrying systematic modifications in the proposed scaffold of the aforementioned candidates led to new candidates with an EC50 better than that of POS (1 nM); however, only in vitro studies have been performed [126].

Since tipifarnib has shown potent anti-T. cruzi activity, it is being considered as a new lead compound for CD treatment [127]. To evaluate this drug, analogs of the antitumor agent were designed and investigated in vitro. They were considered advantageous due to their modest potency for inhibition of human CYP3A4 [128]. One of these analogs, compound 2 was evaluated in vivo, but was not better than POS or BNZ [129]. Lack of adequate pharmacokinetic properties, with short half-life and low exposure, may cause the tipifarnib analog to be ineffective.

A very rational study was performed in the development of 4-aminopyridyl-based CYP51 inhibitors [130]. In this study, compounds were developed and tested for their in vitro activity. To assess their action in vivo, the pharmacokinetic study was previously performed to better understand the compounds disposition and modify the vehicle of administration. Some of the derivatives have demonstrated better in vivo activity when compared to the in vitro studies. For the authors, the disconnection between in vitro and in vivo potency raised questions about prioritizing the CYP inhibitors based only on their in vitro performance. They achieved lead compounds with good potency and oral bioavailability, while they admit the need for optimization of the elimination half-lives.

A similar study design was followed to evaluate analogs of fenarimol [131], where it was observed that compounds with long half-lives, that is, maintaining plasma concentration above the IC50, were those that led to undetectable parasite levels in bloodstream. The compounds that were less effective to reduce the parasitemia were those with fast elimination and plasma concentrations equals or below the IC50. The two lead compounds were highly bioavailable with a long half-life. The in vivo efficacy was superior compared to BNZ and comparable to POS.

Assuming an evolution of the antifungal approach, VNI is an imidazol developed to be specifically active in the inhibition of the CYP51 of T. cruzi. It was able to cure the acute and chronic forms of CD in mice, with 100% survival and no observable side effects. Its pharmacokinetics were considered suitable for further development; however, the presented data were not completely explored [132]. The VNI optimization, VFV, overcame the promising results of VNI not only in the animal model of CD cure rate, but also with better pharmacokinetic properties and more information regarding distribution and biotransformation [133]. The reporting of VFV acute toxicity study have shown no observed adverse effect up to 200 mg/kg in mice, but authors did not present data to assure that the absorption was not saturated, that is, the exposure was proportional to the doses used in the acute toxicity study [134]. These imidazole-containing compounds seem to be the most advanced compounds, but the authors highlight the need for further studies.

5.2.2. Cruzipain

The cysteine proteinase cruzipain is vital for T. cruzi survival, since it is essential for the parasite replication and differentiation. Based on this, cruzipain has been considered as a target for drug discovery, but also for vaccines [135]. Phe-ala-PQ was a prodrug of primaquine, which had the intent to use the proteinase function of cruzipain to release the parent molecule in the parasite [136], but there are others.

Vinyl-sulfone derivatives [137], such as K11777 and WRR-483 have irreversible inhibitory activity in cruzipain, leading them to be effective against T. cruzi in vitro and, also in vivo [138, 139]. On the other hand, Cz007 and Cz008 are vinyl-sulfones containing nitrile with reversible cysteine proteinase activity that showed in vivo anti-T. cruzi action following oral administration in a dose lower than that of BNZ [140, 141]. Authors report bioavailability of both new compounds to be approximately 50%, but with short elimination half-life, what led them to administer the compounds for the efficacy study mixed to powdered chow, while BNZ was administered in water.

Additional compounds focusing on cruzipain have their anti-trypanocidal activity published only in vitro, such as thiazolyl hydrazones [142], thio semicarbazones [143], acylhydrazones [144], dipeptidyl nitrile derivatives [145], and triazine and purine nitriles [146].

5.2.3. Trypanothione reductase

Trypanothione reductase is responsible for the parasite antioxidant protective mechanism, thus inhibitors aiming for this target may disrupt its antioxidant defenses [147]. Some tricyclic compounds already active for other conditions were able to inhibit trypanothione reductase, and clomipramine was the most active with an inhibition constant (Ki) of 6 μM [148]. The testing for new active compounds led to the discovery of inhibitors with a Ki up to 0.33 μM [149]. Among the phenothiazines, thioridazine is one of the most potent irreversible inhibitors of trypanothione reductase [150], and has shown better in vivo activity than the non-treated group [151]. When compared to BNZ, thioridazine animal survival was comparable of BNZ in mice infected with T. cruzi [152].

Moreover the trypanothione reductase inhibition, hydroxymethyl nitrofurazone (NFOH) is able to inhibit cruzipain by 60%, showing in vivo activity and experimental animal survival comparable to BNZ [153]. The NFOH pharmacokinetics were studied in rats and rabbits [154, 155], and indicated that optimization is needed despite the dual action.

Additional compounds able to inhibit trypanothione reductase in vitro include quaternary analogs [156] of chlorpromazine. The dibenzosuberyl-containing analogs of clomipramine were poor inhibitors of trypanothione reductase, while the polyamine derivatives containing dibenzosuberyl were potent inhibitors up to 0.26 μM, even though they were not active in vivo, maybe due to poor pharmacokinetic properties, which were not evaluated [157]. Studying the in vitro anti-T. cruzi activities of ethyl and methyl quinoxaline-7-carboxylate 1,4-di-N-oxide derivatives, three compounds were discovered. However, only one was non-cytotoxic on host macrophage cells [158], and no in vivo study was presented.

5.2.4. Type I nitroreductase

Type I nitroreductase is an enzyme responsible for the differentiation of T. cruzi and for the activation of nitroheterocycles [159]. Nitroreductases are among the enzymes proposed as responsible for the bioactivation of BNZ [159] and NFX [160]. Based on this hypothesis, the impairment of this enzyme would be responsible for resistance [159].

The type I nitroreductase bioactivation approach supports the in vitro action of aziridinyl nitrobenzamide [161] and halogenated nitrobenzylphosphoramide mustards [162] prodrugs; however, the last was assessed in T. brucei parasites. Several nitrotriazole-based compounds demonstrated significant in vitro antichagasic activity [163, 164]. Further studies identified compounds with promising in vivo activity that has shown good in vitro ADME properties [165].

Nitrotriazole compounds with dual action aiming type I nitroreductase and inhibition of CYP51 were able to clear the parasites following a 10-day treatment, better than what was observed in the previous mentioned monofunctional 3-nitrotriazole-based derivatives [166]. Prior to the positive results of the dual action compounds, some experiments led to the understanding that nitroheterocylic compounds are better than those able to inhibit ergosterol biosynthesis [167], while it seems that together, they can work better.

Many preclinical candidates have only proven to be effective in silico or in vitro becoming promising leads or proving the validity of the proposed strategy. Novel drug candidates with in vivo activity, in limited cases have published disposition data, such as bioavailability and elimination half-life. The crucial study before clinical studies is the achievement of the NOAEL, since it is necessary to calculate the first in human dose. However, for compounds not showing clear toxicity without the pharmacokinetic assessment, the NOAEL can be overestimated due to saturated absorption, and this can be an challenge for compounds without this data [168].

