Next Article in Journal
A Full-State Reliability Analysis Method for Remanufactured Machine Tools Based on Meta Action and a Markov Chain Using an Exercise Machine (EM) as an Example
Next Article in Special Issue
In Vitro and In Silico Studies of Antimicrobial Saponins: A Review
Previous Article in Journal
Antimicrobial Resistance of Heterotrophic Bacteria and Enterobacteriaceae Inhabiting an Anthropogenic-Affected River Stretch in Bulgaria
Previous Article in Special Issue
Identification of an Antimicrobial Protease from Acanthamoeba via a Novel Zymogram
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Alternatives in the Fight against Tuberculosis: Possible Targets for Resistant Mycobacteria

by
Eduardo Rodríguez-Bustamante
1,2,
Saúl Gómez-Manzo
3,
Alvaro De Obeso Fernández del Valle
2,
Roberto Arreguín-Espinosa
1,
Clara Espitia-Pinzón
4 and
Eden Rodríguez-Flores
1,*
1
Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
2
Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey 64849, Mexico
3
Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico
4
Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
*
Author to whom correspondence should be addressed.
Processes 2023, 11(9), 2793; https://doi.org/10.3390/pr11092793
Submission received: 12 August 2023 / Revised: 9 September 2023 / Accepted: 13 September 2023 / Published: 20 September 2023

Abstract

:
Tuberculosis (TB) is a bacterial disease that remains a global health threat due to the millions of deaths attributed to it each year. The emergence of drug resistance has exacerbated and further increased the challenges in the fight against this illness. Despite the preventive measures using the application of the Bacillus Calmette-Guérin vaccine, the desired immunization outcome is not as high as expected. Conventional TB treatments exhibit serious limitations, such as adverse effects and prolonged duration, leading to a pressing need for alternative and more effective treatment options. Despite significant efforts, it took nearly four decades for diarylquinoline to become the most recently approved medicine for this disease. In addition, various possibilities, such as the usage of medications used for many other conditions (repurposed drugs), have been explored in order to speed up the process of achieving faster outcomes. Natural compounds derived from various sources (microorganisms, plants, and animals) have emerged as potential candidates for combating TB due to their chemical diversity and their unique modes of action. Finally, efforts towards the generation of novel vaccines have received considerable attention. The goal of this paper was to perform an analysis of the current state of treating drug-resistant TB and to evaluate possible approaches to this complicated challenge. Our focus is centered on highlighting new alternatives that can be used to combat resistant strains, which have potentiated the health crisis that TB represents.

1. The Disease Known as Tuberculosis and Relevant Aspects Associated to It

At the end of October 2022, the World Health Organization (WHO) published its Global Tuberculosis Report, pointing out that worldwide, this disease represents the 13th leading cause of death and the major infectious killer after SARS-CoV-2/COVID-19 and above HIV/AIDS [1]. Tuberculosis (TB) is caused by Mycobacterium tuberculosis (Mtb), whose pathogenic role was demonstrated in 1882 by the research conducted by Koch. TB is transmitted from person to person through the respiratory route, commonly affecting the lungs, but other tissues can also be damaged [2]. Estimations indicate that about a quarter of the world population is infected with Mtb during their lifetimes; however, most people will not develop TB, and in some cases, the infection can even be cleared [1]. Active TB might be manifested with mild symptoms during the first months, developing into a cough with sputum and sometimes blood, chest pains, general fatigue, weight loss, fever, and nocturnal sweating [1]. According to work on TB by Guinn and Rubin [3], published in a “frequently asked questions” (FAQs) format, pneumonia caused by Mtb exhibits two distinctive features: (1) the impairment of the lung tissue rather than the airways and (2) granuloma. Nonetheless, a major challenge against this disease relies on the bacterial capacity to remain latent and surprisingly persist in most infected individuals for a long time.
One distinguishing and, at the same time, outstanding feature of mycobacteria relies on the chemical nature and architecture of their cell wall. Mycobacterial cell walls are mainly composed of a peptidoglycan layer, mycolic acids, and arabinogalactan [4], rendering, in turn, their acid-fastness capacity and the low permeability to antimicrobial drugs (broad-spectrum antibiotics except for rifampicin). The knowledge about the acid-fast reaction of mycobacteria has mostly been derived from the Ziehl-Neelsen stain [5]. This structure plays a critical role for this class of bacteria, as crucial processes such as protection against hostile elements, mechanical resistance of the cells, solute and protein transport, and cell adhesion via the recognition of receptors take place here [6]. In addition to its role in the viability of Mtb, the chemical structure of its cell wall, particularly the peptidoglycan, renders unique molecular features that are important during dormancy [7].
The difference between active and latent TB, better identified as TB disease and latent TB infection, respectively, resides in the symptoms exhibited by the patients and their capacity to infect other people. In a journey through time (over two centuries), Behr et al. [8] addressed the confusion generated by different concepts related to latent TB and highlighted the relevance of using consistent definitions for research, treatments, and public health affairs. In this sense, patients with TB disease require multiple antibiotics/drug treatments for many months, depending on the regimen. Drug susceptible TB is treated by a 6-month standard course of 4 antibiotics: isoniazid (H), rifampicin (R), ethambutol (E), and pyrazinamide (Z); all four drugs are used for the first 2 months, and the remaining 4 are followed by H and R [1]. According to CDC [9], in the United States, treatments for TB disease shift in time length as follows: (1) 4-month rifapentine-moxifloxacin and (2) 6 or 9-month RIPE (rifampicin, isoniazid, pyrazinamide, and ethambutol). Together with rifabutin (Rbt), these four antibiotics represent the first line of drugs against TB [10]. Even though TB is a treatable disease, several complications are associated with its treatment. Adverse reactions to the medications include nausea, vomiting, loss of appetite, skin rashes, liver toxicity, or peripheral neuropathy, side effects that can affect treatment adherence and may require modifications to the drug regimen [11].
Until now, the only prevention against TB is yielded by the licensed but controversial BCG (Bacillus Calmette-Guérin) vaccine. The controversy generated by this vaccine encompasses several topics, such as (1) each country’s policy for its application, (2) candidates for receiving the preventive treatment, (3) conferred time of protection, and (4) efficacy in immunogenicity. This last issue represents a crucial point in the research focused on developing new drug therapies for fighting TB. BCG was developed in 1921 at the Pasteur Institute from an attenuated bacterial strain of Mycobacterium bovis, starting clinical immunization with it this same year [12]. Since 1924, M. bovis BCG has been distributed globally; however, the endemic occurrence of TB, in addition to differences within culture conditions, led to the emergence of genetic variation among strains. At present, more than 20 BCG variants exist worldwide, all with a common genetic loss: the deletion of a region of difference 1 (RD1), an important feature involved in attenuation since the initial M. bovis strain [13]. Today, it is well known that phenotypic and genotypic heterogeneity of the different strains is related to their efficacy for protecting against TB [14].
In summary, the delay in the elimination of TB is caused by multiple factors, such as the complex biology of Mtb and the complications of treatment that have led to the generation of resistant strains. The lack of readily available preventive interventions and the rise in illnesses among vulnerable individuals round out the list. Therefore, the aim of this review paper centers on the state of the art in the development of alternative drugs/medications and new generation vaccines to combat this disease.

