Aerosol fluoroquinolone formulations for improved pharmacokinetics

Information

  • Patent Grant
  • 10722519
  • Patent Number
    10,722,519
  • Date Filed
    Monday, November 19, 2018
    5 years ago
  • Date Issued
    Tuesday, July 28, 2020
    3 years ago
Abstract
The present invention relates to the field of antimicrobial agents. In particular, the present invention relates to the use of aerosolized fluoroquinolones formulated with divalent or trivalent cations and having improved pulmonary availability for the treatment and management of bacterial infections of the lung and upper respiratory tract.
Description
FIELD OF THE INVENTION

The present invention relates to the field of antimicrobial agents. In particular, the present invention relates to the use of aerosolized fluoroquinolones formulated with divalent or trivalent cations with improved pulmonary availability for the treatment and management of bacterial infections of the lung and upper respiratory tract.


BACKGROUND

Gram-negative bacteria are intrinsically more resistant to antibiotics than gram-positive bacteria due to the presence of a second outer membrane, which provides an efficient barrier to both hydrophilic and hydrophobic compounds. Consequently, there are few classes of antibiotics available to treat Gram-negative infections. Indeed, only several representatives of beta-lactams, aminoglycosides and fluoroquinolones have in vitro antibacterial activity against Pseudomonas aeruginosa and have been shown to have clinical utility, not surprisingly, development of resistance to such antibiotics is well-documented.


Respiratory diseases afflict millions of people across the world leading to suffering, economic loss and premature death, including infections of acute, subacute and chronic duration of the nasal cavity or four sinuses (each which have left and right halves, the frontal, the maxillary the ethmoid and the sphenoid), or the larynx, trachea or lung (bronchi, bronchioles, alveoli).


Pulmonary infections caused by gram-negative bacteria represent a particular challenge. Causative agents are usually found in sputum, pulmonary epithelial lining fluid, alveolar macrophages and bronchial mucosa. Acute exacerbations of pulmonary infection, periodically observed in patients with cystic fibrosis, COPD, chronic bronchitis, bronchiectasis, acute and chronic pneumonias, and many other pulmonary infections. Prevention of these exacerbations as well as their treatment is often difficult especially when highly resistant pathogens such as Pseudomonas aeruginosa and Burkholderia cepacia complex are involved. For most treatment protocols, high doses are required to maintain effective concentrations at the site of infection. In the case of aminoglycosides, nephrotoxicity and ototoxicity are also directly related to prolonged elevations of serum antibiotic concentrations. In an attempt to achieve an optimal outcome for the patient, clinicians routinely use a combination of two or more antibiotics such as ceftazidime and tobramycin, which are administered at high doses for 2 weeks, with the aim of achieving antibiotic synergy (J. G. den Hollander, et al., “Synergism between tobramycin and ceftazidime against a resistant Pseudomonas aeruginosa strain, tested in an in vitro pharmacokinetic model” Antimicrob. Agents Chemother. (1997), 41, 95-100). For example, successful treatments require that ceftazidime be administered either every 8 hours or by continuous infusion to maximize the time that the serum concentration is above the minimum inhibitory concentration (M. Cazzola, et al., “Delivering antibacterials to the lungs: Considerations for optimizing outcomes” Am. J. Respir. Med. (2002), 1, 261-272).


Aerosol administration of antibiotics directly to the site of infection, ensuring high local concentrations coupled with low systemic exposure represent an attractive alternative for the treatment of pulmonary infections. Aerosolized tobramycin is used for treatment of pseudomonal bacterial infections in patients with cystic fibrosis. The rationale behind this technique is to administer the drug directly to the site of infection and thereby alleviate the need to produce high serum concentrations by the standard intravenous method. An advantage of aerosol administration is that many patients can self-administer the antibiotic, and this treatment method may negate the need for lengthy hospitalization (M. E. Hodson “Antibiotic treatment: Aerosol therapy”, Chest (1988), 94, 156S-160S; and M. S. Zach “Antibiotic aerosol treatment” Chest (1988), 94, 160S-162S). However, tobramycin is currently the only FDA-approved aerosol antibiotic in the United States. And while it continues to play an important role in the management of recurrent infections in cystic fibrosis patients, its clinical utility is inadvertently being diminished due to development of resistance. In addition, the impact of total high concentrations achieved after aerosol administration is being somewhat diminished due to high binding of tobramycin to the components of cystic fibrosis sputum. Thus, there is a need for improved aerosolized antibiotics.


SUMMARY

The present invention relates to the use of aerosolized fluoroquinolones formulated with divalent or trivalent cations having improved pulmonary availability for the treatment and management of bacterial infections of the lung and upper respiratory tract. Some methods include treating a pulmonary infection including administering to a subject in need thereof, an effective amount of an aerosol solution of levofloxacin or ofloxacin in combination with a divalent or trivalent cation with improved pulmonary availability and exposure to levofloxacin or ofloxacin.


Methods for treating a pulmonary infection are provided. Some such methods include administering to a human having a pulmonary infection an aerosol of a solution comprising levofloxacin or ofloxacin and a divalent or trivalent cation to achieve a maximum lung sputum concentration (Cmax) of at least 1200 mg/L and a lung sputum area under the curve (AUC) of at least 1500 h·mg/L. In more embodiments, methods for treating a chronic lung infection are provided. Some such methods can include administering to a subject having a chronic lung infection an aerosol of a solution comprising levofloxacin or ofloxacin and a divalent or trivalent cation. In more embodiments, pharmaceutical compositions are provided. Some such compositions can include an aqueous solution consisting essentially of from 80 mg/ml to 120 mg/ml levofloxacin or ofloxacin and from 160 mM to 220 mM of a divalent or trivalent cation, wherein the solution has a pH from 5 to 7 and an osmolality from 300 mOsmol/kg to 500 mOsmol/kg.


Some embodiments include methods for treating a pulmonary infection that include administering to a human having said pulmonary infection an aerosol of a solution that includes levofloxacin or ofloxacin and a divalent or trivalent cation to achieve a maximum lung sputum concentration (Cmax) of at least about 1200 mg/L and a lung sputum area under the curve (AUC) of at least about 1500 h·mg/L.


Some embodiments include methods of treating a chronic lung infection that include administering to a subject having a chronic lung infection an aerosol of a solution that includes levofloxacin or ofloxacin and a divalent or trivalent cation.


Some embodiments include pharmaceutical compositions that include an aqueous solution consisting essentially of from about 80 mg/ml to about 120 mg/ml levofloxacin or ofloxacin and from about 160 mM to about 240 mM of a divalent or trivalent cation, wherein the solution has a pH from about 5 to about 7 and an osmolality from about 300 mOsmol/kg to about 500 mOsmol/kg.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows a graph of plasma levofloxacin concentration in rats after intravenous administration, aerosol administration of levofloxacin (LVX) formulated in saline, or aerosol administration of levofloxacin formulated with MgCl2.



FIG. 2 shows a graph of recovery of levofloxacin from lung homogenate in rats after a single 10 mg/kg dose of aerosol levofloxacin formulated with saline, Ca+2, Mg+2 or Zn2+.



FIG. 3 shows a schematic of a pharmacokinetic model for deconvolution of serum levofloxacin concentrations following aerosol administration of a drug.



FIG. 4 shows a graph of plasma levofloxacin (LVX) concentrations after administration of 50 mg LVX in saline by aerosol or intravenous routes.



FIG. 5 shows a graph of the estimated amount of drug remaining in the lung compartment following aerosol administration of 50 mg respirable drug dose levofloxacin in saline, where amount of drug remaining is estimated using deconvolution of serum data.



FIG. 6 shows a graph of serum levofloxacin concentrations in normal healthy volunteers and cystic fibrosis patients after a single intravenous or aerosol dose of levofloxacin administered in saline. Doses are shown as estimated RDD; the 20 mg and 40 mg RDDs represent nebulizer loaded doses of 43.3 and 86.6 mg, respectively.



FIG. 7 shows a graph of deconvolution estimates of the average amount of levofloxacin remaining in the lung in 7 healthy volunteers and 9 CF patients following a single aerosol dose of 43.3 mg loaded into nebulizer (estimated respirable delivered dose (RDD) of 20 mg, and a 86.6 mg dose loaded into the nebulizer (estimated RDD of 40 mg) formulated in normal saline.



FIG. 8A shows a graph of the estimated levofloxacin concentrations in lung epithelial lining fluid following a single 20 mg respirable drug dose (43.3 mg of levofloxacin loaded into the nebulizer) formulated in normal saline in CF patients. FIG. 8B shows a graph of the estimated levofloxacin concentrations in lung epithelial lining fluid following a single 40 mg respirable drug dose (86.6 mg loaded into the nebulizer) formulated in normal saline in CF patients.



FIG. 9 shows a graph of sputum levofloxacin concentrations in CF subjects following a single IV infusion or aerosol dose of levofloxacin formulated in normal saline. Doses are shown as estimated RDD; the 20 mg and 40 mg RDDs represent nebulizer loaded doses of 43.3 and 86.6 mg, respectively.



FIG. 10 shows a graph depicting modeled sputum levofloxacin concentrations in CF subjects following various doses and routes of administration (aerosol doses formulated in normal saline) of levofloxacin.



FIG. 11 shows a graph of sputum levofloxacin (LVX) concentration in cystic fibrosis patients, after aerosol administration of 50 mg/ml LVX formulated with MgCl2 or with saline using an estimated 40 mg respirable drug dose (RDD) which corresponds to a 86.6 mg loaded drug dose.



FIG. 12 shows a graph of the arithmetic mean serum concentrations of levofloxacin in cystic fibrosis patients, after aerosol administration of a 180 mg dose with 50 mg/ml or 100 mg/ml levofloxacin solution, or a 240 mg dose with a 100 mg/ml levofloxacin solution. Treatment was once daily for 7 days. The 50 mg/ml levofloxacin formulation contained 100 mM magnesium chloride and 150 mM lactose, and the 100 mg/ml levofloxacin contained 200 mM magnesium chloride and no lactose.



FIG. 13 shows a graph of the arithmetic mean sputum concentrations of levofloxacin in cystic fibrosis patients, after aerosol administration of a 180 mg dose with 50 mg/ml or 100 mg/ml levofloxacin solution, or a 240 mg dose with a 100 mg/ml levofloxacin solution. Treatment was once daily for 7 days. The 50 mg/ml levofloxacin formulation contained 100 mM magnesium chloride and 150 mM lactose, and the 100 mg/ml levofloxacin contained 200 mM magnesium chloride and no lactose.



FIG. 14 shows a graph of mean sputum levofloxacin levels in cystic fibrosis patients following a single nebulized dose of levofloxacin formulated in saline compared to formulation in a solution of magnesium chloride. Both formulations were nebulized using a Pari eFlow nebulizer using vibrating mesh technology with the same mesh head design and pore size. The nebulizer loaded dose of levofloxacin in saline was 87 mg and the nebulizer loaded dose of levofloxacin was 180 mg for the formulation using magnesium chloride. Data were normalized to an 87 mg dose by multiplying the observed sputum levofloxacin concentrations obtained with the magnesium chloride formulation by 87/180 (0.48).



FIG. 15 shows a graph of the change in log colony forming units (CFU) of K. pneumoniae ATCC 43816/lung in mice, after aerosol administration of 10 mg/kg or 20 mg/kg doses of levofloxacin (LVX) formulated with and without MgCl2.



FIG. 16 shows a graph of the log colony forming units (CFU) of P. aeruginosa ATCC 27853/lung in a mouse acute lung infection model, after aerosol administration of 125 mg/kg, 63 mg/kg, or 32 mg/kg levofloxacin (LVX) with MgCl2, or intraperitoneal administration (IP) of 125 mg/kg, 63 mg/kg, or 32 mg/kg levofloxacin. Values shown are mean±SD log CFU/lung. Treatment groups (n=8) received 2 doses of antibiotics over 24 h. (p<0.05 for comparisons of aerosol vs. IP administration for each dose).



FIG. 17 shows a graph of log colony forming units (CFU) of P. aeruginosa NH57388A/lung in a murine chronic lung infection model, after twice daily aerosol administration of 60 mg/kg, 30 mg/kg, or 15 mg/kg levofloxacin (LVX) with MgCl2, or twice daily intraperitoneal administration of 60 mg/kg, 30 mg/kg, or 15 mg/kg levofloxacin. Aerosol doses of levofloxacin were formulated in magnesium chloride. Values shown are mean±SD log CFU/lung. Treatment groups (n=8) received 2 doses of antibiotics per day for 72 h. (p<0.05 for comparison of aerosol vs intraperitoneal for the same dose).



FIG. 18 shows a graph of log colony forming units (CFU) of P. aeruginosa ATCC 27853/lung in a murine acute lethal lung infection model, after twice daily aerosol administration of 60 mg/kg of levofloxacin, 60 mg/kg tobramycin, or 400 mg/kg aztreonam, or once daily aerosol administration of 120 mg/kg levofloxacin. Aerosol doses of levofloxacin were formulated in magnesium chloride. Treatment groups (n=8) received drug for 48 h. Values shown are mean±SD log CFU/lung. (p<0.05).



FIG. 19 shows a graph of the percent survival over time of mice infected with P. aeruginosa ATCC 27853 in a murine lethal lung infection model, and treated with twice daily aerosol administration of 60 mg/kg of levofloxacin, 60 mg/kg tobramycin, or 400 mg/kg aztreonam, or once daily aerosol administration of 120 mg/kg levofloxacin. Aerosol doses of levofloxacin were formulated in magnesium chloride. Treatment groups of mice (n=10) received drug for 48 h. Survival was monitored through day 9 following infection.



FIG. 20 shows a graph of the log colony forming units (CFU) of P. aeruginosa NH57388A/lung in a murine chronic lung infection model, after twice daily aerosol administration of 60 mg/kg of levofloxacin, 60 mg/kg tobramycin, or 400 mg/kg aztreonam. Treatment groups were treated twice a day for three consecutive days. Aerosol doses of levofloxacin were formulated in magnesium chloride. Values shown are mean±SD log CFU/lung. Aerosol levofloxacin resulted in lower bacterial counts than aztreonam or untreated control mice (p<0.05).





DETAILED DESCRIPTION

The present invention relates to the field of antimicrobial agents. In particular, the present invention relates to the use of aerosolized fluoroquinolones formulated with divalent or trivalent cations having improved pulmonary availability and thus better bactericidal activity for the treatment and management of bacterial infections of the lung and upper respiratory tract.


Many of the problems associated with antimicrobial-resistant pathogens could be alleviated if the concentration of the antimicrobial could be safely increased at the site of infection. For example, pulmonary infections may be treated by administration of the antimicrobial agent, at high concentrations directly to the site of infection without incurring large systemic concentrations of the antimicrobial. Accordingly, some embodiments disclosed herein are improved methods for delivering drug compositions to treat pulmonary bacterial infections. More specifically, described herein are formulations of fluoroquinolones with divalent or trivalent cations that achieve a desirable pharmacokinetic profile of the fluoroquinolone in humans beneficial for increasing efficacy and reducing the emergence of drug resistance.


Accordingly, some embodiments described herein include methods and compositions that include fluoroquinolones where absorption from lung tissue or the upper airway into systemic circulation after aerosol is retarded. In some such embodiments, fluoroquinolones are complexed with divalent cations in a manner that does not significantly diminish their antimicrobial activity. Such complexes may be for the treatment, maintenance or prevention of infection. In addition, such complexes can show higher concentrations of drug at the sites of infection (e.g., the upper and/or lower respiratory system), and higher efficacy, compared to a fluoroquinolone not combined with divalent or trivalent cations.


Some embodiments of the present invention relate to methods for treating a pulmonary infection, and compositions of levofloxacin or ofloxacin formulated with a divalent or trivalent cation. It has been discovered that particular methods and compositions described herein achieve an improved availability of levofloxacin or ofloxacin in the lungs of subjects. An increased availability in the lungs of antimicrobial agents is useful in the treatment of pulmonary infections, and is particularly advantageous in the treatment of conditions such as cystic fibrosis and chronic obstructive pulmonary disorders, including for example, chronic bronchitis, bronchiectasis, and some asthmas.


Improved availability in the lung can be indicated using a variety of pharmodynamic-pharmacokinetic parameters relating to factors such as increased concentration of a drug in the lung and/or length of time a drug is retained in the lung. Such factors can include lung sputum area under curve (AUC), and maximum lung sputum concentration (Cmax).


Typically, administering aerosolized antimicrobial agents to the lungs can provide high concentrations in the lungs without incurring high systemic concentrations. However, the methods and compositions provided herein achieve unexpectedly increased availability in the lungs.


Generally, the compositions provided herein can comprise solutions of levofloxacin or ofloxacin formulated with a divalent or trivalent cation, such as Mg2+. In some embodiments, the compositions can lack a particular excipient, such as lactose. The compositions may be administered using devices such as nebulizers or a microspray aerosol device inserted directly into the trachea of animals, and can be used to treat a wide variety of bacteria. In addition, methods and compositions provided herein can include additional active agents useful in the treatment of pulmonary infections, and disorders associated with pulmonary infections, such as cystic fibrosis, and chronic obstructive pulmonary disease, including chronic bronchitis and some asthmas.


Definitions


The term “administration” or “administering” refers to a method of giving a dosage of an antimicrobial pharmaceutical composition to a vertebrate. The preferred method of administration can vary depending on various factors, e.g., the components of the pharmaceutical composition, the site of the potential or actual bacterial infection, the microbe involved, and the severity of an actual microbial infection.


A “carrier” or “excipient” is a compound or material used to facilitate administration of the compound, for example, to increase the solubility of the compound. Solid carriers include, e.g., starch, lactose, dicalcium phosphate, sucrose, and kaolin. Liquid carriers include, e.g., sterile water, saline, buffers, non-ionic surfactants, and edible oils such as oil, peanut and sesame oils. In addition, various adjuvants such as are commonly used in the art may be included. These and other such compounds are described in the literature, e.g., in the Merck Index, Merck & Company, Rahway, N.J. Considerations for the inclusion of various components in pharmaceutical compositions are described, e.g., in Gilman et al. (Eds.) (1990); Goodman and Gilman's: The Pharmacological Basis of Therapeutics, 8th Ed., Pergamon Press, incorporated by reference herein in its entirety.


A “diagnostic” as used herein is a compound, method, system, or device that assists in the identification and characterization of a health or disease state. The diagnostic can be used in standard assays as is known in the art.


The term “mammal” is used in its usual biological sense. Thus, it specifically includes humans, cattle, horses, dogs, and cats, but also includes many other species.


The term “microbial infection” refers to the undesired proliferation or presence of invasion of pathogenic microbes in a host organism. This includes the excessive growth of microbes that are normally present in or on the body of a mammal or other organism. More generally, a microbial infection can be any situation in which the presence of a microbial population(s) is damaging to a host mammal. Thus, a microbial infection exists when excessive numbers of a microbial population are present in or on a mammal's body, or when the effects of the presence of a microbial population(s) is damaging the cells or other tissue of a mammal.


The term “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.


The term “pharmaceutically acceptable salt” refers to salts that retain the biological effectiveness and properties of the compounds of this invention and, which are not biologically or otherwise undesirable. In many cases, the compounds of this invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, naphtoic acid, oleic acid, palmitic acid, pamoic (emboic) acid, stearic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, ascorbic acid, glucoheptonic acid, glucuronic acid, lactic acid, lactobioic acid, tartaric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases. Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like; particularly preferred are the ammonium, potassium, sodium, calcium and magnesium salts. Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, histidine, arginine, lysine, benethamine, N-methyl-glucamine, and ethanolamine. Other acids include dodecylsufuric acid, naphthalene-1,5-disulfonic acid, naphthalene-2-sulfonic acid, and saccharin.


“Solvate” refers to the compound formed by the interaction of a solvent and fluoroquinolone antimicrobial, a metabolite, or salt thereof. Suitable solvates are pharmaceutically acceptable solvates including hydrates.


In the context of the response of a microbe, such as a bacterium, to an antimicrobial agent, the term “susceptibility” refers to the sensitivity of the microbe for the presence of the antimicrobial agent. So, to increase the susceptibility means that the microbe will be inhibited by a lower concentration of the antimicrobial agent in the medium surrounding the microbial cells. This is equivalent to saying that the microbe is more sensitive to the antimicrobial agent. In most cases the minimum inhibitory concentration (MIC) of that antimicrobial agent will have been reduced. The MIC90 can include the concentration to inhibit growth in 90% of organisms.


By “therapeutically effective amount” or “pharmaceutically effective amount” is meant a fluoroquinolone antimicrobial agent, as disclosed for this invention, which has a therapeutic effect. The doses of fluoroquinolone antimicrobial agent which are useful in treatment are therapeutically effective amounts. Thus, as used herein, a therapeutically effective amount means those amounts of fluoroquinolone antimicrobial agent which produce the desired therapeutic effect as judged by clinical trial results and/or model animal infection studies. In particular embodiments, the fluoroquinolone antimicrobial agent are administered in a predetermined dose, and thus a therapeutically effective amount would be an amount of the dose administered. This amount and the amount of the fluoroquinolone antimicrobial agent can be routinely determined by one of skill in the art, and will vary, depending on several factors, such as the particular microbial strain involved. This amount can further depend upon the patient's height, weight, sex, age and medical history. For prophylactic treatments, a therapeutically effective amount is that amount which would be effective to prevent a microbial infection.


A “therapeutic effect” relieves, to some extent, one or more of the symptoms of the infection, and includes curing an infection. “Curing” means that the symptoms of active infection are eliminated, including the total or substantial elimination of excessive members of viable microbe of those involved in the infection to a point at or below the threshold of detection by traditional measurements. However, certain long-term or permanent effects of the acute or chronic infection may exist even after a cure is obtained (such as extensive tissue damage). As used herein, a “therapeutic effect” is defined as a statistically significant reduction in bacterial load in a host, emergence of resistance, pulmonary function, or improvement in infection symptoms or functional status as measured by human clinical results or animal studies.


“Treat,” “treatment,” or “treating,” as used herein refers to administering a pharmaceutical composition for prophylactic and/or therapeutic purposes. The term “prophylactic treatment” refers to treating a patient who is not yet infected, but who is susceptible to, or otherwise at risk of, a particular infection such that there is a reduced onset of infection. The term “therapeutic treatment” refers to administering treatment to a patient already suffering from an infection that may be acute or chronic. Treatment may eliminate the pathogen, or it may reduce the pathogen load in the tissues that results in improvements measured by patients symptoms or measures of lung function. Thus, in preferred embodiments, treating is the administration to a mammal (either for therapeutic or prophylactic purposes) of therapeutically effective amounts of a fluoroquinolone antimicrobial agent.


Pharmacokinetics (PK) is concerned with the time course of antimicrobial concentration in the body. Pharmacodynamics (PD) is concerned with the relationship between pharmacokinetics and the antimicrobial efficacy in vivo. PK/PD parameters correlate antimicrobial exposure with antimicrobial activity. The rate of killing by antimicrobial is dependent on antimicrobial mode of action and is determined by either the length of time necessary to kill (time-dependent) or the effect of increasing concentrations alone (concentration-dependent) or integrated over time as an area under the concentration-time curve (AUC). To predict the therapeutic efficacy of antimicrobials with diverse mechanisms of action different PK/PD parameters may be used. PK/PD parameters may be used to determine the availability of antimicrobial compositions, for example, availability of a antimicrobial agent in a composition in the pulmonary system, and/or bioavailability of a antimicrobial agent in a composition in plasma/serum.


“AUC/MIC ratio” is one example of a PK/PD parameter. AUC is defined as the area under the plasma/serum or site-of-infection concentration-time curve of an antimicrobial agent in vivo (in animal or human). For example, the site of infection and/or the site where concentration is measured can include portions of the pulmonary system, such as bronchial fluid and/or sputum. Accordingly, AUC may be a serum AUC, or a pulmonary AUC based on concentrations in serum and pulmonary tissues (sputum, epithelial lining fluid, or homogenates of whole tissue). AUC(0-t) can include the area under curve for time zero to a specific time ‘t.’ AUC(0-inf) can include the area under curve from time zero to infinity. AUC/MIC ratio is determined by dividing the 24-hour-AUC for an individual antimicrobial by the MIC for the same antimicrobial determined in vitro. Activity of antimicrobials with the dose-dependent killing (such as fluoroquinolones) is well predicted by the magnitude of the AUC/MIC ratio. The AUC:MIC ratio can also prevent selection of drug-resistant bacteria.


“Cmax:MIC” ratio is another PK:PD parameter. It describes the maximum drug concentration in plasma or tissue relative to the MIC. Fluoroquinolones and aminoglycosides are examples where Cmax:MIC may predict in vivo bacterial killing where resistance can be suppressed.


“Time above MIC” (T>MIC) is another PK/PD parameter. It is expressed a percentage of a dosage interval in which the plasma or site-of-infection level exceeds the MIC. Activity of antimicrobials with the time-dependent killing (such as beta-lactams or monobactam antibiotics) is well predicted by the magnitude of the T>MIC ratio.


The term “dosing interval” refers to the time between administrations of the two sequential doses of a pharmaceutical's during multiple dosing regimens. For example, in the case of orally administered ciprofloxacin, which is administered twice daily (traditional regimen of 400 mg b.i.d) and orally administered levofloxacin, which is administered once a day (500 mg or 750 mg q.d.), the dosing intervals are 12 hours and 24 hours, respectively.


