Skip to main content
Open Forum Infectious Diseases logoLink to Open Forum Infectious Diseases
. 2020 Oct 15;7(11):ofaa460. doi: 10.1093/ofid/ofaa460

Pharmacology, Dosing, and Side Effects of Rifabutin as a Possible Therapy for Antibiotic-Resistant Acinetobacter Infections

Matthew C Phillips 1, Noah Wald-Dickler 1,2, Katherine Loomis 1, Brian M Luna 3, Brad Spellberg 1,
PMCID: PMC7651144  PMID: 33204754

Abstract

Acinetobacter baumannii has among the highest rates of antibiotic resistance encountered in hospitals. New therapies are critically needed. We found that rifabutin has previously unrecognized hyperactivity against most strains of A. baumannii. Here we review the pharmacology and adverse effects of rifabutin to inform potential oral dosing strategies in patients with A. baumannii infections. Rifabutin demonstrates dose-dependent increases in blood levels up to 900 mg per day, but plateaus thereafter. Furthermore, rifabutin induces its own metabolism after prolonged dosing, lowering its blood levels. Pending future development of an intravenous formulation, a rifabutin oral dose of 900–1200 mg per day for 1 week is a rational choice for adjunctive therapy of A. baumannii infections. This dosage maximizes AUC24 to drive efficacy while simultaneously minimizing toxicity. Randomized controlled trials will be needed to definitively establish the safety and efficacy of rifabutin to treat A. baumannii infections.

Keywords: Acinetobacter, pharmacology, optimal dosing, rifabutin


Antibiotic resistance is one of the greatest challenges confronting 21st century clinical medicine. Acinetobacter baumannii is among the most antibiotic-resistant organisms encountered in hospitals and is one of the few bacteria strains that has acquired resistance to all known antibiotics [1, 2]. Extremely drug-resistant (XDR) A. baumannii, defined as resistant to all available antibiotics except for those that are inferior in efficacy or more toxic than alternatives, make up >50% of isolates from US intensive care units [3]. Such infections account for >23000 infections and >10000 deaths per year in the United States alone [3]. Globally, ~75000 XDR A. baumannii infections occur annually, resulting in 30000 deaths [3]. There is a critical need for new options to treat these infections.

We recently found that rifabutin possesses hitherto unknown hyperactivity against most strains of A. baumannii [4]. Rifabutin, which has been approved for use in humans for >30 years, has minimal inhibitory concentrations (MICs) against A. baumannii strains that are >50-fold below those of rifampin. However, this hyperactivity was only detectable in vitro in mammalian culture media, and not in rich broth media, which is traditionally used for MIC testing and new antibiotic screens. This lack of hyperactivity in rich media explains why this phenomenon remained undetected for so many decades. Furthermore, rifabutin’s hyperactivity in nutrient-depleted, mammalian cell culture media was predictive of hyperactivity in vivo as well. Rifabutin was far more effective at protecting mice from lethal bloodstream and lung infection caused by A. baumannii than rifampin, and at doses >10-fold below those of rifampin.

Rifabutin is a spiropiperidyl rifamycin analog with a larger volume of distribution and longer terminal half-life than rifampin. Food and Drug Administration (FDA)–approved in 1992 for the treatment of tuberculosis, rifabutin is typically used to treat mycobacterial infections in patients with advanced HIV, where complex drug–drug interactions often complicate therapy with rifampin. Compared with rifampin, rifabutin’s fewer drug interactions permit its safe co-administration with protease inhibitors; it was widely used in the second and third decades of the HIV pandemic, which led to numerous studies of its clinical and pharmacologic properties [5]. In more recent years, renewed interest has surfaced in rifabutin as salvage therapy in combination with other agents in the treatment of refractory, multidrug-resistant Helicobacter pylori gastritis.

To date, experience is limited in dosing rifabutin to treat critically ill patients with acute pyogenic bacterial infections. To help guide clinicians in treating XDR A. baumannii infections with the currently available oral formulations of rifabutin, we reviewed and amalgamated the published literature on rifabutin pharmacology, clinical efficacy, and toxicity.

METHODS

The PubMed database was searched using the following terms: “rifabutin” in combination with “clinical pharmacokinetics” OR “toxicity.” The terms were used in combination with publication type: “clinical trial” OR “review.” Smaller observational and pilot studies looking at pharmacokinetics were also included. We reviewed references in identified articles, including review articles. Exceedingly rare toxicities reported only in case reports were excluded. Our search revealed 48 clinical trials and studies and 4 review articles [6–9], from which we abstracted data on rifabutin pharmacokinetics and toxicities in the treatment of human infections.

There are no significant differences in rifabutin pharmacokinetics (PK) between healthy subjects and HIV-infected subjects [8]. However, patients with HIV may require treatment with medications that alter PK parameters, including protease inhibitor antiretrovirals, azole antifungals, and certain macrolide antibacterials. For the purpose of this review, the study arms that included drugs with significant effects on PK parameters were excluded, and all other relevant concomitant drugs were listed.

Patient Consent

This is a literature review; no human subjects research was conducted, so there were no factors necessitating consent.

Pharmacology Overview

At the time of this review, rifabutin is only available in oral capsules with no current intravenous (IV) formulation (Tables 1 and 2). Rifabutin is stable at a broad pH of 2–8 and is highly lipophilic with a pKa of 6.9 [6, 35]. It is rapidly absorbed following oral administration and reaches peak plasma concentrations 2–3 hours after ingestion [9, 23]. However, its overall bioavailability is low, at ~20% after a single oral dose [6, 8, 23]. Furthermore, studies show significant interperson variability of bioavailability with standard 300-mg daily oral dosing [23]. When taken with food, particularly high–fat content food, the rate of absorption is significantly prolonged, with a time to maximum plasma concentration (Tmax) of 5.4 hours (fed) vs 3.0 hours (fasting) and a lag time of 1.6 hours (fed) vs 0.4 hours (fasting) [8, 17].

Table 1.