2. Tuberculosis as a Public Health Problem: An Old Challenge Still Present Due to Drug Resistance

From historical records, i.e., Egyptian and Peruvian mummies, it has been postulated that Mtb has infected human beings since very ancient times [12]. TB has a significant economic impact at the individual and social levels: (1) affecting children, the elderly, migrants, prisoners, and people living in poverty and (2) exacerbating health disparities [15,16]. In high-burden countries, many individuals with TB are asymptomatic in a latent state for many years and can be reactivated to cause disease [2]. Another setback of this illness relies on the fact that it affects vulnerable populations such as immunocompromised patients, which include cases positive for HIV/AIDS, malnourished individuals, and/or people with underlying health problems such as diabetes and cancer.
In 1995, “Directly Observed Treatment” (DOT) was launched by WHO to ensure proper medication intake as prescribed and to monitor responses to treatment. Van Deun et al. [17] reported on a standardized program known as “the Bangladesh Regimen” consisting of kanamycin, gatifloxacin, prothionamide, high-dose isoniazid, clofazimine, pyrazinamide, and ethambutol for 9 to 11 months and attaining an 88% of success. Nevertheless, combining several drugs over long periods continues to be a struggle in completing treatment and enhancing the generation of resistant strains. Finally, in 2014, a new action plan called “End TB Strategy” was implemented. However, inadequate health services interfere with the timely diagnosis, treatment of the disease, and follow-up of TB cases, often leading to scheme failure and illness relapse and hindering the success of WHO’s strategies [1].
Drug-resistant TB (DR-TB), which can be broadly divided into intrinsic and acquired, poses an escalating worldwide threat to health security. Intrinsic resistance comprises the innate capacity to render antibacterial compounds less effective; meanwhile, in the specific case of Mtb, the acquired resistance is a result of specific chromosomal mutations [18]. Key mechanisms of drug resistance include loss of enzymatic action in the activation of pro-drugs, drug targets, overexpression of the drug target, and overexpression of the efflux pump. At a molecular level, research focused on finding genes responsible for drug resistance to isoniazid (katG), rifampin (rpoB), pyrazinamide (pncA), and quinolone (gyrA) has been conducted [19]. The emergence of drug-resistant TB, such as multidrug-resistant TB (MDR-TB) and extensively drug-resistant TB (XDR-TB), complicates treatment and control efforts even further. Medicines used to treat drug-resistant strains often exhibit more toxicity, need longer treatment, and are more likely to present adverse effects. To make things worse, drug-resistant strains increase costs; for example, in patients with DR-TB, hospitalization was longer for cases exhibiting this feature compared to susceptible TB: 202 +/− 130 vs. 123 +/− 81 days (mean +/− SD; p = 0.015), respectively [20].
MDR-TB is defined as resistance to the two main first-line anti-TB drugs (isoniazid and rifampicin), while XDR-TB includes resistance to many second-line drugs, such as bedaquiline, linezolid, moxifloxacin, levofloxacin, clofazimine, cycloserine, para-aminosalicylic, propylthiouracil, and amikacin [21]. In addition, XDR-TB encompasses strains resistant to any fluoroquinolones and at least one of three injectable second-line drugs (amikacin, kanamycin, and/or capreomycin) [22,23]. MDR-TB requires the use of specific alternative TB chemotherapy regimens. These regimens involve the combination of second-line anti-tuberculosis drugs, which are typically more expensive and toxic and require longer durations. Alternatively, fluoroquinolones, better tolerated by patients, are central to the treatment of MDR-TB [24]. Prevention of transmission of MDR-TB and XDR-TB- requires robust TB control actions, including early diagnosis, appropriate treatment regimens, infection control measures, and monitoring systems for detection of drug resistance. Fighting against these challenging forms of DR-TB requires (1) ensuring universal access to quality TB care, (2) improving diagnostics, and (3) developing new anti-TB drugs and/or treatment regimens [25].
Of special interest in the field of bacterial resistance are mycobacteria transmitted via close contact with infected animals or via the consumption of contaminated products [26]. The Mycobacterium tuberculosis complex (MtbC) includes some mycobacteria that can cause zoonosis in humans. Alarmingly, among different species of the MtbC, some exhibit resistance to antibiotics used in the treatment of TB. Current research suggests that the transmission of MtbC is not only transmitted via the interfaces between humans and animals but also by the environment, including soil, water, pasture, air, dust, etc. [27]. A recent study found the presence of extracellular DNA (eDNA) in slow-growing mycobacteria, related to the formation of biofilms and the generation of resistance to isoniazid and amikacin [28]. Mtb could survive for extended periods in nature, which would make it easier for other mycobacteria to take eDNA directly from the environment and thus favor the appearance of new resistant strains. Actual methods for the diagnosis of tuberculosis are inefficient in differentiating Mtb from other mycobacteria, and this misleading diagnosis makes proper treatment impossible. The complications for correct diagnosis, the permanence of wild reservoirs, and the persistent infections in cattle have underestimated the number of actual cases of zoonosis, which, in addition, can have a significant impact on the natural course of TB [29].

3. Alternative Treatments and Their Availability

The discovery of new drugs is usually a costly and slow process; in the case of TB, it has been even more complicated. The slow growth rate of the bacteria, the need to work with a biological safety level type 3 (BSL-3), and the long time invested in animal models have led to further delays in this process. On the other hand, the regulatory processes required in preclinical and clinical trials require a minimum of 2 months of culture conversion rate data and 2 years of relapse rate data after treatment. Therefore, more than four decades have been required for the emergence of new effective medications. By 2018, only three new drugs were in the last phase of clinical trials (Phase 3): bedaquiline, delamanid, and pretomanid [10]. Currently, bedaquiline and delamanid have been approved for treating MDR-TB.
Bedaquiline is a diarylquinoline effective against active and non-replicating Mtb, also providing an alternative treatment for latent TB [30]. However, the long-term adverse risks of bedaquiline remain questionable regarding toxicity [31]. New diaryloquinolines that offer a safer alternative to their toxicity are in development, with BTZ-043, PBTZ-169, and OPC-167832 as the most optimistic and highly potent ones [32].
A promising alternative is repurposing old drugs. Since these medications have previously come through the requisite validation processes for their usage, the time required for their approval in other conditions is reduced. The most common cause of this situation is given by acetylsalicylic acid, which was approved as an analgesic and years later as a vascular protector [33]. Tree-repurposed drugs were approved by WHO for the treatment of tuberculosis: linezolid, used in nosocomial pneumonia, clofazimine (an anti-leprosy drug), and meropenem, a β-lactam antibiotic [34]. Table 1 summarizes the mechanisms of action of (1) currently approved drugs, (2) new drugs being introduced into new regimes, and finally, (3) the repurposed drugs that have been reported to be used in combat versus TB.

4. Potential Use of Natural Products as Alternatives against Antibiotic Resistance

The use of natural products (NPs) focused on combating a variety of human diseases has been in practice for many years. Nature has provided a virtually infinite supply of bioactive chemicals whose structural variety and diverse action mechanisms have drawn the attention of researchers worldwide. Currently, NPs constitute a valuable resource for treating complicated diseases like TB, and natural compounds with antimycobacterial potential represent an alternative method to avoid side effects from conventional medications. In recent times, the discovery of numerous and novel NP features for the treatment of TB has been promising; in the last 10 years, at least 1000 different papers on this subject have been published. Selected examples of these compounds are described in this section. However, a deeper look into more NPs with inhibitory properties against mycobacterial enzymatic targets is presented in Table 2.
Various natural and synthetic flavonoids were reported for the effective treatment of TB [85]. Also, some compounds extracted from Haliclona, a marine sponge, exhibited strong activity versus mycobacteria [86]. The potent efficacy of macrolides, such as SEQ-503, SEQ-9, clarithromycin, and sequanamycin A, was demonstrated in both in vitro and in vivo models of TB. SEQ-9 has the potential to enhance a variety of TB regimens by being effective even against MDR-TB [52]. Polyphenols have been exploited as anti-TB agents, and a positive effect was observed when used in conjunction with antibiotics, as they might reduce the exaggerated inflammation derived from traditional treatment [76]. In this regard, NPs have also been an excellent ally in treatments against resistance. Lactococcus produces antibiotics, such as nisin A and lacticin, that are extremely effective against certain MDR-TB strains [87]. In addition, antibacterial activity against Mtb, even with MDR-TB strains, can be obtained via the extraction of essential oils from Micromeria, Eucalyptus, and Juniperus [88]. Epigallocatechin gallate, a major component of green tea catechins, is helpful for the treatment of MDR-TB and XDR-TB [89]. Recently, cyclic peptides from actinomycetes like cyclomarin A, lassomycin, and ecumicin, have shown potent anti-TB effects versus drug-susceptible Mtb, as well as MDR-TB and XDR-TB. These last candidates are very promising because their target molecule, a very different one from those of existing anti-TB drugs, is ClpC1, an essential Mtb protein [61].
NPs also could improve the efficacy of conventional antimycobacterial therapies and be used as adjuvant therapy. For example, cumin, a seed from the parsley plant, improves the bioavailability of rifampicin and affects the permeability of membranes and propolis, improving the binding affinity of anti-TB medications to bacterial cell structures [90]. A natural substance called silymarin, which has antioxidant and hepatoprotective properties, has been suggested as a supplementary medication to lessen the liver damage caused by traditional anti-TB chemotherapy [91].
Treatment of latent tuberculosis infection is essential for TB control. Pyrazinamide (PZA) is the only drug that kills Mtb in a latent state; however, there is a high resistance to PZA. Alternative novel bactericidal against non-replicating Mtb is vital to reduce the transmission in the population. New chemical molecules against active and latent TB have been found in NPs. Halicyclamine A was isolated from the marine sponge Haliclona and showed a high activity against latent Mtb [92]. Trichoderin A, Trichoderin A1, and Trichoderin B are aminolipopeptides isolated from the fungus Trichoderma that showed significant activity against latent mycobacteria [93].
A major disadvantage of the use of NPs continues to be the low distribution and absorption of their active compounds, resulting in poor bioavailability. Bioavailability allows us to determine the percent of an administered dose of a drug/medication able to reach the systemic circulation. Unfortunately, in the case of the main components of NPs with biological activity, i.e., treatment for diseases such as TB, their chemical nature renders low solubility in water, thus lowering their bioavailability. In this sense, research in the areas of biotechnology, combined with nanotechnology, has focused on the development of strategies that help to improve their pharmacokinetic and pharmacodynamic properties [94].