As used herein, the “peak period” of a pharmaceutical's in vivo concentration is defined as that time of the pharmaceutical dosing interval when the pharmaceutical concentration is not less than 50% of its maximum plasma or site-of-infection concentration. In some embodiments, “peak period” is used to describe an interval of antimicrobial dosing.


The estimated “respirable delivered dose” is the dose or amount of drug delivered to the lung of a patient using a nebulizer or other aerosol delivery device. The RDD is estimated from the inspiratory phase of a breath simulation device programmed to the European Standard pattern of 15 breaths per minute, with an inspiration to expiration ratio of 1:1, and measurement of particles emitted from a nebulizer with a size of about 5 microns or less.


Improved Availability


The antibiotic rate of killing is dependent upon antibiotic mode of action and is determined by either the length of time necessary for the antibiotic to kill (time-dependent) or the effect of increasing the antibiotic concentration (concentration-dependent). Fluoroquinolones are characterized by concentration-dependent, time-kill activity where a therapeutic effect requires a high local peak concentration above the MICs of the infecting pathogen.


Fluoroquinolone efficacy in humans, animals and in vitro models of infection is linked to AUC:MIC ratio and Cmax:MIC ratio. A number of in vitro studies have been conducted to determine if high concentrations of levofloxacin with an extremely short half-lives (as predicted from a rat and human PK model) in a target tissues resulted in bacterial killing superior to that seen under conditions with more prolonged residence times. In these studies, levofloxacin concentrations that were 0.018-fold-1024-fold the MIC were evaluated in a standard kill-curve and an in vitro hollow fiber assay. In both of these assays, high concentrations of levofloxacin were rapidly bactericidal and reached their maximum levels of killing in 10-20 minutes. This level of killing was sustained whether levofloxacin was maintained at that level or given a half-life of 10 minutes. In addition, no resistance was observed. Accordingly, high doses and rapid delivery of specially formulated levofloxacin is rapidly bactericidal for susceptible organisms and resistant organisms.


In one embodiment, the concentration of levofloxacin at the site of infection is increased by delivering levofloxacin in combination with divalent or trivalent cations directly to the lung using inhalation therapy, thereby decreasing the amount of time levofloxacin is in the “mutant selection window” (MSW). Such a therapeutic approach achieves broader coverage of pathogens (including levofloxacin resistant strains), prevents further resistance development, and results in shorter courses of levofloxacin therapy.


Some embodiments include compositions of levofloxacin or ofloxacin having an improved pulmonary availability, wherein an increased pulmonary AUC is indicative of the improved pulmonary availability of the levofloxacin or ofloxacin. In some embodiments, the increase can be at least about 10%, 20, 30, 40%, 50%, 75%, 100%, 150%, 200%, 250%, 300%, and 500%. An increase can be relative to, for example, a composition lacking a divalent or trivalent cation, and/or a composition having certain excipients (e.g., lactose), and/or a composition delivered to the lung at a certain rate, and/or a certain respirable delivered dose. In some embodiments, methods are provided that include achieving an improved pulmonary availability indicated by a lung AUC greater than about 400 h·mg/L, about 500 h·mg/L, about 600 h·mg/L, about 700 h·mg/L, about 800 h·mg/L, about 900 h·mg/L, about 1000 h·mg/L, about 1100 h·mg/L, about 1200 h·mg/L, about 1300 h·mg/L, about 1400 h·mg/L, about 1500 h·mg/L, about 1600 h·mg/L, about 1700 h·mg/L, about 1800 h·mg/L, about 1900 h·mg/L, about 2000 h·mg/L, about 2100 h·mg/L, about 2200 h·mg/L, about 2300 h·mg/L, about 2400 h·mg/L, about 2500 h·mg/L, about 2600 h·mg/L, about 2700 h·mg/L, about 2800 h·mg/L, about 2900 h·mg/L, about 3000 h·mg/L, about 3100 h·mg/L, about 3200 h·mg/L, about 3300 h·mg/L, about 3400 h·mg/L, about 3500 h·mg/L, about 3600 h·mg/L, about 3700 h·mg/L, about 3800 h·mg/L, about 3900 h·mg/L, about 4000 h·mg/L, about 4100 h·mg/L, about 4200 h·mg/L, about 4300 h·mg/L, about 4400 h·mg/L, and about 4500 h·mg/L. The increase can be measured for example, in bronchial fluid, homogenates of whole lung tissue, or in sputum.


In more embodiments, an increased pulmonary Cmax can be indicative of an improved pulmonary availability for a formulation of levofloxacin or ofloxacin. In some such embodiments, the increase can be at least about 50%, 75%, 100%, and 150%. An increase can be relative to a composition, for example, lacking a divalent or trivalent cation, and/or a composition having certain excipients (e.g., lactose), and/or a composition delivered to the lung at a certain rate, and/or a certain respirable delivered dose. In some embodiments, methods are provided that include achieving an improved pulmonary availability indicated by a lung Cmax greater than about 300 mg/L, about 400 mg/L, about 500 mg/L, about 600 mg/L, about 700 mg/L, about 800 mg/L, about 900 mg/L, about 1000 mg/L, about 1100 mg/L, about 1200 mg/L, about 1300 mg/L, about 1400 mg/L, about 1500 mg/L, about 1600 mg/L, about 1700 mg/L, about 1800 mg/L, about 1900 mg/L, about 2000 mg/L, about 2100 mg/L, about 2200 mg/L, about 2300 mg/L, about 2400 mg/L, about 2500 mg/L, about 2600 mg/L, about 2700 mg/L, about 2800 mg/L, about 2900 mg/L, about 3000 mg/L, about 3100 mg/L, about 3200 mg/L, about 3300 mg/L, about 3400 mg/L, about 3500 mg/L, about 3600 mg/L, about 3700 mg/L, about 3800 mg/L, about 3900 mg/L, about 4000 mg/L, about 4100 mg/L, about 4200 mg/L, about 4300 mg/L, about 4400 mg/L, about 4500 mg/L, about 4600 mg/L, about 4700 mg/L, about 4800 mg/L, about 4900 mg/L, and 5000 mg/L. The increase can be measured for example, in bronchial secretions, epithelial lining fluid, lung homogenates, and in sputum.


In even more embodiments, a decrease in serum AUC or serum Cmax can be indicative of an increase in the pulmonary availability and prolonged exposure of a levofloxacin or ofloxacin using a formulation. In some such embodiments, the decrease can be at least about 1%, 5%, 10, 20%, or 50%. A decrease can be relative to a composition, for example, lacking a divalent or trivalent cation, and/or a composition having certain excipients (e.g., lactose), and/or a composition delivered to the lung at a certain rate as a solution or other composition. In some embodiments, a formulation of levofloxacin can be characterized by a AUC:MIC90 greater than about 200, 300, 400, 500, 600, 700, 800, 900, and 1000. In some such embodiments, the AUC can be pulmonary AUC.


In some embodiments, the concentrations in lung tissue (sputum, ELF, tissue homogenates) can be characterized by the PK-PD indice Cmax:MIC90 greater than about 20, 40, 60, 80, 100, 120, 140, 160, 180, 200, 220, 240, 260, 280, 300, 320, 340, 340, 360, 380, 400, 420, 440, 460, 480, 500, 520, 540, 560, 580, and 600.


The increase or decrease in a parameter to measure improved availability of a formulation of levofloxacin or ofloxacin can be relative to a formulation of levofloxacin or ofloxacin lacking divalent or trivalent cations, relative to a formulation of levofloxacin or ofloxacin lacking lactose, and/or relative to a formulation of with a lower concentration of levofloxacin or ofloxacin.


Methods of Treatment or Prophylaxis


In some embodiments, a method is provided for treating a microbial infection in an animal, specifically including in a mammal, by treating an animal suffering from such an lung infection with a fluoroquinolone antimicrobial formulated with a divalent or trivalent cation and having improved pulmonary availability. In some embodiments, fluoroquinolone antimicrobials may be administered following aerosol formation and inhalation. Thus, this method of treatment is especially appropriate for the treatment of pulmonary infections involving microbial strains that are difficult to treat using an antimicrobial agent delivered orally or parenterally due to the need for high dose levels (which can cause undesirable side effects), or due to lack of any clinically effective antimicrobial agents. In one such embodiment, this method may be used to administer a fluoroquinolone antimicrobial directly to the site of infection. Such a method may reduce systemic exposure and maximizes the amount of antimicrobial agent to the site of microbial infection. This method is also appropriate for treating infections involving microbes that are susceptible to fluoroquinolone antimicrobials as a way of reducing the frequency of selection of resistant microbes. This method is also appropriate for treating infections involving microbes that are otherwise resistant to fluoroquinolone antimicrobials as a way of increasing the amount of antimicrobial at the site of microbial infection. A subject may be identified as infected with bacteria that are capable of developing resistance by diagnosing the subject as having symptoms that are characteristic of a bacterial infection with a bacteria species known to have resistant strains or a with a bacteria that is a member of group that are known to have resistant strains. Alternatively, the bacteria may be cultured and identified as a species known to have resistant strains or a bacteria that is a member of group that are known to have resistant strains.


In some embodiments, the aerosol fluoroquinolone antimicrobial agent formulated with divalent or trivalent cations is administered at a level sufficient to overcome the emergence resistance in bacteria or increase killing efficiency such that resistance does not have the opportunity to develop.


In some embodiments, the aerosol fluoroquinolone therapy may be administered as a treatment or prophylaxis in combination or alternating therapeutic sequence with other aerosol, oral or parenteral antibiotics. By non-limiting example this may include aerosol tobramycin and/or other aminoglycoside, aerosol aztreonam and/or other beta or monobactam, aerosol ciprofloxacin and/or other fluoroquinolones, aerosol azithromycin and/or other macrolides or ketolides, tetracycline and/or other tetracyclines, quinupristin and/or other streptogramins, linezolid and/or other oxazolidinones, vancomycin and/or other glycopeptides, and chloramphenicol and/or other phenicols, and colisitin and/or other polymyxins.


In addition, compositions and methods provided herein can include the aerosol fluoroquinolone therapy administered as a treatment or prophylaxis in combination or alternating therapeutic sequence with an additional active agent. As discussed above, some such additional agents can include antibiotics. More additional agents can include bronchodilators, anticholinergics, glucocorticoids, eicosanoid inhibitors, and combinations thereof. Examples of bronchodilators include salbutamol, levosalbuterol, terbutaline, fenoterol, terbutlaine, pirbuterol, procaterol, bitolterol, rimiterol, carbuterol, tulobuterol, reproterol, salmeterol, formoterol, arformoterol, bambuterol, clenbuterol, indacterol, theophylline, roflumilast, cilomilast. Examples of anticholinergics include ipratropium, and tiotropium. Examples of glucocorticoids include prednisone, fluticasone, budesonide, mometasone, ciclesonide, and beclomethasone. Examples of eicosanoids include montelukast, pranlukast, zafirlukast, zileuton, ramatroban, and seratrodast. More additional agents can include pulmozyme, hypertonic saline, agents that restore chloride channel function in CF, inhaled beta-agonists, inhaled antimuscarinic agents, inhaled corticosteroids, and inhaled or oral phosphodiesterase inhibitors. More additional agents can include CFTR modulators, for example, VX-770, atluren, VX-809. More additional agents can include agents to restore airway surface liquid, for example, denufosol, mannitol, GS-9411, and SPI-8811 More additional agents can include anti-inflammatory agents, for example, ibuprofen, sildenafil, and simavastatin.


Pharmaceutical Compositions


For purposes of the method described herein, a fluoroquinolone antimicrobial agent formulated with a divalent or trivalent cation having improved pulmonary availability may be administered using an inhaler. In some embodiments, a fluoroquinolone antimicrobial disclosed herein is produced as a pharmaceutical composition suitable for aerosol formation, good taste, storage stability, and patient safety and tolerability. In some embodiments, the isoform content of the manufactured fluoroquinolone may be optimized for tolerability, antimicrobial activity and stability.


Formulations can include a divalent or trivalent cation. The divalent or trivalent cation can include, for example, magnesium, calcium, zinc, copper, aluminum, and iron. In some embodiments, the solution comprises magnesium chloride, magnesium sulfate, zinc chloride, or copper chloride. In some embodiments, the divalent or trivalent cation concentration can be from about 25 mM to about 400 mM, from about 50 mM to about 400 mM, from about 100 mM to about 300 mM, from about 100 mM to about 250 mM, from about 125 mM to about 250 mM, from about 150 mM to about 250 mM, from about 175 mM to about 225 mM, from about 180 mM to about 220 mM, and from about 190 mM to about 210 mM. In some embodiments, the concentration is about 200 mM. In some embodiments, the magnesium chloride, magnesium sulfate, zinc chloride, or copper chloride can have a concentration from about 5% to about 25%, from about 10% to about 20%, and from about 15% to about 20%. In some embodiments, the ratio of fluoroquinolone to divalent or trivalent cation can be 1:1 to 2:1 or 1:1 to 1:2.


Non-limiting fluoroquinolones for use as described herein include levofloxacin, ofloxacin, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, lomefloxacin, moxifloxacin, norfloxacin, pefloxacin, sparfloxacin, garenoxacin, sitafloxacin, and DX-619.


The formulation can have a fluoroquinolone concentration, for example, levofloxacin or ofloxacin, greater than about 50 mg/ml, about 60 mg/ml, about 70 mg/ml, about 80 mg/ml, about 90 mg/ml, about 100 mg/ml, about 110 mg/ml, about 120 mg/ml, about 130 mg/ml, about 140 mg/ml, about 150 mg/ml, about 160 mg/ml, about 170 mg/ml, about 180 mg/ml, about 190 mg/ml, and about 200 mg/ml. In some embodiments, the formulation can have a fluoroquinolone concentration, for example, levofloxacin or ofloxacin, from about 50 mg/ml to about 200 mg/ml, from about 75 mg/ml to about 150 mg/ml, from about 80 mg/ml to about 125 mg/ml, from about 80 mg/ml to about 120 mg/ml, from about 90 mg/ml to about 125 mg/ml, from about 90 mg/ml to about 120 mg/ml, and from about 90 mg/ml to about 110 mg/ml. In some embodiments, the concentration is about 100 mg/ml.


The formulation can have an osmolality from about 300 mOsmol/kg to about 500 mOsmol/kg, from about 325 mOsmol/kg to about 450 mOsmol/kg, from about 350 mOsmol/kg to about 425 mOsmol/kg, and from about 350 mOsmol/kg to about 400 mOsmol/kg. In some embodiments, the osmolality of the formulation is greater than about 300 mOsmol/kg, about 325 mOsmol/kg, about 350 mOsmol/kg, about 375 mOsmol/kg, about 400 mOsmol/kg, about 425 mOsmol/kg, about 450 mOsmol/kg, about 475 mOsmol/kg, and about 500 mOsmol/kg.


The formulation can have a pH from about 4.5 to about 8.5, from about 5.0 to about 8.0, from about 5.0 to about 7.0, from about 5.0 to about 6.5, from about 5.5 to about 6.5, and from 6.0 to about 6.5.


The formulation can comprise a conventional pharmaceutical carrier, excipient or the like (e.g., mannitol, lactose, starch, magnesium stearate, sodium saccharine, talcum, cellulose, sodium crosscarmellose, glucose, gelatin, sucrose, magnesium carbonate, and the like), or auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like (e.g., sodium acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine acetate, triethanolamine oleate, and the like). In some embodiments, the formulation can lack a conventional pharmaceutical carrier, excipient or the like. Some embodiments include a formulation lacking lactose. Some embodiments comprise lactose at a concentration less than about 10%, 5%, 1%, or 0.1%. In some embodiments, the formulation can consist essentially of levofloxacin or ofloxacin and a divalent or trivalent cation.


In some embodiments, a formulation can comprise a levofloxacin concentration between about 75 mg/ml to about 150 mg/ml, a magnesium chloride concentration between about 150 mM to about 250 mM, a pH between about 5 to about 7; an osmolality of between about 300 mOsmol/kg to about 600 mOsmol/kg, and lacks lactose.


In some embodiments, a formulation comprises a levofloxacin concentration of about 100 mg/ml, a magnesium chloride concentration of about 200 mM, a pH of about 6.2, an osmolality of about 383 mOsmol/kg, and lacks lactose. In some embodiments, a formulation consists essentially of a levofloxacin concentration of about 90 mg/ml to about 110 mg/ml, a magnesium chloride concentration of about 180 mM to about 220 mM, a pH of about 5 to about 7, an osmolality of about 300 mOsmol/kg to 500 mOsmol/kg, and lacks lactose.


Administration


The fluoroquinolone antimicrobials formulated with divalent or trivalent cations and having improved pulmonary availability may be administered at a therapeutically effective dosage, e.g., a dosage sufficient to provide treatment for the disease states previously described. The amount of active compound administered will, of course, be dependent on the subject and disease state being treated, the severity of the affliction, the manner and schedule of administration and the judgment of the prescribing physician; for example, a likely dose range for aerosol administration of levofloxacin would be about 20 to 300 mg per day, the active agents being selected for longer or shorter pulmonary half-lives, respectively. In some embodiments, a likely dose range for aerosol administration of levofloxacin would be about 20 to 300 mg BID (twice daily).


Administration of the fluoroquinolone antimicrobial agents disclosed herein or the pharmaceutically acceptable salts thereof can be via any of the accepted modes of administration for agents that serve similar utilities including, but not limited to, aerosol inhalation. Methods, devices and compositions for delivery are described in U.S. Patent Application Publication No. 2006-0276483, incorporated by reference in its entirety.


Pharmaceutically acceptable compositions include solid, semi-solid, liquid and aerosol dosage forms, such as, for example, powders, liquids, suspensions, complexations, liposomes, particulates, or the like. Preferably, the compositions are provided in unit dosage forms suitable for single administration of a precise dose.


The fluoroquinolone antimicrobial agent can be administered either alone or in some alternatives, in combination with a conventional pharmaceutical carrier, excipient or the like (e.g., mannitol, lactose, starch, magnesium stearate, sodium saccharine, talcum, cellulose, sodium crosscarmellose, glucose, gelatin, sucrose, magnesium carbonate, and the like). If desired, the pharmaceutical composition can also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like (e.g., sodium acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine acetate, triethanolamine oleate, and the like). Generally, depending on the intended mode of administration, the pharmaceutical formulation will contain about 0.005% to 95%, preferably about 0.5% to 50% by weight of a compound of the invention. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.


In one preferred embodiment, the compositions will take the form of a unit dosage form such as vial containing a liquid, solid to be suspended, dry powder, lyophilate, or other composition and thus the composition may contain, along with the active ingredient, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like.


Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, etc. an active compound as defined above and optional pharmaceutical adjuvants in a carrier (e.g., water, saline, aqueous dextrose, glycerol, glycols, ethanol or the like) to form a solution or suspension. Solutions to be aerosolized can be prepared in conventional forms, either as liquid solutions or suspensions, as emulsions, or in solid forms suitable for dissolution or suspension in liquid prior to aerosol production and inhalation. The percentage of active compound contained in such aerosol compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the subject. However, percentages of active ingredient of 0.01% to 90% in solution are employable, and will be higher if the composition is a solid, which will be subsequently diluted to the above percentages. In some embodiments, the composition will comprise 1.0%-50.0% of the active agent in solution.


Compositions described herein can be administered with a frequency of about 1, 2, 3, 4, or more times daily, 1, 2, 3, 4, 5, 6, 7 or more times weekly, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more times monthly. In particular embodiments, the compositions are administered twice daily.


Aerosol Delivery


For pulmonary administration, the upper airways are avoided in favor of the middle and lower airways. Pulmonary drug delivery may be accomplished by inhalation of an aerosol through the mouth and throat. Particles having a mass median aerodynamic diameter (MMAD) of greater than about 5 microns generally do not reach the lung; instead, they tend to impact the back of the throat and are swallowed and possibly orally absorbed. Particles having diameters of about 2 to about 5 microns are small enough to reach the upper- to mid-pulmonary region (conducting airways), but are too large to reach the alveoli. Smaller particles, i.e., about 0.5 to about 2 microns, are capable of reaching the alveolar region. Particles having diameters smaller than about 0.5 microns can also be deposited in the alveolar region by sedimentation, although very small particles may be exhaled.


In one embodiment, a nebulizer is selected on the basis of allowing the formation of an aerosol of a fluoroquinolone antimicrobial agent disclosed herein having an MMAD predominantly between about 2 to about 5 microns. In one embodiment, the delivered amount of fluoroquinolone antimicrobial agent provides a therapeutic effect for respiratory infections. The nebulizer can deliver an aerosol comprising a mass median aerodynamic diameter from about 2 microns to about 5 microns with a geometric standard deviation less than or equal to about 2.5 microns, a mass median aerodynamic diameter from about 2.5 microns to about 4.5 microns with a geometric standard deviation less than or equal to about 1.8 microns, and a mass median aerodynamic diameter from about 2.8 microns to about 4.3 microns with a geometric standard deviation less than or equal to about 2 microns. In some embodiments, the aerosol can be produced using a vibrating mesh nebulizer. An example of a vibrating mesh nebulizer includes the PARI E-FLOW® nebulizer or a nebulizer using PARI eFlow technology. More examples of nebulizers are provided in U.S. Pat. Nos. 4,268,460; 4,253,468; 4,046,146; 3,826,255; 4,649,911; 4,510,929; 4,624,251; 5,164,740; 5,586,550; 5,758,637; 6,644,304; 6,338,443; 5,906,202; 5,934,272; 5,960,792; 5,971,951; 6,070,575; 6,192,876; 6,230,706; 6,349,719; 6,367,470; 6,543,442; 6,584,971; 6,601,581; 4,263,907; 5,709,202; 5,823,179; 6,192,876; 6,644,304; 5,549,102; 6,083,922; 6,161,536; 6,264,922; 6,557,549; and 6,612,303 all of which are hereby incorporated by reference in their entireties. More commercial examples of nebulizers that can be used with the formulations described herein include Respirgard II®, Aeroneb®, Aeroneb® Pro, and Aeroneb® Go produced by Aerogen; AERx® and AERx Essence™ produced by Aradigm; Porta-Neb®, Freeway Freedom®, Sidestream, Ventstream and I-neb produced by Respironics, Inc.; and PARI LC-Plus®, PARI LC-Star®, produced by PARI, GmbH. By further non-limiting example, U.S. Pat. No. 6,196,219, is hereby incorporated by reference in its entirety.


The amount of levofloxacin or ofloxacin that can be administered to the lungs with an aerosol dose, such as a respirable drug dose (RDD), that can include at least about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 110 mg, about 120 mg, about 125 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, about 300 mg, about 310 mg, about 320 mg, about 330 mg, about 340 mg, about 350 mg, about 460 mg, about 470 mg, about 480 mg, about 490 mg, about 500 mg, about 510 mg, about 520 mg, about 530 mg, about 540 mg, about 550 mg, about 560 mg, about 570 mg, about 580 mg, about 590 mg, about 600 mg, about 610 mg, about 620 mg, about 630 mg, about 640 mg, about 650 mg, about 660 mg, about 670 mg, about 680 mg, about 690 mg, about 700 mg, about 710 mg, about 720 mg, about 730 mg, about 740 mg, about 750 mg, about 760 mg, about 770 mg, about 780 mg, about 790 mg, and about 800 mg. In some embodiments, the amount of levofloxacin or ofloxacin that can be administered to the lungs with an aerosol dose, such as a respirable drug dose (RDD), that can include at least about 20 mg, 50 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 450 mg, 500 mg, 550 mg, 600 mg, 650 mg, 700 mg, 750 mg, 800 mg, 850 mg, 900 mg, 950 mg, 1000 mg, 1050 mg, 1100 mg, 1150 mg, 1200 mg, 1250 mg, 1300 mg, 1350 mg, 1400 mg, 1450 mg, and 1500 mg.


The aerosol can be administered to the lungs in less than about 10 minutes, about 5 minutes, about 4 minutes, about 3 minutes, about 2 minutes, and about 1 minute.