 Pharmacokinetics of Rifabutin in Individuals Without HIV

Reference Study Population No. Therapy Duration Dose, mg, and Frequency Cmax, μg/mL AUC0-24, μg-h/mL Half-life, h
Allen 1998 [10] Healthy volunteers 11 14 d
24 d
150 QD 0.16 ± 0.03
0.21 ± 0.04
1.61 ± 0.43
1.94 ± 0.48
N/A
Benedetti 1990 [11] Healthy volunteers 7 1 d
10 d
450 QD
450 QD
0.62
0.61
9.29
5.80
45 ± 6
58 ± 7
Ford 2008 [12] Healthy volunteers 15 13 d 300 QD 0.31
(0.27–0.37)
6.11
(5.33–7.01)a
N/A
Ghannad 2019 [13] Healthy volunteers 14 10 d 300 QD 0.54 4.22 N/A
Hamzeh 2003 [14] Healthy volunteers 17 14 d 300 QD 0.40 ± 0.14 3.37 ± 0.86 N/A
Kraft 2004 [15] Healthy volunteers 10
14
10 d
10 d
300 QD
300 QD
0.29
0.29
2.56
2.87
N/A
La Porte 2009 [16] Healthy volunteers 20 Single dose 150 once 0.16
(0.06–0.44)
2.22
(0.90–6.03)
41.3
(11.7–84.8)
Narang 1992 [17] Healthy volunteers 15 Single dose 150 once, fasted (solution)
150 once, with food (capsule)
150 once, fasted (capsule)
0.24
0.16
0.19
2.71b  
2.41b  
2.27b
N/A
Polk 2001 [18] Healthy male volunteers 11 14 d 300 QD 0.38
(0.30–0.48)
3.39
(2.84–4.03)
N/A
Sekar 2010 [19] Healthy volunteers 15 12 d 300 QD 0.57 ± 0.13 4.66 ± 0.97 N/A
van Ingen 2012 [20] Pulmonary MAC 51 N/A 4.80 ± 1.46 mg/kg 0.52 ± 0.29 2.28 ± 1.31c N/A
Zhang 2011 [21] Healthy volunteers 11
9
20 d
20 d
150 QD
150 QD
0.19 ± 0.05
0.18 ± 0.06
1.80 ± 0.40
1.80 ± 0.40
N/A
Zhang 2011 [22] Healthy volunteers 14 10 d 150 QD 0.19 (30) 1.85 (32) N/A

Abbreviations: AUC, area under the curve; Cmax, maximal concentration; QD, once daily; N/A, not reported in study.

aAUC0-48, area under drug-concentration time curve from time of dosing to 48 hours after dosing.

bAUC0-168, area under drug-concentration time curve from time of dosing to 168 hours after dosing.

cAUC0-6/7, area under drug-concentration time curve from time of dosing to 6 or 7 hours after dosing.

Table 2.

 Pharmacokinetics of Rifabutin in HIV-Infected Individuals

Study Number of Subjects Relevant Concurrent Medications Duration of Therapy Rifabutin Daily Dose, mg Cmax,
Mean ± SD or Median (Range), µg/mL
AUC24,
Mean ± SD or Median (Range), µg-h/mL
Half-life, h
Skinner 1989 [23] 5
6
5
5
None Single dose 300 QD
600 QD
900 QD
1200 QD
0.37
0.52
0.90
0.90
5.4 ± 0.6
10.2 ± 0.6
11.7 ± 3.6
13.2 ± 1.2
38 ± 12
38 ± 13
32 ± 8
38 ± 19
Skinner 1989 [23] 5
6
5
5
None 28 d 300 QD
600 QD
900 QD
1200 QD
0.37
0.52
0.90
0.90
3.9 ± 0.6
3.0 ± 0.6
7.2 ± 2.7
9.6 ± 1.2
38 ± 12
38 ± 13
32 ± 8
38 ± 19
Weiser 1989 [24] 4 None 4–66 wk 2250 QD 0.574 N/A N/A
Gatti 1999 [25] 10: wasting syndrome
10: no wasting syndrome
AZT (n = 12)
DDI (n = 9)
TMP-SMX (n = 12)
Single dose
Single dose
300 once
300 once
0.34 ± 0.14
0.55 ± 0.16
7.36a  
7.95a
31.446.0
Benetor 2007 [26] 7 with TB Isoniazid 21 d 300 twice weekly 0.43
(0.34–0.56)
4.10
(3.18–5.27)
N/A
Boulanger 2009 [27] 10 with TB Isoniazid Pyrazinamide Ethambutol 14–28 d 300 3 times weekly 0.30
(0.15–0.55)
2.71
(1.39–3.98)
N/A
Nguyen 2014 [28] 25 with TB Isoniazid Pyrazinamide Ethambutol 14 d
300 QD 0.79
(0.34–1.11)
5.64
(2.72–8.88)
N/A
Moyel 2002 [29] 14 None 14 d 300 QD 0.31 (32.8) 3.01 (28.0) N/A
Trapnell 1996 [30] 12 Zidovudine
Fluconazole
14 d: AZT + fluc
14 d:
AZT only
300 QD 5.4 ± 2.4
3.03 ± 1.12
N/A
Naiker 2014 [31] 15 with TB Isoniazid Pyrazinamide Ethambutol 28 d 300 QD 0.29
(0.25–0.38)
3.05
(2.65–3.43)
N/A
Ramachandran 2013 [32] 16 with TB Isoniazid Pyrazinamide Ethambutol Atazanavir Ritonavir Minimum 14 d 150 3 times weekly 0.33 (0.19–0.48) 4.61 (2.07–5.34) N/A
Ramachandran 2019 [33] 45 with TB Isoniazid Pyrazinamide Ethambutol Atazanavir Ritonavir 300 3 times weekly (n = 36)
150 QD (n = 9)
0.75
(0.52–1.23)
0.58
(0.26–0.81)
9.27
(7.00–12.34)
6.32
(3.91–10.33)
N/A
Siegal 1990 [34] 16 None 4–66 wk 300 QD to
2400 QD (stepwise increase)
0.1
0.6
N/A

Abbreviations: AUC, area under the curve; AZT, zidovudine; fluc, fluconazole; Cmax, maximal concentration; DDI, didanosine; QD, once daily; TB, tuberculosis; TMP/SMX, trimethoprim/sulfamethoxazole.

aAUC0-96, area under drug-concentration time curve from time of dosing to 96 hours after dosing.

Although food appears to affect the rate of absorption, it does not impact the extent or fraction of rifabutin absorbed, and the area under the curve for drug concentrations in blood over 24 hours (AUC24) and maximum plasma concentration (Cmax) remains unaffected by food. As a result, rifabutin can be taken with or without food [8].

Rifabutin has a long elimination half-life, with a mean ranging from 31.4 to 58 hours, allowing for ease of once-daily dosing [23]. It is metabolized in the liver, making it safe for use in patients with renal failure without necessitating dose reduction. Owing to its highly lipophilic nature, rifabutin has a very large volume of distribution (8–9 L/kg) and a relatively low Cmax [6, 8]. Its Cmax is dose-dependent, ranging from 0.16 µg/mL after administration of a single 150-mg capsule to 0.9 µg/mL after a single 900-mg administration. Doses >900 mg/d were not associated with further increases in Cmax, ranging from 0.4 µg/mL to 1 µg/mL [23, 24]. Although its Cmax is low, the rifabutin MICs of most A. baumannii strains tested in nutrient-depleted media were <0.1 µg/mL [4], readily exceedable by standard rifabutin dosing.