5. The Only Preventive Treatment: Novel Generation of Vaccines

Despite the promising features of the treatments described before and as stated previously, vaccination represents the only prevention against TB. Vaccines can be highly effective tools for the fight against antimicrobial resistance by reducing (1) resistant-bacterial infections and (2) drug (antibiotic) consumption [95]. With respect to MDR-TB and XDR-TB, there is no evidence that drug resistance affects susceptibility to immune control; therefore, the development of new TB vaccines offers a crucial alternative in halting the spread of sensitive and drug-resistant Mtb strains [96]. In addition to neonates and infants, novel vaccines should be directed to teenagers and adults as their target population because both represent the most common sources for spreading Mtb.
The constant pressure driven by the necessity of more effective TB vaccines as well as the advances in the field of knowledge of Mtb, have sped up their development. Up to now, four main avenues have been adopted for the synthesis of novel vaccines [97]: (1) immunotherapeutic, (2) immunopreventive, (3) prime-boosting, and (4) priming, classified into four categories according to their immunization mode: whole cell, subunits, viral-based and nucleic acids.
The immunopreventive strategy can be divided into two [12]: (1) preventive pre-exposure (prophylactic) vaccines, which are administered before the first encounter with Mtb and are generally applied to neonates, and (2) preventive post-exposure vaccines, which focus on adolescents or adults, previously immunized by BCG and exhibiting latent TB infection. On the other hand, in conjunction with anti-TB drugs, therapeutic vaccines are employed for treating TB patients. Table 3 summarizes relevant information using selected examples in the field of new generations of TB vaccines.
Due to the novelty represented by nucleic acid-based vaccines and their impact during the COVID-19 pandemic, this category will be discussed separately. RNA/DNA-based vaccines are rapidly gaining attention within the immunization framework due to their several advantages. Despite entering clinical trials, the study conducted by Yonsei University with GX-70, a DNA-based vaccine using four antigen plasmids from Mtb together with Flt3 ligand, was finalized due to safety issues [99]. Very recently, preclinical data by [100] reported RNA platforms as a feasible system for TB vaccines. Certainly, in the near future, more studies will be conducted in this area, and better vaccination outcomes will be attained.

6. Final Remarks

Despite the advances in TB research and the different approaches to the fight against this disease, it still constitutes a worldwide threat to health. The aim of WHO’s “End TB Strategy” for achieving a TB-free world requires global actions, joint efforts from different stakeholders, social and political commitment, and public and private investment. The emergence of Mtb-resistant strains reflects the mismanagement of TB, and its surveillance represents a challenge for TB operations around the world, especially in developing and under-resourced countries, as well as in countries with a prevalence of HIV/AIDS [101]. In this scenario, the importance of co-infections should also be considered; WHO’s Global Tuberculosis Report [1] exhibited the damaging effect of the COVID-19 pandemic on TB diagnosis, treatment, and burden. The global coronavirus emergency affected medical services at all levels, which could disrupt the treatment of thousands of people with TB, including MDR-TB and XDR-TB. Whereas a large proportion of the population is estimated to be infected and living with latent tuberculosis, the damage to the airways and the decrease in the immune system caused by this coronavirus could favor the reactivation of TB in some individuals. Finally, the consequences of COVID-19 are also factors to be considered in the following year regarding the behavior of TB.
A multi-faceted approach is necessary to eradicate this disease as a public health problem. One fundamental pillar for this goal is investing in new tools and approaches to control TB, such as the synthesis of new drugs and vaccines. New medications for combating Mtb have been extensively studied, but the timing for their development continues to be a hindrance to their implementation. In this respect, a trend for repurposing old drugs has been adopted. Nevertheless, in both cases (new and old drugs), adverse effects remain a serious issue for the patient’s safety. Currently, there is a growing body of evidence that natural products can be effective against TB. Natural products may provide a valuable ally in the fight against TB, outstanding for their easy availability and their low cost. However, their use still faces several challenges, so more research is needed to confirm these findings and to determine the optimal dosage and administration route for these products. An interesting and optimistic avenue that constitutes the only prevention strategy lies in the generation of newer and more effective vaccines compared to BCG. Progress in this area has been attained; nevertheless, all clinical phases are mandatory for the approval and use of these alternatives.
Although TB represents a leading cause of death among infectious diseases, it has been it has been ignored or even neglected in some instances. Many questions regarding this illness continue to arise with the control of it as a central topic. Despite the significant improvements in illness understanding, there is still a long way to go considering it to be overcome. In this work, the joint effort of several researchers in the development of the discovery of alternative treatments was compiled in the hope of attaining a world free of TB.

Author Contributions

All authors contributed to the research, writing, and review process. All authors have read and agreed to the published version of the manuscript.