5.2.5. Combined alternatives in preclinical studies

Based on the safety issues of BNZ and NFX, strategies to reduce their administered amount would prove beneficial as it may decrease the adverse effects. To circumvent the decrease in efficacy due to dose reduction, combined therapies have been proposed.

The combination of the diamidine prodrug DB289 and BNZ orally decreased parasitemia by 99%, while alone they led to 70 and 90% for DB289 and BNZ, respectively. When the combination of BNZ and DB766 was administered, the decrease in the parasitemia was at least 99.5%. Both combined treatments provided 100% protection against mice mortality, while BNZ alone provided only 78% protection [169].

The combination of BNZ and ketoconazole were evaluated in a disease mouse model. This led to better results than single treatments with susceptible and moderately resistant (Y) strains of T. cruzi. [170]. The same Y strain of T. cruzi in vivo was used to evaluate the combination of BNZ and itraconazole and the results indicated a fourfold improvement in the disease outcome against each single treatment. Additionally, this combination led to a decrease in cardiotoxicity in the experimental CD [171].

The new compound tetrahydro-β-carboline N-butyl-1-(4-dimethylamino)phenyl-1,2,3,4-tetrahydro-β-carboline-3-carboxamide was superior among its peers in reducing the number of infected cells and the number of internalized parasites, with low cytotoxicity [172]. After this observation, the selected compound was evaluated in combination with BNZ in vitro and in vivo, where a synergic effect was observed, leading to a reduction in parasitemia and mortality rates [173].

An additional compound tested with BNZ was the new drug candidate 2-methyl-1,5-bis(4-nitrophenyl)penta-1,4-dien-3-one, which has shown trypanocidal activity in vitro. Next, it was evaluated in combination with BNZ, ketoconazole, and fluconazole. Although the combination with the last one was ineffective, the combination with BNZ and ketoconazole demonstrated strong synergism [174].

These studies indicate that combination of new compounds or compounds used for alternative conditions have the potential to be a successful approach. A similar strategy has been planned for clinical trials to evaluate the combination of benznidazole and fosravuconazole, despite the absence of advantage in the combined therapy using BNZ and POS.

Advertisement

6. Conclusion

Once no new therapeutic alternatives have been included in its treatment portfolio after benznidazole and nifurtimox introduction there is a need for new compounds able to treat patients of CD. Many new drug candidates are evaluated to provide a better treatment for patients in the chronic phase. Benznidazole and nifurtimox are very effective, but their adverse effects cause many patients to discontinue treatment. Symptomatic treatment of the chronic phase of the disease is as important as the treatment directed in the reduction of the parasite. It is necessary to evaluate the patient conditions correctly to improve the quality of life. It is comprehensible that the chase for new options against this disease begins with a search of new molecules, but it is important to keep in mind that this is just a small step. Significance can only be measured if the following steps are performed and the patient treatment is reached. The combined therapy and the repurposing approaches may discover the next treatment option; however, the research must not be completed until the ideal treatment is achieved.

Advertisement

Acknowledgments

The authors wish to acknowledge the CP 01/2016 Programa Pesquisa para o Sistema Único de Saúde: Gestão Compartilhada em Saúde – PPSUS Edição 2015 Fundação Araucária-PR /SESA-PR/CNPq/MS-Decit (CV 45) and CAPES for their valuable contribution.