Indications


Methods and compositions described herein can be used to treat pulmonary infections and disorders. Examples of such disorders can include cystic fibrosis, pneumonia, and chronic obstructive pulmonary disease, including chronic bronchitis, and some asthmas. Some embodiments include treating an infection comprising one or more bacteria selected from the group consisting of Pseudomonas aeruginosa, Pseudomonas fluorescens, Pseudomonas acidovorans, Pseudomonas alcaligenes, Pseudomonas putida, Stenotrophomonas maltophilia, Aeromonas hydrophilia, Escherichia coli, Citrobacter freundii, Salmonella typhimurium, Salmonella typhi, Salmonella paratyphi, Salmonella enteritidis, Shigella dysenteriae, Shigella flexneri, Shigella sonnei, Enterobacter cloacae, Enterobacter aerogenes, Klebsiella pneumoniae, Klebsiella oxytoca, Serratia marcescens, Morganella morganii, Proteus mirabilis, Proteus vulgaris, Providencia alcalifaciens, Providencia rettgeri, Providencia stuartii, Acinetobacter calcoaceticus, Acinetobacter haemolyticus, Yersinia enterocolitica, Yersinia pestis, Yersinia pseudotuberculosis, Yersinia intermedia, Bordetella pertussis, Bordetella parapertussis, Bordetella bronchiseptica, Haemophilus influenzae, Haemophilus parainfluenzae, Haemophilus haemolyticus, Haemophilus parahaemolyticus, Haemophilus ducreyi, Pasteurella multocida, Pasteurella haemolytica, Helicobacter pylori, Campylobacter fetus, Campylobacter jejuni, Campylobacter coli, Borrelia burgdorferi, Vibrio cholera, Vibrio parahaemolyticus, Legionella pneumophila, Listeria monocytogenes, Neisseria gonorrhoeae, Neisseria meningitidis, Burkholderia cepacia, Francisella tularensis, Kingella, and Moraxella. In some embodiments, the lung infection is caused by a gram-negative anaerobic bacteria. In more embodiments, the lung infection comprises one or more of the bacteria selected from the group consisting of Bacteroides fragilis, Bacteroides distasonis, Bacteroides 3452A homology group, Bacteroides vulgatus, Bacteroides ovalus, Bacteroides thetaiotaomicron, Bacteroides uniformis, Bacteroides eggerthii, and Bacteroides splanchnicus. In some embodiments, the lung infection is caused by a gram-positive bacteria. In some embodiments, the lung infection comprises one or more of the bacteria selected from the group consisting of Corynebacterium diphtherias, Corynebacterium ulcerans, Streptococcus pneumoniae, Streptococcus agalactiae, Streptococcus pyogenes, Streptococcus milleri; Streptococcus (Group G); Streptococcus (Group C/F); Enterococcus faecalis, Enterococcus faecium, Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus saprophyticus, Staphylococcus intermedius, Staphylococcus hyicus subsp. hyicus, Staphylococcus haemolyticus, Staphylococcus hominis, and Staphylococcus saccharolyticus. In some embodiments, the lung infection is caused by a gram-positive anaerobic bacteria. In some embodiments, the lung infection is caused by one or more bacteria selected from the group consisting of Clostridium difficile, Clostridium perfringens, Clostridium tetini, and Clostridium botulinum. In some embodiments, the lung infection is caused by an acid-fast bacteria. In some embodiments, the lung infection is caused by one or more bacteria selected from the group consisting of Mycobacterium tuberculosis, Mycobacterium avium, Mycobacterium intracellulare, and Mycobacterium leprae. In some embodiments, the lung infection is caused by an atypical bacteria. In some embodiments, the lung infection is caused by one or more bacteria selected from the group consisting of Chlamydia pneumoniae and Mycoplasma pneumoniae.


EXAMPLES
Comparative Example 1—Administration of Fluoroquinolones in a Rat Pharmacokinetic Model

This example relates to aerosol and intravenous administration of fluoroquinolones in saline. A rat pharmacokinetic model was used to compare intravenous and pulmonary administration of fluoroquinolones. Male Sprague-Dawley rats (Charles Rivers) were administered 10 mg/kg doses of levofloxacin, ciprofloxacin, gatifloxacin, norfloxacin, or gemifloxacin. Doses were administered via the lateral tail vein, or to the lung just above the tracheal bifurcation using a micro-spray aerosol device (Penn Century, Philadelphia, Pa.). Levofloxacin was prepared in sterile 0.9% saline to concentrations of 5 mg/ml (IV) and 60 mg/ml (aerosol).


Approximately 0.3 ml blood samples were taken from 2-6 rats at each timepoint via an indwelling jugular vein cannula, and collected in lithium heparin tubes. Bronchial alveolar lavage (BAL) and lung tissue were collected following euthanasia. Levofloxacin concentrations in plasma, lung tissue and BAL were determined using a HPLC assay, and the data analyzed using WinNonlin (Pharsight Corporation, v 5.0). Sample concentrations were determined against a standard curve.


Serum AUC(0-inf) (area under the concentration time curve, for time zero to infinity), serum MRT (mean retention time), serum t½ (half-life), BAL AUC, MAT (mean absorption time), and F (bioavailability) were determined and are shown in Table 1.
















TABLE 1







Serum AUC
Serum
Serum
BAL
MAT



Drug
Route
(0-inf)
MRT

AUC
(h)
F, % from Lung vs IV






















Levofloxacin
IV
3.8
0.7
0.5
1.6
NA
NA


Levofloxacin
Aerosol
3.7
0.7
0.5
3.0
0
97%


Ciprofloxacin
IV
2.6
0.76
0.53
3.9
NA
NA


Ciprofloxacin
Aerosol
0.8
1.35
0.93
78.4
  0.59
82%


Gatifloxacin
IV
5.31
1.39
1.06
0.35
NA
NA


Gatifloxacin
Aerosol
5.83
1.34
1.13
3.12
0
100% 


Norfloxacin
IV
4.65
1.59
1.21
0.8
NA
NA


Norfloxacin
Aerosol
4.46
1.29
1.13
24.6
0
100% 


Gemifloxacin
IV
4.54
1.41
1.04
0.9
NA
NA


Gemifloxacin
Aerosol
5.86
2.06
1.68
140.4
  0.65
86%









Aerosol administration of ciprofloxacin, gatifloxacin, norfloxacin, or gemifloxacin resulted in a significant increase in BAL AUC, compared to intravenous administration. Aerosol administration of levofloxacin did not show such a significant increase in BAL AUC, compared to intravenous administration. In addition, levofloxacin showed rapid absorption from the lung into serum. Thus, aerosol administration of levofloxacin in saline did not result in significant increased availability of drug to the lung.


Comparative Example 2—Aerosol Administration of Levofloxacin with Divalent Cations in Rats

This example relates to a series of studies that included aerosol administration of levofloxacin with divalent cations and lactose and IV or aerosol administration of levofloxacin in saline. Rats were administered 10 mg/kg levofloxacin (LVX) in saline or LVX formulated with CaCl2, MgCl2, or Zn+2. Table 2 shows the formulations of levofloxacin used in these studies.















TABLE 2







Levofloxacin
Levofloxacin
Levofloxacin
Levofloxacin
Levofloxacin



(IV)
(Aerosol)
(MgCl2)
(CaCl2)
(ZnCl2)
























Levofloxacin
5 mg/ml
60 mg/ml
60
mg/ml
60
mg/ml
60
mg/ml













MgCl2


120
mM















CaCl2



120
mM














ZnCl2




120
mM













Lactose


150
mM











In one study, pharmacokinetic parameters including Cmax (maximum serum concentration), CL/F (total body clearance/bioavailability) were measured and are shown in Table 3. A graph of plasma concentration of levofloxacin with time is shown in FIG. 1, where levofloxacin was administered by aerosol, by intravenous injection, or by aerosol with MgCl2.











TABLE 3









Mean (+/− SD)













LVX
LVX
LVX


Parameter
Unit
IV
Aerosol
MgCl2





Plasma AUC
hr · mg/L
3.79 (±0.89)
3.69 (±0.14)
3.72 (±0.24)


Plasma Half-life
hr
0.49 (±0.10)
0.52 (±0.09)
0.73 (±0.07)


Plasma Cmax
mg/L
5.54 (±1.51)
6.01 (±1.54)
6.66 (±1.70)


Plasma CL/F
L/hr/kg
2.81 (±0.54)
2.83 (±0.11)
2.68 (±0.17)


Plasma MRT
hr
0.70 (±0.14)
0.71 (±0.08)
0.88 (±0.06)


Plasma MAT
hr
NA
0.01
0.18


Plasma F
%
NA
97.4
98.2


(Bioavailability)


BAL AUC(0-6 h)
hr · mg/L
1.6
3.0
8.3









A two compartment pharmacokinetic model may be used describe the difference in graphs of plasma levofloxacin with time for intravenous and aerosol administration. Plasma AUC after intravenous administration was similar to plasma AUC after administration by aerosol with Mg+2 (3.79 hr·mg/L vs. 3.72 hr·mg/L, respectively). This suggests near 100% bioavailability of the divalent-complex antibiotic from the lung. The mean residence time (MRT) of levofloxacin was greater after aerosol administration compared to after intravenous administration (0.88 vs. 0.70 hours). This delay in absorption was associated with an increase in BAL levofloxacin AUC(0-6h) in BAL (1.6 hr·mg/L vs. 8.3 hr·mg/L for intravenous vs. aerosol dosing, respectively), and an 18-fold increase in the mean absorption time (MAT)


In another study, levofloxacin levels after aerosol administration for formulations containing saline, Zn+2, Ca+2 or Mg+2 were measured and pharmacokinetic parameters were determined. Table 4 and FIG. 2 summarize the results.
















TABLE 4












F, %




Serum




from




AUC
Serum
Serum
BAL
MAT
Lung


Drug
Route
(0-inf)
MRT

AUC
(h)
vs IV






















Levofloxacin
IV
3.8
0.7
0.5
1.6
NA
NA


Levofloxacin
Aerosol
3.7
0.7
0.5
3.0
0
 97%


Levofloxacin
Aerosol
4.4
1.35
1.2
29.6
0.7
116%


(MgCl2)


Levofloxacin
Aerosol
4.3
1.17
0.8
8.3
0.5
116%


(CaCl2)


Levofloxacin
Aerosol
4.4
1.6
1.8
55.6
0.9
100%


(ZnCl2)









Aerosol administration of levofloxacin complexed with Ca+2 and Mg+2 resulted in a longer plasma half-life and longer MAT compared to levofloxacin formulated in saline, indicative of slower lung clearance to plasma (Table 4). Levofloxacin formulated with Ca+2 or Mg+2 produced a 2- to 5-fold higher levofloxacin Cmax and AUC in BAL and lung tissue compared to intravenous levofloxacin or aerosolized levofloxacin formulated in saline (Table 4, FIG. 2). These data suggest that aerosol levofloxacin complexed with divalent cation should result in higher efficacy in the treatment of pulmonary infections.


Example 3—Pharmacokinetic Modeling and Deconvolution Analysis

This example relates to modeling drug concentrations in lung. Pharmacokinetic deconvolution methods are useful to determine the amount of drug remaining in the lung after administration. Such methods are particularly useful where direct measurements are difficult and/or produce variable results, for example, measuring drug concentrations in lung using sputum samples.


Serum and urinary pharmacokinetic parameters can be determined using non-compartmental and compartmental methods, and drug concentrations in the lung over time can be calculated using deconvolution. This approach has been reported for aerosol delivery of tobramycin, where a dose of 5.6 mg/kg showed bioavailability of about 9% and absorption over a 3 hour period, consistent with empirically derived data (Cooney G. F., et al, “Absolute bioavailability and absorption characteristics of aerosolized tobramycin in adults with cystic fibrosis.” J. Clinical Pharmacol. (1994), 34, 255-259, incorporated herein by reference in its entirety).


An example deconvolution method is summarized in FIG. 3. This analysis compares the appearance and elimination of drug following aerosol and intravenous doses to determine the amount of drug remaining in the lung (absorption compartment) over time. To estimate concentrations of drug in lung, amounts were divided by estimates of the epithelial lung fluid (ELF) volume (25 ml) for each subject. Non-compartmental pharmacokinetic analysis was subsequently applied to these projected concentrations of drug in the lung to determine AUCs.


Application of the deconvolution methodology can be accomplished by human or animal aerosol dosing of 50 mg respirable drug dose of levofloxacin in saline or complexed with Mg+2, delivered in a nebulizer or other respiratory delivery device, with resulting plasma drug concentrations profiles and calculated pharmacokinetic parameters as illustrated in FIG. 4 and Tables 5 and 6. Serum levofloxacin concentrations following a 5 minute IV infusion of levofloxacin to a single healthy volunteer were analyzed using WinNonlin and the pharmacokinetic parameters presented in Table 5. On a separate occasion, this volunteer received a single aerosol dose of levofloxacin (RDD=50 mg) by a PARI eFlow vibrating mesh nebulizer. FIG. 4 shows a comparison of the serum levofloxacin concentrations following an IV or aerosol dose. Using the PK model depicted in FIG. 3, serum concentrations of levofloxacin measured in serum following an aerosol dose were deconvoluted using the serum PK data for an IV dose of levofloxacin (PK parameters shown in Table 5). The results are shown in Table 6, which show the estimated amount of levofloxacin (in mg) remaining in the lung over time.













TABLE 5







Parameter
Unit
Estimate




















AUC
hr · mg/L
3.14



K10_HL
hr
0.92



Alpha
1/hr
6.85



Beta
1/hr
0.11



Alpha_HL
hr
0.10



Beta_HL
hr
6.50



A
mg/L
2.05



B
mg/L
0.30



Cmax
mg/L
1.88



CL
L/hr
15.93



AUMC
hr · hr · mg/L
26.78



MRT
hr
8.49



Vss
L
135.29



V2
L
114.03



CLD2
L/hr
111.26






















TABLE 6







Cumulative input

Drug




Input
into serum

remaining



rate
compartment
Input
in lung
Fraction


Hours
(mg/hr)
(mg)
fraction
(mg)
remaining




















0.24
35.79
26.67
0.53
23.33
0.47


0.48
13.80
31.16
0.62
18.84
0.38


0.72
18.25
34.84
0.70
15.16
0.30


0.96
20.55
39.88
0.80
10.12
0.20


1.2
14.98
44.14
0.88
5.86
0.12


1.44
9.21
47.03
0.94
2.97
0.06


1.68
5.13
48.68
0.97
1.32
0.03


1.92
1.81
49.51
0.99
0.49
0.01


2.16
0.45
49.69
0.99
0.31
0.01


2.4
0.36
49.79
1.00
0.21
0.00


2.64
0.27
49.86
1.00
0.14
0.00


2.88
0.18
49.92
1.00
0.08
0.00


3.12
0.11
49.95
1.00
0.05
0.00


3.36
0.08
49.97
1.00
0.03
0.00


3.6
0.05
49.99
1.00
0.01
0.00


3.84
0.02
50.00
1.00
0.00
0.00









These data can be used to calculate the amount (in mg) of levofloxacin remaining in the lung as a function of time. FIG. 5 and Table 6 shows an example for the estimated amount of drug remaining in the lung over time, whereas only 10% of the 50 mg respirable drug dose administered over a 5 minute period remains in the lung after 1.2 hours. This experiment demonstrates the utility of the deconvolution method.


Comparative Example 4—Aerosol and Systemic Administration of Levofloxacin with Saline

This example relates to aerosol and systemic administration of levofloxacin formulated in a saline solution using estimated respirable drug doses of 20 mg or 40 mg (nebulizer loaded doses of 43.3 and 86.6 mg, respectively) of levofloxacin. Single aerosol doses of two dose levels levofloxacin (using the IV formulation Levaquin®) were administered to normal healthy volunteers and stable CF subjects using the PARI eFlow high efficiency nebulizer.


Safety, tolerability, and pharmacokinetics (serum, sputum, and urinary excretion) data were collected after each dose. The nebulizer was loaded with 3.6 ml of a Levaquin® solution diluted in saline to isotonicity, at a concentration of 11.9 mg/ml for the 20 mg respirable drug dose group, and at a concentration of 23.8 mg/ml for the 40 mg respirable drug dose group. These volumes correspond to “load” doses of 43.3 mg and 86.6 mg levofloxacin for the 20 and 40 mg RDD, respectively. Table 7 summarizes nebulizer loaded doses with the corresponding estimated RDD for levofloxacin formulated in saline.











TABLE 7





Levofloxacin Concentration
Nebulizer Loaded
RDD (based on


in Saline (mg/L)
dose (mg)
particles < 5 μm) (mg)

















12
43.3
20


23.8
86.6
40









Novaluzid® (AstraZeneca) was co-administered to minimize the oral absorption of any levofloxacin that was swallowed during inhalation. Each subject received an intravenous dose of levofloxacin and an aerosol saline dose at the first visit to generate pharmacokinetic data for comparison with aerosol levofloxacin doses, and to assess the tolerability of delivering solutions using the eFlow device.


Serum and urine levofloxacin concentrations were analyzed using a validated HPLC assay by Anapharm (Quebec City, Canada). Sputum levofloxacin assays were developed and cross validated using the serum assay.


Serum data: Serum levofloxacin concentrations following the intravenous infusion were fit to a two-compartment open pharmacokinetic model using iteratively reweighted least-squares regression (WinNonlin). A weight of 1/y-observed was applied in the regression. Goodness of fit was assessed by the minimized objective function and inspection of weighted residual plots. Serum levofloxacin concentrations resulting from aerosol administration were analyzed using deconvolution methods to estimate the residence time of the aerosol dose in the lung (Gibaldi M. and Perrier D. Pharmacokinetics, 2nd Edition. Marcel Dekker: New York, 1982, incorporated herein by reference in its entirety). The pharmacokinetic model and approach used for deconvolution analysis is described in Example 3. Briefly, this analysis compares the appearance and elimination of drug following aerosol and intravenous doses to determine the amount of drug remaining in the lung (absorption compartment) over time. To estimate concentrations of drug in lung, amounts were divided by estimates of the epithelial lung fluid (ELF) volume (25 ml) for each subject. Noncompartmental pharmacokinetic analysis was subsequently applied to these projected concentrations of drug in the lung to determine values for AUC.


Sputum data: Sputum concentration data were analyzed using noncompartmental pharmacokinetic methods (Gibaldi M. and Perrier D. Pharmacokinetics, 2nd Edition. Marcel Dekker: New York, 1982, incorporated herein by reference in its entirety). The area under the sputum concentration vs. time curve was estimated using the linear trapezoidal rule. Since sputum was only collected from 0.5 to 8 hrs, forward and backward extrapolation from terminal and initial phases was conducted to generate estimates of secondary pharmacokinetic parameters (Cmax, AUC).


PK-PD parameters such as AUC:MIC, and Cmax:MIC, were generated for lung exposures estimated from deconvolution of serum levofloxacin concentration data. Examples of parameters were calculated at different values for levofloxacin MIC for P. aeruginosa at estimated respirable doses of levofloxacin ranging from 20-120 mg administered twice daily in CF subjects. Levofloxacin MIC distributions (MIC50, MIC90, and mode MIC) were measured for clinical isolates from CF isolates (Traczewski M M and Brown S D. In Vitro activity of doripenem against P. aeruginosa and Burkholderia cepacia isolates from both cystic fibrosis and non-cystic fibrosis patients. Antimicrob Agents Chemother 2006; 50:819-21, incorporated herein by reference in its entirety).


Dosing summary: A total of 7 normal healthy volunteers (NHV) and 9 subjects with CF were enrolled in the study. All subjects completed all phases of the protocol; the dose summary is provided in Table 8. Seven of 9 cystic fibrosis subjects received both the 20 and 40 mg respirable drug dose levels, whereas 2 subjects were re-dosed with the 20 mg dose level with salbutamol pretreatment. Forced expiratory volume during 1 second (FEV1)












TABLE 8









Baseline FEV1
Levofloxacin Doses Studied












Subject
Gender
(% Pred)
20 mg
40 mg
20 mg A*










Normal healthy volunteers












001
M
83%
X
X



002
M
100% 
X
X


003
M
109% 
X
X


004
M
130% 
X
X


006
M
119% 
X
X


008
M
82%
X
X


009
F
104% 
X
X







Cystic fibrosis patients












010
F
58%
X
X



012
M
67%
X

X


013
F
40%
X
X


014
F
40%
X
X


015
M
74%
X
X


016
F
66%
X
X


018
M
63%
X
X


019
M
42%
X
X


022
M
48%
X

X





*A = Repeat study using pretreatment with salbutamol






Levofloxacin pharmacokinetics in serum: FIG. 6 shows mean serum levofloxacin concentrations in normal and CF subjects following IV and aerosol doses. Total levofloxacin clearance was 17.2 and 14.1 L/h in the NHV and CF subjects, respectively. Serum levofloxacin concentrations following aerosol administration generally paralleled those observed with the intravenous dose, particularly after 1 hour post-dosing. Comparison of the levofloxacin AUCs from IV or aerosol dosing using model-independent analysis showed that levofloxacin exposure from aerosol doses relative to the 50 mg IV dose was (mean+/−SD) 35.6+/−9.4% and 59.4+/−16.6% for the low and high aerosol doses, respectively for the normal volunteers, and 27.9+/−3.3% and 51.1+/−11.2% for CF subjects.


Serum deconvolution analysis: Serum levofloxacin concentrations following aerosol administration were successfully deconvoluted in all subjects, permitting estimation of the amount of drug in the absorption (lung) compartment over time (FIG. 7). Absorption from the lung into serum occurred significantly more slowly in CF subjects than in healthy normal volunteers; 50% of the lung dose appeared to remain in the lung for at least 0.5 hours after the dose. Using a literature value for estimation of lung epithelial lining fluid (ELF) volume of 25 ml, the estimated concentrations of levofloxacin in ELF of CF patients following a single aerosol dose is shown in FIGS. 8A and 8B (Rennard, S, G. et al., Estimation of volume of epithelial lining fluid recovered by lavage using urea as a marker of dilution. J. Appl. Physio. 60: 532-8, incorporated by reference herein in its entirety). Mean projected Cmax concentrations in the ELF of CF patients at the conclusion of the low and high doses exceeded 500 μg/ml and 1000 μg/ml, respectively. When integrated over time, the projected mean+/−SD levofloxacin AUC in lung fluid was 365+/−338 and 710+/−471 for the low and high dose in healthy subjects, and 354+/−274 and 1,199+/−1,147 for low and high doses in CF patients.


Levofloxacin pharmacokinetics in sputum: FIG. 9 shows levofloxacin concentrations in sputum following administration of the low and high aerosol dose in CF subjects. Sputum levofloxacin concentrations following both aerosol dose levels were markedly higher for at least 1 hour post-dose with aerosol levofloxacin than those obtained with the 50 mg IV dose. Concentrations tended to fall rapidly during the first 2 hours of administration, consistent with drug absorption from the lung. Sputum levofloxacin concentrations were variable within and between patients, but generally the 86.6 mg loaded dose did provide higher concentrations over the period of observations.


Levofloxacin concentrations in sputum from CF subjects following aerosol administration were averaged and compared with concentrations obtained by other routes of administration. FIG. 10 depicts modeled sputum concentrations in CF subjects for both aerosol dose levels, the 50 mg IV dose to the same subjects infused over 5 minutes, and a 750 mg oral dose (another study not described herein); Table 9 shows Cmax, AUC, and half-life values for measured sputum levofloxacin concentrations.















TABLE 9








50 mg
20 mg
40 mg
750 mg



Parameter
IV
Aerosol
Aerosol
Oral






















Cmax (mg/L)
0.8
86.2
211.5
8.7



AUC (hr · mg/L)
2.5
67.1
171.4
93.4



T1/2 (h)
3.8
0.9
1.3
6.7










While a 750 mg oral levofloxacin dose results in more prolonged drug concentrations in sputum, aerosol doses as low as 20 mg produce peak concentrations 10-fold higher.



FIG. 6 shows serum levofloxacin concentrations following single RDDs of 20 or 40 mg of levofloxacin formulated in saline, and IV doses of Levaquin to normal healthy volunteers and CF patients. These data were used to perform pharmacokinetic deconvolution as previously described. FIG. 7 shows the results of deconvolution, showing the estimated amount of levofloxacin remaining in the lung over time. Levofloxacin remained in the lung for a longer period in CF patients compared to normal healthy volunteers (FIG. 7). Notably, the levofloxacin concentrations observed in sputum are consistent with ELF concentrations projected from the deconvolution analysis (FIGS. 8A and 8B vs. FIG. 9 and Table 9).


PK-PD analysis: Integration of pharmacokinetics with susceptibility data for P. aeruginosa allows for assessment of the expected pharmacodynamic effects in vivo. PK-PD parameters for fluoroquinolones include the 24 hr AUC:MIC and Cmax:MIC ratios. Very high Cmax:MIC ratios appear to be significant for rapid bacterial killing and suppression of drug resistance.


The results of PK-PD analysis with simulated ELF PK data (generated from the amount of levofloxacin in lung divided by the ELF volume) from the deconvolution analysis for twice daily dosing of levofloxacin along with MIC data for P. aeruginosa can be used to calculate levofloxacin PK-PD indices for P. aeruginosa. Table 10 shows predicted PK-PD indices (Cmax:MIC; 24 h AUC:MIC) for particular dosage regimens of levofloxacin.










TABLE 10







Levofloxacin
Cmax:MIC (24 h AUC:MIC) values for


MIC*
Levofloxacin dosage regimens











(mg/L)
20 mg BID
40 mg BID
80 mg BID
120 mg BID





32 
16 (22)
31 (44)
62 (88)
94 (131)


16 (MIC90)
31 (44)
62 (88)
124 (175)
188 (263)


8
62 (88)
124 (175)
248 (350)
375 (525)


4
124 (175)
248 (350)
496 (700)
  750 (1,050)


2
248 (350)
496 (700)
  992 (1,400)
1,500 (2,100)


1 (MIC50)
496 (700)
  992 (1,400)
1,884 (2,800)
3,000 (4,200)


0.5 (mode)
  992 (1,400)
1,884 (2,800)
3,768 (4,200)
6,000 (8,400)


24 h AUC
700
1,400
2,800
4,200


(h · mg/L)


Cmax (mg/L)
500
1,000
2,000
3,000





*MIC50, MIC90, and mode values from Traczewski M M and Brown S D (2006)






For example, a daily dose of 20 mg BID levofloxacin, Cmax:MIC=248; 24 h AUC:MIC=350; and a levofloxacin MIC=2 mg/L. The simulations show that the primary target value of Cmax:MIC>20 would be obtained by all regimens for over 90% of CF isolates of P. aeruginosa. In addition, the secondary PK-PD target value of 24 hr AUC:MIC>300 would be obtained for a majority of strains at the lower doses, but could also cover over 90% of the isolates at the higher doses projected to be evaluated in upcoming clinical studies.


