Skip to main content

Advanced drug delivery and therapeutic strategies for tuberculosis treatment

Abstract

Tuberculosis (TB) remains a significant global health challenge, necessitating innovative approaches for effective treatment. Conventional TB therapy encounters several limitations, including extended treatment duration, drug resistance, patient noncompliance, poor bioavailability, and suboptimal targeting. Advanced drug delivery strategies have emerged as a promising approach to address these challenges. They have the potential to enhance therapeutic outcomes and improve TB patient compliance by providing benefits such as multiple drug encapsulation, sustained release, targeted delivery, reduced dosing frequency, and minimal side effects. This review examines the current landscape of drug delivery strategies for effective TB management, specifically highlighting lipid nanoparticles, polymer nanoparticles, inorganic nanoparticles, emulsion-based systems, carbon nanotubes, graphene, and hydrogels as promising approaches. Furthermore, emerging therapeutic strategies like targeted therapy, long-acting therapeutics, extrapulmonary therapy, phototherapy, and immunotherapy are emphasized. The review also discusses the future trajectory and challenges of developing drug delivery systems for TB. In conclusion, nanomedicine has made substantial progress in addressing the challenges posed by conventional TB drugs. Moreover, by harnessing the unique targeting abilities, extended duration of action, and specificity of advanced therapeutics, innovative solutions are offered that have the potential to revolutionize TB therapy, thereby enhancing treatment outcomes and patient compliance.

Graphical Abstract

Introduction

Tuberculosis (TB), a chronic granulomatous disease caused by Mycobacterium tuberculosis (M. tuberculosis) that typically infects the lungs, is one of the most prevalent contagious infections [1]. This aerosol-based transmissible disease is among the top infectious diseases worldwide [2,3,4]. In India alone, 26% of global cases have been reported, the most significant number of TB cases for any individual country [5]. It is considered the second deadliest infection after COVID-19. This infection is prevalent in all age groups worldwide and is curable as well as preventable. According to the WHO, early diagnosis and proper treatment have saved more than 74 million lives in the past two decades [6]. When the infection is untreated, the TB bacteria multiply and progress to other organs, which can result in fatal outcomes. Many patients diagnosed with TB are prescribed a standardized treatment regimen containing first-line anti-tubercular drugs (ATDs). The nonadherence of patients to ATDs leads to the generation of a newer strain known as multidrug resistant (MDR)-TB [7]. This occurs due to random chromosomal mutations and genetic changes in the bacterium. MDR bacilli are resistant to two important first-line drugs, isoniazid (INH) and rifampicin (RIF), and are treated using second-line ATDs, such as amikacin, capreomycin, and fluoroquinolones. Extensively drug-resistant (XDR)-TB is a more dangerous strain than MDR-TB. The treatment of XDR-TB is more difficult, as patients are resistant to many of the second-line drugs. Highly potent antibiotics, such as thioridazine, have severe side effects and are used for the treatment of XDR-TB [8].

Due to their size, the tubercle bacilli can reach the pulmonary alveoli, further becoming phagocytized by the alveolar macrophages (AM) [9, 10]. The bacilli then multiply in the alveolar sacs. The granulomas formed in these regions have heterogeneous size distributions and varying cellular compositions. When the bacterial load reaches a maximum, it can alter the morphology of granulomas, eventually spreading to other organs through the bloodstream and lymphatic system, resulting in extrapulmonary TB [11]. Thus, apart from the most common form of pulmonary TB, it can practically affect all human body organs, particularly the pleura, lymph nodes, abdomen, genitourinary tract, skin, meninges, joints, and bones [12, 13]. A schematic diagram of the pathogenesis of TB infection is shown in Fig. 1.

Fig. 1
figure 1

Pathogenesis of TB infection. A Pulmonary TB: by inhalation of infected droplet nuclei, M. tuberculosis enters the respiratory tract and alveoli of the lungs. It is ingested by AM, which attempts to destroy the bacilli initially. The development of symbiosis results in logarithmic growth of bacilli. The multicellular host immune response develops, bringing more defensive cells to the site. The infected areas progress into a granuloma that can develop a solid caseous center, where the bacteria survive for years, resulting in latent TB. In the final stage of pathogenesis, liquefaction of the caseous center and granuloma rupture cause the spread of bacilli. Pulmonary TB is caused by bacterial spread in the lungs. Extrapulmonary TB results from the dissemination of bacilli to other tissues and organs via the vascular or lymphatic system. B Extrapulmonary TB; common sites include the pleura, lymph nodes, gastrointestinal tract, urogenital tract, skin, bones, meninges or eye [14, 15]

The treatment of TB patients is performed by using the TB-DOTS (directly observed treatment, short-course) treatment regimen. This regimen involves the use of various ATDs. The majority of the drugs used for therapy have severe adverse effects. The drugs' cure rates are as high as 95% in clinical trials, but they perform significantly worse in clinical conditions. The main reason for this is the lengthy duration of treatment and high dropout rates. The long duration of treatments and the serious adverse effects of the drugs impair patients' physical and mental endurance throughout therapy [16, 17]. Such instances cause patient relapse while also contributing to the development of bacterial resistance. Furthermore, there are several TB subpopulations, each with its physiology within the host. It can exist in two states that react to drugs differently: an active dividing state and a dormant/inactive state. It can alter drug metabolism, which also affects how effectively the therapy cycle functions [18, 19].

Due to tremendous improvements in treatment strategies, the number of patients infected with this disease is declining. The main reason for such a decline is the early and accurate diagnosis of the disease by various conventional and advanced techniques, such as chest X-ray, sputum microscopy, culturing method, nucleic acid amplification, ultralow dose chest CT (Clinical trials.gov identifier: NCT03220464), QuantiFERON (Clinical trials.gov identifier: NCT00982969), Nanodisk MS assay (Clinical trials.gov identifier: NCT03271567), and automated molecular diagnosis platform (Clinical trials.gov identifier: NCT04988984). However, due to the emergence of variant strains, the eradication of TB has not been achievable [20]. In this context, improved treatments with appropriate routes of administration are needed to shorten TB treatment duration, enhance efficacy, reduce adverse effects, and prevent resistance [9].

The significant challenges of clinical efficacy for TB include drug-resistant strains of M. tuberculosis, standard treatment duration (6–9 months), delayed diagnosis, and reduced therapeutic response with immunocompromised patients, which leads to more severe disease and a higher risk of complications that affect clinical outcomes [12, 21]. TB therapy is often delivered through various routes to ensure effective treatment. The main challenges associated with the oral route include slower onset of action, hepatic first-pass metabolism and rapid gastrointestinal absorption [9]. The parenteral and pulmonary routes for TB therapy displayed the highest bioavailability compared to oral administration. In particular, inhaled formulations are considered suitable for improving the pharmacodynamic profile of a drug [22]. Remarkably, lower doses of TB drugs can be delivered through inhalation and still result in effective treatments, thus reducing the chance of toxicity and enhancing localized drug concentrations [23]. With nanotechnology projected to simplify dosing and minimize adverse events, it can contribute significantly to the elimination of TB, especially by eradication of mycobacteria in those who do not have an active disease (latent TB), thereby preventing the progression to active disease [14, 24].

Advanced drug delivery strategies could improve bioavailability, patient compliance and the efficacy of TB treatment. As summarized in Table 1, with the dearth of new drug approvals for ATDs, except for bedaquiline, delamanidin, and pretomanid, which have come up in more than 40 years, innovative drug delivery strategies for existing drugs could be considered promising to enhance patient compliance [25,26,27,28]. Novel drug delivery systems (NDDSs) aid in optimizing the concentration of the active compound in the patient's plasma. Promising strategies for optimizing drug delivery could be based on modern systems such as nanoparticles, liposomes, microemulsions, niosomes, dendrimers and liquid crystalline systems [3, 25, 29]. By developing inexpensive and easy-to-administer delivery systems that offer extended drug release, dosing frequency could be reduced, thereby improving patient adherence. Direct targeting by selectivity toward both AM and tubercle bacilli may counteract the ability of intracellular pathogens to evade antibiotic treatments [30]. Thus, NDDSs can help optimize drug delivery to the target site, maximizing drug absorption and minimizing unwanted side effects [31]. Moreover, drug delivery systems can be optimized for a suitable route of administration to safeguard the therapeutic agents from immediate host metabolism and clearance, which can aid in therapeutic dose reduction [32]. A well-designed drug carrier can also demonstrate controlled drug release characteristics as per different metabolic and physicochemical responses [33]. These delivery systems also have a high possibility of treating nontubercular infections in the future [34, 35].

Table 1 Drugs used in tuberculosis therapy

Here, we have reviewed the recent advances in drug delivery and therapeutics to effectively treat TB by overcoming the existing hurdles associated with TB therapy. We have discussed promising preclinical developments and their future perspectives, along with the challenges that must be addressed for impactful clinical translation.

Advanced drug delivery strategies

Designing NDDSs includes approaches for achieving and optimizing the continuous delivery of drugs in a precise and reproducible manner to the target site. A major focus has been on targeted drug delivery and minimizing undesirable effects. They are of various types depending upon the formulation, dosage form, and mechanism of drug delivery. Various types of delivery systems and routes of administration for TB are represented in Fig. 2.

Fig. 2
figure 2

Modern drug delivery systems for bioactive molecules in TB treatment. Modern drug delivery strategies for existing TB drugs could be promising, as they can offer the flexibility to adopt better routes of administration, multiple drug encapsulation, sustained drug release, targeted drug delivery, enhanced permeability and retention along with a lower incidence of side effects. A Promising strategies for optimizing drug delivery could be based on liposomes, niosomes, microparticles, microemulsions, nanoemulsions, gold nanoparticles, silica nanoparticles, carbon nanomaterials, polymeric micelles, CD, dendrimers, engineered antigens, hydrogels, and liquid crystals. B The promising preclinical developments for TB treatment involve varying routes of administration, such as oral, intranasal, pulmonary, topical, intramuscular, intravenous and subcutaneous routes. Newer approaches include transdermal patches, floating systems, and a one-time large-dose controlled-release gastrointestinal resident delivery system

Nanocarriers offer prominent advantages, such as drug release in the presence of specific triggers, providing temporal control of drug exposure, enhanced drug uptake in target cells, improved efficacy against intracellular pathogens, and protection of labile therapeutic agents from harsh physiological conditions such as low pH or enzymatic degradation [61]. In particular, polymers such as poly(lactic-co-glycolic acid) (PLGA), poly(caprolactone), poly(anhydrides), poly(orthoesters), poly(cyanoacrylates), and poly(amides) have opened avenues to modify drug release patterns by altering the monomer hydrophobicity, polymer chain length and particle size [62]. However, encapsulation of both hydrophobic and hydrophilic drugs is possible with liposomes to achieve sustained release [14].

Recent advances in nanotechnology have brought carriers into the limelight, which can specifically target AM [30]. Modulation of the physical properties of drug carriers, such as surface composition, charge, shape, particle size, hydrophobicity, and zeta potential, can modify drug internalization by AM. Another approach has been to use ligands on the nanocarrier that interact with specific receptors on macrophages, termed active targeting or ligand-mediated targeting. Strategies that do not rely on specific ligands have been called passive targeting. For passive targeting of AM, polymer- and polysaccharide-based carriers, as well as liposomes, solid lipid nanoparticles (SLNs), and gold nanorods, have been utilized for numerous agents, mostly INH and RIF [30]. Polysaccharides and their derivatives, including chitosan, inulin, alginate, and CD, have seen increased applications owing to their biocompatibility, biodegradability, hydrophilicity, mucoadhesive properties, use in modifying carrier surface charge, and target specificity [63,64,65,66].

Despite the beneficial prospects of these modern and fabricated drug delivery systems, safety and toxicity need to be verified, as they comprise different types of material [67, 68]. These technological solutions need to support scalability and reproducibility, aiding clinical translation beyond laboratory optimization by overcoming manufacturing, regulatory and financial challenges [69]. In particular, the reformulation of existing drugs for enhanced efficacy and safety needs to be cost-effective [70]. Furthermore, the synthesis and storage conditions need to be conducive to conditions in low-resource countries [71]. Importantly, process optimization is of utmost significance in the case of nanomedicines that are likely to be 3D constructs of multiple components with preferred spatial arrangements, with any deviation adversely affecting the composition [72, 73]. Table 2 provides a summary of drug delivery systems and their key findings incorporating anti-tubercular drugs.

Table 2 Drug delivery systems for anti-tubercular drugs

Lipid-based drug delivery systems

Liposomes

The efficiency of drug delivery by liposomal systems has mainly been studied using M. avium and M. tuberculosis models. In a mouse model, liposomal rifabutin was demonstrated to slow the pathogenic course of TB infection. Formulations containing dipalmitoyl phosphatidylcholine and dipalmitoyl phosphatidylglycerol were able to reduce TB progression in the lungs and lowered the bacterial loads in the spleen and liver, implying that liposomal-loaded ATDs could be a promising approach for treating extrapulmonary TB [104]. Similarly, liposomal clofazimine showed higher antibacterial activity than free clofazimine against the M. avium complex in mice, even when the treatment was initiated after the dissemination of the infection [108].

RIF and INH-encapsulated liposomes reduced pulmonary inflammation and enhanced the survival of TB-induced mice. Liposome formulation improved RIF penetration across the alveolar epithelium, extending pulmonary residence time and lowering systemic drug toxicity [116]. Mannan-anchored liposomes containing RIF, INH, and pyrazinamide can be delivered to the lungs using a dry powder inhaler (DPI) for the treatment of pulmonary TB with high entrapment efficiency and sustained drug release [117].

The liposome-in-hydrogel technique was found to be promising for treating bone TB locally. Liposomes entrapped with INH served dual functions of pulmonary medication transport and alveolar stabilization. DPIs containing ligand-anchored pH-sensitive liposomes for the simultaneous delivery of INH and ciprofloxacin demonstrated the greatest accumulation in the lung. Liposomes can aid in the pulmonary administration of ATDs, which could be an attractive alternative to improve TB therapy [74, 118, 119].

Asymmetric liposomes made up of phosphatidylserine at the outer surface resembling apoptotic bodies and phosphatidic acid at the inner layer could be used to boost innate antimycobacterial activity in phagocytes while limiting the inflammatory response [120]. Liposomes containing dipalmitoyl phosphatidylcholine and cholesterol were reported to inactivate M. tuberculosis and multidrug-resistant (MDR) -M. tuberculosis, with action dependent on the incubation period and low dose [121]. Phosphatidylcholine-cholesterol-cardiolipin liposome formulation and levofloxacin are efficacious against M. tuberculosis bacilli [103]. Liposomes may potentially be useful as part of a gene vaccine for TB treatment, as evidenced by the efficacy of a peptide-DNA-cationic liposome pseudoternary complex [122].

Niosomes

INH integrated with niosomes was found to be effective against TB, with 62% cellular absorption by macrophages and 65% drug localization to the target organ, compared to 15% with the supplied free INH [123]. Liposomal preparations of azole antifungals such as clotrimazole and econazole were shown to be effective against M. tuberculosis and latent bacilli, while fluoroquinolones such as moxifloxacin accumulated more efficiently in AM when delivered in the context of niosomes [123].

Chowdhury et al. [208] and El-Ridy et al. investigated niosomal formulations of RIF and ofloxacin for the treatment of drug-resistant TB, finding 81.76% entrapment efficiency and regulated release for up to 15 days. El-Ridy et al. formulated ethambutol-containing niosomes that demonstrated regulated release and a reduction in nonspecific toxicity [99]. After intratracheal injection, RIF-loaded niosomes demonstrated enhanced accumulation and 90% RIF release in 48 h, with 65% localization [125]. Niosomes containing cholesterol and Triton X-100 successfully targeted RIF in Wistar rats, INH in J744A.1 mouse macrophages, and ethambutol in Swiss albino mice infected with M. tuberculosis H37Rv [76, 126]. In guinea pigs, pyrazinamide encapsulated in niosomes demonstrated significant drug entrapment efficiency. Subcutaneous treatment with niosomal ethambutol formulated with Span 60, Span 85, cholesterol, diacetyl phosphate, and stearyl amine resulted in prolonged drug release in Swiss albino mouse lungs and lowered bacterial counts in guinea pigs infected with M. tuberculosis H37Rv by i.m. injection [99, 127].

For highly lipophilic drugs, such as BM859, which demonstrated significant antimycobacterial action against M. tuberculosis H37Rv, niosomes are a preferable method of drug delivery. Sadhu et al. developed ethionamide niosomes with sufficient stability for intravenous injection [128, 129]. Niosomes loaded with hydrophilic D-cycloserine and lipophilic ethionamide kill drug-resistant TB by releasing 96% of ethionamide and 97% of D-cycloserine [130]. Antimycobacterial drugs encapsulated in tyloxapol niosomes have a high drug loading efficiency, and the isatin-INH hybrid WF-208 has a fourfold higher MIC against H37Rv M. tuberculosis [131,132,133].

Liquid crystals

Liquid crystals are an ideal mechanism of drug delivery for ATDs. Liquid crystal-based zidovudine, ciprofloxacin, and fluconazole formulations are some of the recent developments in the antimicrobial field, making them a possible approach for delivering ATDs as well [135]. K. Dua et al. synthesized RIF-based liquid crystals with higher solubility and stability in acidic environments than the free drug, allowing for lower dose frequency and increased bioavailability. RIF is poorly water soluble and degrades in the stomach, resulting in limited bioavailability [136]. Tran et al. created two RIF-loaded nanoparticles using neutral lipid monoolein and the cationic lipid N-(1-(2,3-Dioleoyloxy)propyl)-N,N,N-trimethylammonium methyl-sulfate (DOTAP), which decreased the MIC of RIF against S. aureus. The cationic charge helped in increased solubility and greater membrane fusion, as explained in Fig. 3 [91]. Kim et al. developed a liquid crystal-based aptasensor for IFN detection that has higher sensitivity and a lower detection limit than enzyme linked immunosorbent assay (ELISA). They created an improved biosensor that uses immobilized antigen ESAT-6 to detect anti-TB antibodies (anti-ESAT-6) [137]. Bedaquiline-loaded cubosomes based on nanocarriers have been demonstrated to be effective in the treatment of non-small cell lung cancer (NSCLC) by sustained release over 72 h [138].

Fig. 3
figure 3

© Elsevier Inc

Schematic representation of nonlamellar lyotropic liquid crystalline nanoparticles. A Cationic charge effect on the S. aureus cell membrane. B Chemical structures of monoolein (MO), cationic lipid 1,2-dioleyl-3-trimethyl-ammonium-propane (DOTAP), antibiotic rifampicin (Rif), and Pluronic F127 modified and reprinted with permission from [91]

Polymer-based drug delivery systems

Polymer micelles

Because of their capacity to stabilize and protect the drug, prolong the therapeutic activity, and enclose hydrophobic pharmaceuticals, polymeric micelles are used as drug carriers. They can also be utilized to deliver multiple ATDs into infected macrophages while causing fewer negative effects [139, 140]. Tripodo et al. created RIF-delivering micelles based on inulin functionalized with vitamin E (INVITE) and its succinylated derivative (INVITESA). It demonstrated strong mucoadhesion to mucin and equivalent antibacterial activities against gram-positive bacteria [94].

RIF- and INH-loaded N-(2-hydroxypropyl)methacrylamide-poly(lactic acid) micelles allow prolonged drug release, improving effectiveness against resistant and sensitive pathogens. Kaur et al. revealed different polymeric micelle-based delivery methods for ATDs for drug-resistant TB, including polyethylene oxide-polypropylene oxide (PEO-PPO), polyvinyl-caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer (PCL-PVAc-PEG), and chitosan-graft-poly-ε-caprolactone (CS-g-PCL). PEO-PPO decreased the MIC value, PCL-PVAc-PEG improved RIF solubility and physical stability, and CS-g-PCL provided INH with pH-dependent release, improved cellular internalization, and decreased cytotoxicity [141].

Yuan et al. created interconnected hydrogel micelles for the delayed release of weakly water-soluble RIF using biodegradable polymers such as guar gum, chitosan, and polycaprolactone. In vitro release experiments revealed that 90% of the medication was released in 12 days, and 97% of the encapsulation was effective [142]. Sheth et al. encapsulated RIF and INH in pluronic, which increased activity and sustained release against M. tuberculosis. Grotz et al. developed an inhalable nanocarrier based on RIF-loaded polymeric micelles to improve water solubility [96, 143].