References

  1. 1. WHO. Chagas Disease (American Trypanosomiasis). 2017. Available from: http://www.who.int/chagas/disease/en/ [Accessed: Aug 22, 2017]
  2. 2. Kropf S, Sá M. The discovery of Trypanosoma cruzi and Chagas disease (1908–1909): Tropical medicine in Brazil. História, Ciências, Saúde-Manguinhos. 2009;16(Suppl 1):13-34
  3. 3. Malafaia G, Rodrigues ASDL. Centenário do descobrimento da doença de Chagas: Desafios e perspectivas [Centenary of the discovery of Chagas disease: Challenges and perspectives]. Revista da Sociedade Brasileira de Medicina Tropical. 2010;43(5):483-485
  4. 4. Molina I, Gómez i Prat J, Salvador F, Treviño B, Sulleiro E, Serre N, et al. Randomized trial of posaconazole and benznidazole for chronic Chagas' disease. New England Journal of Medicine. 2014;370:1899-1908
  5. 5. do Alexandre JP, Teston APM, Júnior GZ. Tratamento etiológico da doença de Chagas: Um antigo problema de saúde pública [Etiologic treatment of Chagas' disease: An old public health problem]. Revista UNINGÁ Review. 2014;20:91-96
  6. 6. Parker ER, Sethi A. Chagas disease: Coming to a place near you. Dermatologic Clinics. 2011;29:53-62
  7. 7. Bern C. Chagas’ disease. New England Journal of Medicine. 2015;373:456-466
  8. 8. Dias J, Coura J. Clínica e terapêutica da Doença de Chagas, uma abordagem prática para o clínico geral [Clinic and therapy of Chagas disease, a practical approach for the general practitioner]. Revista da Sociedade Brasileira de Medicina Tropical. 1997;30:263
  9. 9. Oliveira M, Dias A, Pontes V, Souza Júnior A, Coelho H, Coelho I. Tratamento etiológico da doença de Chagas no Brasil [Etiologic treatment of Chagas' disease in Brazil]. Revista de Patologia Tropical. 2008;37(3):207-228
  10. 10. Soares Sobrinho JL, de Moraes Medeiros FP, De La Roca MF, Ramos Silva KE, Antunes Lima LN, Rolim Neto PJ. Delineamento de alternativas terapêuticas para o tratamento da doença de Chagas [Design of therapeutic alternatives for the treatment of Chagas' disease]. Revista de Patologia Tropical. 2007;36:103-118
  11. 11. Coura J, Castro S. A critical review on Chagas disease chemotherapy. Memorias do Instituto Oswaldo Cruz. 2002;97:3-24
  12. 12. Mazza S, Cossio R, Zucardi E. Primer caso agudo de enfermedad de Chagas comprobado en Tucuman y en tratamiento con Bayer 7602 [First acute case of Chagas disease verified in Tucuman and in treatment with Bayer 7602]. Mission de Estudios de Patologia Regional de Argentina. 1937;32(3):3-18
  13. 13. Packchanian A. Chemotherapy of African sleeping sickness. I. Chemotherapy of experimental Trypanosoma gambiense infection in mice (Mus musculus) with nitrofurazone. American Journal of Tropical Medicine and Hygiene. 1955;4(4):705-711
  14. 14. Evens F, Niemegeers K, Packchanian A. Nitrofurazone Therapy of Trypanosoma Gambiense Sleeping Sickness in Man. The American Society of Tropical Medicine and Hygiene. 1957;6(4):665-678
  15. 15. Ferreira HO. Forma aguda da doença de Chagas tratada pela nitrofurazona [Acute form of Chagas' disease treated by nitrofurazone]. Revista do Instituto de Medicina Tropical de Sao Paulo. 1961;3:287-289
  16. 16. Ferreira HO. Fase aguda da doença de Chagas [Acute phase of Chagas' disease]. Journal of Hospital. 1962;61:307-311
  17. 17. Coura J, Ferreira L, Saad E, Mortel R, Silva J. Tentativa terapêutica com a nitrofurazona (Furacin) na forma crônica da doença de Chagas. Journal of Hospital. 1961;60:425-429
  18. 18. Coura J, Ferreira L, Silva J. Experiências com nitrofurazona na fase crônica da doença de Chagas [Experiments with nitrofurazone in the chronic phase of Chagas disease]. Journal of Hospital. 1962;62:957-964
  19. 19. Rodriques Coura J, de Castro SL. A critical review on Chagas disease chemotherapy. Memorias do Instituto Oswaldo Cruz. 2002;97(1):3-24
  20. 20. Bern C, Montgomery SP, Herwaldt BL, Rassi Jr A, Marin-Neto JA, Dantas RO, et al. Evaluation and treatment of Chagas disease in the United States: A systematic review. Journal of the American Medical Association. 2007;298(18):2171-2181
  21. 21. Serafim EOP, Chin CM, Ribeiro ML, Araújo DS. Abordagem da latenciação de fármacos como ferramenta para descoberta de novos antichagásicos [Approach of drug latency as a tool for the discovery of new antichagasics]. Revista Uniara. 2011;14:140-157
  22. 22. Palmeiro-Roldán R, Fonseca-Berzal C, Gómez-Barrio A, Arán VJ, Escario JA, Torrado-Durán S, et al. Development of novel benznidazole formulations: Physicochemical characterization and in vivo evaluation on parasitemia reduction in Chagas disease. International Journal of Pharmaceutics. 2014;472:110-117
  23. 23. Priotti J, Ferreira MJG, Lamas MC, Leonardi D, Salomon CJ, Nunes TG. First solid-state NMR spectroscopy evaluation of complexes of benznidazole with cyclodextrin derivatives. Carbohydrate Polymers. 2015;131:90-97
  24. 24. Romina Manarin MCL, Bottasso E, Serra E, Silvia Revelli CJS. Efficacy of novel benznidazole solutions during the experimental infection with Trypanosoma cruzi. Parasitology International. 2013;62(1):79-81
  25. 25. Benziger CP, do Carmo GA, Ribeiro AL. Chagas cardiomyopathy: Clinical presentation and Management in the Americas. Cardiology Clinics. 2017;35(1):31-47
  26. 26. Buschini A, Ferrarini L, Franzoni S, Galati S, Lazzaretti M, Mussi F, et al. Genotoxicity revaluation of three commercial nitroheterocyclic drugs: Nifurtimox, benznidazole, and metronidazole. Journal of Parasitology Research. 2009;2009:1-11
  27. 27. Perdomo VG, Rigalli JP, Villanueva SSM, Ruiz ML, Luquita MG, Echenique CG, et al. Modulation of biotransformation systems and ABC transporters by benznidazole in rats. Antimicrobial Agents and Chemotherapy. 2013;57:4894-4902
  28. 28. Trochine A, Creek DJ, Faral-Tello P, Barrett MP, Robello C. Benznidazole biotransformation and multiple targets in Trypanosoma cruzi revealed by metabolomics. PLoS Neglected Tropical Diseases. 2014;8:e2844
  29. 29. 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;193:17-19
  30. 30. Ministério da Saúde. Guia de Vigilância em saúde [Health Surveillance Guide]. Ministério da Saúde – Guia de Vigilância em saúde. 2016;1:607-619
  31. 31. PAHO. Doença de Chagas: Guia para vigilância, prevênção, controle e manejo clínico da doença de chagas aguda transmitida por alimentos [Chagas disease: A guide for surveillance, prevention, control and clinical management of acute foodborne illness]. Journal of Chemical Information and Modeling. 2009;12:160
