2,606
Views
10
CrossRef citations to date
0
Altmetric
Review

The treatment of melioidosis: is there a role for repurposed drugs? A proposal and review

ORCID Icon, , &
Pages 957-967 | Received 06 Dec 2017, Accepted 29 Jun 2018, Published online: 10 Jan 2019

ABSTRACT

Introduction: Melioidosis is a significant health problem within endemic areas such as Southeast Asia and Northern Australia. The varied presentation of melioidosis and the intrinsic antibiotic resistance of Burkholderia pseudomallei, the causative organism, make melioidosis a difficult infection to manage. Often prolonged courses of antibiotic treatments are required with no guarantee of clinical success.

Areas covered: B. pseudomallei is able to enter phagocytic cells, affect immune function, and replicate, via manipulation of the caspase system. An examination of this mechanism, and a look at other factors in the pathogenesis of melioidosis, shows that there are multiple potential points of therapeutic intervention, some of which may be complementary. These include the directed use of antimicrobial compounds, blocking virulence mechanisms, balancing or modulating cytokine responses, and ameliorating sepsis.

Expert commentary: There may be therapeutic options derived from drugs in clinical use for unrelated conditions that may have benefit in melioidosis. Key compounds of interest primarily affect the disequilibrium of the cytokine response, and further preclinical work is needed to explore the utility of this approach and encourage the clinical research needed to bring these into beneficial use.

1. Introduction

Many pathogens are able to colonize and infect the human host by evading, exploiting, or suppressing the immune response, which leads to a variable spectrum of disease. Sepsis represents one extreme where pathogen-host interactions result in a rapid, biochemically and physiologically violent response with a fatal outcome to the host. There is little opportunity to intervene therapeutically apart from attempting to reduce the pathogen burden as quickly as possible while supporting and correcting physiological derangements. The existence of infection in both chronic and latent disease-states suggests that some form of equilibrium is reached in which the host immune system has accommodated the pathogen. If this equilibrium is not fixed, chronic infection may have profound debilitating effects on the host over time, leading to considerable morbidity and mortality. Latent infection has limited overt effects; however, latent infection potentially represents a pathogenic ‘time-bomb’ typically as the immune system deteriorates with age. Melioidosis, caused by Burkholderia pseudomallei, is an example of such and causes disease that ranges across the entire spectrum from acute to latent [Citation1], and it has been shown that resurgent infections can come from both reinfection and relapse [Citation2].

B. pseudomallei is an important cause of disease in South East Asia, and Northern Australia, where it is a major cause of sepsis [Citation3]. Its geographical distribution, however, is far greater than previously considered due to lack of recognition or under-reporting in some nations [Citation3]; thus, the global burden is unclear but has been predicted to being the cause of as many as 165,000 (95% credible interval 68,000–412,000) and 89,000 (36,000–227,000) deaths [Citation3]. The bacterium B. pseudomallei is associated with soil and fresh water, especially in paddy fields, and causes infection by either direct inoculation following trauma and wound contamination with B. pseudomallei, or via inhalation of contaminated water or soil. Certain individuals appear more susceptible to infection and severe disease, including those with diabetes, established renal failure, and cancer [Citation4]. Management is difficult as B. pseudomallei is intrinsically resistant to many antibiotics and typically treatment requires at least 2 weeks of intravenous antibiotic therapy followed by lengthy oral therapy, often with multiple antibiotics [Citation1,Citation5]. Despite efficacious antibiotic therapy being available, there is still significant mortality and morbidity, and there is no guarantee of clinical success, often due to severe forms of the disease [Citation6]. Furthermore, this brings up the potential for prolonged latent disease, which represents a future problem for the individual infected. B. pseudomallei has also been of significant interest to the biodefence community, due to its attributes of intrinsic antibiotic resistance and its ability to cause severe infections via the respiratory route in healthy individuals [Citation7,Citation8].

This review considers the pathogenesis of B. pseudomallei and considers potential strategies and targets for intervention.

2. Multiple scaled points for therapeutic intervention in melioidosis

In melioidosis, there are a number of points in the disease process where therapeutics could be used to: kill the pathogen, arrest further multiplication and spread, boost, or restore the immune system to kill the pathogen, ameliorate sepsis, and restore normal physiology. There is increasing understanding of the pathogenesis of B. pseudomallei, but gaps in knowledge still remain given the complex nature of the pathogen and considerable variability between strains and in the host response. Some of the bacterial virulence determinants have been described by Stone et al. [Citation9].

2.1. Killing the pathogen

Attempting to kill or cripple a pathogen in the early stages of infection is a tried and tested medical intervention that has been the bedrock of modern medicine and public health for over 200 years in the case of immunization [Citation10] and nearly a century with chemical-based antimicrobial agents [Citation11].

Antimicrobial therapy of melioidosis has been widely researched from in vitro susceptibility testing through to clinical trials with single agents or combinations [Citation12]. However, B. pseudomallei is resistant to many antibiotics, and in the context of a lack of investment into new antibiotic drugs for any bacterial disease, finding a fully efficacious antibiotic is challenging, and this is beginning to have profound effects on healthcare [Citation13].

There are rare reports of new antibiotics [Citation14]. Attempts to develop new antibiotics include the modification of current antibiotics and consideration of antibiotic delivery systems that may offer potential strategies to extend the utility of current antibiotics. For example, dendritic cells have been used as ‘mules’ to take antibiotic directly into granulomas [Citation15], and lipid encapsulation of antibiotics has shown some promise in delivering antibiotics into the lung [Citation16].

Prophylactic immunization primes the immune system to fight off infection. Despite considerable work in animal models with putative candidates, a melioidosis vaccine for use in the clinic remains elusive for the time being [Citation17,Citation18].

Passive immunization introduces preformed antibody from a donor, which could be derived by purification from convalescent serum from a previously infected individual, or from an animal source that has been deliberately challenged with a pathogen or parts of a pathogen [Citation19]. Recently, the use of monoclonal antibodies and de-speciated antibodies (to prevent hypersensitivity reactions) have provided improvements to this strategy, but again in the case of melioidosis, there has been little progress beyond proof-of-concept that such an immunotherapeutic approach can be efficacious using in vivo models [Citation20Citation22]. In contrast, there has been commercial development of a pipeline of immuno-therapeutics for other infections [Citation23]. Perhaps one reason why antibody-based therapies have not been considered more is that antibodies need to be able to bind to bacteria, while in the extracellular space, B. pseudomallei is not only capable of intracellular infection but is likely able to move between cells without entering the intracellular space (reviewed in [Citation24]). This would likely impact on the ability for antibody-based therapies to fully clear infection without assistance from antibiotics that accumulate within cells and/or the cell mediated immune response.

2.2. Prevention of host cell subversion

B. pseudomallei is phagocytozed by granulocytes but resists intracellular killing. Within the cell, the bacteria are able to multiply and spread to adjacent cells [Citation25]. Macrophages can kill B. pseudomallei; however, they need to be in an interferon-γ rich environment [Citation26] and, as discussed later here, are subverted by other mechanisms.

