4,362
Views
48
CrossRef citations to date
0
Altmetric
Review

Potential of dry powder inhalers for tuberculosis therapy: facts, fidelity and future

, , &
Pages S791-S806 | Received 31 May 2018, Accepted 10 Aug 2018, Published online: 11 Oct 2018

References

  • Chetty S, Ramesh M, Singh-Pillay A, et al. Recent advancements in the development of anti-tuberculosis drugs. Bioorg Med Chem Lett. 2017;27:370–386.
  • Hoagland D, Liu J, Lee R, et al. New agents for the treatment of drug-resistant Mycobacterium tuberculosis. Adv Drug Deliv Rev. 2016;102:55–72.
  • WHO, Tuberculosis. Global tuberculosis report. Geneva, Switzerland: WHO; 2017.
  • Giovagnoli S, Schoubben A, Ricci M. The long and winding road to inhaled TB therapy: not only the bug’s fault. Drug Dev Ind Pharm. 2017;43:347–363.
  • Costa A, Pinheiro M, Magalhaes J, et al. The formulation of nanomedicines for treating tuberculosis. Adv Drug Deliv Rev. 2016;102:102–115.
  • Parumasivam T, Chang RY, Abdelghany S, et al. Dry powder inhalable formulations for anti-tubercular therapy. Adv Drug Deliv Rev. 2016;102:83–101.
  • Hickey A, Durham P, Dharmadhikari A, et al. Inhaled drug treatment for tuberculosis: past progress and future prospects. J Control Release. 2016;240:127–134.
  • Mehta P. Imagine the superiority of dry powder inhalers from carrier engineering. J Drug Deliv. 2018;2018:1–19.
  • Mehta P. Dry powder inhalers: a focus on advancements in novel drug delivery systems. J Drug Deliv. 2016;2016:1–17.
  • Lyapustina S. Regulatory pitfalls and opportunities when repurposing for inhalation therapy. Adv Drug Deliv Rev.
  • Sanders M. Inhalation therapy: an historical review. Prim Care Respir J. 2007;16:71–81.
  • Zhou Q, Tang P, Leung S, et al. Emerging inhalation aerosol devices and strategies: where are we headed? Adv Drug Deliv Rev. 2014;75:3–17.
  • Bardania H, Tarvirdipour S, Dorkoosh F. Liposome-targeted delivery for highly potent drugs. Artif Cells Nanomed Biotechnol. 2017;45:1478–1489.
  • Changsan N, Chan H, Separovic F, et al. Physicochemical characterization and stability of rifampicin liposome dry powder formulations for inhalation. J Pharm Sci. 2009;98:628–639.
  • Changsan N, Nilkaeo A, Pungrassami P, et al. Monitoring safety of liposomes containing rifampicin on respiratory cell lines and in vitro efficacy against Mycobacterium bovis in alveolar macrophages. J Drug Target. 2009;17:751–762.
  • Akhter M, Rizwanullah M, Ahmad J, et al. Nanocarriers in advanced drug targeting: setting novel paradigm in cancer therapeutics. Artif Cells Nanomed Biotechnol. 2018;46:873–884.
  • Lemmer Y, Kalombo L, Pietersen R, et al. Mycolic acids, a promising mycobacterial ligand for targeting of nano encapsulated drugs in tuberculosis. J Control Release. 2015;211:94–104.
  • Bhardwaj A, Kumar L, Narang R, et al. Development and characterization of ligand-appended liposomes for multiple drug therapy for pulmonary tuberculosis. Artif Cells Nanomed Biotechnol. 2013;41:52–59.
  • Khan I, Elhissi A, Shah M, et al. Liposome-based carrier systems and devices used for pulmonary drug delivery. Biomaterials and medical tribology research and development. Sawston, Cambridge: Woodhead Publishing Series in Biomaterials; 2013. p. 395–443.
  • Rojanarat W, Changsan N, Tawithong E, et al. Isoniazid proliposome powders for inhalation-preparation, characterization and cell culture studies. Int J Mol Sci. 2011;12:4414–4434.