  32. 32. Rocha MO, Teixeira MM, Ribeiro AL. An update on the management of Chagas cardiomyopathy. Expert Review of Anti-infective Therapy. 2007;5:727-743
  33. 33. Morillo CA, Marin-Neto JA, Avezum A, Sosa-Estani S, Rassi A, Rosas F, et al. Randomized trial of benznidazole for chronic Chagas’ cardiomyopathy. New England Journal of Medicine. 2015;373:1295-1306
  34. 34. Boainain E, Rassi A. Atualização Terapêutica – Terapêutica etiologica da doença de Chagas [therapeutic update – Chagas disease etiological therapy]. Arquivos Brasileiros de Cardiologia. 1979;6:395-399
  35. 35. Cançado JR. Long term evaluation of etiological treatment of Chagas disease with benznidazole. Revista do Instituto de Medicina Tropical de Sao Paulo. 2002;44:29-37
  36. 36. Costa M, Tavares VR, Aquino MVM, Moreira DB. Doença de chagas: uma revisão bibliográfica [Chagas disease: A literature review]. 2013
  37. 37. FDA. FDA Approves First U.S. Treatment for Chagas disease. Accessed: Aug 29, 2017. Available in: https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm573942.htm
  38. 38. Altclas J, Sinagra A, Dictar M, Luna C, Verón MT, De Rissio AM, et al. Chagas disease in bone marrow transplantation: An approach to preemptive therapy. Bone Marrow Transplantation. 2005;36:123-129
  39. 39. Fiorelli AI, Stolf NAG, Honorato R, Bocchi E, Bacal F, Uip D, et al. Later evolution after cardiac transplantation in Chagas’ disease. Transplantation Proceedings. 2005;37:2793-2798
  40. 40. Ministerio da Saúde. Consenso brasileiro em doença de Chagas [Brazilian consensus on Chagas' disease]. Revista da Sociedade Brasileira de Medicina Tropical. 2005;38(3):7-29
  41. 41. Sartori AMC, Ibrahim KY, Westphalen E, Braz L, Oliveira O, Gakiya E, et al. Manifestations of Chagas disease (American trypanosomiasis) in patients with HIV/AIDS. Annals of Tropical Medicine & Parasitology. 2007;101:31-50
  42. 42. Vaidian AK, Weiss LM, Tanowitz HB. Chagas' disease and AIDS. Kinetoplastid Biology and Disease. 2004;3:2
  43. 43. Ribeiro CM, Budni P, Pedrosa RC, Farias MS, Parisotto EB, Dalmarco EM, et al. Antioxidant therapy attenuates oxidative insult caused by benzonidazole in chronic Chagas' heart disease. International Journal of Cardiology. 2010;145:27-33
  44. 44. Ministerio da Saúde. Guia de Vigilância Epidemiológica [Epidemiological Surveillance Guide]. Série A Normas e Manuais Técnicos. 2009;7:819
  45. 45. Schofield CJ, Jannin J, Salvatella R. The future of Chagas disease control. Trends in parasitology. 2006;22(12):583-588
  46. 46. Brener Z. Terapêutica experimental na doença de Chagas. [Experimental therapy in Chagas' disease]. In: Andrade ZB, BarraL-Neto Z, editors. Trypanosoma cruzi e doença de Chagas. 2nd ed. Rio de Janeiro: Guanabara Koogan; 2000. pp. 379-388
  47. 47. CDC. Infectious Disease Laboratory – Formulary: U.S. Department of Health & Human Services. 2016. Available from: https://www.cdc.gov/laboratory/drugservice/formulary.html [updated Sep 22, 2016; cited Oct 15, 2017]
  48. 48. Castro JA, de Mecca MM, Bartel LC. Toxic side effects of drugs used to treat Chagas' disease (American trypanosomiasis). Human & Experimental Toxicology. 2006;25(8):471-479
  49. 49. Coura J, de Abreu L, Willcox H, Petana W. Comparative controlled study on the use of benznidazole, nifurtimox and placebo, in the chronic form of Chagas' disease, in a field area with interrupted transmission. I. Preliminary evaluation. Revista da Sociedade Brasileira de Medicina Tropical. 1997;30(2):139-144
  50. 50. Núñez-Vergara LJ, Squella JA, Aldunate J, Letelier ME, Bollo S, Repetto Y, et al. Nitro radical anion formation from nifurtimox. Part 1: Biological evidences in Trypanosoma cruzi. Bioelectrochemistry and Bioenergetics. 1997;43(1):151-155
  51. 51. 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
  52. 52. Wilkinson SR, Bot C, Kelly JM, Hall BS. Trypanocidal activity of nitroaromatic prodrugs: Current treatments and future perspectives. Current Topics in Medicinal Chemistry. 2011;11(16):2072-2084
  53. 53. Docampo R, Dubin M, Martino EE, Moreno SN, Stoppani AO. Influence of nifurtimox on the glutathione content of the liver and bile in rats. Medicina. 1983;43(1):33-40
  54. 54. Docampo R, Moreno SN. Free radical metabolism of antiparasitic agents. Federation Proceedings. 1986;45(10):2471-2476
  55. 55. Docampo R, Moreno SN. Free radical metabolites in the mode of action of chemotherapeutic agents and phagocytic cells on Trypanosoma cruzi. Reviews of Infectious Diseases. 1984;6(2):223-238
  56. 56. Docampo R, Moreno SN, Stoppani AO, Leon W, Cruz FS, Villalta F, et al. Mechanism of nifurtimox toxicity in different forms of Trypanosoma cruzi. Biochemical Pharmacology. 1981;30(14):1947-1951
  57. 57. Docampo R, Stoppani AO. Generation of superoxide anion and hydrogen peroxide induced by nifurtimox in Trypanosoma cruzi. Archives of Biochemistry and Biophysics. 1979;197(1):317-321
  58. 58. Docampo R, Stoppani AO. Mechanism of the trypanocidal action of nifurtimox and other nitro-derivatives on Trypanosoma cruzi. Medicina. 1980;40(Suppl 1):10-16
  59. 59. Patterson S, Wyllie S. Nitro drugs for the treatment of trypanosomatid diseases: Past, present, and future prospects. Trends in Parasitology. 2014;30(6):289-298
  60. 60. Morello A. The biochemistry of the mode of action of drugs and the detoxication mechanisms in Trypanosoma cruzi. Comparative Biochemistry and Physiology C, Comparative Pharmacology and Toxicology. 1988;90(1):1-12
  61. 61. Boveris A, Sies H, Martino EE, Docampo R, Turrens JF, Stoppani AO. Deficient metabolic utilization of hydrogen peroxide in Trypanosoma cruzi. The Biochemical Journal. 1980;188(3):643-648
  62. 62. Docampo R, de Boiso JF, Boveris A, Stoppani AO. Localization of peroxidase activity in Trypanosoma cruzi microbodies. Experientia. 1976;32(8):972-975
  63. 63. Cançado JR, Salgado AA, Marra UD, Alvares JM, Machado JR. Ensaio terapêutico clínico na doença de Chagas crônica com o nifurtimox era três esquemas de duração prolongada [Clinical therapeutic trial in chronic Chagas' disease with nifurtimox was three schemes of prolonged duration]. Revista do Instituto de Medicina Tropical de São Paulo. 1975;17:111
  64. 64. Cerisola JA, Rohwedder R, Segura EL, del Prado CE, Alvares M, de Martini GJW. El xenodiagnóstico [The xenodiagnosis]. Imprenta dei Instituto Nacional de Investigaciones Cardiovasculares. 1974;1:127
