The Mechanism by which the Phenothiazine ...

15 downloads 134 Views 124KB Size Report
Mar 28, 2012 - rifampicin, the two most effective anti-tuberculosis agents), ...... [106] Mickie, B., Eleftherios, S. Combination therapy for the treatment of cancer.
Send Orders for Reprints to [email protected] Recent Patents on Anti-Infective Drug Discovery, 2013, 8, 000-000

1

The Mechanism by which the Phenothiazine Thioridazine Contributes to Cure Problematic Drug-Resistant Forms of Pulmonary Tuberculosis: Recent Patents for “New Use” Leonard Amaral1*, Ana Martins2,3, Gabriella Spengler2, Attila Hunyadi4 and Joseph Molnar2 1

Center for Malaria and Other Tropical Diseases (CMDT), Institute of Hygiene and Tropical Medicine, Universidade Nova de Lisboa, Rua da Junqueira 100, Lisbon 1349-008, Portugal; 2Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm tér 9, Szeged 6720, Hungary; 3Unidade de Parasitologia e Microbiologia Médica, Institute of Hygiene and Tropical Medicine, Universidade Nova de Lisboa, Lisbon, Portugal, Rua da Junqueira 100, Lisbon 1349-008, Portugal; 4Institute of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, 6720 Szeged, Hungary Received: November 6, 2013; Revised: December 6, 2013; Accepted: December 7, 2013

Abstract: At this moment, over half million patients suffer from multi-drug resistant tuberculosis (MDR-TB) according to the data from the WHO. A large majority is terminally ill with essentially incurable pulmonary tuberculosis. This herein mini-review provides the experimental and observational evidence that a specific phenothiazine, thioridazine, will contribute to cure any form of drug-resistant tuberculosis. This antipsychotic agent is no longer under patent protection for its initial use. The reader is informed on the recent developments in patenting this compound for “new use” with a special emphasis on the aspects of drug-resistance. Given that economic motivation can stimulate the use of this drug as an antitubercular agent, future prospects are also discussed.

Keywords: Curing of XDR TB, mechanism of killing, mycobacterium tuberculosis, thioridazine. INTRODUCTION The frequency of drug resistance among Mycobacterium tuberculosis infections [1-6] and its progression to multidrug resistance (MDR-TB) [3,7-13] and beyond (extensively drug resistance (XDR-TB) [14-21] and the ill-defined total drug resistance (TDR-TB) [21-27]) continue to increase. Generally, the development and progression of bacterial resistance is primarily due to continued ineffective therapy that results in the selection of antibiotic resistant bacteria in patients within a health care facility [13,15]. In addition, drug resistant strains are frequently transmitted within the hospital setting (nosocomial) [15] and the community [8], which, when further treated ineffectively, lead to higher levels of resistance [28-33]. Ineffective therapy involves inappropriate dosage or prescription of antibiotics [34-38], low exposure or non-compliance by the patient [39-42]. The emergence of antibiotic resistant tuberculosis is an actual problem. Taking into consideration that, in the past three decades, only a few safe and effective anti-tuberculosis agents were developed, the increasing number of MDR, XDR and TDR-TB cases are a significant threat to indigent countries as well as to the affluent countries of the globe. Consequently, new antimicrobial agents of alternative classes and from a wide variety of sources (including natural sources such as the microbial *Address correspondence to this author at the Travel Medicine, Centro de Malária Doenças Tropicais, Institute of Hygiene and Tropical Medicine, Universidade Nova de Lisboa, Rua Junqueira 100, 1349-008 Lisbon, Portugal; Tel: +351213652600; Fax: +351213632105; E-mail: [email protected] 1574-891X/13 $100.00+.00

community, animals and plants, chemical modifications of existing antimicrobial agents and de novo syntheses) have been the focus of intensive research that involved not only anti-tuberculosis agents but also agents against resistant bacterial infections in general. In this mini-review we will focus on the possible use of phenothiazines, agents that are usually employed for non-infectious pathologies. This chemical group has for many years been known to possess a wideranging antimicrobial activity, and has recently been subjected to patenting with “new uses”. In addition, further safe and effective phenothiazine derivatives are also going to be introduced – due to their plethora of mechanisms, emerging resistance to these agents is not anticipated. PHENOTHIAZINES AND TUBERCULOSIS Phenothiazines are heterocyclic compounds from which a great number of medicinal compounds have been derived during the last century, for a broad range of different indications including antimalarial, antihistamic, antipsychotic, antiparkinson and antiemetic use. A. Bernthsen produced the first synthetic phenothiazine in 1883 [43] by the reaction of diarylamine with sulfur. However, the first patent for a phenothiazine dye (methylene blue) was issued earlier in December 15, 1877 to the German company BASF [44]. Later on, the synthesis of mauve dyes by Perkin led to an increase in popularity of dyestuffs and in turn encouraged research on the use of aniline derivatives as dye precursors [44]. The fact that such dye derivatives could be used to stain tissues for microscopy was soon realized. Scientists such as Robert © 2013 Bentham Science Publishers