  • Rojanarat W, Nakpheng T, Thawithong E, et al. Inhaled pyrazinamide proliposome for targeting alveolar macrophages. Drug Deliv. 2012;19:334–345.
  • Rojanarat W, Nakpheng T, Thawithong E, et al. Levofloxacin-proliposomes: opportunities for use in lung tuberculosis. Pharmaceutics. 2012;4:385–412.
  • Patil-Gadhe A, Pokharkar V. Single step spray drying method to develop proliposomes for inhalation: a systematic study based on quality by design approach. Pulm Pharmacol Ther. 2014;27:197–207.
  • Dudala T, Yalavarthi P, Vadlamudi H, et al. A perspective overview on lipospheres as lipid carrier systems. Int J Pharm Investig. 2014;4:149–155.
  • Singh C, Koduri L, Singh A, et al. Novel potential for optimization of antitubercular therapy: pulmonary delivery of rifampicin lipospheres. Asian J Pharm Sci. 2015;10:549–562.
  • Singh C, Koduri L, Dhawale V, et al. Potential of aerosolized rifampicin lipospheres for modulation of pulmonary pharmacokinetics and bio-distribution. Int J Pharm. 2015;495:627–632.
  • Kuzmov A, Minko T. Nanotechnology approaches for inhalation treatment of lung diseases. J Control Release. 2015;219:500–518.
  • Zhou Q, Leung S, Tang P, et al. Inhaled formulations and pulmonary drug delivery systems for respiratory infections. Adv Drug Deliv Rev. 2015;85:83–99.
  • Rawal T, Parmar R, Tyagi R, et al. Rifampicin loaded chitosan nanoparticle dry powder presents an improved therapeutic approach for alveolar tuberculosis. Colloids Surf B Biointerfaces. 2017;154:321–330.
  • Noraizaan A, Wong T. Physicochemical effects of lactose microcarrier on inhalation performance of rifampicin in polymeric nanoparticles. Powder Technol. 2017;310:272–281.
  • Garg T, Goyal A, Rath G, et al. Spray-dried particles as pulmonary delivery system of anti-tubercular drugs: design, optimization, in vitro and in vivo evaluation. Pharm Dev Technol. 2016;21:951–960.
  • Pourshahab P, Gilani K, Moazeni E, et al. Preparation and characterization of spray dried inhalable powders containing chitosan nanoparticles for pulmonary delivery of isoniazid. J Microencapsul. 2011;28:605–613.
  • Bhardwaj A, Mehta S, Yadav S, et al. Pulmonary delivery of anti-tubercular drugs using spray-dried lipid-polymer hybrid nanoparticles. Artif Cells Nanomed Biotechnol. 2016;44:1544–1555.
  • Debnath S, Saisivam S, Omri A. PLGA ethionamide nanoparticles for pulmonary delivery: development and in vivo evaluation of dry powder inhaler. J Pharm Biomed Anal. 2017;145:854–859.
  • Ahmad M, Ungphaiboon S, Srichana T. The development of dimple-shaped chitosan carrier for ethambutol dihydrochloride dry powder inhaler. Drug Dev Ind Pharm. 2015;41:791–800.
  • Debnath S, Saisivam S, Debanth M, et al. Development and evaluation of Chitosan nanoparticles based dry powder inhalation formulations of Prothionamide. PLoS One. 2018;13:e0190976.
  • Maretti E, Rustichelli C, Romagnoli M, et al. Solid Lipid Nanoparticle assemblies (SLNas) for an anti-TB inhalation treatment-A design of experiments approach to investigate the influence of pre-freezing conditions on the powder respirability. Int J Pharm. 2016;511:669–679.
  • Maretti E, Costantino L, Rustichelli C, et al. Surface engineering of Solid Lipid Nanoparticle assemblies by methyl α-d-mannopyranoside for the active targeting to macrophages in anti-tuberculosis inhalation therapy. Int J Pharm. 2017;528:440–451.
  • Afsharzadeh M, Hashemi M, Mokhtarzadeh A, et al. Recent advances in co-delivery systems based on polymeric nanoparticle for cancer treatment. Artif Cells Nanomed Biotechnol. 2018;46:1095–1110.