  65. 65. Rassi A, Ferreira HO. Tentativas de tratamento especifico da fase aguda da doença de chagas com nitrofuranos em esquemas de duração prolongada [Specific treatment attempts of the acute phase of chagas disease with nitrofurans in long duration regimens]. Revista da Sociedade Brasileira de Medicina Tropical. 1971;5(2):35
  66. 66. Schenone H, Concha L, Aranda R, Rojas A, Knierim F, Rojo M. Treatment of chronic Chagas' infection with Lampit. Boletin chileno de parasitologia. 1972;27(1):11-14
  67. 67. Silva NN, Kuhn G, Santos JFC, Eye GV, Chaher JAB. Eficácia e tolerância do nitrofurfurilidene na fase crônica da moléstia de Chagas [Efficacy and tolerance of nitrofurfurylidene in the chronic phase of Chagas' disease]. Revista da Sociedade Brasileira de Medicina Tropical. 1974;8:325
  68. 68. Forsyth CJ, Hernandez S, Olmedo W, Abuhamidah A, Traina MI, Sanchez DR, et al. Safety profile of nifurtimox for treatment of Chagas disease in the United States. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America. 2016;63(8):1056-1062
  69. 69. Sanchez G, Coronado X, Zulantay I, Apt W, Gajardo M, Solari S, et al. Monitoring the efficacy of specific treatment in chronic Chagas disease by polymerase chain reaction and flow cytometry analysis. Parasite. 2005;12(4):353-357
  70. 70. Murcia L, Carrilero B, Albajar Vinas P, Segovia M. Nifurtimox chemotherapy: Collateral effects in treated Trypanosoma cruzi infected patients. Revista espanola de quimioterapia : publicacion oficial de la Sociedad Espanola de Quimioterapia. 2012;25(1):74-75
  71. 71. Streiger ML, del Barco ML, Fabbro DL, Arias ED, Amicone NA. Longitudinal study and specific chemotherapy in children with chronic Chagas' disease, residing in a low endemicity area of Argentina. Revista da Sociedade Brasileira de Medicina Tropical. 2004;37(5):365-375
  72. 72. Bestetti RB. Chagas heart failure in patients from Latin America. Journal of Cardiac Failure Reviews. 2016;2(2):90-94
  73. 73. Bestetti RB, Theodoropoulos TA, Cardinalli-Neto A, Cury PM. Treatment of chronic systolic heart failure secondary to Chagas heart disease in the current era of heart failure therapy. American Heart Journal. 2008;156(3):422-430
  74. 74. Cunha-Neto E, Chevillard C. Chagas disease cardiomyopathy: Immunopathology and genetics. Mediators of Inflammation. 2014;2014:683230
  75. 75. Higuchi M d L, Benvenuti LA, Martins Reis M, Metzger M. Pathophysiology of the heart in Chagas' disease: Current status and new developments. Cardiovascular Research. 2003;60(1):96-107
  76. 76. Strasen J, Williams T, Ertl G, Zoller T, Stich A, Ritter O. Epidemiology of Chagas disease in Europe: Many calculations, little knowledge. Clinical Research in Cardiology. 2014;103(1):1-10
  77. 77. Barbosa MP, Carmo AA, Rocha MO, Ribeiro AL. Ventricular arrhythmias in Chagas disease. Revista da Sociedade Brasileira de Medicina Tropical. 2015;48(1):4-10
  78. 78. Botoni FA, Ribeiro AL, Marinho CC, Lima MM, Nunes Mdo C, Rocha MO. Treatment of Chagas cardiomyopathy. BioMed Research International. 2013;2013:849504
  79. 79. de Cardiologia SB. I Latin American guideline for the diagnosis and treatment of Chagas' heart disease. Journal of Sociedade Brasileira de Cardiologia. 2011;97(2):1-48
  80. 80. Rassi Junior A, Rassi A, Marin-Neto J. Chagas heart disease: Pathophysiologic mechanisms, prognostic factors and risk stratification. Memorias do Instituto Oswaldo Cruz. 2009;104(1):152-158
  81. 81. Ribeiro AL, Nunes MP, Teixeira MM, Rocha MO. Diagnosis and management of Chagas disease and cardiomyopathy. Nature Reviews. Cardiology. 2012;9(10):576-589
  82. 82. Andrade J, Marin Neto J, de Paola A, Vilas-Boas A, Oliveira G, Bacal F, et al. I Latin American guidelines for the diagnosis and treatment of Chagas’ heart disease – Executive summary. Arquivos Brasileiros de Cardiologia. 2011;96(6):434-442
  83. 83. Mora G. Chagas cardiomyopathy. Journal of Cardiology Practice. 2016;14(31):1-7
  84. 84. Bilate AM, Cunha-Neto E. Chagas disease cardiomyopathy: Current concepts of an old disease. Revista do Instituto de Medicina Tropical de Sao Paulo. 2008;50(2):67-74
  85. 85. Batlouni M, Barretto AC, Armaganijan D, Vichi FL, Spritzer N, Simoes R, et al. Treatment of mild and moderate cardiac failure with captopril. A multicenter study. Arquivos Brasileiros de Cardiologia. 1992;58(5):417-421
  86. 86. Botoni FA, Poole-Wilson PA, Ribeiro AL, Okonko DO, Oliveira BM, Pinto AS, et al. A randomized trial of carvedilol after renin-angiotensin system inhibition in chronic Chagas cardiomyopathy. American Heart Journal. 2007;153(4):544 e1-544e8
  87. 87. Leon JS, Wang K, Engman DM. Captopril ameliorates myocarditis in acute experimental Chagas disease. Circulation. 2003;107(17):2264-2269
  88. 88. Roberti RR, Martinez EE, Andrade JL, Araujo VL, Brito FS, Portugal OP, et al. Chagas cardiomyopathy and captopril. European Heart Journal. 1992;13(7):966-970
  89. 89. Pitt B, Remme W, Zannad F, Neaton J, Martinez F, Roniker B, et al. Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction. The New England Journal of Medicine. 2003;348(14):1309-1321
  90. 90. Pitt B, Zannad F, Remme WJ, Cody R, Castaigne A, Perez A, et al. The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone evaluation study investigators. The New England Journal of Medicine. 1999;341(10):709-717
  91. 91. Ramires FJ, Salemi VM, Ianni BM, Fernandes F, Martins DG, Billate A, et al. Aldosterone antagonism in an inflammatory state: Evidence for myocardial protection. Journal of the Renin-Angiotensin-Aldosterone System. 2006;7(3):162-167
  92. 92. Bestetti RB, Otaviano AP, Cardinalli-Neto A, da Rocha BF, Theodoropoulos TA, Cordeiro JA. Effects of B-blockers on outcome of patients with Chagas' cardiomyopathy with chronic heart failure. International Journal of Cardiology. 2011;151(2):205-208
  93. 93. Bestetti RB, Theodoropoulos TA, Cardinalli-Neto A. Treating patients with Chagas' cardiomyopathy with chronic heart failure in the contemporary era. American Heart Journal. 2007;154(5):e35. Author reply e3
  94. 94. Issa VS, Amaral AF, Cruz FD, Ferreira SM, Guimaraes GV, Chizzola PR, et al. Beta-blocker therapy and mortality of patients with Chagas cardiomyopathy: A subanalysis of the REMADHE prospective trial. Circulation. Heart Failure. 2010;3(1):82-88
  95. 95. Marin-Neto JA, Rassi Jr A, Avezum Jr A, Mattos AC, Rassi A. The BENEFIT trial: Testing the hypothesis that trypanocidal therapy is beneficial for patients with chronic Chagas heart disease. Memorias do Instituto Oswaldo Cruz. 2009;104:319-324