2 Recent Patents on Anti-Infective Drug Discovery, 2013, Vol. 8, No. 3

Koch and Paul Ehrlich showed that different cell types stained selectively with different dyes due to variances in their chemical structure. Because these dyes could be used as vital stains (staining of living cells), they were soon shown to affect physiological processes, such as the motility of bacteria and protozoa. This encouraged Guttmann and Ehrlich in 1891 to attempt, with success, to treat malaria with methylene blue [45]. Because methylene blue would stain the tissues of the patient receiving this dye, its use was never seriously considered for therapy of malaria. Nevertheless, Roehl, a student of Erhlich’s, continued to work with methylene blue and Plasmodium spp. (organisms that cause malaria) [46], and later, his research was also followed by Schulemann who successfully utilized a phenothiazinium salt for curing malaria in 1932 [47]. However, the phenothiazinium salts were still blue dyes, and this produced a bluish coloration of the skin in the patient. However, because in 1899 the administration of methylene blue to an animal was shown to produce lethargy [48], interest in methylene blue continued, not as a potential antimicrobial agent, but as a possible source for anti-psychotic agents. This became the focus of the biological properties of phenothiazines for the next six decades which eventually resulted in the first neuroleptic chlorpromazine (CPZ), a colorless phenothiazine [49]. The side effects observed during extensive global use of CPZ, suggested that CPZ could also serve as an antimicrobial agent. However, because this era (early 1960’s) was also the “Golden Age of Antibiotics”, there was no interest in the antimicrobial use of CPZ. It was the advent of multidrug resistance of tuberculosis infections during the 1990’s that promoted interest in the earlier studies that showed that CPZ could cure tuberculosis [50-60]. However, because all in vitro studies demonstrated that the concentration of CPZ required for the inhibition of the growth of Mycobacterium tuberculosis was many fold higher than that which could be safely achieved in the patient [61, 62], the use of CPZ for therapy of multi-drug resistant infections was not seriously considered. Moreover, the question of why CPZ could cure patients presenting with tuberculosis at conventional doses employed for therapy of psychosis, was not considered until the studies of Crowle et al. were published in 1992 [63]. These studies showed that exposure of macrophages containing phagocytosed Mycobacterium tuberculosis to clinically reachable concentrations of CPZ resulted in the killing of the mycobacteria. However, because CPZ produces a large number of serious negative side effects [64], the possible use of CPZ for therapy of tuberculosis was still resisted. This situation remained until the studies of Amaral et al showed that thioridazine (TZ) was equipotent to CPZ in vitro [65] and could inhibit the growth of both antibiotic sensitive and MDR Mycobacterium tuberculosis (resistant to isoniazid and rifampicin, the two most effective anti-tuberculosis agents), as well as strains that were resistant to all of the five first line anti-tubercular drugs. TZ is a much milder, and relatively safe neuroleptic as compared to CPZ. And just like CPZ [63], TZ has also been shown to promote the killing of intracellular strains of Mycobacterium tuberculosis regardless of its antibiotic resistance, i.e. multidrug resistant (MDR) [66, 67] or extensively drug resistant (XDR) strains [68]. Although TZ has also been shown to cure mice infected with antibiotic susceptible [69] and multidrug resistant strains of Mycobacterium tuberculosis [70], as shown by a recent

Amaral et al.

study, and confirming what had been predicted earlier by Amaral et al. [65], it does not cure guinea pigs presenting with extracellular infections TZ [71]. Nevertheless, 18 patients infected with XDR-TB have been cured with a combination of TZ and antibiotics to which the strains were initially resistant [72]. Moreover, the in vitro ability of this compound to kill dormant Mycobacterium tuberculosis [73] suggests that TZ could also be used for therapy of latent tuberculosis, alone or as an adjuvant, in combination with existing anti-tuberculosis drugs [74]. Of relevance to these latter studies, Sarathy and co-authors have shown that bactericidal activity of TZ remained comparable in both growing and dormant (nutrient starved) M. tuberculosis [75]. The above cited in vitro studies [73] have support the attempt to employ TZ for monotherapy of essentially terminal patients presenting with XDR TB [76]. Monotherapy with TZ has been shown to improve the quality of life of all patients placed on this “salvage programme”. Within a short period of time (weeks), these patients regained their appetite and gained weight; their night sweats obviated or reduced in frequency; and, due to the neuroleptic activity of TZ, the patients were calmer. However, although TZ can cure XDRTB patients [71, 78], those placed on “salvage therapy” had severe pulmonary damage. Because TZ does not repair or promote repair of the damaged lung parenchyma, all of the patients eventually succumbed to the severity of pulmonary damage caused by the infecting XDR Mycobacterium tuberculosis strain. Nevertheless, because patients were monitored prior to and during therapy with TZ, no detectable cardiac problems such as increased Qt intervals were observed in this [76] or in other studies tghat employed TZ for therapy of XDR-TB [71]. The potential value of employing TZ for therapy of antibiotic resistant infections has received support from the medical press [77-79]. Thioridazine is not the only phenothiazine that has been recommended for therapy of pulmonary tuberculosis. In general, many phenothiazines have been implicated for antitubercular activity [62, 80-86]. Among these are trifluoperazine [87-94], methdilazine [95, 96], promazine [97, 98], promethazine [97, 98], fluphenazin [99], propiomazine [100], and the methylene blue related toluidine blue [101]. Figure 1 summarises the evolution of phenothiazine compounds derived from the parental methylene blue for therapy of pathologies unrelated to tuberculosis, but which compounds also possess antitubercular [44, 48] and/or antimalarial properties [44]. Moreover, derivatives made from any of the phenothiazines that have in vitro activity against Mycobacterium tuberculosis are also active [61, 67, 102, 103], suggesting ample opportunities for patenting of new analogs developed from known, active phenothiazines with even less side effects than those of TZ, as recently suggested by Musuka and co-authors [104]. It is important to mention, that the commercially available phenothiazines such as for example trifluoperazine, methdilazine, promazine, promethazine, fluphenazin and propiomazine are beyond patent protection as initially intended. Nevertheless, these compounds have been patented as adjuvants for the treatment of MDR cancer (patent expired in 2011) [105]; and, right afterwards, a new patent has been filed with a priority date of 28th March, 2012, claiming combination therapy of cancer with a chemotherapeutic agent and a dopamine receptor antagonist

Mechanism by which Thioridazine Enhances Killing of Intracellular

Recent Patents on Anti-Infective Drug Discovery, 2013, Vol. 8, No. 3

N

3

N Me

Me

Me N

S

Me

Me

N

N

N

Me

Me

Paludenblau (antimalarial)

Methylene blue

Antimalarial

N

S

Me

Me

Neuroleptic

H1-antihistamin Me

Me

Et

Me

N

N

HN

Cl

N N Me

Et N

OM e

R

S

N

Mepacrine

Me

R=H: promazine R=Cl: chlorpromazine

N

N

S

S

Promethazine

Methdilazine

Me Me

Et

R N

N

Chloroquine

Me N

N Et

O N

Cl

Me

N

N HN

C F3

SMe

S

Thioridazine

N

CF3

S

R=Me: trifluoperazine R=CH 2 CH 2 OH: fluphenazine

N

S

Propiomazine

Fig. (1). The generation of phenothiazines from methylene blue to clinically important agents.