The opportunities to prevent this exploitation of the host immune system start with preventing bacterial uptake by phagocytic cells, or expression or blockade of bacterial virulence factors. Small molecule inhibitors to blockade the function of virulence determinants have been, and continue to be, widely used in the development of antiviral therapies [Citation27]. To our knowledge, to date there are no small molecule inhibitors that have been licensed against bacterial virulence determinants. However, there is significant interest in this area, and examples of broad spectrum targets of interest include type-III secretion systems [Citation28], quorum sensing [Citation29], and fatty acid synthesis [Citation30]. Each of these are important in the virulence of B. pseudomallei [Citation31Citation33]. The roles of type-III secretion systems to deliver effectors and quorum sensing to alter gene expression based on cell density are self-evident; however, the role of fatty II acid synthesis might need more explanation. Fatty acids are an essential building block of all life, and critically (for the purposes of antimicrobial drug development) the enzymes that perform this function are very different comparing mammals and bacteria [Citation34].

The premise that neutrophils are important in the immune response to melioidosis [Citation35] led to clinical trials using Granulocyte Colony Stimulating Factor (G-CSF, stimulator for neutrophil release) in conjunction with conventional therapies [Citation36]. Unfortunately, G-CSF therapy did not have a significant impact on outcome. While G-CSF therapy increases the numbers of neutrophils (providing their production is not currently at maximum), it is argued that a lack of activation contributed to the apparent clinical failure. Mice deficient in osteopontin, a pleiotropic cytokine that is chemotactic for neutrophils, was shown to be more resistant to B. pseudomallei infection [Citation37].

As already mentioned, B. pseudomallei is able to resist intracellular killing. The process known as xenophagy, effectively an autophagic process, is subverted by many pathogens, and autophagy itself is believed to be important in the formation of tumors and chronic inflammatory conditions. There is considerable research interest in manipulating autophagy pathways as a tumor therapy, and stimulation of autophagy is potentially a strategy that could be employed for melioidosis [Citation38].

2.3. Prevent/mitigate manipulation of caspase systems

A key enzyme in the cell death pathway is caspase-1, which when activated causes cell death and cleaves the pro-inflammatory cytokines IL-1β and IL-18. The role of caspase-1 in HIV has attracted considerable interest. The pyroptotic cell death pathway was found to be subverted in HIV infection, promoting CD4 T-cell destruction, and hence the promotion of inflammation through inhibition of caspase-1 (increasing apoptosis and reducing pyroptosis) was found to be beneficial [Citation39]. In the case of melioidosis, it is known that ablation of caspase-1 will render mice highly susceptible, and this is believed to be because critical interferon-γ production is reduced without the cleaving of IL-18 [Citation40,Citation41]. This would suggest that pyroptosis is important in raising inflammation against B. pseudomallei. The main outputs of pyroptosis (IL-1β and IL-18) were investigated for their utility in protecting against B. pseudomallei infection [Citation42]. The role and importance of IL-18 in inducing IFN-γ was confirmed; however, IL-1β was found to deleterious. Activation of pyroptosis was found to be dependent upon Nod-Like Receptor (NLR) C4 [Citation42,Citation43]. It was also found that NLRP3 contributed to IL-1β production but in a pyroptosis independent manner [Citation42]. This seemingly is the cause of the imbalance where IL-1β is detrimental. Type III secretion is important in the interaction between B. pseudomallei and NLRC4 [Citation44]. Recently, capsase-6 has also been shown to be important in response to B. pseudomallei, where capsase-6 deficient macrophages were shown to be less bactericidal and capsase-6 mice exhibited higher sensitivity alongside raise IL-1β and IL-10 production [Citation45].

2.4. Rebalancing of the cytokine network

An effective immune response to infectious disease must be carefully counterbalanced to effectively kill and eradicate the invading pathogen, yet it must be ‘reined in’ to prevent collateral damage to the host. Immune pathways permitted to continue unopposed by appropriate feedback is the basis of the so-called ‘cytokine storm’ leading to the sepsis pathway, which is associated with high mortality. In the pathogenesis of melioidosis, IL-18 is activated via caspase-1 along with IL-1β; IL-18 then stimulates IFN-γ, which restricts bacterial multiplication. IL-1β can lead to excessive neutrophil recruitment, which in the case of melioidosis provides advantage to the bacteria. Murine studies have shown the benefit of IL-18 production (via IFN-γ) in contrast to worse outcomes associated with overproduction of IL-1β [Citation42]. Further evidence of the detrimental effects of IL-1β is the apparent survival advantage of a small sub-population of diabetic patients with melioidosis that receive the antidiabetic drug glyburide [Citation46]. Study of the patients’ transcriptome showed reduction of many inflammatory markers (including IL-1), correlating with the effects of the drug’s prevention of activation of cryopyrin inflammasome [Citation47]. The benefits of glyburide were also seen in a murine model of infection with reduced bacterial load and inflammation (notably IL-1β) [Citation48]. Another study, however, found that treatment with glyburide worsened disease associated with reducing inflammation [Citation49]. The latter study found that glyburide had an immune-tempering effect that was driven via an IRAK-M specific route, reducing inflammation including IL-1 levels. Further work has shown that glyburide acts upon neutrophilic interaction with B. pseudomallei [Citation50] where again it was demonstrated to reduce IL-1 release. As previously mentioned, IL-1β inhibits IFN-γ release, and IFN-γ is likely to be very beneficial. Murine studies have shown that supplementation with exogenous IFN-γ has a synergistic effect with ceftazidime and improves survival [Citation51], further establishing that IL-1β can be deleterious in melioidosis. Collectively, this evidence suggests that reduction in IL-1β with enhancement of IFN-γ signaling could be of benefit therapeutically.

2.5. Prevention of immunopathology

One retrospective study in Thailand suggested that approximately 9% of individuals with melioidosis will die from sepsis [Citation6]. Given individual variations in responses to pathogens and the complexity of the host response, there may be multiple mechanisms, as yet not all understood, which lead to sepsis. Immuno-therapy to prevent severe sepsis against a variety of pathogens has universally failed to meet expectations [Citation52], and the recognition of the diverse nature of sepsis requires stratification not only of the causative microbe but aspects of the host [Citation53] so that therapy may have to be tailored to the individual [Citation54]. Nonetheless, the science resulting from the sepsis field may still present new therapeutic options for severe melioidosis.

3. Potential therapeutic opportunities

3.1. Therapeutics to kill/inhibit the pathogen

An interesting endeavor might be to select candidates from the pharmacopeia, looking for effects on B. pseudomallei growth and viability as has been attempted for other infections. Kruszewska and colleagues investigated greater than 160 pharmaceuticals for antimicrobial properties against strains of Staphylococcus aureus, Escherichia coli, Pseudomonas aeruginosa, and Candida albicans [Citation55]. Later, Younis and colleagues test 727 pharmaceuticals for activity against strains of Enterococcus faecium, S. aureus, Klebsiella pneumoniae, Acinetobacter baumannii, P. aeruginosa, and Enterobacter cloacae [Citation56]. Two drugs were shown to have efficacy in an in vivo mouse model of S. aureus infection [Citation56]. Between these two studies, numerous in-use pharmaceuticals have been identified to have direct antimicrobial effects () and may be worthy of investigation against B. pseudomallei.

Table 1. In-use pharmaceuticals found to have some direct antimicrobial effects in studies [Citation55,Citation56].