  96. 96. Quiros FR, Morillo CA, Casas JP, Cubillos LA, Silva FA. CHARITY: Chagas cardiomyopathy bisoprolol intervention study: A randomized double-blind placebo force-titration controlled study with Bisoprolol in patients with chronic heart failure secondary to Chagas cardiomyopathy [NCT00323973]. Trials. 2006;7:21
  97. 97. Sousa AS, Xavier SS, Freitas GR, Hasslocher-Moreno A. Prevention strategies of cardioembolic ischemic stroke in Chagas' disease. Arquivos Brasileiros de Cardiologia. 2008;91(5):306-310
  98. 98. Bestetti R. Cardiomiopatia chagásica crônica: diagnóstico e tratamento. Revista da Sociedade de Cardiologia do Estado de São Paulo. 2016;26(4):246-252
  99. 99. de Oliveira AP, Bernardo CR, Camargo AV, Ronchi LS, Borim AA, de Mattos CC, et al. Genetic susceptibility to cardiac and digestive clinical forms of chronic Chagas disease: Involvement of the CCR5 59029 A/G polymorphism. PLoS One. 2015;10(11):e0141847
  100. 100. Prata A. Clinical and epidemiological aspects of Chagas disease. The Lancet Infectious Diseases. 2001;1(2):92-100
  101. 101. Rassi Junior A, Rassi A, Marin-Neto J. Chagas disease. The Lancet. 2010;375:1388-1402
  102. 102. Gullo C, Estofolete C, Gil C, Christiano A, Gomes NJ. Formas digestivas da doença de Chagas e carcinogênese: um estudo de associação [Digestive forms of Chagas' disease and carcinogenesis: an association study]. Revista do Colégio Brasileiro. 2012;39(2):146-150
  103. 103. Matsuda NM, Miller SM, Evora PR. The chronic gastrointestinal manifestations of Chagas disease. Clinics (São Paulo, Brazil). 2009;64(12):1219-1224
  104. 104. Dias JCP, Coura JR. org. Clínica e terapêutica da doença de Chagas: uma abordagem prática para o clínico geral [Clinical and therapeutic of Chagas disease: a practical approach for the general practitioner]. In: Rezende JM, editor. O Aparelho Digestivo na doença de Chagas [The Digestive Tract in Chagas Disease]. Rio de Janeiro: Editora FIOCRUZ. 1997;1:486
  105. 105. Menegheli U. Clinical treatment of the digestive form of Chagas disease. Memorias do Instituto Oswaldo Cruz. 1999;94(1):341-342
  106. 106. Braga C, Guimarães L, Amaral R, Bonani A, Moreira N, Gomes M, et al. Megacólon de Doença de Chagas: Uma Abordagem Interdisciplinar [Megacolon of Chagas disease: An interdisciplinary approach]. Revista de Saúde e Biologia. 2013;8(1):36-43
  107. 107. Posada E, Pell C, Angulo N, Pinazo MJ, Gimeno F, Elizalde I, et al. Bolivian migrants with Chagas disease in Barcelona, Spain: A qualitative study of dietary changes and digestive problems. International Health. 2011;3(4):289-294
  108. 108. Patterson TF. Role of newer azoles in surgical patients. Journal of Chemotherapy. 1999;11(6):504-512
  109. 109. Kwon DS, Mylonakis E. Posaconazole: A new broad-spectrum antifungal agent. Expert Opinion on Pharmacotherapy. 2007;8(8):1167-1178
  110. 110. Molina J, Martins-Filho O, Brener Z, Romanha AJ, Loebenberg D, Urbina JA. Activities of the triazole derivative SCH 56592 (Posaconazole) against drug-resistant strains of the protozoan parasite Trypanosoma (Schizotrypanum) cruzi in immunocompetent and immunosuppressed murine hosts. Antimicrobial Agents and Chemotherapy. 2000;44(1):150-155
  111. 111. Urbina JA, Payares G, Contreras LM, Liendo A, Sanoja C, Molina J, et al. Antiproliferative effects and mechanism of action of SCH 56592 against Trypanosoma (Schizotrypanum) cruzi: In vitro and in vivo studies. Antimicrobial Agents and Chemotherapy. 1998;42(7):1771-1777
  112. 112. Pinazo MJ, Espinosa G, Gallego M, Lopez-Chejade PL, Urbina JA, Gascon J. Successful treatment with posaconazole of a patient with chronic Chagas disease and systemic lupus erythematosus. American Journal of Tropical Medicine and Hygiene. 2010;82(4):583-587
  113. 113. Morillo CA, Waskin H, Sosa-Estani S, del Carmen Bangher M, Cuneo C, Milesi R, et al. Benznidazole and posaconazole in eliminating parasites in asymptomatic T. cruzi carriers. Journal of the American College of Cardiology. 2017;69(8):939-947
  114. 114. Urbina JA, Payares G, Sanoja C, Lira R, Romanha AJ. In vitro and in vivo activities of ravuconazole on Trypanosoma cruzi, the causative agent of Chagas disease. International Journal of Antimicrobial Agents. 2003;21(1):27-38
  115. 115. Torrico F, Gascon J, Ortiz L, Alonso-Vega C, Pinazo MJ, Schijman A, Almeida IC, Alves F, Strub-Wourgaft N, Ribeiro I. Treatment of adult chronic indeterminate Chagas disease with benznidazole and three E1224 dosing regimens: A proof-of-concept, randomised, placebo-controlled trial. The Lancet Infectious Diseases. 2018. DOI:10.1016/S1473-3099(17)30538-8
  116. 116. Bahia MT, Nascimento AFS, Mazzeti AL, Marques LF, Goncalves KR, Mota LWR, et al. Antitrypanosomal activity of fexinidazole metabolites, potential new drug candidates for Chagas disease. Antimicrobial Agents and Chemotherapy. 2014;58(8):4362-4370
  117. 117. DNDi DfNDi. Fexinidazole (Chagas). 2017. Available from: https://www.dndi.org/diseases-projects/portfolio/fexinidazole-chagas/ [updated Aug 2017; cited Aug 23, 2017]
  118. 118. Lepesheva GI, Zaitseva NG, Nes WD, Zhou W, Arase M, Liu J, et al. CYP51 fromTrypanosoma cruzi. Journal of Biological Chemistry. 2006;281(6):3577-3585
  119. 119. Buckner FS, Urbina JA. Recent developments in sterol 14-demethylase inhibitors for Chagas disease. International Journal for Parasitology: Drugs and Drug Resistance. 2012;2:236-242
  120. 120. Urbina JA, Lira R, Visbal G, Bartroli J. In vitro antiproliferative effects and mechanism of action of the new triazole derivative UR-9825 against the protozoan parasite Trypanosoma (Schizotrypanum) cruzi. Antimicrobial Agents and Chemotherapy. 2000;44(9):2498-2502
  121. 121. Guedes PMM, Urbina JA, de Lana M, Afonso LCC, Veloso VM, Tafuri WL, et al. Activity of the new triazole derivative albaconazole against Trypanosoma (Schizotrypanum) cruzi in dog hosts. Antimicrobial Agents and Chemotherapy. 2004;48(11):4286-4292
  122. 122. Schell WA, De Almeida GM, Dodge RK, Okonogi K, Perfect JR. In vitro and in vivo efficacy of the triazole TAK-187 against Cryptococcus neoformans. Antimicrobial Agents and Chemotherapy. 1998;42(10):2630-2632
  123. 123. Corrales M, Cardozo R, Segura MA, Urbina JA, Basombrio MA. Comparative efficacies of TAK-187, a long-lasting ergosterol biosynthesis inhibitor, and benznidazole in preventing cardiac damage in a murine model of Chagas' disease. Antimicrobial Agents and Chemotherapy. 2005;49(4):1556-1560