Like its Cmax, rifabutin’s AUC24 increases in a dose-dependent, approximately proportionate manner up to 900 mg per day (Table 2). At 1200 mg, the AUC24 rises slightly and less than proportionately further. The drug’s AUC24 decreases with chronic use, with a 45% decrease observed in AUC after 21–28 days of daily oral administration [8, 17]. This decline in AUC24 was observed across all doses (300–1200 mg per day) studied [23]. This phenomenon is thought to be due to autoinduction of its own hepatic metabolism, as rifabutin is an inducer of cytochrome P450 [23].

The primary PK/pharmacodynamic (PK/PD) driver of rifampin-mediated antibacterial effect is the ratio of AUC24 to MIC (AUC24:MIC) [36]. Although less well characterized, rifabutin’s PK/PD driver is likely to also be the AUC24/MIC ratio, and indeed in a trial of 169 HIV-positive patients with tuberculous, lower rifabutin AUC24s were associated with a higher rate of treatment failure and development of rifamycin resistance [37]. Thus, chronic dosing may be disadvantageous to its antimicrobial PK/PD driver of AUC24:MIC ratio. High dosing over a short period of time to maximize the AUC/MIC ratio, thus decreasing auto-induction of metabolism that would reduce AUC, may be preferable clinically for acute A. baumannii infections.

Tissue Penetration

The lipophilicity of rifabutin allows it to readily cross cell membranes, resulting in a high degree of tissue and intracellular penetration, including in leukocytes, with an intracellular/plasma concentration ratio of 9 in neutrophils and 15 in monocytes [9, 35, 38]. Rifabutin maintains high tissue-to-plasma drug concentration ratios, highlighting its suitability for the treatment of nonbloodstream tissue bacterial infections.

Given its large volume of distribution, rifabutin has favorable tissue penetration into nearly all organ systems. In lung tissue taken from surgical patients, rifabutin reached concentrations up to 8.6 times that of serum [8]. While penetration of rifabutin into the epithelial lining fluid (ELF) has not been studied, other rifamycins have ELF:plasma ratios between 0.2 and 0.32 [39].

Concentrations of rifabutin in urine and bile have been reported to exceed 100 times that of serum, and levels from tissue in the gastrointestinal (GI) tract exceed that of serum as well [8]. Rifabutin also penetrates muscle tissue, albeit at lower concentrations than in serum [9]. Additionally, rifabutin crosses the blood–brain barrier, with 1 study finding cerebrospinal fluid (CSF) drug concentrations to be 50% of plasma after a 450-mg daily dose (mean CSF concentration, 0.047 µg/mL) [34]. Taken together, these tissue PK data indicate biologic plausibility for rifabutin in the treatment of a variety of life-threatening clinical infections, including pneumonia, meningitis, and hepatobiliary diseases, such as cholangitis and cholecystitis.

Rifabutin Clinical Toxicities

Historical therapeutic uses for rifamycins, including rifabutin, have primarily involved prolonged (eg, many weeks to months) therapy for chronic infections (Table 3). Such infections include mycobacterial infections and staphylococcal prosthetic endovascular device or orthopedic hardware infections. Furthermore, use of rifabutin in the treatment or prevention of mycobacterial diseases has historically involved treatment courses lasting many months to even years. Such prolonged exposure increases the likelihood of developing drug toxicity. However, the majority of published data suggest that many rifabutin-associated toxicities do not develop until very high doses are used and/or until patients are exposed to the drug for many weeks (Table 3). Although serious hematologic and ocular side effects have been reported after prolonged dosing, in many cases adverse events are reversible with discontinuation of therapy and/or supportive care.

Table 3.