Funding

Fund for Scientific Paper Publications from Tecnologico de Monterrey.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO (World Health Organization). Global Tuberculosis Report; Licence: CC BY-NC-SA 3.0 IGO; WHO: Geneva, Switzerland, 2022. [Google Scholar]
  2. Bloom, B.R.; Atun, R.; Cohen, T.; Dye, C.; Fraser, H.; Gomez, G.B.; Knight, G.; Murray, M.; Nardell, E.; Rubin, E.; et al. Tuberculosis. In Major Infectious Diseases, 3rd ed.; Holmes, K.K., Ed.; Chapter 11; The International Bank for Reconstruction and Development/The World Bank: Washington, DC, USA, 2017. [Google Scholar] [CrossRef]
  3. Guinn, K.M.; Rubin, E.J. Tuberculosis: Just the FAQs. mBio 2017, 8, e01910-17. [Google Scholar] [CrossRef] [PubMed]
  4. Maitra, A.; Munshi, T.; Healy, J.; Martin, L.T.; Vollmer, W.; Keep, N.H.; Bhakta, S. Cell wall peptidoglycan in Mycobacterium tuberculosis: An Achilles’ heel for the TB-causing pathogen. FEMS Microbiol. Rev. 2019, 43, 548–575. [Google Scholar] [CrossRef] [PubMed]
  5. Barksdale, L.; Kim, K.S. Mycobacterium . Bacteriol. Rev. 1977, 41, 217–372. [Google Scholar] [CrossRef] [PubMed]
  6. Chiaradia, L.; Lefebvre, C.; Parra, J.; Marcoux, J.; Burlet-Schiltz, O.; Etienne, G.; Tropis, M.; Daffé, M. Dissecting the mycobacterial cell envelope and defining the composition of the native mycomembrane. Sci. Rep. 2017, 7, 12807. [Google Scholar] [CrossRef]
  7. Alderwick, L.J.; Harrison, J.; Lloyd, G.S.; Birch, H.L. The mycobacterial cell wall—Peptidoglycan and arabinogalactan. Cold Spring Harb. Perspect. Med. 2015, 5, a021113. [Google Scholar] [CrossRef]
  8. Behr, M.A.; Kaufmann, E.; Duffin, J.; Edelstein, P.H.; Ramakrishnan, L. Latent tuberculosis: Two centuries of confusion. Am. J. Respir. Crit. Care Med. 2021, 204, 142–148. [Google Scholar] [CrossRef]
  9. CDC (Centers for Disease Control and Prevention). Tuberculosis Treatment. Available online: https://www.cdc.gov/TB/topic/treatment/TBdisease.htm (accessed on 30 July 2023).
  10. Patil, K.; Bagade, S.; Bonde, S.; Sharma, S.; Saraogi, G. Recent therapeutic approaches for the management of tuberculosis: Challenges and opportunities. Biomed. Pharmacother. 2018, 99, 735–745. [Google Scholar] [CrossRef]
  11. Günther, G.; Ruswa, N.; Keller, P.M. Drug-resistant tuberculosis: Advances in diagnosis and management. Curr. Opin. Pulm. Med. 2021, 28, 211–217. [Google Scholar] [CrossRef]
  12. Fatima, S.; Kumari, A.; Das, G.; Dwivedi, V.P. Tuberculosis vaccine: A journey from BCG to present. Life Sci. 2020, 252, 117594. [Google Scholar] [CrossRef] [PubMed]
  13. Ritz, N.; Hanekom, W.A.; Robins-Browne, R.; Britton, W.J.; Curtis, N. Influence of BCG vaccine strain on the immune response and protection against tuberculosis. FEMS Microbiol. Rev. 2008, 32, 821–841. [Google Scholar] [CrossRef]
  14. Wang, J.F.; Dai, F.Y.; Gong, X.L.; Bao, L. Commonly administered bacille Calmette-Guerin strains induce comparable immune response. Int. J. Clin. Exp. Med. 2015, 8, 15834–15839. [Google Scholar] [PubMed]
  15. Mjid, M.; Cherif, J.; Ben Salah, N.; Toujani, S.; Ouahchi, Y.; Zakhama, H.; Louzir, B.; Mehiri-Ben Rhouma, N.; Beji, M. Épidémiologie de la tuberculose [Epidemiology of tuberculosis]. Rev. De Pneumol. Clin. 2015, 71, 67–72. [Google Scholar] [CrossRef] [PubMed]
  16. Lee, J.Y.; Kwon, N.; Goo, G.Y.; Cho, S.I. Inadequate housing and pulmonary tuberculosis: A systematic review. BMC Public Health 2022, 22, 622. [Google Scholar] [CrossRef]
  17. Van Deun, A.; Maug, A.K.; Salim, M.A.; Das, P.K.; Sarker, M.R.; Daru, P.; Rieder, H.L. Short, highly effective, and inexpensive standardized treatment of multidrug-resistant tuberculosis. Am. J. Respir. Crit. Care Med. 2010, 182, 684–692. [Google Scholar] [CrossRef]
  18. Poulton, N.C.; Rock, J.M. Unraveling the mechanisms of intrinsic drug resistance in Mycobacterium tuberculosis. Front. Cell. Infect. Microbiol. 2022, 12, 997283. [Google Scholar] [CrossRef]
  19. Bi, K.; Cao, D.; Ding, C.; Lu, S.; Lu, H.; Zhang, G.; Zhang, W.; Li, L.; Xu, K.; Li, L.; et al. The past, present and future of tuberculosis treatment. J. Zhejiang Univ. Med. Sci. 2022, 51, 657–668. [Google Scholar] [CrossRef]
  20. Eker, B.; Ortmann, J.; Migliori, G.B.; Sotgiu, G.; Muetterlein, R.; Centis, R.; Hoffmann, H.; Kirsten, D.; Schaberg, T.; Ruesch-Gerdes, S.; et al. Multidrug-and extensively drug-resistant tuberculosis, Germany. Emerg. Infect. Dis. 2008, 14, 1700–1706. [Google Scholar] [CrossRef]
  21. Wu, C.; Yi, H.; Hu, Y.; Luo, D.; Tang, Z.; Wen, X.; Zhang, Y.; Tang, M.; Zhang, L.; Wu, S.; et al. Effects of second-line anti-tuberculosis drugs on the intestinal microbiota of patients with rifampicin-resistant tuberculosis. Front. Cell. Infect. Microbiol. 2023, 13, 1127916. [Google Scholar] [CrossRef]
  22. Jain, A.; Mondal, R. Extensively drug-resistant tuberculosis: Current challenges and threats. FEMS Immunol. Med. Microbiol. 2008, 53, 145–150. [Google Scholar] [CrossRef]
  23. Uplekar, M.; Weil, D.; Lonnroth, K.; Jaramillo, E.; Lienhardt, C.; Dias, H.M.; Falzon, D.; Floyd, K.; Gargioni, G.; Getahun, H.; et al. WHO’s new end TB strategy. Lancet 2015, 385, 1799–1801. [Google Scholar] [CrossRef]
  24. Yew, W.W. Management of multidrug-resistant tuberculosis and extensively drug-resistant tuberculosis: Current status and future prospects. Kekkaku 2011, 86, 9–16. [Google Scholar] [PubMed]
  25. Gupta, H.; Kant, S.; Jain, A.; Natu, S.M.; Ahluwalia, S. Initial drug resistance pattern among pulmonary tuberculosis patients. Indian J. Tuberc. 2013, 60, 154–161. [Google Scholar] [PubMed]
  26. Müller, B.; Dürr, S.; Alonso, S.; Hattendorf, J.; Laisse, C.J.; Parsons, S.D.; van Helden, P.D.; Zinsstag, J. Zoonotic Mycobacterium bovis-induced tuberculosis in humans. Emerg. Infect. Dis. 2013, 19, 899–908. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, H.; Liu, M.; Fan, W.; Sun, S.; Fan, X. The impact of Mycobacterium tuberculosis complex in the environment on one health approach. Front. Public Health 2022, 10, 994745. [Google Scholar] [CrossRef] [PubMed]
  28. Ilinov, A.; Nishiyama, A.; Namba, H.; Fukushima, Y.; Takihara, H.; Nakajima, C.; Savitskaya, A.; Gebretsadik, G.; Hakamata, M.; Ozeki, Y.; et al. Extracellular DNA of slow growers of mycobacteria and its contribution to biofilm formation and drug tolerance. Sci. Rep. 2021, 11, 10953. [Google Scholar] [CrossRef] [PubMed]
  29. Abalos, P.; Retamal, P. Tuberculosis: A re-emerging zoonosis? Rev. Sci. Et Tech. (Int. Off. Epizoot.) 2004, 23, 583–594. [Google Scholar] [CrossRef]
  30. Riccardi, N.; Del Puente, F.; Magnè, F.; Taramasso, L.; Di Biagio, A. Bedaquiline: A new hope for shorter and better anti-tuberculosis regimens. Recent Pat. Anti-Infect. Drug Discov. 2018, 13, 3–11. [Google Scholar] [CrossRef]