Chitosan-based polymer drug delivery systems

Chitosan can be used as a vaccine delivery mechanism in the treatment of TB. Negatively charged particles, such as PLGA, can be coated with chitosan to transfer them efficiently to the mucosal membrane [144, 145]. Gu et al. recently proved that a combination of dihydroartemisinin and chitosan might overcome M. tuberculosis RIF resistance. This combination worked best at lower chitosan concentrations, but at greater concentrations, the bacteria were deprived of nutrition [146]. Chitosan biguanidine nanoparticles developed using a one-pot green synthesis method can be used as carrier systems for ATDs. It was demonstrated that chitosan biguanidine nanoparticles have enhanced pharmacological activity owing to targeted delivery [147].

Alginate-based polymer drug delivery systems

For MDR-TB, hydrophilic ATDs such as amikacin and moxifloxacin were encapsulated in alginate-entrapped PLGA nanoparticles. When these nanoparticles were fed to macrophages infected with M. tuberculosis, antibacterial activity was detected [148]. For the administration of RIF in combination with ascorbic acid, a nanocarrier system comprised of alginate coated with Tween 80 and chitosan was created [149]. Alginate-cellulose nanocrystal hybrid nanoparticles have demonstrated significant antimycobacterial action and moderate oral medication delivery difficulties [93]. Anti-TB action has also been shown in nanostructured polyelectrolyte complexes synthesized from sodium alginate and chitosan. Alginate particles have been investigated as a means of encapsulating live mycobacterium particles for use in inhalable vaccinations [150, 151].

Nagpal et al. proposed coating live mycobacterium with alginate to improve dendritic cell activation and maturation [150]. When alginate-coated chitosan nanoparticles were delivered intranasally and subcutaneously, they released the PPE17 antigen, which produced effective immune responses in mice [152]. Pregelatinized sodium alginate and chitosan can be used for the development of nanoparticles of INH and pyrazinamide and could be an interesting approach for TB treatment [153]. To improve stability and long-term release, polypeptidic micelles containing bedaquiline were coated with sodium alginate [154].

Zn-alginate beads show excellent biocompatibility and no fatal cytotoxicity when utilized as carriers for RIF administration [155]. Alginate has been employed as a stabilizer in the manufacture of silver nanoparticles, which have the ability to attack M. tuberculosis and sterilize nonreplicating persistent TB [156]. It has been discovered that calcium ion-sodium alginate-piperine-based microspheres improve entrapment efficiency and extend the release and oral bioavailability of INH [82].

Cyclodextrin-based polymer drug delivery systems

CDs are cyclic oligosaccharides with D-glucose units linked by β-1,4-glucosidic linkages [157]. Because of the constrained rotation around the bonds joining the glucopyranose units, they form a toroidal structure [158,159,160]. β-Cyclodextrins (β-CD) have the potential to be an effective carrier system for ATD delivery. An in vivo investigation found that administering unloaded β-CD via endotracheal or intranasal routes reduced the bacterial burden. Loaded with ethionamide and boosted with BDM43266, the bacilli activity was tenfold increased and selective. This can be used to combine several medications into a single formulation [95].

RIF is insoluble and permeable, but this can be addressed by creating an inclusion combination with hydroxypropyl- β -CD (HP- β -CD). To simplify dose modification and treatment adherence, Javier Suárez-González et al. developed a combined-dose oral pediatric formulation including INH and RIF. HP- β -CD has also been employed to create a powdered RIF dosage form for direct lung-focused distribution [95].

The bioavailability and deposition performance of clofazimine were increased by combining it with β-CD and L-leucine [110]. Curdlan nanoparticles containing RIF and levofloxacin were used to target M. tuberculosis-infected macrophages [161]. In mice, the combination of ethionamide with the booster BDM41906 decreases mycobacterial load [112]. Christian and Werner successfully complexed an INH-hydrazone-phthalocyanine compound in β-CD encapsulated in soybean lecithin liposomes. It demonstrated pH-dependent drug release that is appropriate for site-specific delivery [162]. Anjani et al. discovered that CD inclusion complexes improved antibacterial action, with 60% drug release in 2 h [139].

Dendrimer-based drug delivery systems

Polymers commonly used for the preparation of dendrimers are poly(amidoamine) (PAMAM) and poly(propylene imine) (PPI) [163,164,165]. Others include polyglycerol, poly(ether hydroxylamine) (PEHAM), poly(ester amine) (PEA), and melamine [166]. Dendrimers behave like unimolecular micelles that facilitate the delivery of both hydrophilic and hydrophobic drugs [167,168,169]. Cationic dendrimers can be used as nonviral gene carriers [170].

The RIF-PAMAM complex can be used as a carrier for drugs to acidic sites, as normal RIF can lead to solubility issues [171]. When a maximum of twenty RIF molecules were loaded in fourth-generation PAMAM dendrimers, a sustained release profile was observed at neutral pH, whereas simultaneous release was triggered at acidic pH. Incorporating RIF into G3 PAMAM prolonged its release compared to first- and second-generation PAMAM due to the entrapment of RIF in the branched chains of G3 PAMAM along with the high density, high molecular weight, and size of G3 PAMAM [173].

The solubilization of drugs is due to hydrophobic-hydrophilic interactions, the interaction between ions, and encapsulation of hydrophobic drugs into crevices of dendritic architecture, as demonstrated by Karthikeyan R et al. using known concentrations of PEGylated PPI dendrimers [174]. Fourth- and fifth-generation PEGylated PPIs also demonstrated an increase in the entrapment of RIF [175]. Furthermore, targeting of AM was studied using RIF with fifth-generation ethylenediamine (EDA)-PPI dendrimers based on mannosylation to selectively target macrophages. The outcome was that the concentration of the drug in the macrophages exceeded the plasma concentration, making it a promising approach for the treatment of TB [85].

Poorly aqueous or hydrophobic drugs can be encapsulated within the core of PEHAM dendrimers to improve their solubility, which further increases the bioavailability of the drug [176, 177]. Since the cationic groups present in the PEHAM dendrimers are toxic to RBCs, methods such as glycosylation and acetylation are extensively employed to overcome the toxicity [178, 179]. ATDs can also be encapsulated in polymers to increase their half-life and attain enhanced bioavailability and a sustained release profile [180].

Polymeric-microparticulate drug delivery systems

The inclusion of RIF and INH into polylactic acid microparticles (MPs) at a 1:1 ratio increased the drug concentration in macrophages, lowering the dosage frequency and toxicity [181]. For the detection and treatment of pulmonary TB, monocyte-derived MPs can be utilized to target AM. To release entrapped pharmaceuticals, sodium alginate, a linear copolymer of α-guluronic acid and α-mannuronic acid, creates a meshwork with divalent cations [182]. Recently, β-1,3/1,6 glucan particles (GPs) produced from yeast have been used to deliver anti-TB medicines to macrophages [183, 184]. Macrophages and other phagocytic cells can recognize the β-1,3-D glucan surface makeup. Particulate glucan is biodegradable and biocompatible, and the United states Food and Drug Administration (FDA) considers it generally recognized as safe (GRAS). In a study, RIF DPI was developed that had the ability to overcome drug resistance as well as reduce the time needed for therapy [185].

INH-administered mannitol microspheres containing iron(III) trimesate metal organic framework (MOF) MIL-100 nanoparticles demonstrated adequate encapsulation efficiency and aerodynamics for pulmonary delivery. In vitro testing in human alveolar adenocarcinoma basal epithelial cells revealed effective internalization, indicating that it is suited for deep lung ATD administration [80]. Fucoidan microparticles loaded with ATDs demonstrated good affinity, aerodynamic features, and no cytotoxicity to lung epithelial cells or THP-1 macrophages [186]. ATDs suppressed 95% of microbial growth and triggered cytokine-mediated macrophage activation even when used as a single formulation [187]. Chitosan polymeric MP loaded with INH and rifabutin showed comparable and increased efficacy against M. bovis [188].

Host defense peptide (HDP) microencapsulation with INH demonstrated enhanced and additive antimycobacterial effects, resulting in a lower dosage concentration. Antimycobacterial action was also demonstrated by encapsulated astaxanthin [189, 190]. Following oral dosing, epigallocatechin gallate (EGCG) exhibited a modest therapeutic effect, while microencapsulated EGCG with trehalose sugar (EGCG-t-MS) demonstrated dose-dependent death of TB bacteria in mouse macrophages. In vivo, pulmonary delivery of EGCG for 6 weeks resulted in lower bacterial loads, less inflammation, and fewer granulomas than orally administered EGCG. Combination therapy with EGCG-t-MS with a subtherapeutic dose of regular ATDs demonstrated efficacy comparable to full-dose therapy [191].

The bioavailability of RIF-loaded poly lactic co glycolic acid (PLGA) microsphere powders after intratracheal aerosolization was 92%. Other RIF, INH, pyrazinamide, rifabutin, and linezolid microencapsulated formulations have been studied as prospective NDDSs for prolonged release, infrequent dosage, and adequate bioavailability [192, 193]. Anisimova et al. discovered that INH, streptomycin, and RIF encapsulated in poly(butyl cyanoacrylate) (PBCA) and poly(isobutyl cyanoacrylate) (PIBCA) accumulated more intracellularly in monocytes than free drugs [148]. INH, streptomycin, and RIF encapsulated in PBCA and PIBCA accumulated intracellularly, producing a more effective response [131]. Table 3 represents a summary of drug delivery systems for anti-tubercular drug combinations and new approaches.

Table 3 Drug delivery systems for anti-tubercular drug combinations and new approaches

Floating drug delivery methods are low-density devices that allow the drug to float on top of the stomach juice, boosting retention time and bioavailability. Quercetin-loaded RIF floating microspheres were developed to treat TB and maintain RIF release in the stomach, and they were found to be stable after six months [194]. RIF stability can be increased by integrating it into sustained-release microporous floating microspheres and gastric-resistant INH sustained-release microspheres. Microporous floating sustained release microspheres were created using emulsification and evaporation techniques, resulting in increased RIF bioavailability [195]. Because one drug can be enteric coated to release in the stomach and the other in the ileum, floating delivery systems are useful for ATD fixed-dose combos. Studies have shown that more RIF is absorbed from the stomach even in the presence of INH [196].

Inorganic nanoparticles

Gold nanoparticles

Gold nanoparticles (GNPs) have several applications in targeted drug delivery due to their small size, biocompatibility, and lack of cytotoxicity. Green synthesized GNP with herbs may be beneficial in the treatment of TB. The various types of GNPs and their general synthesis methods are depicted in Fig. 4. GNP exhibits bactericidal activity [216]. With respect to TB, GNP was able to inhibit M. tuberculosis with an MIC of 10 µg/ml but was ineffective against RIF-resistant M. tuberculosis [217]. Gold nanoparticles synthesized using the bacterium Zoogloia ramigera exhibited good antibacterial activity and can be utilized for TB treatment. The antibacterial properties were studied using MIC and minimum bactericidal concentration methods [218]. Mesoporous silica nanoparticles (MSNs) containing gold nanoparticles (MSNs@GNP) can inhibit M. tuberculosis growth and produce a synergistic effect against M. tuberculosis, making it safe for TB treatment [219]. Another important application of GNP is in TB diagnosis. A ferromagnetic GNP-based immune detection system was developed for the detection of M. tuberculosis and to differentiate M. bovis [220]. GNPs loaded with quadruplex DNA motifs can aid in the diagnosis of M. tuberculosis in sputum [15, 221]. Magnetic beads and GNP-based immuno-PCR assays were developed to detect M. tuberculosis antigen [222].

Fig. 4
figure 4

Types of gold nanoparticles and synthesis method. Various types of gold nanoparticles are used for the delivery of ATDs. A GNPs are synthesized in various shapes according to the requirements of the delivery system. B The synthesis of nanoparticles follows a systematic sequence of steps and can be surface modified to target the nanoparticle to the desired site. C The systemic administration of GNPs in preclinical models has been found to enhance the efficiency of drug delivery, thereby improving the action of ATDs

Silica nanoparticles

Silica nanoparticles have the ability to be taken up by macrophages and produce immunological benefits [223]. Polyethyleneimine (PEI)-coated MSNs loaded with RIF exhibited effective targeted intracellular delivery with decreased cytotoxicity [224, 225]. MSNs loaded with first-line ATDs can kill M. tuberculosis-infected macrophages [201, 226]. MSNs containing NZX (mycobacterial peptide) can effectively treat TB by killing MDR strains of M. tuberculosis [227]. Tenland et al. found that MSNs can increase antibacterial activity against M. bovis and M. tuberculosis H37Rv in vitro and in vivo [227]. NapFab, an antimicrobial peptide isolated from bronchoalveolar lavage, showed excellent antimycobacterial activity when introduced into dendritic MSNs. MSNs can be used as carriers for the delivery of silver nanoparticles to target sites because they have very high bactericidal potency. A 2D hexagonal MSN containing silver bromide also showed good antimycobacterial activity [228].

M. tuberculosis produces extracellular vesicles that can cause immunomodulatory responses. Nanodrug delivery systems such as MSNs can mimic endogenous vesicles and act as carriers of the vesicle-associated proteins Ag85B, LprG, and LprA. They have been studied for the development of vaccines against TB [229]. Acetophenone helps MSNs deliver clofazimine to the target site [230]. Oral drug delivery of antitubercular nitroimidazopyrazinone analogs-pretomanid and MCC7433 from the bicyclic nitroimidazole class can be improved by MSNs. The MCM-41 type of SNP was used as a carrier for the transport of poorly water-soluble bicyclic nitroimidazole compounds [231]. MSNs are a promising multifunctional drug delivery system due to their high drug loading capacity and stability [232].

Carbon nanotubes

Carbon nanomaterials have gained popularity due to their unique features, which include physiochemical, thermal, optical, and electrical properties. Carbon nanotubes are the most well-known structures with a continuous cylinder formed of graphene [233]. Zomorodbakhsh et al. 2020 linked INH with multiwall carbon nanotubes (MWCNTs), which demonstrated greater lethality against M. tuberculosis even at considerably lower concentrations than the free drug [79]. Chen et al. developed chitosan nanotubes based on INH nanoparticles to increase medication release time, accelerate TB ulcer healing, and minimize inflammation and cytotoxicity [78]. Tudose et al., Moradi et al. and Pi et al. used a graphene oxide carrier system with various surface modifications for the delivery of ATDs and were found to have superior control over drug release [97, 198, 206].

More et al. developed a graphene oxide-based air-dried hydrogel containing para-amino salicylic acid for targeting MDR TB, whereas Vatanparast et al. revealed that AlN- and AlP-doped graphene quantum dots (GQDs) can be utilized to transport INH [7, 77, 114]. INH- and fluoxetine-conjugated MWCNTs increased the antimycobacterial activity and were capable of regulating the expression of the INH resistance genes inhA and katG, as shown in the schematic representation in Fig. 5 [207]. Carbon nanomaterials have been used to create electrochemical biosensors for M. tuberculosis detection, such as an amperometric DNA biosensor and a microfluidic multiplexed platform based on carbon nanotubes [234].

Fig. 5
figure 5

Chemical structures of the MWCNTs conjugated with therapeutic molecules and aspects of the release of cargos. Reprinted from [207] CC BY license

Emulsion-based drug delivery systems

Microemulsions

Microemulsions can be utilized to deliver targeted drugs, control release, and improve ATD bioavailability. Mehta et al. [202] formulated a highly stable Tween 80 microemulsion using RIF and INH [235]. Tween-based microemulsions of ATDs such RIF, INH, and pyrazinamide were found to be less hazardous and irritating, with INH releasing faster in the continuous phase and RIF releasing faster in the droplet phase [202]. The encapsulation efficiency, shape, antimycobacterial activity, particle size, and zeta potential have all been improved using modified microemulsion procedures [100, 236]. These medication delivery technologies can significantly reduce dose frequency while increasing bioavailability. Kaur et al. developed a Brig 96 microemulsion to work in combination with lipophilic and hydrophilic drugs [203]. Microemulsions of INH, pyrazinamide, and RIF exhibited good antibacterial characteristics [237].

Using the microemulsion process, calcium phosphate nanocontainers can be loaded with 1,3-benzothiazine-4-one, a new antimycobacterial drug that results in an increase in local drug concentration at the site of mycobacterial infection. Eugenol, an active chemical ingredient, was also combined with Tween 20 as a surfactant to improve therapeutic efficacy against M. tuberculosis [238]. According to Talegaonkar et al., microemulsions enhanced drug solubility and absorption, making RIF more effective and less toxic [239]. Microemulsions were also discovered to be promising for the regulated administration of ATDs as well as the destruction of drug-resistant strains of M. tuberculosis [240].

Nanoemulsion

M. tuberculosis can affect the eyes and cause an ocular infection resulting in permanent vision loss [98]. In this case, ATDs have to overcome the obstacle of the blood‒retinal barrier for effective drug movement, decreasing bioavailability. A solution could be the formulation of drugs into nanoemulsions by using excipients such as chitosan and polymyxin B [241]. For instance, a cationic RIF nanoemulsion was produced by high-pressure homogenization in a way that does not affect the therapeutic efficiency of the drug while enhancing bioavailability and other pharmacokinetic parameters [98]. Nanoemulsions were also used to develop a thermostable adjuvanted vaccine against TB by the "design of experiment (DoE)” approach [211].

Hydrogels

Hydrogel-forming microneedle arrays were designed for the transdermal delivery of ATDs, enabling the administration of high doses of ATDs [242]. Transdermal injection of hydrogel-based medicines has been proven to improve antibiotic activity against M. tuberculosis infection. For in vitro permeation, three distinct drug reservoirs were developed and combined with hydrogel-forming microneedle arrays. When the microneedle arrays were paired with polyethylene glycol tablets, immediately compressed tablets, and lyophilized tablets, the maximum penetration of RIF, ethambutol, INH, and pyrazinamide was attained [81].

More et al. formulated a graphene-based hydrogel that contained PAS and had good biocompatibility and antimycobacterial capabilities [7]. Wan et al. synthesized a variety of cationic peptide amphiphiles capable of self-assembling hydrogels [243]. A graphene oxide air-dried hydrogel designed to target M. tuberculosis also showed excellent antibacterial activity [7]. Polyvinyl alcohol (PVA) is nontoxic and has a high effectiveness for the encapsulation of hydrophilic pharmaceuticals, and a PVA-chitosan-tripolyphosphate hydrogel was developed for the extended release of ATDs. In a phosphate buffer solution (pH 7.4), a formulation of 80% PVA—20% chitosan hydrogel matrix demonstrated the maximum rate of drug release in a short period of time, with varied release patterns for RIF, INH, ethambutol, and pyrazinamide [204, 205].

Hydrogel interconnecting micelles made with guar gum/chitosan/polycaprolactone can serve as effective carriers for poorly water-soluble medicines such as RIF [142]. TB-Gel is an injectable and nonimmunogenic amphiphilic-based drug delivery technology with a low molecular weight. In an experimental mouse model, it was found to be more effective than oral delivery of a combination of four medicines in lowering mycobacterial infection [18]. Because of their swelling behavior, wide size range, and biocompatibility, hydrogels are effective in resolving these problems [244].

3D-printed formulations

3D printing techniques could truly advance the application of modern drug delivery systems in TB treatment by precision in the fabrication of scaffolds with well-controlled inner structures and pore morphologies, as depicted in Fig. 6 [245]. The prospects could be enhanced by the use of biocompatible and biodegradable polymers, such as polycaprolactone, as the binder for 3D printing [246].

Fig. 6
figure 6

© American Chemical Society

Possibilities with 3D printing techniques: Various biopolymers and ceramics can be incorporated with therapeutics by utilizing various 3D printing techniques. Modified with permission from [245]

A 3D-printed scaffold containing drugs such as INH and RIF can minimize the occurrence of drug resistance in osteoarticular TB [199]. In osteoarticular TB debridement procedures, drugs are placed into mesoporous and bioactive ceramics that bond with poly(3hydroxybutyrate-co-3-hydroxyhexonate) (PHBHHx) [246].

A bioengineered delivery system, such as 3D-printed tablets with discrete compartmentalization for RIF and INH, has the advantage of reducing drug degradation, allowing for successful combination treatment [247]. Bilayered tablets with two different ATDs, INH and RIF, can be designed and manufactured using a 3D-printed scaffold. The pH-sensitive polymer in which these medicines are contained determines the site of release. These methods can reduce pH-dependent deterioration and improve therapeutic efficiency [200].

Quercetin, a flavonoid, has been found to limit the growth of M. tuberculosis H37Rv. To combat the devastating effects of pulmonary TB, 3D printing technology was employed to create medicinal skin patches. Pharmacokinetic studies in rats revealed the viability of creating 3D-printed medicinal skin patches that may deliver plasma levels for up to 18 days following a single application [248, 249].

Advanced therapeutic strategies

Targeted therapy

Among polymers, biocompatible PLGA has found profound use in the fabrication of controlled ATD delivery systems because of the ease in achieving the desired dose and release kinetics by modification of the lactide to glycolide ratio, molecular weight, and drug concentration [250,251,252]. With regard to tissue-resident macrophage targeting for the delivery of ATDs, the surface of the drug carrier is functionalized with ligands, including mannosylated molecule, β-glucan, curdlan (β-1,3 glucose), folic acid, hyaluronic acid, tuftsin peptide, and phosphoserine conjugate, that can be recognized by corresponding receptors on macrophages, such as mannosyl receptor (CD206), dectin-1 receptor, folate receptor, tuftsin receptor, hyaluronic acid receptor, fucosyl and scavenger receptor, Fc receptor, transferrin receptor, formyl peptide receptor (FPR), and other lectin-like receptors [30, 253, 254] represented in Fig. 7. Among them, drug carriers made of modified mannose are the most common and can be added to liposomes, SLN, and polymer micelles [255,256,257] Studies have shown that cubosomal lipid nanocarriers exhibit higher drug delivery efficiency as well as bioavailability than conventional formulations. They are not only effective against free bacilli but also have the ability to deliver drugs to intracellular bacilli [258]. Thus, the identification of better sets of ligands with higher binding affinity for macrophage-based receptors might result in enhanced targeting efficiency. A promising approach could be heteromultivalent targeting, in which different types of ligands bind to different macrophage receptors simultaneously [259]. With respect to targeting and drug accumulation at sites of infection, future studies need to focus on the translation of preclinical data into humans in relation to the severity of infection, the fraction of drug at off-target sites, and the drug targeting index [14, 260].

Fig. 7
figure 7

Targeting infected alveolar macrophages (AM). In active targeting, ligands are incorporated into the drug carrier, which interacts with specific receptors on AM, leading to ligand‒receptor-mediated phagocytosis. In passive targeting, the surface of the carrier lacks a host-specific ligand. The macrophage surface receptors that can be utilized for active targeting include the formyl peptide receptor, Toll-like receptor, folate receptor, Fc (fragment, crystallizable) receptor, tuftsin receptor, mannose receptor (CD206), complement receptor, hyaluronic acid receptor (CD44), scavenger receptor, fucosyl receptor, Dectin-1 receptor and lectin-like receptors. Common ligands used to target macrophages include a Mannosylated molecule, b Phosphoserine conjugate, c Folic acid, Hyaluronic acid, e Tuftsin peptide, Curdlan (β-1,3 glucose). Concept adopted from [30]

Pulmonary TB is the most common form of TB, and inhalable carriers of ATDs have been a major focus of research and were found to significantly increase targeting in the lungs, reducing undesirable toxic side effects and enabling delivery to AM [9, 14]. Liposomes [261], microparticles [186, 187, 262], microencapsulation [80], liquid crystals [263], hydrogels [244], polymeric micelles [96] and even hybrid systems [264] have been promising for inhalational TB therapy. The formulations need to be optimized to penetrate mucous layers and biofilms and overcome sequestration, rapid deactivation by enzymes, and elimination by coughing [265, 266]. In addition to passing through the acidic gastric environment, pulmonary administration could be particularly beneficial for controlled release within AM. However, for translatable pulmonary administration of ATDs, challenges need to be resolved, including the use of better and safer excipients, drug encapsulation efficiency, process and production scalability, and developing formulations with optimum size and morphology for deep lung deposition [124, 267,268,269]. If sufficient drugs can be delivered through the inhaled route, it would be of immense benefit to TB patients considering the acceptance of portable, cost-effective and easy-to-operate inhalers [23].

Unlike the conventional administration of drugs, nanomaterial-based systems offer significant benefits, such as ease of administration, minimal side effects, and addressing the pharmacodynamics and pharmacokinetic limitations of many potential drug molecules [270]. Recently, transforming growth factor (TGF)-β1-specific siRNA nanoliposomes loaded with INH, RIF, and pyrazinamide have demonstrated the potential for improving spinal TB chemotherapy [271]. With the adoption of a phage-based delivery system for endogenous type III-A CRISPR‒Cas antimicrobials against M. tuberculosis, nanoenabled CRISPR‒Cas-powered strategies might also be developed for the treatment of TB [272,273,274]. Thus, nanotechnology holds immense potential in developing novel and targeted delivery systems for new therapies as well as existing drugs. This could be of interest, particularly in the development of inhaled or orally delivered nanocarriers for extended release of ATDs, which in turn can reduce the required dosing frequency to improve patient adherence. Drug depot systems can be of remarkable benefit, particularly for the treatment of TB in children, if balance can be achieved between the ease and safety of administration [275,276,277].

Long-acting therapeutics

Long-acting therapeutics can be formulated using prodrugs that have low aqueous solubility, inhibit rapid dissolution and drug release, and have a reasonably long half-life, enabling slow elimination from the body and high potency, allowing low drug doses to be injected [278,279,280]. In this regard, drugs such as bedaquiline, which has a longer half-life (24 h), higher lipophilicity (logP 7.3), and lower MIC for M. tuberculosis (0.03 μg/ml), were found to be suitable for use in a long-acting injectable (LAI) formulation [278, 281, 282]. Remarkably, one intramuscular injection of long-acting bedaquiline at 160 mg/kg demonstrated significant antitubercular activity for 12 weeks in p mouse models. Moreover, physiologically based pharmacokinetic modeling identified delamanid and rifapentine as potential LAI candidates suitable for monthly intramuscular administration at doses of 1500 mg and 250 mg, respectively [282]. A promising development is the one-time large-dose controlled release delivery system resident in the gastrointestinal tract. It offers numerous advantages over currently available injectable depot formulations, including ease of administration, lack of immunologic reactions, and the ability to accommodate multigram-level dosing in line with current TB treatment regimens [283].

In another approach, a thermoresponsive matrix containing an extended-release polymer was used to encapsulate drug molecules to improve the duration of action. Significant efficacy was achieved using sustained release intramuscular injection loaded with tin protoporphyrin (SnPPIX), a heme oxygenase-1 inhibitor, in murine models of pulmonary TB [284].

Furthermore, notable technological advancements for TB include developments in oral and subcutaneous systems [285]. Solid nanoparticles (SNPs) are widely studied for the oral delivery of antimicrobials, including SLNs, polymeric nanoparticles, MSNs, and hybrid nanoparticles [286]. SLN has been tested in rodents with the goal of improving the bioavailability of RIF and the combination of INH, RIF, and pyrazinamide [14, 287]. MSNs were able to enhance the activity of orally delivered poorly soluble antibacterial agents against TB, such as pretomanid and MCC7433, a novel nitroimidazopyrazinone analog [231]. SLN was also proposed as a suitable drug delivery platform with short-term sustained release upon intramuscular and subcutaneous administration [288].

Extrapulmonary TB therapy

ATD delivery for TB bone defects has been another area of focus in TB research, as bone TB has the highest incidence among extrapulmonary TB, accounting for approximately 35–50%. Even though surgery is available, the remaining M. tuberculosis around the trauma can multiply, leading to TB ulcer and sinus formation and even causing bone TB recurrence and bacterial infection. This, along with poor local blood supply that causes difficult access to ATDs, complicates the scenario [289]. A probable solution could be loading ATDs into scaffolds through various drug-loading techniques to improve the efficiency of anti-TB treatment [290]. As carbon nanotubes have strong penetrability across physiological barriers to enter tissues, chitosan/carbon nanotube nanoparticles were constructed to achieve slow release of INH. It was found to significantly promote the healing of TB ulcers and could be developed as a new treatment for secondary wounds of bone TB [78].

Another approach that has shown promising potential for osteoarticular TB therapy is biocompatible mesoporous bioactive glass/metal–organic framework (MBG/MOF) scaffolds fabricated by a 3D printing technique using polycaprolactone [246]. MBG is considered a promising material owing to its bone repair potential in relation to its high surface area and better bioactivity, along with its superior drug loading and release ability [291,292,293]. The biomedical applications of MOFs are related to their tunable porosity, biocompatibility and biodegradability, making them an attractive drug delivery system with a modifiable degradation rate for controlled drug delivery [294, 295].

In comparison, the cutaneous administration of ATDs is poorly explored. Skin can be considered a good route for the treatment of cutaneous TB, usually caused by atypical mycobacterium species, namely, M. leprae, M. hemophilum, and M. ulcerans. TB represents only 1–1.5% of extrapulmonary cases, affecting mainly the face, torso, and neck areas [296]. However, the incidence of extrapulmonary TB is increasing in the context of MDR-TB. Recently, van Staden et al. proposed the utility of self-double-emulsifying drug delivery systems (SDEDDS) containing clofazimine for topical delivery in the treatment of cutaneous TB [297]. The benefit of SDEDDS for dermal administration of clofazimine is that, with lower drug concentrations, it could provide consistent drug delivery profiles that will be cytotoxic toward M. tuberculosis, which can help to suppress drug resistance [298]. As a topical delivery, it might be able to reduce the unpleasant discoloration associated with oral administration of clofazimine [299]. The improved drug loading capacity of SDEDDS may be further utilized to treat active TB or resistant TB infections by either including higher concentrations of clofazimine or incorporating fixed-dose drug combinations with other drugs known to act synergistically [300].

The possibility of INH delivery by the skin route has also been evaluated, as this route could avoid the hepatic first-pass effect, thereby reducing hepatotoxicity that leads to poor patient compliance [301]. The selection of excipients is based on the intended use. For example, limonene was found to be the better excipient for transdermal formulations based on the enhancement of INH absorption, while transcutol and menthol were found to be more appropriate for topical systems. Inclusion of transcutol led to increased skin accumulation of the drug, termed the "intracutaneous depot", created by swelling of stratum corneum intercellular lipids that retained the drugs, along with a simultaneous decrease in transdermal permeation. Interestingly, the incorporation of limonene resulted in transdermal absorption of INH that was sufficient to ensure a systemic therapeutic effect [301]. Moreover, to achieve transdermal drug delivery, cutting-edge anti-TB drug delivery systems are being explored, such as 3D printed quercetin-coupled polyvinylpyrrolidone (PVP) skin patches for the treatment of destructive pulmonary TB [249].

Another avenue is ophthalmic drug delivery for the treatment of ocular TB, in which eyes and orbital tissues are affected, leading to ocular morbidity and visual loss [98, 302]. Even though it is a comparatively rare extrapulmonary manifestation, ocular TB may be the first presentation of TB in initially asymptomatic patients, especially since 92% of patients with ocular TB present without evidence of concomitant pulmonary TB [303,304,305]. A RIF-loaded cationic nanoemulsion with specific surface modification employing chitosan and polymyxin B was found to be promising to overcome the hurdle of the blood‒retinal barrier of the eye that hinders the availability of ATD delivered by the systemic route [30, 98].

Phototherapy

An emerging technology with promising application in TB therapy is combining ATDs with other treatment modalities, such as photodynamic and photothermal therapies [306, 307]. For instance, a targeted antibiotic-delivering nanoassembly was shown to exert chemo-photothermal therapy [307]. The core of the nanoassembly was composed of near-infrared (NIR) active gold nanorods (GNRs) coated with MSNs, which served as the carrier for bedaquiline. The assembly was wrapped within a thermosensitive liposome (TSL) conjugated to the mycobacteria-targeting peptide NZX, which mediated adhesion of the final nanoassembly on the mycobacterial surface and had intrinsic antibacterial activity. Upon NIR exposure, TSL undergoes a phase transition, becoming permeable due to the heat generated from the GNRs, releasing encapsulated bedaquiline. Hyperthermia also plays a role in increasing bacterial cell membrane permeability, causing leakage of bacterial cell contents and subsequent bacterial cell death. The final nanoassembly demonstrated remarkable antibacterial activity against M. smegmatis, which was 20-fold more efficacious than the free drug equivalent. Moreover, it successfully inhibited the growth of intracellular mycobacteria residing in lung cells, underlying its potential to treat latent pulmonary TB. The engineered nanoassembly was able to (1) control remote trigger release of encapsulated ATD upon exposure to NIR laser by melting of TSL, (2) increase internalization into infected host cells through TSL coating and offer targeted ATD delivery to the bacterial cell surface by NZX targeting peptide, thereby reducing off-target toxicity, and (3) demonstrate synergistic antibacterial activity due to encapsulated ATD and photothermal activity [307]. Furthermore, combined chemo-photothermal therapy based on an enzyme-responsive nanosystem could be a promising approach to combat drug-resistant bacteria[308]. Even photodynamic therapy could be a new option for the treatment of MDR- and XDR-TB, as it was able to inactivate M. tuberculosis clinical strains regardless of the drug resistance levels of the bacilli [8].

Immunotherapy

The immune system significantly impacts TB recognition, occurrence, development, and outcome. The disease's progression depends on genetics and environmental factors. In the initial stages, innate immune clearance is involved, while macrophages, neutrophils, dendritic cells, T cells, and NK cells form the first line of defense [309]. The interaction between host immunity and mycobacterial invasiveness affects the immune system response. If the invasiveness of bacilli is weak, macrophages eliminate it, generating trained immunity. If mycobacterial invasiveness is balanced with host immunity, bacilli may replicate, spread, and become active TB. TB-specific immunotherapy is needed to regulate the immune system's anti-TB response. Cytokines such as IL-2, IL-24, and IL-32 can be therapeutic targets against TB [310,311,312,313,314]. IL-2, a Th1 immune response cytokine, induces differential gene expression in peripheral blood mononuclear cells (PBMCs) stimulated by TB. Administration of rhuIL-2 immunoadjuvant enhances CD4 + T-cell proliferation and NK cell proliferation, improving the sputum bacterium-negative rate in MDR-TB patients. A multicenter clinical trial on rhuIL-2 as an adjuvant therapy for MDR-TB is being conducted in China (ClinicalTrials.gov Identifier: NCT03069534). IL-24, a novel tumor suppressor, inhibits IL-24 expression in human PBMCs, increasing susceptibility to TB [314,315,316,317]. NK cells, T cells, and macrophages play a crucial role in combating TB. IL-24 activates CD8 + T cells, producing IFN-γ and IL-32, which induce inflammatory cytokines such as IL-1, IL-6, IL-8, and TNF-α. Heat-killed TB stimulates PBMCs to produce IL-32, enhancing clearance by monocyte macrophages. Anti-TB antibodies also have protective effects on anti-TB immunity [318,319,320,321]. Antimicrobial peptides, small molecule peptides, can enter cells through the skin and placenta exhibiting bactericidal and immunotherapeutic effects on TB [322].

Antigens can alert immune cells and precipitate an immunological reaction [323]. If these antigens can be engineered in such a way that they target proteins of M. tuberculosis, such as ESAT-6, CFP-10, and TB 7.7, these antigens can be used effectively against TB bacilli [324]. Accordingly, amino acid polymers that self-assembled to form a hollow core-shaped nanobead were administered to TB patients and produced different cytokines, including IFN-γ, INF-α, IL-2, CCL3, and CCL11 [325]. The biopolyester and polyhydroxybutyrate beads were biocompatible, thereby minimizing adverse reactions. The evaluation of the engineered antigen was performed by interferon release assay [326]. This delivery system could not only be used for ATD delivery but also for TB diagnosis, especially in patients showing tuberculin skin test negative (TSTn) results, as it contains short overlapping synthetic peptides such as in the QuantiFERON-TB Gold in Tube test (QFT-GIT) [327].

Conclusion and future perspectives

In conclusion, advanced drug delivery approaches have the potential to revolutionize TB therapy by addressing the challenges associated with traditional treatment methods. By developing inexpensive and easy-to-administer delivery systems that offer extended drug release, dosing frequency could be reduced, thereby improving patient adherence. Direct targeting by selectivity toward both AM and tubercle bacilli using suitably designed drug carriers and specific ligands may counteract the ability of intracellular pathogens to evade antibiotic treatments. With better penetration of ATDs into lung cavities and necrotic lesions, the success rate of TB therapy could be increased. In recent years, there has been a rise in TB cases, particularly resistant forms, across the globe. Various strategies for combating TB have been established at different levels, including the WHO’s End-TB Strategy and the UN’s Sustainable Development Goals (SDGs) [328]. The ‘3P Project’ aims to unite researchers to develop a treatment strategy that lasts for at most one month for all forms of TB infections [329]. Remarkably, the Medicines Patent Pool (MPP) has facilitated the clinical development of promising investigational treatments for TB, such as sutezolid, a linezolid analog [330]. In addition to the prospect of new drugs for TB, a favorable approach has been to improve the aspects of drug delivery through technologies that can offer the flexibility to adopt better routes of administration, multiple drug encapsulation, sustained drug release, targeted drug delivery, enhanced permeability and retention along with a lower incidence of side effects [331]. This indeed has the potential to overcome patient nonadherence to long and frequent dosing regimens [275]. The challenges that need to be addressed with some of the current ATDs are their poor solubility, instability in gastric acid, and inability to penetrate AM, where the bacilli reside [14, 332].

Recently, a significant breakthrough has been made in the treatment of TB through the use of siRNA-loaded nanoparticles, which effectively silence genes specific to M. tuberculosis. Additionally, a lyophilized formulation of the emulsion-adjuvanted subunit ID93 with GLA-SE, a recombinant subunit antigen combined with a squalene emulsion containing glucopyranosyl lipid A (GLA), has shown promise in a phase 1 clinical trial (Clinical trials.gov identifier: NCT03722472). This thermostable vaccine formulation demonstrated safety and immunogenicity in healthy adults. Similar technology-based formulations have also undergone clinical trials (Clinical trials.gov identifiers: NCT01599897, NCT01927159, NCT02465216, NCT02508376, and NCT03722472), and updates on these trials and development status can be accessed through The Working Group on New TB Vaccines (WGNV) database (https://newtbvaccines.org/tb-vaccine-pipeline/).

Another formulation, a liposome suspension known as RUTI®, containing a mixture of antigens, is actively recruiting patients for phase 2 clinical trials (Clinical trials.gov identifier: NCT04919239). These advancements highlight the potential of advanced drug delivery strategies in addressing the challenges of TB treatment. Despite the promising research activity, progress in clinical trials has been relatively slow.

To further advance the field, it is imperative to focus on addressing research gaps related to drug delivery systems for TB management. These gaps include the need for targeted delivery to specific cells and tissues, enhancing drug bioavailability, optimizing drug release kinetics from delivery systems, ensuring biocompatibility and biodegradability, addressing immunogenicity concerns, enabling personalized medicine approaches, exploring combination therapy benefits, considering cost-effectiveness, navigating regulatory approval processes, promoting successful clinical translations and validations, and fostering interdisciplinary collaboration.

In conclusion, while recent developments in TB treatment using advanced drug delivery strategies are encouraging, continued efforts are required to bridge the gap between research advancements and clinical application. By focusing on the aspects, the field of TB drug delivery can overcome challenges and contribute to more effective and accessible treatment options for patients worldwide.

Data availability

Not applicable.