 Toxicities of Rifabutin

Study Study Participants Receiving Rifabutin Incidence Dose, mg/d Other Antimicrobials Time to Toxicity Reversible?
Leukopenia
Torseth et al. 1989 [40] 15 AIDS patients 0.45
0.20
600–900
1200
- 28 d Yes
Nightingale et al. 1993 [41] 566 AIDS patients 0.02 300 -
Apseloff et al. 1996 [42] 30 healthy adults .78 300 AZM or CLR 10 d Yes
Chien et al. 2014 [43] 221 MTB patients 0.06 300 EMB, INH, and PZA
Gordin et al. 1999 [44] 102 HIV+/MAC+ 0.19 300 CLR and EMB
Benson et al. 2003 [45] 107 HIV+/MAC+ 0.23 450 CLR and EMB
Naiker et al. 2014 [31] 16 HIV+/MTB+ 0.44 150 EMB, INH, PZA, and LPV 28–122 d
Griffith et al. 1995 [46] 26 MAC patients 0.3 600 AZM/CLR, EMB, and STM Mean: 2.4 ± 2.2 mo Yes
Griffith et al. 1996 [47] 53 MAC patients 0.31
0.36
0.14
0.09
600
300
600 (MWF)
300 (MWF)
AZM/CLR 3.9 ± 2 mo
3.7 ± 2.6 mo
4 ± 1.7 mo
4.7 ± 3.5 mo
Yes
Siegal et al. 1990 [34] 16 AIDS patients 0.06 1200–1600 - Yes
Benson et al. 2000 [48] 780 AIDS patients 0.25
0.26
440 -
CLR
Mori et al. 2015 [49] 29 H. pylori patients 0.07 300 PPI and AMX
Ghannad et al. 2019 [13] 14 healthy adults 0.29 300 MVC
Uveitis
Siegal et al. 1990 [34] 16 AIDS patients 0.06
0.06
1200
1800
- 16 wk
14 wk
Yes
Shafran et al. 1994 [50] 59 AIDS patients 0.39 600 FCZ and CLR Mean 65 d
Chien et al. 2014 [43] 221 MTB patients 0.005 300 EMB, INH, and PZA
Havlir et al. 1996 [51] 460 AIDS patients 0.01 300 AZM
May et al. 1997 [52] 72 HIV+/MAC+ .07 450 CLR and EMB
Gordin et al. 1999 [44] 102 HIV+/MAC+ 0.04 300 CLR and EMB
Benson et al. 2003 [45] 107 HIV+/MAC+ 0.07 450 CLR and EMB
Cohn et al. 1999 [53] 42 HIV+/MAC+ 0.02 300 CLR and EMB 8 mo
Naiker et al. 2014 [31] 16 HIV+/MTB+ 0.04 150 EMB, INH, PZA, and LPV 1 mo
Griffith et al. 1995 [46] 26 MAC patients 0.08 600 AZM/CLR, EMB, and STM Mean: 2.4 +/- 2.2 mo Yes
Benson et al. 2000 [48] 780 AIDS patients 0.02
0.09
440 -
CLR
18 wk
Influenza-like illness
Torseth et al. 1989 [40] 15 AIDS patients 0.53 300–1200 - 24 h Yes
Chien et al. 2014 [43] 221 MTB patients 0.03 300 EMB, INH, and PZA
Gastrointestinal symptoms (abdominal pain, nausea, vomiting, diarrhea)
Nightingale et al. 1993 [41] 566 AIDS patients 0.03 300 -
Weiser et al. 1989 [24] 16 AIDS patients 0.06 600 -
Chien et al. 2014 [43] 221 MTB patients 0.04 300 EMB, INH, and PZA
Havlir et al. 1996 [51] 460 AIDS patients 0.01 300 AZM
May et al. 1997 [52] 72 HIV+/MAC+ 0.07 450 CLR and EMB
Benson et al. 2003 [45] 107 HIV+/MAC+ 0.29 450 CLR and EMB
Griffith et al. 1995 [46] 26 MAC patients 0.42 600 AZM/CLR, EMB, and STM Mean: 2.4 +/- 2.2 mo
Benson et al. 2000 [48] 780 AIDS patients 0.07
0.05
440 -
CLR
Havlir et al. 1996 [51] 460 AIDS patients 0.27 300 AZM
Fiorini et al. 2018 [54] 256 H. pylori patients 0.12 150 PPI and AMX
Mori et al. 2015 [49] 29 H. pylori patients 0.17 300 PPI and AMX
Sung et al. 2017 [55] 11 H. pylori patients 0.01 300 PPI and AMX
Ribaldone et al. 2019 [56] 302 H. pylori patients 0.03 150 PPI and AMX
Arthralgias
Weiser et al. 1989 [24] 16 AIDS patients 0.9 >1000 - 8 wk
Chien et al. 2014 [43] 221 MTB patients 0.02 300 EMB, INH, and PZA
Griffith et al. 1995 [46] 26 MAC patients 0.19 600 AZM/CLR, EMB, and STM Mean: 2.4 +/- 2.2 mo Yes
Siegal et al. 1990 [34] 16 AIDS patients 0.9 >1000 - 16–66 wk Yes
Havlir et al. 1996 [51] 460 AIDS patients 0.01 300 AZM
Ghannad et al. 2019 [13] 14 healthy adults 0.14 300 MVC
Dermatologic events (rash, flushing, hyperpigmentation, etc.)
Nightingale et al. 1993 [41] 566 AIDS patients 0.04 300 -
Chien et al. 2014 [43] 221 MTB patients 0.1 300 EMB, INH, and PZA
Gordin et al. 1999 [44] 102 HIV+/MAC+ 0.16 300 CLR and EMB
Griffith et al. 1995 [46] 26 MAC patients 0.15 600 AZM/CLR, EMB, and STM Mean: 2.4 +/- 2.2 mo
Havlir et al. 1996 [51] 460 AIDS patients 0.02 300 AZM
Mori et al. 2015 [49] 29 H. pylori patients 0.1 300 PPI and AMX
Elevated liver function tests
Chien et al. 2014 [43] 221 MTB patients 0.02 300 EMB, INH, and PZA
May et al. 1997 [52] 72 HIV+/MAC+ 0.03 450 CLR and EMB
Benson et al. 2003 [45] 107 HIV+/MAC+ 0.3 450 CLR and EMB
Naiker et al. 2014 [31] 16 HIV+/MTB+ 0.13 150 EMB, INH, PZA, and LPV 96–106 d
Griffith et al. 1995 [46] 26 MAC patients 0.12 600 AZM/CLR, EMB, and STM Mean: 2.4 +/- 2.2 mo
Siegal et al. 1990 [34] 16 AIDS patients 0.83 >1800 - 12 wk Yes
Benson et al. 2000 [48] 780 AIDS patients 0.06
0.08
440 -
CLR
Mori et al. 2015 [49] 29 H. pylori patients 0.17 300 PPI and AMX
Thrombocytopenia
Benson et al. 2003 [45] 107 HIV+/MAC+ 0.09 450 CLR and EMB
Griffith et al. 1995 [46] 26 MAC patients 0.15 600 AZM/CLR, EMB, and STM Mean: 2.4 +/- 2.2 mo Yes
Siegal et al. 1990 [34] 16 AIDS patients 0.06 1200–1600 - Yes
Benson et al. 2000 [48] 780 AIDS patients 0.05
0.05
440 -
CLR
Anemia
Benson et al. 2003 [45] 107 HIV+/MAC+ 0.23 450 CLR and EMB
Siegal et al. 1990 [34] 16 AIDS patients 0.06 1200–1600 - Yes
Benson et al. 2000 [48] 780 AIDS patients 0.02
0.02
440 -
CLR
Taste disturbances
Fiorini et al. 2018 [54] 256 H. pylori patients 0.02 150 PPI and AMX
Mori et al. 2015 [49] 29 H. pylori patients 0.03 300 PPI and AMX
Sung et al. 2017 [55] 11 H. pylori patients 0.01 300 PPI and AMX
Headache
Fiorini et al. 2018 [54] 256 H. pylori patients 0.02 150 PPI and AMX
Mori et al. 2015 [49] 29 H. pylori patients 0.28 300 PPI and AMX
Ribaldone et al. 2019 [56] 302 H. pylori patients <0.01 150 PPI and AMX
Ghannad et al. 2019 [13] 14 healthy adults 0.29 300 MVC
Myalgias
Fiorini et al. 2018 [54] 256 H. pylori patients <0.01 150 PPI and AMX
Ghannad et al. 2019 [13] 14 healthy adults 0.07 300 MVC
Fever
Mori et al. 2015 [49] 29 H. pylori patients 0.28 300 PPI and AMX
Ghannad et al. 2019 [13] 14 healthy adults 0.14 300 MVC

Toxicities included in the table were those reported in 2 or more manuscripts.

Abbreviations: AMX, amoxicillin; AZM, azithromycin; CLR, clarithromycin; EMB, ethambutol; FCZ, fluconazole; INH, isoniazid; LPV, lopinavir; MAC, M. avium complex; MTB, M. tuberculous; MVC, maraviroc; MWF, Monday Wednesday Friday dosing; PPI, proton pump inhibitor; PZA, pyrazinamide; STM, streptomycin.