Table 2. Inhibition of IL-1β can be achieved by a number of drugs.

One strategy is to have B. pseudomallei targeted for killing by the host using of antibodies. There are potential limitations of using antibody therapies to treat an infection that seems to have an intracellular life style. Despite this, monoclonal therapies can have beneficial effects even after pulmonary bacterial challenge [Citation20]. However, such a therapy would need clinical development from first principals, negating the advantage of many of the other therapies discussed in this article.

3.2. Therapeutics to prevent host cell subversion

Given the importance of IL-18 and IFN-γ, and their apparent effectiveness in vivo when given with ceftazidime, their potential use is obvious. Recombinant IFN-γ-1b (Immukin®) is licensed for use in chronic granulomatous disease and severe malignant osteopetrosis and use off-licence in mild to severe atopic dermatitis and has also been trialed for the treatment of Friedreich’s ataxia. Treatment with rIL-18 may play a role in the pathogenesis of adenomyosis, Hashimoto’s thyroiditis and upregulates amyloid-beta production in neurons. It has, however, shown potential in the therapy for age-related wet macular degeneration [Citation57].

Neutrophil (and other phagocytic cell) activation may be a potential pathway as a therapeutic target. In keeping with all bacterial infections, immune reactions to melioidosis are provoked by the interaction of pathogen-associated molecular patterns (PAMPs) and host cell pattern recognition receptors (PRRs), and the resulting signaling cascade. There has been considerable interest in this interaction in the development of vaccine adjuvants, particularly those biasing Th1 responses, and also as adjuvants in cancer therapy [Citation58]. The idea is not new: Coley used a bacterial lysate consisting of killed Streptococcus pyogenes and Serratia marcescens causing regression in some tumors at the end of the 19th century [Citation59]. Continued trials with ‘Coley’s toxin’ have been controversial, but they continue albeit with far more refined and defined moieties such as the CpG oligo-deoxy-nucleotides [Citation60,Citation61]. CpG ODN 1826 was found to be protective in murine models of melioidosis [Citation62]. This finding was confirmed later where an intranasal deliver of class C CpG 2 days prior to an intranasal challenge of B. pseudomallei provided measurable benefits to the animals [Citation63]. There are a number of other adjuvants that are in clinical use such as MF59 [Citation64] that might have benefited in treating an entrenched B. pseudomallei infection by encouraging IFN-γ release and cellular killing.

B. pseudomallei has a variety of virulence factors that provide potential targets for vaccines and chemotherapeutic agents. Vaccine options have been considered extensively [Citation17]. Chemotherapeutic targets include the type-III secretion system [Citation28,Citation65]; quorum sensing [Citation29], and the type II fatty acid biosynthesis pathway [Citation30].

A multitude of small compounds have been considered as putative inhibitors of the Type III secretion system, and these are summarized below from two review articles [Citation28,Citation65]. However, to our knowledge, none of these compounds are in clinical use and would therefore require significant development prior to use in melioidosis patients. The only inhibitor of type III secretion that has been used in clinical trials is an antibody fragment (KB001-A) found to interact with the type III secretion system of P. aeruginosa [Citation66]. Releases in the public media, however, revealed reported commercial concerns with regard to success criteria, and it has since been shelved. Variants of thiazolidinones (a class of small molecule that has been very useful in drug discovery [Citation67]) have been used clinically for unrelated purposes (rosiglitazone and pioglitazone, which are both insulin sensitizers); however, it is clear that the structure of these molecules can to be optimized for effect in inhibiting type III secretion systems in both bacteria from the genus Salmonella and Yersinia [Citation68]. With regard to compounds that can inhibit type II fatty acid synthesis, Anthranilic acid has been used clinically previously [Citation69] and has chemistry associated with other clinically used products. Indole-3-carbinol, which is similar (SB418001), has also been used clinically [Citation70]. Also dithiolethiones have been studied in some detail, more so in the form of Oltipraz, which was tolerated in humans with some adverse effects [Citation71]. 4-aminopyridime (or famridine) bears similarity to the aminopyridine that targets bacteria [Citation72] and has been shown to be tolerated in a clinical trial [Citation73]. The easiest fatty acid synthesis inhibitor to repurpose might be Isoniazid, which is already in clinical use for the treatment of Mycobacterium infection. Another drug that is worthy of investigation is fosmidomycin. This drug is currently used as an antimalarial and targets DXP reductoisomerase, a critical enzyme in the nonmevalonate pathway. This drug also targets this pathway in at least some Gram negative bacteria [Citation74].

B. pseudomallei bacteria are able to escape phagosomes via the type III secretory system and spread intracellularly; avoiding cell mediated killing processes that require compartmentalization of the bacteria. Autophagy is a collective term for the targeted degradation and recycling of cellular components. Autophagy inducing compounds offer an alternative approach as a host-directed therapy to promote killing of intracellular bacteria (xenophagy) by targeting the host autophagic pathway. Examples of autophagy promoting drugs being investigated as host-directed therapies include vitamin D [Citation75], metformin [Citation76,Citation77], statins [Citation78], verapamil [Citation79Citation85], carbamezapine [Citation86,Citation87], and valproic acid [Citation86,Citation87]. Verapamil hydrochloride is a calcium channel antagonist that shows potential as a host-directed autophagy therapy to reduce intracellular Mycobacterium tuberculosis [Citation79Citation85]. Norverapamil is the metabolite of verapamil and shows a similar ability to reduce intracellular bacteria in vitro [Citation79,Citation80]. Verapamil is being investigated as an inhalable therapy for inhalational M. tuberculosis with the verapamil-antibiotic combination demonstrating an enhanced antibacterial effect [Citation88]. Carbamazepine and valproic acid have both demonstrated enhanced M. tuberculosis clearance from RAW 264.7 macrophages in vitro [Citation86]. The diabetic drug metformin is an autophagy inducer and also shows potential as a host-directed autophagy therapy for M. tuberculosis [Citation77]. Given the similarities in intracellular infection characteristics between M. tuberculosis and B. pseudomallei, the autophagy compounds currently being investigated for M. tuberculosis could be an approach for use as a host-directed melioidosis therapy. We have seen antimicrobial effects of verapamil and norverapamil against intracellular Burkholderia thailandensis within an in vitro macrophage assay (A. Taylor, unpublished data).

The receptor COX-2 is a potential target for treatment of melioidosis [Citation89] as the downstream effector from COX-2, prostaglandin E2 (PGE2), aided intracellular survival of B. pseudomallei by biasing macrophage activation toward the alternative pathway. COX-2 inhibitors protected some mice in a murine model of respiratory melioidosis, and it is presumed that this protection was driven by increased classical activation of macrophages [Citation89]. It is also possible that some of the protection afforded by COX-2 inhibition was driven by a reduction in IL-1 as COX-2 can reduce the release of IL-1β via the NLR family, pyrin domain-containing 3 (NLRP3) inflammasome [Citation90]. Currently, celecoxib is the only specific inhibitor of COX-2 that is licensed for use; however, non-steroidal anti-inflammatory drugs (NSIADs) such as diclofenac, aspirin, acetorphan, and ibuprofen all affect COX-2, although these are less specific in their action. It is of note that some members of this class of drugs have antibacterial activity () and may interfere with the type II fatty acid biosynthesis pathway.