Abbreviations

AM:

Alveolar macrophages

ATDs:

Anti-tubercular drugs

CCL11:

Chemokine (C-C motif) ligand 11

CCL3:

Chemokine (C-C motif) ligand 3

CDs:

Cyclodextrins

CFU:

Colony-forming units

CRISPR:

Clustered regularly interspaced short palindromic repeats

CS-g-PCL:

Chitosan-graft-poly-ε-caprolactone

DNA:

Deoxyribose nucleic acid

DoE:

Design of experiment

DOTAP:

1,2-dioleyl-3-trimethyl-ammonium-propane

DOTAP:

N-(1-(2,3-Dioleoyloxy)propyl)-N,N,Ntrimethylammonium methyl-sulfate

DOTS:

Directly observed treatment short-course

DprE1:

Decaprenylphosphoryl-β-d-ribose 2’-epimerase

DPI:

Dry powder inhaler

EGCG:

Epigallocatechin gallate

EGCG-t-MS:

Microencapsulated EGCG with trehalose sugar

ELISA:

Enzyme-linked immunosorbent assay

FDA:

Food and Drug Administration

FPR:

Formyl peptide receptor

GLA:

Glucopyranosyl lipid A

GNPs:

Gold nanoparticles

GNR:

Gold nanorods

GPs:

Glucan particles

GRAS:

Generally Recognized As Safe

GQDs:

Graphene quantum dots

HDP:

Host defence peptide

HP- β -CD:

Hydroxypropyl- β -CD

IFN-γ:

Interferon gamma

IL-2:

Interleukin 2

I.M:

Intramuscular

INH:

Isoniazid

INVITE:

Inulin functionalized with vitamin E

INVITESA:

Inulin functionalized with a vitamin E succinylated derivative

I.V:

Intravenous

LAI:

Long-acting injectables

MBG:

Mesoporous bioactive glass

MDR:

Multidrug resistant

MIC:

Minimum inhibitory concentration

MO:

Monoolein

MOF:

Metal organic framework

MP:

Microparticles

MPP:

Medicines Patent Pool

MSNs:

Mesoporous silica nanoparticles

MWCNTs:

Multiwall carbon nanotubes

NPs:

Nanoparticles

NDDS:

Novel drug delivery systems

NIR:

Near infrared

NSCLC:

Non-small cell lung cancer

PAMAM:

Poly(amidoamine)

PBCA:

Poly(butyl cyanoacrylate)

PBMCs:

Peripheral blood mononuclear cells

PCL:

Poly-caprolactone

PEA:

Poly(ester amine)

PEG:

Polyethylene glycol

PEHAM:

Poly(ether hydroxylamine)

PEI:

Polyethyleneimine

PEO-PPO:

Polyethylene oxide- polypropylene oxide

PHBHHx:

Poly(3hydroxybutyrate-co-3-hydroxyhexonate)

PIBCA:

Poly(isobutyl cyanoacrylate)

PLGA:

Polylactic co glycolic acid

PPI:

Poly (propylene imine)

PVA:

Polyvinyl alcohol

PVP:

Polyvinylpyrrolidone

QFT-GIT:

QuantiFERON-TB Gold in Tube test

RIF:

Rifampicin

SDEDDS:

Self-double-emulsifying drug delivery systems

SDGs:

Sustainable development goals

si RNA:

Small interfering RNA

SLNs:

Solid lipid nanoparticles

SNPs:

Solid nanoparticles

TB:

Tuberculosis

TGF:

Transforming growth factor

TSL:

Thermosensitive liposome

TSTn:

Tuberculin skin test negative

XDR:

Extensive drug resistance

β -CD:

β -Cyclodextrins

References

  1. Zignol M, Gemert Wv, Falzon D, Sismanidis C, Glaziou P, Floyd K, Raviglione M. Surveillance of anti-tuberculosis drug resistance in the world: an updated analysis, 2007–2010. Bull World Health Organ. 2012;90:111–9.

    Article  PubMed  Google Scholar 

  2. Migliori GB, Raviglione MC. Essential tuberculosis. Cham: Springer; 2021.

    Book  Google Scholar 

  3. Shukla R, Sethi A, Handa M, Mohan M, Tripathi PK, Kesharwani P. Dendrimer-based drug delivery systems for tuberculosis treatment. In: Kesharwani Prashant, editor. Nanotechnology based approaches for tuberculosis treatment. Amsterdam: Elsevier; 2020.

    Google Scholar 

  4. Organization WH: Global tuberculosis report 2013: World Health Organization; 2013.

  5. WHO G: Global tuberculosis report 2020. Glob Tuberc Rep 2020.

  6. Tuberculosis. https://www.who.int/news-room/fact-sheets/detail/tuberculosis

  7. More MP, Chitalkar RV, Bhadane MS, Dhole SD, Patil AG, Patil PO, Deshmukh PK. Development of graphene-drug nanoparticle based supramolecular self assembled pH sensitive hydrogel as potential carrier for targeting MDR tuberculosis. Mater Technol. 2019;34(6):324–35.

    Article  CAS  Google Scholar 

  8. Sung N, Back S, Jung J, Kim K-H, Kim J-K, Lee JH, Ra Y, Yang HC, Lim C, Cho S. Inactivation of multidrug resistant (MDR)-and extensively drug resistant (XDR)-Mycobacterium tuberculosis by photodynamic therapy. Photodiagn Photodyn Ther. 2013;10(4):694–702.

    Article  CAS  Google Scholar 

  9. Costa A, Pinheiro M, Magalhães J, Ribeiro R, Seabra V, Reis S, Sarmento B. The formulation of nanomedicines for treating tuberculosis. Adv Drug Deliv Rev. 2016;102:102–15.

    Article  CAS  PubMed  Google Scholar 

  10. Vinod V, Pushkaran AC, Kumar A, Mohan CG, Biswas R. 2021 Interaction mechanism of Mycobacterium tuberculosis GroEL2 protein with macrophage Lectin-like, oxidized low-density lipoprotein receptor-1: an integrated computational and experimental study. Biochimica et Biophysica Acta Gen Subj. 1865;1:129758.

    Google Scholar 

  11. Krishnan N, Robertson BD, Thwaites G. The mechanisms and consequences of the extra-pulmonary dissemination of Mycobacterium tuberculosis. Tuberculosis. 2010;90(6):361–6.

    Article  CAS  PubMed  Google Scholar 

  12. Sia IG, Wieland ML. Current concepts in the management of tuberculosis. In: Beckman Thomas J, editor. Mayo clinic proceedings. Amsterdam: Elsevier; 2011.

    Google Scholar 

  13. Vinod V, Vijayrajratnam S, Vasudevan AK, Biswas R. The cell surface adhesins of Mycobacterium tuberculosis. Microbiol Res. 2020;232:126392.

    Article  CAS  PubMed  Google Scholar 

  14. Kirtane AR, Verma M, Karandikar P, Furin J, Langer R, Traverso G. Nanotechnology approaches for global infectious diseases. Nat Nanotechnol. 2021;16(4):369–84.

    Article  CAS  PubMed  Google Scholar 

  15. Paulose RR, Kumar VA, Sharma A, Damle A, Saikumar D, Sudhakar A, Koshy AK, Venu RP. An outcome-based composite approach for the diagnosis of intestinal tuberculosis: a pilot study from a tertiary care centre in South India. J Royal Coll Phys Edinb. 2021;51(4):344–50.

    Article  Google Scholar 

  16. Saktiawati AM, Sturkenboom MG, Stienstra Y, Subronto YW, Kosterink JG, van der Werf TS, Alffenaar J-WC. Impact of food on the pharmacokinetics of first-line anti-TB drugs in treatment-naive TB patients: a randomized cross-over trial. J Antimicrob Chemother. 2016;71(3):703–10.

    Article  CAS  PubMed  Google Scholar 

  17. Prameswari A. The evaluation of directly observed treatment short-course (DOTS) implementation for TB in hospital X. J Medicoeticolegal dan Manaj Rumah Sakit. 2018;7(2):93–101. https://doi.org/10.18196/jmmr.7261.

    Article  Google Scholar 

  18. Pal S, Soni V, Kumar S, Jha SK, Medatwal N, Rana K, Yadav P, Mehta D, Jain D, Sharma P. A hydrogel-based implantable multidrug antitubercular formulation outperforms oral delivery. Nanoscale. 2021;13(31):13225–30.

    Article  CAS  PubMed  Google Scholar 

  19. Connolly LE, Edelstein PH, Ramakrishnan L. Why is long-term therapy required to cure tuberculosis? PLoS Med. 2007;4(3):e120. https://doi.org/10.1371/journal.pmed.0040120.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Gygli SM, Borrell S, Trauner A, Gagneux S. Antimicrobial resistance in Mycobacterium tuberculosis: mechanistic and evolutionary perspectives. FEMS Microbiol Rev. 2017;41(3):354–73.

    Article  CAS  PubMed  Google Scholar 

  21. Seung KJ, Keshavjee S, Rich ML. Multidrug-resistant tuberculosis and extensively drug-resistant tuberculosis. Cold Spring Harbor Perspect Med. 2015. https://doi.org/10.1101/cshperspect.a017863.

    Article  Google Scholar 

  22. Hickey A, Durham P, Dharmadhikari A, Nardell E. Inhaled drug treatment for tuberculosis: past progress and future prospects. J Control Release. 2016;240:127–34. https://doi.org/10.1016/j.jconrel.2015.11.018.

    Article  CAS  PubMed  Google Scholar 

  23. Braunstein M, Hickey AJ, Ekins S. Why wait? The case for treating tuberculosis with inhaled drugs. Pharm Res. 2019;36(12):1–6.

    Article  Google Scholar 

  24. Organization WH. Latent tuberculosis infection: updated and consolidated guidelines for programmatic management. Geneva: World Health Organization; 2018.

    Google Scholar 

  25. Brhane Y, Gabriel T, Adane T, Negash Y, Mulugeta H, Ayele M. Recent developments and novel drug delivery strategies for the treatment of tuberculosis. Int J Pharm Sci Nanotechnol. 2019;12(3):4524–30.

    CAS  Google Scholar 

  26. Cohen J: Approval of novel TB drug celebrated—with restraint. In: American Association for the Advancement of Science; 2013.

  27. 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.

    Article  CAS  PubMed  Google Scholar 

  28. Keam SJ. Pretomanid: first approval. Drugs. 2019;79(16):1797–803.

    Article  PubMed  Google Scholar 

  29. Kaur K, Gupta A, Narang R, Murthy R. Novel drug delivery systems: desired feat for tuberculosis. J Adv Pharm Technol Res. 2010;1(2):145.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Gairola A, Benjamin A, Weatherston JD, Cirillo JD, Wu HJ. Recent developments in drug delivery for treatment of tuberculosis by targeting macrophages. Adv Ther. 2022. https://doi.org/10.1002/adtp.202100193.

    Article  Google Scholar 

  31. Borah Slater K, Kim D, Chand P, Xu Y, Shaikh H, Undale V. A current perspective on the potential of nanomedicine for anti-tuberculosis therapy. Trop Med Infect Dis. 2023;8(2):100.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Shirsath NR, Goswami AK. Nanocarriers based novel drug delivery as effective drug delivery: a review. Curr Nanomater. 2019;4(2):71–83.

    Article  CAS  Google Scholar 

  33. Dhanjal DS, Mehta M, Chopra C, Singh R, Sharma P, Chellappan DK, Tambuwala MM, Bakshi HA, Aljabali AA, Gupta G. Novel controlled release pulmonary drug delivery systems: current updates and challenges. In: Azar Ahmad Taher, editor. Modeling and control of drug delivery systems. Amsterdam: Elsevier; 2021.

    Google Scholar 

  34. Gopalaswamy R, Shanmugam S, Mondal R, Subbian S. Of tuberculosis and non-tuberculous mycobacterial infections–a comparative analysis of epidemiology, diagnosis and treatment. J Biomed Sci. 2020;27(1):1–17.

    Article  Google Scholar 

  35. Suresh P, Kumar A, Biswas R, Vijayakumar D, Thulasidharan S, Anjaneyan G, Kunoor A, Biswas L. Epidemiology of nontuberculous mycobacterial infection in tuberculosis suspects. Am J Trop Med Hyg. 2021. https://doi.org/10.4269/ajtmh.21-0095.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Singh C, Koduri L, Bhatt TD, Jhamb SS, Mishra V, Gill MS, Suresh S. In vitro-in vivo evaluation of novel co-spray dried rifampicin phospholipid lipospheres for oral delivery. AAPS PharmSciTech. 2017;18(1):138–46.

    Article  CAS  PubMed  Google Scholar 

  37. Denti P, Jeremiah K, Chigutsa E, Faurholt-Jepsen D, PrayGod G, Range N, Castel S, Wiesner L, Hagen CM, Christiansen M. Pharmacokinetics of isoniazid, pyrazinamide, and ethambutol in newly diagnosed pulmonary TB patients in Tanzania. PLoS ONE. 2015;10(10):e0141002.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Saifullah B, Chrzastek A, Maitra A, Naeemullah B, Fakurazi S, Bhakta S, Hussein MZ. Novel anti-tuberculosis nanodelivery formulation of ethambutol with graphene oxide. Molecules. 2017;22(10):1560.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Zhang M, Sala C, Hartkoorn RC, Dhar N, Mendoza-Losana A, Cole ST. Streptomycin-starved Mycobacterium tuberculosis 18b, a drug discovery tool for latent tuberculosis. Antimicrob Agents Chemother. 2012;56(11):5782–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Pijck J, Hallynck T, Soep H, Baert L, Daneels R, Boelaert J. Pharmacokinetics of amikacin in patients with renal insufficiency: relation of half-life and creatinine clearance. J Infect Dis. 1976;134(Supplement_2):S331–41. https://doi.org/10.1093/infdis/135.Supplement_2.S331.

    Article  PubMed  Google Scholar 

  41. Bunn PA. Kanamycin. Med Clin North Amer. 1970;54(5):1245–56. https://doi.org/10.1016/S0025-7125(16)32590-1.

    Article  CAS  Google Scholar 

  42. Stein GE, LeBel M, Flor SC, Zinny M. Bioavailability and pharmacokinetics of oral ofloxacin formulations in normal subjects. Current Med Research Opinion. 1991;12(8):479–84. https://doi.org/10.1185/03007999109111658.

    Article  CAS  Google Scholar 

  43. Fish DN, Chow AT. The clinical pharmacokinetics of levofloxacin. Clin Pharmacokin. 1997;32:101–19. https://doi.org/10.2165/00003088-199732020-00002.

    Article  CAS  Google Scholar 

  44. Naidoo A, Naidoo K, McIlleron H, Essack S, Padayatchi N. A review of moxifloxacin for the treatment of drug-susceptible tuberculosis. J Clin Pharmacol. 2017;57(11):1369–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Begg EJ, Robson RA, Saunders DA, Graham GG, Buttimore RC, Neill AM, Town GI. The pharmacokinetics of oral fleroxacin and ciprofloxacin in plasma and sputum during acute and chronic dosing. British J Clin Pharmacol. 2000;49(1):32–8. https://doi.org/10.1046/j.1365-2125.2000.00105.x.

    Article  CAS  Google Scholar 

  46. Drusano GL, Standiford HC, Plaisance K, Forrest A, Leslie J, Caldwell J. Absolute oral bioavailability of ciprofloxacin. Antimicro Agents Chemo. 1986;30(3):444–6. https://doi.org/10.1128/aac.30.3.444.

    Article  CAS  Google Scholar 

  47. Traunmüller F, Zeitlinger M, Zeleny P, Müller M, Joukhadar C. Pharmacokinetics of single-and multiple-dose oral clarithromycin in soft tissues determined by microdialysis. Antimicro Agents Chemo. 2007;51(9):3185–9. https://doi.org/10.1128/aac.00532-07.

    Article  Google Scholar 

  48. Patel DS, Sharma N, Patel MC, Patel BN, Shrivastav PS, Sanyal M. Development and validation of a selective and sensitive LC–MS/MS method for determination of cycloserine in human plasma: application to bioequivalence study. J Chrom B. 2011;879(23):2265–73. https://doi.org/10.1016/j.jchromb.2011.06.011.

    Article  CAS  Google Scholar 

  49. Peloquin CA, Henshaw TL, Huitt GA, Berning SE, Nitta AT, James GT. Pharmacokinetic evaluation of para-aminosalicylic acid granules. Pharmaco J Human Pharmacol Drug Therapy. 1994;14(1):40–6. https://doi.org/10.1002/j.1875-9114.1994.tb02787.x.

    Article  CAS  Google Scholar 

  50. Abdelwahab MT, Wasserman S, Brust JC, Gandhi NR, Meintjes G, Everitt D, Diacon A, Dawson R, Wiesner L, Svensson EM, Maartens G. Clofazimine pharmacokinetics in patients with TB: dosing implications. J Antimicro Chemo. 2020;75(11):3269–77. https://doi.org/10.1093/jac/dkaa310.

    Article  CAS  Google Scholar 

  51. Dharmadhikari AS, Kabadi M, Gerety B, Hickey AJ, Fourie PB, Nardell E. Phase I, single-dose, dose-escalating study of inhaled dry powder capreomycin: a new approach to therapy of drug-resistant tuberculosis. Antimicrobial agents and chemotherapy. 2013;57(6):2613–9. https://doi.org/10.1128/aac.02346-12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ahmad M, Madni AU, Usman M. In-vitro release and pharmacokinetics of anti-tubercle drug ethionamide in healthy male subjects. J Bioanal Biomed. 2009;1:046–9. https://doi.org/10.4172/1948-593X.1000010.

    Article  CAS  Google Scholar 

  53. Venkatesan K. Clinical pharmacokinetic considerations in the treatment of patients with leprosy. Clin Pharmaco. 1989;16:365–86. https://doi.org/10.2165/00003088-198916060-00003.

    Article  CAS  Google Scholar 

  54. Yun HY, Chang MJ, Jung H, Chang V, Wang Q, Strydom N, Yoon YR, Savic RM. Prothionamide dose optimization using population pharmacokinetics for multidrug-resistant tuberculosis patients. Antimicro Agents Chemo. 2022;66(9):e01893–21. https://doi.org/10.1128/aac.01893-21.

    Article  CAS  Google Scholar 

  55. Holdiness MR. Clinical pharmacokinetics of the antituberculosis drugs. Clin Pharmaco. 1984;9:511–44. https://doi.org/10.2165/00003088-198409060-00003.

    Article  CAS  Google Scholar 

  56. Zitkova L, Tousek J. Pharmacokinetics of cycloserine and terizidone. Chemotherapy. 1974;20(18):28. https://doi.org/10.1159/000221787.

    Article  Google Scholar 

  57. Skinner MH, Hsieh M, Torseth J, Pauloin D, Bhatia GU, Harkonen S, Merigan TC, Blaschke TF. Pharmacokinetics of rifabutin. Antimicro Agents Chemo. 1989;8:1237–41. https://doi.org/10.1128/aac.33.8.1237.

    Article  Google Scholar 

  58. Stalker DJ, Jungbluth GL. Clinical pharmacokinetics of linezolid, a novel oxazolidinone antibacterial. Clin Pharmaco. 2003;42:1129–40. https://doi.org/10.2165/00003088-200342130-00004.

    Article  CAS  Google Scholar 

  59. Chahine EB, Karaoui LR, Mansour H. Bedaquiline: a novel diarylquinoline for multidrug-resistant tuberculosis. Annals Pharmaco. 2014;48(1):107–15. https://doi.org/10.1177/1060028013504087.

    Article  CAS  Google Scholar 

  60. Salinger DH, Subramoney V, Everitt D, Nedelman JR. Population pharmacokinetics of the antituberculosis agent pretomanid. Antimicro Agents Chemo. 2019;63(10):e00907–19. https://doi.org/10.1128/aac.00907-19.

    Article  CAS  Google Scholar 

  61. Biswas B, Misra TK, Ray D, Majumder T, Bandyopadhyay TK, Bhowmick TK. Current therapeutic delivery approaches using nanocarriers for the treatment of tuberculosis disease. Int J Pharm. 2023. https://doi.org/10.1016/j.ijpharm.2023.123018.

    Article  PubMed  Google Scholar 

  62. Langer R. Drug delivery and targeting. Nature. 1998;392(6679 Suppl):5–10.

    CAS  PubMed  Google Scholar 

  63. Mosaiab T, Farr DC, Kiefel MJ, Houston TA. Carbohydrate-based nanocarriers and their application to target macrophages and deliver antimicrobial agents. Adv Drug Deliv Rev. 2019;151:94–129.