Most published literature on rifabutin-associated toxicities comes from patients with HIV, for whom rifabutin was studied as prophylaxis or treatment of disseminated disease from Mycobacterium avium complex (MAC). Early studies of rifabutin sought to replicate its in vitro antiretroviral effects in clinical trials of HIV patients [24, 40]. While ultimately unsuccessful at treating underlying HIV, these trials elucidated prototypic toxicities of rifabutin, such as leukopenia and arthralgias.

Side effects in those early trials of rifabutin monotherapy appeared after prolonged usage at dosages significantly higher than those commonly used today. For example, in a small dose-finding trial of 16 patients with HIV, rifabutin doses of ≥1200 mg resulted in 90% of patients developing arthralgias after the eighth week of the trial [24]. In a separate dose escalation trial, 90% of patients with HIV receiving doses in excess of 1000 mg per day for over 16 weeks also developed arthralgias, which reversed with discontinuation of the drug. Two patients developed reversable uveitis, both after receiving dosages in excess of 1200 mg for 14–16 weeks. A mild, transient leukopenia was seen in 1 of the 16 patients in this dose escalation trial, and a reversable increase in transaminases occurred in 31% of patients after 12 weeks of receiving >1800 mg per day of rifabutin [34]. Leukopenia was also documented in a separate antiretroviral trial in 7% of patients receiving 600 mg of rifabutin, twice per day for 28 days [40].

A multitude of studies investigating rifabutin for MAC prophylaxis and treatment have been conducted in patients with HIV. These trials were conducted with much longer treatment durations, typically 6 months, but with some as long as 2 years. Trials typically utilized dosages of rifabutin between 150 mg and 600 mg per day, with most utilizing the now standard dosage of 300 mg per day. As part of a regimen for prophylaxis against MAC in patients with HIV, rifabutin was used either with or without a macrolide. In trials investigating the use of rifabutin for treatment of pulmonary MAC, rifabutin is typically used as part of a multidrug regimen with ethambutol, macrolides, or aminoglycosides. Side effects in these larger trials most commonly report gastrointestinal intolerance as the most common side effect, occurring in 1 trial in up to 33% of patients receiving rifabutin and azithromycin [51]. Neutropenia was typically seen in dose-dependent frequencies, with up to 31% of patients receiving 600-mg daily doses compared with only 18%–23% of patients receiving dosages between 300 and 450 mg [44–47]. However, it was not seen during the first 1–2 weeks of therapy. Multiple trials also reported arthralgias and elevated transaminases.

As mentioned, uveitis is a well-described, unique, and potentially sight-threatening toxicity associated with rifabutin that is worth special attention. Most reports of rifabutin-associated uveitis occurred only with very high doses over 1200 mg, after many weeks of therapy, and were reversible in most cases with drug discontinuation or topical therapies [34, 50, 57]. In a large trial comparing rifabutin, azithromycin, or the combination of both for MAC prophylaxis, only 5 out of 460 (1.1%) rifabutin recipients developed uveitis [51]. Uveitis was considerably more common when rifabutin was used at a dosage of 450 mg per day. In a trial following nearly 1200 patients over the course of 2 years, 42 patients developed uveitis, primarily those receiving both clarithromycin and rifabutin, between which a significant drug–drug interaction exists. The development of uveitis led to dose reduction of rifabutin from 450 mg to 300 mg. Before reduction, the uveitis event rate was 5.68 events per 100 patient-years; after the dose reduction, the event rate dropped to 1.40 per 100 patient-years [48]. Similarly, a case–control study of 229 HIV patients with MAC bacteremia treated with 600 mg of rifabutin daily (along with ethambutol and clarithromycin) reported a cumulative risk of uveitis of 43% at 6 months. Following a dose reduction to 300 mg daily, the 6-month cumulative risk of uveitis was dramatically reduced to 13% [58]. As for neutropenia and arthralgias, uveitis has never been described to occur during the first 7–14 days of rifabutin therapy.

More recently, rifabutin has been used in shorter-course treatment regimens as an adjunct for drug-resistant Helicobacter pylori infection. These trials primarily used rifabutin in conjunction with a proton pump inhibitor and amoxicillin for between 5 and 14 days of therapy. Side effects in these trials were minimal and consisted primarily of GI toxicities like diarrhea and abdominal pain [49, 54–56]. Leukopenia was relatively rare, only documented in 1 trial [49], and no cases of uveitis have been documented in trials conducted using rifabutin for the treatment of H. pylori.

DISCUSSION

We recently found that rifabutin has hyperactivity against A. baumannii when tested in nutrient-depleted, mammalian cell culture media and that this effect accurately predicts in vivo efficacy in mice [4]. Randomized controlled trials are needed to establish clinical benefit against A. baumannii. However, adjunctive rifampin therapy was shown to significantly reduce bacterial burden when added to standard therapy in a randomized controlled trial of patients with A. baumannii pneumonia, suggesting that adjunctive rifamycin therapy may have some therapeutic benefit [59]. Unfortunately, mortality was not improved in this trial, and hence adjunctive rifampin therapy fell out of favor. But rifabutin is >50-fold more potent than rifampin in vitro when tested in nutrient-depleted media. Furthermore, in vivo rifabutin was far more effective at preventing death during lethal bacteremia and pneumonia in mice, including at doses far below those where rifampin demonstrated some efficacy. Thus, rifabutin has promise to improve outcomes relative to adjunctive rifampin.

Rifabutin is approved for use in humans, is inexpensive and generic, is widely available, has excellent tissue penetration, and has a favorable side effect profile. It is safe to use in patients with renal injury, which is present in up to 57% of patients in the critical care setting [60]. Rifabutin reaches therapeutic concentration in tissues relevant to sites of infection caused by A. baumannii, including pneumonia, bacteremia, urinary tract infections, and soft tissue infections. Thus, experts may be tempted to add rifabutin to the best-available therapy for these infections given their high mortality and limited alternative therapeutic options [2]. We emphasize the importance of conducting randomized controlled trials to affirm or refute the therapeutic benefit of rifabutin in this context and that having an intravenous formulation available would likely result in superior drug levels. However, until such a formulation and clinical trial results are available, clinicians seeking to use adjunctive rifabutin therapy when confronted by XDR A. baumannii infections will need guidance on dosing.