  124. 124. Ueki Y. WO2013157584 A1. Google Patents; 2013
  125. 125. Suryadevara PK, Olepu S, Lockman JW, Ohkanda J, Karimi M, Verlinde CLMJ, et al. Structurally simple inhibitors of Lanosterol 14α-Demethylase are efficacious in a rodent model of acute Chagas disease. Journal of Medicinal Chemistry. 2009;52(12):3703-3715
  126. 126. Suryadevara PK, Racherla KK, Olepu S, Norcross NR, Tatipaka HB, Arif JA, et al. Dialkylimidazole inhibitors of Trypanosoma cruzi sterol 14α-demethylase as anti-Chagas disease agents. Bioorganic & Medicinal Chemistry Letters. 2013;23(23):6492-6499
  127. 127. Hucke O, Gelb MH, Verlinde CLMJ, Buckner FS. The protein farnesyltransferase inhibitor Tipifarnib as a new lead for the development of drugs against Chagas disease. Journal of Medicinal Chemistry. 2005;48(17):5415-5418
  128. 128. Kraus JM, Tatipaka HB, McGuffin SA, Chennamaneni NK, Karimi M, Arif J, et al. Second generation analogues of the cancer drug clinical candidate Tipifarnib for anti-Chagas disease drug discovery. Journal of Medicinal Chemistry. 2010;53(10):3887-3898
  129. 129. Buckner FS, Bahia MT, Suryadevara PK, White KL, Shackleford DM, Chennamaneni NK, et al. Pharmacological characterization, structural studies, and in vivo activities of anti-Chagas disease lead compounds derived from Tipifarnib. Antimicrobial Agents and Chemotherapy. 2012;56(9):4914-4921
  130. 130. Calvet CM, Vieira DF, Choi JY, Kellar D, Cameron MD, Siqueira-Neto JL, et al. 4-Aminopyridyl-based CYP51 inhibitors as anti-Trypanosoma cruzi drug leads with improved pharmacokinetic profile and in Vivo potency. Journal of Medicinal Chemistry. 2014;57(16):6989-7005
  131. 131. Keenan M, Chaplin JH, Alexander PW, Abbott MJ, Best WM, Khong A, et al. Two analogues of fenarimol show curative activity in an experimental model of Chagas disease. Journal of Medicinal Chemistry. 2013;56(24):10158-10170
  132. 132. Villalta F, Dobish MC, Nde PN, Kleshchenko YY, Hargrove TY, Johnson CA, et al. VNI cures acute and chronic experimental Chagas disease. The Journal of infectious diseases. 2013;208(3):504-511
  133. 133. Lepesheva GI, Hargrove TY, Rachakonda G, Wawrzak Z, Pomel S, Cojean S, et al. VFV as a new effective CYP51 structure-derived drug candidate for Chagas disease and visceral Leishmaniasis. Journal of Infectious Diseases. 2015;212(9):1439-1448
  134. 134. Guedes-da-Silva FH, Batista DGJ, Da Silva CF, De Araújo JS, Pavão BP, Simões-Silva MR, et al. Antitrypanosomal activity of sterol 14α-Demethylase (CYP51) inhibitors VNI and VFV in the Swiss mouse models of Chagas disease induced by the Trypanosoma cruzi Y strain. Antimicrobial Agents and Chemotherapy. 2017;61(4):e02098-16
  135. 135. Duschak V, Couto A. Cruzipain, the major cysteine protease of Trypanosoma cruzi: A sulfated glycoprotein antigen as relevant candidate for vaccine development and drug target. A review. Current Medicinal Chemistry. 2009;16(24):3174-3202
  136. 136. Chung MC, Gonçalves MF, Colli W, Ferreira EI, Miranda MTM. Synthesis and in vitro evaluation of potential antichagasic dipeptide prodrugs of primaquine. Journal of Pharmaceutical Sciences. 1997;86(10):1127-1131
  137. 137. Kerr ID, Lee JH, Farady CJ, Marion R, Rickert M, Sajid M, et al. Vinyl sulfones as antiparasitic agents and a structural basis for drug design. Journal of Biological Chemistry. 2009;284(38):25697-25703
  138. 138. Rodriguez A, Chen YT, Brinen LS, Kerr ID, Hansell E, Doyle PS, et al. In vitro and in vivo studies of the trypanocidal properties of WRR-483 against Trypanosoma cruzi. PLoS Neglected Tropical Diseases. 2010;4(9):e825
  139. 139. Doyle PS, Zhou YM, Engel JC, McKerrow JH. A cysteine protease inhibitor cures Chagas' disease in an immunodeficient-mouse model of infection. Antimicrobial Agents and Chemotherapy. 2007;51(11):3932-3939
  140. 140. Ndao M, Beaulieu C, Black WC, Isabel E, Vasquez-Camargo F, Nath-Chowdhury M, et al. Reversible cysteine protease inhibitors show promise for a Chagas disease cure. Antimicrobial Agents and Chemotherapy. 2013;58(2):1167-1178
  141. 141. Beaulieu C, Isabel E, Fortier A, Massé F, Mellon C, Méthot N, et al. Identification of potent and reversible cruzipain inhibitors for the treatment of Chagas disease. Bioorganic & Medicinal Chemistry Letters. 2010;20(24):7444-7449
  142. 142. Leite ACL, Moreira DR de M, Cardoso MV de O, Hernandes MZ, Alves Pereira VR, Silva RO, et al. Synthesis, cruzain docking, and in vitro studies of aryl-4-oxothiazolylhydrazones against Trypanosoma cruzi. ChemMedChem 2007;2(9):1339-1345
  143. 143. Du X, Guo C, Hansell E, Doyle PS, Caffrey CR, Holler TP, et al. Synthesis and structure-activity relationship study of potent trypanocidal thio semicarbazone inhibitors of the trypanosomal cysteine protease cruzain. Journal of Medicinal Chemistry. 2002;45(13):2695-2707
  144. 144. dos Santos Filho JM, Leite AC, de Oliveira BG, Moreira DR, Lima MS, Soares MB, et al. Design, synthesis and cruzain docking of 3-(4-substituted-aryl)-1,2,4-oxadiazole-N-acylhydrazones as anti-Trypanosoma cruzi agents. Bioorganic & Medicinal Chemistry. 2009;17(18):6682-6691
  145. 145. Pollastri MP, Avelar LAA, Camilo CD, de Albuquerque S, Fernandes WB, Gonçalez C, et al. Molecular design, synthesis and trypanocidal activity of dipeptidyl nitriles as cruzain inhibitors. PLOS Neglected Tropical Diseases. 2015;9(7):e0003916
  146. 146. Mott BT, Ferreira RS, Simeonov A, Jadhav A, Ang KK-H, Leister W, et al. Identification and optimization of inhibitors of trypanosomal cysteine proteases: Cruzain, rhodesain, and TbCatB. Journal of Medicinal Chemistry. 2010;53(1):52-60
  147. 147. Flohé L. The trypanothione system and its implications in the therapy of trypanosomatid diseases. International Journal of Medical Microbiology. 2012;302(4–5):216-220
  148. 148. Benson TJ, McKie JH, Garforth J, Borges A, Fairlamb AH, Douglas KT. Rationally designed selective inhibitors of trypanothione reductase. Phenothiazines and related tricyclics as lead structures. The Biochemical Journal. 1992;286(Pt 1):9-11
  149. 149. Richardson JL, Nett IRE, Jones DC, Abdille MH, Gilbert IH, Fairlamb AH. Improved tricyclic inhibitors of trypanothione reductase by screening and chemical synthesis. ChemMedChem. 2009;4(8):1333-1340
  150. 150. Gutierrez-Correa J, Fairlamb AH, Stoppani AO. Trypanosoma cruzi trypanothione reductase is inactivated by peroxidase-generated phenothiazine cationic radicals. Free Radical Research. 2001;34(4):363-378