3.3. Therapeutics to rebalance the cytokine network

The potential therapeutic use of IL-18 and IFN-γ has already been discussed. At present, compounds that act on the inflammasome are restricted to pharmacological tools to elucidate the pathway. As understanding increases, the opportunity for therapeutic intervention may become apparent.

The antidiabetic glyburide [Citation91] clearly has potential in treating melioidosis given the findings from patients who are on the drug, and it has shown (in combination with glucantime) to have some activity against Leishmania major, another intracellular pathogen where IFN-γ production is critical for host defense [Citation91,Citation92].

Thalidomide and its derivatives pomalidomide and lenalidomide enhance CD8+ T cell activity [Citation93], NK cell activity, and the release of IFN-γ [Citation94]. Moreover, these drugs inhibit the release of IL-1, TNF-α, and IL-6 [Citation95]. These effects are all potentially positive for the host during melioidosis, and these cytokines are prognostic markers for melioidosis disease severity [Citation96]. CD8+ T cells and NK cells are thought to contribute to host defense by rapid release of IFN-γ [Citation97]. Moreover, the cytotoxic functions of NK cells and CD8+ T cells are likely to contribute to the control of B. pseudomallei infection in its intracellular phase. In experimental settings, thalidomide has been considered for the treatment of AIDS [Citation98,Citation99] and tuberculosis [Citation100]. In mice, thalidomide has been demonstrated to mitigate K. pneumoniae disease by reducing inflammation [Citation101]. Also, in a hepatitis patient cohort, thalidomide successfully reduced signs of liver damage [Citation102]. In the clinical setting, thalidomide is mainly used to treat leprosy [Citation103]. Thalidomide therapy is not without disadvantages; it has had a long and troubled past with regard to its chiral form’s teratogenetic affect [Citation104] and is a known contraindication of a drug of interest to this manuscript (anakinra).

Another approach that has recently shown promise is the use of the anticancer therapeutic hexamethylene bisacetamide (HMBA) [Citation105]. This therapeutic was found to increase the production of IL-12 and IFN-γ but not TNF-α, IL-6, and IL-8 in human peripheral blood mononuclear cells infected with B. pseudomallei.

Anakinra is a recombinant form of human IL-1 receptor antagonist (RA), a protein that competes with IL1 for binding to the IL-1R, thereby blocking signal transduction. IL-1RA has been previously considered for the tempering of severe sepsis, and, despite interesting preliminary data, the clinical trial was halted. A non-statistically significant 9% enhancement of survival was observed in the IL-1RA treated sepsis patients [Citation106]. Anakinra has been shown to have efficacy in a murine model of respiratory melioidosis [Citation42,Citation107]. Other drugs exist that perform the same function (table 2).

Chloroquine, which has a long history of use in the treatment of malaria, also inhibits IL-1β release [Citation108] as well as conferring other immune suppressive functions [Citation109] that are being considered in trials in autoimmune disease. It also increases the pH on the phagolysosome to the detriment of intracellular pathogens, and hence it is used in chronic Query (Q)-fever. Chloroquine also has been demonstrated to show similar suppressive effects on the virulence of B. pseudomallei through mediation of pH in the phagolysosome [Citation110].

Recent studies have indicated that the Damage Associated Molecular Pattern (DAMP) HMGB1 may also contribute to immune dysregulation, and two studies have demonstrated that antibody ablation of this target can be beneficial to a murine host under conditions of B. pseudomallei infection [Citation111,Citation112]. As yet there are no known drugs that are in clinical use that target HMGB1 or the pathways immediately below HMGB1.

3.4. Therapeutics to prevent, ameliorate, or treat immunopathology

The key focus in preventing sepsis is by attempting to quantify predictive derangements in physiology in a timely manner. Unfortunately, the presenting signs and symptoms vary considerably with age and are often non-specific, mimicked by many other medical and surgical problems. A lack of a consistent and specific and sensitive diagnostic system exacerbates the problem. Preventing the immune system spiraling into a catastrophic positive feedback loop has been an area of sepsis research for many years, with many established and novel therapeutics being investigated and trialed and in the main failing [Citation53]. Meta-analysis of immunoglobin therapy suggests benefit in sepsis, but as for antibiotic administration, immunotherapy has to be given in a timely manner, and further research is needed [Citation113]. Recombinant activated protein C (also known as autoprothrombin IIA and blood coagulation factor XIV) treatment was initially promising; however, meta-analysis has shown this to be ineffectual [Citation114]. Recently, beta-blockers have been considered [Citation115].

At present, antibiotics, supportive therapy to maintain blood pressure, end-stage organ oxygenation and perfusion are the mainstay in the care of the sepsis patient [Citation116]. Hypoxic injury to the brain, kidneys, liver, or heart has obvious deleterious consequences. Furthermore, injury to the intestine may exacerbate an already desperate situation, where there may be a sudden release of huge amounts of PAMPs into the blood from translocation of the gut flora. Moreover, as the integrity of the gut is lost due to hypoxic damage, release of proteolytic enzymes from the gut can contribute to pathology [Citation117].

Ultimately, what can be critical in the therapeutic success of treating melioidosis is the quality of the supportive care offered. This can include a range of healthcare, such as the supportive therapy discussed above, to ensuring antibiotic therapy is taken. The effectiveness of supportive care is demonstrated by a comparison of melioidosis case fatality rates between countries with different healthcare systems []. While better therapeutics might lead to faster recovery for all melioidosis patients, they are unlikely to translate to improved survival in areas with good healthcare (where most individuals survive) but may reduce the period of treatment and thereby reduce the likelihood of complications and acquired resistance in the microbiota driven by prolonged exposure to antibiotics.

4. Conclusion

In conclusion, the pathogenesis of melioidosis (and indeed, other bacterial infections) provides a number of targets and processes that may be amenable to therapeutic intervention. Traditional strategies have been discussed briefly with the premise of a number of experimental pharmacological agents exploiting new avenues for intervention such as the type III secretion pathway, fatty acid synthesis and quorum sensing, some of which may have close analogs currently in clinical use. It is clear from this review that a number of drugs, particularly from certain classes, notably NSAIDs, cancer chemotherapeutics, and disease-modifying drugs and various others such as rapamycin, thalidomide (and derivatives), chloroquine, and glyburide, have the potential to be repurposed for melioidosis therapy. Further work is needed to evaluate these candidates. However, these drugs have a wide spectrum of biological effects, not only those they were designed to fulfill but also some side-effects. For this reason, consideration needs to be given to the overall pharmacological effect. For example, anesthetic drugs that are bactericidal may also cause numerous extraneous effects.

With the constant threat of antimicrobial resistance, and its potential to render treatable diseases untreatable, the melioidosis community has an opportunity to be the forerunners of new anti-infective therapies that might have a benefit across the whole gambit of infectious diseases. Repurposing drugs that are in clinical use to correct/re-direct the immune response represents a real opportunity that is potentially low in risk and might deliver rapidly. Moreover, this approach has the potential to be impervious to standard microbial resistance mechanisms, as the whole spectrum of the immune response will be brought to bear on the invading B. pseudomallei. We therefore urge that these anti-infective strategies be tested further in preclinical studies to build the case for use in the treatment of sufferers of this infection.