Based on our review of available pharmacokinetic and clinical toxicity data, a high-dose, short-course regimen could achieve the desired balance between maximizing the drug’s AUC:MIC ratio to enhance microbial killing, avoiding auto-induction, which diminishes rifabutin blood levels, and minimizing toxicity. Multiple randomized controlled trials have found that 1 week of therapy is adequate to treat nosocomial pneumonia, complicated urinary tract infections, complicated intra-abdominal infections, and severe skin/soft tissue infections [61, 62]. Severe side effects from rifabutin, such as uveitis, arthralgias/arthritis, and leukopenia, have not been described to occur within the first week of therapy, even when very high doses (up to 2400 mg per day) were administered. Thus, dosing for no more than 1 week should enable minimization of side effects, minimizing selection for resistance and still providing adequate potential therapeutic benefit.

The primary question, which dose to administer for that week of therapy, is difficult to define in the absence of controlled clinical trials. Pharmacological studies have found dose-dependent increases in Cmax and AUC24 with dose escalation from 150 to 900 mg once per day. With the caveat that only limited published data sets are available, Cmax did not further increase beyond 900 mg, but AUC24 slightly increased at a dose of 1200 mg per day. As such, 900–1200 mg once per day for no more than 7 days may be a rational dose to balance the desire to maximize Cmax and AUC24 as potential efficacy drivers with the need to minimize toxicity. We emphasize that rifabutin ELF levels are not well established, and this should be a focus of future clinical investigations to help inform pivotal clinical trial conduct. Nevertheless, until such data are available, the plasma:ELF ratios of other rifamycins (0.2–0.32) underscore the need to maximize plasma AUC levels to attempt to achieve adequate ELF levels.

Based on available data, adverse events from such a regimen might reasonably be anticipated to include mild gastrointestinal symptoms and slight increases in hepatic transaminases. Caution would be advised for close laboratory and clinical monitoring for neutropenia and uveitis, particularly in those receiving concurrent CYP3A substrates and in those with underlying liver disease. Many patients studied in the AIDS era with disseminated MAC infections were very seriously ill. However, it is conceivable that acutely ill patients on mechanical ventilators in the intensive care unit may have different propensities for adverse events, as rifabutin and its pharmacokinetics have not been studied in these patients. Thus, as with any off-label drug use, caution should be exercised, with monitoring of hepatic enzymes if adjunctive rifabutin therapy is attempted, particularly in critically ill patients. Furthermore, as an inducer of CYP3A, rifabutin may lower serum concentrations of other medications, including commonly used analgesics in ventilated patients such as fentanyl. Thus careful drug interaction checking should be conducted before using rifabutin in critically ill patients.

Given its relatively poor bioavailability, the development of an intravenous formulation of rifabutin would be ideal given that similar dosages given intravenously (with 100% bioavailability) would result in a 5-fold higher AUC as compared with oral administration. This increased AUC would directly improve the AUC:MIC-dependent killing capacity of rifabutin, thus increasing its efficacy. Despite this limitation, adding rifabutin as an adjunct to current antibiotic therapy in XDR A. baumannii infections presents promising potential in addressing the critical need for new therapies to treat infection from this life-threatening pathogen. Rifabutin has already been approved by the FDA, and our review suggests its ideal candidacy for phase III efficacy studies of its role as an adjunctive therapy for infections caused by A. baumannii, particularly those resistant to preferred antimicrobial regimens.

Acknowledgments

Financial support. This work was supported by National Institute of Allergy and Infectious Diseases (NIAID) grants R01AI139052 (to B.L.), R01AI130060 (to B.S.), and R01AI117211 (to B.S.) and by the Food and Drug Administration (BAA Contract HHSF223201710199C to B.L.).

Potential conflicts of interest. B.L. and B.S. are inventors on a patent for rifabutin therapy for A. baumannii infections and own equity in ExBaq, which has licensed the technology for development as an intravenous formulation. The University of Southern California owns intellectual property related to these development efforts. The other authors have no conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

Author contributions. B.S. was responsible for the conception of this manuscript. Literature review was conducted by M.P., N.W.D., K.L., and B.S. All authors contributed to the writing and editing of the manuscript. Financial support was provided by B.S. and B.L.