  151. 151. Lo Presti MS, Rivarola HW, Bustamante JM, Fernández AR, Enders JE, Fretes R, et al. Thioridazine treatment prevents cardiopathy in Trypanosoma cruzi infected mice. International Journal of Antimicrobial Agents. 2004;23(6):634-636
  152. 152. Bustamante JM, Presti MSL, Rivarola HW, Fernández AR, Enders JE, Fretes RE, et al. Treatment with benznidazole or thioridazine in the chronic phase of experimental Chagas disease improves cardiopathy. International Journal of Antimicrobial Agents. 2007;29(6):733-737
  153. 153. Davies C, Cardozo RM, Negrette OS, Mora MC, Chung MC, Basombrio MA. Hydroxymethylnitrofurazone is active in a murine model of Chagas' disease. Antimicrobial Agents and Chemotherapy. 2010;54(9):3584-3589
  154. 154. Nogueira Filho MA, Padilha EC, Campos ML, Pontes Machado DV, Davanço MG, Pestana KC, et al. Pharmacokinetics of hydroxymethylnitrofurazone and its parent drug nitrofurazone in rabbits. Drug Metabolism Letters. 2013;7(1):58-64
  155. 155. Serafim EOP, ATdAe S, Moreno AH, Vizioli EO, Ferreira EI, Peccinini RG, et al. Pharmacokinetics of hydroxymethylnitrofurazone, a promising new Prodrug for Chagas' disease treatment. Antimicrobial Agents and Chemotherapy. 2013;57(12):6106-6109
  156. 156. Parveen S, Khan MOF, Austin SE, Croft SL, Yardley V, Rock P, et al. Antitrypanosomal, antileishmanial, and antimalarial activities of quaternary arylalkylammonium 2-amino-4-chlorophenyl phenyl sulfides, a new class of trypanothione reductase inhibitor, and of N-acyl derivatives of 2-amino-4-chlorophenyl phenyl sulfide. Journal of Medicinal Chemistry. 2005;48(25):8087-8097
  157. 157. O’Sullivan MC, Durham TB, Valdes HE, Dauer KL, Karney NJ, Forrestel AC, et al. Dibenzosuberyl substituted polyamines and analogs of clomipramine as effective inhibitors of trypanothione reductase; molecular docking, and assessment of trypanocidal activities. Bioorganic & Medicinal Chemistry. 2015;23(5):996-1010
  158. 158. Villalobos-Rocha JC, Sánchez-Torres L, Nogueda-Torres B, Segura-Cabrera A, García-Pérez CA, Bocanegra-García V, et al. Anti-Trypanosoma cruzi and anti-leishmanial activity by quinoxaline-7-carboxylate 1,4-di-N-oxide derivatives. Parasitology Research. 2014;113(6):2027-2035
  159. 159. Wilkinson SR, Taylor MC, Horn D, Kelly JM, Cheeseman I. A mechanism for cross-resistance to nifurtimox and benznidazole in trypanosomes. Proceedings of the National Academy of Sciences. 2008;105(13):5022-5027
  160. 160. Kubata BK, Kabututu Z, Nozaki T, Munday CJ, Fukuzumi S, Ohkubo K, et al. A key role for old yellow enzyme in the metabolism of drugs by Trypanosoma cruzi. The Journal of Experimental Medicine. 2002;196(9):1241-1251
  161. 161. Bot C, Hall BS, Bashir N, Taylor MC, Helsby NA, Wilkinson SR. Trypanocidal activity of aziridinyl nitrobenzamide prodrugs. Antimicrobial Agents and Chemotherapy. 2010;54(10):4246-4252
  162. 162. Hall BS, Wu X, Hu L, Wilkinson SR. Exploiting the drug-activating properties of a novel trypanosomal nitroreductase. Antimicrobial Agents and Chemotherapy. 2009;54(3):1193-1199
  163. 163. Papadopoulou MV, Trunz BB, Bloomer WD, McKenzie C, Wilkinson SR, Prasittichai C, et al. Novel 3-nitro-1H-1,2,4-triazole-based aliphatic and aromatic amines as anti-chagasic agents. Journal of Medicinal Chemistry. 2011;54(23):8214-8223
  164. 164. Papadopoulou MV, Bloomer WD, Rosenzweig HS, Chatelain E, Kaiser M, Wilkinson SR, et al. Novel 3-nitro-1H-1,2,4-triazole-based amides and Sulfonamides as potential antitrypanosomal agents. Journal of Medicinal Chemistry. 2012;55(11):5554-5565
  165. 165. Papadopoulou MV, Bloomer WD, Rosenzweig HS, Ashworth R, Wilkinson SR, Kaiser M, et al. Novel 3-nitro-1H-1,2,4-triazole-based compounds as potential anti-chagasic drugs:In vivostudies. Future Medicinal Chemistry. 2013;5(15):1763-1776
  166. 166. Papadopoulou MV, Bloomer WD, Lepesheva GI, Rosenzweig HS, Kaiser M, Aguilera-Venegas B, et al. Novel 3-Nitrotriazole-based amides and carbinols as bifunctional antichagasic agents. Journal of Medicinal Chemistry. 2015;58(3):1307-1319
  167. 167. Moraes CB, Giardini MA, Kim H, Franco CH, Araujo-Junior AM, Schenkman S, et al. Nitroheterocyclic compounds are more efficacious than CYP51 inhibitors against Trypanosoma cruzi: Implications for Chagas disease drug discovery and development. Scientific Reports. 2014;4(1):1-11
  168. 168. Guidance for Industry: Estimating the Maximum Safe Starting Dose in Initial Clinical Trials for Therapeutics in Adult Healthy Volunteers. U.S. Department of Health and Human Services. Food and Drug Administration. Center for Drug Evaluation and Research (CDER); 2005
  169. 169. Moreno SN, Batista DDGJ, Batista MM, Oliveira GMD, Britto CC, Rodrigues ACM, et al. Combined treatment of heterocyclic analogues and benznidazole upon Trypanosoma cruzi in vivo. PLoS One. 2011;6(7):e22155
  170. 170. Araujo MS, Martins-Filho OA, Pereira ME, Brener ZA. Combination of benznidazole and ketoconazole enhances efficacy of chemotherapy of experimental Chagas' disease. The Journal of Antimicrobial Chemotherapy. 2000;45(6):819-824
  171. 171. Costa FTM, Assíria Fontes Martins T, de Figueiredo Diniz L, Mazzeti AL, da Silva do Nascimento ÁF, Caldas S, et al. Benznidazole/Itraconazole combination treatment enhances anti-Trypanosoma cruzi Activity in experimental Chagas disease. PLoS One. 2015;10(6):e0128707
  172. 172. Valdez RH, Tonin LTD, Ueda-Nakamura T, Filho BPD, Morgado-Diaz JA, Sarragiotto MH, et al. Biological activity of 1,2,3,4-tetrahydro-β-carboline-3-carboxamides against Trypanosoma cruzi. Acta Tropica. 2009;110(1):7-14
  173. 173. Valdez RH, Tonin LTD, Ueda-Nakamura T, Silva SO, Dias Filho BP, Kaneshima EN, et al. In vitro and in vivo trypanocidal synergistic activity of N-butyl-1-(4-dimethylamino)phenyl-1,2,3,4-tetrahydro-carboline-3-carboxamide associated with benznidazole. Antimicrobial Agents and Chemotherapy. 2011;56(1):507-512
  174. 174. Peron F, Lazarin-Bidóia D, Ud Din Z, Rodrigues-Filho E, Ueda-Nakamura T, SDO S, et al. Effects of (1E,4E)-2-methyl-1,5-bis(4-nitrophenyl)penta-1,4-dien-3-one on Trypanosoma cruzi and its combinational effect with benznidazole, ketoconazole, or fluconazole. BioMed Research International. 2017;2017:1-11

Written By

Wilton H. Kawaguchi, Leticia Bonancio Cerqueira, Mariana Millan Fachi, Michel L. Campos, Iara J. Messias Reason and Roberto Pontarolo

Submitted: 13 June 2017 Reviewed: 05 February 2018 Published: 13 March 2018