(e.g. TZ), with a special emphasis on cancers stem cells (CSCs) [106]. Taking into account that intrinsic MDR is considered as one of the key properties of CSCs [107], the subject to be covered is indeed related. According to the MDR, XDR and TDR Mycobacterium tuberculosis, subjects of this herein paper, the initial step for actually reaching those in need has been made: a patent has been published in December, 2007, for the use of TZ and its derivatives for reversing anti-microbial drug resistance [108]. We must note, however, that, despite the six years passed since, we were unable to find any related clinical trials, which would certainly be of outmost importance and urgency in order to proceed towards an effective therapy of highly resistant mycobacterial infections. MECHANISM OF ACTION OF TZ: WHY IT CURES MULTI-DRUG, EXTENSIVELY DRUG RESISTANT AND PROBABLY TOTALLY DRUG RESISTANT TUBERCULOSIS Over-expressed efflux pumps of Mycobacterium tuberculosis render the organism multi-drug resistant [13]. Special attention has been given to those coded by the mmpL7, p55, efpA, mmr, Rv1258c and Rv2459 genes [109]. The activity of these efflux pumps can be suppressed by concentrations of TZ that have no effect on the viability of Mycobacterium tuberculosis rendering the organism susceptible to the antibiotic to which it was initially resistant as a consequence of the over-expression of its efflux pumps [109]. TZ has also been shown to inhibit the activity of the main efflux pumps of bacteria belonging to other species such as Escherichia

coli [110-115], Salmonella enterica serovar Typhimurium [116, 117], Enterobacter aerogenes [118], Staphylococcus aureus [119] as well as the ABCB1 efflux pump of cancer cells [120], suggesting that the activity of TZ against efflux pumps is quite broad. With respect to M. tuberculosis, the use of TZ has been suggested as an adjunct for therapy of MDR-TB infections whose MDR phenotype is due in part or completely to the presence of over-expressed efflux pumps [74, 121]. This suggestion has received further support from the studies of Dutta et al., who demonstrated that TZ has strong inhibitory activity against the genes that code for essential proteins of M. tuberculosis [122-124]. Consequently, we may conclude that the in vitro activity of TZ involves the inhibition of the efflux pumps of M. tuberculosis and that the in vitro exposure of this organism to TZ renders the organism susceptible to antibiotics to which it was initially resistant as a consequence of over-expressed efflux pumps [21]. Phenothiazines such as CPZ, TZ, trifluoperazine, etc., also inhibit the binding of calcium to calcium binding proteins such as calmodulin in eukaryotes [125], and interfere with other proteins involved in the regulation of cellular activity [126]. They inhibit the transport of calcium and potassium systems in eukaryotic cells [127-129] as well as in mycobacteria [89, 130] and E. coli [113]. In fact, in the later case, calcium was shown essential to the continuous activity of the thioridiazine sensitive efflux system [113]. The killing activity of the human macrophage as well as that of the neutrophil is dependent upon the retention of calcium and potassium within the phagolysosome of the cell [131]. These ions are needed for the activity of vacuolar ATPases, which in

4 Recent Patents on Anti-Infective Drug Discovery, 2013, Vol. 8, No. 3

Amaral et al.

turn reduce the pH of the phagolysosome and, consequently, induce hydrolysis of the phagolysosome-entrapped bacterium [130]. Because agents such as oubain that inhibit transport of potassium, enhance the killing of intracellular M. tuberculosis [127], the mechanism by which TZ, an inhibitor of potassium transport, enhances the killing of intracellular M. tuberculosis is supposed to be due to the inhibition of the efflux of these ions from the phagolysosome of the nonkilling human macrophage, thereby activating its killing machinery [127]. This concept of enhancing the killing activity of the non-killing human macrophage of the alveolar structure of the lung is new and significant since it targets the macrophage [132]. Because targeting the intracellular M. tuberculosis with an antibiotic may be soon followed by a mutational response of the bacterium that renders it resistant to the antibiotic, the enhanced intracellular killing capacity of the macrophage promoted by TZ would bypass any mutational response by the organism. Moreover, if TZ is used as an adjunct with antibiotics that are effluxed by the intracellular M. tuberculosis, TZ will also inhibit the efflux of the antibiotic, rendering the organism susceptible. Furthermore, because macrophages are rich in lysosomes that concentrate this phenothiazine [133, 134], the concentration of intracellular TZ may reach levels associated with its minimal inhibitory concentration (MIC) or even its minimum bactericidal concentration [135-138] within the macrophage, without the need of giving those concentrations to the patient, which would otherwise be toxic.

conduct one or more well-designed, double-blind clinical trial(s), which, based on the previous experiences on patients treated with this drug, will most likely confirm the expected efficacy. Unfortunately, as of today, the patent protection of TZ and its derivatives as MDR reversal agents [108] does not appear to be utilized. NeverthelesFs, many other commercially available phenothiazines have in vitro activities as cited in this mini-review and the above mentioned patent focuses on TZ and its derivatives [108], consequentially not covering all prospective candidates. Considering this, several alternative choices are available for patenting under “new use”, which would allow a “fresh start” for the compound to be developed. However, the needed experimental proof that these phenothiazine agents have activity at the pulmonary macrophage of the alveolar unit (the site where the causative organism of pulmonary tuberculosis resides) is still absent.

Other studies also suggest that TZ, as well as other phenothiazine analogs, specifically inhibit respiratory chain related enzymes and affect key cellular mechanisms essential to the survival of M. tuberculosis under low-oxygen conditions (for example during intracellular infection) [84, 139]. Among those are; inhibition of NADH; menaquinone oxidoreductase activity [139], type II NADH-ubiquinone dehydrogenase [84] and the integral membrane succinate dehydrogenase [84], as well as inhibition of oxygen consumption and decrease of intracellular ATP levels of M. tuberculosis [84].

The authors acknowledge the Szeged Foundation for Cancer Research, the European Social Fund (TÁMOP 4.2.2/B-10/1-2010-0012 and TAMOP-4.2.2A-11/1/KONV2012-0035), and the Fundação para a Ciência e a Tecnologia, Portugal (PEsT-OE/SAU/UI0074/2011) for financial support. AM acknowledges the grant SFRH/BPD/81118/2011 provided by the FCT, Portugal.