References

  • 1. Shlaes DM, Sahm D, Opiela C, Spellberg B. The FDA reboot of antibiotic development. Antimicrob Agents Chemother  2013; 57:4605–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Wong D, Nielsen TB, Bonomo RA, et al.  Clinical and pathophysiological overview of Acinetobacter infections: a century of challenges. Clin Microbiol Rev  2017; 30:409–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Spellberg B, Rex JH. The value of single-pathogen antibacterial agents. Nat Rev Drug Discov  2013; 12:963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Luna B, Trebosc V, Lee B, et al.  A nutrient-limited screen unmasks rifabutin hyperactivity for extensively drug-resistant Acinetobacter baumannii. Nat Microbiol  2020; 5:1134–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Hennig S, Svensson EM, Niebecker R, et al.  Population pharmacokinetic drug–drug interaction pooled analysis of existing data for rifabutin and HIV PIs. J Antimicrob Chemother  2016; 71:1330–1340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Gisbert JP, Calvet X. Review article: rifabutin in the treatment of refractory Helicobacter pylori infection. Aliment Pharmacol Ther  2012; 35:209–21. [DOI] [PubMed] [Google Scholar]
  • 7. Brogden RN, Fitton A. Rifabutin. A review of its antimicrobial activity, pharmacokinetic properties and therapeutic efficacy. Drugs  1994; 47:983–1009. [DOI] [PubMed] [Google Scholar]
  • 8. Blaschke TF, Skinner MH. The clinical pharmacokinetics of rifabutin. Clin Infect Dis  1996; 22(Suppl 1:S15–21; discussion S21–2. [PubMed] [Google Scholar]
  • 9. Crabol Y, Catherinot E, Veziris N, et al.  Rifabutin: where do we stand in 2016?  J Antimicrob Chemother  2016; 71:1759–71. [DOI] [PubMed] [Google Scholar]
  • 10. Cato A 3rd, Cavanaugh J, Shi H, Hsu A, Leonard J, Granneman R. The effect of multiple doses of ritonavir on the  pharmacokinetics of rifabutin. Clin Pharmacol Ther  1998; 63:414–21. [DOI] [PubMed] [Google Scholar]
  • 11. Benedetti MS, Efthymiopoulos C, Sassella D, Moro E, Repetto M. Autoinduction of rifabutin metabolism in man. Xenobiotica  1990; 20:1113–9. [DOI] [PubMed] [Google Scholar]
  • 12. Ford SL, Chen YC, Lou Y, et al.  Pharmacokinetic interaction between fosamprenavir-ritonavir and rifabutin in healthy subjects. Antimicrob Agents Chemother  2008; 52:534–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Ghannad M, Dennehy M, la Porte C, et al.  A drug interaction study investigating the effect of rifabutin on the pharmacokinetics of maraviroc in healthy subjects. PLoS One  2019; 14:e0223969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Hamzeh FM, Benson C, Gerber J, et al. ; AIDS Clinical Trials Group 365 Study Team Steady-state pharmacokinetic interaction of modified-dose indinavir and rifabutin. Clin Pharmacol Ther  2003; 73:159–69. [DOI] [PubMed] [Google Scholar]
  • 15. Kraft WK, McCrea JB, Winchell GA, et al.  Indinavir and rifabutin drug interactions in healthy volunteers. J Clin Pharmacol  2004; 44:305–13. [DOI] [PubMed] [Google Scholar]
  • 16. la Porte CJ, Sabo JP, Elgadi M, Cameron DW. Interaction studies of tipranavir-ritonavir with clarithromycin, fluconazole, and rifabutin in healthy volunteers. Antimicrob Agents Chemother  2009; 53:162–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Narang PK, Lewis RC, Bianchine JR. Rifabutin absorption in humans: relative bioavailability and food effect. Clin Pharmacol Ther  1992; 52:335–41. [DOI] [PubMed] [Google Scholar]
  • 18. Polk RE, Brophy DF, Israel DS, et al.  Pharmacokinetic Interaction between amprenavir and rifabutin or rifampin in healthy males. Antimicrob Agents Chemother  2001; 45:502–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Sekar V, Lavreys L, Van de Casteele T, et al.  Pharmacokinetics of darunavir/ritonavir and rifabutin coadministered in HIV-negative healthy volunteers. Antimicrob Agents Chemother  2010; 54:4440–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. van Ingen J, Egelund EF, Levin A, et al.  The pharmacokinetics and pharmacodynamics of pulmonary Mycobacterium avium complex disease treatment. Am J Respir Crit Care Med  2012; 186:559–65. [DOI] [PubMed] [Google Scholar]
  • 21. Zhang X, Fettner S, Zwanziger E, et al.  Pharmacokinetic interaction study of ritonavir-boosted saquinavir in combination with rifabutin in healthy subjects. Antimicrob Agents Chemother  2011; 55:680–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Zhang J, Zhu L, Stonier M, et al.  Determination of rifabutin dosing regimen when administered in combination with ritonavir-boosted atazanavir. J Antimicrob Chemother  2011; 66:2075–82. [DOI] [PubMed] [Google Scholar]
  • 23. Skinner MH, Hsieh M, Torseth J, et al.  Pharmacokinetics of rifabutin. Antimicrob Agents Chemother  1989; 33:1237–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Weiser B, Burger H, Eilbott DJ, et al.  Efficacy of rifabutin in the treatment of AIDS-related complex. AIDS  1989; 3:823–7. [DOI] [PubMed] [Google Scholar]
  • 25. Gatti G, Di Biagio A, De Pascalis CR, et al.  Pharmacokinetics of rifabutin in HIV-infected patients with or without wasting syndrome. Br J Clin Pharmacol  1999; 48:704–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Benator DA, Weiner MH, Burman WJ, et al. ; Tuberculosis Trials Consortium Clinical evaluation of the nelfinavir-rifabutin interaction in patients with tuberculosis and human immunodeficiency virus infection. Pharmacotherapy  2007; 27:793–800. [DOI] [PubMed] [Google Scholar]
  • 27. Boulanger C, Hollender E, Farrell K, et al.  Pharmacokinetic evaluation of rifabutin in combination with lopinavir-ritonavir in patients with HIV infection and active tuberculosis. Clin Infect Dis  2009; 49:1305–11. [DOI] [PubMed] [Google Scholar]
  • 28. Nguyen Thi Ngoc L, Thi Nguyet Thu N, Barrail-Tran A, et al.  Randomised pharmacokinetic trial of rifabutin with lopinavir/ritonavir-antiretroviral therapy in patients with HIV-associated tuberculosis in Vietnam. 2014; 9:e84866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Moyle GJ, Buss NE, Goggin T, et al.  Interaction between saquinavir soft-gel and rifabutin in patients infected with HIV. Br J Clin Pharmacol  2002; 54:178–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Trapnell CB, Narang PK, Li R, Lavelle JP. Increased plasma rifabutin levels with concomitant fluconazole therapy in HIV-infected patients. Ann Intern Med  1996; 124:573–6. [DOI] [PubMed] [Google Scholar]
  • 31. Naiker S, Connolly C, Wiesner L, et al.  Randomized pharmacokinetic evaluation of different rifabutin doses in African HIV- infected tuberculosis patients on lopinavir/ritonavir-based antiretroviral therapy. BMC Pharmacol Toxicol  2014; 15:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Ramachandran G, Bhavani PK, Hemanth Kumar AK, et al.  Pharmacokinetics of rifabutin during atazanavir/ritonavir co-administration in HIV-infected TB patients in India. Int J Tuberc Lung Dis  2013; 17:1564–8. [DOI] [PubMed] [Google Scholar]
  • 33. Ramachandran G, Hemanth Kumar AK, Kannan T, et al.  Pharmacokinetics of rifabutin during atazanavir/ritonavir co-administration in HIV-infected TB patients. Indian J Tuberc  2019; 66:129–33. [DOI] [PubMed] [Google Scholar]