5. Expert commentary

The question of profitability continues to be an albatross around the neck of people trying to devise any new anti-infective strategies. Melioidosis is a fascinating example of this because it impacts two groups of people.

The first group are those people living in endemic areas who suffer from this disease, who are often from low-income groups working on the land and thus exposed to the bacteria. If we were to compare melioidosis to tuberculosis (which does have a significant amount of investment), the former is disadvantaged for funding, for a number of reasons. Firstly, the number of at-risk people is simply lower. Secondly, there is no evidence for horizontal transfer, indicating that only those performing high-risk activities are susceptible and those in more affluent areas are not. This means that any novel therapeutic is unlikely to be either profitable for pharma or affordable for those who suffer from this disease.

The other groups interested in melioidosis are the biodefence and military medicine communities, who have the capability to repurpose or support the licensure of some anti-infective therapies.

These communities are working together to accelerate development of therapeutics that is affordable for sufferers/at-risk people and suitable for biodefence. So far, no novel therapeutic has transitioned beyond the ‘bench.’ However, a significant benefit is that there is now a substantial body of scientific research that informs on the biology of melioidosis and possible targets to treat it.

The issue with leveraging this information to create novel ways to treat endemic infection and provide biodefence is the lack of investment in the translation of laboratory science into clinically useable therapeutics. This brings us to the second part of the review that addresses whether much of the cost of translation might be bypassed by repurposing drugs used to treat unrelated infections. We draw the reader’s attention to a great variety of in-use pharmaceutics that have significant potential in the treatment of melioidosis. Drug repurposing would have the advantage of using drugs for which patents have expired and that can be produced cheaply as ‘generics.’ However, this route will almost surely require public funding; there is little profit to be made from expanding the use of a ‘generic’ pharmaceutical.

In conclusion, this review addresses how a treatment for melioidosis might be arrived at within a short time frame, in the hope that investment might be found for this strategy.

6. Five-year review

While melioidosis continues to kill people, it is incumbent on the scientific community to transition therapeutic concepts into real benefits. We hope to see either new antimicrobial drugs (perhaps the fruits of labors to counter the rise of antibiotic resistance) or a return to repurposing therapies from other areas of medicine as a real option to treat this disease.

Key issues

  • Melioidosis is a tropical disease caused by infection with the Gram negative bacteria B. pseudomallei. These infections are problematic due to the intrinsic resistance of B. pseudomallei to many antibiotics.

  • Melioidosis research suffers from the disease not affecting affluent groups, but some additional funding is leveraged because it is considered a biothreat agent.

  • Repurposing therapies used elsewhere in medicine might provide a cost-effective way to transition recent findings regarding pathogenesis into the clinic.

  • Disease is a result of biological interactions that can be scaled from the bacteria’s capacity to grow in the body and how it causes immune dysregulation and ultimately sepsis and potentially death.

  • There are varying amounts of evidence that a variety of ‘off-the-shelf’ medicines can target these biological processes with potentially beneficial effects.

  • More work in this area is needed.

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosure

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

The manuscript was funded by the UK Ministry of Defence.