    Article  PubMed  Google Scholar 

  64. Afinjuomo F, Abdella S, Youssef SH, Song Y, Garg S. Inulin and its application in drug delivery. Pharmaceuticals. 2021;14(9):855.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Putri KSS, Ramadhani LS, Rachel T, Suhariyono G, Surini S. Promising chitosan-alginate combination for rifampicin dry powder inhaler to target active and latent tuberculosis. J Appl Pharm Sci. 2022;12(5):098–103.

    CAS  Google Scholar 

  66. Longuinho MM, Leitão SG, Silva RS, Silva PE, Rossi AL, Finotelli PV. Lapazine loaded alginate/chitosan microparticles: enhancement of anti-mycobacterium activity. J Drug Deliv Sci Technol. 2019;54:101292.

    Article  CAS  Google Scholar 

  67. Wolfram J, Zhu M, Yang Y, Shen J, Gentile E, Paolino D, Fresta M, Nie G, Chen C, Shen H. Safety of nanoparticles in medicine. Curr Drug Targets. 2015;16(14):1671–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Khairnar SV, Pagare P, Thakre A, Nambiar AR, Junnuthula V, Abraham MC, Kolimi P, Nyavanandi D, Dyawanapelly S. Review on the scale-up methods for the preparation of solid lipid nanoparticles. Pharmaceutics. 2022;14(9):1886.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Junnuthula V, Kolimi P, Nyavanandi D, Sampathi S, Vora LK, Dyawanapelly S. Polymeric micelles for breast cancer therapy: recent updates, clinical translation and regulatory considerations. Pharmaceutics. 2022;14(9):1860.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Sundar S, Chakravarty J. Liposomal amphotericin B and leishmaniasis: dose and response. J Global Infect Dis. 2010;2(2):159.

    Article  Google Scholar 

  71. Mitchell SL, Carlson EE. Tiny things with enormous impact: nanotechnology in the fight against infectious disease. ACS Infect Dis. 2018;4(10):1432–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Desai N. Challenges in development of nanoparticle-based therapeutics. AAPS J. 2012;14(2):282–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Ioannidis J, Kim B, Trounson A. How to design preclinical studies in nanomedicine and cell therapy to maximize the prospects of clinical translation. Nat Biomed Eng. 2018;2(11):797–809.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Chimote G, Banerjee R. In vitro evaluation of inhalable isoniazid-loaded surfactant liposomes as an adjunct therapy in pulmonary tuberculosis. J Biomed Mater Res B Appl Biomater. 2010;94(1):1–10.

    Article  CAS  PubMed  Google Scholar 

  75. Karki R, Mamatha G, Subramanya G, Udupa N. Preparation, characterization and tissue disposition of niosomes containing isoniazid. Rasayan J Chem. 2008;1(2):224–7.

    CAS  Google Scholar 

  76. Singh G, Dwivedi H, Saraf SK, Saraf SA. Niosomal delivery of isoniazid-development and characterization. Trop J Pharm Res. 2011. https://doi.org/10.4314/tjpr.v10i2.66564.

    Article  Google Scholar 

  77. Vatanparast M, Shariatinia Z. Computational studies on the doped graphene quantum dots as potential carriers in drug delivery systems for isoniazid drug. Struct Chem. 2018;29(5):1427–48.

    Article  CAS  Google Scholar 

  78. Chen G, Wu Y, Yu D, Li R, Luo W, Ma G, Zhang C. Isoniazid-loaded chitosan/carbon nanotubes microspheres promote secondary wound healing of bone tuberculosis. J Biomater Appl. 2019;33(7):989–96.

    Article  CAS  PubMed  Google Scholar 

  79. Zomorodbakhsh S, Abbasian Y, Naghinejad M, Sheikhpour M. The effects study of isoniazid conjugated multi-wall carbon nanotubes nanofluid on Mycobacterium tuberculosis. Int J Nanomed. 2020;15:5901.

    Article  CAS  Google Scholar 

  80. Fernández-Paz C, Fernández-Paz E, Salcedo-Abraira P, Rojas S, Barrios-Esteban S, Csaba N, Horcajada P, Remuñán-López C. Microencapsulated isoniazid-loaded metal-organic frameworks for pulmonary administration of antituberculosis drugs. Molecules. 2021;26(21):6408.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Anjani QK, Permana AD, Cárcamo-Martínez Á, Domínguez-Robles J, Tekko IA, Larrañeta E, Vora LK, Ramadon D, Donnelly RF. Versatility of hydrogel-forming microneedles in in vitro transdermal delivery of tuberculosis drugs. Eur J Pharm Biopharm. 2021;158:294–312.

    Article  CAS  PubMed  Google Scholar 

  82. Telange DR, Pandharinath RR, Pethe AM, Jain SP, Pingale PL. Calcium ion-sodium alginate-piperine-based microspheres: evidence of enhanced encapsulation efficiency, bio-adhesion, controlled delivery, and oral bioavailability of isoniazid. AAPS PharmSciTech. 2022;23(4):1–18.

    Article  Google Scholar 

  83. Jain C, Vyas S. Preparation and characterization of niosomes containing rifampicin for lung targeting. J Microencapsul. 1995;12(4):401–7.

    Article  CAS  PubMed  Google Scholar 

  84. Jain C, Vyas S, Dixit V. Niosomal system for delivery of rifampicin to lymphatics. Indian J Pharm Sci. 2006. https://doi.org/10.4103/0250-474X.29622.

    Article  Google Scholar 

  85. Kumar PV, Asthana A, Dutta T, Jain NK. Intracellular macrophage uptake of rifampicin loaded mannosylated dendrimers. J Drug Target. 2006;14(8):546–56.

    Article  CAS  PubMed  Google Scholar 

  86. Takenaga M, Ohta Y, Tokura Y, Hamaguchi A, Igarashi R, Disratthakit A, Doi N. Lipid microsphere formulation containing rifampicin targets alveolar macrophages. Drug Deliv. 2008;15(3):169–75.

    Article  CAS  PubMed  Google Scholar 

  87. Patil JS, Devi VK, Devi K, Sarasija S. A novel approach for lung delivery of rifampicin-loaded liposomes in dry powder form for the treatment of tuberculosis. Lung India. 2015;32(4):331.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Bellini RG, Guimarães AP, Pacheco MA, Dias DM, Furtado VR, de Alencastro RB, Horta BA. Association of the anti-tuberculosis drug rifampicin with a PAMAM dendrimer. J Mol Graph Model. 2015;60:34–42.

    Article  CAS  PubMed  Google Scholar 

  89. Parmar R, Misra R, Mohanty S. In vitro controlled release of Rifampicin through liquid-crystalline folate nanoparticles. Colloids Surf, B. 2015;129:198–205.

    Article  CAS  Google Scholar 

  90. Rajabnezhad S, Casettari L, Lam JK, Nomani A, Torkamani MR, Palmieri GF, Rajabnejad MR, Darbandi MA. Pulmonary delivery of rifampicin microspheres using lower generation polyamidoamine dendrimers as a carrier. Powder Technol. 2016;291:366–74.

    Article  CAS  Google Scholar 

  91. Tran N, Hocquet M, Eon B, Sangwan P, Ratcliffe J, Hinton TM, White J, Ozcelik B, Reynolds NP, Muir BW. Non-lamellar lyotropic liquid crystalline nanoparticles enhance the antibacterial effects of rifampicin against Staphylococcus aureus. J Colloid Interface Sci. 2018;519:107–18.

    Article  CAS  PubMed  Google Scholar 

  92. Ola M, Bhaskar R, Patil GR. Liquid crystalline drug delivery system for sustained release loaded with an antitubercular drug. J Drug Deliv Ther. 2018;8(4):93–101.

    Google Scholar 

  93. Thomas D, Latha M, Thomas KK. Synthesis and in vitro evaluation of alginate-cellulose nanocrystal hybrid nanoparticles for the controlled oral delivery of rifampicin. J Drug Deliv Sci Technol. 2018;46:392–9.

    Article  CAS  Google Scholar 

  94. Tripodo G, Perteghella S, Grisoli P, Trapani A, Torre ML, Mandracchia D. Drug delivery of rifampicin by natural micelles based on inulin: physicochemical properties, antibacterial activity and human macrophages uptake. Eur J Pharm Biopharm. 2019;136:250–8.

    Article  CAS  PubMed  Google Scholar 

  95. Suárez-González J, Santoveña-Estévez A, Soriano M, Fariña JB. Design and optimization of a child-friendly dispersible tablet containing isoniazid, pyrazinamide, and rifampicin for treating tuberculosis in pediatrics. Drug Develop Indus Pharm. 2020;46(2):309–17. https://doi.org/10.1080/03639045.2020.1717516.

  96. Grotz E, Tateosian NL, Salgueiro J, Bernabeu E, Gonzalez L, Manca ML, Amiano N, Valenti D, Manconi M, García V. Pulmonary delivery of rifampicin-loaded soluplus micelles against Mycobacterium tuberculosis. J Drug Deliv Sci Technol. 2019;53:101170.

    Article  CAS  Google Scholar 

  97. Pi J, Shen L, Shen H, Yang E, Wang W, Wang R, Huang D, Lee B-S, Hu C, Chen C. Mannosylated graphene oxide as macrophage-targeted delivery system for enhanced intracellular M. tuberculosis killing efficiency. Mater Sci Eng C. 2019;103:109777.

    Article  CAS  Google Scholar 

  98. Henostroza MAB, Melo KJC, Yukuyama MN, Löbenberg R, Bou-Chacra NA. Cationic rifampicin nanoemulsion for the treatment of ocular tuberculosis. Colloids Surf A. 2020;597:124755.

    Article  Google Scholar 

  99. El-Ridy MS, Yehia SA, Kassem MA-E-M, Mostafa DM, Nasr EA, Asfour MH. Niosomal encapsulation of ethambutol hydrochloride for increasing its efficacy and safety. Drug Deliv. 2015;22(1):21–36.

    Article  CAS  PubMed  Google Scholar 

  100. Nemati E, Mokhtarzadeh A, Panahi-Azar V, Mohammadi A, Hamishehkar H, Mesgari-Abbasi M, Ezzati Nazhad Dolatabadi J, de la Guardia M. Ethambutol-loaded solid lipid nanoparticles as dry powder inhalable formulation for tuberculosis therapy. AAPS PharmSciTech. 2019;20(3):1–9.

    Article  Google Scholar 

  101. Vladimirsky M, Ladigina G. Antibacterial activity of liposome-entrapped streptomycin in mice infected with Mycobacterium tuberculosis. Biomed Pharmacotherap. 1982;36(8–9):375–7.

    CAS  Google Scholar 

  102. Cynamon MH, Swenson CE, Palmer GS, Ginsberg RS. Liposome-encapsulated-amikacin therapy of Mycobacterium avium complex infection in beige mice. Antimicrob Agents Chemother. 1989;33(8):1179–83. https://doi.org/10.1128/aac.33.8.1179.

  103. Gaidukevich S, Mikulovich YL, Smirnova T, Andreevskaya S, Sorokoumova G, Chernousova L, Selishcheva A, Shvets V. Antibacterial effects of liposomes containing phospholipid cardiolipin and fluoroquinolone levofloxacin on Mycobacterium tuberculosis with extensive drug resistance. Bull Exp Biol Med. 2016;160(5):675–8.

    Article  CAS  PubMed  Google Scholar 

  104. Gaspar M, Cruz A, Penha A, Reymão J, Sousa A, Eleutério C, Domingues S, Fraga A, Longatto Filho A, Cruz M. Rifabutin encapsulated in liposomes exhibits increased therapeutic activity in a model of disseminated tuberculosis. Int J Antimicrob Agents. 2008;31(1):37–45.

    Article  CAS  PubMed  Google Scholar 

  105. Gaspar MM, Neves S, Portaels F, Pedrosa J, Silva MT, Cruz MEM. Therapeutic efficacy of liposomal rifabutin in a Mycobacterium avium model of infection. Antimicrob Agents Chemother. 2000;44(9):2424–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Sandler ED, Ng V, Hadley W. Clofazimine crystals in alveolar macrophages from a patient with the acquired immunodeficiency syndrome. Arch Pathol Lab Med. 1992;116(5):541–3.

    CAS  PubMed  Google Scholar 

  107. Mehta RT. Liposome encapsulation of clofazimine reduces toxicity in vitro and in vivo and improves therapeutic efficacy in the beige mouse model of disseminated Mycobacterium avium-M. intracellulare complex infection. Antimicrob Agents Chemother. 1996;40(8):1893–902.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Kansal RG, Gomez-Flores R, Sinha I, Mehta RT. Therapeutic efficacy of liposomal clofazimine against Mycobacterium avium complex in mice depends on size of initial inoculum and duration of infection. Antimicrob Agents Chemother. 1997;41(1):17–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Adams LB, Sinha I, Franzblau SG, Krahenbuhl JL, Mehta RT. Effective treatment of acute and chronic murine tuberculosis with liposome-encapsulated clofazimine. Antimicrob Agents Chemother. 1999;43(7):1638–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. de Castro RR, Todaro V, da Silva LCRP, Simon A, do Carmo FA, de Sousa VP, Rodrigues CR, Sarmento B, Healy AM, Cabral LM. Development of inhaled formulation of modified clofazimine as an alternative to treatment of tuberculosis. J Drug Deliv Sci Technol. 2020;58:101805.

    Article  Google Scholar 

  111. Kisich K, Gelperina S, Higgins M, Wilson S, Shipulo E, Oganesyan E, Heifets L. Encapsulation of moxifloxacin within poly (butyl cyanoacrylate) nanoparticles enhances efficacy against intracellular Mycobacterium tuberculosis. Int J Pharm. 2007;345(1–2):154–62.

    Article  CAS  PubMed  Google Scholar 

  112. Costa-Gouveia J, Pancani E, Jouny S, Machelart A, Delorme V, Salzano G, Iantomasi R, Piveteau C, Queval CJ, Song O-R. Combination therapy for tuberculosis treatment: pulmonary administration of ethionamide and booster co-loaded nanoparticles. Sci Rep. 2017;7(1):1–14.

    Article  CAS  Google Scholar 

  113. Garcia-Contreras L, Padilla-Carlin DJ, Sung J, VerBerkmoes J, Muttil P, Elbert K, Peloquin C, Edwards D, Hickey A. Pharmacokinetics of ethionamide delivered in spray-dried microparticles to the lungs of guinea pigs. J Pharm Sci. 2017;106(1):331–7.

    Article  CAS  PubMed  Google Scholar 

  114. De Maio F, Palmieri V, Santarelli G, Perini G, Salustri A, Palucci I, Sali M, Gervasoni J, Primiano A, Ciasca G. Graphene oxide-linezolid combination as potential new anti-tuberculosis treatment. Nanomaterials. 2020;10(8):1431.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Sercombe L, Veerati T, Moheimani F, Wu S, Sood A, Hua S. 2015 Advances and challenges of liposome assisted drug delivery. Front Pharmacol. 2015;6:286.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Orozco LC, Quintana FO, Beltrán RM, de Moreno I, Wasserman M, Rodriguez G. The use of rifampicin and isoniazid entrapped in liposomes for the treatment of murine tuberculosis. Tubercle. 1986;67(2):91–7.

    Article  CAS  PubMed  Google Scholar 

  117. Bhardwaj A, Kumar L, Narang RK, Murthy RS. Development and characterization of ligand-appended liposomes for multiple drug therapy for pulmonary tuberculosis. Artificial cells, nanomedicine, and biotechnology. 2013;41(1):52–59. https://doi.org/10.3109/10731199.2012.702316.

  118. Liu P, Guo B, Wang S, Ding J, Zhou W. A thermo-responsive and self-healing liposome-in-hydrogel system as an antitubercular drug carrier for localized bone tuberculosis therapy. Int J Pharm. 2019;558:101–9.

    Article  CAS  PubMed  Google Scholar 

  119. Bhardwaj A, Grobler A, Rath G, Kumar Goyal A, Kumar Jain A, Mehta A. Pulmonary delivery of anti-tubercular drugs using ligand anchored pH sensitive liposomes for the treatment of pulmonary tuberculosis. Curr Drug Deliv. 2016;13(6):909–22.

    Article  CAS  PubMed  Google Scholar 

  120. Greco E, Quintiliani G, Santucci MB, Serafino A, Ciccaglione AR, Marcantonio C, Papi M, Maulucci G, Delogu G, Martino A. Janus-faced liposomes enhance antimicrobial innate immune response in Mycobacterium tuberculosis infection. Proc Natl Acad Sci. 2012;109(21):E1360–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Miretti M, Juri L, Cosiansi MC, Tempesti TC, Baumgartner MT. Antimicrobial effects of ZnPc delivered into liposomes on multidrug resistant (MDR)-mycobacterium tuberculosis. ChemistrySelect. 2019;4(33):9726–30.

    Article  CAS  Google Scholar 

  122. Rosada RS, Silva CL, Santana MHA, Nakaie CR, de la Torre LG. Effectiveness, against tuberculosis, of pseudo-ternary complexes: peptide-DNA-cationic liposome. J Colloid Interface Sci. 2012;373(1):102–9.

    Article  CAS  PubMed  Google Scholar 

  123. Bekraki AI. Liposomes-and niosomes-based drug delivery systems for tuberculosis treatment. In: Kesharwani Prashant, editor. Nanotechnology based approaches for tuberculosis treatment. Amsterdam: Elsevier; 2020.

    Google Scholar 

  124. Patil K, Bagade S, Bonde S, Sharma S, Saraogi G. Recent therapeutic approaches for the management of tuberculosis: challenges and opportunities. Biomed Pharmacother. 2018;99:735–45. https://doi.org/10.1016/j.biopha.2018.01.115.

    Article  CAS  PubMed  Google Scholar 

  125. Kaur IP, Singh H. Nanostructured drug delivery for better management of tuberculosis. J Control Release. 2014;184:36–50.

    Article  CAS  PubMed  Google Scholar 

  126. Bibhas C, Narahari N. Exploring the use of lipid based nano-formulations for the management of tuberculosis. J Nanosci Curr Res. 2017;2(112):2572–813.

    Google Scholar 

  127. Bibhas C, Subas C, Gitanjali M, Narahari N. Exploring the use of lipid based nano-formulations for the management of tuberculosis. J Nanosci Curr Res. 2017;2(112):2572–813.

    Google Scholar 

  128. Hanieh PN, Consalvi S, Forte J, Cabiddu G, De Logu A, Poce G, Rinaldi F, Biava M, Carafa M, Marianecci C. Nano-based drug delivery systems of potent MmpL3 inhibitors for tuberculosis treatment. Pharmaceutics. 2022;14(3):610.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Sadhu PK, Saisivam S, Debnath SK. Design and characterization of niosomes of ethionamide for multi drug resistance tuberculosis. 2019.

  130. Kulkarni P, Rawtani D, Barot T. Formulation and optimization of long acting dual niosomes using box-Behnken experimental design method for combinative delivery of ethionamide and D-cycloserine in tuberculosis treatment. Colloids Surf A. 2019;565:131–42.

    Article  CAS  Google Scholar 

  131. Hussain A, Singh S, Das SS, Anjireddy K, Karpagam S, Shakeel F. Nanomedicines as drug delivery carriers of anti-tubercular drugs: from pathogenesis to infection control. Curr Drug Deliv. 2019;16(5):400–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Van Zyl L, Viljoen JM, Haynes RK, Aucamp M, Ngwane AH, du Plessis J. Topical delivery of artemisone, clofazimine and decoquinate encapsulated in vesicles and their in vitro efficacy against Mycobacterium tuberculosis. AAPS PharmSciTech. 2019;20(1):1–11.

    Google Scholar 

  133. Eldehna WM, El Hassab MA, Abdelshafi NA, Sayed FA-Z, Fares M, Al-Rashood ST, Elsayed ZM, Abdel-Aziz MM, Elkaeed EB, Elsabahy M. Development of potent nanosized isatin-isonicotinohydrazide hybrid for management of Mycobacterium tuberculosis. Int J Pharm. 2022;612:121369.

    Article  CAS  PubMed  Google Scholar 

  134. Emami F, Vatanara A, Park EJ, Na DH. Drying technologies for the stability and bioavailability of biopharmaceuticals. Pharmaceutics. 2018;10(3):131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Marzaman AN, Roska TP, Sartini S, Utami RN, Sulistiawati S, Enggi CK, Manggau MA, Rahman L, Shastri VP, Permana AD. Recent advances in pharmaceutical approaches of antimicrobial agents for selective delivery in various administration routes. Antibiotics. 2023;12(5):822. https://doi.org/10.3390/antibiotics12050822.

  136. Dua K, Rapalli VK, Shukla SD, Singhvi G, Shastri MD, Chellappan DK, Satija S, Mehta M, Gulati M, Pinto TDJA. Multi-drug resistant Mycobacterium tuberculosis & oxidative stress complexity: emerging need for novel drug delivery approaches. Biomed Pharmacother. 2018;107:1218–29.

    Article  CAS  PubMed  Google Scholar 

  137. Kim SY, Park MS, Kim YS, Kim SK, Chang J, Lee HJ, Cho SN, Kang YA. The responses of multiple cytokines following incubation of whole blood from TB patients, latently infected individuals and controls with the TB antigens ESAT‐6, CFP‐10 and TB 7.7. Scand J Immunol. 2012;76(6):580–86. https://doi.org/10.1111/j.1365-3083.2012.02776.x.

  138. Patil SM, Sawant SS, Kunda NK. Inhalable bedaquiline-loaded cubosomes for the treatment of non-small cell lung cancer (NSCLC). Int J Pharm. 2021;607:121046.

    Article  CAS  PubMed  Google Scholar 

  139. Anjani QK, Domínguez-Robles J, Utomo E, Font M, Martínez-Ohárriz MC, Permana AD, Cárcamo-Martínez Á, Larrañeta E, Donnelly RF. Inclusion complexes of rifampicin with native and derivatized cyclodextrins: in silico modeling, formulation, and characterization. Pharmaceuticals. 2021;15(1):20.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Amarnath Praphakar R, Sam Ebenezer R, Vignesh S, Shakila H, Rajan M. Versatile pH-responsive chitosan-g-polycaprolactone/maleic anhydride–isoniazid polymeric micelle to improve the bioavailability of tuberculosis multidrugs. ACS Appl Bio Mater. 2019;2(5):1931–43.

    Article  CAS  PubMed  Google Scholar 

  141. Kaur J, Mishra V, Singh SK, Gulati M, Kapoor B, Chellappan DK, Gupta G, Dureja H, Anand K, Dua K. Harnessing amphiphilic polymeric micelles for diagnostic and therapeutic applications: breakthroughs and bottlenecks. J Control Release. 2021;334:64–95.

    Article  CAS  PubMed  Google Scholar 

  142. Yuan X, Praphakar RA, Munusamy MA, Alarfaj AA, Kumar SS, Rajan M. Mucoadhesive guargum hydrogel inter-connected chitosan-g-polycaprolactone micelles for rifampicin delivery. Carbohyd Polym. 2019;206:1–10.

    Article  CAS  Google Scholar 

  143. Sheth U, Tiwari S, Bahadur A. Preparation and characterization of anti-tubercular drugs encapsulated in polymer micelles. J Drug Deliv Sci Technol. 2018;48:422–8.

    Article  CAS  Google Scholar 

  144. Garg NK, Dwivedi P, Jain A, Tyagi S, Sahu T, Tyagi RK. Development of novel carrier (s) mediated tuberculosis vaccine: more than a tour de force. Eur J Pharm Sci. 2014;62:227–42.

    Article  CAS  PubMed  Google Scholar 

  145. Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Préat V. PLGA-based nanoparticles: an overview of biomedical applications. J Control Release. 2012;161(2):505–22.

    Article  CAS  PubMed  Google Scholar 

  146. Gu X, Cheng Q, He P, Zhang Y, Jiang Z, Zeng Y. Dihydroartemisinin-loaded chitosan nanoparticles inhibit the rifampicin-resistant mycobacterium tuberculosis by disrupting the cell wall. Front Microbiol. 2021. https://doi.org/10.3389/fmicb.2021.735166/full.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Abdallah HM, Elella MHA, Abdel-Aziz MM. One-pot green synthesis of chitosan biguanidine nanoparticles for targeting M. tuberculosis. Int J Biol Macromol. 2023;232:123394.

    Article  CAS  PubMed  Google Scholar 

  148. Anisimova YV, Gelperina SI, Peloquin CA, Heifets LB. Nanoparticles as antituberculosis drugs carriers: effect on activity against Mycobacterium tuberculosis in human monocyte-derived macrophages. J Nanopart Res. 2000;2:165–71. https://doi.org/10.1023/A:1010061013365.

  149. Scolari IR, Páez PL, Sánchez-Borzone ME, Granero GE. Promising chitosan-coated alginate-tween 80 nanoparticles as rifampicin coadministered ascorbic acid delivery carrier against Mycobacterium tuberculosis. AAPS PharmSciTech. 2019;20(2):1–21.

    Article  Google Scholar 

  150. Nagpal PS, Kesarwani A, Sahu P, Upadhyay P. Aerosol immunization by alginate coated mycobacterium (BCG/MIP) particles provide enhanced immune response and protective efficacy than aerosol of plain mycobacterium against M. tb. H37Rv infection in mice. BMC Infect Dis. 2019;19(1):1–14.

    Article  CAS  Google Scholar 

  151. Kesarwani A, Sahu P, Jain K, Sinha P, Mohan KV, Nagpal PS, Singh S, Zaidi R, Nagarajan P, Upadhyay P. The safety and efficacy of BCG encapsulated alginate particle (BEAP) against M. tb H37Rv infection in macaca mulatta: a pilot study. Sci Rep. 2021;11(1):1–10.

    Article  Google Scholar 

  152. Najafi A, Ghazvini K, Sankian M, Gholami L, Amini Y, Zare S, Khademi F, Tafaghodi M. T helper type 1 biased immune responses by PPE17 loaded core-shell alginate-chitosan nanoparticles after subcutaneous and intranasal administration. Life Sci. 2021;282:119806.

    Article  CAS  PubMed  Google Scholar 

  153. Kushwaha K, Dwivedi H. Interfacial phenomenon based biocompatible alginate-chitosan nanoparticles containing isoniazid and pyrazinamide. Pharm Nanotechnol. 2018;6(3):209–17.

    Article  CAS  PubMed  Google Scholar 

  154. Soria-Carrera H, Lucía A, De Matteis L, Aínsa JA, de la Fuente JM, Martín-Rapún R. Polypeptidic micelles stabilized with sodium alginate enhance the activity of encapsulated bedaquiline. Macromol Biosci. 2019;19(4):1800397.

    Article  Google Scholar 

  155. Latha M, Kurienthomas K. Zinc-alginate beads for the controlled release of rifampici. Orient J Chem. 2018;34(1):428.

    Article  Google Scholar 

  156. Chen C-C, Chen Y-Y, Yeh C-C, Hsu C-W, Yu S-J, Hsu C-H, Wei T-C, Ho S-N, Tsai P-C, Song Y-D. Alginate-capped silver nanoparticles as a potent anti-mycobacterial agent against mycobacterium tuberculosis. Front Pharmacol. 2021. https://doi.org/10.3389/fphar.2021.746496/full.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Liu Z, Ye L, Xi J, Wang J, Feng Z-G. Cyclodextrin polymers: structure, synthesis, and use as drug carriers. Prog Poly Sci. 2021;118:101408.

    Article  CAS  Google Scholar 

  158. Abdellatif FHH, Abdellatif MM. Utilization of sustainable biopolymers in textile processing. In: Ibrahim Nabil, Hussain Chaudhery Mustansar, editors. Green chemistry for sustainable textiles. Amsterdam: Elsevier; 2021.

    Google Scholar 

  159. Tiwari G, Tiwari R, Rai AK. Cyclodextrins in delivery systems: applications. J Pharm Bioall Sci. 2010;2(2):72–9.

    Article  CAS  Google Scholar 

  160. Crini G. A history of cyclodextrins. Chem Rev. 2014;114(21):10940–75.

    Article  CAS  PubMed  Google Scholar 

  161. Basha RY, TS SK, Doble M. Dual delivery of tuberculosis drugs via cyclodextrin conjugated curdlan nanoparticles to infected macrophages. Carbohydr Polym. 2019;218:53–62.

    Article  Google Scholar 

  162. Nkanga CI, Krause RWM. Encapsulation of isoniazid-conjugated phthalocyanine-in-cyclodextrin-in-liposomes using heating method. Sci Rep. 2019;9(1):1–16.

    Article  CAS  Google Scholar 

  163. Maiti PK, Çaǧın T, Wang G, Goddard WA. Structure of PAMAM dendrimers: generations 1 through 11. Macromolecules. 2004;37(16):6236–54.

    Article  CAS  Google Scholar 

  164. Kaur D, Jain K, Mehra NK, Kesharwani P, Jain NK. A review on comparative study of PPI and PAMAM dendrimers. J Nanopart Res. 2016;18(6):1–14.

    Article  CAS  Google Scholar 

  165. Bapat RA, Joshi CP, Bapat P, Chaubal TV, Pandurangappa R, Jnanendrappa N, Gorain B, Khurana S, Kesharwani P. The use of nanoparticles as biomaterials in dentistry. Drug Discov Today. 2019;24(1):85–98.

    Article  CAS  PubMed  Google Scholar 

  166. Madaan K, Kumar S, Poonia N, Lather V, Pandita D. Dendrimers in drug delivery and targeting: drug-dendrimer interactions and toxicity issues. J Pharm Bioall Sci. 2014;6(3):139.

    Article  Google Scholar 

  167. Chauhan AS. Dendrimers for drug delivery. Molecules. 2018;23(4):938.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Menjoge AR, Kannan RM, Tomalia DA. Dendrimer-based drug and imaging conjugates: design considerations for nanomedical applications. Drug Discov Today. 2010;15(5–6):171–85.

    Article  CAS  PubMed  Google Scholar 

  169. Wang J, Li B, Qiu L, Qiao X, Yang H. Dendrimer-based drug delivery systems: history, challenges, and latest developments. J Biol Eng. 2022;16(1):1–12.

    Article  Google Scholar 

  170. Parekh H. The advance of dendrimers-a versatile targeting platform for gene/drug delivery. Curr Pharm Des. 2007;13(27):2837–50.

    Article  CAS  PubMed  Google Scholar 

  171. Choudhary S, Gupta L, Rani S, Dave K, Gupta U. Impact of dendrimers on solubility of hydrophobic drug molecules. Front Pharmacol. 2017;16(8):261. https://doi.org/10.3389/fphar.2017.00261.

    Article  CAS  Google Scholar 

  172. Bodewein L, Schmelter F, Di Fiore S, Hollert H, Fischer R, Fenske M. Differences in toxicity of anionic and cationic PAMAM and PPI dendrimers in zebrafish embryos and cancer cell lines. Toxicol Appl Pharmacol. 2016;305:83–92.

    Article  CAS  PubMed  Google Scholar 

  173. Bellini RG, Guimarães AP, Pacheco MA, Dias DM, Furtado VR, de Alencastro RB, Horta BA. Association of the anti-tuberculosis drug rifampicin with a PAMAM dendrimer. J Mol Graph Model. 2015;60:34–42. https://doi.org/10.1016/j.jmgm.2015.05.012.

    Article  CAS  PubMed  Google Scholar 

  174. Karthikeyan R, Koushik O, Kumar V. Surface modification of cationic dendrimers eases drug delivery of anticancer drugs. Nanosci Nanotechnol. 2016;10:108.

    Google Scholar 

  175. Kaur M, Garg T, Narang R. A review of emerging trends in the treatment of tuberculosis. Artif Cells Nanomed Biotechnol. 2016;44(2):478–84.

    Article  CAS  PubMed  Google Scholar 

  176. Srinivasan M, Rajabi M, Mousa SA. Multifunctional nanomaterials and their applications in drug delivery and cancer therapy. Nanomaterials. 2015;5(4):1690–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Zohuri G. Polymer science: a comprehensive reference. Amsterdam: Elsevier; 2012.

    Google Scholar 

  178. Leng Z-H, Zhuang Q-F, Li Y-C, He Z, Chen Z, Huang S-P, Jia H-Y, Zhou J-W, Liu Y, Du L-B. Polyamidoamine dendrimer conjugated chitosan nanoparticles for the delivery of methotrexate. Carbohyd Polym. 2013;98(1):1173–8.

    Article  CAS  Google Scholar 

  179. Jain A, Jain K, Mehra NK, Jain N. Lipoproteins tethered dendrimeric nanoconstructs for effective targeting to cancer cells. J Nanopart Res. 2013;15(10):1–18.

    Article  Google Scholar 

  180. Bernkop-Schnürch A, Scholler S, Biebel RG. Development of controlled drug release systems based on thiolated polymers. J Control Release. 2000;66(1):39–48.

    Article  PubMed  Google Scholar 

  181. Muttil P, Kaur J, Kumar K, Yadav AB, Sharma R, Misra A. Inhalable microparticles containing large payload of anti-tuberculosis drugs. Eur J Pharm Sci. 2007;32(2):140–50.

    Article  CAS  PubMed  Google Scholar 

  182. Sharma S, Khuller G, Garg S. Alginate-based oral drug delivery system for tuberculosis: pharmacokinetics and therapeutic effects. J Antimicrob Chemother. 2003;51(4):931–8.

    Article  Google Scholar 

  183. Soto E, Kim YS, Lee J, Kornfeld H, Ostroff G. Glucan particle encapsulated rifampicin for targeted delivery to macrophages. Polymers. 2010;2(4):681–9.

    Article  CAS  Google Scholar 

  184. Upadhyay TK, Fatima N, Sharma D, Saravanakumar V, Sharma R. Preparation and characterization of beta-glucan particles containing a payload of nanoembedded rifabutin for enhanced targeted delivery to macrophages. EXCLI J. 2017;16:210.

    PubMed  PubMed Central  Google Scholar 

  185. Rawal T, Kremer L, Halloum I, Butani S. Dry-powder inhaler formulation of rifampicin: an improved targeted delivery system for alveolar tuberculosis. J Aero Med Pulm Drug Delivery. 2017;30(6):388–98. https://doi.org/10.1089/jamp.2017.1379.

    Article  CAS  Google Scholar 

  186. Cunha L, Rosa da Costa AM, Lourenço JP, Buttini F, Grenha A. Spray-dried fucoidan microparticles for pulmonary delivery of antitubercular drugs. J Microencapsul. 2018;35(4):392–405.

    Article  CAS  PubMed  Google Scholar 

  187. Cunha L, Rodrigues S, Rosa da Costa AM, Faleiro ML, Buttini F, Grenha A. Inhalable fucoidan microparticles combining two antitubercular drugs with potential application in pulmonary tuberculosis therapy. Polymers. 2018;10(6):636.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Cunha L, Rodrigues S, da Costa AMR, Faleiro L, Buttini F, Grenha A. Inhalable chitosan microparticles for simultaneous delivery of isoniazid and rifabutin in lung tuberculosis treatment. Drug Dev Ind Pharm. 2019. https://doi.org/10.1080/03639045.2019.1608231.

    Article  PubMed  Google Scholar 

  189. Sharma A, Vaghasiya K, Ray E, Gupta P, Singh AK, Gupta UD, Verma RK. Mycobactericidal activity of some micro-encapsulated synthetic host defense peptides (HDP) by expediting the permeation of antibiotic: a new paradigm of drug delivery for tuberculosis. Int J Pharm. 2019;558:231–41.

    Article  CAS  PubMed  Google Scholar 

  190. Vasudevan S, Venkatraman A, Yahoob SAM, Jojula M, Sundaram R, Boomi P. Biochemical evaluation and molecular docking studies on encapsulated astaxanthin for the growth inhibition of Mycobacterium tuberculosis. J Appl Biol Biotechnol. 2021;9(1):3–9.

    Google Scholar 

  191. Sharma A, Vaghasiya K, Ray E, Gupta P, Gupta UD, Singh AK, Verma RK. Targeted pulmonary delivery of the green tea polyphenol epigallocatechin gallate controls the growth of mycobacterium tuberculosis by enhancing the autophagy and suppressing bacterial burden. ACS Biomater Sci Eng. 2020;6(7):4126–40.

    Article  CAS  PubMed  Google Scholar 

  192. Gaspar MC, Grégoire N, Sousa JJ, Pais AA, Lamarche I, Gobin P, Olivier J-C, Marchand S, Couet W. Pulmonary pharmacokinetics of levofloxacin in rats after aerosolization of immediate-release chitosan or sustained-release PLGA microspheres. Eur J Pharm Sci. 2016;93:184–91.

    Article  CAS  PubMed  Google Scholar 

  193. Vidyadevi B. Direct lungs targeting: an alternative treatment approach for pulmonary tuberculosis. Asian J Pharm (AJP). 2021. https://doi.org/10.2237/ajp.v15i04.4212.

    Article  Google Scholar 

  194. Pingale PL, Amrutkar SV. Quercetin loaded rifampicin-floating microspheres for improved stability and invitro drug release. Pharmacophore. 2021;12(3):95–9. https://doi.org/10.51847/yBXnl2bSUH.

    Article  Google Scholar 

  195. Mwila C, Walker RB. Improved stability of rifampicin in the presence of gastric-resistant isoniazid microspheres in acidic media. Pharmaceutics. 2020;12(3):234.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Luciani-Giacobbe LC, Lorenzutti AM, Litterio NJ, Ramírez-Rigo MV, Olivera ME. Anti-tuberculosis site-specific oral delivery system that enhances rifampicin bioavailability in a fixed-dose combination with isoniazid. Drug Delivery Trans Re. 2021;11:894–908. https://doi.org/10.1007/s13346-020-00847-9.

    Article  CAS  Google Scholar 

  197. Upadhyay S, Khan I, Gothwal A, Pachouri PK, Bhaskar N, Gupta UD, Chauhan DS, Gupta U. Conjugated and entrapped HPMA-PLA nano-polymeric micelles based dual delivery of first line anti TB drugs: improved and safe drug delivery against sensitive and resistant Mycobacterium tuberculosis. Pharm Res. 2017;34(9):1944–55.

    Article  CAS  PubMed  Google Scholar 

  198. Moradi S, Taran M, Mohajeri P, Sadrjavadi K, Sarrami F, Karton A, Shahlaei M. Study of dual encapsulation possibility of hydrophobic and hydrophilic drugs into a nanocarrier based on bio-polymer coated graphene oxide using density functional theory, molecular dynamics simulation and experimental methods. J Mol Liq. 2018;262:204–17.

    Article  CAS  Google Scholar 

  199. Zhu M, Li K, Zhu Y, Zhang J, Ye X. 3D-printed hierarchical scaffold for localized isoniazid/rifampin drug delivery and osteoarticular tuberculosis therapy. Acta Biomater. 2015;16:145–55.

    Article  CAS  PubMed  Google Scholar 

  200. Tabriz AG, Nandi U, Hurt AP, Hui H-W, Karki S, Gong Y, Kumar S, Douroumis D. 3D printed bilayer tablet with dual controlled drug release for tuberculosis treatment. Int J Pharm. 2021;593:120147.

    Article  Google Scholar 

  201. Clemens DL, Lee B-Y, Xue M, Thomas CR, Meng H, Ferris D, Nel AE, Zink JI, Horwitz MA. Targeted intracellular delivery of antituberculosis drugs to Mycobacterium tuberculosis-infected macrophages via functionalized mesoporous silica nanoparticles. Antimicrob Agents Chemother. 2012;56(5):2535–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Mehta S, Kaur G, Bhasin K. Tween-embedded microemulsions—physicochemical and spectroscopic analysis for antitubercular drugs. AAPS PharmSciTech. 2010;11(1):143–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Kaur G, Mehta S, Kumar S, Bhanjana G, Dilbaghi N. Coencapsulation of hydrophobic and hydrophilic antituberculosis drugs in synergistic Brij 96 microemulsions: a biophysical characterization. J Pharm Sci. 2015;104(7):2203–12.

    Article  CAS  PubMed  Google Scholar 

  204. Mulia K, Chadarwati S, Rahyussalim A, Krisanti E. Preparation and characterization of polyvinyl alcohol-chitosan-tripolyphosphate hydrogel for extended release of anti-tuberculosis drugs. IOP Conf Ser Mater Sci Eng. 2019. https://doi.org/10.1088/1757-899X/703/1/012010.