  • Pawar A, Rajalakshmi S, Mehta P, et al. Strategies for formulation development of andrographolide. RSC Adv. 2016;6:69282–69300.
  • Vadakkan M, Annapoorna K, Sivakumar K, et al. Dry powder cationic lipopolymeric nano micelle inhalation for targeted delivery of antitubercular drug to alveolar macrophage. Int J Nanomedicine. 2013;8:2871–2885.
  • Weber S, Zimmer A, Pardeike J. Solid Lipid Nanoparticles (SLN) and Nanostructured Lipid Carriers (NLC) for pulmonary application: a review of the state of the art. Eur J Pharm Biopharm. 2014;86:7–22.
  • Nagpal K, Mohan A, Thakur S, et al. Dendritic platforms for biomimicry and biotechnological applications. Artif Cells Nanomed Biotechnol. 2018;1–15.
  • Rajabnezhad S, Casettari L, Lam J, et al. Pulmonary delivery of rifampicin microspheres using lower generation polyamidoamine dendrimers as a carrier. Powder Technol. 2016;291:366–374.
  • Bohr A, Water J, Beck-Broichsitter M, et al. Nanoembedded microparticles for stabilization and delivery of drug-loaded nanoparticles. Curr Pharm Des. 2015;21:5829–5844.
  • Torge A, Grützmacher P, Mücklich F, et al. The influence of mannitol on morphology and disintegration of spray-dried nano-embedded microparticles. Eur J Pharm Sci. 2017;104:171–179.
  • Goyal A, Garg T, Rath G, et al. Development and characterization of nano embedded microparticles for pulmonary delivery of antitubercular drugs against experimental tuberculosis. Mol Pharm. 2015;12:3839–3850.
  • Gaspar D, Gaspar M, Eleutério C, et al. Microencapsulated solid lipid nanoparticles as a hybrid platform for pulmonary antibiotic delivery. Mol Pharm. 2017;14:2977–2990.
  • Ohashi K, Kabasawa T, Ozeki T, et al. One-step preparation of rifampicin/poly(lactic-co-glycolic acid) nanoparticle-containing mannitol microspheres using a four-fluid nozzle spray drier for inhalation therapy of tuberculosis. J Control Release. 2009;135:19–24.
  • Sharma V, Jain A, Soni V. Nano-aggregates: emerging delivery tools for tumor therapy. Crit Rev Ther Drug Carrier Syst. 2013;30:535–563.
  • Kaur R, Garg T, Malik B, et al. Development and characterization of spray-dried porous nanoaggregates for pulmonary delivery of anti-tubercular drugs. Drug Deliv. 2016;23:882–887.
  • Sung J, Padilla D, Garcia-Contreras L, et al. Formulation and pharmacokinetics of self-assembled rifampicin nanoparticle systems for pulmonary delivery. Pharm Res. 2009;26:1847–1855.
  • Durham P, Zhang Y, German N, et al. Spray dried aerosol particles of pyrazinoic acid salts for tuberculosis therapy. Mol Pharm. 2015;12:2574–2581.
  • Eedara B, Tucker I, Das S. Phospholipid-based pyrazinamide spray-dried inhalable powders for treating tuberculosis. Int J Pharm. 2016;506:174–183.
  • Pham D, Grégoire N, Couet W, et al. Pulmonary delivery of pyrazinamide-loaded large porous particles. Eur J Pharm Biopharm. 2015;94:241–250.
  • Kaewjan K, Srichana T. Nano spray-dried pyrazinamide-L-leucine dry powders, physical properties and feasibility used as dry powder aerosols. Pharm Dev Technol. 2016;21:68–75.
  • Vadakkan M, Kumar G. Cryo-crystallization under a partial anti-solvent environment as a facile technology for dry powder inhalation development. RSC Adv. 2015;5:73020–73027.
  • Parikh R, Dalwadi S. Preparation and characterization of controlled release poly-ε-caprolactone microparticles of isoniazid for drug delivery through pulmonary route. Powder Technol. 2014;264:158–165.
  • Maretti E, Rossi T, Bondi M, et al. Inhaled Solid Lipid Microparticles to target alveolar macrophages for tuberculosis. Int J Pharm. 2014;462:74–82.
