Expired activity
Please go to the PowerPak homepage and select a course.

Current and Emerging Therapies for Non-Cystic Fibrosis Nontuberculous Mycobacterial Lung Disease: What Pharmacists Need to Know

INTRODUCTION

Human infections due to Mycobacterium tuberculosis (known as tuberculosis or TB) are well known by medical providers, but nontuberculous mycobacterial (NTM) infections, also known as mycobacterium other than tuberculosis, are not as well known. Similar to TB, NTM infections can manifest in myriad syndromes, including skin and soft tissue infections and lymphadenitis, but they occur most often as pulmonary infections. Treatment of mycobacterial infections involves complex medication regimens taken for long periods of time, and pharmacists have ample opportunities to be involved in the care processes of patients with mycobacterial disease. This review will describe the microbiology, epidemiology, and treatment of pulmonary NTM disease caused by the 3 most common isolated species, with a focus on potential contributions of pharmacists.

MICROBIOLOGY

Mycobacteria are aerobic, acid-fast positive organisms with a thick, lipid-rich, hydrophobic cell wall.1 The impermeability of the cell wall, along with its ability to form biofilms, renders mycobacteria resistant to antimicrobials, high temperatures, and disinfectants.2 NTM are ubiquitous in the environment, but they have a preference for water and soil reservoirs. The mode of transmission is not well established, but inhalation and ingestion are the leading hypotheses of transmission: direct person- to-person transmission is not thought to play a significant role. Hospital water systems, hemodialysis centers, peat-rich potting soil, and hot tubs are commonly colonized with NTM and, in some cases, can lead to nosocomial outbreaks.3,4

NTM comprises more than 100 species, many of which were discovered only in recent years due to advances in culturing and molecular techniques.5 The various species have classically been divided into rapid (< 7 days) and slow (> 7 days) growers. Of those known to cause human disease, notable rapid growers include Mycobacterium abscessus complex (MABC), M. fortuitium, and M. chelonae; slow growers are M. avium complex (MAC) and M. kansasii. MABC comprises subspecies abscessus, bolletii, and massiliense, and MAC is further divided into M. avium, M. intracellulare, and M. chimaera.5 Among these isolates, MAC is the most common cause of pulmonary disease, followed by M. kansasii, and M. abscessus.6 The vast majority of cultures positive for NTM are from pulmonary sources, and skin and soft tissue infections are the second-most common source of isolates. Identification of the mycobacterial species is key to the proper management of the associated disease.

EPIDEMIOLOGY

The true incidence of NTM infections is difficult to determine, since positive cultures do not always indicate the presence of clinical disease. Given the nearly universal presence of NTM in the environment, each culture result needs to be weighed carefully in the context of several factors, including imaging and patient symptoms, to determine its relevance. Another consideration making determination of disease incidence difficult is that reporting of NTM to public health officials is not mandated, as it is with TB.

Despite these barriers to identification and reporting of NTM infections, there is a general consensus that NTM infections are increasing throughout the world7: a review that evaluated the use of laboratory results along with patient characteristics reported an NTM incidence of 4.1 to 14.1 cases per 100,000 patient years.8 However, it has been a matter of debate whether the disease incidence is truly increasing or if improved ability to detect disease through newer culture techniques and clinician awareness is the cause of the increase.9 Proposed reasons for a rise in NTM infections include air pollution, an aging population, increases in comorbidities, and the increasing use of immunosuppressive therapies.4,10

Host factors

Most people are likely exposed to NTM in their lifetimes, though only a few people will develop an active infection. The factors predisposing certain patients to NTM infection are not well understood, but certain demographic characteristics are common among those diagnosed with clinical disease. One patient group with a known predisposition to NTM infection is slender, postmenopausal women with mitral valve prolapse.11,12 Pulmonary NTM infection is also strongly associated with pre-existing lung disease such as bronchiectasis, chronic obstructive pulmonary disease (COPD), cystic fibrosis, and pneumonitis.13 However, the question of which comes first—the bronchiectasis or the NTM infection— has been difficult to delineate: bronchiectasis is considered a risk factor for NTM, though it may be a clinical manifestation of the disease itself. Interestingly, cigarette smoking in the absence of COPD is not an established predisposing factor for NTM infection. The immunosuppressed population, including patients with HIV/AIDS, those who received a solid organ transplant, and those on anti-tumor necrosis factor alpha therapy, is an established at-risk group. The factors influencing these increased risks are outside the scope of this monograph and will not be discussed in detail.

PRESENTATION AND DIAGNOSIS

The American Thoracic Society (ATS) and the Infectious Diseases Society of America (IDSA) jointly published a guideline to aid in the diagnosis and management of NTM disease.6 Diagnosis involves the synthesis of patient signs and symptoms, symptom severity, radiologic results, and microbiologic cultures along with the exclusion of other potential causes of disease. Clinical symptoms are variable and not present in all patients, but commonly reported symptoms are chronic cough with or without sputum production, weight loss, fatigue/malaise, and night sweats. A hallmark radiographic sign of pulmonary NTM is the "tree in bud" finding of small nodules of less than 0.5 mm in diameter. Other signs include fibrocavitary disease, parenchymal destruction, and diffuse consolidations due to hypersensitivity pneumonitis.5

Since mycobacteria are ubiquitous in the environment, the microbiology criteria for diagnosis mandates at least 2 or more positive sputum cultures, at least one culture from a bronchial washing or lavage, or biopsy positive with histologic features of mycobacterium.6 Most laboratories use at least a 6- week incubation period for mycobacterial cultures, which results in the need for ongoing culture surveillance from the provider suspecting NTM disease. Other disease states, including TB, malignancy, and invasive fungal or bacterial infections, should be ruled out prior to initiation of NTM therapy.14

TREATMENT

Treatment of NTM lung disease depends on several factors: the causative species, disease severity, and patient-specific risk factors and considerations. Dosing frequency, potential adverse effects drug interactions, and goals of care are also important issues that the care team must consider when choosing therapy for NTM infections. Two guidelines are available to guide the selection of treatment.