Based upon the evidence presented, it is reasonable to expect that there are at least four mechanisms which together insure that TZ will be effective against any type of antibiotic resistant pulmonary tuberculosis infection; 1) enhanced killing activity of the human macrophage; 2) inhibition of the over-expressed efflux pump system of the antibiotic resistant Mycobacterium tuberculosis infecting the organism; 3) concentration of TZ to levels compatible with the MIC and MBC of the agent; and 4) inhibition of the oxygen consumption of the mycobacterium.

Consequently, we still urge the medical community to seriously consider thioridazine as an anti-tuberculotic agent, or, as a somewhat less preferable alternative, to conduct rapid development of another legislated phenothiazine drug for therapy of problematic tuberculosis infections. CONFLICT OF INTEREST The authors confirm that this article content has no conflicts of interest. ACKNOWLEDGEMENTS

REFERENCES [1] [2]

[3] [4] [5]

[6]

CURRENT & FUTURE DEVELOPMENTS It is important to consider that economic factors motivate the development of new and effective compounds that can safely cure any form of drug resistant tuberculosis infections. However this process if conducted de novo will take many years, which is not compatible with the urgent needs of those infected with MDR, XDR and TDR-TB now. Thioridazine is safe when used as recommended by others [71, 76, 77] and it is effective [71, 76, 77]. In order to facilitate its legislation for this therapeutic indication, there is an urgent need to

[7] [8] [9] [10]

[11]

Devirgiliis C, Zinno P, Perozzi G. Update on antibiotic resistance in foodborne Lactobacillus and Lactococcus species. Front Microbiol 2013; 4: 301. Fournier PE, Drancourt M, Colson P, Rolain JM, La Scola B, Raoult D. Modern clinical microbiology: New challenges and solutions. Nat Rev Microbiol 2013; 11: 574-85. Mathur P, Singh S. Multidrug Resistance in Bacteria: A serious patient safety challenge for India. J Lab Phys 2013; 5: 5-10. Gatta L, Vakil N, Vaira D, Scarpignato C. Global eradication rates for Helicobacter pylori infection: Systematic review and metaanalysis of sequential therapy. BMJ 2013; 347: f4587. Furuta T, Sugimoto M, Uotani T, Sahara S, Ichikawa H. [Present status and future prospects of third line rescue regimens for H. pylori infection]. Nihon Rinsho 2013; 71: 1404-12. Pagès JM, Amaral L. Mechanisms of drug efflux and strategies to combat them: Challenging the efflux pump of Gram-negative bacteria. Biochim Biophys Acta 2009; 1794: 826-33. Shalimar, Acharya SK. Difficult to treat spontaneous bacterial peritonitis. Trop Gastroenterol 2013; 34: 7-13. Neveur M. [Multi-drug resistant bacteria in the home] Rev Infirm 2013; 192: 24-6. French. Nair GB, Niederman MS. Nosocomial pneumonia: Lessons learned. Crit Care Clin 2013; 29: 521-46. Ji Y, Lei T. Antisense RNA regulation and application in the development of novel antibiotics to combat multidrug resistant bacteria. Sci Prog 2013; 96(Pt 1): 43-60. Belardinelli JM, Morbidoni HR. Recycling and refurbishing old antitubercular drugs: The encouraging case of inhibitors of mycolic acid biosynthesis. Expert Rev Anti Infect Ther 2013; 11: 429-40.

Mechanism by which Thioridazine Enhances Killing of Intracellular [12]

[13]

[14] [15]

[16]

[17] [18]

[19] [20]

[21]

[22] [23] [24] [25]

[26]

[27] [28] [29] [30] [31] [32] [33]

[34] [35] [36]

[37]

Boyanova L, Mitov I. Antibiotic resistance rates in causative agents of infections in diabetic patients: Rising concerns. Expert Rev Anti Infect Ther 2013; 11: 411-20. Viveiros M, Martins M, Rodrigues L, Machado D, Couto I, Ainsa J, et al. Inhibitors of mycobacterial efflux pumps as potential boosters for anti-tubercular drugs. Expert Rev Anti Infect Ther 2012; 10: 983-98. Llamas-González YY, Flores-Valdez MA. [Extensively drugresistant tuberculosis (XDR-TB): Successful therapies used in the clinic to the disease]. Rev Invest Clin 2013; 65: 255-62. Barbier F, Andremont A, Wolff M, Bouadma L. Hospital-acquired pneumonia and ventilator-associated pneumonia: Recent advances in epidemiology and management. Curr Opin Pulm Med 2013; 19: 216-28. Karaiskos I, Galani L, Baziaka F, Giamarellou H. Intraventricular and intrathecal colistin as the last therapeutic resort for the treatment of multidrug-resistant and extensively drug-resistant Acinetobacter baumannii ventriculitis and meningitis: A literature review. Int J Antimicrob Agents 2013; 41: 499-508. Unemo M, Nicholas RA. Emergence of multidrug-resistant, extensively drug-resistant and untreatable gonorrhea. Future Microbiol 2012; 7: 1401-22. Cattoir V, Leclercq R. Twenty-five years of shared life with vancomycin-resistant enterococci: Is it time to divorce?. J Antimicrob Chemother 2013; 68: 731-42. Vila J, Pachón J. Therapeutic options for Acinetobacter baumannii infections: An update. Expert Opin Pharmacother 2012; 13: 231936. Whiley DM, Goire N, Lahra MM, Donovan B, Limnios AE, Nissen MD, et al. The ticking time bomb: Escalating antibiotic resistance in Neisseria gonorrhoeae is a public health disaster in waiting. J Antimicrob Chemother 2012; 67: 2059-61. Amaral L, Viveiros M. Why thioridazine in combination with antibiotics cures extensively drug-resistant Mycobacterium tuberculosis infections. Int J Antimicrob Agents 2012; 39: 376-80. Walsh FM, Amyes SG. Microbiology and drug resistance mechanisms of fully resistant pathogens. Curr Opin Microbiol 2004; 7: 439-44. Berger-Bächi B. Resistance mechanisms of gram-positive bacteria. Int J Med Microbiol 2002; 292: 27-35. Leclercq R. Enterococci acquire new kinds of resistance. Clin Infect Dis 1997; 24 Suppl 1: S80-4. Review. Jensenius M, von der Lippe B, Melby K, Steinbakk M. [The end of antibiotics? Increasing bacterial resistance in global and Norwegian perspective]. Tidsskr Nor Laegeforen 1995; 115: 3382-6. Amaral L, Kristiansen JE. Phenothiazines: An alternative to conventional therapy for the initial management of suspected multidrug resistant tuberculosis. A call for studies. Int J Antimicrob Agents 2000; 14: 173-6. Amaral L. Thioridazine: An old neuroleptic effective against totally drug resistant tuberculosis. Acta Med Port 2012; 25: 118-21. Udwadia ZF, Amale RA, Ajbani KK, Rodrigues CS. Nomenclature of drug-resistant tuberculosis. Lancet Infect Dis 2013; 13: 917. Udwadia ZF. Totally drug-resistant tuberculosis in India: Who let the djinn out? Respirology 2012; 17(5): 741-2. Udwadia ZF. MDR, XDR, TDR tuberculosis: Ominous progression. Thorax 2012; 67: 286-8. Mégraud F. Current recommendations for Helicobacter pylori therapies in a world of evolving resistance. Gut Microbes 2013; 4(6), in press. Stanton TB. A call for antibiotic alternatives research. Trends Microbiol 2013; 21: 111-3. Kimang'a AN. A situational analysis of antimicrobial drug resistance in Africa: Are we losing the battle? Ethiop J Health Sci 2012; 22: 135-43. Rice LB. Mechanisms of resistance and clinical relevance of resistance to -lactams, glycopeptides, and fluoroquinolones. Mayo Clin Proc 2012; 87: 198-208. Deasy J. The antibiotic challenge: Changing clinical management of infections. JAAPA 2009; 22: 22-6. Kollef MH. Broad-spectrum antimicrobials and the treatment of serious bacterial infections: Getting it right up front. Clin Infect Dis 2008; 47 Suppl 1: S3-13. Hayden MK. Insights into the epidemiology and control of infection with vancomycin-resistant enterococci. Clin Infect Dis 2000; 31: 1058-65.

Recent Patents on Anti-Infective Drug Discovery, 2013, Vol. 8, No. 3 [38] [39]

[40] [41]

[42] [43]

[44] [45] [46] [47] [48]

[49] [50] [51] [52]

[53] [54]

[55] [56]

[57]

[58] [59]

[60] [61]

[62] [63]

[64]

5

Nathwani D, Wood MJ. Penicillins. A current review of their clinical pharmacology and therapeutic use. Drugs 1993; 45: 866-94. Hurst JM, Bosso JA. Antimicrobial stewardship in the management of community-acquired pneumonia. Curr Opin Infect Dis 2013; 26: 184-8. Baguley D, Lim E, Bevan A, Pallet A, Faust SN. Prescribing for children -taste and palatability affect adherence to antibiotics: A review. Arch Dis Child 2012; 97: 293-7. Haggard M. Poor adherence to antibiotic prescribing guidelines in acute otitis media--obstacles, implications, and possible solutions. Eur J Pediatr 2011; 170: 323-32. Leung CC, Rieder HL, Lange C, Yew WW. Treatment of latent infection with Mycobacterium tuberculosis: Update 2010. Eur Respir J 2011; 37(3): 690-711. Bernthsen A. Zur Kenntniss des Methylenblau und verwandter Farbstoffe. Berichte der deutschen chemischen Gesellschaft 1883; 16: 2896-904. Wainwright M, Amaral L. The phenothiazinium chromophore and the evolution of antimalarial drugs. Trop Med Int Health 2005; 10: 501-11. Guttmann P & Ehrlich P. U¨ ber die Wirkung des Methylenblau bei Malaria. Berliner Klinische Wochenschrift 1891; 28: 953–6. Roehl W Foundations of chemotherapy. Deutsche Medizinische Wochenschrift 1926; 52, 2017–21. Schulemann W. Synthetic anti-malarial preparations. Proc R Soc Med 1932; 25: 897–905. Kristiansen JE and Amaral L. The potential management of resistant infections with non-antibiotics. J Antimicrob Chemoth 1997; 40: 319–27. Bodoni M. Dellàzione sedative delblue di metilene in vaire forme di psicosi. Clinica Medica Italiana 1899; 24: 217-22. López-Muñoz F, Alamo C, Cuenca E, Shen WW, Clervoy P, Rubio G. History of the discovery and clinical introduction of chlorpromazine. Ann Clin Psychiatry 2005; 17: 113-5. Pleasure H. Chlorpromazine (thorazine) for mental illness in the presence of pulmonary tuberculosis. Psychiatr Q 1956; 30: 23-30. Aguet F, Bossy A, Ducommun P, Favez G. [Non-specific therapeutic stimulation in evolutive pulmonary tuberculosis. I. Properties of chlorpromazine and phenylsemicarbazide]. Schweiz Med Wochenschr 1956; 86: 1239-45. Marchand H and Reuter C. [Phenothiazine derivatives in treatment of pulmonary tuberculosis]. Tuberkulosearzt 1957; 11: 19-27. Shubin H, Heiken CA, Glaskin A, Pennes E, Arsenian J. Chlorpromazine as an adjunct in managing tuberculous patients. Int Rec Med Gen Pract Clin 1957; 170: 369-73. Hyvert M, Le Cloarec J, Carrer J. [Chlorpromazine & atypical tuberculosis in 578 chronic mental patients; Need for combined therapy]. Ann Med Psychol (Paris) 1957; 115: 296-300. Zamfir D, Ionesco G. [Healing of tuberculous caverns of the lung, with persistence of the cavitary image, after treatment by antibiotics associated with chlorpromazine and phenylsemicarbazide]. Poumon Coeur 1958; 14: 1093-7. Hollister LE, Eokenberry DT, Raffel S. Chlorpromazine in nonpsychotic patients with pulmonary tuberculosis. Am Rev Respir Dis 1960; 81: 562-6. Raffel S, Kochan I, Poland N and Hollister LE. The action of chlorpromazine upon Mycobacterium tuberculosis. Am Rev Respir Dis 1960; 81: 555-61. Viktorov IT, Galina S. [Experience with aminazin therapy of mental patients suffering from pulmonary tuberculosis]. Vopr Psikhiatr Nevropatol 1961; 7: 293-304. Kamiska M. [Role of chlorpromazine in the treatment of pulmonary tuberculosis in psychiatric patients]. Folia Med Cracov 1967; 9: 115-43. Molnár J, Béládi I, Földes I. Studies on antituberculotic action of some phenothiazine derivatives in vitro. Zentralbl Bakteriol Orig A 1977; 239: 521-6. Kristiansen JE, Vergmann B. The antibacterial effect of selected phenothiazines and thioxanthenes on slow-growing mycobacteria. Acta Pathol Microbiol Immunol Scand B 1986; 94: 393-8. Crowle AJ, Douvas GS, May MH. Chlorpromazine: A drug potentially useful for treating mycobacterial infections. Chemotherapy 1992; 38(6): 410-9. Amaral L, Kristiansen JE, Viveiros M, Atouguia J. Activity of phenothiazines against antibiotic-resistant Mycobacterium tuberculosis: A review supporting further studies that may elucidate the