    Article  Google Scholar 

  205. Krisanti EA, Gofara TZ, Rahyussalim AJ, Mulia K. Polyvinyl alcohol (PVA)/chitosan/sodium tripolyphosphate (STPP) hydrogel formulation with freeze-thaw method for anti-tuberculosis drugs extended release. AIP Conf Proc. 2021. https://doi.org/10.1063/5.0063175.

    Article  Google Scholar 

  206. Tudose M, Anghel EM, Culita DC, Somacescu S, Calderon-Moreno J, Tecuceanu V, Dumitrascu FD, Dracea O, Popa M, Marutescu L. Covalent coupling of tuberculostatic agents and graphene oxide: a promising approach for enhancing and extending their antimicrobial applications. Appl Surf Sci. 2019;471:553–65.

    Article  CAS  Google Scholar 

  207. Sheikhpour M, Delorme V, Kasaeian A, Amiri V, Masoumi M, Sadeghinia M, Ebrahimzadeh N, Maleki M, Pourazar S. An effective nano drug delivery and combination therapy for the treatment of tuberculosis. Sci Rep. 2022;12(1):1–11.

    Article  Google Scholar 

  208. Chowdhury P, Shankar U. Formulation and evaluation of Rifampicin and Ofloxacin niosomes for Drugresistant TB on Logarithmic-phase cultures of Mycobacterium tuberculosis. Int J Res Pharma Sci (IJRPS). 2016;3(4):628–33.

    Google Scholar 

  209. Ferraz-Carvalho RS, Pereira MA, Linhares LA, Lira-Nogueira MC, Cavalcanti IM, Santos-Magalhães NS, Montenegro LM. Effects of the encapsulation of usnic acid into liposomes and interactions with antituberculous agents against multidrug-resistant tuberculosis clinical isolates. Mem Inst Oswaldo Cruz. 2016;111:330–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Yousefi A, Khodaverdi E, Atyabi F, Dinarvand R. Thermosensitive drug permeation through liquid crystal-embedded cellulose nitrate membranes. PDA J Pharm Sci Technol. 2010;64(1):54–62.

    CAS  PubMed  Google Scholar 

  211. Kramer RM, Archer MC, Orr MT, Cauwelaert ND, Beebe EA, Po-wei DH, Dowling QM, Schwartz AM, Fedor DM, Vedvick TS. Development of a thermostable nanoemulsion adjuvanted vaccine against tuberculosis using a design-of-experiments approach. Int J Nanomed. 2018;13:3689.

    Article  CAS  Google Scholar 

  212. de Almeida A, Caleffi-Ferracioli K, de Regiane B, Scodro L, Baldin VP, Montaholi DC, Spricigo LF, Nakamura-Vasconcelos SS, Hegeto LA, Sampiron EG, Costacurta GF, dos Diego A, Yamazaki S, de Gauze FG, Siqueira VL, Cardoso RF. Eugenol and derivatives activity against Mycobacterium tuberculosis, nontuberculous mycobacteria and other bacteria. Future Microbiol. 2019;14:331–44.

    Article  PubMed  Google Scholar 

  213. Zhang G, Sheng L, Hegde P, Li Y, Aldrich CC. 8-cyanobenzothiazinone analogs with potent antitubercular activity. Med Chem Res. 2021;30(2):449–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Kumar U, Singh RK. Clinical efficacy of beta-sitosterol as adjuvant therapy for the treatment of tuberculosis in children. Int J Paediatr Geriatr. 2021;4(1):141–3. https://doi.org/10.33545/26643685.2021.v4.i1c.144.

    Article  Google Scholar 

  215. Rudolph D, Redinger N, Schaible UE, Feldmann C. Transport of lipophilic anti-tuberculosis drug benzothiazone-043 in Ca3 (PO4) 2 nanocontainers. ChemNanoMat. 2021;7(1):7–16.

    Article  CAS  Google Scholar 

  216. Gupta A, Pandey S, Yadav JS. A review on recent trends in green synthesis of gold nanoparticles for tuberculosis. Adv Pharm Bull. 2021;11(1):10.

    Article  PubMed  Google Scholar 

  217. Govindaraju K, Vasantharaja R, Suganya KU, Anbarasu S, Revathy K, Pugazhendhi A, Karthickeyan D, Singaravelu G. Unveiling the anticancer and antimycobacterial potentials of bioengineered gold nanoparticles. Process Biochem. 2020;96:213–9.

    Article  CAS  Google Scholar 

  218. Srivastava N, Mukhopadhyay M. Biosynthesis and characterization of gold nanoparticles using Zooglea ramigera and assessment of its antibacterial property. J Cluster Sci. 2015;26(3):675–92.

    Article  CAS  Google Scholar 

  219. Sun C, Zhang X, Wang J, Chen Y, Meng C. Novel mesoporous silica nanocarriers containing gold; a rapid diagnostic tool for tuberculosis. BMC Complement Med Ther. 2021;21(1):1–7.

    Article  Google Scholar 

  220. Gilbride B, Moreira GMSG, Hust M, Cao C, Stewart L. Catalytic ferromagnetic gold nanoparticle immunoassay for the detection and differentiation of Mycobacterium tuberculosis and Mycobacterium bovis. Anal Chim Acta. 2021;1184:339037.

    Article  CAS  PubMed  Google Scholar 

  221. Li J, Hu K, Zhang Z, Teng X, Zhang X. Click DNA cycling in combination with gold nanoparticles loaded with quadruplex DNA motifs enable sensitive electrochemical quantitation of the tuberculosis-associated biomarker CFP-10 in sputum. Microchim Acta. 2019;186(9):1–7.

    Article  Google Scholar 

  222. Singh N, Dahiya B, Radhakrishnan VS, Prasad T, Mehta PK. Detection of Mycobacterium tuberculosis purified ESAT-6 (Rv3875) by magnetic bead-coupled gold nanoparticle-based immuno-PCR assay. Int J Nanomed. 2018;13:8523.

    Article  CAS  Google Scholar 

  223. Sadanandan P, Payne NL, Sun G, Ashokan A, Gowd SG, Lal A, Kumar MKS, Pulakkat S, Nair SV, Menon KN. Exploiting the preferential phagocytic uptake of nanoparticle-antigen conjugates for the effective treatment of autoimmunity. Nanomed Nanotechnol Biol Med. 2022;40:102481.

    Article  CAS  Google Scholar 

  224. Saravanan V, Ramachandran M, Prasanth V: Exploring various Silver Nanoparticles and Nanotechnology. 2022.

  225. Mamaeva V, Sahlgren C, Lindén M. Mesoporous silica nanoparticles in medicine—recent advances. Adv Drug Deliv Rev. 2013;65(5):689–702.

    Article  CAS  PubMed  Google Scholar 

  226. Hwang J, Son J, Seo Y, Jo Y, Lee K, Lee D, Khan MS, Chavan S, Park C, Sharma A. Functional silica nanoparticles conjugated with beta-glucan to deliver anti-tuberculosis drug molecules. J Ind Eng Chem. 2018;58:376–85.

    Article  CAS  Google Scholar 

  227. Tenland E, Pochert A, Krishnan N, Umashankar Rao K, Kalsum S, Braun K, Glegola-Madejska I, Lerm M, Robertson BD, Lindén M. Effective delivery of the anti-mycobacterial peptide NZX in mesoporous silica nanoparticles. PLoS ONE. 2019;14(2):e0212858.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Beitzinger B, Gerbl F, Vomhof T, Schmid R, Noschka R, Rodriguez A, Wiese S, Weidinger G, Ständker L, Walther P. Antimicrobial peptides: delivery by dendritic mesoporous silica nanoparticles enhances the antimicrobial activity of a napsin-derived peptide against intracellular Mycobacterium tuberculosis (Adv. Healthcare Mater. 14/2021). Adv Healthcare Mater. 2021;10(14):2170066.

    Article  Google Scholar 

  229. Montalvo-Quirós S, Gómez-Graña S, Vallet-Regí M, Prados-Rosales RC, González B, Luque-Garcia JL. Mesoporous silica nanoparticles containing silver as novel antimycobacterial agents against Mycobacterium tuberculosis. Colloids Surf, B. 2021;197:111405.

    Article  Google Scholar 

  230. Chen W, Cheng C-A, Lee B-Y, Clemens DL, Huang W-Y, Horwitz MA, Zink JI. Facile strategy enabling both high loading and high release amounts of the water-insoluble drug clofazimine using mesoporous silica nanoparticles. ACS Appl Mater Interfaces. 2018;10(38):31870–81.

    Article  CAS  PubMed  Google Scholar 

  231. Ang CW, Tan L, Qu Z, West NP, Cooper MA, Popat A, Blaskovich MA. Mesoporous silica nanoparticles improve oral delivery of antitubercular bicyclic nitroimidazoles. ACS Biomater Sci Eng. 2021. https://doi.org/10.1021/acsbiomaterials.1c00807.

    Article  PubMed  PubMed Central  Google Scholar 

  232. Selvarajan V, Obuobi S, Ee PLR. Silica nanoparticles—a versatile tool for the treatment of bacterial infections. Front Chem. 2020;8:602.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Bhushan B, Luo D, Schricker SR, Sigmund W, Zauscher S. Handbook of nanomaterials properties. Berlin: Springer; 2014.

    Book  Google Scholar 

  234. Chen Y, Guo S, Zhao M, Zhang P, Xin Z, Tao J, Bai L. Amperometric DNA biosensor for Mycobacterium tuberculosis detection using flower-like carbon nanotubes-polyaniline nanohybrid and enzyme-assisted signal amplification strategy. Biosens Bioelectron. 2018;119:215–20.

    Article  CAS  PubMed  Google Scholar 

  235. Buya AB, Witika BA, Bapolisi AM, Mwila C, Mukubwa GK, Memvanga PB, Makoni PA, Nkanga CI. Application of lipid-based nanocarriers for antitubercular drug delivery: a review. Pharmaceutics. 2021;13(12):2041.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Mehta S, Kaur G, Bhasin K. Entrapment of multiple anti-Tb drugs in microemulsion system: quantitative analysis, stability, and in vitro release studies. J Pharm Sci. 2010;99(4):1896–911.

    Article  CAS  Google Scholar 

  237. Rajput A, Mandlik S, Pokharkar V. Nanocarrier-based approaches for the efficient delivery of anti-tubercular drugs and vaccines for management of tuberculosis. Front Pharmacol. 2021. https://doi.org/10.3389/fphar.2021.749945/full.

    Article  PubMed  PubMed Central  Google Scholar 

  238. Eleleemy M, Amin BH, Nasr M, Sammour OA. A succinct review on the therapeutic potential and delivery systems of Eugenol. Arch Pharm Sci Ain Shams University. 2020;4(2):290–311.

    Google Scholar 

  239. Talegaonkar S, Azeem A, Ahmad FJ, Khar RK, Pathan SA, Khan ZI. Microemulsions: a novel approach to enhanced drug delivery. Recent Patents Drug Delivery Form. 2008;2(3):238–57. https://doi.org/10.2174/187221108786241679.

    Article  CAS  Google Scholar 

  240. Sheikh BA, Bhat BA, Alshehri B, Mir RA, Mir WR, Parry ZA, Mir MA. Nano-drug delivery systems: possible end to the rising threats of tuberculosis. J Biomed Nanotechnol. 2021;17(12):2298–318.

    Article  CAS  PubMed  Google Scholar 

  241. Kompella UB, Kadam RS, Lee VH. Recent advances in ophthalmic drug delivery. Ther Deliv. 2010;1(3):435–56.

    Article  CAS  PubMed  Google Scholar 

  242. Raina N, Pahwa R, Bhattacharya J, Paul AK, Nissapatorn V, de Lourdes PM, Oliveira SM, Dolma KG, Rahmatullah M, Wilairatana P. Drug delivery strategies and biomedical significance of hydrogels: translational considerations. Pharmaceutics. 2022;14(3):574.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Wan Y, Liu L, Yuan S, Sun J, Li Z. pH-responsive peptide supramolecular hydrogels with antibacterial activity. Langmuir. 2017;33(13):3234–40.

    Article  CAS  PubMed  Google Scholar 

  244. Ahmad N, Pandey M, Mohamad N, Chen XY, Amin MCIM. Hydrogels for pulmonary drug delivery. In: Dua Kamal, Hansbro Philip M, Wadhwa Ridhima, Haghi Mehra, Pont Lisa G, Williams Kylie A, editors. Targeting chronic inflammatory lung diseases using advanced drug delivery systems. Amsterdam: Elsevier; 2020.

    Google Scholar 

  245. Guvendiren M, Molde J, Soares RM, Kohn J. Designing biomaterials for 3D printing. ACS Biomater Sci Eng. 2016;2(10):1679–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  246. Zhao S, Zhu M, Zhang J, Zhang Y, Liu Z, Zhu Y, Zhang C. Three dimensionally printed mesoporous bioactive glass and poly (3-hydroxybutyrate-co-3-hydroxyhexanoate) composite scaffolds for bone regeneration. J Mater Chem B. 2014;2(36):6106–18. https://doi.org/10.1039/c4tb00838c.

    Article  CAS  PubMed  Google Scholar 

  247. Malakar TK, Chaudhari VS, Dwivedy SK, Murty US, Banerjee S. 3D printed housing devices for segregated compartmental delivery of oral fixed-dose anti-tubercular drugs adopting print and fill strategy. Print Addit Manuf. 2021. https://doi.org/10.1089/3dp.2021.0037.

    Article  Google Scholar 

  248. Sasikumar K, Ghosh AR, Dusthackeer A. Antimycobacterial potentials of quercetin and rutin against Mycobacterium tuberculosis H37Rv. 3 Biotech. 2018;8(10):1–6.

    Article  Google Scholar 

  249. Chaudhari VS, Malakar TK, Murty US, Banerjee S. Extruded filaments derived 3D printed medicated skin patch to mitigate destructive pulmonary tuberculosis: design to delivery. Expert Opin Drug Deliv. 2021;18(2):301–13.

    Article  CAS  PubMed  Google Scholar 

  250. Marcianes P, Negro S, Barcia E, Montejo C, Fernández-Carballido A. Potential active targeting of gatifloxacin to macrophages by means of surface-modified PLGA microparticles destined to treat tuberculosis. AAPS PharmSciTech. 2020;21(1):1–14.

    Article  Google Scholar 

  251. Shah S, Cristopher D, Sharma S, Soniwala M, Chavda J. Inhalable linezolid loaded PLGA nanoparticles for treatment of tuberculosis: design, development and in vitro evaluation. J Drug Deliv Sci Technol. 2020;60:102013.

    Article  CAS  Google Scholar 

  252. Operti MC, Bernhardt A, Grimm S, Engel A, Figdor CG, Tagit O. PLGA-based nanomedicines manufacturing: technologies overview and challenges in industrial scale-up. Int J Pharm. 2021;605:120807.

    Article  CAS  PubMed  Google Scholar 

  253. Baranyai Z, Soria-Carrera H, Alleva M, Millán-Placer AC, Lucía A, Martín-Rapún R, Aínsa JA, de la Fuente JM. Nanotechnology-based targeted drug delivery: an emerging tool to overcome tuberculosis. Adv Ther. 2021;4(1):2000113.

    Article  Google Scholar 

  254. Roy A, Agnivesh PK, Sau S, Kumar S, Kalia NP. Tweaking host immune responses for novel therapeutic approaches against Mycobacterium tuberculosis. Drug Discov Today. 2023. https://doi.org/10.1016/j.drudis.2023.103693.

    Article  PubMed  Google Scholar 

  255. Hamed A, Osman R, Al-Jamal KT, Holayel SM, Geneidi A-S. Enhanced antitubercular activity, alveolar deposition and macrophages uptake of mannosylated stable nanoliposomes. J Drug Deliv Sci Technol. 2019;51:513–23.

    Article  CAS  Google Scholar 

  256. Vieira AC, Chaves LL, Pinheiro M, Lima SAC, Ferreira D, Sarmento B, Reis S. Mannosylated solid lipid nanoparticles for the selective delivery of rifampicin to macrophages. Artif Cells Nanomed Biotechnol. 2018;46(sup1):653–63.

    Article  CAS  PubMed  Google Scholar 

  257. Galdopórpora JM, Martinena C, Bernabeu E, Riedel J, Palmas L, Castangia I, Manca ML, Garcés M, Lázaro-Martinez J, Salgueiro MJ. Inhalable mannosylated rifampicin-curcumin co-loaded nanomicelles with enhanced in vitro antimicrobial efficacy for an optimized pulmonary tuberculosis therapy. Pharmaceutics. 2022;14(5):959.

    Article  PubMed  PubMed Central  Google Scholar 

  258. Sarkar S, Dyett B, Lakic B, Ball AS, Yeo LY, White JF, Soni S, Drummond CJ, Conn CE. Cubosome lipid nanocarriers as a drug delivery vehicle for intracellular mycobacterium tuberculosis infections. ACS Appl Mater Interfaces. 2023;15(18):21819–29.

    Article  CAS  PubMed  Google Scholar 

  259. Worstell NC, Singla A, Saenkham P, Galbadage T, Sule P, Lee D, Mohr A, Kwon JS-I, Cirillo JD, Wu H-J. Hetero-multivalency of Pseudomonas aeruginosa lectin LecA binding to model membranes. Sci Rep. 2018;8(1):1–11.

    Article  CAS  Google Scholar 

  260. Siegel RA, Kirtane AR, Panyam J. Assessing the benefits of drug delivery by nanocarriers: a partico/pharmacokinetic framework. IEEE Trans Biomed Eng. 2016;64(9):2176–85.

    Article  PubMed  Google Scholar 

  261. Garcia-Contreras L, Sethuraman V, Kazantseva M, Hickey A. Efficacy of combined rifampicin formulations delivered by the pulmonary route to treat tuberculosis in the guinea pig model. Pharmaceutics. 2021;13(8):1309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Sharma PR, Dravid AA, Kalapala YC, Gupta VK, Jeyasankar S, Goswami A, Agarwal R. Cationic inhalable particles for enhanced drug delivery to M. tuberculosis infected macrophages. Biomater Adv. 2022;133:112612.

    Article  PubMed  Google Scholar 

  263. Gangadhar KN, Changsan V, Buatong W, Srichana T. Phase behavior of rifampicin in cholesterol-based liquid crystals and polyethylene glycol. Eur J Pharm Sci. 2012;47(5):804–12.

    Article  CAS  PubMed  Google Scholar 

  264. Truzzi E, Capocefalo A, Meneghetti F, Maretti E, Mori M, Iannuccelli V, Domenici F, Castellano C, Leo E. Design and physicochemical characterization of novel hybrid SLN-liposome nanocarriers for the smart co-delivery of two antitubercular drugs. J Drug Deliv Sci Technol. 2022;70:103206.

    Article  CAS  Google Scholar 

  265. Roy I, Vij N. Nanodelivery in airway diseases: challenges and therapeutic applications. Nanomed Nanotechnol Biol Med. 2010;6(2):237–44.

    Article  CAS  Google Scholar 

  266. Han C, Romero N, Fischer S, Dookran J, Berger A, Doiron AL. Recent developments in the use of nanoparticles for treatment of biofilms. Nanotechnol Rev. 2017;6(5):383–404.

    Article  CAS  Google Scholar 

  267. Misra A, Hickey AJ, Rossi C, Borchard G, Terada H, Makino K, Fourie PB, Colombo P. Inhaled drug therapy for treatment of tuberculosis. Tuberculosis. 2011;91(1):71–81.

    Article  CAS  PubMed  Google Scholar 

  268. Pham D-D, Fattal E, Tsapis N. Pulmonary drug delivery systems for tuberculosis treatment. Int J Pharm. 2015;478(2):517–29.

    Article  CAS  PubMed  Google Scholar 

  269. Tan ZM, Lai GP, Pandey M, Srichana T, Pichika MR, Gorain B, Bhattamishra SK, Choudhury H. Novel approaches for the treatment of pulmonary tuberculosis. Pharmaceutics. 2020;12(12):1196.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Ghosh S, Ghosh S, Sil PC. Role of nanostructures in improvising oral medicine. Toxicol Rep. 2019;6:358–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. Yang Z, Niu N, Lou C, Wang X, Wang C, Shi Z. Preparation, characterrization, and in-vitro cytotoxicity of nanoliposomes loaded with anti-tuberculous drugs and TGF-β1 siRNA for improving spinal tuberculosis therapy. BMC Infect Dis. 2022. https://doi.org/10.1186/s12879-022-07791-8.