Mycobacterium avium complex

Pulmonary MAC disease is usually classified as 1 of 2 clinical entities: cavitary disease or nodular/bronchiectatic disease. The more severe form of progressive apical fibrocavitary lung disease tends to present in middle-aged males who have a history of cigarette smoking and alcohol consumption. The less aggressive disease presents with nodular infiltrates and tends to occur in post- menopausal white women.

Treatment for MAC pulmonary disease follows 1 of 2 treatment paradigms: daily drug therapy or thrice-weekly therapy. Outcomes are comparable between daily and thrice-weekly therapy in patients with the less severe form of pulmonary disease (nodular/bronchiectatic MAC lung disease).15-17 Reduced overall pill burden and the potential for decreased adverse effects and toxicity with lower drug exposure makes 3-times-weekly therapy a desirable approach, especially for older patients—a group that accounts for a significant portion of the infected population. However, in patients with cavitary disease, a 3-times-weekly drug dosing regimen with a macrolide, ethambutol, and rifampin has shown a very low rate of culture conversion (only 4%) at 12 months of therapy.16 The recommend regimens for initial treatment of MAC pulmonary disease from the 2007 ATS/IDSA and 2017 British Thoracic Society (BTS) guidelines are outlined in Table 1.6,18

Table 1: Drug Therapy for Pulmonary Mycobacterium avium Complex Infection6,18
  Drug and dose
Disease Macrolidea Ethambutolb Rifampinc Aminoglycosided
Bronchiectatic/nodular disease (mild-moderate) 3x weekly:
clarithromycin 1000 mg or azithromycin 500-600 mg
3x weekly: 25 mg/kg 3x weekly: 600 mg none
Cavitary disease (moderate-severe) Daily:
clarithromycin 1000 mg or azithromycin 250-300 mg
Daily: 15 mg/kg Daily: 600 mg Daily: 15 mg/kg

3x weekly: 25 mg/kg
nebulized AMK
Abbreviations: AMK, amikacin.

a Give clarithromycin in 2 equal, divided doses. May consider 500 mg total daily dose for patients with body weight < 50 kg on daily therapy.
b Dose using ideal body weight (IBW).
c May consider rifampin 450 mg if patient weighs < 50 kg.
d Streptomycin or amikacin. Intravenous (IV) aminoglycoside dosing recommendations vary. British Thoracic Society (BTS) guidelines recommend the use of dosing body weight for obese patients (patient weighs > 20% over IBW) with target Cmax of 35-45 mcg/mL for 15 mg/kg dose and Cmax of 65-80 mcg/mL for 25 mg/kg dose. BTS guidelines offer nebulized amikacin as an option in place of IV therapy.

Macrolides are the primary class of medications used to treat pulmonary MAC infection. Clarithromycin and azithromycin have excellent in vitro activity against MAC; minimum inhibitory concentration (MIC) values are attainable in serum with standard doses of clarithromycin and azithromycin; and clarithromycin and azithromycin provide enhanced concentrations at the site of infection in both phagocytes and tissue.19-21

Two early, open-label, non-comparative trials demonstrated that azithromycin and clarithromycin monotherapy yielded favorable microbiologic and clinical responses. Azithromycin and clarithromycin monotherapy provided rates of microbiologic conversion of 38% and 58%, with notable reductions in sputum microbiologic growth of 76% and 79%, respectively.15,20,22 However, macrolide resistance developed with clarithromycin monotherapy.15

Multi-drug therapy with a macrolide backbone provides more favorable outcomes than macrolide monotherapy. Ethambutol and rifamycins have synergy against MAC isolates23 and have provided protection against the development of macrolide resistance in disseminated MAC disease,24 which has led to its use in combination with macrolide therapy. Clinically, combination therapy with clarithromycin, a rifamycin, ethambutol, and an early period of 2 to 4 months of streptomycin yielded 36 of 39 (92%) patients with sputum conversion and prolonged maintenance of negative cultures after therapy completion.21 Enhanced clinical improvement and maintenance of culture negativity were also achieved when azithromycin was combined with rifabutin, ethambutol, and an initial 2 months of streptomycin.25

Rifampin is the preferred rifamycin for the treatment of MAC pulmonary disease. Rifabutin has microbiologic activity against MAC isolates and has more potent in vitro activity against MAC than rifampin.26 However, rifabutin's adverse effects (e.g., arthralgias, neutropenias, cytopenias, uveitis) are notable in older patents. As such, rifabutin is poorly tolerated compared to rifampin in a common patient population with MAC disease, which has led to the use of rifampin as the preferred initial rifamycin for MAC pulmonary disease.15,21,22,25,27,28

An initial period of 2 to 3 months of an injectable agent (usually streptomycin or amikacin) is provided as an option in severe MAC disease. The lack of a strong recommendation for routine use in all cases is attributable to a lack of definitive clinical outcome enhancement when used as an adjunct to therapy. For instance, when intramuscular (IM) streptomycin was administered 3 times weekly for the first 3 months of therapy (added to rifampin, ethambutol, and clarithromycin), it yielded a statistically significant improvement in microbiologic sputum conversion (71.2% vs. 50.7%) at 2 years, though clinical relapse rate, clinical symptom improvement, and radiologic findings did not otherwise differ.29 Further, the IM administration route can be a deterrent to streptomycin use. Amikacin has a similar kinetic profile and similar safety data to streptomycin,30 so it is often the selected aminoglycoside agent in severe cavitary disease.

Mycobacterium kansasii

M. kansasii infections tend to have a similar presentation and clinical course to TB. Patients can have fever, chills, weight loss, and night sweats. Cavitary disease often has a predilection for the upper lobes of the lungs, though non-cavitary bronchiectatic disease is also seen. Surgery does not have a role in M. kansasii infections because drug therapy achieves excellent clinical and microbiologic responses.6

Treatment for M. kansasii is daily therapy with a 3-drug combination. The recommended treatment regimen described in the 2007 ATS/IDSA and 2017 BTS guidelines is outlined in Table 2.6,18 Historically, when investigating drug therapy for M. kansasii, drugs used for TB were evaluated, leading to early clinical data supporting the use of rifampin, ethambutol, and isoniazid (INH). One consideration of M. kansasii drug sensitivity is the MIC results for INH: this drug is useful in multi-drug regimens to treat M. kansasii and clinical data support its use, but the MICs of INH tend to be quite high, though this may not always translate into lack of response. Resistance may be reported at an MIC of 1 mcg/mL, but resistant isolates have been shown to respond to multi-drug regimens containing rifampin.31,32 Macrolides have been shown to have excellent in vitro activity against M. kansasii isolates and efficacy has been demonstrated in a small trial,33 leading to the incorporation of macrolide therapy as a third option in place of INH in the BTS guidelines. Similar to its role in TB, rifampin is of critical importance to the success of the treatment regimen for M. kansasii.