6 Recent Patents on Anti-Infective Drug Discovery, 2013, Vol. 8, No. 3

[65]

[66]

[67]

[68]

[69] [70]

[71]

[72]

[73] [74]

[75]

[76]

[77] [78]

[79] [80]

[81] [82]

[83] [84]

[85]

potential use of thioridazine as anti-tuberculosis therapy. J Antimicrob Chemother 2001; 47: 505-11. Amaral L, Kristiansen JE, Abebe LS, Millett W. Inhibition of the respiration of multi-drug resistant clinical isolates of Mycobacterium tuberculosis by thioridazine: Potential use for initial therapy of freshly diagnosed tuberculosis. J Antimicrob Chemother 1996; 38: 1049-53. Ordway D, Viveiros M, Leandro C, Bettencourt R, Almeida J, Martins M, et al. Clinical concentrations of thioridazine kill intracellular multidrug-resistant Mycobacterium tuberculosis. Antimicrob Agents Chemother 2003; 47: 917-22. Martins M, Schelz Z, Martins A, Molnar J, Hajös G, Riedl Z, et al. In vitro and ex vivo activity of thioridazine derivatives against Mycobacterium tuberculosis. Int J Antimicrob Agents 2007; 29: 33840. Martins M, Viveiros M, Ramos J, Couto I, Molnar J, Boeree M, et al. SILA 421, an inhibitor of efflux pumps of cancer cells, enhances the killing of intracellular extensively drug-resistant tuberculosis (XDR-TB). Int J Antimicrob Agents 2009; 33: 479-82. Martins M, Viveiros M, Kristiansen JE, Molnar J, Amaral L. The curative activity of thioridazine on mice infected with Mycobacterium tuberculosis. In vivo 2007; 21: 771-5 van Soolingen D, Hernandez-Pando R, Orozco H, Aguilar D, Magis-Escurra C, Amaral L, et al. The antipsychotic thioridazine shows promising therapeutic activity in a mouse model of multidrug-resistant tuberculosis. PLoS One 2010; 5(9), e12640. Dutta NK, Pinn ML, Zhao M, Rudek MA, Karakousis PC. Thioridazine lacks bactericidal activity in an animal model of extracellular tuberculosis. J Antimicrob Chemother 2013; 68(6): 1327-30 Abbate E, Vescovo M, Natiello M, Cufré M, García A, Gonzalez Montaner P, et al. Successful alternative treatment of extensively drug-resistant tuberculosis in Argentina with a combination of linezolid, moxifloxacin and thioridazine. J Antimicrob Chemother 2012; 67: 473-7. Sohaskey C. Latent tuberculosis: Is there a role for thioridazine?. Recent Pat Antiinfect Drug Discov 2011; 6: 139-46. Alsaad N, Wilffert B, van Altena R, de Lange WCM, van der Werf TS, Kosterink JGW, et al. Potential antimicrobial agents for the treatment of MDR-TB. ERJ 2013; doi:10.1183/09031936.00113713. Sarathy J, Dartois V, Dick T, Gengenbacher M. Reduced drug uptake in phenotypically resistant nutrient-starved nonreplicating Mycobacterium tuberculosis. Antimicrob Agents Chemother 2013; 57(4): 1648-53. Udwadia ZF, Sen T, Pinto LM. Safety and efficacy of thioridazine as salvage therapy in Indian patients with XDR-TB. Recent Pat Antiinfect Drug Discov 2011; 6: 88-91. Amaral L, Udwadia Z, Abbate E, van Soolingen D. The added effect of thioridazine in the treatment of drug-resistant tuberculosis. Int J Tuberc Lung Dis 2012; 16: 1706-2. Amaral L. It is anticipated that as is usually the case, financial motivation will move TZ from its current status “potential antiMDR/XDR agent,” to one that will cure MDR and XDR TB. Foreword. Recent Pat Antiinfect Drug Discov 2011; 6: 76. Boeree MJ. Global clinical trials for the treatment of TB with thioridazine. Recent Pat Antiinfect Drug Discov 2011; 6: 99-103. Warman AJ, Rito TS, Fisher NE, Moss DM, Berry NG, O'Neill PM, et al. Antitubercular pharmacodynamics of phenothiazines. J Antimicrob Chemother 2013; 68: 869-80. Field SK, Fisher D, Jarand JM, Cowie RL. New treatment options for multidrug-resistant tuberculosis. Ther Adv Respir Dis 2012; 6: 255-68. Sharma S, Singh A. Phenothiazines as anti-tubercular agents: Mechanistic insights and clinical implications. Expert Opin Investig Drugs 2011; 20: 1665-76. Gillespie SH, Singh K. XDR-TB, what is it?; how is it treated?; and why is therapeutic failure so high? Recent Pat Antiinfect Drug Discov 2011; 6: 77-83. Boshoff HI, Myers TG, Copp BR, McNeil MR, Wilson MA, Barry CE 3rd. The transcriptional responses of Mycobacterium tuberculosis to inhibitors of metabolism: Novel insights into drug mechanisms of action. J Biol Chem 2004; 279: 40174-84. Ratnakar P, Rao SP, Sriramarao P, Murthy PS. Structureantitubercular activity relationship of phenothiazine-type calmodulin antagonists. Int Clin Psychopharmacol 1995; 10: 39-43.