  • 34. Siegal FP, Eilbott D, Burger H, et al.  Dose-limiting toxicity of rifabutin in AIDS-related complex: syndrome of arthralgia/arthritis. AIDS  1990; 4:433–41. [DOI] [PubMed] [Google Scholar]
  • 35. Kunin CM. Antimicrobial activity of rifabutin. Clin Infect Dis  1996; 22(Suppl 1:S3–13; discussion S13–4. [DOI] [PubMed] [Google Scholar]
  • 36. Gumbo T, Louie A, Deziel MR, et al.  Concentration-dependent Mycobacterium tuberculosis killing and prevention of resistance by rifampin. Antimicrob Agents Chemother  2007; 51:3781–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Weiner M, Benator D, Burman W, et al. ; Tuberculosis Trials Consortium Association between acquired rifamycin resistance and the pharmacokinetics of rifabutin and isoniazid among patients with HIV and tuberculosis. Clin Infect Dis  2005; 40:1481–91. [DOI] [PubMed] [Google Scholar]
  • 38. Kurashima A, Mori T, Tomono Y, et al.  A new anti-mycobacterial agent, rifabutin [in Japanese]. Kekkaku  2010; 85:743–56. [PubMed] [Google Scholar]
  • 39. Rodvold KA, Yoo L, George JM. Penetration of anti-infective agents into pulmonary epithelial lining fluid: focus on antifungal, antitubercular and miscellaneous anti-infective agents. Clin Pharmacokinet  2011; 50:689–704. [DOI] [PubMed] [Google Scholar]
  • 40. Torseth J, Bhatia G, Harkonen S, et al.  Evaluation of the antiviral effect of rifabutin in AIDS-related complex. J Infect Dis  1989; 159:1115–8. [DOI] [PubMed] [Google Scholar]
  • 41. Nightingale SD, Cameron DW, Gordin FM, et al.  Two controlled trials of rifabutin prophylaxis against Mycobacterium avium complex infection in AIDS. N Engl J Med  1993; 329:828–33. [DOI] [PubMed] [Google Scholar]
  • 42. Apseloff G, Fluids G, LaBoy-Goral L, et al.  Severe neutropenia caused by recommended prophylactic doses of rifabutin. Lancet  1996; 348:685. [DOI] [PubMed] [Google Scholar]
  • 43. Chien JY, Chien ST, Huang SY, Yu CJ. Safety of rifabutin replacing rifampicin in the treatment of tuberculosis: a single-centre retrospective cohort study. J Antimicrob Chemother  2014; 69:790–6. [DOI] [PubMed] [Google Scholar]
  • 44. 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; 28:1080–5. [DOI] [PubMed] [Google Scholar]
  • 45. Benson CA, Williams PL, Currier JS, et al. ; AIDS Clinical Trials Group 223 Protocol Team A prospective, randomized trial examining the efficacy and safety of clarithromycin in combination with ethambutol, rifabutin, or both for the treatment of disseminated Mycobacterium avium complex disease in persons with acquired immunodeficiency syndrome. Clin Infect Dis  2003; 37:1234–43. [DOI] [PubMed] [Google Scholar]
  • 46. 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; 21:594–8. [DOI] [PubMed] [Google Scholar]
  • 47. 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; 23:1321–2. [DOI] [PubMed] [Google Scholar]
  • 48. Benson CA, Williams PL, Cohn DL, et al.  Clarithromycin or rifabutin alone or in combination for primary prophylaxis of Mycobacterium avium complex disease in patients with AIDS: a randomized, double-blind, placebo-controlled trial. The AIDS Clinical Trials Group 196/Terry Beirn Community Programs for Clinical Research on AIDS 009 Protocol Team. J Infect Dis  2000; 181:1289–97. [DOI] [PubMed] [Google Scholar]
  • 49. Mori H, Suzuki H, Matsuzaki J, et al.  Rifabutin-based 10-day and 14-day triple therapy as a third-line and fourth-line regimen for Helicobacter pylori eradication: A pilot study. United European Gastroenterol J  2016; 4:380–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Shafran SD, Deschênes J, Miller M, et al.  Uveitis and pseudojaundice during a regimen of clarithromycin, rifabutin, and ethambutol. MAC Study Group of the Canadian HIV Trials Network. N Engl J Med  1994; 330:438–9. [DOI] [PubMed] [Google Scholar]
  • 51. Havlir DV, Dubé MP, Sattler FR, et al.  Prophylaxis against disseminated Mycobacterium avium complex with weekly azithromycin, daily rifabutin, or both. California Collaborative Treatment Group. N Engl J Med  1996; 335:392–8. [DOI] [PubMed] [Google Scholar]
  • 52. May T, Brel F, Beuscart C, et al.  Comparison of combination therapy regimens for treatment of human immunodeficiency virus-infected patients with disseminated bacteremia due to Mycobacterium avium. ANRS Trial 033 Curavium Group. Agence Nationale de Recherche sur le Sida. Clin Infect Dis  1997; 25:621–9. [DOI] [PubMed] [Google Scholar]
  • 53. Cohn DL, Fisher EJ, Peng GT, et al.  A prospective randomized trial of four three-drug regimens in the treatment of disseminated Mycobacterium avium complex disease in AIDS patients: excess mortality associated with high-dose clarithromycin. Terry Beirn Community Programs for Clinical Research on AIDS. Clin Infect Dis  1999; 29:125–33. [DOI] [PubMed] [Google Scholar]
  • 54. Fiorini G, Zullo A, Vakil N, et al.  Rifabutin triple therapy is effective in patients with multidrug-resistant strains of Helicobacter pylori. J Clin Gastroenterol  2018; 52:137–40. [DOI] [PubMed] [Google Scholar]
  • 55. Sung J, Kim N, Park YH, et al.  Rifabutin-based fourth and fifth-line rescue therapy in patients with for Helicobacter pylori eradication failure. Korean J Gastroenterol  2017; 69:109–18. [DOI] [PubMed] [Google Scholar]
  • 56. Ribaldone DG, Fagoonee S, Astegiano M, et al.  Rifabutin-based rescue therapy for Helicobacter pylori eradication: a long-term prospective study in a large cohort of difficult-to-treat patients. J Clin Med  2019; 8:199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Dautzenberg B, Truffot C, Mignon A, et al.  Rifabutin in combination with clofazimine, isoniazid and ethambutol in the treatment of AIDS patients with infections due to opportunist mycobacteria. Groupe d’Etude et de Traitement des Infections à Mycobacteries Résistantes. Tubercle  1991; 72:168–75. [DOI] [PubMed] [Google Scholar]
  • 58. Shafran SD, Singer J, Zarowny DP, et al.  Determinants of rifabutin-associated uveitis in patients treated with rifabutin, clarithromycin, and ethambutol for Mycobacterium avium complex bacteremia: a multivariate analysis. Canadian HIV Trials Network Protocol 010 Study Group. J Infect Dis  1998; 177:252–5. [DOI] [PubMed] [Google Scholar]
  • 59. Durante-Mangoni E, Signoriello G, Andini R, et al.  Colistin and rifampicin compared with colistin alone for the treatment of serious infections due to extensively drug-resistant Acinetobacter baumannii: a multicenter, randomized clinical trial. Clin Infect Dis  2013; 57:349–58. [DOI] [PubMed] [Google Scholar]
  • 60. Hoste EA, Bagshaw SM, Bellomo R, et al.  Epidemiology of acute kidney injury in critically ill patients: the multinational AKI-EPI study. Intensive Care Med  2015; 41:1411–23. [DOI] [PubMed] [Google Scholar]
  • 61. Wald-Dickler N, Spellberg B. Short-course antibiotic therapy-replacing constantine units with “shorter is better.”  Clin Infect Dis  2019; 69:1476–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Spellberg B. The new antibiotic mantra—“shorter is better.”  JAMA Intern Med  2016; 176:1254–5. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Open Forum Infectious Diseases are provided here courtesy of Oxford University Press

RESOURCES