Comparative Example 5—Aerosol Administration of 30 mg/ml and 50 mg/ml Solutions of Levofloxacin Formulated with MgCl2

This example relates to aerosol administration to CF patients of 30 mg/ml and 50 mg/ml solutions of levofloxacin formulated with MgCl2. Table 11 shows the formulations of levofloxacin with MgCl2 and lactose.













TABLE 11








30 mg/ml
50 mg/ml






















Levofloxacin, mg/ml (mM)
30
(81.6)
50
(136)



Magnesium, mg/ml (mM)
1.5
(60)
2.4
(100)



Chloride, mg/ml (mM)
4.3
(120)
7.1
(200)



Lactose, mg/ml (mM)
51.4
(150)
51.4
(150)











pH
6.3
6.3



Osmolality, mOsm/kg
314
400










Eight stable CF patients received loaded doses of 78 mg, 175 mg, and 260 mg (corresponding to RDD of 40 mg, 80 mg, and 120 mg, respectively) of levofloxacin formulated with MgCl2 using an eFlow high efficiency nebulizer (PART Pharma, Munich, Germany). Escalated doses were administered 1 week apart. A separate group of 7 CF patients were administered a single dose of 750 mg oral levofloxacin at weekly intervals for 4 consecutive weeks. Serum and sputum samples were assayed for levofloxacin by HPLC. Serum and sputum levofloxacin concentration data were analyzed using non-compartmental pharmacokinetic methods. Mean pharmacokinetic parameters are shown in Table 12.











TABLE 12









Dose













Aerosol
Aerosol
Aerosol





78 mg
175 mg
260 mg



RDD:
RDD:
RDD:
Oral
IV


Parameter
40 mg
80 mg
120 mg
750 mg
50 mg
















Sputum
Cmax
388
714
1112
8.7
1.05



(mg/L)



Cmax: MIC90*
49
89
139
1.1
0.1



AUC(0-inf)
851
656
1448
93.4
5.70



(h · mg/L)



t½ (h)
3.09
1.61
2.51
6.70
3.5


Serum
Cmax
0.48
0.95
1.30
7.30
2.55



(mg/L)



AUC(0-inf)
2.08
4.45
6.54
76.6
3.91



(h · mg/L)



t1/2 (h)
5.69
6.50
6.20
7.60
5.89



MAT**
1.06
1.61
1.30
ND
ND





*P. aeruginosa MIC90 for CF isolates is 8 μg/ml


MAT = Mean absorption time form the lung.






PK-PD data have previously shown that for fluoroquinolones, Cmax:MIC ratio is a PK-PD parameter associated with optimal bacterial killing and prevention of resistance. Aerosol administration of levofloxacin with MgCl2 provides concentrations in sputum that achieve Cmax:MIC ratios for P. aeruginosa >40. In contrast, an oral levofloxacin dose of 750 mg produces a ratio of 1.1. These data show that aerosolized doses of levofloxacin with MgCl2 provide high exposures in sputum that are greater than those achievable with oral levofloxacin.


Comparative Example 6—Comparison of Aerosol Administration of a 40 mg RDD of Levofloxacin Formulated in Saline or MgCl2 in CF Patients

This example relates to aerosol administration of levofloxacin with MgCl2 or in saline using estimated respirable drug doses (RDD) of 40 mg levofloxacin. The concentrations of levofloxacin in saline are 23.8 mg/ml and 30 mg/ml in a formulation containing MgCl2/lactose (see Table 11). CF patients received 40 mg respirable drug doses of levofloxacin by aerosol delivery: 7 patients received levofloxacin formulated in saline; 10 patients received the same estimated RDD received levofloxacin formulated with MgCl2. Sputum samples were taken at various times up to 24 hours and levofloxacin concentrations determined using a HPLC/fluorescence method. Mean levofloxacin concentrations measured in sputum over time are shown in FIG. 11. Levofloxacin delivered with MgCl2 is retained in sputum for a longer period and at higher concentrations than the same dose of levofloxacin delivered in saline.


Further comparison of the PK parameters in CF sputum for aerosol administration of levofloxacin in saline (Example 4-Table 8) indicate that both a significantly higher sputum Cmax and AUC are achieved by complexation with magnesium (e.g., Cmax is 211.5 mg/L levofloxacin vs. 388 for levofloxacin:Mg and AUC is 171.4 h·mg/L levofloxacin/saline vs. 851 h·mg/L levofloxacin:Mg for 40 mg respirable dose).


Example 7: Pharmacokinetics of Levofloxacin in CF Patients Following Aerosol Administration of Formulations Containing MgCl2 Plus Lactose for Up to 14 Days

CF patients received respirable delivered doses of approximately 40 mg, 80 mg, or 120 mg per treatment (loaded doses of 78 mg, 175 mg, or 260 mg per treatment) on day 1 followed by twice daily dosing for 14 days. Formulations shown in Table 11 were used. Standard non-compartmental and compartmental PK methods were used to generate serum, sputum, and urinary PK parameters (Gibaldi M, Perrier B. Pharmacokinetics. 2nd ed. New York:Marcel-Dekker; 1982, incorporated by reference herein in its entirety). PK parameters were determined for serum and sputum and are shown in Tables 13 and 14, respectively. Comparison with the administration of levofloxacin in saline (Example 4) indicate that both a significantly higher sputum Cmax and AUC are achieved by complexation with magnesium (e.g., Cmax is 211.5 mg/L levofloxacin vs. 448.97 for levofloxacin:Mg and AUC is 171.4 h·mg/L levofloxacin vs. 420.54 h·mg/L levofloxacin:Mg (day 1) for 40 mg respirable dose).











TABLE 13









Loaded Levofloxacin Dose (Mean ± SD)











78 mg (n = 10)
175 mg (n = 10)
260 mg (n = 10)










Parameter
RDD: 40 mg
RDD: 80 mg
RDD: 120 mg














Day 1
Serum Cmax (mg/L)
0.36 ± 0.6 
1.05 ± 0.29
1.34 ± 0.42



Serum Tmax (h)
0.21
1.00
0.29



Serum AUC(0-t) (h · mg/L)
2.2.61 ± 1.81
9.12 ± 1.89
10.24 ± 3.08 



Serum AUC(inf) (h · mg/L)
3.40 ± 1.69
9.94 ± 2.30
11.44 ± 2.86 



Serum t½ (h)
7.21 ± 1.89
6.60 ± 0.91
7.36 ± 2.42


Day 15
Serum Cmax (mg/L)
0.58 ± 0.35
1.37 ± 0.56
2.39 ± 0.56



Serum Tmax (h)
0.53
0.98
0.22



Serum AUC(0-t) (h · mg/L)
5.16 ± 1.22
12.95 ± 5.75 
18.12 ± 12.00



Serum t½ (h)
8.48 ± 2.42
6.52 ± 0.87
6.62 ± 1.16


















TABLE 14









Loaded Levofloxacin Dose (Mean ± SD)











78 mg (n = 10)
175 mg (n = 10)
260 mg (n = 10)










Parameter
RDD: 40 mg
RDD: 80 mg
RDD: 120 mg














Day 1
Sputum Cmax (mg/L)
448.97 ± 875.02
1333.96 ± 1146.55
1766.23 ± 1493.52



Sputum Tmax (h)
0.52
0.53
0.54



Sputum AUC(0-t) (h · mg/L)
420.54 ± 994.99
1468.60 ± 1420.04
1779.23 ± 1223.12



Sputum t½ (h)
1.54 ± 0.56
2.56 ± 1.94
5.04


Day 15
Sputum Cmax (mg/L)
 612.06 ± 1440.13
1258.82 ± 1888.15
1721.51 ± 1511.15



Sputum Tmax (h)
0.52
0.53
0.50



Sputum AUC(0-t) (h · mg/L)
 637.56 ± 1280.39
1642.81 ± 2849.76
1272.76 ± 795.19 



Sputum t½ (h)
9.96 ± 13.9
4.10 ± 1.93
2.73 ± 1.58









Example 8—Aerosol Administration of 50 mg/ml and 100 mg/ml Solutions of Levofloxacin Formulated with MgCl2

This example relates to aerosol administration to CF patients of 50 mg/ml and 100 mg/ml solutions of levofloxacin formulated with MgCl2 at doses of 180 mg and 240 mg. Table 15 shows the formulations of levofloxacin with MgCl2.












TABLE 15







50 mg/ml
100 mg/ml




















Levofloxacin, mg/ml
50
100



Magnesium, mg/ml (mM)
2.4 (100)
 4.9 (200)



Chloride, mg/ml (mM)
7.1 (200)
14.2 (400) 



Lactose, mg/ml (mM)
51.4 (150) 
0 (0)



pH
6-8
6-8



Osmolality, mOsm/kg
300-500
300-500










Levofloxacin with MgCl2 was administered by inhalation using a PARI eFlow nebulizer using vibrating mesh technology with the 35 L head configuration. Subjects received, in an order specified by a randomization schedule, a single 180 mg dose of a particular formulation (50 mg/ml or 100 mg/ml) in Period 1 of the study, followed by a 7-day wash-out period and a single 180 mg dose of the other formulation (50 mg/ml or 100 mg/ml) in Period 2. This was followed by 7 consecutive days of a once-daily 240 mg dose during Period 3. Serum and sputum concentrations of levofloxacin were measured using an HPLC/fluorescence method.


With respect to serum concentrations of levofloxacin, the arithmetic mean serum concentrations of levofloxacin after administration of 180 mg with the 100 mg/ml formulation were slightly higher than after administration with the 50 mg/ml formulation (FIG. 12). Table 16 summarizes pharmacokinetic parameters for levofloxacin after administration of single 180 mg doses as a 50 mg/ml or 100 mg/ml solution for inhalation, and after administration of 240 mg as a 100 mg/ml solution for inhalation once daily for 7 days to patients with CF. The mean Cmax and AUC(inf) for the 100 mg/ml formulation were 35% and 22% higher than the corresponding values for the 50 mg/ml formulation.












TABLE 16









Dose: 180 mg
Dose: 240 mg










Parameter1
50 mg/ml
100 mg/ml
100 mg/ml





Serum Cmax (ng/ml)
952 ±
1,284 ±
1,707 ±



617 (10)
642 (10)
624 (10)


Serum Tmax (h)
0.25 (10)
0.17 (10)
0.3 (10)


Serum Cmin (ng/ml)
58.5 ±
73.5 ±
145 ±



60.4 (10)
45.8 (10)
172 (10)


Serum AUC(0-t) (h · ng/ml)
7,074 ±
9,054 ±
14,771 ±



3,625 (10)
3,411 (10)
9,969 (10)


Serum AUC(inf) (h · ng/ml)
8,058 ±
9,848 ±
16,930 ±



3,704 (9)
3,813 (10)
13,631 (10)


Serum t½ (h)
6.40 ±
6.78 ±
7.49 ±



1.27 (9)
1.61 (10)
2.89 (10)






1Arithmetic mean ± standard deviation (N) except for Tmax for which the median (N) is reported.







Based on a mean t½ of 6.78 h after administration of 180 mg with the 100 mg/ml formulation, the accumulation with once-daily dosing should be about 9%. There was a 1.33-fold increase in the mean Cmax after administration of 240 mg with the 100 mg/ml formulation, similar to the increase in level of dose. AUC(0-t) on Day 7 after administration of 240 mg QD×7 days is AUC(0-24), or the AUC over the dosing interval, which should be equivalent to AUC(inf) after a single dose. Correcting the 14,771 h·ng/ml mean AUC(0-t) of the 240 mg dose level to the 180 mg dose level, results in an estimate of 11,078 h·ng/ml, comparable to the observed AUC(inf) of 9,848±3,813 h·ng/ml after administration of a single 180 mg dose of the same formulation. This demonstrates the linearity of the pharmacokinetics of levofloxacin after single and multiple aerosol doses of levofloxacin with the 100 mg/ml formulation. The arithmetic mean t½ was comparable for all three treatments, ranging from 6.40 h to 7.49 h.


With respect to sputum concentrations of levofloxacin, mean values for arithmetic sputum concentration, Cmax, and AUC were similar after administration of 180 mg with either the 50 mg/ml or 100 mg/ml formulation (FIG. 13). Table 17 summarizes sputum pharmacokinetic parameters for levofloxacin after administration of single 180 mg doses as a 50 mg/ml or 100 mg/ml solution for inhalation, and after administration of 240 mg as a 100 mg/ml solution for inhalation once daily for 7 days to patients with CF.












TABLE 17









Loaded Dose: 180 mg
Loaded Dose: 240 mg










Parameter1
50 mg/ml
100 mg/ml
100 mg/ml





Sputum Cmax
2,563,119 ±
2,932,121 ±
4,690,808 ±


(ng/ml)
1,411,715 (10)
2,559,422 (10)
4,515,727 (10)


Sputum Tmax
0.27 (10)
0.28 (10)
0.29 (10)


(h)


Sputum Cmin
398 ±
278 ±
697 ±


(ng/ml)
482 (10)
192 (10)
939 (10)


Sputum
1,889,669 ±
1,958,819 ±
4,507,180 ±


AUC(0-t)
1,252,341 (10)
2,109,909 (10)
6,593,884 (10)


(h · ng/ml)


Sputum
1,890,699 ±
1,960,771 ±
4,517,439 ±


AUC(inf)
1,252,486 (10)
2,110,392 (10)
6,611,353 (10)


(h · ng/ml)


Sputum t½
3.55 ±
4.34 ±
4.58 ±


(h)
2.69 (10)
1.80 (10)
2.54 (10)






1Arithmetic mean ± standard deviation (N) except for Tmax for which the median (N) is reported.







There was a 1.6-fold increase in Cmax between the 180 mg and 240 mg doses of the 100 mg/ml formulation, of 2,932,121 ng/ml to 4,690,808 ng/ml (Table 17). In view of the small number of patients and variability between subjects, this increase is reasonably consistent with a predicted increase of about 1.33-fold. In contrast, there was a 2.3-fold increase in AUC, from 1,960,771 h·ng/ml [AUC(inf)] to 4,507,180 h·ng/ml [AUC(0-24)]. The arithmetic mean t½ was comparable for all three treatments, ranging from 3.55 h to 4.58 h (Table 16).


These results show that levofloxacin exposure in sputum was orders of magnitude higher than that in serum (Tables 16 and 17). However, the ratio of levofloxacin exposure in sputum to that in serum was relatively independent of the formulation and the dose, and averaged approximately 260,000% for Cmax, and 25,000% for AUC (Table 18).











TABLE 18









Sputum/Serum Ratio










Dose: 180 mg
Dose: 240 mg












Parameter
50 mg/ml
100 mg/ml
100 mg/ml
















Cmax (ng/ml)
269,336
228,271
274,796



AUC (h · ng/ml)1
23,462
19,911
30,514








1AUC(inf) for the single 180 mg doses and AUC(0-t) for the multiple 240 mg dose.







Sputum exposure is similar for both formulations. Taking into account potential accumulation from the 240 mg QD×7-day regimen, the systemic and sputum exposure after administration of 180 mg and 240 mg as the 100 mg/ml formulation appear to be proportional to dose and consistent between single and multiple doses.


Table 19 compares levofloxacin AUC and Cmax results following nebulization of formulations shown in Examples 4 and 8 as the raw results or normalized to the RDD or nebulizer loaded dose for each formulation tested.











TABLE 19









Dose-normalized Sputum Levofloxacin PK Parameters in CF Patients



Example










Example 4 formulations
Example 8 formulations









Formulation/Dose















Dose Level C







Levofloxacin
Dose Level C
Dose Level D



Dose Level A
Dose Level B
(50 mg/ml) with
Levofloxacin
Levofloxacin



Levofloxacin
Levofloxacin
MgCl2 and
(100 mg/ml)
(100 mg/ml)



(12 mg/ml)
(23.8 mg/ml)
Lactose
with MgCl2
with MgCl2
















Loaded Dose
43.3
86.6
180
180
240


Estimated RDD
20
40
92
98
131


Cmax (ng/ml)
86,200
211,500
2,563,119
2,932,121
4,690,808


AUC (hr · ng/ml)
67,100
171,400
1,890,699
1,960,771
4,517,439


Loaded Dose-
1,991
2,442
14,240
16,290
19,545


normalized Cmax


(ng/ml per mg


dose)


Loaded Dose-
1,550
1,979
10,504
10,893
18,823


normalized AUC


(hr · ng/ml per mg


dose)


RDD- normalized
4,310
5,288
27,866
29,862
35,830


Cmax (ng/ml per mg


dose)


RDD- normalized
3,355
4,285
20,556
19,969
34,505


AUC (hr · ng/ml per


mg dose)









The dose-normalized AUC and Cmax PK parameters show the significantly increased exposures of levofloxacin in sputum using the formulations of Example 8 that include levofloxacin formulated with Mg+2 over the formulations of Example 4 that lack Mg+2. The differences in sputum concentrations of levofloxacin between Example 4 and Example 8 formulations are further shown in FIG. 14.


Example 9—Mouse Lung Infection Model

A mouse lung infection model was used to compare the efficacy of intravenous administration with pulmonary administration of fluoroquinolones. Eight mice per group were infected with Klebsiella pneumoniae ATCC 43816 by intra-tracheal instillation. Twenty-four hours after infection, mice were administered aerosol doses of 10 or 20 mg/kg twice daily (BID) using a microspray aerosol generation device (PennCentury, Philadelphia, Pa.). Twenty-four hours after beginning treatment, animals were sacrificed and their lungs were removed, homogenized, and plated to determine colony counts. Table 20 shows the formulations used in this study.












TABLE 20









Levofloxacin in saline
Levofloxacin with MgCl2











Dose (mg/kg)
10
20
10
20














Levofloxacin (mg/mL)
4
8
4
8


MgCl2 (mM)
0
0
8
16


Saline (%)
0.9
0.9
0
0


Lactose (mM)
0
0
12
24









Levofloxacin formulated with MgCl2 produced 1 log greater bacterial killing than levofloxacin formulated in saline at each dose tested (FIG. 15). This result is consistent with the increased lung concentrations determined in the rat in Example 2.


Example 10—Efficacy of Aerosol Levofloxacin Formulated with MgCl2 in Mouse Lung Infection Models

This example relates to aerosol administration of levofloxacin with MgCl2, and intraperiteneal administration of levofloxacin in saline. The purpose of the following studies was to determine the efficacy of these therapies in acute and chronic lung infection models due to P. aeruginosa.


Antimicrobial agents: Levofloxacin (LKT Laboratories, St. Paul, Minn.), tobramycin (Sicor pharmaceuticals, Irvine, Calif.), and aztreonam (MP Biomedicals, Solon, Ohio) were purchased from independent vendors. Prior to the initiation of each experiment, fresh stock solutions of each antibiotic were prepared. Levofloxacin formulated with MgCl2 was diluted in water; levofloxacin and tobramycin were diluted in 0.9% saline, aztreonam was diluted in 7% sodium bicarbonate in water. Table 21 shows formulations used in this study.













TABLE 21









Levofloxacin in Saline

Levofloxacin with MgCl2









Dose (mg/kg)
















32
63
125
32
63
125

















Levofloxacin
1.5
3
6
6
12
24


(mg/mL)


MgCl2 (mM)
0
0
0
12
24
48


Saline (%)
0.9
0.9
0.9
0
0
0









Bacterial strains MIC testing: P. aeruginosa ATCC 27853 and NH57388A were used in these studies. MICs were determined by a broth microdilution assay according to CLSI reference methods (Methods for dilution of antimicrobial susceptibility test for bacteria that grow aerobically. Seventh Edition: Clinical and Laboratory Standards Institute (2006) M&-A7, incorporated by reference in its entirety). Assays were performed in a final volume of 100 μl. The bacterial suspensions were adjusted to yield a cell density of 5×105 CFU/ml. Antibiotics were prepared at a concentration equivalent to twofold the highest desired final concentration in culture medium and were then diluted directly into 96-well microtiter plates. Microtiter plates were incubated for 24 h at 35° C. and were read by using a microtiter plate reader (Molecular Devices) at 600 nm as well as by visual observation by using a microtiter plate reading mirror. The MIC was defined as the lowest concentration of antibiotic at which the visible growth of the organism is completely inhibited.


Mice: Female Swiss mice (5-6 wk of age) were obtained from Harlan West Coast (Germantown, Calif.). All studies were performed under protocols approved by an Institutional Animal Care and Use Committee.


Preparation of pseudomonal alginate: P. aeruginosa NH57388A was cultured in 50 ml Mueller-hinton broth (MHB) for 24-28 h at 37° C. with shaking (170 rpm). Bacterial cells were harvested by centrifugation (23,000×g, 30 min, at 4° C.) and resuspended in 3-6 ml of MHB. The supernatant was collected and placed in 80° C. water-bath for 30 min. Alginate was precipitated by adding the supernatant to 150 ml of ice-cold 99% ethanol. The precipitated alginate was collected with a sterile bacterial loop and washed several times in sterile saline. The purified alginate was then resuspended in 10 ml of sterile saline and stirred vigorously to form a homogeneous suspension. The alginate concentration was measured and adjusted to a concentration of 2-3 mg/ml.


Aerosol Administration of antibiotics: Antibiotics were aerosolized using a microspray aerosol device (MicroSprayer Model IA-C, PennCentury, Philadelphia, Pa.) attached to a FMJ-250 High-Pressure Syringe (PennCentury, Philadelphia, Pa.). This device produces a 16-22 μM Mass Medium Diameter spray. For administration, each mouse was anesthetized (5% isoflurane in oxygen running at 4 L/min) and positioned securely at a 45-50° angle by the upper teeth, the microspray aerosol tip was inserted to the bifurcation and a 50 μl volume was administered.


Pharmacokinetics: Mice (n=3/timepoint) were administered single 60 mg/kg aerosol dose of levofloxacin formulated with MgCl2 or a 20 mg/kg IP dose of levofloxacin. Mice were sacrificed at 0.08, 0.16, 0.25, 0.5, 0.75, 1.0, 2.0, 3.0, and 4.0 h after dosing and their lungs collected. Levofloxacin lung homogenate concentrations administered as levofloxacin or levofloxacin formulated with MgCl2 were measured using an HPLC method. Analytical standards (0.05 to 100 mg/L) were prepared in fresh mouse lung homogenate collected from untreated animals. Lung homogenate or standards for both compounds were mixed with double the volume of 4% trichloroacetic acid, vortexed and then centrifuged at 12,000 rpm for 10 min using a refrigerated Eppendorf 5415c centrifuge set at 4-10° C. Aliquots of the supernatant (25 μl) were injected directly onto the HPLC using a temperature-controlled autoinjector set at 10° C. Standard curves were constructed of the peak area versus standard concentration, and the data were fit using weighted linear regression (Microsoft Excel, Seattle, Wash.). The concentrations of levofloxacin in the lung homogenate were calculated from these standard curves. The lung pharmacokinetic parameters were determined using WinNonlin (Pharsight, Mountain View, Calif.).


Acute Mouse Lung Infection Model: P. aeruginosa ATCC 27853 was grown overnight in MHB at 35° C. The bacterial suspensions were adjusted to approximately 1-6×105 CFU/ml by correlation of the absorbance at 600 nm with predetermined plate counts. Female Swiss mice were made neutropenic by the intraperitoneal (IP) injection of 150 mg/kg cyclophosphamide (Baxter, Deerfield) on days 1 and 3. On day 4, mice were infected by intratracheal (IT) instillation of 0.05 ml of inoculum using a curved oral gavage tip attached to a 1 ml syringe. Antibiotic treatments started 24 h post-infection and were administered once or twice daily for 24 or 48 h. Antibiotics were aerosolized using a microspray aerosol device. All infections and aerosol treatments were performed under isoflurane anesthesia (5% isoflurane in oxygen running at 4 L/min). An untreated group of mice (n=8) was sacrificed prior to the initiation of treatment to determine baseline bacterial counts. The treated animals (n=8) were sacrificed 12-16 h following the last antibiotic dose by carbon dioxide asphyxiation. The lungs were removed aseptically and homogenized (Pro200 homogenizer, Pro Scientific, Monroe, Conn.) in 1 ml of sterile saline. Serial 10-fold dilutions of the homogenized lung were plated on Mueller-hinton agar (MHA), and colonies counted. For survival studies, mice (n=10) were observed for 7 days after the end of treatment or a total of 9 days post-infection.


Chronic Mouse Lung Infection Model: P. aeruginosa NH57388A was cultured in 50 ml MHB for 24-28 h at 37° C. with shaking (170 rpm). Bacterial cells were harvested by centrifugation (23,000×g, 30 min, at 4° C.) and resuspended in 3-6 ml of MHB (Hoffmann, N. T. B. et al. 2005. Novel mouse model of chronic Pseudomonas aeruginosa lung infection mimicking cystic fibrosis. Infect Immun 73:2504-14, incorporated herein by reference in its entirety). The bacterial suspension was diluted (1:10) in the alginate suspension to yield about 108 CFU/ml. Initial establishment of infection was achieved by a transient neutropenia using a single 150 mg/kg IP dose of cyclophosphamide 4 days prior to infection. On day 4, the mice were infected using a curved bead-tipped oral gavage attached to a 1 ml syringe while under isoflurane anesthesia. Antibiotic treatments started 24 h post-infection and were administered twice daily for three consecutive days with various concentrations of antibiotics either by the IP route or by aerosol using a microspray device. 12-16 h following the last treatment, mice were sacrificed and colony counts in the lung determined as described herein.


Statistical Analysis: Survival and lung bacterial counts were analyzed by log-rank and the Mann-Whitney U test (GraphPad Prism version of 4.03), respectively. A P value of <0.05 was considered statistically significant.


Minimal Inhibitory Concentration of Antibiotics


The minimal inhibitory concentration (MIC) of the P. aeruginosa strains used in animal studies are shown in Table 22. Tobramycin was the most potent antibiotic in vitro, with MICs of <1 μg/ml, levofloxacin formulated with MgCl2 and levofloxacin had MICs of 1 and 2 μg/ml, and aztreonam had MICs of 4 μg/ml against both strains











TABLE 22









MIC (μg/ml)












Levofloxacin






P. aeruginosa

formulated


strain
with MgCl2
Levofloxacin
Tobramycin
Aztreonam














ATCC27853
1
1
0.25
4


NIH57388A
2
2
0.5
4


CF










Mouse Pharmacokinetics


Normalized lung pharmacokinetic parameters for levofloxacin formulated with MgCl2 and levofloxacin are shown in Table 23. Aerosol administration of 60 mg/kg levofloxacin formulated with MgCl2 produced values for levofloxacin AUC and Cmax that were 9 and 30-fold higher than those achieved with dose normalized intraperitoneal administration of levofloxacin.













TABLE 23








Levofloxacin
Levofloxacin




formulated
formulated



Parameter
with MgCl2
in Saline









Route of Administration
Aerosol
IP



Dose (mg/kg)
60
20



Cmax (mg/kg)
550
6.2 (18.6)



(Normalized to 60 mg/kg)



AUC (hr · mg/kg)
106
4.1 (12.3)



(Normalized to 60 mg/kg)











Aerosol Levofloxacin Formulated with MgCl2 vs. Systemic Levofloxacin in Acute and Chronic Lung Infection Models


In the acute lung infection model, aerosol treatment with 125, 62.5, and 32 mg/kg of levofloxacin formulated with MgCl2 produced 5.9, 4.3, and 2.3 log CFU reductions in lung bacterial counts, respectively (FIG. 16). Systemic treatment with 125, 62.5, and 32 mg/kg of levofloxacin produced 3.5, 2.7, and 0.65 log CFU reductions, respectively. The reduction in bacterial counts with aerosol levofloxacin formulated with MgCl2 was greater than that observed with IP levofloxacin on a per dose basis (p<0.05).


In the chronic lung infection model, intraperitoneal treatment with 60, 30, and 15 mg/kg of levofloxacin in saline produced a 0.15, 0.32, and 0.83 log increase in bacterial counts, respectively (FIG. 17). In contrast, aerosol dosing with 60, 30, and 15 mg/kg of levofloxacin formulated with MgCl2 produced 1.26, 0.62, and 0.07 log decreases in bacterial counts, respectively. Overall, bacterial load in the lung was significantly lower in mice treated with aerosolized levofloxacin formulated with MgCl2 compared to systemic levofloxacin on a dose per dose basis in both infection models (p<0.05 for levofloxacin formulated with MgCl2 vs. systemic levofloxacin).


Aerosol Levofloxacin, Tobramycin, and Aztreonam in an Acute Lethal Lung Infection Model


To compare the effects of levofloxacin formulated with MgCl2, tobramycin, and aztreonam in the acute lung infection model, mice were infected with P. aeruginosa ATCC 27853 and treated by the aerosol route twice a day for 2 consecutive days. Due to toxicity, tobramycin was limited to a 60 mg/kg maximum dose and aztreonam was limited to 400 mg/kg maximum dose. In addition, due to the need for anesthesia for treatment, the maximum number of daily doses was limited to two.


As shown in FIG. 18, aerosol dosing with levofloxacin formulated with MgCl2, tobramycin, and aztreonam produced mean reductions of 4.10, 2.70, and 0.24 log CFU per lung, respectively (p<0.05 for comparisons of levofloxacin formulated with MgCl2 with aztreonam). Notably, administration of the same total daily dose of levofloxacin formulated with MgCl2 as single or twice daily doses resulted in similar reductions in P. aeruginosa counts in the lung.


Survival was monitored over 9 days. As shown in FIG. 19, all untreated mice succumbed to the infection after 3 days. Treatment with 800 mg/kg/day (400 mg/kg BID) aerosolized aztreonam had the lowest survival rate among the antibiotics used in this study (20%) and was not significantly different from untreated mice (p>0.05). Treatment with 120 mg/kg/day (60 mg/kg BID) tobramycin produced a 60% survival rate which was statistically different than controls (p<0.05). Treatment with 120 mg/kg/day levofloxacin formulated with MgCl2 as either 120 mg/kg QD or 60 mg/kg BID produced 100% survival which was significantly different from untreated controls or aztreonam (p<0.05), but not significantly different from tobramycin (p=0.056).


Aerosol Levofloxacin, Tobramycin, and Aztreonam in a Chronic Lung Infection Model


Aerosolized levofloxacin formulated with MgCl2, tobramycin and aztreonam produced mean log CFU reductions of 3.3, 2.9, and 1.25, respectively (FIG. 20). Aerosolized doses of either tobramycin or levofloxacin formulated with MgCl2 produced significantly lower bacterial counts compared to aztreonam, or untreated control groups (p<0.05).


These in vivo studies show that aerosol dosing of levofloxacin formulated with MgCl2 produces greater antibacterial killing than systemic dosing in both acute and chronic P. aeruginosa lung infection models. Notably, twice daily dosing with levofloxacin formulated with MgCl2 reduced the lung bacterial load by an extent similar to or greater than that observed with aerosolized tobramycin and aztreonam (FIG. 18). This reduction in bacterial load in the lungs translated to improved survival (FIG. 19).


In addition, comparisons of single-versus twice-daily dosing of levofloxacin formulated with MgCl2 showed comparable bacterial killing and survival, suggesting that once-daily treatment with levofloxacin formulated with MgCl2 may be possible in patients. Once daily administration of a medicament is particularly advantageous over multiple administrations, where multiple administrations are inconvenient to patients and can result in poor adherence to treatment.


While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.


All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.

Claims
  • 1. A method of treating a Mycobacterium pulmonary infection in a subject in need thereof, the method comprising administering to the lungs of the subject an aerosol of a solution comprising from about 75 mg/ml to about 150 mg/ml levofloxacin or ofloxacin and from about 160 mM to about 240 mM of a divalent or trivalent cation, wherein the solution has a pH from about 5 to about 7 and an osmolality from about 300 mOsmol/kg to about 500 mOsmol/kg.
  • 2. The method of claim 1, wherein the Mycobacterium is Mycobacterium tuberculosis, Mycobacterium avium, Mycobacterium intracellulare, and Mycobacterium leprae, Chlamydia pneumoniae, Mycoplasma pneumoniae.
  • 3. The method of claim 2, wherein the Mycobacterium is Mycobacterium avium, Mycobacterium intracellulare, or a combination thereof.
  • 4. The method of claim 1, wherein the levofloxacin or ofloxacin is present at a concentration from about 80 mg/ml to about 120 mg/ml.
  • 5. The method of claim 1, wherein the solution consists essentially of from about 90 mg/ml to about 110 mg/ml levofloxacin and from about 180 mM to about 220 mM of a divalent or trivalent cation.
  • 6. The method of claim 1, wherein the solution has a pH from about 6.0 to about 6.5 and an osmolality from about 350 mOsmol/kg to about 400 mOsmol/kg.
  • 7. The method of claim 1, wherein the solution consists of 80 mg/ml to 120 mg/ml levofloxacin and from about 160 mM to about 240 mM of a divalent or trivalent cation.
  • 8. The method of claim 1, wherein the divalent or trivalent cation is selected from magnesium, calcium, zinc, copper, aluminum, and iron.
  • 9. The method of claim 1, wherein the divalent or trivalent cation is selected from magnesium chloride, magnesium sulfate, zinc chloride, and copper chloride.
  • 10. The method of claim 1, wherein the aqueous solution lacks a conventional pharmaceutical carrier or excipient selected from the group consisting of mannitol, lactose, starch, magnesium stearate, sodium saccharine, talcum, cellulose, sodium crosscarmellose, glucose, gelatin, sucrose, and magnesium carbonate.
RELATED APPLICATIONS

This application is a continuation of U.S. application Ser. No. 15/635,818 filed Jun. 28, 2017, issued as U.S. Pat. No. 10,149,854, which is a continuation of U.S. application Ser. No. 15/132,122 filed Apr. 18, 2016, issued as U.S. Pat. No. 9,717,738, which is a continuation of U.S. application Ser. No. 14/333,583 filed Jul. 17, 2014, issued as U.S. Pat. No. 9,326,936, which is a continuation of U.S. application Ser. No. 12/574,680 filed Oct. 6, 2009, issued as U.S. Pat. No. 8,815,838, which claims priority to U.S. Application No. 61/103,501 filed Oct. 7, 2008, which is hereby expressly incorporated by reference in its entirety.

US Referenced Citations (254)
Number Name Date Kind
2587215 Priestly Feb 1952 A
2858691 Hoffman Nov 1958 A
2868691 Irving Jan 1959 A
3014844 Thiel Dec 1961 A
3456644 Thiel Jul 1969 A
3456645 Brock Jul 1969 A
3456646 Phillips Jul 1969 A
3507277 Altounyan Apr 1970 A
3565070 Hanson Feb 1971 A
3598294 Hedrick Aug 1971 A
3635219 Altounyan Jan 1972 A
3636949 Kropp Jan 1972 A
3669113 Altounyan Jun 1972 A
3732864 Thompson May 1973 A
3789843 Armstrong Feb 1974 A
3807400 Cocozza Apr 1974 A
3826255 Havstad Jul 1974 A
3906950 Cocozza Sep 1975 A
3948264 Wilke Apr 1976 A
3971377 Damani Jul 1976 A
3991761 Cocozza Nov 1976 A
4013075 Cocozza Mar 1977 A
4046146 Rosskamp Sep 1977 A
4147166 Hansen Apr 1979 A
4253468 Lehmbeck Mar 1981 A
4263907 Lindsey Apr 1981 A
4268460 Boiarski May 1981 A
4353365 Hallworth Oct 1982 A
4382892 Hayakawa May 1983 A
4470412 Nowacki Sep 1984 A
4510929 Bordoni Apr 1985 A
4517359 Kobrehel May 1985 A
4534345 Wetterlin Aug 1985 A
4624251 Miller Nov 1986 A
4648393 Landis Mar 1987 A
4649911 Knight Mar 1987 A
4664107 Wass May 1987 A
4667668 Wetterlin May 1987 A
4688218 Blineau Aug 1987 A
4730000 Chu Mar 1988 A
4790305 Zoltan Dec 1988 A
4805811 Wetterlin Feb 1989 A
4807814 Douche Feb 1989 A
4809692 Nowacki Mar 1989 A
4811731 Newell Mar 1989 A
4832015 Nowacki May 1989 A
4844902 Grohe Jul 1989 A
4857311 Domb Aug 1989 A
4889144 Tateno Dec 1989 A
4907538 Helmle et al. Mar 1990 A
4926852 Zoltan May 1990 A
4955371 Zamba Sep 1990 A
4977154 Sanchez Dec 1990 A
4985557 Hayakawa Jan 1991 A
4994599 Chu Feb 1991 A
5012803 Foley May 1991 A
5012804 Foley May 1991 A
5024467 Truchet Jun 1991 A
5027806 Zoltan Jul 1991 A
5033463 Cocozza Jul 1991 A
5040527 Larson Aug 1991 A
5053407 Hayakawa Oct 1991 A
5060643 Rich Oct 1991 A
5113855 Newhouse May 1992 A
5119806 Palson Jun 1992 A
5142046 Hayakawa Aug 1992 A
5164740 Ivri Nov 1992 A
5217004 Blasnik Jun 1993 A
5284133 Burns Feb 1994 A
5304559 Rozier Apr 1994 A
5334589 Al-Razzak Aug 1994 A
5347998 Hodson Sep 1994 A
5364838 Rubsamen Nov 1994 A
5385140 Smith Jan 1995 A
5388572 Mulhauser Feb 1995 A
5404781 Chen Apr 1995 A
5404871 Goodman Apr 1995 A
5427089 Kraemer Jun 1995 A
5437270 Braithwaite Aug 1995 A
5478578 Arnold Dec 1995 A
5508269 Smith Apr 1996 A
5532239 Pruna Jul 1996 A
5549102 Lintl Aug 1996 A
5563155 Domagala Oct 1996 A
5586550 Ivri Dec 1996 A
5642730 Baran Jul 1997 A
5645049 Foley Jul 1997 A
5688792 Barbachyn Nov 1997 A
5694920 Abrams Dec 1997 A
5709202 Lloyd Jan 1998 A
5740794 Smith Apr 1998 A
5756506 Copeland May 1998 A
5758637 Ivri Jun 1998 A
5775320 Patton Jul 1998 A
5785049 Smith Jul 1998 A
5816240 Komesaroff Oct 1998 A
5820873 Choi Oct 1998 A
5823179 Grychowski Oct 1998 A
5829434 Ambrosio Nov 1998 A
5840279 Narodylo Nov 1998 A
5906202 Schuster May 1999 A
5918594 Asking Jul 1999 A
5934272 Lloyd Aug 1999 A
5960792 Lloyd Oct 1999 A
5971951 Ruskewicz Oct 1999 A
5988160 Foley Nov 1999 A
6003512 Gerde Dec 1999 A
6006747 Eisele Dec 1999 A
6026807 Puderbaugh Feb 2000 A
6026809 Abrams Feb 2000 A
6029662 Marcon Feb 2000 A
6070575 Gonda Jun 2000 A
6083922 Montgomery Jul 2000 A
6161536 Redmon Dec 2000 A
6192876 Denyer Feb 2001 B1
6196219 Hess Mar 2001 B1
6223746 Jewett May 2001 B1
6230706 Gonda May 2001 B1
6264922 Wood Jul 2001 B1
6268489 Allen Jul 2001 B1
6288080 Barsuhn Sep 2001 B1
6294178 Weinstein Sep 2001 B1
6333044 Santus Dec 2001 B1
6333045 Yasueda Dec 2001 B1
6338443 Piper Jan 2002 B1
6349719 Gonda Feb 2002 B2
6350199 Williams Feb 2002 B1
6367470 Denyer Apr 2002 B1
6406880 Thornton Jun 2002 B1
6427682 Klimowicz Aug 2002 B1
6435177 Schmidt Aug 2002 B1
6468967 Oleson, Jr. Oct 2002 B1
6492328 Lehrer Dec 2002 B2
6503953 Vyden Jan 2003 B2
6518239 Kuo Feb 2003 B1
6523536 Fugelsang Feb 2003 B2
6543442 Gonda Apr 2003 B2
6544555 Rudnic Apr 2003 B2
6557549 Schmidt May 2003 B2
6561186 Casper May 2003 B2
6576224 Osbakken Jun 2003 B1
6579854 Mitchell Jun 2003 B1
6584971 Denyer Jul 2003 B1
6585958 Keller Jul 2003 B1
6586008 Batycky Jul 2003 B1
6601581 Babaev Aug 2003 B1
6605609 Barbachyn Aug 2003 B2
6608078 De Souza Aug 2003 B2
6612303 Grychowski Sep 2003 B1
6626173 Genova Sep 2003 B2
6644304 Grychowski Nov 2003 B2
6663890 Rudnic Dec 2003 B2
6663891 Rudnic Dec 2003 B2
6664239 Mitchell Dec 2003 B2
6667042 Rudnic Dec 2003 B2
6667057 Rudnic Dec 2003 B2
6669948 Rudnic Dec 2003 B2
6672304 Casper Jan 2004 B1
6681768 Haaije De Boer Jan 2004 B2
6689769 Gordeev Feb 2004 B2
6716819 Welsh Apr 2004 B2
6723341 Rudnic Apr 2004 B2
6730320 Rudnic May 2004 B2
6756369 Mitchell Jun 2004 B2
6806256 Ulrich Oct 2004 B2
6835372 Kuo Dec 2004 B2
6838552 Mitchell Jan 2005 B1
6869965 Gordeev Mar 2005 B2
6878713 De Souza Apr 2005 B2
6884784 Mitchell Apr 2005 B1
6890526 Stratton May 2005 B2
6962151 Knoch Nov 2005 B1
6987094 Malvolti Jan 2006 B2
7148404 Hoegenhaug Dec 2006 B2
7838532 Surber Nov 2010 B2
7893020 Glinka Feb 2011 B2
8357696 Surber Jan 2013 B2
8524734 Surber Sep 2013 B2
8524735 Surber Sep 2013 B2
8546423 Surber Oct 2013 B2
8629139 Dudley Jan 2014 B2
8815838 Griffith Aug 2014 B2
9326936 Griffith May 2016 B2
9700564 Loutit Jul 2017 B2
9717738 Griffith Aug 2017 B2
9951013 Zankel Apr 2018 B2
10149854 Griffith Dec 2018 B2
10231975 Loutit Mar 2019 B2
20010049366 Singh Dec 2001 A1
20020061281 Osbakken May 2002 A1
20020086867 Dubois Jul 2002 A1
20020142050 Straub Oct 2002 A1
20020197212 Osbakken Dec 2002 A1
20030012814 Rudnic Jan 2003 A1
20030028025 Raghavan Feb 2003 A1
20030032600 Ulrich Feb 2003 A1
20030078517 Kensey Apr 2003 A1
20030138403 Drustrup Jul 2003 A1
20030143265 Araki et al. Jul 2003 A1
20030171340 Isbister Sep 2003 A1
20030186894 Kuo Oct 2003 A1
20040009126 Pilkiewicz Jan 2004 A1
20040009989 Niddam-Hildesheim Jan 2004 A1
20040014750 Michaelis Jan 2004 A1
20040025876 Miller Feb 2004 A1
20040037781 McCormack, Jr. Feb 2004 A1
20040045546 Hirsh Mar 2004 A1
20040152701 Reddy Aug 2004 A1
20050036951 Henderson Feb 2005 A1
20050106151 Shapiro May 2005 A1
20050139211 Alston Jun 2005 A1
20050147567 Kuo Jul 2005 A1
20050208124 Araki Sep 2005 A1
20050235987 Smaldone Oct 2005 A1
20050260099 Xia Nov 2005 A1
20050288302 Niddam-Hildesheim Dec 2005 A1
20060003944 Glinka Jan 2006 A1
20060025355 Duddu Feb 2006 A1
20060062738 Hofmann Mar 2006 A1
20060223751 Mygind Oct 2006 A1
20060258677 Amir Nov 2006 A1
20060276416 Sinclair Dec 2006 A1
20060276463 Sharma Dec 2006 A1
20060276473 Bostion Dec 2006 A1
20060276483 Surber Dec 2006 A1
20060276563 Osterod Dec 2006 A1
20060286574 Romesberg Dec 2006 A1
20070003753 Asgari Jan 2007 A1
20070071686 Lintz Mar 2007 A1
20070155715 Van Duzer Jul 2007 A1
20070197548 Murthy Aug 2007 A1
20070248693 Mazzio Oct 2007 A1
20080276935 Wang Nov 2008 A1
20090025713 Keller Jan 2009 A1
20090197212 Masen Aug 2009 A1
20090247458 Watson Oct 2009 A1
20100037890 Surber Feb 2010 A1
20100040560 Surber Feb 2010 A1
20100087386 Dudley Apr 2010 A1
20100087416 Griffith Apr 2010 A1
20100158957 Surber Jun 2010 A1
20100166673 Surber Jul 2010 A1
20100204470 Wieser Aug 2010 A1
20120035166 Dudley Feb 2012 A1
20120121593 Levy May 2012 A1
20120237564 Dudley Sep 2012 A1
20120276153 Loutit Nov 2012 A1
20140066441 Surber Mar 2014 A1
20140105985 Dudley Apr 2014 A1
20140329810 Griffith Nov 2014 A1
20160279138 Surber Sep 2016 A1
20160287606 Griffith Oct 2016 A1
20170029376 Zankel Feb 2017 A1
20180085462 Dudley Mar 2018 A1
Foreign Referenced Citations (88)
Number Date Country
0892357 Jul 1982 BE
2440412 Sep 2002 CA
2739897 Apr 2010 CA
2773033 Mar 2011 CA
1312076 Apr 2007 CN
101222927 Jul 2008 CN
0047005 Mar 1982 EP
206283 Dec 1986 EP
0211595 Feb 1987 EP
0298650 Jan 1989 EP
0347779 Dec 1989 EP
0455463 Nov 1991 EP
0467172 Jan 1992 EP
0470667 Feb 1992 EP
0855183 Jul 1998 EP
1092430 Apr 2001 EP
1319399 Jun 2003 EP
1459739 Sep 2004 EP
1223915 Dec 2005 EP
2344129 Jul 2011 EP
2346509 Jul 2011 EP
901107 Jul 1962 GB
60202822 Oct 1985 JP
63188627 Aug 1988 JP
2003513046 Apr 2003 JP
2003300882 Oct 2003 JP
2004277431 Oct 2004 JP
2004535370 Nov 2004 JP
2008540676 Nov 2008 JP
2009526003 Jul 2009 JP
2012505222 Mar 2012 JP
2012505223 Mar 2012 JP
2013502416 Jan 2013 JP
2013502579 Jan 2013 JP
2013503907 Feb 2013 JP
6099609 Mar 2017 JP
2126000 Feb 1999 RU
628930 Oct 1978 SU
8705213 Sep 1987 WO
9007351 Jul 1990 WO
9013327 Nov 1990 WO
9209322 Jun 1992 WO
9312831 Jul 1993 WO
9324165 Dec 1993 WO
9507271 Mar 1995 WO
9511666 May 1995 WO
9623485 Aug 1996 WO
9703649 Feb 1997 WO
9723217 Jul 1997 WO
9803217 Jan 1998 WO
1999059566 Nov 1999 WO
9962495 Dec 1999 WO
2000018388 Apr 2000 WO
2001002024 Jan 2001 WO
2001032181 May 2001 WO
0224167 Mar 2002 WO
2002018345 Mar 2002 WO
0227167 Apr 2002 WO
2002072102 Sep 2002 WO
03030868 Apr 2003 WO
2003035030 May 2003 WO
2003066064 Aug 2003 WO
2003075889 Sep 2003 WO
2004019912 Mar 2004 WO
2004069253 Aug 2004 WO
2005035036 Apr 2005 WO
2005037256 Apr 2005 WO
2005089738 Sep 2005 WO
2006011051 Feb 2006 WO
2006033713 Mar 2006 WO
2006078925 Jul 2006 WO
2006100875 Sep 2006 WO
2006125132 Nov 2006 WO
2007085057 Aug 2007 WO
2007090123 Aug 2007 WO
2007090646 Aug 2007 WO
2007095156 Aug 2007 WO
2007095187 Aug 2007 WO
2008025560 Mar 2008 WO
2009044202 Apr 2009 WO
2009140587 Nov 2009 WO
2010042549 Apr 2010 WO
2010042553 Apr 2010 WO
2010124141 Oct 2010 WO
2011022074 Feb 2011 WO
2011022075 Feb 2011 WO
2011029059 Mar 2011 WO
2014032184 Mar 2014 WO
Non-Patent Literature Citations (414)
Entry
“Fungal Lung Disease,” In Breathing in America: Diseases, Progress, and Hope, chapter 9, pp. 92 and 95, published online in 2010 by the American Thoracic Society, accessed on Jan. 2, 2013 at http://www.thoracic.org/education/breaething-in-america/resources/chapter- -9-fungal-lung-disease.pdf.
“Hypersensitivity Pneumonitis,” by the American Lung Association, Retrieved from the Internet at http://www.lung.org/lung-disease/hypersensitivity-pneumonitis; 1 page, 2015.
“Physician's Desk Reference”, 52.sup.nd ed., Medical Economics, Montvale, N.J. (1998).
“Understanding Sarcoidosis” by the American Lung Association. Retrieved from the Internet at http://www.lung.org/lung-disease/sarcoidosis/understanding-sarcoidosis.ht-ml; 4 pages, 2015.
Abusriwil et al. “The interaction of host and pathogen factors in chronic obstructive pulmonary disease exacerbations and their role in tissue damage”, Proc. Am. Thorac. Soc. (2007) 4(8):611-617.
Aggarwal et al., “Predictors of mortality and resource utilization in cirrhotic patients admitted to the medical ICU,” Chest, vol. 119, No. 5, (May 2001), pp. 1489-1497.
Ambrose, et al., “Pharmacokinetics—Pharmacodynamics of Antimicrobial Therapy: It's Not Just for Mice Anymore,” Antimicrobial Resistance, vol. 44, (Jan. 1, 2007); pp. 79-86.
Amsden, “Anti-Inflammatory Effects of Macrolides—an Underappreciated Benefit in the Treatment of Community-Acquired Respiratory Tract Infections and Chronic Inflammatory Pulmonry Conditions?”, Journal of Antimicrobial Chemotherapy, 55:10-21, (2005).
Anonymous, “Mpex Candidate, MP-376, Granted U.S. Orphan Drug Status for the Treatment of Cystic Fibrosis”, Medical News Today, Internet Citation, Mar. 5, 2008, 3 pages, XP002560239, Retreived from the Internet: URL: http://www.medicalnewstoday.com/articles/99488.php.
Anonymous, “MPEX Pharmaceuticals Initiates Multi-Dose Clinical Trial in the U.S. with MP-376 in Patients with Cystic Fibrosis” Science Letter (2007) 2 pages.
Anonymous, MP-376 safe and effective for treatment of P. aeruginosa in CF patients, May 16, 2010 at http://www.eurekalert.org/pub.sub.--releases/2010-05/ats-msa051010.php; 2 pages.
Araujo, et al., “Effect of Moxifloxacin on Secretion of Cytokines by Human Monocytes Stimulated with Lipopolysaccharide”, Clin. Microbiol. Infect., 8:26-30, (2002).
Araujo, et al., “Gemifloxacin Inhibits Cytokine Secretion by Lipopolysaccharide Stimulated Human Monocytes at the Post-Transcriptional Level”, Clin. Microbiol. Infect., 10:213-9, (2004).
Arzte, Zeitung De, www.aerztezeitunq.de/extras/druckansicht/?sid=347342&pid=351267 (retrieved online Dec. 11, 2009), XP002560241. (Machine Translation Provided); 6 pages.
Atkins et al., “The Design and Development of Inhalation Drug Delivery Systems”, Pharmaceutical Inhalation Aerosol Technology, Marcel Dekker, Inc., New York, NY (1992) 6: p. 155-185.
Australian Patent Examination Report No. 1, corresponding to Australian Patent Application No. 2010289326, dated May 7, 2014; 4 pages.
Australian Patent Examination Report No. 1, corresponding to Australian Patent Application No. 2014203364, dated Jul. 6, 2015; 4 pages.
Australian Patent Examination Report No. 1, dated May 5, 2014, corresponding to to Australian Patent Application No. 2010238765; 4 pages.
Australian Patent Examination Report No. 1, dated Jul. 25, 2014, corresponding to Australian Patent Application No. 2009302478; 4 pages.
Australian Patent Examination Report No. 1, dated Jun. 9, 2015, corresponding to Australian Patent Application No. 2013203605; 4 pages.
Baker, et al., “A Prodrug Approach Toward the Deveolpment of Water Soluble Fluoroquinolnes and Structure-Activity Relationships of Quinolone-3-Carboxylic Acids”, J. Med. Chem., 47:4693-709, (2004).
Banerjee et al., “The treatment of respiratory pseudomonas infection in cystic fibrosis: what drug and which way?” Drugs (2000) 60(5):1053-64. (Abstract Only).
Barry, et al., “Novel Agents in the Management of Mycobacterium Tuberculosis Disease”, Current Medical Chemistry (Netherlands), 14(18):2000-8, (2007), (Abstract only).
Bartlett, J., “Overview of Pneumonia”, Merck Manual Home Edition article accessed at<http://www.merckmanuals.com/home/lung-and-airway-disorders/pneumonia/overview-of-pneumonia>; 5 pages.
Bartlett, Clinical Microbiology, “Anaerobic bacterial infection of the lung,” Anaerobe 18 (Elsevier) (2012); pp. 235-239.
Battram, et al., “In Vitro and In Vivo Pharmacological Characterization of 5-[(R)-2(5,6-diethyl-indian-2ylamino)-1-hydroxy-ethyl]-8-hydroxy-1H-quinolin-2-one (indacterol), a Novel Inhaled Beta(2) Adrenoceptor Agonist with a 25-h Duration of Action”, J. Pharmacol. Exp. Ther., 317(2):762-70, (2006), (Abstract only).
Beasley, et al., “Adverse Reactions to the Non-Drug Constituents of Nebuliser Solutions”, Br. J. clin. Pharmac., 25:283-7, (1988).
Benko, et al., “Pharmacokinetics and Pharmacodynamics of Levofloxacin in Critically Ill Patients with Ventilator-Associted Pneumonia”, International Journal of Antimicrobial Agents (Netherlands), 30(2):162-8, (2007), (Abstract only).
Berg, “Combination products are spotlighted at Drug/Device Summit,” Pulmonary drug delivery systems,The BBI Newsletter (May 1, 2005); 2 pages.
Bide, et al., “Allometric Respiration/Body Mass DAta for Animals to be Used for Estimates of Inhalation Toxicity to Young Adult Humans,” Journal of Applied Toxicology, (J. Appl. Toxicol.) vol. 20, (2000); pp. 273-290.
Blaser et al., “Influence of Medium and Method on the In Vitro Susceptibility of Pseudomonas Aeruginosa and Other Bacteria to Ciprofloxacin and Enoxacin Antimicrobial Agents and Chemotherapy”, American Society for Microbiology (1986) 29(5):927-929.
Blau, et al., “Moxifloxacin but not Ciprofloxacin or Azithromycin Selectively Inhibits IL-8, IL-6,ERK1/2, JNK, and NF-kB Activation in a Cystic Fibrosis Epithelial Cell Line,” American Journal of Physiology—Lung Cellular and Molecular, vol. 292, (Jan. 2007); pp. L343-L352.
Blitz, et al., “Aerosolized Magnesium Sulfate for Acute Asthma: A Systematic Review”, Chest the Cardiopulmonary and Critical Care Journal, 128(1):337-44, (Jul. 2005).
Boehnke, et al., “High-dose riboflavin treatment is efficacious in migraine prophylaxis: an open study in a tertiary care centre,” European Journal of Neurology, vol. 11, (2004); pp. 475-477.
Braga, et al., “Chem. Commun., Making Crystals from Crystals: A Green Route to Crystal Engineering and Polymorphism”, 3635-45, (2005).
Brouillard, et al., “Antibiotic Selection and Resistance Issues with Flouroquinolones and Doxycycline Against Bioterrorism Agents”, Pharmacotherapy, Special Article, United States, 26(1):3-14, (2006).
Bryskier, “Bacillue anthracis and antibacterial agents,” Clinical Microbiology and Infection—the official publication of the European Society of Clinical Microbiology and Infectious Diseases (France), vol. 8, No. 8, (2002) pp. 467-478.
Burger, AM. Preclinical Screening for New Anticancer Agents. Springer. 2014, p. 23.
Calbo, E. et al., “Systemic expression of cytokine production in patients with severe pneumococcal pneumonia: Effects of treatment with a beta-lactam versus a fluoroquinolone”, Antimicrobial Agents and Chemotherapy, (2008), 52(7):2395-402, ISSN 0066-4804, XP002560242.
Canadian Office Action and Examination Search Report dated Aug. 11, 2015, corresponding to Candian Patent Application No. 2,739,897; 3 pages.
Canadian Office Action dated Jul. 29, 2014, corresponding to Canadian Application No. 2,608,273; 2 pages.
Canadian Office Action dated Jul. 31, 2015, corresponding to Canadian Patent Application No. 2,739,893; 3 pages.
Carratala, et al., “Clinical Experience in the Management of Community-Acquired Pneumonia: Lessons from the Use of Fluoroquinolones”, Clinical Microbiology and Infection—The Official Publication of the European Society of Clinical Microbiology and Infectious Diseases (France), 12(3):2-11, (2006), (Abstract only).
Cazzola, et al., “Delivering antibacterials to the lungs: considerations for optimizing outcomes”, Am. J. Respir. Med. (2002) 1(4):261-272.
Celli et al., “The body-mass index, airflow obstruction, dyspnea, and exercise capacity index in chronic obstructive pulmonary disease”, N Engl J Med. (2004) 350(10):1005-1012.
Chang et al., “Properties of the Drug Molecule in Nasal Systemic Drug Delivery,” 1989, pp. 49-51, Chapter 3, Marcel Dekker, Inc.
Chhabra, et al., “Evaluation of Three Scales of Dyspnea in Chronic Obstructive Pulmonary Disease,” Annals of Thoracic Medicine, vol. 4, No. 3, (2009); pp. 128-132.
Chien, Yie W., et al., “Propeties of the Drug Molecule in Nasal Systemic Drug Delivery”, Chapter 3, pp. 63-68, Marcel Dekker, Inc., (1989).
Chilean Office Action (no English translation), dated Mar. 5, 2014, corresponding to Chilean Patent Application No. 2011-002649; 9 pages.
Chilean Office Action (with no English translation), corresponding to Chilean Patent Application No. 00586-2012, dated Dec. 5, 2014; 8 pages.
Chilean Office Action; 9 pages.
Chilean Second Examination Report (No English translation), dated Jun. 1, 2015, corresponding to Chilean Patent No. 00586-2012; 4pages.
Chilean Second Examination Report (No English translation), dated Nov. 3, 2014, corresponding to Chilean Patent Application No. 2011-02649; 6 pages.
Chinese First Office Action, (with English translation) dated Apr. 6, 2010, corresponding to Chinese Patent Application No. 200680026156.0; 9 total pages.
Chinese Office Action (no English translation), dated Dec. 10, 2014, corresponding to Chinese Patent Application No. 200980142471.3; 3 pages.
Chinese Office Action (No English Translation), dated Sep. 15, 2014, corresponding to Chinese Patent Application No. 201080018022.0; 5 pages.
Chinese Office Action (with English translation) dated Mar. 3, 2015, corresponding to Chinese Patent Application No. 200680026156.0; 14 pages.
Chinese Office Action (with English translation), dated Sep. 1, 2014, corresponding to Chinese Patent Application No. 200680026156.0; 7 pages.
Chinese Office Action (with no English translation), corresponding to Chinese Patent Application No. 201080048091.6, dated Jul. 25, 2014; 6 pages.
Chinese Office Action (with No English translation), dated Feb. 12, 2015, corresponding to Chinese Application No. 201080048091.6; 7 pages.
Chodosh S., “Clinical Significance of the Infection-Free Interval in the Management of Acute Bacterial Exacerbations of Chronic Bronchitis,” Chest, 127(6):2231-6, (Jun. 2005).
Choi et al., “Effect of moxifloxacin on production of proinflammatory cytokines from human peripheral blood mononuclear cells”, Antimicrobial Agents and Chemotherapy (Dec. 2003) 47(12):3704-3707.
Christ-Crain, M. et al., “Biomarkers in Respiratory Tract Infections: Diagnostic Guides to Antibiotic Prescription, Prognostic Markers and Mediators”, Eur Respir J., 30(3):556-73, (2007).
Chung, K. et al., “Multifaceted Mechanisms in COPD: Inflammation, Immunity, and Tissue Repair and Destruction”, Eur Respir J., 31(6):1334-56, (2008).
Cigana, et al., “Azithromycin Selectively Reduces Tumor Necrosis Factor Alpha Levels in Cystic Fibrosis Airway Epithelial Cells,” Antimicrobial Agents and Chemotherapy, vol. 51, No. 3, (Mar. 2007); pp. 975-981.
Clancy, et al., “Results of a phase IIa study of VX-809, an investigational CFTR corrector compound, in subjects with cystic fibrosis homozygous for the F508del-CFTR mutation,” Thorax 2012, vol. 67; pp. 12-18.
ClinicalTrials.gov archive, Linking patients to medical research, A service of the U.S. National Institutes of Health, “Phase II, Multi-Center, Randomized, Double-Blind, Placebo-Controlled, Study to Evaluate the Safety, Tolerability and Efficacy of Three Dosage Regimens of MP-376 Solution for Inhalation Given for 28 Days to Stable CF Patients,” (May 13, 2008); 7 pages.
Conrad, “Mpex 204 Phase 2,” Stanford School of Medicine (retrieved online Dec. 11, 2009), Sep. 3, 2008, pp. 1-7 (PCT ISR/WO provided a partial reference, and the full reference is no longer available); 4 pages.
Conte et al., “Intrapulmonary pharmacodynamics of high-dose levofloxacin in subjects with chronic bronchitis or chronic obstructuctive pulmonary disease”, International Journal of Antimicrobial Agents, Elsevier Science, Amsterdam, NL (2007) 30:422-427.
Cooney, et al., “Absolute Bioavailability and Absorption Characteristics of Aerosolized Tobramycin in Adults with Cystic Fibrosis”, The Journal of Clinical Pharmacology, 34:255-9, (1994).
Crombleholme, MD, William R. “Preeclampsia-Eclampsia” Ch. 18 Obstetrics In: Current Medical Treatment and Diagnosis. 37th edited by Tierney, Jr. Lawrence M., McPhee, Stephen J., Papadakis, Maxine A.; Appleton and Lange, Stamford, CT 1998 pp. 731-734, (1998); (PDF is from 2004 43rd edition).
Dal Negro et al., “Tobramycin Nebulizer Solution in Severe COPD Patients Colonized with Pseudomonas Aeruginosa: Effects on Bronchial Inflammation” Advances in Therapeutics (2008) vol. 20 pp. 1019-1030.
Dalhoff et al., “Immunomodulatory effects of quinolones,” The Lancet Infectious Diseases, vol. 3, (Jun. 2003); pp. 359-371.
Dalhoff, “Immunomodulatory activities of fluoroquinolones”, Infection (2005) 33(Suppl 2):55-70.
Den Hollander, et al., “Synergism Between Tobramycin and Ceftazidime Against a Resistant Pseudomonas aeruginosa Strain, Tested in an In Vitro Pharmacokinetic Model,” Antimicrobial Agents and Chemotherapy, vol. 41, No. 1, (Jan. 1997); pp. 95-100.
Derbacher, et al., “Physical Properties of Nebulized Solutions”, Poster (1994) 381-382 (English Translation included).
DeRyke, et al., “Pharmacodynamic target attainment of six beta-lactams and two fluoroquinolones against Pseudomonas aeruginosa, Acinetobacter baumannii, Escherichia coli, and Klebsiella species collected from United States intensive care units in 2004,” Pharmacotherapy (United States), vol. 27, No. 3, (Mar. 2007), pp. 333-342; Abstract Only.
Deterding, R. et al., “Phase 2 Randomized Safety and Efficacy Trial of Nebulized Denufosol Tetrasodiumin Cystic Fibrosis”, Am J Respir Crit Care Med, 176(4):362-9.
Diakov, et al., “The Chemotherapeutic Efficacy of Ciprofloxacin and Lomefloxacin in the Inhalation Method of Infecting White Mice with Tularemia., Khimioterapevticheskaia Effektivnost′ Tsiprofloksatsina i Lomefloksatsina Pri Ingaliatsionnom Sposobe Zarazheniia Tuliaremiei Belykh Myshei”, Antibiotiki i Khimioterapii a = Antibiotcs and Chemotherapy sic / Ministerstvo Meditsinskoi i Mikrobiologicheskoi Promoyshlennosti SSSR (Russia), 45(6):17-20, (2000), (Abstract only).
Djurdjevic et al, “Study of Solution Equilibria Between Gadolinium (III) Ion and Moxifloxacin” Acta Chim. Slov. 2010, 57, pp. 386-397.
Djurdjevic, P. et al., “Study of solution equilibria between aluminum(III) ion and ofloxacin”, Journal of Pharmaceutical and Biomedical Analysis, vol. 19(3-4):501-510, (Mar. 1999).
Donnarumma, et al., “Anti-inflammatory Effects of Moxifloxacin and Human Beta-Defensin 2 Association in Human Lung Epithelial Cell Line (A549) Stimulated with Lipopolysaccharide”, Peptides, 28:2286-92, (2007).
Doring et al., “Antibiotic therapy against Pseudomonas aeruginosa in cystic fibrosis: a European consensus” [comment in Eur Respir J. Oct. 2000; 16(4):585-7], Euro Respir J. Oct. 2000; 16(4):749-67. (Abstract Only).
Drevensek, et al., “X-Ray Crystallographic, NMR and Antimicrobial Activity Studies of Magnesium Complexes of Flouroquinolones—Racemic Ofloxacin and it's S-form, Levofloxacin”, Journal of Inorganic Biochemistry, 100:1755-63, (2006).
Drevensek, et al., “Influence of Copper(II) and Magnesium(II) ions on the Ciprofloxacin Binding to DNA,” Journal of Inorganic Biochemistry, vol. 96, (2003); pp. 407-415.
Drusano et al., “Pharmacodynamics of a Fluoroquinolone Antimicrobial Agent in a Neutropenic Rat Model of Pseudomonas Sepsis” Antimicrob. Agents & Chemother. (Mar. 1993) 37(3):483-490.
Dudley, M.N. et al. (Dec. 2008, e-published Nov. 25, 2008). “Aerosol antibiotics: considerations in pharmacological and clinical evaluation,” Curr Opin Biotechnol 19(6):637-643.
Elizur, et al., “Airway Inflammation in Cystic Fibrosis”, Chest, 133:489-95, (2008).
English language translation of WO 2006/100875 A1 obtained on Dec. 17, 2012.
English translation of Israeli First Examination Report, corresponding to Israeli Applicaiton No. 218458, dated Dec. 8, 2014; 3 pages.
English Translation summary of Japanese Office Action, corresponding to Japanese Application No. 2012-528109, dated Aug. 19, 2014; 3 pages.
EPO, Office Action dated Oct. 20, 2010 from European Patent Application 06 760 146.8-2123 filed on May 18, 2008.
European Communication dated Jun. 11, 2015, corresponding to European Patent Application No. 06760146.8; 5 pages.
European Communication/Examination Report, dated Jul. 7, 2015, corresponding to European Patent Application No. 10814595.4; 4 pages.
European Communication/Examination Report, dated Jul. 17, 2015, corresponding to European Patent Application No. 10767800.5; 5 pages.
European Office Action (Decision to Refuse a European Patent Application), dated Jul. 15, 2014, corresponding to European Application No. 09 793 326.1; 12 pages.
European Search Report dated Nov. 6, 2013 and European Communication dated Dec. 9, 2013, corresponding to European Application No. 12007354.9; 6 total pages.
European Search Report, dated Dec. 17, 2012, corresponding to European Application No. 10810290.6; 5 pages.
European Search Report, dated, Oct. 4, 2013, corresponding to European Application No. 10810291.4; 6 pages.
Extended European Search Report, dated Jan. 8, 2014, corresponding to European Patent Application No. 10 76 7800.5; 5 pages.
Extended Supplemental European Search Report, dated Apr. 17, 2013, corresponding to European Application No. 10814595.4; 9 pages.
File, Jr., “A New Dosing Paradigm: High-Dose, Short-Course Fluoroquinolone Therapy for Community-Acquired Pneumonia,” Clinical Cornerstone—Supplemental 3, (2003); pp. S21-S28.
Flume et al., “Cystic Fibrosis Pulmonary Guidelines: Chronic Medications for Maintenance of Lung Health,” Am J Respir Crit Care Med (2007) 176:957-969.
Fuchs et al., “Effect of Aerosolized Recombinant Human DNase on Exacerbations of Respiratory Symptoms and on Pulmonary Function in Patients with Cystic Fibrosis” The New England Journal of Medicine, Sep. 8, 1994, vol. 331, No. 10 pp. 637-642.
Garrity et al., “Bergey's Manual of Systematic Bacteriology,” Editor-in-chief: Garrity, George M. Boone, David R.; Castenholz, Richard W. (Eds.) Originally published by Williams & Wilkins, 1984, 2nd ed. (2001).
Gavilanes, et al., “Azithromycin Fails to Reduce Increased Expression of Neutrophil-Related Cytokines in Primary-Cultured Epithelial Cells from Cystic Fibrosis Mice”, J. Cystic Fibrosis, 10(1016):1-8, (2009).
Geddes, D. M., “5. Bronchiectasis and Cystic Fibrosis” Airways Obstruction (published 1981 by MTP Press Ltd) pp. 40-50.
Geller et al., “Levofloxacin Inhalation Solution (MP-376) in Patients with Cystic Fibrosis with Pseudomonas aeruginosa”, Am J Respir Crit Care Med, (20110000), vol. 183, pp. 1510-1516, XP055403234 [X] 7, (8-10)/7 * ; abstract, p. 1511, col. 1, para 4, p. 1511, col. 2, para 6 * [Y] (8-10)/(1-6), 59.
Geller, D.E. et al. (2008). “A Phase 1 Safety, Tolerability and Pharmacokinetic (PK) Study of MP-376 (Levofloxacin Solution for Inhalation) in Stable Cystic Fibrosis (CF) Patients,” Pediatr Pulmonol Suppl 31, Abstract 321, 2 pages.
Geller, D.E. et al., “Pharmacokinetics and Safety of MP-376 (Levofloxacin Inhalation Solution) in Cystic Fibrosis Subjects”, Antimicrobial agents and chemotherapy, vol. 55, No. 6, Jun. 1, 2011, pp. 2636-2640, XP55031818, ISSN: 0066-4804, DOI: 10.1128/AAC.01744-10.
Gennaro, “Remington: Practice of The Science and Pharmacy”, 19.sup.th ed., Williams & Williams, (1995).
Gibaldi et al., “Pharmacokinetics”, 2nd Edition, Marcel Dekker: New York (1982).
Goh, et al., “Current Status of Topical Nasal Antimicrobial Agents,” The Laryngoscope, vol. 110 (Jun. 2000); pp. 875-880.
Griese, et al., “Amphotericin B and Pulmonary Surfactant,” European Journal of Medical Research,vol. 3, No. 8, (Aug. 18, 1998); pp. 383-386—(Abstract Only).
Griffith et al., “Pharmacodynamics of Levofloxacin Against Pseudomonas Aeruginosa with Reduced Susceptibility Due to Different Efflux Pumps: Do Elevated MICs Always Predict Reduced in Vivo Efficacy?” Antimicrobial Agents and Chemotherapy, May 2006, vol. 50 No. 5 pp. 1628-1632.
Griffith, D. et al, “Pharmacokinetics and Safety of MP 376 (Levofloxacin solution for inhalation) in Normal Healthy Volunteers and Cystic fibrosis Patients”, Pediatr. Pulmonol., (Aug. 29, 2007), vol. 42, No. S30, p. 303, XP002560243.
Griffith, et al., “Efficacy of Fluoroquinolones Against Leptospira Interrogans in a Hamster Model”, Antimicrobial Agents and Chemotherapy (United States), 51(7):2615-2617, (2007), (Abstract only).
Griffith, et al., (C1-1954) “In vitro Activity of Levofloxacin (LVX) and Other Antibiotics Administered by the Aerosol Route in Cystic Fibrosis (CF) Against Pseudomonas aeruginosa (Pa) Under Anaerobic Conditions,” IDSA, Oct. 27, 2008; Abstract Only.
Griffith, et. al., “Single-Dose Pharmacokenetics of Aerosol MP-376 (Levofloxacin Solution for Inhalation) in Cystic Fibrosis Patients: PK-PD implacations”, Journal of Cystic Fibrosis, (Jun. 2008), abstract 104(7):S26, (abstract).
Guina, et al., “Quantitative Proteomic Analysis Indicates Increased Synthesis of a Quinolone by Pseudomonas Aeruginosa Isolates from Cystic Fibrosis Airways”, PNAS, 100(5):2771-6, (Mar. 4, 2003).
Handbook of Pharmaceutical Excipients, published jointly by the American Pharmaceutical Association and The Pharmaceutical Society of Great Britain (The Pharmaceutical Press, 1986).
Hart et al., “Cross-over assessment of serum bactericidal activity of moxifloxacin and levofloxacin versus penicillin-susceptible and penicillin-resistant Streptococcus pneumoniae in healthy volunteers”, Diagnostic Microbiology and Infectious Disease (United States) (Jul. 2007) 58(3):375-8. (Abstract Only).
Harutyunyan, “Mpex Pharmaceuticals Presents Data on MP-376 in Cystic Fibrosis” EmaxHealth Oct. 24, 2008 (http://www.emaxhealth.com/2/95/25752/mpex-pharmaceuticals-presents-data--mp-376-cystic-fibrosis.html).
Hashimoto, et al., “Grepafloxacin Inhibits Tumor Necrosis Factor-alpha-induced Interleukin-8 Expression in Human Airway Epithelial Cells,” Life Sciences, vol. 66, No. 5, (2000); pp. 77-82—(Abstract Only).
Hecht, et al., “In Vitro Activities of 15 Antimicrobial Agents Against 110 Toxigenic Clostridium difficile Clinical Isolates Collected from 1983 to 2004,” Antimicrobial Agents and Chemotherapy, vol. 51, No. 8, (Aug. 2007); pp. 2716-2719.
Heine, et al., Major Article, “Comparison of 2 Antibiotics That Inhibit Protein Synthesis for the Treatment of Infection with Yersinia pestis Delivered by Aerosol in a Mouse Model of Pneumonic Plague,” Journal of Infectious Diseases, vol. 196, No. 5, (2007); pp. 782-787.
Hodson, “Antibiotic Treatment: Aerosol Therapy,” Chest, vol. 94, (1988); pp. 156S-160S.
Hoffmann, et al., “Novel Mouse Model of Chronic Pseudomonas Aeruginosa Lung Infection Mimicking Cystic Fibrosis”, Infect. Immun., 73(4):2504-14, (2005).
Honeybourne, “Antibiotic Penetration in the Respiratory Tract and Implications for the selection of Antimicrobial Therapy”, Current Opinion of Pulmonary Medicine, 3(2):170-4, (1997), (Abstract only).
Hoogkamp-Korstanje, “In-Vitro Activities of Ciprofloxacin, Levofloxacin, Iomefloxacin, Ofloxacin, Pefloxacin, Sparfloxacin and Travofloxacin Against Gram-Positive and Gram-Negative Pathogens from Respiratory Tract Infections”, Journal of Antimicrobial Chemotherapy, 40:427-31, (1997).
Horiguchi, et al., “Usefulness of sparfloxacin against Chlamydia pneumoniae infection in patients with bronchial asthma,” Journal of International Medical Research (England), vol. 33, No. 6, (Nov.-Dec. 2005); pp. 668-676, (Abstract Only).
Hrkach, et al., “Synthesis of poly(L-lactic acid-co-L-lysine) Graft Copolymers,” Macromolecules, vol. 28, No. 13, (1995); pp. 4736-4739—(Abstract Only).
Huang et al., “Oxidation of fluoroquinolone antibacterials and structurally related amines with manganese oxide” National Meeting—American Chemical Society Division of Environmental Chemistry (2003) 43(2)(5):1257-1260.
Huang, H. et al., “Comparing the Protective Effects of Ciprofloxacin, Moxifloxacin and Levofloxacin in Mice with Lipopolysaccharide-Induced Acute Lung Injuries”, Respirology, 13(6):47-52, (Jan. 2008).
Hung, et al., “Evaluation of Two Commercial Jet Nebulisers and Three Compressors for the Nebulisation of Antibiotics,” Archives of Diesase in Childhood, 71(4):335-8, (Oct. 1994).
Hutschala, et al., “In Vivo Measurement of Levofloxacin Penetration into Lung Tissue: CPB versus OPCAB”, European Journal of Anaesthesiology, 49(12):5107-11, (2005).
Indian First Examination Report (English translation only), dated Oct. 15, 2013, corresponding to Indian Patent Application No. 9505/DELNP/2007; 3 pages.
International Application No. PCT/US2016/044607; International Preliminary Report on Patentability dated Jan. 30, 2018; 9 pages.
International Application No. PCT/US2016/044607; International Search Report and Written Opinion of the International Search Authority, dated Dec. 12, 2016; 14 pages.
International Application No. PCT/US2017/016161; International Preliminary Report on Patentability (Ch I), dated Aug. 7, 2018; 9 pages.
International Application No. PCT/US2017/016161; International Search Report and Written Opinion of the International Search Authority, dated Jun. 9, 2017; 12 pages.
International Patent Application No. PCT/US2010/032128; International Preliminary Report on Patentability, dated Oct. 25, 2011; 8 pages.
International Patent Application No. PCT/US2010/032128; International Search Report and Written Opinion of the International Search Authority, dated Jul. 30, 2010; 11 pages.
International Preliminary Report on Patentability, dated Apr. 12, 2011 and Written Opinion of the International Searching Authority and International Search Report, dated Dec. 17, 2009, corresponding to International Patent Application No. PCT/US2009/059744; 15 total pages.
International Preliminary Report on Patentability, dated Aug. 19, 2008 and Written Opinion of the International Searching Authority and International Search Report, dated Oct. 25, 2007, corresponding to International Patent Application No. PCT/US2007/003649; 21 total pages.
International Preliminary Report on Patentability, dated Aug. 31, 2010 and Written Opinion of the International Searching Authority and International Search Report, dated Jan. 21, 2010, corresponding to International Patent Application No. PCT/US2009/059740; 18 total pages.
International Preliminary Report on Patentability, dated Feb. 21, 2012 and Written Opinion of the International Searching Authority and International Search Report, dated Oct. 20, 2010, corresponding to International Patent Application No. PCT/US2010/002307; 17 total pages.
International Preliminary Report on Patentability, dated Mar. 6, 2012 and Written Opinion of the International Searching Authority and International Search Report, dated Dec. 7, 2010, corresponding to International Patent Application No. PCT/US2010/047903; 23 total pages.
International Preliminary Report on Patentability, dated Nov. 20, 2007 and Written Opinion of the International Searching Authority and International Search Report, dated Oct. 20, 2006, corresponding to International Patent Application No. PCT/US2006/019351; 15 total pages.
International Search Report and Written Opinion corresponding to International Application No. PCT/US2010/002306, dated Nov. 8, 2010 by the Astralian Patent Office in its capacity as International Searching Authority; 10 total pages.
IPOS Search Report, Written Opinion and Invitation to Response to Written Opinion, dated Aug. 12, 2010 for Singapore Patent Application No. 200717702-5.
Israel Official Action (No English Translation), dated Dec. 8, 2013, corresponding to Israeli Patent Application No. 212190; 4 pages.
Israeli Examination Report (English translation only), dated Sep. 11, 2014, corresponding to Israeli Patent Application No. 215777; 2 pages.
Israeli Office Action (with No English tranlsation), dated Jan. 5, 2015, corresponding to Israeli Patent Application No. 212190; 3 pages.
Jacquot et al., “Airway epithelial cell inflammatory signalling in cystic fibrosis,” The International Journal of Biochemistry & Cell Biology (2008) 40:1703-15.
Japanese Decision of Rejection (English translation only), dated Jun. 5, 2014, corresponding to Japanese Application No. 2011-531125; 3 pages.
Japanese Decision of Rejection (with English Translation), dated Apr. 15, 2015, corresponding to Japanese Patent No. 2012-528109; 5 pages.
Japanese Decision of Rejection (with English translation), dated Nov. 6, 2014, corresponding to Japanese Patent Application No. 2011-531126; 5 total pages.
Japanese Decision of Rejection with English translation, dated Jun. 1, 2015, corresponding to Japanese Patent Application No. 2012-507400; 4 total pages.
Japanese Notice of Reasons for Rejection (English Translation only), dated Dec. 24, 2013, corresponding to Japanese Patent Application No. 2011-531126; 6 pages.
Japanese Notice of Reasons for Rejection (with English translation), dated Apr. 27, 2015, corresponding to Japanese Patent Application No. 2014-095077; 10 total pages.
Japanese Office Action (with English translation), dated Jun. 10, 2014, corresponding to Japanese Patent Application No. 2012-507400; 5 pages.
Japanese Office Action dated Mar. 11, 2014 (with English translation), corresponding to Japanese Application No. 2013-002399; 7 total pages.
Japanese Office Action dated Oct. 20, 2014 (with English translation), corresponding to Japanese Application No. 2013-002399; 6 pages.
Jarraud, et al., “Legionnaires Disease (Legionellose)”, Presse Medicale (Paris, France—1983) (France), 36(2 parts):279-87, (2007), (Abstract only).
Jensen, et al., “The efficacy and safety of ciprofloxacin and ofloxacin in chronic Pseudomonas aeruginosa infection in cystic fibrosis,” Journal of Antimicrobial Chemotherapy, vol. 20, No. 4, (1987); pp. 585-594.
Jones and Helm, “Emerging Treatments in Cystic Fibrosis,” Drugs 2009, vol. 69, No. 14; pp. 1903-1910.
Jones et al., “Quantifying of severity of exacerbations in chronic obstructive pulmonary disease: adaptations to the definition to allow quantification”, Proc Am Thorac Soc. (2007) 4(8):597-601.
Jones, et al., “St. George's Respiratory Questionnaire Manual”, St. George's University of London, (Jun. 2009), 17 pages.
Jumbe et al., “Application of a Mathematical Model to Prevent In Vivo Amplification of Antibiotic-Resistant Bacterial Populations During Therapy” The Journal of Clinical Investigation, Jul. 2003, vol. 112, No. 2, pp. 275-285.
Kays, et al., “Levofloxacin treatment failure in a patient with fluoroquinolone-resistant Streptococcus pneumoniae pneumonia”, Pharmacotherapy, Mar. 2002, vol. 22, No. 3, pp. 395-399.
Kearns, et al., “Poster No. 88: Levofloxacin Pharmacokinetics (PK) after Administration of MP-376 (Levofloxacin Inhalation Solution; Aeroquin.RTM.) in Children with Cystic Fibrosis (CF)” 34th European Cystic Fibrosis Conference Jun. 8-11, 2011.
Khan, et al., “Effect of Travofloxacin on Production of Cytokines by Human Monocytes”, Antimicrobial Agents and Chemotherapy, 42(7):1713-7, (1998).
Khan, et al., “Protection Against Lipopolysaccharide-Induced Death by Fluoroquinolones”, Antimicrobial Agents and Chemotherapy, 44(11):3169-73, (2000).
King et al, “In Vitro Pharmacodynamics of Levofloxacin and Other Aerosolized Antibiotics under Multiple Conditions Relevant to Chronic Pulmonary Infection in Cystic Fibrosis” Antimicrobial Agents and Chemotherapy, Jan. 2010, vol. 54, No. 1, pp. 143-148.
King et al., “Effect of oxygen limitation on the in vitro activity of levofloxacin and other antibiotics administered by the aerosol route against Pseudomonas aeruginosa from cystic fibrosis patients”, Diagn Microbiol Infect Dis. Feb. 2010;66(2)181-6. Epub Oct. 13, 2009.
Kitazawa, et al., “Biophasic Regulation of Levofloxacin on Lipopolysaccharide-Induced IL-1B Production”, Life Sciences, 80:1572-7, (2007).
Kobayashi, et al., “Antibacterial Activity of Tosufloxacin Against Major Organisms Detected by Patients with Respiratory Infections”, Japanese Journal of Antibiotics (Japan), 60(2):98-106, (2007), (Abstract only).
Kohyama et al., “Fourteen-member macrolides inhibit interleukin-8 release by human eosinophils from atopic donors,” Antimicrobial Agents and Chemotherapy, vol. 43, No. 4, (Apr. 1999); pp. 907-911.
Korean Office Action (English translation only), dated Aug. 27, 2014, corresponding to Korean Patent Application No. 10-2007-7029629; 1 page.
Korean Office Action (with English translation) dated Aug. 29, 2014, corresponding to Korean Patent Application No. 10-2007-7016750; 7 pages.
Korean Office Action (with English translation) dated Mar. 28, 2014, corresponding to Korean Application No. 10-2007-7029629; 8 pages.
Korean Office Action, (No English translation), dated Oct. 31, 2014, corresponding to Korean Patent Applcation No. 10-2014-7002452; 3 pages.
Kraynack, et al., “Improving Care at Cystic Fibrosis Centers Through Quality Improvement”, Seminars in Respiratory and Critical Care Medicine, 30(5):547-58, (2009).
Kuhn, “Formulation of aerosolized therapeutics”, Chest, The Cardiopulmonary and Critical Care Journal (2001) 120(3):94S-98S. (Abstract Only).
Kurosaka et al., “DX-619, a novel Des-F(6)-quinolone: Pharmacodynamics (PD) Activity and Thereapeutic Efficacy in Animal Infection Models”, 43rd Interscience Conference on Antimicrobial Agents and Chemotherapy (p. 241 43rd ICAAC Abstracts) ( Sep. 14-17, 2003) Chicago, Illinois.
Lacy et al, Drug Information Handbook American Pharmaceutical Association—1999-2000, Lexi-Comp Inc.: Hudson, Ohio pp. 589-590 and 749-750.
Lammerts Van Bueren et al.: “Structural and Thermodynamic Analyses of alpha-L-Fucosidase Inhibitors”, Chembiochem, vol. 11, No. 14, Sep. 24, 2010 (Sep. 24, 2010), pp. 1971-1974, XP009168983, ISSN: 1439-4227 [retrieved on Jul. 27, 2010].
LaPlante, et al., “Fluoroquinolone resistance in Streptococcus pneumoniae: area under the concentration-time curve/MIC ratio and resistance development with gatifloxacin, gemifloxacin, levofloxacin, and moxifloxacin” Antimicrobial Agents and Chemotherapy (United States) (Apr. 2007), vol. 51, No. 4: pp. 1315-1320. (Abstract Only).
Le Conte, et al., “Lung Distribution and Pharmacokinetics of Aerosolized Tobramycin”, American Review of Respiratory Disease, (1993) vol. 147, pp. 1279-1282.
Lee, et al., “Levofloxacin Pharmacokinetics in Adult Cystic Fibrosis”, Chest, vol. 131, No. 3, (2007); pp. 796-802.
Legssyer, et al., “Azithromycin reduces spontaneous and induced inflammation in F508 cystic fibrosis mice,” Respiratory Research, vol. 7, No. 134, (2006); pp. 1-13.
Leiva, et al., “Effects of Telithromycin in In Vitro and In Vivo Models of Lipopolysaccharide-Induced Airway Inflammation,” Chest, vol. 134, No. 1, (Jul. 2008); pp. 20-29.
Leonard et al., “Topical Antibiotic Therapy for Recalcitrant Sinusitis,” The Laryngoscope, vol. 109, No. 4, (Apr. 1999); pp. 668-670.
Lode, N. et al., “Levofloxacin Versus Clarithromycin in COPD Exacerbation: Focus on Exacerbation-Free Interval”, European Respiratory Journal, 24(6):947-53, (2004).
Louie, et al., “Impact of Resistance Selection and Mutant Growth Fitness on the Relative Efficacies of the Streptomycin and Levofloxacin for Plague Therapy”, Antimicrobial Agents and Chemotherapy, 51(8), (Aug. 2007), (Abstract only).
MacMillan Encyclopedia of Physics, vol. 4, Simon & Schuster: London, 1996, pp. 1677.
Mandell, et al., “Safety of fluoroquinolones: An update,” The Canadian Journal of Infectious Diseases, vol. 13, No. 1, Jan.-Feb. 2002; pp. 54-61.
Martin, Physical Pharmacy (4th Edition), Physical Chemical Principles in the Pharmaceutical Sciences, Chapter II, Complexation and Protein Binding, pp. 261-263, and p. 265; Published by Lippincott Williams & Wilkins, Philadelphia, PA, 1993; 6 total pages.
Martinez et al, “Appropriate outpatient treatment of acute bacterial exacerbations of chronic bronchitis”, American Journal of Medicine,, (Jul. 1, 2005), vol. 118, No. 7, ISSN 0002-9343, pp. 39-44, XP005148473.
Matthys, “Inhalation Delivery of Asthma Drugs”, Lung., 168:645-52, (1990), (Abstract only).
Mazurek, H. et al, “Cystic Fibrosis lung disease: infection, inflammation, or both? Helicobacter pylori seroprevalence in patients with cystic fibrosis”, European Respiratory Annual Congress 2006 as accessed Jan. 24, 2014 from http://www.ers-education.org/home/browse-all-content.aspx?idPar-ent=7958.
McCoy, et al., “Inhaled Aztreonam Lysine for Chronic Airway Pseudomonas Aeruginosa in Cystic Fibrosis”, American Journal of Respiratory and Critical Care Medicine, 178:921-8, (2008).
McGraw-Hill Encyclopedia of Science & Technology, 9th edition, McGraw-Hill: New York, 2002, pp. 303.
MedlinePlus Medical Encyclopedia, “Cystic Fibrosis,” at p. 1 accessed on Jul. 11, 2008 at www.nlm.nih.gov/medlineplus/ency/article/000107.htm.
Meguro, et al., “Development and Validation of an Improved, COPD-Specific Version of the St. George Respiratory Questionnaire,” Chest, vol. 132, No. 2, (Aug. 2007); pp. 456-463.
Mexican Office Action (with English translation), dated Jan. 14, 2015, corresponding to Mexican Patent Application No. MX/a/2011/011190; 8 total pages.
Mexican Office Action (with No English tranlsation), corresponding to Mexican Patent Application No. MX/a/2011/003745, dated Dec. 1, 2014; 4 pages.
Mexican Office Action, dated Aug. 12, 2014 (no English translation), corresponding to Mexican Application No. MX/a/2011/007566; 2 pages.
Mexican Office Action, dated Jun. 11, 2014 (no English translation), corresponding to Mexican Application No. MX/a/2011/007566; 2 pages.
Mexican Official Action (no English Translation) dated May 27, 2014, corresponding to Mexican Patent Application No. MX/a/2011/003745; 2 pages.
Miller, et al., “Standardisation of spirometry,” American Thoracic Society/European Respiratory Society (ATS/ERS) Spirometry Standards, Eur Respir J 2005, vol. 26 No. 2; pp. 319-338.
Mohammed, et al., “Intravenous and Nebulised Magnesium Sulphate for Acute Asthma: Systematic Review and Meta-Analysis”, Emergency Medicine Journal, 24:823-30, (2007).
Mori, et al., “Influence of Prescription days by Simultaneous Combined Use of New Quinolone and Drugs Containing Metal Cation”, Japanese Society of Hospital Pharmacists Journal, 35(4):469-72, (1999).
Moss, “Administration of Aerosolized Antibiotics in Cystic Fibrosis Patients”, Chest, 120(3):107S-13S, (2001), (Abstract only).
Movassaghi, M. et al. Total Synthesis and Absolute Stereochemical Assignment of (+)- and (−)-Galbulimima Alkaloid 13-Supplementary Information. JACS Communications. 2006, vol. 128, p. S54.
Movassaghi, M. et al. Total Synthesis and Absolute Stereochemical Assignment of (+)- and (−)-Galbulimima Alkaloid 13. JACS Communications. 2006, vol. 128, p. 8126.
Mpex Pharmaceuticals Presents New Data on MP-376 in Cystic Fibrosis, (http://www2.prnewswire.com/cgi-bin/stories.pl?ACCT=104&STORY=/www/story/-10-23-2008/0004910076&EDate=); Oct. 23, 2008; 4 pages.
Murphy, et al., “Pseudomonas Aeruginosa in Chronic Obstructive Pulmonary Disease”, American Journal of Respiratory and Clinical Care Medicine, (177):853-60, (2008).
Murray, “Lung Inflammation Treatment,” by eHow Health. [retrieved on Mar. 7, 2013]. Retrieved from the Internet at http://www.ehow.com/about.sub.--5417681.sub.--lung-inflammation-treatment-.html; 4 pages.
Nakanishi, et al., “A case of cystic fibrosis in a Japanese student”, Nihon Kyobu Shikkan Gakkai Zasshi (Japan), vol. 33, No. 7, (1995); pp. 771-774—(Abstract Only).
Navarro, et al., “Oral Absorption of Ofloxacin Administered Together with Aluminum”, Antimicrobial Agents and Chemotherapy, 1994, vol. 38, No. 10, p. 2510-2512.
NCBI Bookshelf Glossary, Appendix D, definition of “Microbe,” accessed at http://www.ncbi.nlm.nih.gov/books/NBK54258/; 1 page, 2010.
Neu, “The Effects of Cations Upon the Activity of Quinolone Agents”, Quinolones Bulletin, Reports on Gyrase Inhibitors (1985).
Neu, et al., “In Vitro Activity of S-Ofloxacin,” Antimicrobial Agents and Chemotherapy, American Society for Microbiology, vol. 33, No. 7, (1989); pp. 1105-1107.
New Zealand First Examination Report, dated Sep. 30, 2014, corresponding to New Zealand Patent Application No. 631469; 2 pages.
New Zealand First Examination, dated Oct. 14, 2013, corresponding to New Zealand Patent Application No. 616438; 2 pages.
New Zealand First Examintion Report, dated Feb. 25, 2013, corresponding to New Zealand Patent Application No. 607408; 3 pages.
New Zealand Further Examination Report dated Dec. 10, 2014, corresponding to New Zealand Patent Application No. 616438, 2 pages.
New Zealand Further Examination Report, dated Sep. 5, 2014, corresponding to New Zealand Patent Application No. 607408; 3 pages.
Newman, “Aerosols and the Lung:Clinical and Experimental Aspects,” Butterworth & Co. Ltd., London, England (1984); pp. 197-224.
Noel GJ, et al, Coparative Safety Profile of Levofloxacin in 2523 Children with a Focus on Four Specific Musculoskeletal Disorders. Pediatr. Infect Dis J. Oct. 2007; 26(10):879-91—Abstract only—found in the Internet Oct. 9, 2014—http://www.ncbi.nlm.nih.gov/pubmed/17901792.
Nouira Semir et al, “Once daily oral ofloxacin in chronic obstructive pulmonary disease exacerbation requiring mechanical ventilation: A randomised placebo-controlled trial”, LANCET (North American Edition), (Dec. 15, 2001), vol. 358, No. 9298, ISSN 0099-5355, pp. 2020-2025, XP004805687.
O-Lee et al, “Fluoroquinolone-induced arthralgia and myalgia in the treatment of sinusitis”, Am. J. Rhinol. (2005) 19(4):395-399.
Office Action dated Apr. 27, 2010 from Russian Patent Application No. 2007146972 filed on May 18, 2006.
Ono et al., “Effect of grepafloxacin on cytokine production in vitro”, Journal of Antimicrobial Chemotherapy, (2000) 46:91-94.
Ortho-Mcneil Pharmaceutical, Inc., OMP Division, Text of Proposed Labeling for Levaquin.RTM. (2004) 1-52.
Ortho-McNeil Pharmaceutical, Inc., Package Insert for Levaquin.RTM., (2006) 15 pages.
Palmer, K. et al., “Membrane-Bound Nitrate Reductase is Required for Anaerobic Growth in Cystic Fibrosis Sputum”, J Bacteriol., 189(12):4449-55, (2007).
Pavlinova et al., “Estimation of the Modern Mucolytic Therapy Efficacy in Children Suffering From Mucoviscidosis (two-year experience of dornase alfe application),” Voprosy Sovremennoi Pediatrii, vol. 2007, No. 2, (2007), pp. 102-106—(with English translation of relevant parts).
Pellegrino, et al., “Interpretative strategies for lung function tests,” European Respiratory Journal, vol. 26, No. 5, (2005); pp. 948-968.
Perez, et al., “CFTR Inhibition Mimics the Cystic Fibrosis Inflammatory Profile”, Am. J. Physiol. Lung Cell Mol Physiol, 292(2):383-95, (2007), (Abstract only).
Polenakovik, et al., “The use of ivacaftor in an adult with severe lung disease due to cystic fibrosis,” Journal of Cystic Fibrosis, Elsevier, (2013); pp. 1 and 2.
Preston et al., “Pharmacodynamics of levofloxacin: a new paradigm for early clinical trials,” JAMA. (1998) 279(2):125-129.
Pringle, C., “Influenza,” Merck Manual Home Edition article, accessed at http://www.merckmanuals.com/home/infections/viral-infections/influenza-flu; 8 pages.
Quan, et al., “A Two-Year Randomized, Placebo-Controlled Trial of Dornase Alfa in Young Patients with Cystic Fibrosis with Mild Lung Function Abnormalities”, the Journal of Pediatrics, 139(6):813-20; (Dec. 2001).
Querol-Ribelles, et al., “Discrepancy Between Antibiotics Administered in Acute Exacerbations of Chronic Bronchitis and Susceptibility of Isolated Pathogens in Respiratory Samples: Multicentre Study in Primary Care Setting”, International Journal of Antimicrobial Agents, 28(5):472-6, (Nov. 2006), (Abstract only).
Rapp, “Fluoroquinolone Positioning in Hospital Antimicrobial Stewardship Programs,” U.S. Pharmacist, vol. 32, No. 12, (2007); pp. HS-10 to HS-17 (6 pages).
Ratcliffe et al., “Effects of Magnesium on the Activity of 4-Quinolone Antibacterial Agents”, Journal of Pharmacy and Pharmacology, 1983, p. 61, vol. 35, Supplement Dec. 1983, The Pharmaceutical Society of Great Britain.
Reato, et al., “Immunomodulating effect on antimicrobial agents on cytokine production by human polymorphonuclear neutrophils,” International Journal of Antimicrobial Agents, vol. 23, No. 2, (Feb. 2004); pp. 150-154—(Abstract Only).
Rennard, Stephen I. “COPD: Overview of Definitions, Epidemiology, and Factors Influencing Its Development” Chest / 113 / 4/ Apr. 1998 p. 235S-241S.
Romano et al., “[The use of ofloxacin in cystic fibrosis patients.] Uso dell'ofloxacin nei pazienti con fibrosi cistica”, Minerva Pediatr. (Mar. 1992) 44(3):79-86. (Abstract Only).
Rosell, A. et al., “Microbiologic determinants of exacerbation in chronic obstructive pulmonary disease” Arch Intern Med 165: 2005, p. 891-897 (printed from http://archinte.jamanetwork.com/article.aspx?articleid=486514).
Rosenfeld et al, “Defining a Pulmonary Exacerbation in Cystic Fibrosis” The Journal of Pediatrics, Sep. 2001, vol. 139(3), pp. 359-365 (from http://ovidsp.tx.ovid.com/sp-3.7.1b/ovidweb.cgi).
Ross, et al., “Physicochemical properties of the fluoroquinolone antimicrobials V. effect of fluoroquinolone structure and pH on the complexation of various fluoroquinolones with magnesium and calcium ions,” International Journal of Pharmaceutics, XP25565729 (1993) vol. 93; pp. 121-129.
Russian Office Action (with English translation), corresponding to Russian Patent Application No. 2012111458/15, dated Jan. 26, 2015; 9 pages.
Russian Office Action (with English translation), corresponding to Russian Patent Application No. 2012111458/15, dated Sep. 12, 2014; 16 total pages.
Russian Office Action (with English Translation), dated Apr. 23, 2015, corresponding to Russian Patent Application No. 2011118619/15; 8 total pages.
Sabet et al., “Efficacy of Aerosol MP-378, a levofloxacin inhalation solution, in models of mouse lung infection due to Pseudomonas aeruginosa, Antimicrobial Agents and Chemotherapy”, (2009) 53(9):3923-3928.
Sabet et al., “In-Vivo Antibacterial Activity of Aerosol MP-376 in Mouse Models of Pulmonary Infection”, #288 The 21st Annual North American Cystic Fibrosis Conference (2007) p. 304.
Sagel et al., “Sputum biomarkers of inflammation in cystic fibrosis lung disease”, Proc. Am. Thorac. Soc. (2007) 4:406-417.
Saito, et al., “New drugs used for Infection Disease Synthetic Antibacterials Levofloxacin”, Clinical and Drug Therapy, 13(2):187-92, (1994).
Sakai, et al., “Comparison of the complexation of fluoroquinolone antimicrobials with metal ions by nuclear magnetic resonance spectroscopy,” Journal of Pharmaceutical and Biomedical Analysis, 18 (Elsevier) (1999); pp. 1057-1067.
Salvatore, D. et al. (Jul. 2002). “Effects of salmeterol on arterial oxyhemoglobin saturations in patients with cystic fibrosis,” Pediatr Pulmonol 34(1):11-15.
Sandor, P.S., et al., “Prophylactic Treatment of Migraine With b-Blockers and Riboflavin: Differential Effects on the Intensity Dependence of Auditory Evoked Cortical Potentials” Headache 40(1), Jan. 2000, pp. 30-35.
Sato et al, “Antimicrobial Activity of DU-6859, a New Potent Fluoroquinolone, Against Clinical Isolates” Antimicrobial Agents and Chemotherapy, vol. 36, No. 7, Jul. 1992, p. 1491-1498.
Scheinberg, et al., “Nebulized Antibiotics for the Treatment of Acute Exacerbations of Chronic Rhinosinusitis,” ENT—Ear, Nose & Throat Journal, vol. 81, No. 9, (Sep. 2002), pp. 648-652.
Schoenen, et al., “Effectiveness of high-dose riboflavin in migraine prophylaxis—A randomized controlled trial,” American Accademy of Neurology, vol. 50, No. 2, (Feb. 1998); pp. 466-470.
Seddon, “Pseudopolymorph: a polemic”, Crystal Growth & Design (2004) 4(6):1087, web release date Oct. 19, 2004.
Seeimungal, et al., “Long-term erythromycin therapy is associated with decreased chronic obstructive pulmonary disease exacerbations,” American Journal of Respiratory and Critical Care Medicine, vol. 178, (2008), pp. 1139-1147.
Sethi, et al. “Poster No. 27964: A Phase 2 Study to Evaluate the Safety, Tolerability, and Efficacy of Levofloxacin Inhalation Solution (MP-376) Administered for 5 Days Every 28 Days to Prevent Acute Exacerbations in High Risk COPD Patients” American Thoracic Society 2012 International Conference, May 18-23, 2012; 1 page.
Sethi, et al., “New Strains of Bacteria and Exacerbations of Chronic Obstructive Pulmonary Disease,” The New England Journal of Medicine, vol. 347, No. 7, (Aug. 15, 2002); pp. 465-471.
Shalit, et al., “Anti-Inflammatory Effects of Moxifloxacin on IL-8, IL-1B and TNF-a Secretion and NFkB and MAP-kinase Activation in Human Monocytes Stimulated with Aspergillus Fumigatus”, Journal of Antibacterial Chemotherapy, 57:230-5, (2006).
Shalit, et al., “Immunomodulatory and protective effects of moxifloxacin against candida albicans-induced bronchopneumonia in mice injected with cyclophosphamide,” Antimicrobial Agents and Chemotherapy, vol. 46, No. 8, (2002); pp. 2442-2449.
Shinkai et al., “Clarithromycin has an immunomodulatory effect on ERK-mediated inflammation induced by Pseudomonas aeruginosa flagellin”, Journal of Antimicrobial Chemotherapy (2007) 59:1096-1101.
Shinkai, et al,, “Macrolide antibiotics as immunomodulatory medications: Proposed mechanisms of action,” Pharmacology & Therapeutics, vol. 117, (2008); pp. 393-405.
Singapore Written Opinion and Search Report, dated Jun. 2, 2015, issued by the Intellectual Property Office of Singapore (IPOS), corresponding to Singapore Patent Application No. 201202482-4; 11 total pages.
Skauge et al., “Interaction Between Ciprofloxacin and DNA Mediated by Mg2+-ions”, Inorganica Chimica Acta (2002) 339: 239-247.
Smith et al., “Chemistry and Mechanisms of Action of the Quinolone Antibacterials”, Chapter 2 of “The Quinolones” Academic Press Limited, Harcourt Brace Janovich, Publishers (1988) pp. 23-82.
Smith, “Interactions Between 4-Quinolone Antibacterials and Multivalent Metal Ions,” Microbiology Section, Department of Pharmaceutics, The School of Pharmacy, University of London, Brunswick Square, London, England. Journal of Chemotherapy (Florence, Italy) 1989, 1(4 Suppl): pp. 134-135.
Soler, N. et al, “Airway Inflammation and Bronchial Microbial Patterns in Patients with Stable Chronic Obstructive Pulmonary Diseae” European Respiratory Journal 14 (1999) p. 1015-1022.
Stephenson, “Applications of X-Ray Powder Diffraction in the Pharmaceutical Industry,” The Rigaku Journal, vol. 22, No. 1, (2005); pp. 2-15.
Stockley, et al., “Relationship of Sputum Color to Nature and Outpatient Management of Acute Exacerbations of COPD”, Chest 117(6):1638-45, (Jun. 2000).
Strieter, “Interleukin-8: A Very Important Chemokine of the Human Airway Epithelium”, Journal of Physiology—Lung Cell Molecular Physiology, 283:L688-9, (2002).
Suman, et al., “Comparison of Nasal Deposition and Clearance of Aerosol Generated by a Nebulizer and an Aqueous Spray Pump”, Pharmaceutical Research, 16(10):1648-52, (1999).
Suman, et al., “Validity of In Vitro Tests on Aqueous Spray Pumps as Surrogates for Nasal Deposition,” Pharmaceutical Research, vol. 19, No. 1, (Jan. 2002); pp. 1-6—(Abstract Only).
Suri, R. et al. (Feb. 2007, e-published Jun. 27, 2006). “Assessing the usefulness of outcomes measured in a cystic fibrosis treatment trial,” Respir Med 101(2):254-260.
Suzuki, et al., “Histopathological Study of the Effects of a Single Intratracheal Instillation of Surface Active Agents on Lung in Rats,” The Journal of Toxicological Sciences, vol. 25, No. 1, (2000); pp. 49-55—(Abstract Only).
Tabaru, et al., P4677—“Various Aspects of Respiratory Epidemiology: Helicobacter pylori infection in COPD,” accessed at http://lrp.ersnet.org/abstract.sub.--print.sub.--10/files/407.pdf; p. 858s, 2010.
Takeyama, et al., “The 6-Fluoro-8-Methoxy Quinolone Gatifloxacin Down-Regulates Interleukin-8 Production in Prostate Cell Line PC-3,” Antimicrobial Agents and Chemotherapy, vol. 51, No. 1, (Jan. 2007); pp. 162-168.
Takizawa, et al., “Erthromycin Modulates IL-8 Expression in Normal and Inflamed Human Bronchial Epithelial Cells”, American Journal of Respiratory and Clinical Care Medicine, (156):266-71, (1997).
Tanaka et al., “Antimicrobial Activity of DV-7751a, a New Fluoroquinolone” Antimicrobial Agents and Chemotherapy, Oct. 1993, vol. 37, No. 10, p. 2112-2118.
The Engineering ToolBox, “Dynamic, Absolute, Kinematic Viscosity” accessed online at http://www.engineeringtoolbox.com/dynamic-absolute-kinematic-vi-scosity-d.sub.--412.html (6 pages).
The Engineering Toolbox, “Surface Tension,”—accessed at http://www.engineeringtoolbox.com/surface-tension-d.sub.--962.html (3 pages), 2015.
Tirouvanziam, et al, Rapid Communication—“Inflammation and Infection in Naive Human Cystic Fibrosis Airway Grafts,” American Journal of Respiratory Cell and Molecular biology, vol. 23, (2000); pp. 121-127.
Traczewski et al., “In Vitro Activity of Doripenem Against Pseudomonas aeruginosa and Burkholderia cepacia Isolates from Both Cystic Fibrosis and Non-Cystic Fibrosis Patients,” Antimicrobial Agents and Chemotherapy, (Feb. 2006) 50:819-821.
Tsai, et al., “Azitromycin Blocks Neutrophil Recruitment in Pseudomonas Endobronchial Infection”, Am. J. Respir Crit. Care Med., 170:1331-9, (2004).
Tsapis et al., “Direct lung delivery of para-aminosalicylic acid by aerosol particles” Tuberculosis (Edinburgh, Scotland) (England) (2003) 83(6):379-85. (Abstract Only).
Tu et al.: “Development of fucosyltransferase and fucosidase inhibitors”, Chem. Soc. Rev., Apr. 15, 2013 (Apr. 15, 2013), pp. 1-17, XP009168995, ISSN: 0303-0012, DOI: 10.1039/c3cs60056d [retrieved on Apr. 15, 2013] p. 2, section 1; pp. 12-14, section 7, including figures 11 to 13. Cited as common general knowledge.
Turel et al., “Biological activity of some magnesium(II) complexes of quinolones”, The Synthesis and Biological Activity of Some Magnesium (II) Complexes of Quinolones—Metal-Based Drugs (2000) 7(2):101-104.
Turel, “The Interactions of Metal Ions with Quinolone Antibacterial Agents”, Coordinatuion Chemistry Reviews, 232:27-47, (Oct. 2002).
U.S. Appl. No. 13/278,706; Advisory Action dated Mar. 7, 2016; 4 pages.
U.S. Appl. No. 13/278,706; Advisory Action dated May 12, 2017; 3 pages.
U.S. Appl. No. 13/278,706; Applicant Initiated Interview Summary dated Dec. 12, 2012; 3 pages.
U.S. Appl. No. 13/278,706; Applicant Initiated Interview Summary dated Feb. 5, 2013; 3 pages.
U.S. Appl. No. 13/278,706; Declaration of Michael Dudley, David Griffith, and Olga Rodny Under 37 C.F.R. Section 1.132; signed Feb. 22, 2016; 2 pages.
U.S. Appl. No. 13/278,706; Examiner Initiated Interview Summary dated Sep. 22, 2014; 2 pages.
U.S. Appl. No. 13/278,706; Final Office Action dated Mar. 2, 2017; 24 pages.
U.S. Appl. No. 13/278,706; Final Office Action dated Nov. 2, 2015; 20 pages.
U.S. Appl. No. 13/278,706; Final Office Action dated Oct. 18, 2012; 31 pages.
U.S. Appl. No. 13/278,706; Non-Final Office Action dated Mar. 5, 2015; 15 pages.
U.S. Appl. No. 13/278,706; Non-Final Office Action dated May 23, 2012; 22 pages.
U.S. Appl. No. 13/278,706; Non-Final Office Action dated Sep. 21, 2106; 20 pages.
U.S. Appl. No. 13/278,706; Notice of Allowance dated Feb. 19, 2013; 9 pages.
U.S. Appl. No. 13/278,706; Notice of Appeal dated Aug. 31, 2017; 2 pages.
U.S. Appl. No. 13/278,706; Notice of Appeal dated Feb. 24, 2016; 1 page.
U.S. Appl. No. 15/623,168; Corrected Notice of Allowability, dated Dec. 5, 2018; 8 pages.
U.S. Appl. No. 15/623,168; Non-Final Office Action dated Mar. 12, 2018; 12 pages.
U.S. Appl. No. 15/623,168; Notice of Allowance dated Nov. 1, 2018; 22 pages.
U.S. Appl. No. 13/412,423; Non-Final Office Action dated Apr. 1, 2015; 25 pages.
U.S. Appl. No. 13/412,423; Non-Final Office Action dated May 11, 2016; 18 pages.
U.S. Appl. No. 13/412,423; Notice of Allowance dated Mar. 6, 2017; 4 pages.
U.S. Appl. No. 13/412,423; Notice of Allowance dated Mar. 3, 2017; 8 pages.
U.S. Appl. No. 12/574,666; Applicant Initiated Interview Summary dated Jun. 12, 2013; 3 pages.
U.S. Appl. No. 12/574,666; Final Office Action dated May 17, 2012; 22 pages.
U.S. Appl. No. 12/574,666; Non-Final Office Action dated Mar. 13, 2013; 23 pages.
U.S. Appl. No. 12/574,666; Non-Final Office Action dated Nov. 29, 2011; 21 pages.
U.S. Appl. No. 12/574,666; Notice of Allowance dated Sep. 23, 2013; 12 pages.
U.S. Appl. No. 11/436,875; Applicant Initiated Interview Summary dated Apr. 30, 2010; 1 page.
U.S. Appl. No. 11/436,875; Applicant Initiated Interview Summary dated Jul. 27, 2010; 3 pages.
U.S. Appl. No. 11/436,875; Examiner Interview Summary Report dated Apr. 21, 2010; 4 pages.
U.S. Appl. No. 11/436,875; Examiner Interview Summary Report dated Aug. 5, 2010; 2 pages.
U.S. Appl. No. 11/436,875; Non-Final Office Action dated Feb. 5, 2010; 29 pages.
U.S. Appl. No. 11/436,875; Notice of Allowance dated Aug. 5, 2010; 4 pages.
U.S. Appl. No. 11/436,875; Notice of Allowance dated Aug. 5, 2010; 5 pages.
U.S. Appl. No. 12/574,680; Final Office Action dated Jun. 5, 2012; 26 pages.
U.S. Appl. No. 12/574,680; Non-Final Office Action dated Nov. 4, 2011; 21 pages.
U.S. Appl. No. 12/574,680; Notice of Allowance dated Jun. 17, 2014; 9 pages.
U.S. Appl. No. 12/604,324; Final Office Action dated Nov. 5, 2012; 16 pages.
U.S. Appl. No. 12/604,324; Non-Final Office Action dated Apr. 12, 2012; 27 pages.
U.S. Appl. No. 12/604,324; Notice of Allowance dated Jan. 25, 2013; 8 pages.
U.S. Appl. No. 12/604,324; Notice of Allowance dated May 1, 2013; 9 pages.
U.S. Appl. No. 12/604,340; Non-Final Office Action dated Jan. 7, 2013; 16 pages.
U.S. Appl. No. 12/604,340; Non-Final Office Action dated Sep. 26, 2012; 11 pages.
U.S. Appl. No. 12/604,340; Notice of Allowance dated Jun. 3, 2013; 7 pages.
U.S. Appl. No. 12/604,347; Applicant Initiated Interview Summary dated Jan. 22, 2013; 3 pages.
U.S. Appl. No. 12/604,347; Non-Final Office Action dated Oct. 12, 2012; 14 pages.
U.S. Appl. No. 12/604,347; Notice of Allowance dated Apr. 30, 2013; 10 pages.
U.S. Appl. No. 12/604,347; Notice of Allowance dated Jan. 14, 2013; 10 pages.
U.S. Appl. No. 12/695,981; Final Office Action dated Jul. 5, 2012; 18 pages.
U.S. Appl. No. 12/695,981; Non-Final Office Action dated Nov. 16, 2011; 36 pages.
U.S. Appl. No. 12/695,981; Notice of Allowance dated Nov. 18, 2012; 7 pages.
U.S. Appl. No. 13/412,423; Advisory Action dated Apr. 7, 2016; 3 pages.
U.S. Appl. No. 13/412,423; Final Office Action dated Dec. 17, 2015; 29 pages.
U.S. Appl. No. 13/412,423; Final Office Action dated Oct. 16, 2014; 27 pages.
U.S. Appl. No. 13/412,423; Final Office Action dated Oct. 22, 2013; 21 pages.
U.S. Appl. No. 13/412,423; Non-Final Office Action dated Mar. 22, 2013; 22 pages.
U.S. Appl. No. 14/012,307; Advisory Action dated Feb. 2, 2016; 3 pages.
U.S. Appl. No. 14/012,307; Final Office Action dated Oct. 7, 2015; 26 pages.
U.S. Appl. No. 14/012,307; Non-Final Office Action dated Jun. 16, 2016; 26 pages.
U.S. Appl. No. 14/012,307; Non-Final Office Action dated May 20, 2015; 19 pages.
U.S. Appl. No. 14/012,307; Notice of Appeal dated Jan. 19, 2016; 1 page.
U.S. Appl. No. 14/134,348; Advisory Action dated May 9, 2016; 5 pages.
U.S. Appl. No. 14/134,348; Applicant Initiated Interview Summary dated Sep. 2, 2016; 3 pages.
U.S. Appl. No. 14/134,348; Final Office Action dated Mar. 4, 2016; 13 pages.
U.S. Appl. No. 14/134,348; Final Office Action dated Oct. 13, 2016; 10 pages.
U.S. Appl. No. 14/134,348; Non-Final Office Action dated Jul. 15, 2015; 17 pages.
U.S. Appl. No. 14/134,348; Non-Final Office Action dated Jul. 29, 2016; 11 pages.
U.S. Appl. No. 14/134,348; Notice of Appeal dated Apr. 11, 2017; 1 page.
U.S. Appl. No. 14/333,583; Examiner Initiated Interview Summary dated Mar. 21, 2016; 1 page.
U.S. Appl. No. 14/333,583; Non-Final Office Action dated Nov. 12, 2015; 11 pages.
U.S. Appl. No. 14/333,583; Notice of Allowance dated Mar. 21, 2016; 10 pages.
U.S. Appl. No. 15/132,122; Non-Final Office Action dated Oct. 4, 2016; 16 pages.
U.S. Appl. No. 15/132,122; Notice of Allowance dated Mar. 28, 2017; 9 pages.
U.S. Appl. No. 15/173,372; Final Office Action dated Jul. 17, 2018; 26 pages.
U.S. Appl. No. 15/173,372; Non-Final Office Action dated Nov. 13, 2017; 13 pages.
U.S. Appl. No. 15/225,188; Examiner Initiated Interview Summary dated Dec. 13, 2017; 2 pages.
U.S. Appl. No. 15/225,188; Non-Final Office Action dated Jul. 21, 2017; 15 pages.
U.S. Appl. No. 15/225,188; Notice of Allowance dated Dec. 13, 2017; 9 pages.
U.S. Appl. No. 15/635,818; Non-Final Office Action dated Mar. 28, 2018; 8 pages.
U.S. Appl. No. 15/729,930; Non-Final Office Action dated Mar. 20, 2019; 67 pages.
U.S. Appl. No. 16/250,520; Application as filed dated Jan. 17, 2019; 196 pages.
U.S. Appl. No. 16/263,886; Application as filed dated Jan. 31, 2019; 73 pages.
U.S. Appl. No. 13/412,423; Final Office Action dated Oct. 20, 2016; 16 pages.
U.S. Appl. No. 13/278,706; Non-Final Office Action, dated Dec. 12, 2018; 44 pages.
U.S. Appl. No. 13/412,423; Non-Final Office Action dated Feb. 20, 2014; 20 pages.
Urban C. et al., “Fluoroquinolone-Resistant Streptococcus pneumoniae Associated with Levofloxacin Therapy”, The Journal of Infectious Diseases, 2001, vol. 184, No. 6, pp. 794-798.
Vaughan, “Nebulization of Antibiotics in Management of Sinusitis”, Current Infectious Disease Reports, 6(3):187-90, (2004), (Abstract only).
Vaughan, et al., “Use of Nebulized Antibiotics for Acute Infections in Chronic Sinusitis”, Otolaryngology—Head and Neck Surgery, 127:558-68, (2002).
Villeneuve, et al., “Nebulized Magnesium Sulfate in the Management of Acute Exacerbations of Asthma,” The Annals of Pharmacotherapy, vol. 40, (Jun. 2006); p. 1118—(Abstract Only).
Vippagunta, et al. “Crystalline Solids,” Advanced Drug Delivery Reviews, May 16, 2001, vol. 48, No. 1; pp. 3-26—(Abstract Only).
Wacker, J. et al., “Riboflavin deficiency and preeclampsia”, Obstet Gynecol. 2000;96(1):38-44.
Wada et al., “Immunomodulatory Effect of Gatifloxacin on Mouse Peritoneal Macrophages in vitro and in Models of Endotoxin-Induced Rat Conjunctivitis and Rabbit Bacterial Keratitis,” Opthalmic Research, vol. 40, (2008); pp. 54-60.
Wagner et al., (Clin Rev Allerg Immunol 2008, 35, 124-134).
Wahl et al. “New Medical Management Techniques for Acute Exacerbations of Chronic Rhinosinusitis”, Current Opinion in Otolaryngology & Head and Neck Surgery (2003) 11:27-32.
Wang et al. “Synthesis and crystal structure of a new copper (II) complex containing fluoroquinolone”, Inter'l Symposium on Solid State Chemistry in China; Frontiers of Solid State Chemistry, World Scientific (2002) 327-332.
Wang et al.: “Synthesis and Biological Evaluation of Glycosidase Inhibitors: gem—Difluoromethylenated Nojirimycin Analogues”, J. Med. Chem., vol. 49, No. 10, May 1, 2006 pp. 2989-2997, XP55301961, US ISSN: 0022-2623, DOI: 10,1021/jm060066q p. 2989, left column, paragraphs 1-2; table 1: compound 41.
Weber A. et.al., “Effect of Nebulizer Type and Antibiotic Concentration on Device Performance”, Pediatric Pulmonology, 23(4):249-60, (Apr. 1997).
Weber et al., “Nebulizer Delivery of Tobramycin to the Lower Respiratory Tract”, Pediatric Pulmonology, Wiley-Liss, Inc. (1994) 17: p. 331-339.
Weiss, et al., “Anti-Inflammatory Effects of Moxifloxacin on Activated Human Monocytic Cells: Inhibition of NF-.kappa.B and Mitogen-Activated Protein Kinase Activiation and of Synthesis of Proinflammatory Cytokines”, Antimicrobial Agents and Chemotherapy, 48(6):1974-82, (2004).
Werber, et al., “Moxifloxacin Cytokine-Induced MAP Kinase and NF-.kappa.B Activation as well as Nitric Oxide Synthesis in a Human Respiratory Epithelial Cell Line”, Journal of Antimicrobial Chemotherapy, 55(3):293-300, (2005).
Wilkinson, et al., “Airway Bacterial Load and FEV1 Decline in Patients with Chronic Obstructive Pulmonary Disease”, American Journal of Respiratory and Critical Care Medicine, 167:1090-5, (2003).
Wilkinson, et al., “Effect of Interactions Between Lower Airway Bacterial and Rhinoviral Infection in Exacerbations of COPD,” Chest, vol. 129, No. 2, (Feb. 2006); pp. 317-324.
Williams, Jr., “Fluoroquinolones for respiratory infections: too valuable to overuse”, Chest (2001) 120(6):1771-5.
Wise, R., Merck online Manual Home Edition article, entitled, “Chronic Obstructive Pulmonary Disease,” accessed on Mar. 21, 2010 at www.merck.com/mmhe/print/sec04/ch045/ch045a.html.
Wu et al.: “Rapid Diversity-Oriented Synthesis in Microtiter Plates for In Situ Screening: Discovery of Potent and Selective [alpha]-Fucosidase Inhibitors”, Angew. Chem. Int. Ed., vol. 42, No. 38, Oct. 6, 2003, pp. 4661-4664, XP055301569, DE ISSN: 1433-7851, DOI: 10.1002/anie, left column, paragraph 2; schemes 1 and 2; table 1.
Wu et al.: “Structural basis of alpha-fucosidase inhibition by iminocyclitols with K(i) values in the micro-to picomolar range”, Angew. Chem. Int. Ed., vol. 49, No. 2, Jan. 8, 2010 (Jan. 8, 2010), pp. 337-340, XP009168699, ISSN: 1521-3773 [retrieved on Dec. 3, 2009] cited in the application.
Yamamoto, et al., “Treatment of respiratory and urinary tract infections in elderly inmates at a nursing home by selective antimicrobial agents based on the sensitivity of the isolated bacteria,” Nippon Ronen Igakkai Zasshi, Japanese Journal of Geriatrics, vol. 44, No. 3, (2007); pp. 359-366—(Abstract Only).
Zach, M., “Discussion”, Chest, vol. 94, No. 2, (Aug. 1988); pp. 160S-162S.
Zhang, et al., “Besifloxacin, A Novel Fluoroquinolone Antimicrobial Agent, Exhibits Potent Inhibition of Pro-Inflammatory Cytokines in Human THP-1 Monocytes”, Journal of Antimicrobial Chemotherapy, 61:111-6, (2008).
Zhao, et al., “Description and Clinical Treatment of an Early Outbreak of Severe Acute Respiratory Syndrome (SARS) in Guangzhou, PR China,” Journal of Medical Microbiology, vol. 52, No. 8, (2003); pp. 715-720.
Zheng, et al., “Pulmonary delivery of a dopamine D-1 agonist, ABT-431, in dogs and humansm” International Journal of Pharmaceutics, vol. 191, No. 2, (Nov. 30, 1999); pp. 131-140—(Abstract Only).
Zimmermann, et al., “Anti-Inflammatory Effects of Antibacterials on Human Bronchial Epithelial Cells”, Respiratory Research, 10(89)1-8, (2009).
Zolkina, T.D. et al, “Cytochemical indicators of lymphocytes after inhalation of riboflavin-nucleotide and calcium pantothenate in children with bronchial asthma” Pediatriia, vol. 8 (1987) pp. 108-109.
Related Publications (1)
Number Date Country
20190321371 A1 Oct 2019 US
Provisional Applications (1)
Number Date Country
61103501 Oct 2008 US
Continuations (4)
Number Date Country
Parent 15635818 Jun 2017 US
Child 16194954 US
Parent 15132122 Apr 2016 US
Child 15635818 US
Parent 14333583 Jul 2014 US
Child 15132122 US
Parent 12574680 Oct 2009 US
Child 14333583 US