    Article  PubMed  PubMed Central  Google Scholar 

  272. Jiang Z, Wei J, Peng N, Li Y. Genetic engineering of a phage-based delivery system for endogenous III-A CRISPR-cas system against mycobacterium tuberculosis. In: Tofazzal Islam M, Molla Kutubuddin Ali, editors. CRISPR-cas methods. New York: Springer; 2021.

    Google Scholar 

  273. Dubey AK, Kumar Gupta V, Kujawska M, Orive G, Kim N-Y, Li C-Z, Kumar Mishra Y, Kaushik A. Exploring nano-enabled CRISPR-Cas-powered strategies for efficient diagnostics and treatment of infectious diseases. J Nanostruct Chem. 2022. https://doi.org/10.1007/s40097-022-00472-7.

    Article  Google Scholar 

  274. Babunovic GH, DeJesus MA, Bosch B, Chase MR, Barbier T, Dickey AK, Bryson BD, Rock JM, Fortune SM. CRISPR interference reveals that all-trans-retinoic acid promotes macrophage control of mycobacterium tuberculosis by limiting bacterial access to cholesterol and propionyl coenzyme A. MBio. 2022;13(1):e03683-e3621.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  275. Verma M, Furin J, Langer R, Traverso G. Making the case: developing innovative adherence solutions for the treatment of tuberculosis. BMJ Glob Health. 2019;4(1):e001323.

    Article  PubMed  PubMed Central  Google Scholar 

  276. Furin J, Tommasi M, Garcia-Prats AJ. Drug-resistant tuberculosis: will grand promises fail children and adolescents? Lancet Child Adolesc Health. 2018;2(4):237–8.

    Article  PubMed  Google Scholar 

  277. Harausz EP, Garcia-Prats AJ, Seddon JA, Schaaf HS, Hesseling AC, Achar J, Bernheimer J, Cruz AT, D’Ambrosio L, Detjen A. New and repurposed drugs for pediatric multidrug-resistant tuberculosis Practice-based recommendations. Am J Respir Crit Care Med. 2017;195(10):1300–10.

    Article  CAS  PubMed  Google Scholar 

  278. Swindells S, Siccardi M, Barrett SE, Olsen DB, Grobler JA, Podany AT, Nuermberger E, Kim P, Barry C, Owen A. Long-acting formulations for the treatment of latent tuberculous infection: opportunities and challenges. Int J Tuberc Lung Dis. 2018;22(2):125–32.

    Article  CAS  PubMed  Google Scholar 

  279. Park EJ, Amatya S, Kim MS, Park JH, Seol E, Lee H, Shin Y-H, Na DH. Long-acting injectable formulations of antipsychotic drugs for the treatment of schizophrenia. Arch Pharmacal Res. 2013;36(6):651–9.

    Article  CAS  Google Scholar 

  280. Kaushik A, Ammerman NC, Tyagi S, Saini V, Vervoort I, Lachau-Durand S, Nuermberger E, Andries K. Activity of a long-acting injectable bedaquiline formulation in a paucibacillary mouse model of latent tuberculosis infection. Antimicrob Agents Chemother. 2019;63(4):e00007-00019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  281. Diacon A, Donald P, Pym A, Grobusch M, Patientia R, Mahanyele R, Bantubani N, Narasimooloo R, De Marez T, Van Heeswijk R. Randomized pilot trial of eight weeks of bedaquiline (TMC207) treatment for multidrug-resistant tuberculosis: long-term outcome, tolerability, and effect on emergence of drug resistance. Antimicrob Agents Chemother. 2012;56(6):3271–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  282. Rajoli RK, Podany AT, Moss DM, Swindells S, Flexner C, Owen A, Siccardi M. Modelling the long-acting administration of anti-tuberculosis agents using PBPK: a proof of concept study. Int J Tuberc Lung Dis. 2018;22(8):937–44.

    Article  CAS  PubMed  Google Scholar 

  283. Verma M, Vishwanath K, Eweje F, Roxhed N, Grant T, Castaneda M, Steiger C, Mazdiyasni H, Bensel T, Minahan D. A gastric resident drug delivery system for prolonged gram-level dosing of tuberculosis treatment. Sci Transl Med. 2019;11(483):6267.

    Article  Google Scholar 

  284. Adeleke OA, Fisher L, Moore IN, Nardone GA, Sher A. A long-acting thermoresponsive injectable formulation of tin protoporphyrin sustains antitubercular efficacy in a murine infection model. ACS Pharmacol Transl Sci. 2020;4(1):276–87.

    Article  PubMed  PubMed Central  Google Scholar 

  285. Sadeghi I, Byrne J, Shakur R, Langer R. Engineered drug delivery devices to address global health challenges. J Control Release. 2021;331:503–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  286. Raza A, Sime FB, Cabot PJ, Maqbool F, Roberts JA, Falconer JR. Solid nanoparticles for oral antimicrobial drug delivery: a review. Drug Discov Today. 2019;24(3):858–66.

    Article  CAS  PubMed  Google Scholar 

  287. Singh H, Jindal S, Singh M, Sharma G, Kaur IP. Nano-formulation of rifampicin with enhanced bioavailability: development, characterization and in-vivo safety. Int J Pharm. 2015;485(1–2):138–51.

    Article  CAS  PubMed  Google Scholar 

  288. Elbrink K, Van Hees S, Chamanza R, Roelant D, Loomans T, Holm R, Kiekens F. Application of solid lipid nanoparticles as a long-term drug delivery platform for intramuscular and subcutaneous administration: in vitro and in vivo evaluation. Eur J Pharm Biopharm. 2021;163:158–70.

    Article  CAS  PubMed  Google Scholar 

  289. Pigrau-Serrallach C, Rodríguez-Pardo D. Bone and joint tuberculosis. Eur Spine J. 2013;22(4):556–66.

    Article  PubMed  Google Scholar 

  290. Hua L, Qian H, Lei T, Liu W, He X, Zhang Y, Lei P, Hu Y. Anti-tuberculosis drug delivery for tuberculous bone defects. Expert Opin Drug Deliv. 2021;18(12):1815–27.

    Article  CAS  PubMed  Google Scholar 

  291. Zhang J, Zhao S, Zhu Y, Huang Y, Zhu M, Tao C, Zhang C. Three-dimensional printing of strontium-containing mesoporous bioactive glass scaffolds for bone regeneration. Acta Biomater. 2014;10(5):2269–81.

    Article  CAS  PubMed  Google Scholar 

  292. Zhao S, Zhang J, Zhu M, Zhang Y, Liu Z, Ma Y, Zhu Y, Zhang C. Effects of functional groups on the structure, physicochemical and biological properties of mesoporous bioactive glass scaffolds. J Mater Chem B. 2015;3(8):1612–23.

    Article  CAS  PubMed  Google Scholar 

  293. Wu C, Chang J. Multifunctional mesoporous bioactive glasses for effective delivery of therapeutic ions and drug/growth factors. J Control Release. 2014;193:282–95.

    Article  CAS  PubMed  Google Scholar 

  294. Miller SR, Heurtaux D, Baati T, Horcajada P, Grenèche J-M, Serre C. Biodegradable therapeutic MOFs for the delivery of bioactive molecules. Chem Commun. 2010;46(25):4526–8.

    Article  CAS  Google Scholar 

  295. Giménez-Marqués M, Hidalgo T, Serre C, Horcajada P. Nanostructured metal–organic frameworks and their bio-related applications. Coord Chem Rev. 2016;307:342–60.

    Article  Google Scholar 

  296. Semaan R, Traboulsi R, Kanj S. Primary Mycobacterium tuberculosis complex cutaneous infection: report of two cases and literature review. Int J Infect Dis. 2008;12(5):472–7.

    Article  PubMed  Google Scholar 

  297. van Staden D, Haynes RK, Viljoen JM. Adapting clofazimine for treatment of cutaneous tuberculosis by using self-double-emulsifying drug delivery systems. Antibiotics. 2022;11(6):806.

    Article  PubMed  PubMed Central  Google Scholar 

  298. Xu Y, Wu J, Liao S, Sun Z. Treating tuberculosis with high doses of anti-TB drugs: mechanisms and outcomes. Ann Clin Microbiol Antimicrob. 2017;16(1):1–13.

    Article  Google Scholar 

  299. Ammerman NC, Swanson RV, Bautista EM, Almeida DV, Saini V, Omansen TF, Guo H, Chang YS, Li S-Y, Tapley A. Impact of clofazimine dosing on treatment shortening of the first-line regimen in a mouse model of tuberculosis. Antimicrob Agents Chemother. 2018;62(7):e00636-e618.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  300. van Staden D, Haynes RK, Viljoen JM. Adapting clofazimine for treatment of cutaneous tuberculosis by using self-double-emulsifying drug delivery systems. Antibiotics. 2022;11(6):806. https://doi.org/10.3390/antibiotics11060806.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  301. Caon T, Campos CEM, Simões CMO, Silva MAS. Novel perspectives in the tuberculosis treatment: administration of isoniazid through the skin. Int J Pharm. 2015;494(1):463–70.

    Article  CAS  PubMed  Google Scholar 

  302. Basu S, Monira S, Modi RR, Choudhury N, Mohan N, Padhi TR, Balne PK, Sharma S, Panigrahi SR. Degree, duration, and causes of visual impairment in eyes affected with ocular tuberculosis. J Ophthalmic Inflam Infect. 2014;4(1):1–5.

    Article  Google Scholar 

  303. Bennett JE, Dolin R, Blaser MJ: Mandell, douglas, and bennett’s principles and practice of infectious diseases E-book: Elsevier Health Sciences; 2019.

  304. Agrawal R, Gunasekeran DV, Raje D, Agarwal A, Nguyen QD, Kon OM, Pavesio C, Gupta V. Global variations and challenges with tubercular uveitis in the collaborative ocular tuberculosis study. Invest Ophthalmol Vis Sci. 2018;59(10):4162–71.

    Article  PubMed  Google Scholar 

  305. Agrawal R, Ludi Z, Betzler BK, Testi I, Mahajan S, Rousellot A, Kempen JH, Smith JR, McCluskey P, Nguyen QD. The collaborative ocular tuberculosis study (COTS) calculator—a consensus-based decision tool for initiating antitubercular therapy in ocular tuberculosis. Eye. 2022. https://doi.org/10.1038/s41433-022-02147-7.

    Article  PubMed  Google Scholar 

  306. Zhang Z, Liu J, Wan C, Liu P, Wan H, Guo Z, Tong J, Cao X. Successful treatment of tuberculosis verrucosa cutis with fester as primary manifestation with photodynamic therapy and anti-tubercular drugs. Photodiagn Photodyn Ther. 2022;38:102763.

    Article  CAS  Google Scholar 

  307. Patel U, Rathnayake K, Jani H, Jayawardana KW, Dhakal R, Duan L, Jayawardena SN. Near-infrared responsive targeted drug delivery system that offer chemo-photothermal therapy against bacterial infection. Nano Select. 2021;2(9):1750–69.

    Article  CAS  Google Scholar 

  308. Liu Y, Lin A, Liu J, Chen X, Zhu X, Gong Y, Yuan G, Chen L, Liu J. Enzyme-responsive mesoporous ruthenium for combined chemo-photothermal therapy of drug-resistant bacteria. ACS Appl Mater Interfaces. 2019;11(30):26590–606.

    Article  CAS  PubMed  Google Scholar 

  309. Sia JK, Georgieva M, Rengarajan J. Innate immune defenses in human tuberculosis: an overview of the interactions between Mycobacterium tuberculosis and innate immune cells. J Immunol Res. 2015. https://doi.org/10.1155/2015/747543.

    Article  PubMed  PubMed Central  Google Scholar 

  310. Cadena AM, Flynn JL, Fortune SM. The importance of first impressions: early events in Mycobacterium tuberculosis infection influence outcome. MBio. 2016;7(2):e00342-e316.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  311. Divangahi M, Aaby P, Khader SA, Barreiro LB, Bekkering S, Chavakis T, van Crevel R, Curtis N, DiNardo AR, Dominguez-Andres J. Trained immunity, tolerance, priming and differentiation: distinct immunological processes. Nat Immunol. 2021;22(1):2–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  312. Gong W, Wu X. Differential diagnosis of latent tuberculosis infection and active tuberculosis: a key to a successful tuberculosis control strategy. Front Microbiol. 2021. https://doi.org/10.3389/fmicb.2021.745592.

    Article  PubMed  PubMed Central  Google Scholar 

  313. Khan N, Vidyarthi A, Javed S, Agrewala JN. Innate immunity holding the flanks until reinforced by adaptive immunity against Mycobacterium tuberculosis infection. Front Microbiol. 2016;7:328.

    Article  PubMed  PubMed Central  Google Scholar 

  314. Mi J, Liang Y, Liang J, Gong W, Wang S, Zhang J, Li Z, Wu X. The research progress in immunotherapy of tuberculosis. Front Cell Infect Microbiol. 2021. https://doi.org/10.3389/fcimb.2021.763591.

    Article  PubMed  PubMed Central  Google Scholar 

  315. Johnson B, Bekker LG, Ress S, Kaplan G. Recombinant interleukin 2 adjunctive therapy in multidrug-resistant tuberculosis. In: Chadwick Derek J, Cardew Gail, editors. Genetics and tuberculosis: novartis foundation symposium 217. Hoboken: Wiley Online Library; 1998.

    Google Scholar 

  316. Kim YG, Baltabekova AZ, Zhiyenbay EE, Aksambayeva AS, Shagyrova ZS, Khannanov R, Ramanculov EM, Shustov AV. Recombinant vaccinia virus-coded interferon inhibitor B18R: expression, refolding and a use in a mammalian expression system with a RNA-vector. PLoS ONE. 2017;12(12):e0189308.

    Article  PubMed  PubMed Central  Google Scholar 

  317. Ma Y, Chen H-D, Wang Y, Wang Q, Li Y, Zhao Y, Zhang X-L. Interleukin 24 as a novel potential cytokine immunotherapy for the treatment of Mycobacterium tuberculosis infection. Microbes Infect. 2011;13(12–13):1099–110.

    Article  CAS  PubMed  Google Scholar 

  318. Netea MG, Lewis EC, Azam T, Joosten LA, Jaekal J, Bae S-Y, Dinarello CA, Kim S-H. Interleukin-32 induces the differentiation of monocytes into macrophage-like cells. Proc Natl Acad Sci. 2008;105(9):3515–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  319. Li W, Deng W, Xie J. The biology and role of interleukin-32 in tuberculosis. J Immunol Res. 2018. https://doi.org/10.1155/2018/1535194.

    Article  PubMed  PubMed Central  Google Scholar 

  320. Teitelbaum R, Glatman-Freedman A, Chen B, Robbins JB, Unanue E, Casadevall A, Bloom BR. A mAb recognizing a surface antigen of Mycobacterium tuberculosis enhances host survival. Proc Natl Acad Sci. 1998;95(26):15688–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  321. Hamasur B, Haile M, Pawlowski A, Schröder U, Källenius G, Svenson SB. A mycobacterial lipoarabinomannan specific monoclonal antibody and its F (ab′) 2 fragment prolong survival of mice infected with Mycobacterium tuberculosis. Clin Exp Immunol. 2004;138(1):30–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  322. AlMatar M, Makky EA, Yakıcı G, Var I, Kayar B, Köksal F. Antimicrobial peptides as an alternative to anti-tuberculosis drugs. Pharmacol Res. 2018;128:288–305.

    Article  PubMed  Google Scholar 

  323. Saeed AF, Wang R, Ling S, Wang S. Antibody engineering for pursuing a healthier future. Front Microbiol. 2017;8:495.

    Article  PubMed  PubMed Central  Google Scholar 

  324. Gutiérrez-Ortega A, Moreno DA, Ferrari SA, Espinosa-Andrews H, Ortíz EP, Milián-Suazo F, Alvarez AH. High-yield production of major T-cell ESAT6-CFP10 fusion antigen of M. tuberculosis complex employing codon-optimized synthetic gene. Int J Biol Macromol. 2021;171:82–8. https://doi.org/10.1016/j.ijbiomac.2020.12.179.

    Article  CAS  PubMed  Google Scholar 

  325. Bianchi L, Galli L, Moriondo M, Veneruso G, Becciolini L, Azzari C, Chiappini E, de Martino M. Interferon-gamma release assay improves the diagnosis of tuberculosis in children. Pediatr Infect Dis J. 2009;28(6):510–4.

    Article  PubMed  Google Scholar 

  326. Zhang X, Liu X-Y, Yang H, Chen J-N, Lin Y, Han S-Y, Cao Q, Zeng H-S, Ye J-W. A polyhydroxyalkanoates-based carrier platform of bioactive substances for therapeutic applications. Front Bioeng Biotechnol. 2021. https://doi.org/10.3389/fbioe.2021.798724/full.

    Article  PubMed  PubMed Central  Google Scholar 

  327. Saracino A, Scotto G, Fornabaio C, Martinelli D, Faleo G, Cibelli D, Tartaglia A, Di Tuwo R, Fazio V, Prato R. QuantiFERON®-TB gold in-tube test (QFT-GIT) for the screening of latent tuberculosis in recent immigrants to Italy. New Microbiol. 2009;32(4):369.

    PubMed  Google Scholar 

  328. Patil TS, Deshpande AS. Innovative strategies in the diagnosis and treatment of tuberculosis: a patent review (2014–2017). Expert Opin Ther Pat. 2018;28(8):615–23.

    Article  CAS  PubMed  Google Scholar 

  329. Brigden G, Castro JL, Ditiu L, Gray G, Hanna D, Low M, Matsoso MP, Perry G, Spigelman M, Swaminathan S. Tuberculosis and antimicrobial resistance–new models of research and development needed. Bull World Health Organ. 2017;95(5):315–315.

    Article  PubMed  PubMed Central  Google Scholar 

  330. Pool MP. The medicines patent pool announces first license for tuberculosis treatment. Geneva: UNITAID; 2017.

    Google Scholar 

  331. Bekale RB, Du Plessis S-M, Hsu N-J, Sharma JR, Sampson SL, Jacobs M, Meyer M, Morse GD, Dube A. Mycobacterium tuberculosis and interactions with the host immune system: opportunities for nanoparticle based immunotherapeutics and vaccines. Pharm Res. 2019;36(1):1–15.

    Article  CAS  Google Scholar 

  332. Verma N, Arora V, Awasthi R, Chan Y, Jha NK, Thapa K, Jawaid T, Kamal M, Gupta G, Liu G. Recent developments, challenges and future prospects in advanced drug delivery systems in the management of tuberculosis. J Drug Deliv Sci Technol. 2022. https://doi.org/10.1016/j.jddst.2022.103690.

    Article  Google Scholar 

  333. ID93 + GLA-SE vaccine [https://classic.clinicaltrials.gov/ct2/results?cond=&term=ID93+%2B+GLA-SE+vaccine&cntry=&state=&city=&dist=%20cite%20link%20and%20add%20date]

Download references

Acknowledgements

AYN AG AMN RKS AJ AKV PS would like to acknowledge the support by Amrita Vishwa Vidyapeetham. VJ expresses his gratitude to the Finnish Cultural Foundation (Ingrid, Toini and Olavi Martelius foundation) and the Helsinki University Library for funding.

Funding

Open Access funding provided by University of Helsinki (including Helsinki University Central Hospital).

Author information

Authors and Affiliations

Authors

Contributions

AYN AG AMN RKS AJ VJ performed the literature search and data analysis. AYN AG AMN RKS AJ AKV PS VJ drafted the manuscript. AKV PS VJ created the backbone, and AKV PS VJ SD critically revised the work. All authors agreed on the submission.

Corresponding authors

Correspondence to Vijayabhaskarreddy Junnuthula, Athira K. V. or Prashant Sadanandan.

Ethics declarations

Competing interests

The authors declare no competing interests, financial or otherwise.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nair, A., Greeny, A., Nandan, A. et al. Advanced drug delivery and therapeutic strategies for tuberculosis treatment. J Nanobiotechnol 21, 414 (2023). https://doi.org/10.1186/s12951-023-02156-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-023-02156-y

Keywords