  31. Gaida, R.; Truter, I.; Peters, C.A. Adverse effects of bedaquiline in patients with extensively drug-resistant tuberculosis. S. Afr. J. Infect. Dis. 2020, 35, 23. [Google Scholar] [CrossRef]
  32. Edwards, B.D.; Field, S.K. The struggle to end a millennia-long pandemic: Novel candidate and repurposed drugs for the treatment of tuberculosis. Drugs 2022, 82, 1695–1715. [Google Scholar] [CrossRef]
  33. Bliden, K.P.; Tantry, U.S.; Chaudhary, R.; Byun, S.; Gurbel, P.A. Extended-release acetylsalicylic acid for secondary prevention of stroke and cardiovascular events. Expert Rev. Cardiovasc. Ther. 2016, 14, 779–791. [Google Scholar] [CrossRef]
  34. Sharma, K.; Ahmed, F.; Sharma, T.; Grover, A.; Agarwal, M.; Grover, S. Potential repurposed drug candidates for tuberculosis treatment: Progress and update of drugs identified in over a decade. ACS Omega 2023, 8, 17362–17380. [Google Scholar] [CrossRef] [PubMed]
  35. O’Connor, C.; Brady, M.F. Isoniazid. In StatPearls; StatPearls Publishing: St. Petersburg, FL, USA, 2022. [Google Scholar]
  36. MacVinish, S.; McMaster, D.; Moledina, T.; Tamne, S.K.; Ashworth, J.; Anderson, S.R. Ethambutol and visual assessment in England: Current practice and recommendations. Eye 2023, 1–6. [Google Scholar] [CrossRef] [PubMed]
  37. Stein, G.E. Pharmacokinetics and pharmacodynamics of newer fluoroquinolones. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 1996, 23, S19–S24. [Google Scholar] [CrossRef] [PubMed]
  38. Peloquin, C.A.; Davies, G.R. The treatment of tuberculosis. Clin. Pharmacol. Ther. 2021, 110, 1455–1466. [Google Scholar] [CrossRef] [PubMed]
  39. Diacon, A.H.; Pym, A.; Grobusch, M.; Patientia, R.; Rustomjee, R.; Page-Shipp, L.; Pistorius, C.; Krause, R.; Bogoshi, M.; Churchyard, G.; et al. The diarylquinoline TMC207 for multidrug-resistant tuberculosis. N. Engl. J. Med. 2009, 360, 2397–2405. [Google Scholar] [CrossRef]
  40. Liu, Y.; Matsumoto, M.; Ishida, H.; Ohguro, K.; Yoshitake, M.; Gupta, R.; Geiter, L.; Hafkin, J. Delamanid: From discovery to its use for pulmonary multidrug-resistant tuberculosis (MDR-TB). Tuberculosis 2018, 111, 20–30. [Google Scholar] [CrossRef]
  41. Lee, S.F.K.; Laughon, B.E.; McHugh, T.D.; Lipman, M. New drugs to treat difficult tuberculous and nontuberculous mycobacterial pulmonary disease. Curr. Opin. Pulm. Med. 2019, 25, 271–280. [Google Scholar] [CrossRef]
  42. Manjunatha, U.; Boshoff, H.I.; Barry, C.E. The mechanism of action of PA-824: Novel insights from transcriptional profiling. Commun. Integr. Biol. 2009, 2, 215–218. [Google Scholar] [CrossRef]
  43. Sbardella, G.; Mai, A.; Artico, M.; Loddo, R.; Setzu, M.G.; La Colla, P. Synthesis and in vitro antimycobacterial activity of novel 3-(1H-pyrrol-1-yl)-2-oxazolidinone analogues of PNU-100480. Bioorg. Med. Chem. Lett. 2004, 14, 1537–1541. [Google Scholar] [CrossRef]
  44. Zhang, Y.; Shi, W.; Zhang, W.; Mitchison, D. Mechanisms of pyrazinamide action and resistance. Microbiol. Spectr. 2014, 2, MGM2-0023-2013. [Google Scholar] [CrossRef]
  45. Andries, K.; Verhasselt, P.; Guillemont, J.; Göhlmann, H.W.; Neefs, J.M.; Winkler, H.; Van Gestel, J.; Timmerman, P.; Zhu, M.; Lee, E.; et al. Diarylquinoline drug active on the ATP synthase of Mycobacterium tuberculosis. Science 2005, 307, 223–227. [Google Scholar] [CrossRef] [PubMed]
  46. Pethe, K.; Bifani, P.; Jang, J.; Kang, S.; Park, S.; Ahn, S.; Jiricek, J.; Jung, J.; Jeon, H.K.; Cechetto, J.; et al. Discovery of Q203, a potent clinical candidate for the treatment of tuberculosis. Nat. Med. 2013, 19, 1157–1160. [Google Scholar] [CrossRef] [PubMed]
  47. Meshnick, S.R. Artemisinin: Mechanisms of action, resistance and toxicity. Int. J. Parasitol. 2002, 32, 1655–1660. [Google Scholar] [CrossRef] [PubMed]
  48. Kaushik, A.; Makkar, N.; Pandey, P.; Parrish, N.; Singh, U.; Lamichhane, G. Carbapenems and rifampin exhibit synergy against Mycobacterium tuberculosis and Mycobacterium abscessus. Antimicrob. Agents Chemother. 2015, 59, 6561–6567. [Google Scholar] [CrossRef] [PubMed]
  49. Matt, U.; Selchow, P.; Dal Molin, M.; Strommer, S.; Sharif, O.; Schilcher, K.; Andreoni, F.; Stenzinger, A.; Zinkernagel, A.S.; Zeitlinger, M.; et al. Chloroquine enhances the antimycobacterial activity of isoniazid and pyrazinamide by reversing inflammation-induced macrophage efflux. Int. J. Antimicrob. Agents 2017, 50, 55–62. [Google Scholar] [CrossRef]
  50. Barry, V.C.; Conalty, M.L.; Gaffney, E.E. Antituberculosis activity in the phenazine series; Isomeric pigments obtained by oxidation of o-phenylenediamine derivatives. J. Pharm. Pharmacol. 1956, 8, 1089–1096. [Google Scholar] [CrossRef]
  51. Campoli-Richards, D.M.; Sorkin, E.M.; Heel, R.C. Inosine pranobex. A preliminary review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy. Drugs 1986, 32, 383–424. [Google Scholar] [CrossRef]
  52. Zhang, J.; Lair, C.; Roubert, C.; Amaning, K.; Barrio, M.B.; Benedetti, Y.; Cui, Z.; Xing, Z.; Li, X.; Franzblau, S.G.; et al. Discovery of natural-product-derived sequanamycins as potent oral anti-tuberculosis agents. Cell 2023, 186, 1013–1025. [Google Scholar] [CrossRef]
  53. Diacon, A.H.; van der Merwe, L.; Barnard, M.; von Groote-Bidlingmaier, F.; Lange, C.; García-Basteiro, A.L.; Sevene, E.; Ballell, L.; Barros-Aguirre, D. β-lactams against tuberculosis--new trick for an old dog? N. Engl. J. Med. 2016, 375, 393–394. [Google Scholar] [CrossRef]
  54. Singh, R.; Manjunatha, U.; Boshoff, H.I.; Ha, Y.H.; Niyomrattanakit, P.; Ledwidge, R.; Dowd, C.S.; Lee, I.Y.; Kim, P.; Zhang, L.; et al. PA-824 kills nonreplicating Mycobacterium tuberculosis by intracellular NO release. Science 2008, 322, 1392–1395. [Google Scholar] [CrossRef]
  55. Lee, R.E.; Hurdle, J.G.; Liu, J.; Bruhn, D.F.; Matt, T.; Scherman, M.S.; Vaddady, P.K.; Zheng, Z.; Qi, J.; Akbergenov, R.; et al. Spectinamides: A new class of semisynthetic antituberculosis agents that overcome native drug efflux. Nat. Med. 2014, 20, 152–158. [Google Scholar] [CrossRef] [PubMed]
  56. Pollo, L.A.E.; Martin, E.F.; Machado, V.R.; Cantillon, D.; Wildner, L.M.; Bazzo, M.L.; Waddell, S.J.; Biavatti, M.W.; Sandjo, L.P. Search for antimicrobial activity among fifty-two natural and synthetic compounds identifies anthraquinone and polyacetylene classes that inhibit Mycobacterium tuberculosis. Front. Microbiol. 2021, 11, 622629. [Google Scholar] [CrossRef] [PubMed]
  57. Igarashi, M.; Takahashi, Y.; Shitara, T.; Nakamura, H.; Naganawa, H.; Miyake, T.; Akamatsu, Y. Caprazamycins, novel lipo-nucleoside antibiotics, from Streptomyces sp. II. Structure elucidation of caprazamycins. J. Antibiot. 2005, 58, 327–337. [Google Scholar] [CrossRef] [PubMed]