  • Garcia Contreras L, Sung J, Ibrahim M, et al. Pharmacokinetics of inhaled rifampicin porous particles for tuberculosis treatment: insight into rifampicin absorption from the lungs of guinea pigs. Mol Pharm. 2015;12:2642–2650.
  • Takeuchi I, Taniguchi Y, Tamura Y, et al. Effects of L-leucine on PLGA microparticles for pulmonary administration prepared using spray drying: fine particle fraction and phagocytotic ratio of alveolar macrophages. Colloids Surf A Physicochem Eng Asp. 2018;537:411–417.
  • Brunaugh A, Jan S, Ferrati S, et al. Excipient-free pulmonary delivery and macrophage targeting of clofazimine via air jet micronization. Mol Pharm. 2017;14:4019–4031.
  • Verma R, Germishuizen W, Motheo M, et al. Inhaled microparticles containing clofazimine are efficacious in treatment of experimental tuberculosis in mice. Antimicrob Agents Chemother. 2013;57:1050–1052.
  • Garcia-Contreras L, Padilla-Carlin D, Sung J, et al. Pharmacokinetics of ethionamide delivered in spray-dried microparticles to the lungs of guinea pigs. J Pharm Sci. 2017;106:331–337.
  • Parumasivam T, Ashhurst A, Nagalingam G, et al. Inhalation of respirable crystalline rifapentine particles induces pulmonary inflammation. Mol Pharm. 2017;14:328–335.
  • Durham P, Young E, Braunstein M, et al. A dry powder combination of pyrazinoic acid and its n-propyl ester for aerosol administration to animals. Int J Pharm. 2016;514:384–391.
  • Ibrahim M, Hatipoglu M, Garcia-Contreras L. SHetA2 dry powder aerosols for tuberculosis: formulation design and optimization using quality by design. Mol Pharm. 2018;15:300–313.
  • Schoubben A, Giovagnoli S, Tiralti M, et al. Capreomycin inhalable powders prepared with an innovative spray-drying technique. Int J Pharm. 2014;469:132–139.
  • Kulkarni V, Shaw C. Microscopy techniques. Essential chemistry for formulators of semisolid and liquid dosages. 1st ed. ‎Amsterdam, Netherlands: Elsevier; 2016. p. 183–192.
  • Pai R, Jain R, Bannalikar A, et al. Development and evaluation of chitosan microparticles based dry powder inhalation formulations of rifampicin and rifabutin. J Aerosol Med Pulm Drug Deliv. 2016;29:179–195.
  • Onoshita T, Shimizu Y, Yamaya N, et al. The behavior of PLGA microspheres containing rifampicin in alveolar macrophages. Colloids Surf B Biointerfaces. 2010;76:151–157.
  • Doan T, Couet W, Olivier J. Formulation and in vitro characterization of inhalable rifampicin-loaded PLGA microspheres for sustained lung delivery. Int J Pharm. 2011;414:112–117.
  • Liu Z, Li X, Xiu B, et al. A novel and simple preparative method for uniform-sized PLGA microspheres: preliminary application in antitubercular drug delivery. Colloids Surf B Biointerfaces. 2016;145:679–687.
  • Sethuraman V, Hickey A. Powder properties and their influence on dry powder inhaler delivery of an antitubercular drug. AAPS PharmSciTech. 2002;3:E28.
  • Liu C, Kong C, Wu G, et al. Uniform and amorphous rifampicin microspheres obtained by freezing induced LLPS during lyophilization. Int J Pharm. 2015;495:500–507.
  • Park J, Jin H, Kim D, et al. Chitosan microspheres as an alveolar macrophage delivery system of ofloxacin via pulmonary inhalation. Int J Pharm. 2013;441:562–569.
  • Hwang S, Kim D, Chung S, et al. Delivery of ofloxacin to the lung and alveolar macrophages via hyaluronan microspheres for the treatment of tuberculosis. J Control Release. 2008;129:100–106.
  • Parumasivam T, Leung S, Quan D, et al. Rifapentine-loaded PLGA microparticles for tuberculosis inhaled therapy: Preparation and in vitro aerosol characterization. Eur J Pharm Sci. 2016;88:1–11.
  • Gupta A, Pant G, Mitra K, et al. Inhalable particles containing rapamycin for induction of autophagy in macrophages infected with Mycobacterium tuberculosis. Mol Pharm. 2014;11:1201–1207.
  • Brooks B, Brooks A. Therapeutic strategies to combat antibiotic resistance. Adv Drug Deliv Rev. 2014;78:14–27.
  • Weers J. Inhaled antimicrobial therapy - barriers to effective treatment. Adv Drug Deliv Rev. 2015;85:24–43.
  • Kadota K, Senda A, Tagishi H, et al. Evaluation of highly branched cyclic dextrin in inhalable particles of combined antibiotics for the pulmonary delivery of anti-tuberculosis drugs. Int J Pharm. 2017;517:8–18.
  • Chan J, Chan H, Prestidge C, et al. A novel dry powder inhalable formulation incorporating three first-line anti-tubercular antibiotics. Eur J Pharm Biopharm. 2013;83:285–292.
  • Kumar Verma R, Mukker J, Singh R, et al. Partial biodistribution and pharmacokinetics of isoniazid and rifabutin following pulmonary delivery of inhalable microparticles to rhesus macaques. Mol Pharm. 2012;9:1011–1016.
  • Momin MAM, Tucker I, Doyle C, et al. Manipulation of spray-drying conditions to develop dry powder particles with surfaces enriched in hydrophobic material to achieve high aerosolization of a hygroscopic drug. Int J Pharm. 2018;543:318–327.
  • Momin MAM, Tucker I, Doyle C, et al. Co-spray drying of hygroscopic kanamycin with the hydrophobic drug rifampicin to improve the aerosolization of kanamycin powder for treating respiratory infections. Int J Pharm. 2018;541:26–36.
  • Rodrigues S, Alves A, Cavaco J, et al. Dual antibiotherapy of tuberculosis mediated by inhalable locust bean gum microparticles. Int J Pharm. 2017;529:433–441.
  • Lau J, Dunn M. Therapeutic peptides: historical perspectives, current development trends, and future directions. Bioorg Med Chem. 2018;26:2700–2707.
  • Depreter F, Pilcer G, Amighi K. Inhaled proteins: challenges and perspectives. Int J Pharm. 2013;447:251–280.
  • Chaulagain B, Jain A, Tiwari A, et al. Passive delivery of protein drugs through transdermal route. Artif Cells Nanomed Biotechnol. 2018;1–16.
  • Mehta P, Pawar V. Electrospun nanofiber scaffolds: technology and applications. Applications of nanocomposite materials in drug delivery. Sawston, Cambridge: Woodhead Publishing Series in Biomaterials; 2018. p. 509–573.
  • Kwok P, Grabarek A, Chow M, et al. Inhalable spray-dried formulation of D-LAK antimicrobial peptides targeting tuberculosis. Int J Pharm. 2015;491:367–374.
  • Tyne A, Chan J, Shanahan E, et al. TLR2-targeted secreted proteins from Mycobacterium tuberculosis are protective as powdered pulmonary vaccines. Vaccine. 2013;31:4322–4329.
  • Muralidharan P, Hayes D, Jr, Mansour H. Dry powder inhalers in COPD, lung inflammation and pulmonary infections. Expert Opin Drug Deliv. 2015;12:947–962.
  • Mehta PP. Multi-dose dry powder inhaler: advance technology for drug delivery to airways. India Drugs. 2018. Available from: https://www.indiandrugsonline.org/accepted-article-details?id=MTE0MDM=
  • Rogueda P, Traini D. The future of inhalers: how can we improve drug delivery in asthma and COPD? Expert Rev Respir Med. 2016;10:1041–1044.
  • Mehta P, Pawar A, Mahadik K, et al. Emerging novel drug delivery strategies for bioactive flavonol fisetin in biomedicine. Biomed Pharmacother. 2018;106:1282–1291.
  • Frick M. Funding for tuberculosis research-an urgent crisis of political will, human rights, and global solidarity. Int J Infect Dis. 2017;56:21–24.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.