Abstract

Non-tuberculous mycobacteria (NTM) cause pulmonary infections in patients with structural lung damage, impaired immunity, or other risk factors. Delivering antibiotics to the sites of these infections is a major hurdle of therapy because pulmonary NTM infections can persist in biofilms or as intracellular infections within macrophages. Inhaled treatments can improve antibiotic delivery into the lungs, but efficient nebulization delivery, distribution throughout the lungs, and penetration into biofilms and macrophages are considerable challenges for this approach. Therefore, we developed amikacin liposome inhalation suspension (ALIS) to overcome these challenges. Nebulization of ALIS has been shown to provide particles within the respirable size range that distribute to both central and peripheral lung compartments in humans. The in vitro and in vivo efficacy of ALIS against NTM has been demonstrated previously. The key mechanistic questions are whether ALIS penetrates NTM biofilms and enhances amikacin uptake into macrophages. We found that ALIS effectively penetrated throughout NTM biofilms and concentration-dependently reduced the number of viable mycobacteria. Additionally, we found that ALIS improved amikacin uptake by ∼4-fold into cultured macrophages compared with free amikacin. In rats, inhaled ALIS increased amikacin concentrations in pulmonary macrophages by 5- to 8-fold at 2, 6, and 24 h post-dose and retained more amikacin at 24 h in airways and lung tissue relative to inhaled free amikacin. Compared to intravenous free amikacin, a standard-of-care therapy for refractory and severe NTM lung disease, ALIS increased the mean area under the concentration-time curve in lung tissue, airways, and macrophages by 42-, 69-, and 274-fold. These data demonstrate that ALIS effectively penetrates NTM biofilms, enhances amikacin uptake into macrophages, both in vitro and in vivo, and retains amikacin within airways and lung tissue. An ongoing Phase III trial, adding ALIS to guideline based therapy, met its primary endpoint of culture conversion by month 6. ALIS represents a promising new treatment approach for patients with refractory NTM lung disease.

Highlights

  • Non-tuberculous mycobacteria (NTM) are commonly found throughout the environment and cause pulmonary infections in patients with structural lung damage, impaired immunity, or other risk factors (Johnson and Odell, 2014; van Ingen and Kuijper, 2014)

  • Amikacin liposome inhalation suspension effectively penetrated M. avium biofilms to reduce the number of viable bacteria; this was true for 100% amikacin liposome inhalation suspension (ALIS) as well as 70% ALIS/30% free amikacin

  • Retained more amikacin at 24 h in airways and lung tissue compared to free amikacin, which would increase the duration of amikacin exposure at the sites of biofilm infections within mucus and alveolar walls

Read more

Summary

Introduction

Non-tuberculous mycobacteria (NTM) are commonly found throughout the environment and cause pulmonary infections in patients with structural lung damage, impaired immunity, or other risk factors (Johnson and Odell, 2014; van Ingen and Kuijper, 2014). Clinical case reports have described intracellular MAC infections in humans, with M. avium detected inside peripheral blood leukocytes and bone marrow aspirate (Graham et al, 1984; Moffie et al, 1989). These findings indicate that MAC species can live and replicate within macrophages. Delivering effective antibiotic concentrations into NTM biofilms and cells infected with NTM are important components of treatment

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call