References

  • Wuthiekanun V, Peacock SJ. Management of melioidosis. Expert Rev Anti Infect Ther. 2006 Jun;4(3):445–455.
  • Maharjan B, Chantratita N, Vesaratchavest M, et al. Recurrent melioidosis in patients in northeast Thailand is frequently due to reinfection rather than relapse. J Clin Microbiol. 2005 Dec;43(12):6032–6034.
  • Limmathurotsakul D, Golding N, Dance DAB, et al. Predicted global distribution of burkholderia pseudomallei and burden of melioidosis [Letter]. Nat Microbiol. 2016 01 11;1:15008. online.
  • Limmathurotsakul D, Peacock SJ. Melioidosis: a clinical overview. Br Med Bull. 2011;99:125–139.
  • Estes DM, Dow SW, Schweizer HP, et al. Present and future therapeutic strategies for melioidosis and glanders. Expert Rev Anti Infect Ther. 2010 Mar;8(3):325–338.
  • Churuangsuk C, Chusri S, Hortiwakul T, et al. Characteristics, clinical outcomes and factors influencing mortality of patients with melioidosis in southern Thailand: A 10-year retrospective study. Asian Pac J Trop Med. 2016 Mar;9(3):256–260.
  • Gilad J, Harary I, Dushnitsky T, et al. Burkholderia mallei and burkholderia pseudomallei as bioterrorism agents: national aspects of emergency preparedness. Isr Med Assoc Journal: IMAJ. 2007 Jul;9(7):499–503.
  • Schweizer HP. When it comes to drug discovery not all gram-negative bacterial biodefence pathogens are created equal: burkholderia pseudomallei is different. Microb Biotechnol. 2012 Sep;5(5):581–583.
  • Stone JK, DeShazer D, Brett PJ, et al. Melioidosis: molecular aspects of pathogenesis. Expert Rev Anti Infect Ther. 2014 Dec;12(12):1487–1499.
  • Hajj Hussein I, Chams N, Chams S, et al. Vaccines through centuries: major cornerstones of global health. Frontiers in Public Health. 2015;3:269.
  • Aminov R. History of antimicrobial drug discovery - major classes and health impact. Biochem Pharmacol. 2017 Jun;1(133):4–19.
  • Dance D. Treatment and prophylaxis of melioidosis. Int J Antimicrob Agents. 2014 Apr;43(4):310–318.
  • Laxminarayan R, Matsoso P, Pant S, et al. Access to effective antimicrobials: a worldwide challenge. Lancet (London, England). 2016 Jan 9;387(10014):168–175.
  • Zipperer A, Konnerth MC, Laux C, et al. Human commensals producing a novel antibiotic impair pathogen colonization. Nature. 2016 Jul 28;535(7613):511–516.
  • Montes-Worboys A, Brown S, Regev D, et al. Targeted delivery of amikacin into granuloma. Am J Respir Crit Care Med. 2010 Dec 15;182(12):1546–1553.
  • Merchant Z, Buckton G, Taylor KM, et al. A new era of pulmonary delivery of nano-antimicrobial therapeutics to treat chronic pulmonary infections. Curr Pharm Des. 2016;22(17):2577–2598.
  • Silva EB, Dow SW. Development of burkholderia mallei and pseudomallei vaccines. Front Cell Infect Microbiol. 2013;3:10.
  • Titball RW, Burtnick MN, Bancroft GJ, et al. Burkholderia pseudomallei and burkholderia mallei vaccines: are we close to clinical trials? Vaccine. 2017 Oct 20;35(44):5981–5989.
  • Graham BS, Ambrosino DM. History of passive antibody administration for prevention and treatment of infectious diseases. Curr Opin HIV AIDS. 2015 May;10(3):129–134.
  • AuCoin DP, Reed DE, Marlenee NL, et al. Polysaccharide specific monoclonal antibodies provide passive protection against intranasal challenge with burkholderia pseudomallei. PloS One. 2012;7(4):e35386.
  • Zhang S, Feng SH, Li B, et al. In vitro and in vivo studies of monoclonal antibodies with prominent bactericidal activity against burkholderia pseudomallei and burkholderia mallei. Clinical Vaccine Immunology: CVI. 2011 May;18(5):825–834.
  • Jones SM, Ellis JF, Russell P, et al. Passive protection against burkholderia pseudomallei infection in mice by monoclonal antibodies against capsular polysaccharide, lipopolysaccharide or proteins. J Med Microbiol. 2002 Dec;51(12):1055–1062.
  • Czaplewski L, Bax R, Clokie M, et al. Alternatives to antibiotics-a pipeline portfolio review. Lancet Infect Dis. 2016 Feb;16(2):239–251.
  • Allwood EM, Devenish RJ, Prescott M, et al. Strategies for intracellular survival of burkholderia pseudomallei. Front Microbiol. 2011 08 22;2:170.
  • Willcocks SJ, Denman CC, Atkins HS, et al. Intracellular replication of the well-armed pathogen burkholderia pseudomallei. Curr Opin Microbiol. 2016 Jan;21(29):94–103.
  • Miyagi K, Kawakami K, Saito A. Role of reactive nitrogen and oxygen intermediates in gamma interferon-stimulated murine macrophage bactericidal activity against burkholderia pseudomallei. Infect Immun. 1997 Oct;65(10):4108–4113.
  • Debing Y, Jochmans D, Neyts J. Intervention strategies for emerging viruses: use of antivirals. Curr Opin Virol. 2013 Apr;3(2):217–224.
  • Gu L, Zhou S, Zhu L, et al. Small-molecule inhibitors of the type iii secretion system. Molecules (Basel, Switzerland). 2015;20(9):17659–17674.
  • Reuter K, Steinbach A, Helms V. Interfering with bacterial quorum sensing. Perspect Medicin Chem. 2016;8:1–15.
  • Miesel L, Greene J, Black TA. Genetic strategies for antibacterial drug discovery. Nat Reviews Genet. 2003 Jun;4(6):442–456.
  • Kang W-T, Vellasamy KM, Chua E-G, et al. Functional characterizations of effector protein bipc, a type iii secretion system protein, in burkholderia pseudomallei pathogenesis. J Infect Dis. 2015 Mar 1;211(5):827–834.
  • Gutierrez MG, Pfeffer TL, Warawa JM. Type 3 secretion system cluster 3 is a critical virulence determinant for lung-specific melioidosis. PLoS Negl Trop Dis. 2015 Jan;9(1):e3441.
  • Cummings JE, Kingry LC, Rholl DA, et al. The Burkholderia pseudomallei enoyl-acyl carrier protein reductase FabI1 is essential for in vivo growth and is the target of a novel chemotherapeutic with efficacy. Antimicrob Agents Chemother. 2014;58(2):931–935.
  • Heath RJ, White SW, Rock CO. Inhibitors of fatty acid synthesis as antimicrobial chemotherapeutics. Appl Microbiol Biotechnol. 2002 May;58(6):695–703.
  • Easton A, Haque A, Chu K, et al. A critical role for neutrophils in resistance to experimental infection with Burkholderia pseudomallei. J Infect Dis. 2007 Jan 1;195(1):99–107.
  • Cheng AC, Limmathurotsakul D, Chierakul W, et al. A randomized controlled trial of granulocyte colony-stimulating factor for the treatment of severe sepsis due to melioidosis in Thailand. Clinical Infectious Diseases: an Official Publication of the Infectious Diseases Society of America. 2007 Aug 1;45(3):308–314.
  • van der Windt GJ, Wiersinga WJ, Wieland CW, et al. Osteopontin impairs host defense during established gram-negative sepsis caused by Burkholderia pseudomallei (melioidosis). PLoS Negl Trop Dis. 2010 Aug 31;4:(8)
  • Devenish RJ, Lai SC. Autophagy and burkholderia. Immunol Cell Biol. 2015 Jan;93(1):18–24.
  • Doitsh G, Galloway NL, Geng X, et al. Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection. Nature. 2014 Jan 23;505(7484):509–514.
  • Breitbach K, Sun GW, Kohler J, et al. Caspase-1 mediates resistance in murine melioidosis. Infect Immun. 2009 Apr;77(4):1589–1595.
  • Sun GW, Lu J, Pervaiz S, et al. Caspase-1 dependent macrophage death induced by Burkholderia pseudomallei. Cell Microbiol. 2005 Oct;7(10):1447–1458.
  • Ceballos-Olvera I, Sahoo M, Miller MA, et al. Inflammasome-dependent pyroptosis and IL-18 protect against Burkholderia pseudomallei lung infection while IL-1beta is deleterious. PLoS Pathog. 2011 Dec;7(12):e1002452.