  58. Tousif, S.; Singh, D.K.; Mukherjee, S.; Ahmad, S.; Arya, R.; Nanda, R.; Ranganathan, A.; Bhattacharyya, M.; Van Kaer, L.; Kar, S.K.; et al. Nanoparticle-formulated curcumin prevents posttherapeutic disease reactivation and reinfection with Mycobacterium tuberculosis following isoniazid therapy. Front. Immunol. 2017, 8, 739. [Google Scholar] [CrossRef] [PubMed]
  59. Schmitt, E.K.; Riwanto, M.; Sambandamurthy, V.; Roggo, S.; Miault, C.; Zwingelstein, C.; Krastel, P.; Noble, C.; Beer, D.; Rao, S.P.; et al. The natural product cyclomarin kills Mycobacterium tuberculosis by targeting the ClpC1 subunit of the caseinolytic protease. Angew. Chem. 2011, 50, 5889–5891. [Google Scholar] [CrossRef] [PubMed]
  60. Mullowney, M.W.; Hwang, C.H.; Newsome, A.G.; Wei, X.; Tanouye, U.; Wan, B.; Carlson, S.; Barranis, N.J.; hAinmhire, E.; Chen, W.L.; et al. Diaza-anthracene antibiotics from a freshwater-derived actinomycete with selective antibacterial activity toward Mycobacterium tuberculosis. ACS Infect. Dis. 2015, 1, 168–174. [Google Scholar] [CrossRef]
  61. Lee, H.; Suh, J.W. Anti-tuberculosis lead molecules from natural products targeting Mycobacterium tuberculosis ClpC1. J. Ind. Microbiol. Biotechnol. 2016, 43, 205–212. [Google Scholar] [CrossRef]
  62. Wang, G.; Dong, W.; Lu, H.; Lu, W.; Feng, J.; Wang, X.; Chen, H.; Liu, M.; Tan, C. Enniatin A1, a natural compound with bactericidal activity against Mycobacterium tuberculosis in vitro. Molecules 2019, 25, 38. [Google Scholar] [CrossRef]
  63. Lin, G.; Li, D.; Chidawanyika, T.; Nathan, C.; Li, H. Fellutamide B is a potent inhibitor of the Mycobacterium tuberculosis proteasome. Arch. Biochem. Biophys. 2010, 501, 214–220. [Google Scholar] [CrossRef]
  64. Pawar, A.; Jha, P.; Chopra, M.; Chaudhry, U.; Saluja, D. Screening of natural compounds that targets glutamate racemase of Mycobacterium tuberculosis reveals the anti-tubercular potential of flavonoids. Sci. Rep. 2020, 10, 949. [Google Scholar] [CrossRef]
  65. Kling, A.; Lukat, P.; Almeida, D.V.; Bauer, A.; Fontaine, E.; Sordello, S.; Zaburannyi, N.; Herrmann, J.; Wenzel, S.C.; König, C.; et al. Targeting DnaN for tuberculosis therapy using novel griselimycins. Science 2015, 348, 1106–1112. [Google Scholar] [CrossRef]
  66. Efremenko, Y.V.; Arjanova, O.V.; Prihoda, N.D.; Yurchenko, L.V.; Sokolenko, N.I.; Mospan, I.V.; Pylypchuk, V.S.; Rowe, J.; Jirathitikal, V.; Bourinbaiar, A.S.; et al. Clinical validation of sublingual formulations of Immunoxel (Dzherelo) as an adjuvant immunotherapy in treatment of TB patients. Immunotherapy 2012, 4, 273–282. [Google Scholar] [CrossRef]
  67. Ōmura, S.; Crump, A. Lactacystin: First-in-class proteasome inhibitor still excelling and an exemplar for future antibiotic research. J. Antibiot. 2019, 72, 189–201. [Google Scholar] [CrossRef]
  68. Intorasoot, S.; Intorasoot, A.; Tawteamwong, A.; Butr-Indr, B.; Phunpae, P.; Tharinjaroen, C.S.; Wattananandkul, U.; Sangboonruang, S.; Khantipongse, J. In vitro antimycobacterial activity of human lactoferrin-derived peptide, d-hlf 1-11, against susceptible and drug-resistant Mycobacterium tuberculosis and its synergistic effect with rifampicin. Antibiotics 2022, 11, 1785. [Google Scholar] [CrossRef]
  69. Gavrish, E.; Sit, C.S.; Cao, S.; Kandror, O.; Spoering, A.; Peoples, A.; Ling, L.; Fetterman, A.; Hughes, D.; Bissell, A.; et al. Lassomycin, a ribosomally synthesized cyclic peptide, kills Mycobacterium tuberculosis by targeting the ATP-dependent protease ClpC1P1P2. Chem. Biol. 2014, 21, 509–518. [Google Scholar] [CrossRef]
  70. Hou, X.M.; Wang, C.Y.; Gerwick, W.H.; Shao, C.L. Marine natural products as potential anti-tubercular agents. Eur. J. Med. Chem. 2019, 165, 273–292. [Google Scholar] [CrossRef]
  71. Daniel, T.M. The history of tuberculosis. Respir. Med. 2006, 100, 1862–1870. [Google Scholar] [CrossRef] [PubMed]
  72. Caminero, J.A.; Sotgiu, G.; Zumla, A.; Migliori, G.B. Best drug treatment for multidrug-resistant and extensively drug-resistant tuberculosis. Lancet Infect. Dis. 2010, 10, 621–629. [Google Scholar] [CrossRef] [PubMed]
  73. Mazlun, M.H.; Sabran, S.F.; Mohamed, M.; Abu Bakar, M.F.; Abdullah, Z. Phenolic compounds as promising drug candidates in tuberculosis therapy. Molecules 2019, 24, 2449. [Google Scholar] [CrossRef] [PubMed]
  74. Jeon, D.; Jeong, M.C.; Jnawali, H.N.; Kwak, C.; Ryoo, S.; Jung, I.D.; Kim, Y. Phloretin exerts anti-tuberculosis activity and suppresses lung inflammation. Molecules 2017, 22, 183. [Google Scholar] [CrossRef]
  75. Quan, D.; Nagalingam, G.; Payne, R.; Triccas, J.A. New tuberculosis drug leads from naturally occurring compounds. Int. J. Infect. Dis. IJID Off. Publ. Int. Soc. Infect. Dis. 2017, 56, 212–220. [Google Scholar] [CrossRef] [PubMed]
  76. Arrigoni, R.; Ballini, A.; Topi, S.; Bottalico, L.; Jirillo, E.; Santacroce, L. Antibiotic resistance to Mycobacterium tuberculosis and potential use of natural and biological products as alternative anti-mycobacterial agents. Antibiotics 2022, 11, 1431. [Google Scholar] [CrossRef] [PubMed]
  77. Banerjee, A.; Dubnau, E.; Quemard, A.; Balasubramanian, V.; Um, K.S.; Wilson, T.; Collins, D.; de Lisle, G.; Jacobs, W.R. InhA, a gene encoding a target for isoniazid and ethionamide in Mycobacterium tuberculosis. Science 1994, 263, 227–230. [Google Scholar] [CrossRef] [PubMed]
  78. Levy, C.W.; Roujeinikova, A.; Sedelnikova, S.; Baker, P.J.; Stuitje, A.R.; Slabas, A.R.; Rice, D.W.; Rafferty, J.B. Molecular basis of triclosan activity. Nature 1999, 398, 383–384. [Google Scholar] [CrossRef] [PubMed]
  79. Giddens, A.C.; Nielsen, L.; Boshoff, H.I.; Tasdemir, D.; Perozzo, R.; Kaiser, M.; Copp, B.R. Natural product inhibitors of fatty acid biosynthesis: Synthesis of the marine microbial metabolites pseudopyronines A and B and evaluation of their anti-infective activities. Tetrahedron 2008, 64, 1242–1249. [Google Scholar] [CrossRef]
  80. Safwat, N.A.; Kashef, M.T.; Aziz, R.K.; Amer, K.F.; Ramadan, M.A. Quercetin 3-O-glucoside recovered from the wild Egyptian Sahara plant, Euphorbia paralias L.; inhibits glutamine synthetase and has antimycobacterial activity. Tuberculosis 2018, 108, 106–113. [Google Scholar] [CrossRef]
  81. Chen, D.N.; Chen, H.; She, Z.G.; Lu, Y.J. Identification of bostrycin derivatives as potential inhibitors of Mycobacterium tuberculosis Protein Tyrosine Phosphatase (MptpB). Med. Chem. 2016, 12, 296–302. [Google Scholar] [CrossRef]
  82. Smolarz, H.D.; Swatko-Ossor, M.; Ginalska, G.; Medyńska, E. Antimycobacterial effect of extract and its components from Rheum rhaponticum. J. AOAC Int. 2013, 96, 155–160. [Google Scholar] [CrossRef]
  83. Qi, Y.K.; Tang, X.; Wei, N.N.; Pang, C.J.; Du, S.S.; Wang, K. Discovery, synthesis, and optimization of teixobactin, a novel antibiotic without detectable bacterial resistance. J. Pept. Sci. Off. Publ. Eur. Pept. Soc. 2022, 28, e3428. [Google Scholar] [CrossRef]