Amaral et al. [86] [87]

[88]

[89] [90]

[91]

[92] [93]

[94] [95]

[96] [97]

[98]

[99] [100]

[101] [102] [103]

[104]

[105] [106] [107]

[108] [109]

Meindl W, Laske R. [Antimycobacterial properties of phenothiazine type compounds]. Arch Pharm (Weinheim) 1989; 322: 133-6. Advani MJ, Siddiqui I, Sharma P, Reddy H. Activity of trifluoperazine against replicating, non-replicating and drug resistant M. Tuberculosis. PLoS One 2012; 7: e44245. Kunciw DL, Liechty JJ, Mitchell MO, Wan B, Franzblau SG. Structural requirements for the antitubercular quaternized triflupromazine pharmacophore. Bioorg Med Chem Lett 2012; 22: 5679-80. Koul S, Somayajulu A, Advani MJ, Reddy H. A novel calcium binding protein in Mycobacterium tuberculosis--potential target for trifluoperazine. Indian J Exp Biol 2009; 47: 480-8. Gadre DV, Talwar V. In vitro susceptibility testing of Mycobacterium tuberculosis strains to trifluoperazine. J Chemother 1999; 11: 203-6. Gadre DV, Talwar V, Gupta HC, Murthy PS. Effect of trifluoperazine, a potential drug for tuberculosis with psychotic disorders, on the growth of clinical isolates of drug resistant Mycobacterium tuberculosis. Int Clin Psychopharmacol 1998; 13: 129-31. Reddy MV, Nadadhur G, Gangadharam PR. In-vitro and intracellular antimycobacterial activity of trifluoperazine. J Antimicrob Chemother 1996; 37: 196-7. Ratnakar P, Murthy PS. Trifluoperazine inhibits the incorporation of labeled precursors into lipids, proteins and DNA of Mycobacterium tuberculosis H37Rv. FEMS Microbiol Lett 1993; 110: 291-4. Ratnakar P, Murthy PS. Antitubercular activity of trifluoperazine, a calmodulin antagonist. FEMS Microbiol Lett 1992; 76: 73-6. Dutta NK, Mazumdar K, DasGupta A, Dastidar SG. Activity of the phenothiazine methdilazine alone or in combination with isoniazid or streptomycin against Mycobacterium tuberculosis in mice. J Med Microbiol 2009; 58: 1667-8. Chakrabarty AN, Bhattacharya CP, Dastidar SG. Antimycobacterial activity of methdilazine (Md), an antimicrobic phenothiazine. APMIS 1993; 101: 449-54. Bate AB, Kalin JH, Fooksman EM, Amorose EL, Price CM, Williams HM, et al. Synthesis and antitubercular activity of quaternized promazine and promethazine derivatives. Bioorg Med Chem Lett 2007; 17: 1346-8. Bettencourt MV, Bosne-David S, Amaral L. Comparative in vitro activity of phenothiazines against multidrug-resistant Mycobacterium tuberculosis. Int J Antimicrob Agents 2000; 16: 69-71. Molcan J, Floreánová L, Kukucová H, Motlová E, Polák L. Therapeutic effect of fluphenazin in various doses and forms. Act Nerv Super (Praha) 1971; 13: 182-3. Henson EC, Brunson JG. Effects of epinephrine and propiomazine used alone and as a mixture on granuloma formation. J Reticuloendothel Soc 1970; 7: 109-20. Grosse-Schönepauck D. [Toluidine blue--a tuberculostatic substance?]. Dtsch Med Wochenschr 1970; 95: 786-7. Madrid PB, Polgar WE, Toll L, Tanga MJ. Synthesis and antitubercular activity of phenothiazines with reduced binding to dopamine and serotonin receptors. Bioorg Med Chem Lett 2007; 17: 3014-7. Yano T, Li LS, Weinstein E, Teh JS, Rubin H. Steady-state kinetics and inhibitory action of antitubercular phenothiazines on mycobacterium tuberculosis type-II NADH-menaquinone oxidoreductase (NDH-2). J Biol Chem 2006; 281: 11456-63. Musuka S, Srivastava S, Dona CWS, Meek C, Leff R, Pasipanody J, et al. Thioridazine pharmacokinetic - Pharmacodynamic parameters “Wobble” during treatment of Tuberculosis: A theoretical basis for shorter-duration curative monotherapy with congeners. Antimicrob Agents Chemother 2013; 57(12): 5870-7. Bruno, C., Philippe, G. Anticancer compsns. - consists of a cytotoxic agent with a agent to prevent multi: Drug resistance, e.g. in liposome(s). CH681780 (1993) Mickie, B., Eleftherios, S. Combination therapy for the treatment of cancer. WO2013143000 (2013) Moitra, K., Lou, H., Dean, M. Multidrug efflux pumps and cancer stem cells: Insights into multidrug resistance and therapeutic development. Clin Pharmacol Ther 2011; 89: 491-502. Christensen, J.B., Hendricks, O., Kristiansen, J. Thioridazine and derivatives thereof for reversing anti-microbial drug resistance. US20070287702 (2007) Machado D, Couto I, Perdigão J, Rodrigues L, Portugal I, Baptista P, et al. Contribution of efflux to the emergence of isoniazid and multidrug resistance in Mycobacterium tuberculosis. PLoS One 2012; 7(4): e34538.

Mechanism by which Thioridazine Enhances Killing of Intracellular [110]

[111]

[112] [113]

[114]

[115]

[116]

[117] [118]

[119]

[120]

[121]

[122]

[123] [124]

Martins A, Hunyadi A, Amaral L. Mechanisms of resistance in bacteria: An evolutionary approach. Open Microbiol J 2013; 7: 538. Varga ZG, Armada A, Cerca P, Amaral L, Mior Ahmad Subki MA, Savka MA, et al. Inhibition of quorum sensing and efflux pump system by trifluoromethyl ketone proton pump inhibitors. In vivo 2012; 26: 277-85. Martins A, Amaral L. Screening for efflux pump systems of bacteria by the new acridine orange agar method. In vivo 2012; 26(2): 203-6. Martins A, Machado L, Costa S, Cerca P, Spengler G, Viveiros M, et al. Role of calcium in the efflux system of Escherichia coli. Int J Antimicrob Agents 2011; 37: 410-4. Viveiros M, Rodrigues L, Martins M, Couto I, Spengler G, Martins A, et al. Evaluation of efflux activity of bacteria by a semiautomated fluorometric system. Methods Mol Biol 2010; 642: 15972. Paixão L, Rodrigues L, Couto I, Martins M, Fernandes P, de Carvalho CC, et al. Fluorometric determination of ethidium bromide efflux kinetics in Escherichia coli. J Biol Eng 2009; 3: 18. doi: 10.1186/1754-1611-3-18. Spengler G, Rodrigues L, Martins A, Martins M, McCusker M, Cerca P, et al. Genetic response of Salmonella enterica serotype Enteritidis to thioridazine rendering the organism resistant to the agent. Int J Antimicrob Agents 2012; 39: 16-21. Lawler AJ, Ricci V, Busby SJ, Piddock LJ. Genetic inactivation of acrAB or inhibition of efflux induces expression of ramA. J Antimicrob Chemother 2013; 68: 1551-7. Spengler G, Martins A, Schelz Z, Rodrigues L, Aagaard L, Martins M, et al. Characterization of intrinsic efflux activity of Enterococcus faecalis ATCC29212 by a semi-automated ethidium bromide method. In vivo 2009; 23: 81-7. Klitgaard JK, Skov MN, Kallipolitis BH, Kolmos HJ. Reversal of methicillin resistance in Staphylococcus aureus by thioridazine. J Antimicrob Chemother 2008; 62: 1215-21. Spengler G, Molnar J, Viveiros M, Amaral L. Thioridazine induces apoptosis of multidrug-resistant mouse lymphoma cells transfected with the human ABCB1 and inhibits the expression of Pglycoprotein. Anticancer Res 2011; 31: 4201-5. Amaral L, Spengler G, Martins A, Armada A, Handzlik J, KiecKononowicz K, et al. Inhibitors of bacterial efflux pumps that also inhibit efflux pumps of cancer cells. Anticancer Res 2012; 32: 2947-57. Dutta NK, Mazumdar K, Dastidar SG, Karakousis PC, Amaral L. New patentable use of an old neuroleptic compound thioridazine to combat tuberculosis: A gene regulation perspective. Recent Pat Antiinfect Drug Discov 2011; 6: 128-38. Mehra S, Dutta NK, Mollenkopf HJ, Kaushal D. Mycobacterium tuberculosis MT2816 encodes a key stress-response regulator. J Infect Dis 2010; 202: 943-53. Dutta NK, Mehra S, Kaushal D. A Mycobacterium tuberculosis sigma factor network responds to cell-envelope damage by the promising anti-mycobacterial thioridazine. PLoS One 2010; 5: e10069.

Recent Patents on Anti-Infective Drug Discovery, 2013, Vol. 8, No. 3 [125]

[126]

[127] [128]

[129]

[130]

[131]

[132]

[133]

[134] [135]

[136]

[137]

[138]

[139]

7

Mayur YC, Jagadeesh S, Thimmaiah KN. Targeting calmodulin in reversing multi-drug resistance in cancer cells. Mini Rev Med Chem 2006; 6: 1383-9. Sheaves RM. Calmodulin: A multifunctional protein for the control of cellular activity and homeostasis. A review of its physicochemical properties and pharmacological manipulation. Braz J Med Biol Res 1982; 15: 1-9. Martins M, Viveiros M, Amaral L. Inhibitors of Ca2+ and K+ transport enhance intracellular killing of M. tuberculosis by non-killing macrophages. In vivo 2008; 22: 69-75. Wittekindt OH, Schmitz A, Lehmann-Horn F, Hänsel W, Grissmer S. The human Ca2+-activated K+ channel, IK, can be blocked by the tricyclic antihistamine promethazine. Neuropharmacology 2006; 50: 458-67. Rocha JB, Wolosker H, Souza DO, de Meis L. Alteration of Ca2+ fluxes in brain microsomes by K+ and Na+: Modulation by sulfated polysaccharides and trifluoperazine. J Neurochem 1996; 66: 772-8. Reddy PT, Prasad CR, Reddy PH, Reeder D, McKenney K, Jaffe H, et al. Cloning and expression of the gene for a novel protein from Mycobacterium smegmatis with functional similarity to eukaryotic calmodulin. J Bacteriol 2003; 185: 5263-8. Ahluwalia J, Tinker A, Clapp LH, Duchen MR, Abramov AY, Pope S, et al. The large-conductance Ca2+-activated K+ channel is essential for innate immunity. Nature 2004; 427(6977): 853-858. Retraction in: Nature. 2010; 468: 122. Martins M. Targeting the human macrophage with combinations of drugs and inhibitors of Ca2+ and K+ transport to enhance the killing of intracellular multi-drug resistant Mycobacterium tuberculosis (MDR-TB)--a novel, patentable approach to limit the emergence of XDR-TB. Recent Pat Antiinfect Drug Discov 2011; 6: 110-7. Martins M, Viveiros M, Couto I, Amaral L. Targeting human macrophages for enhanced killing of intracellular XDR-TB and MDR-TB. Int J Tuberc Lung Dis 2009; 13: 569-73. Wójcikowski J, Daniel WA. Thioridazine-fluoxetine interaction at the level of the distribution process in vivo. Pol J Pharmacol 2002; 54: 647-54. Daniel WA. Mechanisms of cellular distribution of psychotropic drugs. Significance for drug action and interactions. Prog Neuropsychopharmacol Biol Psychiatry 2003; 27: 65-73. Martins M, Bleiss W, Marko A, Ordway D, Viveiros M, Leandro C, et al. Clinical concentrations of thioridazine enhance the killing of intracellular methicillin-resistant Staphylococcus aureus: An in vivo, ex vivo and electron microscopy study. In vivo 2004; 18: 78794. Ordway D, Viveiros M, Leandro C, Arroz MJ, Molnar J, Kristiansen JE, et al. Chlorpromazine has intracellular killing activity against phagocytosed Staphylococcus aureus at clinical concentrations. J Infect Chemother 2002; 8: 227-31. Ordway D, Viveiros M, Leandro C, Arroz MJ, Amaral L. Intracellular activity of clinical concentrations of phenothiazines including thioridiazine against phagocytosed Staphylococcus aureus. Int J Antimicrob Agents 2002; 20: 34-43. Weinstein EA, Yano T, Li LS, Avarbock D, Avarbock A, Helm D, et al. Inhibitors of type II NADH: Menaquinone oxidoreductase represent a class of antitubercular drugs. Proc Natl Acad Sci U S A 2005; 102(12): 4548-53.