  • West TE, Myers ND, Chantratita N, et al. NLRC4 and TLR5 each contribute to host defense in respiratory melioidosis. PLoS Negl Trop Dis. 2014 Sep;8(9):e3178.
  • Bast A, Krause K, Schmidt IH, et al. Caspase-1-dependent and -independent cell death pathways in Burkholderia pseudomallei infection of macrophages. PLoS Pathog. 2014 Mar;10(3):e1003986.
  • Bartel A, Gohler A, Hopf V, et al. Caspase-6 mediates resistance against Burkholderia pseudomallei infection and influences the expression of detrimental cytokines. PloS One. 2017;12(7):e0180203.
  • Koh GC, Maude RR, Schreiber MF, et al. Glyburide is anti-inflammatory and associated with reduced mortality in melioidosis. Clinical Infectious Diseases: an Official Publication of the Infectious Diseases Society of America. 2011 Mar 15;52(6):717–725.
  • Lamkanfi M, Mueller JL, Vitari AC, et al. Glyburide inhibits the Cryopyrin/Nalp3 inflammasome. J Cell Biol. 2009 Oct 5;187(1):61–70.
  • Koh GC, Weehuizen TA, Breitbach K, et al. Glyburide reduces bacterial dissemination in a mouse model of melioidosis. PLoS Negl Trop Dis. 2013;7(10):e2500.
  • Liu X, Foo G, Lim WP, et al. Sulphonylurea usage in melioidosis is associated with severe disease and suppressed immune response. PLoS Negl Trop Dis. 2014 Apr;8(4):e2795.
  • Kewcharoenwong C, Rinchai D, Utispan K, et al. Glibenclamide reduces pro-inflammatory cytokine production by neutrophils of diabetes patients in response to bacterial infection. Sci Rep. 2013;3:3363.
  • Propst KL, Troyer RM, Kellihan LM, et al. Immunotherapy markedly increases the effectiveness of antimicrobial therapy for treatment of Burkholderia pseudomallei infection. Antimicrob Agents Chemother. 2010 May;54(5):1785–1792.
  • Marshall JC. Sepsis: rethinking the approach to clinical research. J Leukoc Biol. 2008 Mar;83(3):471–482.
  • Marshall JC. Why have clinical trials in sepsis failed? Trends Mol Med. 2014 Apr;20(4):195–203.
  • Pinheiro Da Silva F, Cesar Machado MC. Personalized medicine for sepsis. Am J Med Sci. 2015 Nov;350(5):409–413.
  • Kruszewska H, Zareba T, Tyski S. Search of antimicrobial activity of selected non-antibiotic drugs. Acta Pol Pharm. 2002 Nov-Dec;59(6):436–439.
  • Younis W, Thangamani S, Seleem MN. Repurposing non-antimicrobial drugs and clinical molecules to treat bacterial infections. Curr Pharm Des. 2015;21(28):4106–4111.
  • Doyle SL, Ozaki E, Brennan K, et al. IL-18 attenuates experimental choroidal neovascularization as a potential therapy for wet age-related macular degeneration. Sci Transl Med. [2014 Apr 2];6(230):230ra44.
  • Cooper CL, Davis HL, Morris ML, et al. CPG 7909, an immunostimulatory TLR9 agonist oligodeoxynucleotide, as adjuvant to Engerix-B HBV vaccine in healthy adults: a double-blind phase I/II study. J Clin Immunol. 2004 Nov;24(6):693–701.
  • Coley WB. The treatment of malignant tumors by repeated inoculations of erysipelas. with a report of ten original cases. 1893. Clin Orthop Relat Res. 1991 Jan;(262):3–11.
  • Vollmer J, Krieg AM. Immunotherapeutic applications of CpG oligodeoxynucleotide TLR9 agonists. Adv Drug Deliv Rev. 2009 Mar 28;61(3):195–204.
  • Freidag BL, Melton GB, Collins F, et al. CpG oligodeoxynucleotides and interleukin-12 improve the efficacy of Mycobacterium bovis BCG vaccination in mice challenged with M. Tuberculosis. Infection and Immunity 2000 May;68(5):2948–2953.
  • Wongratanacheewin S, Kespichayawattana W, Intachote P, et al. Immunostimulatory CpG oligodeoxynucleotide confers protection in a murine model of infection with Burkholderia pseudomallei. Infect Immun. 2004 Aug;72(8):4494–4502.
  • Judy BM, Taylor K, Deeraksa A, et al. Prophylactic application of CpG oligonucleotides augments the early host response and confers protection in acute melioidosis. PloS One. 2012;7(3):e34176.
  • Black S. Safety and effectiveness of MF-59 adjuvanted influenza vaccines in children and adults. Vaccine. 2015 Jun 8;33(Suppl 2):B3–5.
  • Duncan MC, Linington RG, Auerbuch V. Chemical inhibitors of the type three secretion system: disarming bacterial pathogens. Antimicrob Agents Chemother. 2012 Nov;56(11):5433–5441.
  • Warrener P, Varkey R, Bonnell JC, et al. A novel anti-PcrV antibody providing enhanced protection against Pseudomonas aeruginosa in multiple animal infection models. Antimicrob Agents Chemother. 2014 Aug;58(8):4384–4391.
  • Kaur Manjal S, Kaur R, Bhatia R, et al. Synthetic and medicinal perspective of thiazolidinones: A review. Bioorg Chem. 2017;75:406–423.
  • Felise HB, Nguyen HV, Pfuetzner RA, et al. An inhibitor of gram-negative bacterial virulence protein secretion. Cell Host Microbe. 2008 Oct 16;4(4):325–336.
  • Shioda H. A double blind controlled trial of N-(3ʹ,4ʹ-dimethoxycinnamoyl) anthranilic acid on children with bronchial asthma. N-5ʹ Study Group in Children. Allergy. 1979 Aug;34(4):213–219.
  • Naik R, Nixon S, Lopes A, et al. A randomized phase II trial of indole-3-carbinol in the treatment of vulvar intraepithelial neoplasia. International Journal of Gynecological Cancer: Official Journal of the International Gynecological Cancer Society. 2006 Mar-Apr;16(2):786–790.
  • Zhang Y, Munday R. Dithiolethiones for cancer chemoprevention: where do we stand? Mol Cancer Ther. 2008;7(11):3470–3479.
  • Miller WH, Seefeld MA, Newlander KA, et al. Discovery of aminopyridine-based inhibitors of bacterial enoyl-acp reductase (FabI). J Med Chem. [2002 07 01];45(15):3246–3256.
  • Judge SI, Bever CT Jr. Potassium channel blockers in multiple sclerosis: neuronal Kv channels and effects of symptomatic treatment. Pharmacology & Therapeutics. 2006 Jul;111(1):224–259.
  • Davey MS, Tyrrell JM, Howe RA, et al. A promising target for treatment of multidrug-resistant bacterial infections. Antimicrob Agents Chemother. 2011;55(7):3635–3636. . PubMed PMID: PMC3122435
  • Wallis RS, Zumla A. Vitamin D as adjunctive host-directed therapy in tuberculosis: A systematic review. Open Forum Infectious Diseases. 2016 Sep;3(3):ofw151.
  • Wallis RS, Hafner R. Advancing host-directed therapy for tuberculosis. Nat Reviews Immunol. 2015 Apr;15(4):255–263.
  • Degner NR, Wang JY, Golub JE, et al. Metformin use reverses the increased mortality associated with diabetes mellitus during tuberculosis treatment. Clinical Infectious Diseases: an Official Publication of the Infectious Diseases Society of America. 2018 Jan 6;66(2):198–205.
  • Dutta NK, Bruiners N, Pinn ML, et al. Statin adjunctive therapy shortens the duration of TB treatment in mice. J Antimicrob Chemother. 2016 Jun;71(6):1570–1577.
  • Abate G, Ruminiski PG, Kumar M, et al. New verapamil analogs inhibit intracellular mycobacteria without affecting the functions of mycobacterium-specific t cells. Antimicrob Agents Chemother. 2015 Dec 7;60(3):1216–1225.
  • Adams KN, Szumowski JD, Verapamil RL. its metabolite norverapamil, inhibit macrophage-induced, bacterial efflux pump-mediated tolerance to multiple anti-tubercular drugs. J Infect Dis. 2014 Aug 1;210(3):456–466.
  • de Knegt GJ, van Der Meijden A, de Vogel CP, et al. Activity of moxifloxacin and linezolid against Mycobacterium tuberculosis in combination with potentiator drugs verapamil, timcodar, colistin and SQ109. Int J Antimicrob Agents. 2017 Mar;49(3):302–307.
  • Demitto Fde O, Do Amaral RC, Maltempe FG, et al. In vitro activity of rifampicin and verapamil combination in multidrug-resistant Mycobacterium tuberculosis. PloS One. 2015;10(2):e0116545.
  • Gupta A, Pant G, Mitra K, et al. Inhalable particles containing rapamycin for induction of autophagy in macrophages infected with Mycobacterium tuberculosis. Mol Pharm. 2014 Apr 7;11(4):1201–1207.
  • Gupta S, Tyagi S, Almeida DV, et al. Acceleration of tuberculosis treatment by adjunctive therapy with verapamil as an efflux inhibitor. Am J Respir Crit Care Med. 2013 Sep 1;188(5):600–607.
  • Gupta S, Tyagi S, Bishai WR. Verapamil increases the bactericidal activity of bedaquiline against Mycobacterium tuberculosis in a mouse model. Antimicrob Agents Chemother. 2015 Jan;59(1):673–676.
  • Schiebler M, Brown K, Hegyi K, et al. Functional drug screening reveals anticonvulsants as enhancers of mTOR-independent autophagic killing of Mycobacterium tuberculosis through inositol depletion. EMBO Mol Med. 2015 Feb;7(2):127–139.
  • Juarez E, Carranza C, Sanchez G, et al. Loperamide restricts intracellular growth of mycobacterium tuberculosis in lung macrophages. Am J Respir Cell Mol Biol. 2016 Dec;55(6):837–847.
  • Parumasivam T, Chan JG, Pang A, et al. In vitro evaluation of inhalable verapamil-rifapentine particles for tuberculosis therapy. Mol Pharm. 2016 Mar 7;13(3):979–989.
  • Asakrah S, Nieves W, Mahdi Z, et al. Post-exposure therapeutic efficacy of COX-2 inhibition against Burkholderia pseudomallei. PLoS neglected tropical diseases. 2013 May 9;7(5):e2212.. * Demonstrates the utility of targeting Cox-2 (a highly druggable target) for treatment of melioidosis; 2013.
  • Hua KF, Chou JC, Ka SM, et al. Cyclooxygenase-2 regulates NLRP3 inflammasome-derived IL-1beta production. J Cell Physiol. 2015 Apr;230(4):863–874.
  • Ponte-Sucre A, Mendoza-Leon A, Moll H. Experimental leishmaniasis: synergistic effect of ion channel blockers and interferon-gamma on the clearance of Leishmania major by macrophages. Parasitol Res. 2001 Jan;87(1):27–31.
  • Padron-Nieves M, Diaz E, Machuca C, et al. Glibenclamide modulates glucantime activity and disposition in Leishmania major. Exp Parasitol. 2009 Apr;121(4):331–337.
  • Haslett PA, Hanekom WA, Muller G, et al. Thalidomide and a thalidomide analogue drug costimulate virus-specific CD8+ T cells in vitro. J Infect Dis. 2003 Mar 15;187(6):946–955.
  • Davies FE, Raje N, Hideshima T, et al. Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma. Blood. 2001 Jul 1;98(1):210–216.
  • Bodera P, Stankiewicz W. Immunomodulatory properties of thalidomide analogs: pomalidomide and lenalidomide, experimental and therapeutic applications. Recent Pat Endocr Metab Immune Drug Discov. 2011 Sep;5(3):192–196.
  • Simpson AJ, Smith MD, Weverling GJ, et al. Prognostic value of cytokine concentrations (tumor necrosis factor-alpha, interleukin-6, and interleukin-10) and clinical parameters in severe melioidosis. J Infect Dis. 2000 Feb;181(2):621–625.
  • Lertmemongkolchai G, Cai G, Hunter CA, et al. Bystander activation of CD8+ T cells contributes to the rapid production of IFN-gamma in response to bacterial pathogens. Journal of Immunology (Baltimore, Md: 1950). 2001 Jan 15;166(2):1097–1105.
  • Brunel AS, Reynes J, Tuaillon E, et al. Thalidomide for steroid-dependent immune reconstitution inflammatory syndromes during AIDS. AIDS (London, England). 2012 Oct 23;26(16):2110–2112.
  • Lim H, Kane L, Schwartz JB, et al. Lenalidomide enhancement of human T cell functions in human immunodeficiency virus (HIV)-infected and HIV-negative CD4 T lymphocytopenic patients. Clin Exp Immunol. 2012 Aug;169(2):182–189.
  • Fu LM, Fu-Liu CS. Thalidomide and tuberculosis. The international journal of tuberculosis and lung disease. The Official Journal of the International Union against Tuberculosis and Lung Disease. 2002 Jul;6(7):569–572.
  • Kumar V, Harjai K, Chhibber S. Thalidomide treatment modulates macrophage pro-inflammatory function and cytokine levels in Klebsiella pneumoniae B5055 induced pneumonia in BALB/c mice. Int Immunopharmacol. 2010 Jul;10(7):777–783.
  • Milazzo L, Biasin M, Gatti N, et al. Thalidomide in the treatment of chronic hepatitis C unresponsive to alfa-interferon and ribavirin. Am J Gastroenterol. 2006 Feb;101(2):399–402.
  • Vargesson N. Thalidomide-induced teratogenesis: history and mechanisms. Birth Defects Research Part C, Embryo Today: Reviews. 2015 Jun;105(2):140–156.
  • Gamage AM, Lee KO. Anti-cancer drug hmba acts as an adjuvant during intracellular bacterial infections by inducing type I IFN through STING. J Immunol. 2017 Oct 1;199(7):2491–2502.
  • Opal SM, Fisher CJ Jr., Dhainaut JF, et al. Confirmatory interleukin-1 receptor antagonist trial in severe sepsis: a phase III, randomized, double-blind, placebo-controlled, multicenter trial. the interleukin-1 receptor antagonist sepsis investigator group. Crit Care Med. 1997 Jul;25(7):1115–1124.
  • Weehuizen TA, Lankelma JM, De Jong HK, et al. Therapeutic administration of a monoclonal anti-il-1beta antibody protects against experimental melioidosis. Shock (Augusta, Ga). 2016 Nov;46(5):566–574.
  • Jang CH, Choi JH, Byun MS, et al. Chloroquine inhibits production of TNF-alpha, IL-1beta and IL-6 from lipopolysaccharide-stimulated human monocytes/macrophages by different modes. Rheumatology (Oxford, England). 2006 Jun;45(6):703–710.
  • Savarino A, Shytaj IL. Chloroquine and beyond: exploring anti-rheumatic drugs to reduce immune hyperactivation in HIV/AIDS. Retrovirology. 2015;12:51.
  • Chua J, Senft JL, Lockett SJ, et al. pH alkalinization by chloroquine suppresses pathogenic burkholderia type 6 secretion system 1 and multinucleated giant cells. Infect Immun. 2017 Jan;85(1):e00586–16.
  • Charoensup J, Sermswan RW, Paeyao A, et al. High HMGB1 level is associated with poor outcome of septicemic melioidosis. International Journal of Infectious Diseases: IJID: Official Publication of the International Society for Infectious Diseases. 2014;28:111–116.
  • Laws TR, Clark GC, D’Elia RV. Immune profiling of the progression of a BALB/c mouse aerosol infection by Burkholderia pseudomallei and the therapeutic implications of targeting HMGB1. International Journal of Infectious Diseases: IJID: Official Publication of the International Society for Infectious Diseases. 2015 Nov;40:1–8.
  • Busani S, Damiani E, Cavazzuti I, et al. Intravenous immunoglobulin in septic shock: review of the mechanisms of action and meta-analysis of the clinical effectiveness. Minerva Anestesiol. 2016 May;82(5):559–572.
  • Marti-Carvajal AJ, Sola I, Lathyris D, et al. Human recombinant activated protein C for severe sepsis. The Cochrane Database of Systematic Reviews. 2012;3:Cd004388.
  • Sanfilippo F, Santonocito C, Morelli A, et al. Beta-blocker use in severe sepsis and septic shock: a systematic review. Curr Med Res Opin. 2015;31(10):1817–1825.
  • Chawla S, DeMuro JP. Current controversies in the support of sepsis. Curr Opin Crit Care. 2014 Dec;20(6):681–684.
  • Altshuler AE, Kistler EB, Schmid-Schonbein GW. Autodigestion: proteolytic degradation and multiple organ failure in shock. Shock (Augusta, Ga). 2016 May;45(5):483–489.
  • Cheng AC, West TE, Limmathurotsakul D, et al. Strategies to reduce mortality from bacterial sepsis in adults in developing countries. PLoS Med. 2008 Aug 19;5(8):e175.