Table 2: Drug Therapy for Mycobacterium kansasii Infection6,18

Rifampin Ethambutol Third agent (choose one)
600 mg daily 15 mg/kg daily Isoniazid 300 mg daily Macrolide: clarithromycin 500 mg twice daily or azithromycin 250 mg daily

Mycobacterium abscessus

M. abscessus pulmonary infection has been notoriously challenging to treat: isolates have large variability in antibiotic sensitivity and outcomes for drug therapy (particularly macrolides) vary in the literature. Many factors contribute to these challenges, but the variation in antibiotic responses of different subspecies plays a significant role in the difficult nature of M. abscessus treatment. The predominant difference contributing to macrolide response among the subspecies of M. abscessus (M. abscessus subspecies abscessus, M. abscessus subspecies bolletii, and M. abscessus subspecies massiliense)—collectively referred to as MABC—is the presence of a functional inducible erm gene for macrolide resistance. Further, no multi-drug regimen to treat M. abscessus pulmonary disease has been found that reliably leads to cure or culture conversion for an extended duration. This must be considered when determining goals of therapy for patients: antibiotic therapy is often aimed more at improving quality of life and achieving symptomatic improvement through suppression of microbial growth than at curing the infection. However, with limited disease and surgery, prolonged culture conversion is possible.34

Drug sensitivity results, patient tolerability, and a patient's ultimate goals of care will dictate the selection of a treatment course for M. abscessus pulmonary disease. The clinical treatment paradigms highlighted by the 2007 ATS/IDSA guidelines and 2017 BTS guidelines are similar. Both recommendations advocate the use of an initial intensive phase of intravenous (IV) therapy with an oral macrolide followed by a continuation phase of oral antimicrobials that are selected on the basis of patient tolerability and the antibiotic sensitivity profile. Table 3 lists the recommendations for the initial intensive phase of therapy for M. abscessus treatment. Neither the ATS/IDSA nor the BTS guidelines' determinations are based on randomized controlled trial data. Most data were from observational studies, case series, and case reports; guideline recommendations also incorporate expert opinions.6,18

Table 3: Drug Therapy Recommendations for Mycobacterium abscessus Infection6,18
PO Macrolidea IV Amikacinb IV b-lactam IV Tigecycline
Clarithromycin 500 mg twice daily
or azithromycin 250 mg daily
Daily: 15 mg/kg

3x weekly: 25 mg/kg
Imipenem 1 gm twice daily or cefoxitin 200 mg/kg/day (max 12 gm) divided in 3 doses 50 mg twice daily
Abbreviations: IV, intravenous; PO, oral.
aPending sensitivity profile.
bConsideration for inhaled amikacin if IV administration is contraindicated or impractical.

The regimen selected for initial therapy within the ATS/IDSA and BTS guidelines constitute agents shown in the majority of large case series to result in clinical improvement, microbiological improvement, radiological improvement, and symptom improvement. The guidelines advocate the use of these initial agents for a period of at least 4 weeks up to 4 months, though they acknowledge that tolerability may challenge patients and limit treatment duration.6,18 The BTS guidelines further advocate for consideration of extending initial IV therapy up to 3 to 6 months if tolerated by patients with inducible or constitutional macrolide resistance, as outcomes are worse in this setting.18

Clinical outcomes are improved when M. abscessus isolates are macrolide susceptible and macrolides are included in the treatment program.35,36 However, subspecies have varying macrolide resistance profiles. Subspecies bolletii and abscessus have copies of a functional erm41 gene for inducible resistance; subspecies massiliense has a nonfunctional erm gene and lacks inducible resistance.37 MABC isolates can also have acquired (rrl) resistance. Erm41 induction leads to methylation of the macrolide binding site on the 23S rRNA, and rrl mutations change the structure of the 23S rRNA, permanently preventing macrolide binding to the active site. Acquired resistance can be seen on standard resistance panels, but inducible resistance is not detectable by this method38 and requires culture sensitivity assessment at day 3 to 5 and again at day 14 for detection. However, DNA sequencing techniques can identify a functional erm41 gene sooner.36 Despite the presence of inducible resistance, macrolides may retain in vivo efficacy in 20% of M. abscessus subsp. abscessus isolates.39 Furthermore, azithromycin may be preferred over clarithromycin, since it has less potential to induce the erm41 gene.40

After the initial, intensive phase of IV therapy, 2 to 4 oral agents with activity according to the sensitivity panel are typically chosen on the basis of patient tolerability, drug interactions, and patient- specific factors with goals of care in mind. Potential agents include nebulized amikacin, ongoing oral macrolide (clarithromycin/azithromycin), clofazimine, linezolid, minocycline, moxifloxacin, and sulfamethoxazole-trimethoprim.6,18

NTM treatment duration and goals of therapy

For pulmonary disease caused by MAC or M. kansasii, where curative potential is a possibility, the duration of antimicrobial therapy is usually 18 to 24 months. This includes the continuation of therapy for 12 months after culture conversion.6,18 M. abscessus pulmonary disease is more challenging and curative potential may not be possible for many patients, except for cases of minimal disease with surgical intervention. In this circumstance, goals of care should be decided between patient and provider. Often, goals are designed to minimize symptoms through bacterial burden reduction and ongoing antimicrobial suppression and balance the benefits of therapy with the tolerability of the selected antibiotic regimen. Therapeutic failure is determined in cases of MAC or M. kansasii if patients do not have clinical response after 6 months and/or lack culture conversion by 6 to 12 months.

In cases of M. abscessus, the clinical case and goals of care should be ascertained by an experienced NTM provider to determine if the symptom-guided goals are appropriately met by the antimicrobial program, assuming adjustments have been made to the regimen accordingly. For patients who undergo surgery with curative intent, antibiotic regimens are typically continued for 12 months after surgery and culture conversion.41,42

Table 4 lists the product information, standard dosing, and adverse effects for the agents used to treat pulmonary NTM disease.