  84. Ling, L.L.; Schneider, T.; Peoples, A.J.; Spoering, A.L.; Engels, I.; Conlon, B.P.; Mueller, A.; Schäberle, T.F.; Hughes, D.E.; Epstein, S.; et al. A new antibiotic kills pathogens without detectable resistance. Nature 2015, 520, 388. [Google Scholar] [CrossRef]
  85. Rabaan, A.A.; Alhumaid, S.; Albayat, H.; Alsaeed, M.; Alofi, F.S.; Al-Howaidi, M.H.; Turkistani, S.A.; Alhajri, S.M.; Alahmed, H.E.; Alzahrani, A.B.; et al. Promising antimycobacterial activities of flavonoids against Mycobacterium sp. drug targets: A comprehensive review. Molecules 2022, 27, 5335. [Google Scholar] [CrossRef] [PubMed]
  86. Khan, M.T.; Kaushik, A.C.; Bhatti, A.I.; Zhang, Y.J.; Zhang, S.; Wei, A.J.; Malik, S.I.; Wei, D.Q. Marine natural products and drug resistance in latent tuberculosis. Mar. Drugs 2019, 17, 549. [Google Scholar] [CrossRef] [PubMed]
  87. Oliveira, G.S.; Costa, R.P.; Gomes, P.; Gomes, M.S.; Silva, T.; Teixeira, C. Antimicrobial peptides as potential anti-tubercular leads: A concise review. Pharmaceuticals 2021, 14, 323. [Google Scholar] [CrossRef]
  88. El Omari, K.; Hamze, M.; Alwan, S.; Osman, M.; Jama, C.; Chihib, N.E. In-vitro evaluation of the antibacterial activity of the essential oils of Micromeria barbata, Eucalyptus globulus and Juniperus excelsa against strains of Mycobacterium tuberculosis (including MDR-TB), Mycobacterium kansasii and Mycobacterium gordonae. J. Infect. Public Health 2019, 12, 615–618. [Google Scholar] [CrossRef] [PubMed]
  89. Sharma, S.K.; Kumar, G.; Kapoor, M.; Surolia, A. Combined effect of epigallocatechin gallate and triclosan on enoyl-ACP reductase of Mycobacterium tuberculosis. Biochem. Biophys. Res. Commun. 2008, 368, 12–17. [Google Scholar] [CrossRef]
  90. Maiolini, M.; Gause, S.; Taylor, J.; Steakin, T.; Shipp, G.; Lamichhane, P.; Deshmukh, B.; Shinde, V.; Bishayee, A.; Deshmukh, R.R. The war against tuberculosis: A review of natural compounds and their derivatives. Molecules 2020, 25, 3011. [Google Scholar] [CrossRef]
  91. Rodríguez-Flores, E.M.; Mata-Espinosa, D.; Barrios-Payan, J.; Marquina-Castillo, B.; Castañón-Arreola, M.; Hernández-Pando, R. A significant therapeutic effect of silymarin administered alone, or in combination with chemotherapy, in experimental pulmonary tuberculosis caused by drug-sensitive or drug-resistant strains: In vitro and in vivo studies. PLoS ONE 2019, 14, e0217457. [Google Scholar] [CrossRef]
  92. Arai, M.; Sobou, M.; Vilchéze, C.; Baughn, A.; Hashizume, H.; Pruksakorn, P.; Ishida, S.; Matsumoto, M.; Jacobs, W.R.; Kobayashi, M. Halicyclamine A, a marine spongean alkaloid as a lead for anti-tuberculosis agent. Bioorg. Med. Chem. 2008, 16, 6732–6736. [Google Scholar] [CrossRef]
  93. Pruksakorn, P.; Arai, M.; Kotoku, N.; Vilchèze, C.; Baughn, A.D.; Moodley, P.; Jacobs, W.R.; Kobayashi, M. Trichoderins, novel aminolipopeptides from a marine sponge-derived Trichoderma sp., are active against dormant mycobacteria. Bioorg. Med. Chem. Lett. 2010, 20, 3658–3663. [Google Scholar] [CrossRef]
  94. Sachdeva, A.; Dhawan, D.; Jain, G.K.; Yerer, M.B.; Collignon, T.E.; Tewari, D.; Bishayee, A. Novel Strategies for the Bioavailability Augmentation and Efficacy Improvement of Natural Products in Oral Cancer. Cancers 2022, 15, 268. [Google Scholar] [CrossRef]
  95. Frost, I.; Sati, H.; Garcia-Vello, P.; Hasso-Agopsowicz, M.; Lienhardt, C.; Gigante, V.; Beyer, P. The role of bacterial vaccines in the fight against antimicrobial resistance: An analysis of the preclinical and clinical development pipeline. Lancet. Microbe 2023, 4, e113–e125. [Google Scholar] [CrossRef] [PubMed]
  96. WHO (World Health Organization). Preferred Product Characteristics for New Tuberculosis Vaccines; Licence: CC BY-NC-SA 3.0 IGO; WHO: Geneva, Switzerland, 2018. [Google Scholar]
  97. Gong, W.; Liang, Y.; Wu, X. The current status, challenges, and future developments of new tuberculosis vaccines. Hum. Vaccines Immunother. 2018, 147, 1697–1716. [Google Scholar] [CrossRef] [PubMed]
  98. Brazier, B.; McShane, H. Towards new TB vaccines. Semin. Immunopathol. 2020, 42, 315–331. [Google Scholar] [CrossRef] [PubMed]
  99. Zhuang, L.; Ye, Z.; Li, L.; Yang, L.; Gong, W. Next-Generation TB Vaccines: Progress, Challenges, and Prospects. Vaccines 2023, 11, 1304. [Google Scholar] [CrossRef] [PubMed]
  100. Larsen, S.E.; Baldwin, S.L.; Coler, R.N. Tuberculosis vaccines update: Is an RNA-based vaccine feasible for tuberculosis? Int. J. Infect. Dis. 2023, 130, S47–S51. [Google Scholar] [CrossRef]
  101. Caminero, J.A.; García-García, J.M.; Cayla, J.A.; García-Pérez, F.J.; Palacios, J.J.; Ruiz-Manzano, J. Drug resistant tuberculosis: New WHO definitions and their implication in the SEPAR Guideline. Tuberculosis con resistencia a fármacos: Nuevas definiciones de la OMS y su implicación en la Normativa de SEPAR. Arch. De Bronconeumol. 2022, 58, 87–89. [Google Scholar] [CrossRef]
Table 1. Common names of current, alternative, and repurposed drugs used for the treatment of TB, including their action mode.
Table 1. Common names of current, alternative, and repurposed drugs used for the treatment of TB, including their action mode.
NameDrug TypeMechanism of ActionReference
Approved Drugs
IsoniazidAntibiotic, highly specific against mycobacteriaInhibits the biosynthesis of mycolic acids[35]
EthambutolAntibioticBlocks the arabinosyl transferases involved in cell wall biosynthesis[36]
MoxifloxacinThird-generation quinoloneInhibits the DNA gyrases[37]
RifampicinAntibioticInhibits the DNA-dependent RNA polymerase of Mtb[38]
Alternative new drugs
BedaquilineDiarylquinolineInhibits mycobacterial ATP synthase[39]
DelamanidNitro-dihydro-imidazooxadole derivativeInhibits the synthesis of mycolic acids[40]
Macozinone (PBTZ1698)Benzothiazinone Inhibits cell wall synthesis; has synergic effect with first-line antibiotics[41]
PretomanidNitroimidazo oxazinInhibits respiratory chain and synthesis of mycolic acids [42]
PNU-100480OxazolidinonesInhibits translation of the initiation phase of bacterial protein synthesis by selectively inhibiting ribosomal peptidyl transferase[43]
PyrazinamideAntiuricosuric drugTargets the mycobacterial fatty acid synthase I gene[44]
R-207910Diarylquinolones Inhibits mycobacterial ATP synthase[45]
TelacebecAnti-TB drugInhibits the mycobacterial cytochrome bc1 complex[46]
Repurposed drugs
ArtemisininTreatment against malaria Isolated from the wormwood plant Artemisia annua; works by forming free radicals[47]
BiapenemCarbapenem antibiotic used for Pseudomonas spp.Inhibits cell wall synthesis[48]
Chloroquine AntifungalInhibits efflux pump[49]
ClofazimineAnti-leprosyBinds to the guanine bases of bacterial DNA, blocking the template function of DNA[50]
IsoprinosineAnti-viral Stimulates host immune system[51]