Table 4: Product Information, Standard Dosing, and Adverse Effects of Agents Used to Treat Pulmonary Non-tuberculous Mycobacterial Disease
Agent Dosage form/availability Standard dose Adverse effects
Amikacin sulfate
(parenteral)
IV/IM injection solution: 250 mg/mL Daily: 15 mg/kg1
3x weekly: 25 mg/kg1
Ototoxocity: vestibular and auditory
Nephrotoxicity
Electrolyte abnormalities (K+, Mg2+, Ca2+)
  Amikacin sulfate
(inhaled)*
IV/IM injection solution: 250 mg/mL 250-500 mg once or twice daily Dysphonia
Hoarseness
Cough
Oral pain/discomfort
Amikacin liposome
inhalation suspension (ALIS)
ALIS solution: 590 mg 590 mg inhaled via nebulizer once daily Dysphonia
Bronchiectasis exacerbation
Cough
Oral pain
Azithromycin Oral tablet: 250 mg, 500 mg, 600 mg
Oral powder for suspension
IV powder for solution
Daily: 250-300 mg
3x weekly (MAC): 500 mg
GI effects: abdominal pain, diarrhea, flatulence, nausea, vomiting
Headache
Rare: hearing loss, QT prolongation
Bedaquiline fumarate Oral tablet: 100 mg Weeks 1-2: 400 mg once daily with food
Weeks 3-24: 200 mg 3x weekly with food (doses at least 48 hours apart) for a total dose of 600 mg per week
Nausea
Arthralgia
Headache
Chest pain
QT prolongation
Cefoxitin IV powder for solution
IV solution: 1 gm/50 mL, 2 gm/50 mL
200 mg/kg/day (max 12 g/day) in 3 divided doses Rash
Leukopenia
Thrombocytopenia
Transaminitis
Clarithromycin Oral tablet: 250 mg, 500 mg
Oral powder for suspension
Oral tablet (XR): 500 mg
500 mg twice daily GI effects: metallic taste, diarrhea, nausea, vomiting, indigestion, abdominal pain
Headache
Rare: hearing loss, QT prolongation
Clofazimine Oral capsule: 50 mg, 100 mg 100 mg daily GI effects: nausea, vomiting, diarrhea, anorexia, abdominal pain, weight loss
Orange discoloration of skin and body fluids2
Photosensitivity, rash, pruritus, dryness/scaling of skin
Rare: QT prolongation, hepatitis, jaundice
Ethambutol Oral tablet: 100 mg, 400 mg Daily: 15 mg/kg
3x weekly: 25 mg/kg
Ocular toxicity: optic neuritis, often manifested as decreased visual acuity or decreased red-green color discrimination
Rare: peripheral neuropathy
Imipenem/cilastatin (I/C) Lyophilized powder (for reconstitution) in a 1:1 ratio of (I/C) for IV administration Imipenem 1000 mg/cilastatin 1000 mg IV twice daily Generally well tolerated
Low risk of GI effects: nausea, vomiting, diarrhea
Rare: seizure (noted with CNS infection)
Isoniazid (INH)3 Oral tablet: 50 mg, 100 mg, 300 mg
Oral syrup: 50 mg/mL
Aqueous solution: 100 mg/mL for IV/IM administration
5 mg/kg daily (max 300 mg) Asymptomatic transaminase elevations (up to 3-5x the upper limit of normal)
Hepatitis
Peripheral neurotoxicity
Rash
Rare: Hypersensitivity reactions
Linezolid3 Oral tablet: 600 mg
Oral suspension:
100 mg/5 mL
IV solution: 2 mg/mL
600 mg daily GI effects: nausea, vomiting, diarrhea
Headache
Myelosuppression: decreased platelets, decreased white blood cells, and/or anemia
Rare: optic neuritis, peripheral neuropathy, lactic acidosis, seizure
Moxifloxacin Oral tablet: 400 mg
IV solution:
400 mg/250 mL
400 mg daily GI effects: nausea, bloating, diarrhea
CNS effects: headache, dizziness, insomnia, tremulousness, agitation
QTc prolongation
Rare: hepatotoxicity, tendon rupture, arthralgias (can usually be treated symptomatically), peripheral neuropathy, seizures
Rifabutin Oral capsule: 150 mg 5 mg/kg daily (typical max 300 mg) Uveitis, arthralgias
Hematologic/immunologic toxicity: leukopenia, neutropenia
Rash/pruritis (generally self-limited)
Orange discoloration/staining of body fluids: sweat, tears, urine, stool, saliva
GI upset: nausea, indigestion, abdominal pain
Hepatotoxicity: cholestatic picture with asymptomatic elevated bilirubins more common
Rifampin Oral capsule:
150 mg, 300 mg
Powder for reconstitution and IV administration
Daily: 10 mg/kg (max 600 mg)
3x weekly: 10 mg/kg (max 600 mg)
Flu-like syndrome
Rash/pruritis (generally self-limited)
Orange discoloration/staining of body fluids: sweat, tears, urine, stool, saliva
GI upset: nausea, anorexia, abdominal pain
Hepatotoxicity: cholestatic picture with asymptomatic elevated bilirubin common
Hematologic/immunologic toxicity: thrombocytopenia, hemolytic anemia
Streptomycin 1-gm vials of aqueous solution for IV/IM injection Daily: 15 mg/kg1
3x weekly: 25 mg/kg1
Ototoxicity: vestibular and auditory
Nephrotoxicity
Electrolyte abnormalities (K+, Mg2+, Ca2+)
Tedizolid Oral tablet: 200 mg
IV powder for solution
200 mg daily GI effects: nausea, vomiting, diarrhea
Headache
Myelosuppression: decreased level of platelets, white blood cells, and/or anemia
Rare: peripheral neuropathy
Tigecycline IV powder for solution Loading dose: 100 mg
Maintenance dose: 50 mg every 12h
Nausea, vomiting, diarrhea, anorexia, dyspepsia, rash, dizziness
*The parenteral formulation of amikacin is delivered as a solution for inhalation.
Abbreviations: CNS, central nervous system; GI, gastrointestinal; IM, intramuscular; IV, intravenous.

1For obese patients, use adjusted body weight (ABW) instead of ideal body weight (IBW) for dosing: ABW = IBW + 0.4 (ABW – IBW). Serum levels should be monitored to guide appropriate dosing, optimize efficacy, and minimize toxicity.
2 Resolution of skin discoloration with clofazimine may take 6 months or longer. Monitor for changes in mood/depression associated with skin discoloration.
3 Vitamin B6 supplementation
      a INH can cause vitamin B6 depletion, which can lead to neuropathy. Vitamin B6 25-50 mg daily should be considered in all patients, but
      specially in patients at high risk of neuropathy such as women who are pregnant or breastfeeding; patients with HIV, diabetes,
      malnourishment, or alcoholism; and patients over the age of 65 years.
      b Linezolid: Vitamin B6 supplementation may decrease cytopenias, though data is limited. Consider vitamin B6 25 mg daily.

REFRACTORY NTM DISEASE

The treatment of pulmonary NTM disease is notably limited by a lack of randomized controlled trial data. Refractory disease treatment is even more limited, with small case series, observational studies, and retrospective reports providing the majority of information on treatment options. Often, MIC data yielded from antibiotic sensitivity assessments in patients with refractory disease suggest that in vitro activity may offer the possibility of clinical benefit. Typically, 1 or 2 agents are selected to add to a current regimen on the basis of patient-specific factors, end organ function, potential adverse drug reactions, drug interactions, or medication availability. Data regarding some of the agents used in refractory pulmonary NTM disease are outlined below. The use of inhaled liposomal amikacin in the clinical setting is increasing and ongoing randomized trials are investigating its applications in NTM treatment, so this agent is discussed in detail.

Inhaled amikacin

Amikacin sulfate in a concentration of 250 mg/mL is often used to compound the IV formulation of administered amikacin. Dosing is variable, but, commonly, 1 mL of this concentration of amikacin is added to 3 mL saline; this mixture is then administered via nebulizer once daily over 20 to 30 minutes. The dose is usually increased after 1 to 2 weeks to 250 mg twice daily. Then, as tolerability allows, the dose can be further increased to 500 mg twice daily, which is the typical maximum recommended dose. Inhaled amikacin has several benefits over parenteral amikacin: 1) the theoretical advantage of delivering a high concentration of drug directly to the site of action favors efficacy of the antimicrobial; 2) direct delivery via the lung reduces systemic exposure and minimizes toxicity; and 3) durations of therapy can be extended due to the lower risk of systemic toxicity that limits the chronic use of IV aminoglycosides.

Amikacin liposome inhalation suspension (ALIS) consists of small, charge-neutral liposomal particles; the amikacin sulfate salt form provides amikacin in its water-soluble, positively-charged form. Amikacin liposome inhalation suspension offers potential benefits over the sulfate form. First, with a neutral charge, the liposomal particles can theoretically penetrate macrophages more efficiently and provide higher intracellular amikacin directly to the site of mycobacteria. Second, the charge-neutral liposomes should theoretically be able to more effectively penetrate electrically charged biofilm than the amikacin salt. Finally, the liposomal delivery to intracellular lung tissue should serve to create a drug depot where drug action is sustained over time and systemic exposure may be even less than with the amikacin salt form.43,44

Two small, retrospective case series of 20 and 25 patients with refractory MAC or MABC pulmonary infection showed benefit with the addition of inhaled amikacin sulfate to current antibiotics.45,46 Inhaled amikacin sulfate led to sputum conversion in 25% to 44% of patients with 30% improvement in computed tomography imaging of the chest and 27% to 45% improvement in symptom scores. Adverse effects varied, and no patients discontinued treatment in one of the studies and only mild oral discomfort/hoarseness was reported.46 However, 30% of patients in the other study discontinued treatment, with ototoxicity, hemoptysis, nephrotoxicity, dysphonia, and vertigo cited as the reasons for discontinuations.45

A phase 2 randomized controlled trial involving 89 patients evaluated the addition of ALIS to current therapy in refractory MAC or MABC pulmonary disease against placebo. While a decrease was seen in mean semi-quantitative mycobacterial culture (the primary outcome), this change did not reach clinical significance (p = 0.072). However, there were statistically significant improvements in the secondary outcomes of sputum conversion (32% vs. 9%, p = 0.006) and the 6-minute walk test. In all, 15% of patients discontinued ALIS in this study, most commonly due to bronchiectasis exacerbation and dyspnea.47 A larger phase 3 study with ALIS is currently underway.

Other agents for the treatment of refractory disease

Moxifloxacin 400 mg daily demonstrated successful culture conversion when added to patients' antimicrobial regimen in the setting of refractory MAC pulmonary disease. Of 41 patients evaluated in a retrospective case series, 12 (29%) ultimately had culture conversion after moxifloxacin addition.48

Bedaquiline has shown potent in vitro activity against both MAC and MABC isolates. Currently, bedaquiline is approved in the United States (U.S.) for TB treatment, but it lacks approval for NTM. In a retrospective case evaluation of 10 patients from Texas with refractory MAC or MABC, bedaquiline demonstrated favorable clinical results. Six (60%) patients had improvement in semi-quantitative cultures, 4 (40%) had radiographic improvement, and 9 (90%) had symptom improvement.49 The high cost of bedaquiline is a considerable barrier to its use.

Linezolid has in vitro data and limited outcome data in NTM infections, but tolerability is a concern. An evaluation of 102 patients with NTM infections (majority were pulmonary infections, 44% MABC and 33% MAC) noted that 46 patients experienced adverse effects (e.g., peripheral neuropathy, thrombocytopenia, anemia) at a median of 20 weeks, with 90% of the patients discontinuing therapy due to the adverse effects.50 Tedizolid appears to have better in vitro activity against NTM and may offer a more favorable adverse effect profile, though this is yet to be demonstrated in clinical trials.51

Clofazimine has in vitro activity against both MAC and MABC. A 2003 study evaluated a regimen of clofazimine, ethambutol, and a macrolide (clarithromycin/azithromycin) in 30 patients with MAC pulmonary disease. Four patients discontinued treatment early due to clarithromycin intolerability, but all 26 patients who completed the regimen demonstrated sputum conversion.52 Data were also published that supported clofazimine use in refractory NTM pulmonary disease. A total of 41 (50%) of 82 patients converted sputum within 12 months of therapy.53 Further retrospective data support clofazimine use in MABC refractory lung disease.54

THE PHARMACIST'S ROLE IN NTM TREATMENT

Pharmacists can assist at multiple points in the care continuum of patients with NTM disease, both in traditional and progressive ways. Some of the well-established functions that pharmacists provide are easily extrapolated to NTM therapy (Table 5).

Table 5. Pharmacist Services in the Care of Non-tuberculous Mycobacterial Disease
Antimicrobial dosing and monitoring recommendations
Therapeutic drug monitoring when indicated
Drug interaction screening and management
Adverse effect and administration counseling
Adherence counseling
Dose titration instructions and/or tables
Drug procurement assistance as needed

Pharmacists can be resources for patients and offer advice on what to expect from extended antibiotic therapy; they can also help ensure that patients are aware of monitoring requirements, provide insight on common drug interactions, and assist with mitigation of some adverse effects (Table 4). For example, pharmacists can inform patients that the rifamycins will stain body fluids orange. Additionally, the pharmacist should offer a recommendation to contact lens wearers who will take a rifamycin to consider daily-wear contacts or glasses to avoid the appearance of eye discoloration that may occur with staining of extended wear contact lenses. Pharmacists can ensure that patients are aware of ways to monitor for ethambutol eye toxicity by advising monthly self-assessments for visual acuity (e.g., looking at a phone book or novel at the same distance once monthly to determine any changes from baseline) and color discrimination (e.g., performing a monthly ishihara test, which is available online and for mobile devices). Further, the pharmacist can offer spacing instructions for fluoroquinolones and divalent cation supplements, such as calcium and iron, to optimize medication absorption and counsel patients to take macrolides with food to minimize some of the gastrointestinal side effects. As drug therapy experts, pharmacists can and should provide tips and advice for drug administration that will help avoid common drug interactions and adverse effects of NTM medication therapy.

Pharmacists are well equipped to support the care team with therapeutic drug monitoring (TDM). Given the high risk of adverse effects of amikacin such as ototoxicity and nephrotoxicity, TDM is a standard of care for patients receiving amikacin. A calculated maximum concentration (Cmax) should be determined using 2-hour and 6-hour serum levels drawn after the end of the infusion. The goal Cmax target level is 35 to 45 mcg/mL (for the 15 mg/kg daily dose) or 65 to 80 mcg/mL (for the 25 mg/kg thrice-weekly dose) for patients with normal renal function. Target troughs are undetectable (Figure 1). If multiple post-dose levels are not feasible, a 1-hour post-infusion 'peak' may be obtained, with a goal level of 25 to 35 mcg/mL for a 15-mg/kg dose for patients with normal renal function. Once the Cmax goal is reached with a given dosing regimen, trough levels and serum creatinine should be monitored at least once weekly. Baseline and periodic audiograms should also be obtained.

Figure 1. Therapeutic Drug Monitoring of Amikacin

Table 6 includes the optimal timing and levels for common doses of oral agents used in NTM therapy. TDM of oral agents in the treatment of NTM is less well established, but, given the extended duration of antimicrobial treatment, consideration should be given to optimizing drug exposure and minimizing toxicity through serum level assessment. Charles Peloquin, PharmD, an expert in mycobacterial pharmacotherapy, provides a detailed review of TDM in the setting of NTM disease that can serve as a useful resource for pharmacists.55 While TDM of oral antimicrobials is not standard of care for all patients, it should be considered in the following circumstances: concern for adequate drug absorption (e.g., surgical history of Roux-en-Y, graft-versus-host disease of the gut, severe gastrointestinal disease such as Crohn's Disease), drug interactions in which increased or decreased antimicrobial exposure is expected, renal/hepatic dysfunction, or lack of clinical/microbiologic response to therapy.

Table 6. Optimal Timing and Target Levels of Monitoring for Drugs Used in the Treatment of Non-tuberculous Mycobacterial Disease55
Antimicrobial agent and dose Timing for Cmax
(hours)

(obtain at steady state*)
Target Cmax (mcg/mL)
Azithromycin 250 mg daily 2-3 0.2-0.7
Clarithromycin 500 mg twice daily 2-3 2-7
Clofazimine 100 mg daily 2 2-7
Ethambutol 25 mg/kg daily 2-3 2-6
Isoniazid 300 mg daily 1-2 3-6
Levofloxacin 750 mg daily 1-2 8-13
Linezolid 600 mg daily 2 12-26
Moxifloxacin 400 mg daily 2 3-5
Rifampin 600 mg daily 2 8-24
Rifabutin 300 mg daily 3 0.45-0.9
*Steady state occurring at > 1-2 weeks according to individual drug half-life. May obtain additional 6-hour post-dose levels to determine peak plasma concentrations more accurately and to determine if there is delayed absorption verses malabsorption.

Pharmacists can also assist the care team with obtaining medications. Bedaquiline and clofazimine require additional steps for procurement for patient use. Bedaquiline is approved by the U.S. Food and Drug Administration (FDA) for TB treatment but not for NTM treatment.49 Given the high cost of the drug, insurance prior authorization can be a barrier to acquisition for treatment of NTM disease, but pharmacists can help delineate the indications for use and assist with prior authorization for the care team when necessary. Clofazimine is FDA approved for leprosy, but it is not available through traditional U.S. pharmaceutical distribution. Clofazimine use requires institutional review board (IRB) approval and submission of a drug application through the FDA. The drug application component can be accomplished by submission of a single-patient investigational new drug application directly to the FDA or individual patients can be added to an intermediate-sized investigational new drug application through the drug manufacturer's expanded access program.56 Pharmacists can be engaged in the IRB approval process and submission of the drug applications as key participants in the care team.

Exciting roles for pharmacists to become more involved in different aspects of care for patients with NTM disease are developing and expanding, including research and publication (particularly in the area of TDM), expansion of medication therapy management services for NTM patients, monitoring and dosing of NTM therapies and/or culture results in outpatient parenteral antimicrobial therapy clinics staffed by pharmacists, outpatient antimicrobial stewardship efforts, and the development of monographs, evidence-based reviews, and formulary recommendations. Pharmacists perform many of these roles already and their involvement will continue to improve the overall care of patients with NTM disease.

The monograph was reviewed on October 1, 2018 and January 15, 2019 and no updates are needed.

REFERENCES

  1. Chan ED, Iseman MD. Underlying host risk factors for nontuberculous mycobacterial lung disease. Semin Respir Crit Care Med. 2013;34(1):110-23.
  2. Jarlier V, Nikaido H. Mycobacterial cell wall: structure and role in natural resistance to antibiotics. FEMS Microbiol Lett. 1994;123(1-2):11-8.
  3. Falkinham JO 3rd. Growth in catheter biofilms and antibiotic resistance of Mycobacterium avium. J Med Microbiol. 2007;56(Pt 2):250-4.
  4. van Ingen J, Hoefsloot W, Dekhuijzen PN, et al. The changing pattern of clinical Mycobacterium avium isolation in the Netherlands. Int J Tuberc Lung Dis. 2010; 14(9):1176-80.
  5. Johnson MM, Odell JA. Nontuberculous mycobacterial pulmonary infections. J Thorac Dis. 2014;6(3): 210-20.
  6. Griffith DE, Aksamit T, Brown-Elliott BA, et al; ATS Mycobacterial Diseases Subcommittee; American Thoracic Society; Infectious Diseases Society of America. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med. 2007;175(4):367-416.
  7. Larsson LO, Polverino E, Hoefsloot W, et al. Pulmonary disease by non-tuberculous mycobacteria - clinical management, unmet needs and future perspectives. Expert Rev Respir Med. 2017;11(12):977-89.
  8. Kirschner RA Jr, Parker BC, Falkinham JO 3rd. Epidemiology of infection by nontuberculous mycobacteria. Mycobacterium avium, Mycobacterium intracellulare, and Mycobacterium scrofulaceum in acid, brown-water swamps of the southeastern United States and their association with environmental variables. Am Rev Respir Dis. 1992;145(2 Pt 1):271-5.
  9. McShane PJ, Glassroth J. Pulmonary disease due to nontuberculous mycobacteria: current state and new insights. Chest. 2015;148(6):1517-27.
  10. Al-Houqani MF, Jamieson M, Mehta M, et al. Aging, COPD, and other risk factors do not explain the increased prevalence of pulmonary Mycobacterium avium complex in Ontario. Chest. 2012;141(1):190-7.
  11. Iseman MD, Buschman DL, Ackerson LM. Pectus excavatum and scoliosis. Thoracic anomalies associated with pulmonary disease caused by Mycobacterium avium complex. Am Rev Respir Dis. 1991;144(4):914-6.
  12. Honda JR, Knight V, Chan ED. Pathogenesis and risk factors for nontuberculous mycobacterial lung disease. Clin Chest Med. 2015;36(1):1-11.
  13. Griffith DE, Aksamit TR. Bronchiectasis and nontuberculous mycobacterial disease. Clin Chest Med. 2012;33(2):283-95.
  14. Gadkowski LB, Stout JE. Cavitary pulmonary disease. Clin Microbiol Rev. 2008;21(2):305-33.
  15. Griffith DE, Brown BA, Girard WM, et al. Azithromycin activity against Mycobacterium avium complex lung disease in patients who were not infected with human immunodeficiency virus. Clin Infect Dis. 1996;23(5):983-9.
  16. Lam PK, Griffith DE, Aksamit TR, et al. Factors related to response to intermittent treatment of Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2006;173(11):1283-9.
  17. Jeong BH, Jeon K, Park HY, et al. Intermittent antibiotic therapy for nodular bronchiectatic Mycobacterium avium complex lung disease. Am J Respir Crit Care Med. 2015;191(1):96-103.
  18. Haworth CS, Banks J, Capstick T, et al. British Thoracic Society Guideline for the management of non-tuberculous mycobacterial pulmonary disease (NTM-PD). BMJ Open Respir Res. 2017;4(1):e000242.
  19. Eisenberg E, Barza M. Azithromycin and clarithromycin. Curr Clin Top Infect Dis. 1994;14:52-79.
  20. Kanatani MS, Guglielmo BJ. The new macrolides. Azithromycin and clarithromycin. West J Med. 1994;160(1):31-7.
  21. Wallace RJ Jr, Brown BA, Griffith DE, et al. Clarithromycin regimens for pulmonary Mycobacterium avium complex. The first 50 patients. Am J Respir Crit Care Med. 1996;153(6 Pt 1):1766-72.
  22. Wallace RJ Jr, Brown BA, Griffith DE, et al. Initial clarithromycin monotherapy for Mycobacterium avium-intracellulare complex lung disease. Am J Respir Crit Care Med. 1994;149(5):1335-41.
  23. Heifets LB, Iseman MD, Lindholm-Levy PJ. Combinations of rifampin or rifabutine plus ethambutol against Mycobacterium avium complex. Bactericidal synergistic, and bacteriostatic additive or synergistic effects. Am Rev Respir Dis. 1988;137(3):711-5.
  24. Gordin FM, Sullam PM, Shafran SD, et al. A randomized, placebo-controlled study of rifabutin added to a regimen of clarithromycin and ethambutol for treatment of disseminated infection with Mycobacterium avium complex. Clin Infect Dis. 1999;28(5):1080-5.
  25. Griffith DE, Brown BA, Girard WM, et al. Azithromycin-containing regimens for treatment of Mycobacterium avium complex lung disease. Clin Infect Dis. 2001;32(11):1547-53.
  26. Kunin CM. Antimicrobial activity of rifabutin. Clin Infect Dis. 1996;22 Suppl 1: S3-13; discussion S13-4.
  27. Griffith DE, Brown BA, Girard WM, Wallace RJ Jr. Adverse events associated with high-dose rifabutin in macrolide-containing regimens for the treatment of Mycobacterium avium complex lung disease. Clin Infect Dis. 1995;21(3):594-8.
  28. Griffith DE, Brown BA, Wallace RJ Jr. Varying dosages of rifabutin affect white blood cell and platelet counts in human immunodeficiency virus--negative patients who are receiving multidrug regimens for pulmonary Mycobacterium avium complex disease. Clin Infect Dis. 1996;23(6):1321-2.
  29. Kobashi Y, Matsushima T, Oka M. A double-blind randomized study of aminoglycoside infusion with combined therapy for pulmonary Mycobacterium avium complex disease. Respir Med. 2007;101(1):130-8.
  30. Peloquin CA, Berning SE, Nitta AT, et al. Aminoglycoside toxicity: daily versus thrice-weekly dosing for treatment of mycobacterial diseases. Clin Infect Dis. 2004;38(11):1538-44.
  31. Ahn CH, Lowell JR, Ahn SS, et al. Chemotherapy for pulmonary disease due to Mycobacterium kansasii: efficacies of some individual drugs. Rev Infect Dis. 1981;3(5):1028-34.
  32. Brown-Elliott BA, Nash KA, Wallace RJ Jr. Antimicrobial susceptibility testing, drug resistance mechanisms, and therapy of infections with nontuberculous mycobacteria. Clin Microbiol Rev. 2012;25(3):545-82.
  33. Griffith DE, Brown-Elliott BA, Wallace RJ Jr. Thrice-weekly clarithromycin-containing regimen for treatment of Mycobacterium kansasii lung disease: results of a preliminary study. Clin Infect Dis. 2003;37(9):1178-82.
  34. Jarand J, Levin A, Zhang L, et al. Clinical and microbiologic outcomes in patients receiving treatment for Mycobacterium abscessus pulmonary disease. Clin Infect Dis. 2011;52(5):565-71.
  35. Koh WJ, Jeon K, Lee NY, et al. Clinical significance of differentiation of Mycobacterium massiliense from Mycobacterium abscessus. Am J Respir Crit Care Med. 2011;183(3):405-10.
  36. Koh WJ, Jeong BH, Kim SY, et al. Mycobacterial characteristics and treatment outcomes in Mycobacterium abscessus lung disease. Clin Infect Dis. 2017;64(3):309-16.
  37. Stout JE, Floto RA. Treatment of Mycobacterium abscessus: all macrolides are equal, but perhaps some are more equal than others. Am J Respir Crit Care Med. 2012;186(9):822-3.
  38. Novosad S, Henkle E, Winthrop KL. The challenge of pulmonary nontuberculous mycobacterial infection. Curr Pulmonol Rep. 2015;4(3):152-61.
  39. Brown-Elliott BA, Vasireddy S, Vasireddy R, et al. Utility of sequencing the erm(41) gene in isolates of Mycobacterium abscessus subsp. abscessus with low and intermediate clarithromycin MICs. J Clin Microbiol. 2015;53(4):1211-5.
  40. Choi GE, Shin SJ, Won CJ, et al. Macrolide treatment for Mycobacterium abscessus and Mycobacterium massiliense infection and inducible resistance. Am J Respir Crit Care Med. 2012;186(9): 917-25.
  41. Koh WJ, Kim YH, Kwon OJ, et al. Surgical treatment of pulmonary diseases due to nontuberculous mycobacteria. J Korean Med Sci. 2008;23(3):397-401.
  42. Kang HK, Park HY, Kim D, et al. Treatment outcomes of adjuvant resectional surgery for nontuberculous mycobacterial lung disease. BMC Infect Dis. 2015;15:76.
  43. Meers P, Neville M, Malinin V, et al. Biofilm penetration, triggered release and in vivo activity of inhaled liposomal amikacin in chronic Pseudomonas aeruginosa lung infections. J Antimicrob Chemother. 2008;61(4):859-68.
  44. Rose SJ, Neville ME, Gupta R, Bermudez LE. Delivery of aerosolized liposomal amikacin as a novel approach for the treatment of nontuberculous mycobacteria in an experimental model of pulmonary infection. PLoS One. 2014;9(9):e108703.
  45. Olivier KN, Shaw PA, Glaser TS, et al. Inhaled amikacin for treatment of refractory pulmonary nontuberculous mycobacterial disease. Ann Am Thorac Soc. 2014;11(1):30-5.
  46. Yagi K, Ishii M, Namkoong H, et al. The efficacy, safety, and feasibility of inhaled amikacin for the treatment of difficult-to-treat non-tuberculous mycobacterial lung diseases. BMC Infect Dis. 2017;17(1): 558.
  47. Olivier KN, Griffith DE, Eagle G, et al. Randomized trial of liposomal amikacin for inhalation in nontuberculous mycobacterial lung disease. Am J Respir Crit Care Med. 2017;195(6):814-23.
  48. Koh WJ, Hong G, Kim SY, et al. Treatment of refractory Mycobacterium avium complex lung disease with a moxifloxacin-containing regimen. Antimicrob Agents Chemother. 2013;57(5):2281-5.
  49. Philley JV, Wallace RJ Jr, Benwill JL, et al. Preliminary results of bedaquiline as salvage therapy for patients with nontuberculous mycobacterial lung disease. Chest. 2015;148(2):499-506.
  50. Winthrop KL, Ku JH, Marras JT, et al. The tolerability of linezolid in the treatment of nontuberculous mycobacterial disease. Eur Respir J. 2015;45(4):1177-9.
  51. Brown-Elliott BA, Wallace RJ Jr. In vitro susceptibility testing of tedizolid against nontuberculous mycobacteria. J Clin Microbiol. 2017;55(6):1747-54.
  52. Field SK, Cowie RL. Treatment of Mycobacterium avium-intracellulare complex lung disease with a macrolide, ethambutol, and clofazimine. Chest. 2003;124(4):1482-6.
  53. Martiniano SL, Wagner BD, Levin A, et al. Safety and effectiveness of clofazimine for primary and refractory nontuberculous mycobacterial Infection. Chest. 2017;152(4):800-9.
  54. Yang B, Jhun BW, Moon SM, et al. Clofazimine-containing regimen for the treatment of Mycobacterium abscessus lung disease. Antimicrob Agents Chemother. 2017;61(6).
  55. Peloquin C. The role of therapeutic drug monitoring in mycobacterial infections. Microbiol Spectr. 2017;5(1).
  56. McGuffin SA, Pottinger PS, Harnisch JP. Clofazimine in nontuberculous mycobacterial infections: a growing niche. Open Forum Infectious Diseases. 2017;4(3).

Back Top