LinezolidUsed to treat infections, including pneumonia, and infections of the skinInhibits protein synthesis by targeting the 50S ribosome; active against XDR-TB[52]
Meropen/ClavulanateRiminophenazineInhibit the enzyme β-lactamase[53]
PA-824 and OPC-67683 (delamanid)Derivative of metroniazoleInhibit the synthesis of ketimycolates, component of mycobacterial cell wall[54]
SpectinamidesTreatment of gonorrhea in patients who are allergic (or resistant) to penicillinReduces susceptibility of the former to drug efflux; active against MDR-TB and XDR-TB clinical isolates[55]
Table 2. Natural products with inhibitory properties against mycobacteria, including their source and action mode.
Table 2. Natural products with inhibitory properties against mycobacteria, including their source and action mode.
Natural ProductSourceMechanism of ActionReference
Anthraquinone and polyacetylenePlant-derived secondary metaboliteInhibit Mtb at a level similar to the first-line anti-TB drug, rifampicin[56]
CaprazamycinsLiponucleoside antibiotics isolated from StreptomycesInhibition of the biosynthetic enzyme MraY (translocase I)[57]
CurcuminNatural polyphenol derived from turmericSignificantly decreases hepatotoxicity with INH and is used as an adjuvant therapy regimen; used also against MDR-TB and XDR-TB[58]
CyclomarinsCycloheptapeptides from marine streptomyceteInhibit the ATPase ClpC1[59]
DiazaquinomycinsDiaza-anthracene from StreptomycesInhibit thymidylate synthase[60]
EcumicinCyclic tridecapeptide from NonomuraeaInhibits the ATPase ClpC1[61]
Ennaitin A1Obtained from Quercus pathogen Gnomonia errabundaSynergistic effects against Mtb with first and second-line anti-TB agents[62]
Fellutamide BPeptide aldehydes from the marine fungus Penicillium fellutanumPotent proteasome inhibitor of Mtb[63]
Flavonoids (naringenin and quercetin)Citrus-derived flavonoidsInduce membrane damage of Mtb and decrease the functional activity of Mtb-MurI protein; activity against MDR-TB[64]
GriselimycinsCyclic peptide from StreptomycesInhibit the DNA polymerase sliding
clamp (DnaN). Shown to be equally effective as rifampicin
[65]
Immunoxel (Dzherelo)Extract of medicinal plantsUsed in immunotherapy for TB and TB/HIV co-infection[66]
LactacystinProduced by StreptomycesAn irreversible proteasomal
inhibitor (specifically at the 20S proteasome β-subunit)
[67]
LactoferrinHuman antimicrobial peptideAdditive effect with isoniazide and rifampin[68]
LassomycinHexadecapeptide from actinomycetesInhibits the ATPase ClpC1; activity against MDR-TB and XDR-TB isolates[69]
ManzaminesObtained from marine spongesInsecticidal, cytotoxic, anti-inflammatory, antimicrobial, and anti-viral activities.[70]
MycinsSourced from actinobacteriab-lactams, tetracylines, and
aminoglycosides; exhibit potent activity against MDR-TB
[71]
PhenazinesAromatic compounds for many species of actinobacteriaClofazimine is currently used for MDR-TB[72]
Phenolic compoundsPhytophenolic compoundsEfflux system inhibition, protease inhibition, and mycolic acid biosynthesis inhibition[73]
PhloretinPolyphenolic compounds found in fruits, vegetables, legumes, etc.Inhibits growth of Mtb H37Rv, MDR-TB, and XDR-TB isolates[74]
PiperidinesHeterocycline amines extracted from black pepperVasodilators, antipsychotics, neuroleptics, and opioids, active against MDR-TB and XDR-TB; work synergistically with bedaquiline, pretomanid, and moxifloxacin[75]
PolyphenolsContained in fruits, vegetables, cereals, red wine, and extra virgin olive oilAntioxidant, anti-inflammatory, and microbicidal activities[76]
PolyphenolsIsolated from tea plant Camellia sinensisInhibition of InhA[77]
PyridomycinIsolated from StreptomycesInhibition of NADH-dependent enoyl-[acyl-carrier-protein] reductase InhA[78]
PyronesObtained from
the bacterium Pseudomonas sediment
Inhibition of InhA (enoyl-ACP-reductase, related to the synthesis of cell wall)[79]
QuercetinFlavanols, isolated from apples, berries, capers, grapes, and teaInhibit Mtb growth by blocking isocitrate lyase and exhibit hepatoprotective effects[80]
QuinonesObtained from fungi like Bostrichonema and NigrosporaInhibit MptpB and impair
mycobacterial survival in macrophages
[81]
ResveratrolStilbenoid polyphenol, found in peanuts, wine, and cranberries.Activates abyssinone II, possible target for therapy or prevention of TB[82]
Sequanamycin AMacrolide produced by Allokutzneria albata (actinomycete)Inhibits bacterial ribosome; derived macrolide antibiotic active on MDR-TB[52]
TeixobactinCyclic depsipeptide obtained from uncultured soil bacteriaBlocks cell wall biosynthesis; activity against TB, including drug-resistant strains[83]
Tiacumicin BGlycosylated macrolide tiacumicin from
the soil bacterium Dactylosporangium
Bacterial RNA polymerase inhibitor[84]
Table 3. Different types of vaccines intended for their use in the combat against TB a.
Table 3. Different types of vaccines intended for their use in the combat against TB a.
Vaccine CategoryMain Group TypeMain Features
Whole cell: inactivated
RUTI®TherapeuticDetoxified and fragmented Mtb cells delivered in liposomes
Immuvac/MIPTherapeuticHeat-killed Mycobacterium indicus pranii
Whole cell: live attenuated
MTBVACPre- and post-exposureAttenuated Mtb
Whole cell: recombinant live
VPM1002Three types (pre-exposure, post-exposure, and therapeutic)Recombinant BCG (rBCG) expressing listeriolysin gene for lysosome escape and lacking urease gene
Adjuvant protein subunit
M72/AS01E or Mtb72FBooster, pre-, and post-exposureImmunogenic fusion protein (M72) derived from Mtb antigens Mtb32A and Mtb39A, and AS01E adjuvant
Viral-based (recombinant)
Ad5Ag85APre- and post-exposureRecombinant adenovirus
MVA85ARecombinant vaccinia virus
a This table compiled data from [12,95,97,98,99].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rodríguez-Bustamante, E.; Gómez-Manzo, S.; De Obeso Fernández del Valle, A.; Arreguín-Espinosa, R.; Espitia-Pinzón, C.; Rodríguez-Flores, E. New Alternatives in the Fight against Tuberculosis: Possible Targets for Resistant Mycobacteria. Processes 2023, 11, 2793. https://doi.org/10.3390/pr11092793

AMA Style

Rodríguez-Bustamante E, Gómez-Manzo S, De Obeso Fernández del Valle A, Arreguín-Espinosa R, Espitia-Pinzón C, Rodríguez-Flores E. New Alternatives in the Fight against Tuberculosis: Possible Targets for Resistant Mycobacteria. Processes. 2023; 11(9):2793. https://doi.org/10.3390/pr11092793

Chicago/Turabian Style

Rodríguez-Bustamante, Eduardo, Saúl Gómez-Manzo, Alvaro De Obeso Fernández del Valle, Roberto Arreguín-Espinosa, Clara Espitia-Pinzón, and Eden Rodríguez-Flores. 2023. "New Alternatives in the Fight against Tuberculosis: Possible Targets for Resistant Mycobacteria" Processes 11, no. 9: 2793. https://doi.org/10.3390/pr11092793

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop