Cystic fibrosis (CF), also called mucoviscidosis, is an autosomal, recessive, hereditary disease of the exocrine glands. It affects the lungs, sweat glands and the digestive system, causing chronic respiratory and digestive problems. It is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) protein. It is the most common fatal autosomal recessive diseases amongst Caucasions.
The first manifestation of CF is sometimes meconium ileus, occurring in 16% of infants who develop CF. Other symptoms of CF manifest during early childhood. Both lungs and pancreas produce abnormally viscous mucus. This mucus begins to build up and starts to clog the opening to the pancreas and the lungs. Pulmonary problems start from the constant presence of thick, sticky mucus and are one of the most serious complications of CF. The mucus in the lungs can become a growth medium for bacteria, resulting in chronic respiratory infections and eventual permanent damage to the lung tissue. During the end stage of CF, the patient experiences increased chest congestion, activity intolerance, increased crackles, and increased cough, which often contains sputum mixed with blood (hemoptysis) due to the bronchiole bleeding from the lung arteries. A chronic and loose sounding cough is common in people with CF. These thick secretions also obstruct the pancreas, preventing digestive enzymes from reaching the intestines to help break down and absorb food. Frequent and foul smelling stools are often an early sign of CF along with fatty oil that is visible in the stool. This can compromise growth and overall nutrition if proper treatment to aid digestion is not utilized early in life. As lung function deteriorates, CF patients can develop pulmonary hypertension, chronic bronchitis, and chronic dilation of the bronchioles (bronchiectasis). Lung abscess are very common. Death usually occurs from severe infection, pneumonia, or heart failure.
Cystic fibrosis is exclusively heritable as both parents must carry the recessive genes for a child to acquire the disease. At the genetic level, cystic fibrosis is most often the result of an in-frame deletion of three base pairs in the DNA. Cystic fibrosis results from the production of an abnormal form of a protein called cystic fibrosis transmembrane conductance regulator (CFTR). CFTR functions in transporting chloride ions across epithelial cells found in the lung and intestinal tract. In CF patients, CFTR does not function properly, causing accumulation of ions inside epithelial cells. Since water follows ions by osmosis, this results in water depletion and viscous mucus on the surface of alveoli. The most common CFTR protein abnormality is a mutation termed ΔF508, which is characterized by the 3-bp deletion of the DNA basepair sequence at chromosome location 7q31.1-31.2 that codes for the amino acid, phenylalanine.
In addition to pulmonary infections, most people with CF also have problems with digestion, particularly the digestion of fats. This leads to malabsorption and difficulty gaining and maintaining weight, which in turn affects overall health. This is due to the abnormally sticky mucus that blocks the release of digestive enzymes from the pancreas. Pancreatic insufficiency is treated with supplemental enzymes. Usually water-miscible forms of the fat-soluble vitamins A, D, E, and K are required as the decreased fat absorption can lead to deficiencies of these vitamins.
CF patients also have an increased incidence of diabetes mellitus because of the pancreatic blockage. The chronic blocking causes the Islets of Langerhans to degrade over time and decrease insulin production, causing hyperglycemia. There is also evidence that patients with CF become more resistant to the insulin that is produced, this can be triggered by infections or treatment with corticosteroids. Diabetes in CF patients is commonly referred to as CFRD, cystic fibrosis related diabetes. A typical diabetic diet is not feasible and therefore insulin doses are instead adjusted to fit the typical high-calorie/high-fat CF diet.
Many CF patients, to some degree, experience the widening of the tips of their fingers, known as “clubbing”. The condition affects fingers and toes, and results in the tip of the digit being round and enlarged. This can also be seen in people with COPD or severe heart disease. Since people with CF are prone to poor absorption of nutrients, osteoporosis can occur in early adulthood due to low bone density. It is important for people with CF to have regular dual energy X-ray absorptiometry (DEXA) scans to measure bone density and begin treatment if needed. When diagnosed early, treatment can help prevent more serious complications.
Some CF patients have hearing loss as a side effect of long-term use of the -mycin/-micin group of drugs, such as Tobramycin, which is used to combat lung infections. Although this side-effect is well-known and understood, these particular antibiotics are of high value in the treatment of CF patients, and often the hearing loss must be considered a necessary trade-off in order to preserve life and health. CF occurs primarily in individuals of central and western European origin. In the United States, the median age at death has increased from 8.4 years of age in 1969 to 14.3 years of age in 1998. The mean age of death has increased from 14 years in 1969 to 32.4 years of age in 2003 (Cystic Fibrosis Foundation). A major contributor to the significant increase in life expectancy is improved antibiotic treatment of chronic respiratory tract infections in CF subjects (Goss and Rosenfeld 2004) as well as improved nutrition and earlier diagnosis.
A major factor in the respiratory health of CF subjects is acquisition of chronic Pseudomonas aeruginosa infections. The infection rate with P. aeruginosa increases with age and by age 18 years, 80% of CF subjects in the U.S. are infected. The difficulties treating this infection are multifactorial, including poor penetration of antibiotics into sites of infection including mucus plugs, inactivation of antibiotics by CF sputum, growth of bacteria in a biofilm, changes in phenotype including conversion to a mucoid form of P. aeruginosa, and emergence of multi-drug resistance (Chmiel and Davis 2003; Gibson, Burns et al. 2003). The cornerstone of pulmonary therapy is optimizing treatment of P. aeruginosa as infection with this pathogen is associated with a poor clinical outcome (Doring, Conway et al. 2000; Chmiel and Davis 2003; Gibson, Burns et al. 2003; Gibson, Emerson et al. 2003).
One of the current approaches to management of chronic P. aeruginosa infection in humans with CF includes the use of suppressive therapy with inhaled tobramycin (TOBI®). Inhaled tobramycin, 300 mg, administered twice a day for cycles of 28 days followed by 28 days off drug has been shown to reduce P. aeruginosa colony counts, increase FEV1% predicted, reduce hospitalizations, and decrease antibiotic use (Ramsey, Pepe et al. 1999). Nevertheless, patients have to be dosed twice a day for approximately 15-20 minute inhalation periods per dose.
Daily chest physiotherapy and aerosol breathing treatments are very commonly prescribed for CF patients. Typical physical therapy involves manual chest percussion (pounding), positive pressure techniques and/devices or possibly using a device such as the ThAIRapy Vest or the Intrapulmonary Percussive Ventilator (IPV) to achieve the same effect: loosening of the thick mucus. Aerosolized medicines commonly given include albuterol, ipratropium bromide and Pulmozyme to loosen secretions and decrease inflammation. It was found that CFers who surf were healthier; consequently, some hospitals use a nebulized 6%-10% Saline solution on those CFers who do not have asthma to loosen the secretions. Inhaled aminoglycoside antibiotics are sometimes given to fight infections. A number of pharmacological agents that help mucosal clearance are being used. N-acetylcysteine that solubilizes mucus glycoprotein, however, has not proved to be significantly effective. Recombinant human DNAse decreases the viscosity of sputum by degrading the concentrated amount of DNA in the sputum of CF patients. DNAse treatment has been beneficial in increasing airflow during short-term use, and has also prolonged the interval between episodes of pulmonary exacerbations.
CF patients are typically hospitalized somewhat regularly, often every 6 months depending on the severity of the case. Patients often have intravenous antibiotics through a PICC line, Central Line, or Port-a-Caths.
Cystic fibrosis can also lead to bronchiectasis. Bronchiectasis is an abnormal stretching and enlarging of the respiratory passages caused by mucus blockage. When the body is unable to get rid of mucus, mucus becomes stuck and accumulates in the airways. The blockage and accompanying infection cause inflammation, leading to the weakening and widening of the passages. The weakened passages can become scarred and deformed, allowing more mucus and bacteria to accumulate, resulting in a cycle of infection and blocked airways. Bronchiectasis is a disease that causes localized, irreversible dilatation of part of the bronchial tree. Involved bronchi are dilated, inflamed, and easily collapsible, resulting in airflow obstruction and impaired clearance of secretions. Bronchiectasis is associated with a wide range of disorders, but it usually results from necrotizing bacterial infections, such as infections caused by the Staphylococcus or Klebsiella species or Bordatella pertussis.
Bronchiectasis is one of the chronic obstructive pulmonary diseases (COPD) and it can be complicated by emphysema and bronchitis. The disease is commonly misdiagnosed as asthma or pneumonia. Bronchiectasis can develop at any age, begins most often in childhood, but symptoms may not be apparent until much later. Bronchiectasis can occur as part of a birth defect, such as primary ciliary dyskinesia or cystic fibrosis. About 50% of all cases of bronchiectasis in the U.S. result from cystic fibrosis. It can also develop after birth as a result of injury or other diseases, like tuberculosis, pneumonia and influenza.
Dilation of the bronchial walls results in airflow obstruction and impaired clearance of secretions because the dilated areas interrupt normal air pressure of the bronchial tubes, causing sputum to pool inside the dilated areas instead of being pushed upward. The pooled sputum provides an environment conducive to the growth of infectious pathogens, and these areas of the lungs are thus very vulnerable to infection. The more infections that the lungs experience, the more damaged the lung tissue and alveoli become. When this happens, the bronchial tubes become more inelastic and dilated, which creates a perpetual, destructive cycle within this disease.
There are three types of bronchiectasis, varying by level of severity. Fusiform (cylindrical) bronchiectasis (the most common type) refers to mildly inflamed bronchi that fail to taper distally. In varicose bronchiectasis, the bronchial walls appear beaded, because areas of dilation are mixed with areas of constriction. Saccular (cystic) bronchiectasis is characterized by severe, irreversible ballooning of the bronchi peripherally, with or without air-fluid levels. Chronic productive cough is prominent, occurring in up to 90% of patients with bronchiectasis. Sputum is produced on a daily basis in 76% of patients.
In addition to CF, other genetic causes or contributing factors to bronchiectasisis include Kartagener syndrome, Young's syndrome, alpha 1-antitrypsin deficiency, and Primary immunodeficiencies. Acquired bronchiectasis occurs more frequently, with one of the biggest causes being tuberculosis. A especially common cause of the disease in children is Acquired Immunodeficiency Syndrome, stemming from the human immunodeficiency virus. Other causes of bronchiectasis include respiratory infections, obstructions, inhalation and aspiration of ammonia, and other toxic gases, pulmonary aspiration, alcoholism, heroin use and allergies. Cigarette smoking may also contribute to bronchiectasis.
The diagnosis of bronchiectasis is based on the review of clinical history and characteristic patterns in high-resolution CT scan findings. Such patterns include “tree-in-bud” abnormalities and cysts with definable borders. Bronchiectasis may also be diagnosed without CT scan confirmation if clinical history clearly demonstrates frequent, respiratory infections, as well confirmation of an underlying problem via blood work and sputum culture samples.
Symptoms include coughing (worsened when lying down), shortness of breath, abnormal chest sounds, weakness, weight loss, and fatigue. With infections the mucus may be discolored, foul smelling and may contain blood. Symptom severity varies widely from patient to patient and occasionally, a patient is asymptomatic.
Treatment of bronchiectasis is aimed at controlling infections and bronchial secretions, relieving airway obstruction, and preventing complications. This includes prolonged usage of antibiotics to prevent detrimental infections, as well as eliminating accumulated fluid with postural drainage and chest physiotherapy. Surgery may also be used to treat localized bronchiectasis, removing obstructions that could cause progression of the disease.
Inhaled steroid therapy that is consistently adhered to can reduce sputum production and decrease airway constriction over a period of time will prevent progression of bronchiectasis. One commonly used therapy is beclometasone dipropionate, also used in asthma treatment. Use of inhalers such as Albuterol (Salbutamol), Fluticasone (Flovent/Flixotide) and Ipratropium (Atrovent) may help reduce likelihood of infection by clearing the airways and decreasing inflammation.
Mannitol dry inhalation powder, under the name Bronchitol, has been approved by the FDA for use in Cystic Fibrosis patients with Bronchiectasis. The original orphan drug indication approved in February 2005 allowed its use for the treatment of bronchiectasis. The original approval was based on the results of phase 2 clinical studies showing the product to be safe, well-tolerated, and effective for stimulating mucus hydration/clearance, thereby improving quality of life in patients with chronic obstructive lung diseases like Bronchiectasis. Long-term studies are underway as of 2007 to ensure the safety and effectiveness of the treatment.
Bronchiectasis patients are often given antibiotics for infection and bronchodilator medicines to open passages. Sometimes antibiotics are prescribed for a long period to prevent recurring infections, especially in people who have cystic fibrosis. There are also physical therapy techniques to help clear mucus. Lung transplants are also an option for severe cases. Fatalities are uncommon but may result from massive hemorrhage. If lung infections are treated immediately, bronchiectasis is less likely to develop.
Pneumonia is an illness of the lungs and respiratory system in which the alveoli (microscopic air-filled sacs of the lung responsible for absorbing oxygen from the atmosphere) become inflamed and flooded with fluid. Pneumonia can result from a variety of causes, including infection with bacteria, viruses, fungi, or parasites, and chemical or physical injury to the lungs. Typical symptoms associated with pneumonia include cough, chest pain, fever, and difficulty in breathing. Diagnostic tools include x-rays and examination of the sputum.
Therefore, there is a need for therapies to treat pulmonary disorders, including CF, pulmonary infections, COPD, bronchiectasis and others. Additionally, there is a need to improve lung function in patients having such disorders.
The present invention relates in part to a method of treating a pulmonary disorder in a patient comprising administering to the patient an effective dose of a nebulized liposomal amikacin formulation for at least one treatment cycle, wherein:
In some embodiments, the treatment cycle is administered to the patient at least twice. In some embodiments, the administration period is 15 to 35 days, or 20 to 35 days. In other embodiments, the administration period is about 28 days. In some embodiments, the off period is 15 to 35 days, or 20 to 35 days. In other embodiments, the off period is about 28 days. In still other embodiments, the off period is of 25 to 75 days, 35 to 75 days, or 45 to 75 days. In other embodiments, the off period is about 56 days.
In some embodiments, the administration period is about 28 days and the off period is about 28 days, while in other embodiments, the administration period is about 28 days and the off period is about 56 days.
In some embodiments, the effective dose comprises 250 to 1,500 mg of amikacin, 250 to 1000 mg of amikacin, or about 280 to about 560 mg of amikacin. In other embodiments, the effective dose is about 280 or about 560 mg of amikacin.
In some embodiments, the pulmonary disorder is selected from the group consisting of chronic obstructive pulmonary disease, bronchiectasis, pulmonary infection, cystic fibrosis, alpha-1-antitrypsin enzyme deficiency and a combination thereof. In other embodiments, the pulmonary condition is a bacterial pulmonary infection, such as a P. aeruginosa infection. In some embodiments, the pulmonary condition is bronchiectasis.
In some embodiments, the patient has a serum Cmax of amikacin of less than about 10 mcg/mL during the administration period. In other embodiments, the patient has a sputum Cmax of amikacin of at least 1000 mcg per gram of sputum either during the administration, for at least 15 days after the administration.
In some embodiments, the patient has a reduction in log10 CFU of the bacterial infection in the lungs of at least 0.5 for at least 15 days after the administration period ends. In other embodiments, the reduction in the logo CFU is at least 1.0.
In some embodiments, the patient experiences an improvement in lung function for at least 15 days after the administration period ends. For example, the patient may experience an increase in FEV1, an increase in blood oxygen saturation, or both. In some embodiments, the patient has an FEV1 that is increased by at least 5% over the FEV1 prior to the treatment cycle. In other embodiments, FEV1 is increased by 5 to 50%. In other embodiments, FEV1 is increased by 25 to 500 mL over FEV1 prior to the treatment cycle. In some embodiments, blood oxygen saturation is increased by at least 1% over oxygen saturation prior to the treatment cycle.
In some embodiments, the length of time to a pulmonary exacerbation is at least 20 days from the last day of administration. In other embodiments, the length of time to a rescue treatment is at least 25 days from the last day of the administration.
In some embodiments, the liposomal amikacin formulation comprises a lipid selected from the group consisting of egg phosphatidylcholine (EPC), egg phosphatidylglycerol (EPG), egg phosphatidylinositol (EPI), egg phosphatidylserine (EPS), phosphatidylethanolamine (EPE), phosphatidic acid (EPA), soy phosphatidylcholine (SPC), soy phosphatidylglycerol (SPG), soy phosphatidylserine (SPS), soy phosphatidylinositol (SPI), soy phosphatidylethanolamine (SPE), soy phosphatidic acid (SPA), hydrogenated egg phosphatidylcholine (HEPC), hydrogenated egg phosphatidylglycerol (HEPG), hydrogenated egg phosphatidylinositol (HEPI), hydrogenated egg phosphatidylserine (HEPS), hydrogenated phosphatidylethanolamine (HEPE), hydrogenated phosphatidic acid (HEPA), hydrogenated soy phosphatidylcholine (HSPC), hydrogenated soy phosphatidylglycerol (HSPG), hydrogenated soy phosphatidylserine (HSPS), hydrogenated soy phosphatidylinositol (HSPI), hydrogenated soy phosphatidylethanolamine (HSPE), hydrogenated soy phosphatidic acid (HSPA), dipalmitoylphosphatidylcholine (DPPC), dimyristoylphosphatidylcholine (DMPC), dimyristoylphosphatidylglycerol (DMPG), dipalmitoylphosphatidylglycerol (DPPG), distearoylphosphatidylcholine (DSPC), distearoylphosphatidylglycerol (DSPG), dioleylphosphatidyl-ethanolamine (DOPE), palmitoylstearoylphosphatidyl-choline (PSPC), palmitoylstearolphosphatidylglycerol (PSPG), mono-oleoyl-phosphatidylethanolamine (MOPE), cholesterol, ergosterol, lanosterol, tocopherol, ammonium salts of fatty acids, ammonium salts of phospholipids, ammonium salts of glycerides, myristylamine, palmitylamine, laurylamine, stearylamine, dilauroyl ethylphosphocholine (DLEP), dimyristoyl ethylphosphocholine (DMEP), dipalmitoyl ethylphosphocholine (DPEP) and distearoyl ethylphosphocholine (DSEP), N-(2, 3-di-(9-(Z)-octadecenyloxy)-prop-1-yl-N,N,N-trimethylammonium chloride (DOTMA), 1, 2-bis(oleoyloxy)-3-(trimethylammonio)propane (DOTAP), phosphatidyl-glycerols (PGs), phosphatidic acids (PAs), phosphatidylinositols (PIs), phosphatidyl serines (PSs), distearoylphosphatidylglycerol (DSPG), dimyristoylphosphatidylacid (DMPA), dipalmitoylphosphatidylacid (DPPA), distearoylphosphatidylacid (DSPA), dimyristoylphosphatidylinositol (DMPI), dipalmitoylphosphatidylinositol (DPPI), distearoylphospatidylinositol (DSPI), dimyristoylphosphatidylserine (DMPS), dipalmitoylphosphatidylserine (DPPS), distearoylphosphatidylserine (DSPS), and mixtures thereof. In other embodiments, the liposomal amikacin formulation comprises a phospholipid and a sterol, such as DPPC and cholesterol. In other embodiments, the liposomal amikacin formulation comprises DPPC and cholesterol in about a 2 to 1 ratio by weight. In some embodiments, the liposomal amikacin formulation has a lipid to drug ratio of about 0.5 to about 1.0, about 0.5 to 0.7, or about 0.6 by weight.
For convenience, before further description of the present invention, certain terms employed in the specification, examples and appended claims are collected here. These definitions should be read in light of the remainder of the disclosure and understood as by a person of skill in the art. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art.
The term “pulmonary disorder” refers to any disease, ailment, or other unhealthy condition related to the respiratory tract of a subject, particularly the lungs of a subject. Generally pulmonary distress results in difficulty of breathing.
The term “treating” is art-recognized and refers to curing as well as ameliorating at least one symptom of any condition or disease.
The term “prophylactic” or “therapeutic” treatment is art-recognized and refers to administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, i.e., it protects the host against developing the unwanted condition, whereas if administered after manifestation of the unwanted condition, the treatment is therapeutic (i.e., it is intended to diminish, ameliorate or maintain the existing unwanted condition or side effects therefrom).
The terms “therapeutically effective dose” and “therapeutically effective amount” refer to that amount of a compound that results in prevention or amelioration of symptoms in a patient or a desired biological outcome, e.g., improved clinical signs, delayed onset of disease, reduced levels of bacteria, etc.
The term “FEV1” is well known in the art as a measure of lung function, and refers to the forced expiratory volume in one second. The FEV1 values used herein are measured in mLs, and also in terms of percent change from baseline, e.g., a change from pre-treatment values.
A “patient,” “subject” or “host” to be treated by the subject method may mean either a human or non-human animal.
The term “mammal” is known in the art, and exemplary mammals include humans, primates, bovines, porcines, canines, felines, and rodents (e.g., mice and rats).
The term “bioavailable” is art-recognized and refers to a form of the subject invention that allows for it, or a portion of the amount administered, to be absorbed by, incorporated to, or otherwise physiologically available to a subject or patient to whom it is administered.
The term “pharmaceutically-acceptable salts” is art-recognized and refers to the relatively non-toxic, inorganic and organic acid addition salts of compounds, including, for example, those contained in compositions of the present invention.
The term “pharmaceutically acceptable carrier” is art-recognized and refers to a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting any subject composition or component thereof from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the subject composition and its components and not injurious to the patient. Some examples of materials which may serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations.
Liposomal amikacin formulations useful in the presently disclosed methods can be prepared as described, for example, in U.S. Publication No. 20060073198 or 20080089927, both of which are hereby incorporated by reference. Generally, amikacin is used in the form of a pharmaceutically acceptable salt, for example the sulfate salt of amikacin.
The lipids used in the compositions of the present invention can be synthetic, semisynthetic or naturally-occurring lipids, including phospholipids, tocopherols, steroids, fatty acids, glycoproteins such as albumin, anionic lipids and cationic lipids. The lipids may be anionic, cationic, or neutral. In one embodiment, the lipid formulation is substantially free of anionic lipids, substantially free of cationic lipids, or both. In one embodiment, the lipid formulation comprises only neutral lipids. In another embodiment, the lipid formulation is free of anionic lipids or cationic lipids or both. In another embodiment, the lipid is a phospholipid. Phospholipids include egg phosphatidyl choline (EPC), egg phosphatidylglycerol (EPG), egg phosphatidylinositol (EPI), egg phosphatidylserine (EPS), phosphatidylethanolamine (EPE), and egg phosphatidic acid (EPA); the soya counterparts, soy phosphatidyl choline (SPC); SPG, SPS, SPI, SPE, and SPA; the hydrogenated egg and soya counterparts (e.g., HEPC, HSPC), other phospholipids made up of ester linkages of fatty acids in the 2 and 3 of glycerol positions containing chains of 12 to 26 carbon atoms and different head groups in the 1 position of glycerol that include choline, glycerol, inositol, serine, ethanolamine, as well as the corresponding phosphatidic acids. The chains on these fatty acids can be saturated or unsaturated, and the phospholipid can be made up of fatty acids of different chain lengths and different degrees of unsaturation. In particular, the compositions of the formulations can include dipalmitoylphosphatidylcholine (DPPC), a major constituent of naturally-occurring lung surfactant as well as dioleoylphosphatidylcholine (DOPC). Other examples include dimyristoylphosphatidylcholine (DMPC) and dimyristoylphosphatidylglycerol (DMPG) dipalmitoylphosphatidcholine (DPPC) and dipalmitoylphosphatidylglycerol (DPPG) distearoylphosphatidylcholine (DSPC) and distearoylphosphatidylglycerol (DSPG), dioleylphosphatidylethanolamine (DOPE) and mixed phospholipids like palmitoylstearoylphosphatidylcholine (PSPC) and palmitoylstearoylphosphatidylglycerol (PSPG), driacylglycerol, diacylglycerol, seranide, sphingosine, sphingomyelin and single acylated phospholipids like mono-oleoyl-phosphatidylethanol amine (MOPE).
The lipids used can include ammonium salts of fatty acids, phospholipids and glycerides, phosphatidylglycerols (PGs), phosphatidic acids (PAs), phosphotidylcholines (PCs), phosphatidylinositols (PIs) and the phosphatidylserines (PSs). The fatty acids include fatty acids of carbon chain lengths of 12 to 26 carbon atoms that are either saturated or unsaturated. Some specific examples include: myristylamine, palmitylamine, laurylamine and stearylamine, dilauroyl ethylphosphocholine (DLEP), dimyristoyl ethylphosphocholine (DMEP), dipalmitoyl ethylphosphocholine (DPEP) and distearoyl ethylphosphocholine (DSEP), N-(2, 3-di-(9 (Z)-octadecenyloxy)-prop-1-yl-N,N,N-trimethylammonium chloride (DOTMA) and 1, 2-bis(oleoyloxy)-3-(trimethylammonio)propane (DOTAP). Examples of PGs, PAs, PIs, PCs and PSs include DMPG, DPPG, DSPG, DMPA, DPPA, DSPA, DMPI, DPPI, DSPI, DMPS, DPPS and DSPS, DSPC, DPPG, DMPC, DOPC, egg PC.
In another embodiment, the liposome comprises a lipid selected from the group consisting of phosphatidyl cholines (PCs), phosphatidyl-glycerols (PGs), phosphatidic acids (PAs), phosphatidylinositols (PIs), and phosphatidyl serines (PSs).
In another embodiment, the lipid is selected from the group consisting of: egg phosphatidylcholine (EPC), egg phosphatidylglycerol (EPG), egg phosphatidylinositol (EPI), egg phosphatidylserine (EPS), phosphatidylethanolamine (EPE), phosphatidic acid (EPA), soy phosphatidylcholine (SPC), soy phosphatidylglycerol (SPG), soy phosphatidylserine (SPS), soy phosphatidylinositol (SPI), soy phosphatidylethanolamine (SPE), soy phosphatidic acid (SPA), hydrogenated egg phosphatidyl choline (HEPC), hydrogenated egg phosphatidylglycerol (HEPG), hydrogenated egg phosphatidylinositol (HEPI), hydrogenated egg phosphatidylserine (HEPS), hydrogenated phosphatidylethanolamine (HEPE), hydrogenated phosphatidic acid (HEPA), hydrogenated soy phosphatidylcholine (HSPC), hydrogenated soy phosphatidylglycerol (HSPG), hydrogenated soy phosphatidylserine (HSPS), hydrogenated soy phosphatidylinositol (HSPI), hydrogenated soy phosphatidylethanolamine (HSPE), hydrogenated soy phosphatidic acid (HSPA), dipalmitoylphosphatidylcholine (DPPC), dimyristoylphosphatidylcholine (DMPC), dimyristoylphosphatidylglycerol (DMPG), dipalmitoylphosphatidylglycerol (DPPG), distearoylphosphatidylcholine (DSPC), distearoylphosphatidylglycerol (DSPG), dioleylphosphatidyl-ethanolamine (DOPE), palmitoylstearoylphosphatidyl-choline (PSPC), palmitoylstearolphosphatidylglycerol (PSPG), mono-oleoyl-phosphatidylethanolamine (MOPE), tocopherol, ammonium salts of fatty acids, ammonium salts of phospholipids, ammonium salts of glycerides, myristylamine, palmitylamine, laurylamine, stearylamine, dilauroyl ethylphosphocholine (DLEP), dimyristoyl ethylphosphocholine (DMEP), dipalmitoyl ethylphosphocholine (DPEP) and distearoyl ethylphosphocholine (DSEP), N-(2, 3-di-(9-(Z)-octadecenyloxy)-prop-1-yl-N,N,N-trimethylammonium chloride (DOTMA), 1, 2-bis(oleoyloxy)-3-(trimethylammonio)propane (DOTAP), distearoylphosphatidylglycerol (DSPG), dimyristoylphosphatidylacid (DMPA), dipalmitoylphosphatidylacid (DPPA), distearoylphosphatidylacid (DSPA), dimyristoylphosphatidylinositol (DMPI), dipalmitoylphosphatidylinositol (DPPI), distearoylphospatidylinositol (DSPI), dimyristoylphosphatidylserine (DMPS), dipalmitoylphosphatidylserine (DPPS), distearoylphosphatidylserine (DSPS), and mixtures thereof.
In another embodiment, the liposome comprises a phosphatidyl choline. The phosphatidyl choline may be unsaturated, such as DOPC or POPC, or saturated, such as DPPC. In another embodiment, the liposome does not include a sterol. In one embodiment, the liposome consists essentially of a phosphatidyl choline and a sterol. In another embodiment, the liposome consists essentially of DPPC and cholesterol.
Liposomes or lipid antiinfective formulations composed of phosphatidylcholines, such as DPPC, aid in the uptake by the cells in the lung such as the alveolar macrophages and helps to sustain release of the antiinfective agent in the lung (Gonzales-Rothi et al. (1991)). The negatively charged lipids such as the PGs, PAs, PSs and PIs, in addition to reducing particle aggregation, can play a role in the sustained release characteristics of the inhalation formulation as well as in the transport of the formulation across the lung (transcytosis) for systemic uptake.
While not being bound by any particular theory, it is believed that when the lipid comprises a neutral lipid, and does not comprise a negatively charged or positively charged phospholipid, the liposomal formulation has improved uptake by the lungs. For example, the liposome my have improved penetration into a biofilm or mucus layer when the lipid comprises only neutral lipids. Exemplary neutral lipids include the aforementioned phosphatidylcholines, such as DPPC and sterols, such as cholesterol.
The present invention is directed to methods of treating a pulmonary condition in a subject need thereof comprising administering to the subject and effective amount of any one of the aforementioned liposomal antibiotic formulations. In some embodiments, the pulmonary condition is a bacterial infection. In some embodiments, the method comprises administering to a patient in need thereof an effective amount of a liposomal amikacin formulation (also referred to herein as “liposomal amikacin”) by inhalation daily. In some embodiments, the administration by inhalation comprises nebulizing the liposomal formulation.
In some embodiments, the liposomal amikacin formulation is administered daily for a period of time, followed by second period of time (an “off” period) wherein no liposomal formulation is administered. For example, in some embodiments, the method of treating a pulmonary disorder comprises administering to the patient an effective dose of a nebulized liposomal amikacin formulation for at least one treatment cycle, wherein:
In some embodiments, the aforementioned treatment cycle is administered to the patient at least twice. In other embodiments, the treatment cycle may be administered 3, 4, 5, 6, or more times.
During the administration period, liposomal amikacin is administered daily. In some embodiments, liposomal amikacin can be administered every other day or every third day during the administration period. As explained above, the administration period can be 15 to 75 days. In some embodiments, the administration period is 15 to 35 days, or 20 to 35 days. In other embodiments, the administration period is 20 to 30 days, 25 to 35 days or 25 to 30 days. In other embodiments, the administration period is about 25, 26, 27, 28, 29 or 30 days. In another embodiment, the administration period is about 28 days.
During the off period the liposomal amikacin formulation is not administered to the patient. In some embodiments, the off period is 15 days or longer, for example, 15 to 75 days, 15 to 35 days, or 20 to 35 days. In other embodiments, the off period is 20 to 30 days, 25 to 35 days or 25 to 30 days. In other embodiments, the off period is about 25, 26, 27, 28, 29 or 30 days. In other embodiments, the off period is about 28 days, while in still other embodiments, the off period is at least 29 days.
In some embodiments, the off period is of 25 to 75 days, 35 to 75 days, or 45 to 75 days. In other embodiments, the off period is 50 to 75 days, 50 to 70 days, 50 to 65 days or 50 to 60 days. In other embodiments, the off period is about 50, 51, 52, 53, 54, 55, 56, 57, 58, 59 or 60 days, while in other embodiments, the off period is about 56 days.
In some embodiments, the administration period is about 28 days and the off period is about 28 days, while in other embodiments, the administration period is about 28 days and the off period is about 56 days.
In some embodiments, the effective dose comprises 250 to 1,500 mg of amikacin, 250 to 1000 mg of amikacin, 250 to 750 mg of amikacin, or 250 to 700 mg amikacin each day of the administration period. In other embodiments, the effective dose is about 280 to about 560 mg of amikacin. In other embodiments, the effective dose is about 230 mg to about 330 mg, or about 510 mg to about 610 mg. In other embodiments, the effective dose of amikacin is about 100, 150, 200, 250, 300, 250, 400, 450, 500, 550, 600, 650, 700 or 750 mg of amikacin daily. In other embodiments, the effective dose is about 280 or about 560 mg of amikacin.
In some embodiments, the administration period is about 28 days, and the dose is about 280 to about 560 mg of amikacin. In other embodiments, the administration period is about 28 days, the off period is about 28 days, and the dose is about 280 to about 560 mg. In other embodiments, the administration period is about 28 days, the off period is about 56 days, and the dose is about 280 to about 560 mg.
In some embodiments, the pulmonary disorder is selected from the group consisting of chronic obstructive pulmonary disease, bronchiectasis, pulmonary infection, cystic fibrosis, alpha-1-antitrypsin enzyme deficiency and a combination thereof. In some embodiments, the pulmonary condition is cystic fibrosis. In other embodiments, the pulmonary condition is a bacterial pulmonary infection, Pseudomonas (e.g., P. aeruginosa, P. paucimobilis, P. putida, P. fluorescens, and P. acidovorans), staphylococcal, Methicillin-resistant Staphylococcus aureus (MRSA), streptococcal (including by Streptococcus pneumoniae), Escherichia coli, Klebsiella, Enterobacter, Serratia, Haemophilus, Yersinia pesos, Burkholderia pseudomallei, B. cepacia, B. gladioli, B. multivorans, B. vietnamiensis, Mycobacterium tuberculosis, M. avium complex (MAC) (M. avium and M. intracellulare), M kansasii, M. xenopi, M marinum, M. ulcerans, or M. fortuitum complex (M. fortuitum and M. chelonei) infections. In some embodiments, the infection is a P. aeruginosa infection, while in other embodiments, the infection is a non-tuberculous mycobacterial infection. The pulmonary infection may or may not be associated with cystic fibrosis. Thus, in some embodiments, the pulmonary condition is both cystic fibrosis and a pulmonary infections such as P. aeruginosa. In other embodiments, the pulmonary conditions is bronchiectasis. The bronchiectasis may or may not be associated with cystic fibrosis.
The present method provides advantageous levels of amikacin at the site of the pulmonary disorder, while limiting systemic exposure to the drug, and also provides a sustained benefit to the subject for surprisingly extended periods of time. While not being bound by any particular theory, it is believed that administration of liposomal amikacin in accordance the with methods described herein results a “depot” effect in the lungs of the subject. Specifically, it is believed that the liposome particles are small enough and contain an appropriate lipid formulation to penetrate and diffuse through CF sputum and into the bacterial biofilm. The liposomes shield the entrapped cationic amikacin in neutral liposomes to minimize electrostatic interaction with the negatively charged sputum/biofilm, which would otherwise reduce its bioavailability. In addition, there are P. aeruginosa derived virulence factors (rhamnolipids) (Davey et al. 2003), which release amikacin the liposomes. Therefore, it is hypothesized that relatively high concentrations of drug can be delivered locally to the bacterial macro-colony environment.
Additionally, it is believed that inhalation of liposomal amikacin leads to a dose dependent recruitment of macrophages as an adaptive response to inhalation of drug/lipid formulation. The presence of alveolar macrophages (which have been shown to be functionally normal in liposomal amikacin treated rats) may be particularly beneficial in CF patients. CF patients are known to have reduced number of macrophages in their lungs and possibly with poor functionality, which may contribute to the chronicity of P. aeruginosa lung infection, and to the higher prevalence of non-tuberculous mycobacterial infection in this population. The dose dependent recruitment of macrophages may also contribute to the sustained effects observed using the methods of the present invention. Specifically, the macrophages in the lung may take up liposomal amikacin, and then remain in the lung for a period of time, followed by release of the liposomal amikacin by the macrophages. A clinical study (described in the exemplification below) of liposomal amikacin in CF patients chronically infected with P. aeruginosa has demonstrated safety, tolerability and dose dependent improvement in lung function and respiratory symptoms; and reduction of sputum bacterial density at the end of 28 days of treatment. This improvement in lung function was sustained for at least 28 days after completion of treatment (Day 56) with a 560 mg dose of liposomal amikacin, indicating a sustained treatment effect.
The present method thus provides, in some embodiments, advantageous levels of amikacin in the blood and in the sputum. For example, the methods provides relatively low systemic exposure to amikacin, while providing high, sustained levels of amikacin at the site of the pulmonary condition. For example, in some embodiments, the patient has a serum Cmax of amikacin of less than about 25 mcg/mL during the administration period. In other embodiments, the serum Cmax is less than 20, 15, 10, 5 or 2 mcg/mL during the administration period.
In some embodiments, the patient has a sputum Cmax of amikacin of at least about 500 mcg per gram of sputum either during the administration, or for a sustained period of time, such as at least 15 days, after the administration. In other embodiments, the sputum Cmax of amikacin is at least 750, 1000, 1500, 2000, 2500, 3000 or 3500 mcg per gram of sputum.
When the pulmonary disorder includes a pulmonary infection, the present invention also provides a reduction in the colony forming units of the bacteria in the lung for a sustained period of time. For example, the CFU's are reduced compared to a baseline value. In some embodiments, the patient has a reduction in log10 CFU of the bacterial infection in the lungs of at least about 0.5 for at least 15 days after the administration period ends. In other embodiments, the reduction in the log10 CFU is at least by 1.0, 1.5, 2.0 or 2.5. Pseudomonas infections, in particular, can form large colonies, known as “mucoid” Pseudomonas, particularly in patients with cystic fibrosis. In some embodiments, the CFU's are reduced as described above in a mucoid strain of a Pseudomonas infection.
In some embodiments, the patient experiences an improvement in lung function for at least 15 days after the administration period ends. For example, the patient may experience an increase in the forced expiratory volume in one second (FEV1), an increase in blood oxygen saturation, or both. In some embodiments, the patient has an FEV1 that is increased by at least 5% or at least 10% over the FEV1 prior to the treatment cycle. In other embodiments, FEV1 is increased by 5 to 50%, 5 to 25%, or 5 to 20%. In other embodiments, FEV1 is increased by 5 to 15% or 5 to 10%. In other embodiments, FEV1 is increased by 10 to 50%, 10 to 40%, 10 to 30% or 10 20%. FEV1 is frequently measured in mL. Accordingly, in some embodiments, FEV1 is increased by at least 25 mL when compared to FEV1 prior to the treatment. In some embodiments, FEV1 is increased by 25 to 500 mL, 25 to 400, 25 to 300 or 25 to mL. In other embodiments, FEV1 is increased by 50 to 500 mL, 50 to 400 mL, 50 to 300 mL, 50 to 200 mL or 50 to 100 mL.
In some embodiments, blood oxygen saturation is increased in the subject compared to the blood oxygen saturation levels prior to the administration. In some embodiments, blood oxygen saturation is increased by at least 1% or by at least 2% for at least 15 days after the administration period. In other embodiments, the blood oxygen saturation levels are increased by about 1 to 50%, 1 to 25%, 1 to 20%, 1 to 15%, 1 to 10% or 1 to 5%. In other embodiments, the blood oxygen saturation levels are increased by about 2 to 10% or 2 to 5%.
The aforementioned sustained periods of time may be at least 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70 or 75 days after the administration period. In other embodiments, the sustained period of time is at least 28, 35, 42, 48 or 56 days after the administration period. In other embodiments, sustained period of 15 to 75 days, 15 to 35 days, or 20 to 35 days. In other embodiments, the sustained period of time is 20 to 30 days, 25 to 35 days or 25 to 30 days. In other embodiments, the sustained period of time is about 25, about 26, about 27, about 28, about 29 or about 30 days, or about 28 days, or at least 29 days. In other embodiments, the sustained period of time during is 25 to 75 days, 35 to 75 days, or 45 to 75 days. In other embodiments, the sustained period is 50 to 75 days, 50 to 70 days, 50 to 65 days or 50 to 60 days. In other embodiments, the sustain period is about 50, about 51, about 52, about 53, about 54, about 55, about 56, about 57, about 58, about 59 or about 60 days, while in other embodiments, the sustained period is about 56 days.
In some embodiments, the aforementioned methods advantageously provide a reduced incidence of pulmonary exacerbations in the patient. The method also advantageously increases the length of time to pulmonary exacerbation. For example, in some embodiments, the length of time to pulmonary exacerbation is at least about 20 days. In other embodiments, the length of time is 20 to 100 days. In other embodiments, the length of time is 25 to 100 days, 30 to 100 days, 35 to 100 days or 40 to 100 days. In other embodiments, the length of time is 25 to 75 days, 30 to 75 days, 35 to 75 days or 40 to 75 days. In other embodiments, the length of time is 30 to 60 days.
In some embodiments, the incidence of rescue treatment is reduced. In other embodiments, the length of time to rescue treatment is reduced, for example when the patient has a pulmonary infection, the time to anti-infective rescue treatment is reduced. In some embodiments, the length of time is 20 to 100 days. In other embodiments, the length of time is 25 to 100 days, 30 to 100 days, 35 to 100 days or 40 to 100 days. In other embodiments, the length of time is 25 to 75 days, 30 to 75 days, 35 to 75 days or 40 to 75 days. In other embodiments, the length of time is 30 to 60 days.
In some embodiments, the liposomal amikacin formulation used in the aforementioned methods comprises amikacin and any of the lipids described above. In some embodiments, the liposomal amikacin formulation comprises a phospholipid and a sterol, such as DPPC and cholesterol. In other embodiments, the liposomal amikacin formulation comprises DPPC and cholesterol in about a 2 to 1 ratio by weight. In some embodiments, the liposomal amikacin formulation has a lipid to drug ratio of about 0.5 to about 1.0, about 0.5 to 0.7, or about 0.6 by weight. In other embodiments, the liposomal amikacin formulation has a lipid to drug ratio of about 0.3 to about 1.0 by weight, while in other embodiments, the lipid to drug ratio is about 0.5 to 0.7 by weight, or about 0.65 by weight. The liposomes in the formulation may have a amend diameter of 100 to 1000 nm, 100 to 500 nm, 200 to 500 nm, or about 300 nm. In some embodiments, the total concentration of amikacin in the liposomal amikacin formulation is about 20 to 100 mg/mL, 20 to 90 mg, mL, 30 to 90 mg/mL, 30 to 80 mg/mL, or 40 to 80 mg/mL. In other embodiments, the concentration is about 30, 40, 50, 60, 70, 80 or 90 mg/mL.
In some embodiments, aforementioned method comprises:
administering to the patient an effective dose of a nebulized liposomal amikacin formulation for at least one treatment cycle, wherein:
the treatment cycle comprises an administration period of about 28 days, followed by an off period of about 28 days;
the effective dose comprises about 280 to about 560 mg of amikacin daily during the administration period; and
the liposomal amikacin formulation comprises DPPC and cholesterol in about a 2:1 ratio, and a lipid to amikacin ratio of about 0.5 to about 0.7.
In other embodiments, the method comprises:
administering to the patient an effective dose of a nebulized liposomal amikacin formulation for at least one treatment cycle, wherein:
the treatment cycle comprises an administration period of about 28 days, followed by an off period of about 56 days;
the effective dose comprises about 280 to about 560 mg of amikacin daily during the administration period; and
the liposomal amikacin formulation comprises DPPC and cholesterol in about a 2:1 ratio, and a lipid to amikacin ratio of about 0.5 to about 0.7.
In other embodiments, the present invention relates to a method of providing a sustained treatment effect in a subject comprising: administering to the patient an effective dose of a nebulized liposomal amikacin formulation for at least one treatment cycle, wherein: the treatment cycle comprises an administration period of 15 to 75 days, followed by an off period of 15 to 75 days; and the effective dose comprises 100 to 2500 mg of amikacin daily during the administration period.
In another embodiment, the present invention relates to a method of improving oxygen saturation levels in a patient with a pulmonary condition comprising: administering to the patient an effective dose of a nebulized liposomal amikacin formulation for at least one treatment cycle, wherein: the treatment cycle comprises an administration period of 15 to 75 days, followed by an off period of 15 to 75 days; and the effective dose comprises 100 to 2500 mg of amikacin daily during the administration period.
In another embodiment, the present invention relates to a method of improving FEV1 in a patient with a pulmonary condition comprising: administering to the patient an effective dose of a nebulized liposomal amikacin formulation for at least one treatment cycle, wherein: the treatment cycle comprises an administration period of 15 to 75 days, followed by an off period of 15 to 75 days; and the effective dose comprises 100 to 2500 mg of amikacin daily during the administration period.
In another embodiment, the present invention relates to a method of reducing bacterial density in the lung or sputum of a patient with a bacterial pulmonary infection comprising: administering to the patient an effective dose of a nebulized liposomal amikacin formulation for at least one treatment cycle, wherein: the treatment cycle comprises an administration period of 15 to 75 days, followed by an off period of 15 to 75 days; and the effective dose comprises 100 to 2500 mg of amikacin daily during the administration period, and wherein the bacterial density remains reduced for at least 15 days after the last day of the administration.
Introduction to Materials and Methods
Lipid based or liposomal aminoglycoside, such as amikacin, formulations for inhalation are sustained-release formulations of aminoglycosides encapsulated inside nanoscale liposomal carriers designed for administration via inhalation. Sustained-release targeting of high concentrations of amikacin in the lungs and biofilm penetration properties of these formulations have several advantages over inhalation of the “free” antibiotic, e.g., inhaled tobramycin. Amikacin can be encapsulated in liposomes composed of dipalmitoylphosphatidylcholine (DPPC) and cholesterol, at a targeted lipid-to-drug ratio of about 0.6-0.7:1 (w/w). An example of a ˜70 mg/mL liposomal amikacin formulation useful in the aforementioned methods is presented below:
1Added to the formulation as Amikacin sulfate, USP.
These formulations can be prepared according to the methods described in U.S. Publication No. 20060073198 or 20080089927, both of which are hereby incorporated by reference.
These formulations have several advantages in treating pulmonary conditions, for example, CF subjects with chronic infection caused by P. aeruginosa, including:
Preclinical pharmacokinetics have demonstrated that the AUC (0-48 hr) of amikacin in the lungs of rats that received a 60 mg/kg dose aerosol of Liposomal Amikacin was five-fold higher than the AUC of tobramycin in the lungs of rats that received an equal dose of tobramycin by inhalation. Generally, 10% of the administered antibiotic is deposited in the lungs for rats. Conversely, the AUC of drug in the kidneys of rats that received an equal dose of tobramycin was significantly higher than the kidney AUC of rats that received aerosols of Liposomal Amikacin. Additionally, data from 30-day inhalation toxicology studies in rats and dogs suggest that there will be no safety pharmacology issues with inhaled Liposomal Amikacin.
In 14 days rat model studies of pseudomonas infection, it was noted that 60 mg/kg of Liposomal Amikacin (75 mg/mL) administered every other day for 14 days (Q2D×7), which effectively delivered half the cumulative dose of aminoglycoside than the other groups, was as effective as 60 mg/kg of Liposomal Amikacin given once per day, and Tobramycin given twice per day daily for 14 days. With 28 day dosing in this model, there were equivalent reductions in CFUs in animals receiving Liposomal Amikacin dosed daily at ˜60 mg/kg or dosed every other day at ˜120 mg/kg. Liposomal Amikacin administered at 120 mg/kg once a day for 14 days was as effective as Tobramycin 60 mg/kg/day (administered twice a day) for 28 days, which suggests a higher AUC and possibly a prolonged post-antibiotic effect with Liposomal Amikacin at 120 mg/kg dosed once per day (see Example 5).
The administration of Liposomal Amikacin via inhalation in the animal model resulted in increased lung (AUC) above the MIC of the bacteria, and demonstrated sustained therapeutic effect, with a reduced frequency, and duration of dosing as compared to Tobramycin. Importantly, the preclinical data for Liposomal Amikacin appear supportive of the hypothesis that this specific formulation may be advantageous over other inhalation products that are hindered by a rapid clearance from lung tissue, necessitating frequent dosing (Geller, Pitlick et al. 2002), which poses a burden for patients and might limit patient compliance.
Additionally, clinical experience demonstrated that nebulized Liposomal Amikacin 50 mg/mL administered as 500 mg once per day for 14 days is well tolerated, and elicits a clinically relevant effect on pulmonary function and decrease in P. aeruginosa density in CF patients. Also, evaluation of the PK data indicates the systemic exposure to Liposomal Amikacin, even at the 500 mg dose, is very low. By either Cmax or AUC or mg of aminoglycoside which is recovered in the urine, the observed systemic exposure to amikacin, associated with Liposomal Amikacin, given by inhalation is approximately ⅕ to ¼ the exposure seen with 600 mg/d of TOBI and is less than 1/200 compared to normal parenteral doses of amikacin. The data further indicate high levels of amikacin are achieved in the sputum. Median AUC values for sputum were 290 and 980 fold greater than the median AUC values for serum on day 1 and day 14 respectively.
Inhaled liposomal amikacin maintains prolonged targeted lung exposures and enhance the uptake of drug to the site of infection. Using data from a human clinical Phase 1b/2a study in which CF patients who were chronically infected with P. aeruginosa received multiple doses of Liposomal Amikacin 50 mg/ml, the objectives of the analyses described herein were three-fold: (1) to use population pharmacokinetic (PK) modeling to characterize amikacin systemic exposure, including approximate systemic bioavailability; (2) to characterize the disposition of liposomal amikacin in sputum; and 3) to characterize the pharmacokinetic-pharmacodynamic (PK-PD) relationship between change in forced expiratory volume in one second (FEV1), change in percent predicted forced expiratory volume in one second (FEV1% predicted), forced expired flow between 25-75% of forced vital capacity (FEF25-75%), and forced vital capacity (FVC), in P. aeruginosa colony forming units (CFU) relative to baseline at Days 7 and 14, and amikacin exposure.
Preclinical Studies with Liposomal Amikacin
Several preclinical studies were conducted with the 20 and 50 mg/mL formulations. Anti-pseudomonas activity of Liposomal Amikacin in in vitro and in vivo models was demonstrated. Additionally, studies confirmed that virulence factors secreted by Pseudomonas facilitate the further release of amikacin from the liposomes, and characterized the deposition and sustained release of amikacin in the lungs of rats, and dogs. The safety of a 30 day administration of Liposomal Amikacin in two species was also established.
Nonclinical pharmacokinetics have demonstrated that the AUC (0-48 hr) of amikacin in the lungs of rats that received a 60 mg/kg dose of Liposomal Amikacin via nebulization, was five-fold higher than the AUC of tobramycin in the lungs of rats that received an equal dose of tobramycin by inhalation. High levels of amikacin were sustained in the lung (>250 μg/mL through 150 hr), suggesting a depot effect. In contrast, lung levels of tobramycin were undetectable within 6 hours of cessation of administration. Conversely, the AUC of drug in the kidneys of rats that received an equal dose of tobramycin was significantly higher than the AUC of rats that received aerosols of Liposomal Amikacin. There were no significant differences in the AUC of aminoglycosides in the serum and urine of the animals; serum levels were undetectable after 24 hr. This profile supports the intended sustained release and depot effect of amikacin in the lung following administration of nebulized Liposomal Amikacin, potentially representing an enhanced efficacy profile. These data for Liposomal Amikacin appear supportive of the hypothesis that this specific formulation may be advantageous over other inhalation products that are hindered by a rapid clearance from lung tissue, necessitating frequent dosing (Geller, Pitlick et al. 2002), and placing a burden on patients. Additionally, toxicokinetic data from 30-day inhalation GLP toxicology studies in rats and dogs showed that there is a 15 fold increase in lung deposition of amikacin dogs as compared to the free amikacin treated group, with comparable plasma and urine levels, indicating high lung concentrations with low systemic exposure.
The pharmacodynamic effect of Liposomal Amikacin was evaluated in vivo in a rat model of chronic pulmonary infection with Pseudomonas (Cash, Woods et al. 1979). In a 14 days Pseudomonas infection model, 60 mg/kg of Liposomal Amikacin (75 mg/mL) was administered every other day for 14 days (Q2D×7). This regimen was as effective as 60 mg/kg of Liposomal Amikacin (given once per day for 14 days), and tobramycin (given twice per day for 14 days). When dosing was extended to 28 days, there were equivalent reductions in CFUs for animals receiving Liposomal Amikacin dosed daily at ˜60 mg/kg or dosed every other day at ˜120 mg/kg. Also, in this experiment, Liposomal Amikacin administered at 120 mg/kg once a day for 14 days was as effective as tobramycin 60 mg/kg/day (administered twice a day) for 28 days. This indicated a higher AUC and a prolonged post-antibiotic effect with Liposomal Amikacin at 120 mg/kg dosed once per day. The preclinical pharmacodynamic data were thus consistent with a sustained antimicrobial benefit enhanced by the site-specific delivery of drug to the lungs via inhalation.
Thus, administration of Liposomal Amikacin via inhalation resulted in increased lung concentrations (AUC) several fold above the MIC of the bacteria, with the potential to provide a sustained therapeutic effect with a reduced frequency and duration of dosing, particularly as compared to Tobramycin.
Data used for this population PK analysis were obtained from two human clinical Phase 1b/2a studies in which CF patients, chronically infected with P. aeruginosa, were administered a total of 500 mg of Liposomal Amikacin daily (in two 20 minute sessions with a 5 minute rest period in between) for 14 days.
Amikacin serum samples were obtained pre-dose, and 1, 2, 4, 6, 8, 12 and 24 hours post-dose on Days 1 and 14, while urine samples were collected over 6 hour intervals on Day 1 and Day 14 for a period of 24 hours. Sputum samples were also collected on Day 1 and Day 14, soon after the dose was administered, between 4 and 6 hours after dosing and prior to dose administration on the following day, as well as on Days 14, 21, and 28. Serum, sputum and urine samples were assayed for amikacin using Liquid Chromatography-Mass Spectrometry/Mass Spectrometry (LC-MS/MS).
Pulmonary function tests (PFT) were carried out during screening from Day −14 to 0) and at baseline (i.e., prior to dose administration on Day 1) and on Day 1, 7, 14, 21, 28, 35, and 42. Sputum samples for microbiology were also collected at baseline and on each of these days. Additional PFTs were carried out 1.5 hours and 3 hours post-dose on Day 1 and Day 14.
Pharmacokinetic Analysis
The data were fit by candidate PK models, using Monte Carlo Parametric Expectation Maximization (MC-PEM), as is implemented in S-ADAPT 1.53, initially fitting the plasma concentrations, then co-modeling the serum and urine data. Model discrimination was based on the fit of the data and change in objective function. The 24 hour area under the curve (AUC) at steady state for serum amikacin values were calculated using the post-hoc parameter estimates from the final population PK model. Covariate relationships between patient demographics and individual post-hoc parameters were assessed first graphically, then by means of statistical models created using SYSTAT® 11 (SYSTAT Software, Inc., Richmond, Calif.). Sputum AUC values from 0 to 24 hours on Day 1 and Day 14 were obtained using the linear trapezoidal rule.
Dependent variables for the PK-PD analysis included the change in PFT values for FEV1, FEV1% predicted, FEF25-75% and FVC, on Day 7 and 14 relative to baseline (prior to dose administration on Day 1) and the change in log10 CFU on each of these days relative to baseline. Independent variables evaluated included the ratio of the average 24 hour AUC for serum and sputum to the baseline minimum inhibitory concentration (MIC), AUC:MIC ratio for P. aeruginosa. The average 24 hour serum and sputum AUC was computed by taking the average of the Day 1 and Day 14 AUC values.
Using a one-sample t-test, the statistical significance of mean changes from baseline for each of the above-described dependent variables was assessed. Using Spearman's rank correlation (rs), the direction and strength of the relationship between each of the dependent variables and AUC:MIC ratio for serum and sputum was assessed. The direction and strength of the relationship between change in each of the PFT values from baseline and change in log10 CFU from baseline were also assessed.
Results
A total of 24 patients completed the two studies with 13 patients from Study 1 and 11 patients from Study 2. The median (min, max) age of all the patients was 23.7 (14, 38) years with a median (range) creatinine clearance (CrCL) at baseline of 126 (76.8, 173) mLmin/1.73 m2.
The most robust fit to the serum concentration data was obtained using a two-compartment model (one absorption site, the lung, and the central compartment) with zero-order drug input into the lungs, a first-order process from lungs to the central compartment and linear elimination. Allowing inter-occasional variation on apparent total clearance (CLt/F) and apparent central volume of distribution (Vc/F) between Day 1 and Day 14 improved the objective function statistically. Urine data was modeled by fitting the amounts of amikacin recovered in the collection intervals, as a function of serum concentrations and renal clearance (CLr). Table 1 is a summary of the fitted PK parameter values.
The goodness of fit for observed versus Bayesian post-hoc individual fitted serum concentration data was excellent, with an overall r2 of 0.98.
The AUC values for the serum and sputum data are shown in Tables 2 and 3, respectively. Median AUC values for sputum were 286 and 978 fold greater than the median AUC values for serum on Day 1 and Day 14, respectively. As evidenced by the higher CV % values, greater variability was evident in sputum (117% on Day 1 and 91.2% on Day 14) compared to serum AUC (51.9% on Day 1 and 42.4% on Day 14) values.
1AUC values in mcg/mL · hr
1AUC values are in mcg/mL · hr
Serum (r2=0.98) and urine (r2=0.38) concentrations were well and modestly fit by model, respectively. On Day 7, 14 and 21, the observed change for FEF25-75% was 0.49 (p<0.001), 0.42 (p=0.02) and 0.34 L/sec (p=0.04), respectively. On Day 7 and 14, the observed change for FEV1 was 0.24 (p=0.002) and 0.13 L (p=0.10), respectively, and was 7.49 (p<0.001) and 4.38 L/sec (p=0.03) for FEV1% predicted. Significant relationships (p≤0.05) between log10 CFU and serum AUC:MIC ratio, and between changes in log10 CFU and FEV1, FEV1% predicted and FVC were identified.
Baseline and Day 14 PFT data were available for all 24 patients and for PFTs carried out on Day 7 and 21, such data were available for 23 patients. Microbiology data were available for all 24 patients. Since MIC values collected prior to dosing on Day 1 for Study 2 were not reported, the screening MIC values as well as CFU counts were used as baseline values.
Using a one-sample t-test, the statistical significance of mean changes from baseline for each of the above-described dependent variables was assessed. Using Spearman's rank correlation (rs), the direction and strength of the relationship between each of the dependent variables and AUC:MIC ratio for serum and sputum was assessed.
Mean changes in PFT values on Day 7 relative to baseline were statistically significant for all PFT endpoints. Mean changes in FEV1% predicted and FEF25-75% on Day 14 relative to baseline were also statistically significant (p=0.029 and p=0.016, respectively). By Day 21, mean change in FEF25-75% relative to baseline was the single PFT that remained statistically significant (p=0.036). Regardless of the study day considered, mean change in log10 CFU from baseline was not statistically significant.
As shown in Table 4, correlations between change in PFT values from baseline and either sputum or serum AUC:MIC ratio were not statistically significant, regardless of whether changes on Day 7 or 14 were evaluated. As shown in Table 5, the correlation between change in log10 CFU from baseline and serum AUC:MIC ratio was statistically significant for both Day 7 or 14. Increasing serum AUC:MIC ratios were associated with larger decreases in log10 CFU on Day 7 (rs=−0.46, p=0.048) and 14 (rs=−0.45, p=0.048) relative to baseline.
Correlations between change in both PFT value and log10 CFU on Day 7 and 14 relative to baseline were statistically significant for FEV1, FEV1% predicted, and FVC (p<0.05).
While mean change in log10 CFU of P. aeruginosa from baseline on both Day 7 and 14 was not statistically significant, the correlation between change in log10 CFU from baseline at both of these time points and serum AUC:MIC ratio was statistically significant; increases in serum AUC:MIC ratio were associated with decreases in log10 CFU. In contrast, this relationship did not hold with sputum AUC:MIC and confirms the large variability in sputum kinetics of Liposomal Amikacin, that is also shown with TOBI (Geller, Pitlick et al. 2002).
The significant relationships between changes in log10 CFU and serum AUC:MIC ratio, and between changes in PFT values and log10 CFU, and the lack of significant decrease in log10 CFU of P. aeruginosa during the two weeks of treatment with liposomal amikacin for inhalation suggests that higher doses may be required to be more reliably effective in a large patient population.
Summary of the Phase 1a/2b Study
Two Phase 1b/2a studies using the Liposomal Amikacin 50 mg/mL have been completed. The two studies were similar in design. A total of 24 CF patients (with FEV1≥40% of predicted) received 500 mg Liposomal Amikacin daily for 14 days. The drug was administered using a PARI LC Star nebulizer, over a period of two 20-minute inhalation sessions with a 5 minute rest period between sessions. There were 13 patients enrolled in Study land 11 patients in Study 2. Patient demographics were similar, with the exception of Pseudomonas MICs at baseline. In Study 1, the mean MIC (μg/mL) was 8 (range 1.5-16) and in Study 2, the mean MIC was 41 μg/mL (range 8-192). The patients enrolled in Study 2 had prior experience with inhalation antibiotics, and per protocol, were permitted to resume treatment with TOBI®/Colistin after Day 28 of the study. The patients in Study 1 were naïve to inhalation antibiotics, and did not receive additional inhalation antibiotics during the follow-up period. The 500 mg dose of Liposomal Amikacin (50 mg/mL) was well tolerated, and in select patients improved pulmonary function and decreased the density of P. aeruginosa in sputum. The details of patient demographics for Studies 1 and 2 (combined) are shown in Table 6.
All efficacy analyses in these human clinical Phase 1b/2a studies were exploratory in nature. The efficacy endpoints included:
Change from Baseline in density of P. aeruginosa (log10 CFU/g) in sputum;
Change from Baseline in pulmonary function tests (FEV1, FEV1% predicted, FVC, and FEF(25-75%)
Changes in P. aeruginosa sputum density, FEV1, and FEV1% predicted at Day 14 were identified as the primary efficacy endpoints.
Quantitative culture of sputum samples and subsequent amikacin susceptibility testing of each morphologically distinct P. aeruginosa were performed. The MIC of amikacin for the isolates with the highest MIC cultured from each subject at screening and Day 14 was documented. The density (CFU per gram of sputum) of P. aeruginosa in sputum was calculated as the log10 value for the sum of all morphotypes.
A summary of the baseline characteristics for the combined population (n=24) are shown in Table 7.
Study 1:
In this study CF patients infected with P. aeruginosa isolates sensitive to amikacin (amikacin MIC <64 μg/mL), and those subjects naïve to inhaled antibiotics were enrolled. Administration of Liposomal Amikacin 500 mg once daily for 2 weeks showed a mean change in log sum of counts of P. aeruginosa from baseline to Day 14 of 1.09 (n=13; 95% confidence interval, 2.09 to 0.09). The reductions in counts were observed in 9 of the 13 subjects. Treatment with Liposomal Amikacin did not result in selection of resistant strains of P. aeruginosa. The mean P. aeruginosa amikacin MIC was 8.04 μg/mL at Day 0 and 30.79 μg/mL at Day 14. On Day 14, a single isolate in one subject had a non sensitive MIC (>25 μg/mL); all other Day 14 isolates were sensitive to amikacin. No human was hospitalized or received intravenous anti-Pseudomonas antibiotics. Additionally, there was improvement in lung function as measured by an increase in FEV1 from baseline to Day 14 of +260 mL (n=13; 95% confidence interval, +30 mL to +500 mL). The corresponding change in FEV1% predicted from baseline to Day 14 was +7.32%. Increases in FEV1 were observed in 9 of the 13 subjects. Also noted were increases in FEF(25-75%) (mean: 570 mL) and FVC (mean: 180 mL).
Study 2:
Study 2 was conducted in a population of CF patients who were infected with P. aeruginosa, and were inhalation antibiotic treatment experienced. In these patients, the administration of Liposomal Amikacin 500 mg q.d. for 2 weeks did not show any significant change in P. aeruginosa density during the study (p-values ≥0.297 for change from Day 1). The proportion of patients with mucoid P. aeruginosa remained constant throughout the study. No statistically significant changes in FEV1, FEV1% predicted, FVC, and FEF(25-75%) were observed after administration of Liposomal Amikacin 500 mg. Nevertheless, trends suggesting improvement in FEV1% predicted, FVC, and FEF(25-75%) were observed at Day 7, Day 14 (end of treatment), and Day 15.
Integrated Efficacy Summary: Studies 1 and 2
Data from the combined population of 24 patients in studies 1 and 2 are summarized below in Tables 8, 9, 10, and 11. The microbiologic end-point of change in log CFU of P. aeruginosa, demonstrated a reduction in bacterial density in the combined population, but this did not achieve statistical significance. But, when data were analyzed from the inhalation antibiotic naïve patients (study 1), a statistically significant reduction in CFU was observed at end of treatment. Factors that might explain this effect are the inherent variability in sputum samples, the inter-laboratory variability in methodology, and reporting of quantitative microbiology, and the enrollment of patients with higher MICs (including resistant isolates) in study 2. All of the above are further compounded by the small sample size of each study.
Assessment of clinical benefit by measurement of pulmonary function tests showed a statistically significant improvement in lung function as measured by an increase in FEV1 from baseline to Day 7 of +240 mL (n=23; p-value 0.0024). The effect at day 14 was a 126 mL increase from baseline in FEV1, which was not statistically significant. A corresponding statistically significant increase in FEV1% predicted from baseline to Day 7 was +7.49% (n=24; p-value 0.0002), and at Day 14 was +4.37% (n=24; p-value 0.0285). The improvement in lung function was also noted with the assessment of small airways as measured by FEF(25-75%) at day 7, an increase in +494 mL (n=23; p-value 0.001), and at Day 14, +423 mL (n=24; p-value 0.0162). These data support a clinically meaningful improvement in lung function in CF patients with chronic Pseudomonas infection who have received a 14 day course of treatment with Liposomal Amikacin.
Two Phase 1 single dose clinical studies were completed with 20 and 50 mg/mL formulations of Liposomal Amikacin in healthy volunteers and in CF patients, respectively. Six healthy volunteers received a single dose of 120 mg of Liposomal Amikacin and tolerated it well, and exhibited prolonged retention of the radiolabeled liposomes in the lungs, with a measured half-life of 46 hours.
Liposomal Amikacin was administered to CF subjects with chronic P. aeruginosa infections in a human clinical Phase I study (Study 3). Single doses of 90 mg (n=6), 270 mg (n=6), or 500 mg (n=4) were administered to CF subjects to evaluate the safety, tolerability and pharmacokinetics of liposomal amikacin for inhalation. A total of 24 patient dosing sessions of a single dose administration of Liposomal Amikacin or placebo by inhalation via the Pari LC Star nebulizer were evaluated. Two serious adverse events were reported (both occurring in placebo group). Both events recovered without sequelae. A total of 41 adverse events (AEs) were experienced by 17 of the 24-patient sessions dosed (71%) during the trial. Of the AEs reported, 10 of the 16 patients (62.5%) who reported adverse events were in the active group and 7 of the 8 patients (87.5%) were in the placebo group. Headache was the most common AE reported in the active group and no patients were discontinued from the study due to AEs. Liposomal Amikacin was well tolerated and safe up to a single dose of 500 mg administered via inhalation.
Additionally, the PK data confirm minimal systemic drug levels, and high sputum levels of drug, and pharmacodynamic modeling estimates long elimination half life presumably due to slow release from liposomes.
The study design is summarized in
In summary, daily administration of 280 mg and 560 mg liposomal amikacin for 28 days appeared safe and well-tolerated. Administration of liposomal amikacin at 280 mg and 560 mg for 28 days results in a dose-dependent improvement in lung function, which is sustained at least for 28 days after the completion of the dosing. The patients receiving liposomal amikacin experienced fewer pulmonary exacerbations (7.14%) compared to those receiving a placebo (18.18%). Additionally, the time to exacerbation was prolonged in the amikacin groups (41 days) compared to the placebo (19 days). The groups receiving amikacin experienced no pulmonary exacerbations during the 28 day treatment period. Patients receiving liposomal amikacin demonstrated greater clinical benefit compared to the placebo group as measured by improvement in the quality of life CFQR-respiratory scale.
The amikacin group had the following change in lung function values (mL):
A comparison of the change in FEV1 from baseline (measured in mL) for all patients in the 560 mg, 280 mg and placebo groups is depicted in
The data from the study also demonstrated a significant reduction in CFU's in patients receiving liposomal amikacin compared to the placebo, and this reduction was sustained at least to day 35. The reduction in CFU was more pronounced for the group receiving 560 mg of amikacin compared to the 280 mg group, as seen in
The pharmacokinetic data revealed high levels of amikacin in the sputum of patients receiving liposomal amikacin, with the mean Cmax (CV) of 3496 (0.973) mcg/g. The mean area under the curve (AUC) value was 13,120 (1.63) mcg/g*hr for the 280 mg group, while the mean AUC was 22,445 (0.831) mcg/g*hr). The serum pharmacokinetic data, on the other hand, demonstrated low systemic exposure to amikacin, with the Cmax mean (SD) of 2.27 (1.58) mcg/mL.
Patients receiving liposomal amikacin also had a reduced frequency and time to pulmonary exacerbation. Table 14:
As seen in Table 14, the percentage of exacerbations in patients treated with liposomal amikacin (including both the 280 mg and 560 mg groups) was lower compared to the placebo group. Moreover, the time to exacerbation was much longer in patients receiving liposomal amikacin (40.6 days) compared to 19.3 days in the placebo group.
Anti-Pseudomonal rescue treatments was also reduced in patients receiving inhaled liposomal amikacin, compared to the placebo group, as seen in Table 15.
As seen in table 15, a lower percentage of patients receiving inhaled liposomal amikacin required anti-Pseudomonal rescue treatment, compared to the placebo group.
Additionally, the time before rescue treatment was needed was reduced in the liposomal amikacin patients (43.0 days) compared to the placebo group (21.3 days).
The aerosol properties of Liposomal Amikacin produced from the eFlow 40 L are shown in Table 15. When compared to nebulizate generated from the LC Star, the mass median aerodynamic diameter (MMAD) values for the eFlow are ˜0.5 μm larger. The actual size dependent mass distributions from both ACI (with eFlow) and NGI (with LC Star) cascade impactors for nebulized Liposomal Amikacin are shown in
The Andersen cascade impactor was used at a flow rate of 28.3 L/min, 18° C., and 50% humidity. The NGI impactor was used at a flow rate of 15 L/min and 5° C. to achieve >60% humidity. *Percent mass of the nominal drug dose that is less than 5 μm in diameter.
The efficacy of Liposomal Amikacin for Inhalation was studied using a model for chronic pulmonary infection (Cash, Woods et al. 1979) where P. aeruginosa, embedded in an agarose bead matrix, was instilled in the trachea of rats. This mucoid Pseudomonas animal model was developed to resemble the chronic Pseudomonas infections seen in CF patients (Cantin and Woods 1999). Rat lungs were inoculated with 104 CFUs of a mucoid P. aeruginosa strain (mucoid strain 3064) originally isolated from a CF patient. Three days later, 60 mg/kg Liposomal Amikacin (75 mg/mL) was administered by inhalation once daily for 14 doses (Q1D×14) or every other day for 7 doses (Q2D×7) (6 mg/kg per dose). For comparison, tobramycin was administered by inhalation BID for 14 days (30 mg/kg per dose for a total of 60 mg/kg daily). There was a significant reduction in bacterial density in all three treatment groups as compared to the saline control (see
As shown in
A multi-cycle, open-label study was conducted in order to evaluate the long-term effects of multiple cycles of treatment with ARIKACE® (liposomal amikacin). The lipid component of ARIKACE® comprises the neutral lipids DPPC and cholesterol.
Patients 6 years of age and above who were previously enrolled in a placebo-controlled Phase 2 study of ARIKACE® were consented to participate in this multiple cycle, 18-month study. The design of the study is shown in
As shown in Table 17, 49 patients were enrolled in the study, and 45 patients completed six treatment cycles. Patient demographics and other baseline data are shown in Table 18. The FEV1 (% predicted) at baseline was 59.2%.
ARIKACE® administered once daily for 6 cycles was well tolerated, as adverse events were consistent with those expected in a population of CF patients. Additionally, no unexpected adverse events were observed. Overall adverse events are shown in Table 19 and Table 20 shows a listing of all adverse events by descending frequency.
The estimated relative change from baseline in FEV1 to end of treatment (day 28) during cycles 1-6 was 7.9% (95% CI +4.3, +11.5%, p<0.0001). This effect was also sustained at the end of the off-treatment period (56 days) during cycles 1-6, as the estimated relative change in FEV1 was 5.7% (95% CI+3.0, +8.5%, p=0.0001,
Additionally, the results indicated that a significant reduction in Pseudomonas aeruginosa density occurred, including for mucoid strains. This effect was sustained over the treatment period of 6 cycles, with each cycle including the 56 day off period (
There was also no significant shift in minimum inhibitory concentration (MIC90) (
The results of the study indicated that 560 mg ARIKACE® given once daily for 28 days followed by 56 days off drug for 6 cycles was well tolerated in cystic fibrosis patients. An increase in FEV1(%) significantly above baseline was observed during the treatment period of each cycle, and was sustained during the 56 day off-drug period.
A study was conducted to determine the biodistribution, retention, and clearance of amikacin and liposomes in rats following multiple inhalation treatments of 90 mg/kg or 10 mg/kg liposomal amikacin (ARIKACE®). Treatment was evaluated by comparing tissue concentrations of amikacin, fluorescence of TAMRA and DilC18(5)-DS and by microscopic evaluation of fluorescence in various sections of the lung.
CDIG female rats were randomized into three groups and received either (1) 90 mg/kg via inhalation for 27 consecutive days, (2) 10 mg/kg of liposomal amikacin via inhalation for 27 consecutive days, or (3) no treatment.
On day 28, rats in each amikacin group were administered a mixture of liposomal amikacin and fluorescently labeled liposomal amikacin (amikacin-TAMRA and DilC18(5)-DS liposomes; 90 mg/kg) by inhalation. At 0 and 4 hours post inhalation and at 1, 3, 7, 14, 21, and 28 days post inhalation, lungs, sera, and urine were collected from 3 rats per group. The concentration of amikacin in tissues was determined by immunopolarization assays. Fluorescence was measured by spectraphotometry. TAMRA and DilC18(5)-DS fluorescence was assessed in sera, urine, and lungs.
Lungs were separated into 3 individual right lobes and into 3 segments of each left lobe. The right caudal lobes of the lungs were frozen and later cut into thin sections (7 μm) for microscopic analysis. At each time point, unfixed tissue sections were examined for the deposition of fluorescently labeled amikacin-TAMRA and DilC18(5)-DS liposomes using a fluorescence microscope. Images of tissues were photographed and examined for differences in fluorescence intensity.
Exposure of healthy adult rats to 28 daily multiple doses of liposomal amikacin via inhalation resulted in the uniform deposition of amikacin and liposomes throughout the lung in both 90 mg/kg and 10 mg/kg groups. The amount of amikacin deposited per gram of lung tissue was dose dependent. For each dose, the amount of amikacin deposited per gram of lung tissue was the same regardless of which lung lobe was evaluated or what section of a lobe was assayed. Amikacin was cleared from the lung uniformly. Liposomes labeled with DilC18(5)-DS were cleared more slowly from the lungs than amikacin, resulting in the lack of measureable levels of DilC18(5)-DS in the serum or urine. Sequenced microscopic evaluations of lung tissues dosed with dual fluorescently labeled ARIKACE® revealed that the lipid DilC18(5)-DS within the liposomes was primarily sequestered in the macrophages within the alveolar spaces and parenchyma during the entire observation period. Amikacin-TAMRA presented as both diffused and macrophage associated fluorescence during the entire observation period.
The results of the study indicated that exposure to multiple doses of aerosolized liposomal amikacin, regardless of the dose, resulted in a uniform deposition of amikacin and liposomes throughout the entire lung followed by uniform clearance of amikacin from the lung of normal rats.
All of the U.S. patents and U.S. published patent applications cited herein are hereby incorporated by reference.
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
This application claims priority from U.S. Provisional Application No. 61/489,940, filed May 25, 2011, and is a continuation of U.S. application Ser. No. 14/809,128, filed Jul. 24, 2015, now U.S. Pat. No. 9,737,555, which is a continuation of U.S. application Ser. No. 13/480,246, filed May 24, 2012, now U.S. Pat. No. 9,119,783, which is a continuation in part of U.S. application Ser. No. 12/250,412, filed on Oct. 13, 2008, now U.S. Pat. No. 9,114,081, which is a continuation in part of International Application No. PCT/US08/062868, filed on May 7, 2008, which claims priority from U.S. Provisional Application No. 60/916,342, filed on May 7, 2007, all of which are hereby incorporated by reference in their entirety.
Number | Name | Date | Kind |
---|---|---|---|
3091572 | Luedemann et al. | May 1963 | A |
3136704 | Charney | Jun 1964 | A |
4235871 | Paphadjopoulos et al. | Nov 1980 | A |
4372949 | Kodama et al. | Feb 1983 | A |
4394448 | Szoka, Jr. et al. | Jul 1983 | A |
4396630 | Riedl et al. | Aug 1983 | A |
4451447 | Kaplan et al. | May 1984 | A |
4515736 | Deamer | May 1985 | A |
4522803 | Lenk et al. | Jun 1985 | A |
4547490 | Ecanow et al. | Oct 1985 | A |
4588578 | Fountain et al. | May 1986 | A |
4606939 | Frank et al. | Aug 1986 | A |
4684625 | Eppstein et al. | Aug 1987 | A |
4693999 | Axelsson et al. | Sep 1987 | A |
4721612 | Janoff et al. | Jan 1988 | A |
4767874 | Shima et al. | Aug 1988 | A |
4833134 | Kishimoto et al. | May 1989 | A |
4857311 | Domb et al. | Aug 1989 | A |
4895452 | Yiournas et al. | Jan 1990 | A |
4895719 | Radhakrishnan et al. | Jan 1990 | A |
4897384 | Janoff et al. | Jan 1990 | A |
4933121 | Law et al. | Jun 1990 | A |
4952405 | Yau-Young | Aug 1990 | A |
4963367 | Ecanow | Oct 1990 | A |
4975282 | Cullis et al. | Dec 1990 | A |
4981692 | Popescu et al. | Jan 1991 | A |
5000958 | Fountain et al. | Mar 1991 | A |
5006343 | Benson et al. | Apr 1991 | A |
5008050 | Cullis et al. | Apr 1991 | A |
5023087 | Yau-Young | Jun 1991 | A |
5030453 | Lenk et al. | Jul 1991 | A |
5041278 | Janoff et al. | Aug 1991 | A |
5049388 | Knight et al. | Sep 1991 | A |
5049389 | Radhakrishnan | Sep 1991 | A |
5059421 | Loughrey et al. | Oct 1991 | A |
5059591 | Janoff et al. | Oct 1991 | A |
5077056 | Bally et al. | Dec 1991 | A |
5169637 | Lenk et al. | Dec 1992 | A |
5178876 | Khokhar et al. | Jan 1993 | A |
5192549 | Barenolz et al. | Mar 1993 | A |
5211955 | Legros et al. | May 1993 | A |
5252339 | Cristofori et al. | Oct 1993 | A |
5264618 | Felgner et al. | Nov 1993 | A |
5269979 | Fountain | Dec 1993 | A |
5279833 | Rose | Jan 1994 | A |
5316771 | Barenholz et al. | May 1994 | A |
5320906 | Eley et al. | Jun 1994 | A |
5334761 | Gebeyehu et al. | Aug 1994 | A |
5401511 | Margalit | Mar 1995 | A |
5409704 | Bally et al. | Apr 1995 | A |
5415867 | Minchey et al. | May 1995 | A |
5459127 | Felgner et al. | Oct 1995 | A |
5508269 | Smith et al. | Apr 1996 | A |
5540936 | Coe et al. | Jul 1996 | A |
5543152 | Webb et al. | Aug 1996 | A |
5549102 | Lintl et al. | Aug 1996 | A |
5569464 | Endo et al. | Oct 1996 | A |
5578320 | Janoff et al. | Nov 1996 | A |
5580575 | Unger et al. | Dec 1996 | A |
5596982 | Blaha-Schnabel | Jan 1997 | A |
5610198 | Barry, III et al. | Mar 1997 | A |
5614216 | Janoff | Mar 1997 | A |
5616334 | Janoff et al. | Apr 1997 | A |
5616341 | Mayer et al. | Apr 1997 | A |
5631018 | Zalipsky et al. | May 1997 | A |
5641662 | Debs et al. | Jun 1997 | A |
5643599 | Lee et al. | Jul 1997 | A |
5662929 | Legace et al. | Sep 1997 | A |
5665383 | Grinstaff et al. | Sep 1997 | A |
5723147 | Kim et al. | Mar 1998 | A |
5736155 | Bally et al. | Apr 1998 | A |
5740966 | Blaha-Schnabel | Apr 1998 | A |
5741516 | Webb et al. | Apr 1998 | A |
5753613 | Ansell et al. | May 1998 | A |
5756120 | Hersch et al. | May 1998 | A |
5756121 | Bracken | May 1998 | A |
5756353 | Debs | May 1998 | A |
5759571 | Hersch et al. | Jun 1998 | A |
5766627 | Sankaram et al. | Jun 1998 | A |
5785987 | Hope et al. | Jul 1998 | A |
5795589 | Mayer et al. | Aug 1998 | A |
5814335 | Webb et al. | Sep 1998 | A |
5820848 | Boni et al. | Oct 1998 | A |
5823178 | Lloyd et al. | Oct 1998 | A |
5837279 | Janoff et al. | Nov 1998 | A |
5837282 | Fenske et al. | Nov 1998 | A |
5840702 | Bedwell | Nov 1998 | A |
5843473 | Woodle et al. | Dec 1998 | A |
5849490 | Schonwetter et al. | Dec 1998 | A |
5861159 | Pardoll et al. | Jan 1999 | A |
5871710 | Bogdanov et al. | Feb 1999 | A |
5875776 | Vaghefi | Mar 1999 | A |
5883074 | Boggs et al. | Mar 1999 | A |
5891468 | Martin et al. | Apr 1999 | A |
5922350 | Janoff et al. | Jul 1999 | A |
5939096 | Clerc et al. | Aug 1999 | A |
5945122 | Abra et al. | Aug 1999 | A |
5958449 | Hersch et al. | Sep 1999 | A |
5967389 | Wunderlich et al. | Sep 1999 | A |
5965549 | Purwar et al. | Oct 1999 | A |
5972379 | Guo et al. | Oct 1999 | A |
5993850 | Sankaram et al. | Nov 1999 | A |
6000394 | Blaha-Schnabel et al. | Dec 1999 | A |
6045828 | Bystrom et al. | Apr 2000 | A |
6051251 | Zalipsky et al. | Apr 2000 | A |
6051549 | Roberts et al. | Apr 2000 | A |
6085741 | Becker | Jul 2000 | A |
6086851 | Boni et al. | Jul 2000 | A |
6090407 | Knight et al. | Jul 2000 | A |
6106479 | Wunderlich et al. | Aug 2000 | A |
6106858 | Ye et al. | Aug 2000 | A |
6124273 | Drohan et al. | Sep 2000 | A |
6147060 | Zasloff et al. | Nov 2000 | A |
6162462 | Bolotin et al. | Dec 2000 | A |
6176237 | Wunderlich et al. | Jan 2001 | B1 |
6197333 | Onyuksel et al. | Mar 2001 | B1 |
6211162 | Dale et al. | Apr 2001 | B1 |
6221385 | Camu et al. | Apr 2001 | B1 |
6221388 | Hersch et al. | Apr 2001 | B1 |
6228346 | Zhang et al. | May 2001 | B1 |
6228393 | DiCosmo et al. | May 2001 | B1 |
6235177 | Borland et al. | May 2001 | B1 |
6274175 | Gombotz et al. | Aug 2001 | B1 |
6316024 | Allen et al. | Nov 2001 | B1 |
6338859 | Leroux et al. | Jan 2002 | B1 |
6348069 | Vacanti et al. | Feb 2002 | B1 |
6352996 | Cao et al. | Mar 2002 | B1 |
6355267 | Collins | Mar 2002 | B1 |
6387886 | Montgomery et al. | May 2002 | B1 |
6419901 | Placke et al. | Jul 2002 | B2 |
6440393 | Waldrep et al. | Aug 2002 | B1 |
6443898 | Unger et al. | Sep 2002 | B1 |
6447753 | Edwards et al. | Sep 2002 | B2 |
6451784 | Placke et al. | Sep 2002 | B1 |
6458373 | Lambert et al. | Oct 2002 | B1 |
6468532 | Hsei et al. | Oct 2002 | B1 |
6475779 | Mathiowitz et al. | Nov 2002 | B2 |
6481438 | Gallem et al. | Nov 2002 | B1 |
6492560 | Wilbur et al. | Dec 2002 | B2 |
6497901 | Royer | Dec 2002 | B1 |
6509323 | Davis et al. | Jan 2003 | B1 |
6511676 | Boulikas | Jan 2003 | B1 |
6513727 | Jaser et al. | Feb 2003 | B1 |
6518243 | Kahne et al. | Feb 2003 | B1 |
6521211 | Unger et al. | Feb 2003 | B1 |
6521736 | Watterson et al. | Feb 2003 | B2 |
6534018 | Baker et al. | Mar 2003 | B1 |
6546927 | Litherland et al. | Apr 2003 | B2 |
6554201 | Klimowicz et al. | Apr 2003 | B2 |
6596305 | Edgerly-Plug | Jul 2003 | B1 |
6599912 | Au et al. | Jul 2003 | B1 |
6606990 | Stapleton et al. | Aug 2003 | B2 |
6613352 | Lagace et al. | Sep 2003 | B2 |
6615824 | Power | Sep 2003 | B2 |
6623671 | Coe et al. | Sep 2003 | B2 |
6629646 | Ivri | Oct 2003 | B1 |
6676034 | Tanaka et al. | Jan 2004 | B2 |
6679251 | Gallem et al. | Jan 2004 | B1 |
6759057 | Weiner et al. | Jul 2004 | B1 |
6770291 | Smyth-Templeton et al. | Aug 2004 | B2 |
6843942 | Katinger et al. | Jan 2005 | B2 |
6845770 | Klimowicz et al. | Jan 2005 | B2 |
6855296 | Baker et al. | Feb 2005 | B1 |
6890555 | Desai et al. | May 2005 | B1 |
6900184 | Cohen et al. | May 2005 | B2 |
6915962 | Power et al. | Jul 2005 | B2 |
6916490 | Garver et al. | Jul 2005 | B1 |
6948491 | Loeffler et al. | Sep 2005 | B2 |
6962151 | Knoch et al. | Nov 2005 | B1 |
6983747 | Gallem et al. | Jan 2006 | B2 |
6991809 | Anderson | Jan 2006 | B2 |
7059320 | Feiner et al. | Jun 2006 | B2 |
7063860 | Chancellor et al. | Jun 2006 | B2 |
7077126 | Kummer et al. | Jul 2006 | B2 |
7100600 | Loeffler et al. | Sep 2006 | B2 |
7104463 | Litherland et al. | Sep 2006 | B2 |
7131440 | Sonntag | Nov 2006 | B2 |
7244413 | Barbera-Guillem | Jul 2007 | B2 |
7252085 | Kunschir | Aug 2007 | B2 |
7255106 | Gallem et al. | Aug 2007 | B2 |
7297344 | Fleischer et al. | Nov 2007 | B1 |
7331339 | Smith et al. | Feb 2008 | B2 |
7368102 | Tarara et al. | May 2008 | B2 |
D583928 | Knoch | Dec 2008 | S |
7458372 | Feiner et al. | Dec 2008 | B2 |
7472701 | Pfichner et al. | Jan 2009 | B2 |
7544369 | Boni et al. | Jun 2009 | B2 |
7600511 | Power et al. | Oct 2009 | B2 |
7686014 | Boehm et al. | Mar 2010 | B2 |
7718189 | Boni et al. | May 2010 | B2 |
7748377 | Smith et al. | Jul 2010 | B2 |
7758886 | Jauernig et al. | Jul 2010 | B2 |
7771642 | Power et al. | Aug 2010 | B2 |
7779838 | Hetzer et al. | Aug 2010 | B2 |
7879351 | Li et al. | Feb 2011 | B2 |
7891352 | Gallem et al. | Feb 2011 | B2 |
7931212 | Urich et al. | Apr 2011 | B2 |
D638117 | Eckstein et al. | May 2011 | S |
7958887 | Kelliher et al. | Jun 2011 | B2 |
7930247 | Boehm et al. | Jul 2011 | B2 |
7971588 | Fink et al. | Jul 2011 | B2 |
8006693 | Boehm et al. | Aug 2011 | B2 |
8071127 | Cipolla et al. | Dec 2011 | B2 |
D652908 | Eckstein et al. | Jan 2012 | S |
8100162 | Joern et al. | Jan 2012 | B2 |
8113194 | Boehm et al. | Feb 2012 | B2 |
8119156 | Cipolla et al. | Feb 2012 | B2 |
D656804 | Eckstein et al. | Mar 2012 | S |
8226975 | Weers | Jul 2012 | B2 |
8263645 | Keller | Sep 2012 | B2 |
8268347 | Cipolla et al. | Sep 2012 | B1 |
8333187 | Gallem et al. | Dec 2012 | B2 |
8342171 | Boehm et al. | Jan 2013 | B2 |
8347878 | Schuschnig et al. | Jan 2013 | B2 |
8387895 | Stangl | Mar 2013 | B2 |
8398001 | Borland et al. | Mar 2013 | B2 |
D680214 | Eckstein et al. | Apr 2013 | S |
8414915 | Cipolla et al. | Apr 2013 | B2 |
8459252 | Gallem et al. | Jun 2013 | B2 |
8511581 | Urich et al. | Aug 2013 | B2 |
8596264 | Sommer | Dec 2013 | B2 |
8616195 | Power et al. | Dec 2013 | B2 |
8632804 | Weers | Jan 2014 | B2 |
8642075 | Weers | Feb 2014 | B2 |
8671933 | Boehm et al. | Mar 2014 | B2 |
8673348 | Weers | Mar 2014 | B2 |
8673349 | Weers | Mar 2014 | B2 |
8679532 | Weers | Mar 2014 | B2 |
8720432 | Borgschulte et al. | May 2014 | B2 |
8720435 | Gallem et al. | May 2014 | B2 |
8739777 | Kreutzmann et al. | Jun 2014 | B2 |
8802137 | Boni et al. | Aug 2014 | B2 |
8852557 | Keller et al. | Oct 2014 | B2 |
8985100 | Minocchieri et al. | Mar 2015 | B2 |
9016272 | Gallem et al. | Apr 2015 | B2 |
9027548 | Borgschulte et al. | May 2015 | B2 |
9028864 | Cipolla et al. | May 2015 | B2 |
9046092 | Boehm et al. | Jun 2015 | B2 |
9061303 | Waldner et al. | Jun 2015 | B2 |
9072464 | Haartsen et al. | Jul 2015 | B2 |
9078897 | Cipolla et al. | Jul 2015 | B1 |
9084862 | Blakey et al. | Jul 2015 | B2 |
9095676 | Gallem et al. | Aug 2015 | B2 |
9108211 | Ivri | Aug 2015 | B2 |
9114081 | Gupta | Aug 2015 | B2 |
9119783 | Gupta | Sep 2015 | B2 |
9119930 | Kreutzmann et al. | Sep 2015 | B2 |
9149588 | Gordon et al. | Oct 2015 | B2 |
9161963 | Keller et al. | Oct 2015 | B2 |
9168556 | Pumm et al. | Oct 2015 | B2 |
9198859 | Keller et al. | Dec 2015 | B2 |
9259424 | Cipolla et al. | Feb 2016 | B2 |
9265900 | Loenner et al. | Feb 2016 | B2 |
9333214 | Gupta | May 2016 | B2 |
9402845 | Weers | Aug 2016 | B2 |
9511082 | Weers | Dec 2016 | B2 |
9549925 | Weers | Jan 2017 | B2 |
9549939 | Weers | Jan 2017 | B2 |
9724301 | Gupta | Aug 2017 | B2 |
9737555 | Gupta | Aug 2017 | B2 |
9827317 | Boni et al. | Nov 2017 | B2 |
9895385 | Eagle et al. | Feb 2018 | B2 |
9925205 | Malinin | Mar 2018 | B2 |
20010006660 | Legace et al. | Jul 2001 | A1 |
20020035061 | Krieger et al. | Mar 2002 | A1 |
20020052390 | Ponikau | May 2002 | A1 |
20020086852 | Cantor et al. | Jul 2002 | A1 |
20020187105 | Zou et al. | Dec 2002 | A1 |
20030039615 | Katz | Feb 2003 | A1 |
20030059375 | Perez-Soler et al. | Mar 2003 | A1 |
20030096774 | Gonda et al. | May 2003 | A1 |
20030099697 | Panzner et al. | May 2003 | A1 |
20030118636 | Friesen et al. | Jun 2003 | A1 |
20030138481 | Zadi | Jul 2003 | A1 |
20030148964 | Dunne | Aug 2003 | A1 |
20030224039 | Boni et al. | Dec 2003 | A1 |
20040009126 | Pilkiewicz et al. | Jan 2004 | A1 |
20040032037 | Katinger et al. | Feb 2004 | A1 |
20040091541 | Unger | May 2004 | A1 |
20040101553 | Lee et al. | May 2004 | A1 |
20040142025 | MacLachlan et al. | Jul 2004 | A1 |
20040142026 | Boni et al. | Jul 2004 | A1 |
20040156888 | Jensen et al. | Aug 2004 | A1 |
20040180082 | Kang et al. | Sep 2004 | A1 |
20050019926 | Gonda et al. | Jan 2005 | A1 |
20050025822 | Wong et al. | Feb 2005 | A1 |
20050042341 | Thomas et al. | Feb 2005 | A1 |
20050113337 | Taneja et al. | May 2005 | A1 |
20050214224 | Weers et al. | Sep 2005 | A1 |
20050217666 | Fink et al. | Oct 2005 | A1 |
20050220752 | Charmot et al. | Oct 2005 | A1 |
20050249795 | Zhang et al. | Nov 2005 | A1 |
20060062738 | Hofmann et al. | Mar 2006 | A1 |
20060067998 | Kurzrock et al. | Mar 2006 | A1 |
20060073198 | Boni et al. | Apr 2006 | A1 |
20060110441 | Wong et al. | May 2006 | A1 |
20060217603 | Nagai et al. | Sep 2006 | A1 |
20060286038 | Rairkar et al. | Dec 2006 | A1 |
20070065367 | Condos et al. | Mar 2007 | A1 |
20070077290 | Li et al. | Apr 2007 | A1 |
20070081963 | Oh et al. | Apr 2007 | A1 |
20070105758 | May et al. | May 2007 | A1 |
20070196461 | Weers | Aug 2007 | A1 |
20070267010 | Fink et al. | Nov 2007 | A1 |
20080089927 | Malinin | Apr 2008 | A1 |
20080108104 | Eckstein et al. | May 2008 | A1 |
20080131497 | Perkins et al. | Jun 2008 | A1 |
20080246472 | Igney et al. | Oct 2008 | A1 |
20090104256 | Gupta | Apr 2009 | A1 |
20090104257 | Li et al. | Apr 2009 | A1 |
20090105126 | Li et al. | Apr 2009 | A1 |
20090269396 | Cipolla et al. | Oct 2009 | A1 |
20090274754 | Cipolla et al. | Nov 2009 | A1 |
20100068257 | Boni et al. | Mar 2010 | A1 |
20100196455 | Malinin | Aug 2010 | A1 |
20100260829 | Boni et al. | Oct 2010 | A1 |
20110064796 | Cipolla et al. | Mar 2011 | A1 |
20110150983 | Cipolla et al. | Jun 2011 | A1 |
20110159079 | Li et al. | Jun 2011 | A1 |
20110256175 | Hope et al. | Oct 2011 | A1 |
20120010162 | Norling | Jan 2012 | A1 |
20120077786 | Byron et al. | Mar 2012 | A1 |
20120192861 | Surber | Aug 2012 | A1 |
20120244206 | Cipolla et al. | Sep 2012 | A1 |
20130028960 | Weers | Jan 2013 | A1 |
20130034534 | Kroneberg et al. | Feb 2013 | A1 |
20130052260 | Weers | Feb 2013 | A1 |
20130064883 | Weers | Mar 2013 | A1 |
20130071468 | Weers | Mar 2013 | A1 |
20130071469 | Weers | Mar 2013 | A1 |
20130087480 | Stark et al. | Apr 2013 | A1 |
20130089598 | Gupta | Apr 2013 | A1 |
20130121918 | Hong et al. | May 2013 | A1 |
20130136788 | Gupta | May 2013 | A1 |
20130177629 | Martin et al. | Jul 2013 | A1 |
20130330400 | Perkins et al. | Dec 2013 | A1 |
20130330440 | Fulgham | Dec 2013 | A1 |
20140018431 | Wade et al. | Jan 2014 | A1 |
20140072620 | Weers | Mar 2014 | A1 |
20140248335 | Malinin | Sep 2014 | A1 |
20140308304 | Manoharan et al. | Oct 2014 | A1 |
20140314835 | Boni et al. | Oct 2014 | A1 |
20140371293 | Brown et al. | Dec 2014 | A1 |
20150110855 | Cipolla et al. | Apr 2015 | A1 |
20150272880 | Seidel et al. | Oct 2015 | A1 |
20150283076 | Cipolla et al. | Oct 2015 | A1 |
20150283133 | Gonda et al. | Oct 2015 | A1 |
20150306173 | Chen et al. | Oct 2015 | A1 |
20150314002 | Perkins et al. | Nov 2015 | A1 |
20150328244 | Eagle et al. | Nov 2015 | A1 |
20160113927 | Weers | Apr 2016 | A1 |
20160120806 | Cipolla et al. | May 2016 | A1 |
20160143849 | Gupta | May 2016 | A1 |
20160151402 | Gupta | Jun 2016 | A1 |
20160184301 | Weers | Jun 2016 | A1 |
20160184302 | Weers | Jun 2016 | A1 |
20160271125 | Boni et al. | Sep 2016 | A1 |
20160317563 | Weers | Nov 2016 | A1 |
20160317564 | Weers | Nov 2016 | A1 |
20160354371 | Weers | Dec 2016 | A1 |
20170014342 | Li et al. | Jan 2017 | A1 |
20170087155 | Weers | Mar 2017 | A1 |
20170100420 | Boni et al. | Apr 2017 | A1 |
20170165374 | Perkins et al. | Jun 2017 | A1 |
20170196900 | Perkins et al. | Jul 2017 | A1 |
20170360816 | Eagle et al. | Dec 2017 | A1 |
Number | Date | Country |
---|---|---|
2174803 | Oct 1997 | CA |
2101241 | Dec 1998 | CA |
2215716 | Dec 1999 | CA |
2614764 | Jan 2007 | CA |
2838111 | Jun 2007 | CA |
1747738 | Mar 2006 | CN |
0069307 | Jan 1983 | EP |
0274431 | May 1994 | EP |
0652008 | May 1995 | EP |
1083881 | Mar 2001 | EP |
1083886 | Mar 2001 | EP |
1190705 | Mar 2002 | EP |
1332755 | Aug 2003 | EP |
0825852 | Jul 2004 | EP |
1559431 | Aug 2005 | EP |
2457609 | May 2012 | EP |
2145107 | Mar 1985 | GB |
S63-500175 | Jan 1988 | JP |
S63-239213 | Oct 1988 | JP |
6-345663 | Dec 1994 | JP |
H10-511363 | Nov 1998 | JP |
11-080022 | Mar 1999 | JP |
2006-028069 | Feb 2006 | JP |
2006-514016 | Apr 2006 | JP |
2006-514682 | May 2006 | JP |
27298 | Oct 2007 | UA |
27804 | Nov 2007 | UA |
WO 8500968 | Mar 1985 | WO |
WO 8606959 | Dec 1986 | WO |
WO 8700043 | Jan 1987 | WO |
WO 8702219 | Apr 1987 | WO |
WO 8804573 | Jun 1988 | WO |
WO 9109616 | Jul 1991 | WO |
WO 9116882 | Nov 1991 | WO |
WO 9312240 | Jun 1993 | WO |
WO 9412155 | Jun 1994 | WO |
WO 9412156 | Jun 1994 | WO |
WO 9422430 | Oct 1994 | WO |
WO 9608235 | Mar 1996 | WO |
WO 9619199 | Jun 1996 | WO |
WO 9619972 | Jul 1996 | WO |
WO 1996037194 | Nov 1996 | WO |
WO 9729851 | Aug 1997 | WO |
WO 9930686 | Jun 1999 | WO |
WO 9951202 | Oct 1999 | WO |
WO 9961003 | Dec 1999 | WO |
WO 9965466 | Dec 1999 | WO |
WO 0027359 | May 2000 | WO |
WO 0029103 | May 2000 | WO |
WO 0045791 | Aug 2000 | WO |
WO 0100173 | Jan 2001 | WO |
WO 0105373 | Jan 2001 | WO |
WO 0115678 | Mar 2001 | WO |
WO 0118280 | Mar 2001 | WO |
WO 0132246 | May 2001 | WO |
WO 2002032400 | Apr 2002 | WO |
WO 2002043699 | Jun 2002 | WO |
WO 2003045965 | Jun 2003 | WO |
WO 2003075889 | Sep 2003 | WO |
WO 2003075890 | Sep 2003 | WO |
WO 2004002453 | Jan 2004 | WO |
WO 2004047802 | Jun 2004 | WO |
WO 2004054499 | Jul 2004 | WO |
WO 2004091623 | Oct 2004 | WO |
WO 2004110346 | Dec 2004 | WO |
WO 2004110493 | Dec 2004 | WO |
WO 2006108556 | Oct 2006 | WO |
WO 2007011940 | Jan 2007 | WO |
WO 2007012191 | Feb 2007 | WO |
WO 2007067520 | Jun 2007 | WO |
WO 2007117509 | Oct 2007 | WO |
WO 2007117550 | Oct 2007 | WO |
WO 2008039989 | Apr 2008 | WO |
WO 2008063341 | May 2008 | WO |
WO 2008137717 | Nov 2008 | WO |
WO 2008137917 | Nov 2008 | WO |
WO 2009045116 | Apr 2009 | WO |
WO 2009055568 | Apr 2009 | WO |
WO 2009055571 | Apr 2009 | WO |
WO 2009126502 | Oct 2009 | WO |
WO 2010045209 | Apr 2010 | WO |
WO 2010111641 | Sep 2010 | WO |
WO 2011050206 | Apr 2011 | WO |
WO 2012050945 | Apr 2012 | WO |
WO 2012069531 | May 2012 | WO |
WO 2012159103 | Nov 2012 | WO |
WO 2013177226 | Nov 2013 | WO |
WO 2014052634 | Apr 2014 | WO |
WO 2014085526 | Jun 2014 | WO |
WO 2015017807 | Feb 2015 | WO |
WO 2015175939 | Nov 2015 | WO |
WO 2017008076 | Jan 2017 | WO |
Entry |
---|
Office Action for U.S. Appl. No. 12/598,830, dated Mar. 7, 2012, 5 pages. |
Office Action for U.S. Appl. No. 12/598,830, dated Oct. 23, 2012, 7 pages. |
Office Action for U.S. Appl. No. 12/598,830, dated Apr. 18, 2014, 5 pages. |
Office Action for U.S. Appl. No. 12/598,830, dated Nov. 28, 2014, 8 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2008/062469, dated Sep. 18, 2008, 9 pages. |
International Preliminary Report on Patentability for International Application No. PCT/US2008/062469, dated Nov. 10, 2009, 7 pages. |
Office Action for U.S. Appl. No. 12/250,412, dated Dec. 2, 2011, 7 pages. |
Office Action for U.S. Appl. No. 12/250,412, dated Jun. 27, 2011, 6 pages. |
Office Action for U.S. Appl. No. 12/250,412, dated Mar. 24, 2014, 9 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2008/062868, dated Sep. 18, 2008, 7 pages. |
International Preliminary Report on Patentability for International Application No. PCT/US2008/062868, dated Nov. 10, 2009, 5 pages. |
Examination Report for Australian Patent Application No. 2009303542, dated Jun. 20, 2012, 3 pages. |
Office Action for Canadian Application No. 2,739,954, dated Apr. 28, 2015, 4 pages. |
Office Action for Canadian Application No. 2,739,954, dated Mar. 4, 2016, 7 pages. |
Office Action for Chilean Application No. 814-2011, dated Apr. 8, 2014, 6 pages. |
Office Action for Chilean Application No. 814-2011, dated Nov. 4, 2014, 5 pages. |
Office Action for Chinese Patent Application No. 200980140740.2, dated Jul. 3, 2012, 2 pages. |
Office Action for Chinese Patent Application No. 200980140740.2, dated Jun. 4, 2013, 7 pages. |
Office Action for Chinese Application No. 200980140740.2, dated Mar. 5, 2014, 5 pages. |
Notification of Reexamination for Chinese Application No. 200980140740.2, dated Aug. 26, 2015, 11 pages. |
Decision of Reexamination for Chinese Application No. 200980140740.2, dated Feb. 14, 2016, 25 pages. |
Supplementary European Search Report for European Application No. 09821103.0, dated Aug. 12, 2015, 10 pages. |
Office Action for Indonesian Application No. W00201101412, dated Feb. 28, 2014, 1 page. |
Office Action for Israel Application No. 212268, dated Dec. 9, 2013, 2 pages. |
Office Action for Israel Application No. 212268, dated Apr. 28, 2015, 2 pages. |
First Examination Report for Indian Application No. 2600/DELNP/2011, dated Aug. 24, 2015, 2 pages. |
Office Action for Japanese Application No. 2011-532180, dated May 1, 2014, 7 pages. |
Office Action for Japanese Application No. 2011-532180, dated Jan. 23, 2014, 4 pages. |
Notice of Grounds for Rejection for Korean Application No. 2011-7008430, dated Sep. 3, 2015, 3 pages. |
Notice of Grounds for Rejection for Korean Application No. 2011-7008430, dated Jul. 21, 2016, 4 pages. |
Office Action for New Zealand Patent Application No. 592217, dated Sep. 1, 2011, 2 pages. |
Office Action for New Zealand Patent Application No. 592217, dated Feb. 5, 2013, 2 pages. |
Office Action for Philippine Application No. 12011500726, dated Apr. 24, 2014, 1 page. |
Office Action for Philippine Application No. 12011500726, dated Nov. 27, 2015, 3 pages. |
Office Action for Russian Application No. 2011119018, dated Mar. 5, 2014, 3 pages. |
Search Report and Written Opinion for Singapore Application No. 201102419-7, dated Sep. 7, 2012, 12 pages. |
Second Written Opinion for Singapore Application No. 201102419-7, dated Sep. 4, 2014, 13 pages. |
Office Action for Ukraine Application No. 201105955, dated Jun. 7, 2013, 2 pages. |
Written Opinion for International Application No. PCT/US2009/060468, dated Jun. 24, 2010, 3 pages. |
International Preliminary Report on Patentability for International Application No. PCT/US2009/060468, dated Apr. 19, 2011, 4 pages. |
Office Action for Australian Application No. 2014201765, dated Apr. 9, 2015, 3 pages. |
First Office Action for Chinese Application No. 201410564453.7, dated Nov. 28, 2016, 6 pages. |
Office Action for Japanese Application No. 2014-075240, dated Mar. 9, 2015, 2 pages. |
Office Action for Japanese Application No. 2014-075240, dated Dec. 10, 2015, 5 pages. |
Office Action for Japanese Application No. 2014-075240, dated Aug. 23, 2016, 7 pages. |
First Examination Report for New Zealand Patent Application No. 606383, dated Feb. 5, 2013, 2 pages. |
Office Action for U.S. Appl. No. 13/480,246, dated Jan. 10, 2013, 7 pages. |
Office Action for U.S. Appl. No. 13/480,246, dated Apr. 10, 2014, 7 pages. |
Office Action for U.S. Appl. No. 13/566,707, dated May 27, 2015, 11 pages. |
Office Action for U.S. Appl. No. 14/809,127, dated Jun. 16, 2016, 10 pages. |
Office Action for Australian Patent Application No. 2003304204, dated Jun. 25, 2008, 3 pages. |
Office Action for Canadian Patent Application No. 2504317, dated Jan. 27, 2011, 2 pages. |
Office Action for Canadian Patent Application No. 2504317, dated Jun. 16, 2010, 3 pages. |
First Office Action for Chinese Patent Application No. 200380106534.2, dated Aug. 11, 2006, 3 pages. |
Second Office Action for Chinese Patent Application No. 200380106534.2 [no date], 4 pages. |
Third Office Action for Chinese Patent Application No. 200380106534.2, dated May 22, 2009, 4 pages. |
Supplementary European Search Report for European Application No. 03816990.0, dated Jan. 12, 2009, 5 pages. |
Summons to Attend Oral Hearing for European Application No. 03816990.0, dated Dec. 21, 2011, 7 pages. |
Office Action for European Application No. 03816990.0, dated Jun. 17, 2011, 5 pages. |
Office Action for European Application No. 03816990.0, dated Apr. 24, 2009, 6 pages. |
Office Action for European Application No. 03816990.0, dated Jun. 5, 2012, 3 pages. |
Office Action for Israel Application No. 168279, dated Mar. 19, 2014, 2 pages. |
Office Action for Israel Patent Application No. 168279, dated Nov. 3, 2010, 1 page. |
Office Action for Israel Patent Application No. 168279, dated Aug. 17, 2009, 2 pages. |
Office Action for Israel Patent Application No. 168279, dated Jun. 23, 2008, 1 page. |
Office Action for Israel Application No. 168279, dated Jun. 6, 2012, 1 page. |
Office Action for Indian Patent Application No. 2219/DELNP/2005, dated Jan. 3, 2007, 2 pages. |
Decision of Refusal for Japanese Patent Application No. 2005-500829, dated Feb. 14, 2012, 2 pages. |
Notification of Reasons for Refusal for Japanese Patent Application No. 2005-500829, dated Feb. 15, 2011, 3 pages. |
Notification of Reasons for Refusal for Japanese Patent Application No. 2005-500829, dated Jul. 6, 2010, 3 pages. |
Office Action for Korean Patent Application No. 10-2005-7007679, dated Dec. 26, 2011, 10 pages. |
Office Action for Korean Patent Application No. 10-2005-7007679, dated Jan. 18, 2011, 13 pages. |
Office Action for Korean Patent Application No. 10-2005-7007679, dated Dec. 26, 2012, 3 pages. |
Office Action for Mexican Patent Application No. PA/a/2005/004580, dated Aug. 25, 2009, 3 pages. |
Third Office Action for Mexican Patent Application No. PA/a/2005/004580, dated Dec. 10, 2008, 3 pages. |
Second Office Action for Mexican Patent Application No. PA/a/2005/004580, dated May 9, 2008, 2 pages. |
First Office Action for Mexican Patent Application No. PA/a/2005/004580, dated Jan. 30, 2008, 2 pages. |
Office Action for New Zealand Patent Application No. 540087, dated Jan. 4, 2008, 2 pages. |
Office Action for New Zealand Patent Application No. 540087, dated Sep. 14, 2006, 2 pages. |
Office Action for U.S. Appl. No. 10/696,389, dated Nov. 14, 2008, 6 pages. |
Office Action for U.S. Appl. No. 10/696,389, dated Mar. 28, 2008, 11 pages. |
Office Action for U.S. Appl. No. 10/696,389, dated Oct. 10, 2007, 10 pages. |
Office Action for U.S. Appl. No. 10/696,389, dated Apr. 2, 2007, 9 pages. |
International Search Report for International Application No. PCT/US2003/034240, dated Jul. 12, 2005, 1 page. |
International Preliminary Report on Patentability for International Application No. PCT/US2003/034240, dated May 6, 2013, 5 pages. |
Office Action for Japanese Patent Application No. 2011-001318, dated Feb. 12, 2013, 3 pages. |
Official Action for Korean Application No. 10-2013-7010499, dated Sep. 2, 2013, 4 pages. |
Office Action for Mexican Patent Application No. MX/a/2010/000195, dated Feb. 1, 2012, 2 pages. |
Office Action for Mexican Patent Application No. MX/a/2010/000195, dated Jul. 27, 2011, 2 pages. |
Office Action for Mexican Patent Application No. MX/a/2010/000195, dated Oct. 2, 2012, 3 pages. |
Office Action for Mexican Patent Application No. MX/a/2010/000195, dated May 31, 2013, 2 pages. |
Office Action for New Zealand Patent Application No. 564543, dated Jan. 4, 2008, 2 pages. |
Examiner's First Report for Australian Patent Application No. 2006270008, dated Dec. 10, 2010, 3 pages. |
Office Action for Canadian Patent Application No. 2,614,764, dated Nov. 14, 2012, 4 pages. |
First Office Action for Chinese Patent Application No. 200680034397.X, dated Jan. 22, 2010, 24 pages. |
Second Office Action for Chinese Patent Application No. 200680034397.X, dated Feb. 9, 2011, 21 pages. |
Third Office Action for Chinese Patent Application No. 200680034397.X, dated Mar. 12, 2012, 5 pages. |
Fourth Office Action for Chinese Patent Application No. 200680034397.X, dated Dec. 4, 2012, 3 pages. |
Office Action for Columbian Patent Application No. 08016117, dated Jun. 26, 2012, 8 pages. |
Office Action for Columbian Patent Application No. 08016117, dated Jan. 14, 2013, 11 pages. |
Office Action for Costa Rican Patent Application No. 9736, dated Apr. 22, 2013, 7 pages. |
Office Action for Egyptian Patent Application No. PCT 84/2008, dated Oct. 24, 2012, 3 pages. |
Supplementary European Search Report for European Application No. 06787716.7, dated Dec. 29, 2011, 7 pages. |
Office Action for European Application No. 06787716.7, dated Oct. 26, 2012, 3 pages. |
Generics [UK] Ltd.'s Notice of Opposition for European Application No. 06787716.7, filed Jun. 4, 2014, 17 pages. |
Patentee's Response to Notice of Opposition and Declaration of Lee Leserman for European Application No. 06787716.7, filed Jan. 16, 2015, 58 pages. |
Office Action for Israel Patent Application No. 188406, dated Jun. 13, 2011, 4 pages. |
Office Action for Israel Patent Application No. 188406, dated Apr. 26, 2010, 1 page. |
Office Action for Israel Patent Application No. 188406, dated Jan. 6, 2013, 1 page. |
Office Action for Israel Patent Application No. 188406, dated Aug. 3, 2015, 2 pages. |
Office Action for Indian Application No. 353/DELNP/2008, dated Jul. 26, 2013, 2 pages. |
Office Action for Japanese Patent Application No. 2008-522895, dated Apr. 17, 2012, 4 pages. |
Office Action for Japanese Application No. 2008-522895, dated Aug. 27, 2013, 5 pages. |
Office Action for Korean Patent Application No. 10-2008-7002031, dated Dec. 21, 2012, 7 pages. |
Office Action for Mexican Patent Application No. MX/a/2008/000425, dated Jun. 2, 2010, 2 pages. |
Office Action for New Zealand Patent Application No. 565300, dated Feb. 24, 2011, 2 pages. |
Office Action for New Zealand Patent Application No. 565300, dated Nov. 11, 2009, 2 pages. |
Office Action for New Zealand Patent Application No. 565300, dated May 31, 2011, 2 pages. |
Office Action for U.S. Appl. No. 11/185,448, dated Dec. 17, 2009, 11 pages. |
Office Action for U.S. Appl. No. 11/185,448, dated Jun. 30, 2009, 10 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2006/027859, dated Aug. 14, 2007, 8 pages. |
International Preliminary Report on Patentability for International Application No. PCT/US2006/027859, dated Jan. 22, 2008, 6 pages. |
Office Action for Canadian Patent Application No. 2,646,255, dated Feb. 4, 2013, 5 pages. |
Office Action for Chinese Application No. 201310149581.0, dated Apr. 21, 2014, 6 pages. |
Office Action for Chinese Application No. 201310149581.0, dated Oct. 17, 2014, 7 pages. |
Office Action for Chinese Application No. 201310149581.0, dated Jun. 23, 2015, 8 pages. |
Supplementary European Search Report for European Application No. 07754853, dated Jan. 16, 2013, 8 pages. |
Office Action for European Application No. 07754853.5, dated Feb. 3, 2014, 4 pages. |
Office Action for Israel Application No. 216401, dated May 27, 2014, 2 pages. |
Office Action for Israel Application No. 216401, dated Aug. 3, 2015, 3 pages. |
Office Action for Japanese Patent Application No. 2009-504281, dated Sep. 4, 2012, 2 pages. |
Office Action for Japanese Application No. 2009-504281, dated Nov. 1, 2013, 5 pages. |
Office Action for Japanese Application No. 2009-504281, dated Aug. 3, 2015 , 4 pages. |
Office Action for Korean Application No. 10-2013-7031379, dated Feb. 25, 2014, 3 pages. |
Office Action for Mexican Patent Application No. MX/a/2008/012684, dated Jul. 8, 2011, 2 pages. |
Office Action for Mexican Patent Application No. MX/a/2008/012684, dated Apr. 5, 2011, 2 pages. |
Office Action for U.S. Appl. No. 11/398,859, dated Jun. 4, 2010, 7 pages. |
Office Action for U.S. Appl. No. 11/398,859, dated Sep. 11, 2009, 12 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2007/008404, dated Sep. 26, 2008, 6 pages. |
International Preliminary Report on Patentability for International Application No. PCT/US2007/008404, dated Oct. 21, 2008, 4 pages. |
Office Action for Canadian Application No. 2,838,108, dated Jan. 6, 2015, 3 pages. |
European Search Report for European Patent Application No. 11159754.8, dated Jun. 22, 2011, 5 pages. |
Office Action for Japanese Application No. 2013-146934, dated May 26, 2014, 6 pages. |
Office Action for Korean Application No. 10-2014-7005780, dated Jun. 26, 2014, 4 pages. |
Office Action for U.S. Appl. No. 12/424,177, dated Jul. 21, 2014, 10 pages. |
Office Action for U.S. Appl. No. 12/424,177, dated Mar. 16, 2012, 15 pages. |
Office Action for U.S. Appl. No. 12/424,177, dated Aug. 31, 2011, 10 pages. |
Office Action for U.S. Appl. No. 12/424,177, dated Dec. 24, 2013, 10 pages. |
Office Action for U.S. Appl. No. 12/424,177, dated Oct. 29, 2015, 7 pages. |
Office Action for U.S. Appl. No. 12/424,177, dated May 20, 2016, 5 pages. |
Office Action for Canadian Application No. 2,853,611, dated Mar. 5, 2015, 3 pages. |
European Search Report for European Patent Application No. 13175824.5, dated Sep. 16, 2013, 8 pages. |
Office Action for Japanese Application No. 2013-167610, dated Aug. 8, 2014, 4 pages. |
Office Action for U.S. Appl. No. 12/748,756, dated Jan. 27, 2012, 9 pages. |
Office Action for U.S. Appl. No. 12/748,756, dated Aug. 23, 2012, 8 pages. |
Office Action for Japanese Application No. 2014-040222, dated Feb. 10, 2015, 1 page. |
Office Action for U.S. Appl. No. 12/983,659, dated May 20, 2016, 6 pages. |
Office Action for U.S. Appl. No. 12/983,659, dated Dec. 2, 2013, 14 pages. |
Office Action for U.S. Appl. No. 12/983,659, dated Jun. 17, 2014, 11 pages. |
Office Action for U.S. Appl. No. 12/983,659, dated Oct. 29, 2015, 9 pages. |
European Search Report for European Application No. 14183066.1, dated Dec. 16, 2014, 11 pages. |
Office Action for European Application No. 14183066.1, dated Oct. 11, 2016, 6 pages. |
Office Action for Japanese Application No. 2014-196130, dated Jul. 14, 2015, 4 pages. |
Office Action for U.S. Appl. No. 14/319,018, dated Dec. 2, 2016, 8 pages. |
Notice of Reasons for Rejection for Japanese Application No. 2014-222230, dated Aug. 1, 2016, 6 pages. |
Office Action for U.S. Appl. No. 15/093,180, dated Jun. 24, 2016, 8 pages. |
Office Action for U.S. Appl. No. 15/093,180, dated Oct. 21, 2016, 11 pages. |
Office Action for Australian Patent Application No. 2006322076, dated Sep. 23, 2011, 2 pages. |
Office Action for Canadian Patent Application No. 2,631,872, dated Dec. 7, 2012, 2 pages. |
Supplementary European Search Report for European Application No. 06847502.9, dated Dec. 5, 2012, 7 pages. |
Office Action for European Patent Application No. 06847502.9, dated Oct. 14, 2013, 6 pages. |
Office Action for European Patent Application No. 06847502.9, dated Jul. 16, 2014, 4 pages. |
Office Action for Japanese Patent Application No. 2008-544430, dated May 26, 2012, 3 pages. |
Office Action for U.S. Appl. No. 11/634,343, dated Jan. 17, 2012, 8 pages. |
Office Action for U.S. Appl. No. 11/634,343, dated Aug. 4, 2011, 17 pages. |
Office Action for U.S. Appl. No. 11/634,343, dated Apr. 5, 2011, 21 pages. |
Office Action for U.S. Appl. No. 11/634,343, dated Sep. 14, 2010, 22 pages. |
Office Action for U.S. Appl. No. 11/634,343, dated Feb. 23, 2010, 16 pages. |
Office Action for U.S. Appl. No. 11/634,343, dated Jun. 19, 2009, 11 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2006/046360, dated Oct. 17, 2007, 5 pages. |
International Preliminary Report on Patentability for International Application No. PCT/US2006/046360, dated Jun. 11, 2008, 5 pages. |
Office Action for Canadian Application No. 2,838,111, dated Nov. 27, 2014, 4 pages. |
European Search Report for European Application No. 16156100.6, dated Jul. 25, 2016, 6 pages. |
Office Action for U.S. Appl. No. 13/527,213, dated Mar. 11, 2013, 12 pages. |
Office Action for Canadian Application No. 2,896,083, dated Jun. 3, 2016, 6 pages. |
European Search Report for European Application No. 16156099.0, dated Jul. 25, 2016, 7 pages. |
Office Action for U.S. Appl. No. 13/664,181, dated Feb. 12, 2013, 12 pages. |
Office Action for U.S. Appl. No. 13/664,181, dated Aug. 22, 2013, 9 pages. |
Office Action for U.S. Appl. No. 13/666,420, dated Mar. 5, 2013, 10 pages. |
Office Action for U.S. Appl. No. 13/675,559, dated Mar. 19, 2013, 13 pages. |
Office Action for U.S. Appl. No. 13/675,559, dated Aug. 20, 2013, 9 pages. |
Office Action for U.S. Appl. No. 13/675,587, dated Apr. 4, 2013, 11 pages. |
Office Action for U.S. Appl. No. 13/675,587, dated Aug. 21, 2013, 9 pages. |
Office Action for U.S. Appl. No. 14/080,922, dated Jun. 12, 2014, 11 pages. |
Office Action for U.S. Appl. No. 14/080,922, dated Mar. 3, 2015, 16 pages. |
Office Action for U.S. Appl. No. 14/080,922, dated Aug. 10, 2015, 17 pages. |
Office Action for U.S. Appl. No. 14/080,922, dated Dec. 21, 2016, 15 pages. |
Office Action for U.S. Appl. No. 14/987,508, dated Apr. 1, 2016, 12 pages. |
Office Action for U.S. Appl. No. 15/066,346, dated Jul. 21, 2016, 9 pages. |
Office Action for U.S. Appl. No. 15/066,360, dated Jul. 20, 2016, 10 pages. |
Office Action for U.S. Appl. No. 15/205,918, dated Sep. 26, 2016, 16 pages. |
Office Action for U.S. Appl. No. 15/205,925, dated Oct. 27, 2016, 10 pages. |
Office Action for U.S. Appl. No. 15/241,439, dated Oct. 17, 2016, 13 pages. |
Supplementary European Search Report and Written Opinion for European Application No. 07754936.8, dated Jan. 18, 2013, 9 pages. |
Office Action for European Application No. 07754936.8, dated Sep. 30, 2016, 6 pages. |
Office Action for Japanese Patent Application No. 2009-504301, dated Sep. 4, 2012, 4 pages. |
Office Action for Japanese Patent Application No. 2009-504301, dated Oct. 31, 2013, 5 pages. |
Office Action for U.S. Appl. No. 11/696,343, dated Oct. 21, 2011, 16 pages. |
Office Action for U.S. Appl. No. 11/696,343, dated May 10, 2011, 11 pages. |
Office Action for U.S. Appl. No. 11/696,343, dated May 23, 2014, 18 pages. |
Office Action for U.S. Appl. No. 11/696,343, dated Feb. 3, 2015, 11 pages. |
Office Action for U.S. Appl. No. 11/696,343, dated Jun. 10, 2016, 11 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2007/008500, dated Sep. 26, 2008, 9 pages. |
International Preliminary Report on Patentability for International Application No. PCT/US2007/008500, dated Oct. 21, 2008, 8 pages. |
Office Action for Japanese Application No. 2014-165736, dated Jul. 9, 2015, 10 pages. |
First Office Action for Chinese Application No. 201380030763.4, dated Jan. 6, 2016, 7 pages. |
Supplementary European Search Report for European Application No. 13793204.2, dated Sep. 25, 2015, 5 pages. |
Office Action for European Application No. 13793204.2, dated Jul. 15, 2016, 8 pages. |
Office Action for New Zealand Application No. 700983, dated Oct. 5, 2015, 4 pages. |
Office Action for New Zealand Application No. 700983, dated Jun. 2, 2016, 2 pages. |
Office Action for U.S. Appl. No. 13/899,457, dated Apr. 7, 2016, 33 pages. |
Office Action for U.S. Appl. No. 13/899,457, dated Jul. 28, 2016, 38 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2013/042113, dated Sep. 4, 2013 , 13 pages. |
International Preliminary Report on Patentability for International Application No. PCT/US2013/042113, dated Nov. 25, 2014, 9 pages. |
Allen, T. M. et al., “Effect of liposome size and drug release properties of pharmacokinetics of encapsulated drug to rats,” The Journal of Pharmacology and Experimental Therapeutics, 226(2):539-544 (1983). |
Alton et al., “Cationic lipid-mediated CFTR gene transfer to the lungs and nose of patients with cystic fibrosis: a double-blind placebo-controlled trial,” The Lancet, 353(9157):947-954 (1999). |
Andrews, J. M., “Determination of minimum inhibitory concentrations,” Journal of Antimicrobial Chemotherapy, 48(S1):5-14 (2001). |
Antos, M. et al., “Antibacterial activity of liposomal amikacin against Pseudomonas aeruginosa in vitro,” Pharmacological Research, 32(1/2):84-87 (1995). |
Bakker-Woudenberg, I. et al., “Efficacy of gentamicin or ceftazidine entrapped in liposomes with prolonged blood circulation and enhanced localization in Klebsiella pneumoniae-infected lung tissue,” The Journal Infectious Diseases, 171:938-947 (1995). |
Bakker-Woudenberg et al. (2002). Ciprofloxacin in polyethylene glycol-coated liposomes: efficacy in rat models of acute or chronic Pseudomonas aeruginosa infection. Antimicrobial Agents and Chemotherapy 46(8), pp. 2575-2581. |
Bakker-Woudenberg et al. (2001). Improved efficacy of ciprofloxacin administered in polyethylene glycol-coated liposomes for treatment of Klebsiella pneumoniae pneumonia in rats. Antimicrobial Agents and Chemotherapy 45(5), pp. 1487-1492. |
Ball, V. et al., “Complexation mechanism of bovine serum albumin and poly(allylamine hydrochloride),” J. Phys. Chem. B., 106(9):2357-2364 (2002). |
Bangham, A. D. et al., “Diffusion of univalent ions across the lamellae of swollen phospholipids,” J. Mol. Biol., 13(1):238-252 (1965). |
Bangham, A. D., Introduction, “Liposomes: An Historical Perspective,” in: Liposomes, Ostro, M. J. (ed.), pp. 1-25, Marcel Dekker, Inc., New York (1983). |
Bargoni, A. et al., “Transmucosal transport of tobramycin incorporated in solid lipid nanoparticles (SLN) after duodenal administration to rats. Part II—Tissue distribution,” Pharmacological Research, 43(5):497-502 (2001). |
Beaulac, C. et al., “Eradication of Mucoid Pseudomonas aeruginosa with Fluid Liposome-Encapsulated Tobramycin in an Animal Model of Chronic Pulmonary Infection,” Antimicrobial Agents and Chemotherapy, 40(3):665-669 (1996). |
Beaulac, C. et al., “In-vitro bactericidal efficacy of sub-MIC concentrations of liposome-encapsulated antibiotic against Gram-negative and Gram-positive bacteria,” Journal of Antimicrobial Chemotherapy, 41:35-41 (1998). |
Beaulac, C. et al., “Aerolization of low phase transition temperature liposomal tobramycin as a dry powder in an animal model of chronic pulmonary infection caused by Pseudomonas aeruginosa,” Journal Drug Targeting, 7(1):33-41 (1999). |
Beaulac, C. et al., “In vitro kinetics of drug release and pulmonary retention of microencapsulated antibiotic in liposomal formulations in relation to the lipid composition,” Journal Microencapsulation 14(3):335-348 (1997). |
Bedard et al. (1989). Interaction of the fluoroquinolone antimicrobial agents ciprofloxacin and enoxacin with liposomes. Antimicrobial Agents and Chemotherapy 33(8), pp. 1379-1382. |
Bermudez, L. E. et al., “Treatment of disseminated Mycobacterium avium complex infection of beige mice with liposome-encapsulated aminoglycosides,” The Journal of Infectious Diseases, 161(6):1262-1268 (1990). |
Bhavane et al. (2003). Agglomerated vesicle technology: a new class of particles for controlled and modulated pulmonary drug delivery. Journal of Controlled Release 93, pp. 15-28. |
Bhavane (2006). Nanoparticle agglomerates for pulmonary drug delivery. A dissertation presented to the faculty of the University of Texas Health Science Center at Houston of Health Information Sciences. UMI No. 3237380. |
Bilodeau, M. et al., “Kanamycin aerosol therapy in 200 cases of bronchopulmonary suppurations,” Can. Med. Assoc. J., 89:537-541 (1963) (with English Abstract). |
Blaser, J. et al., “Once daily dosing of aminoglycosides,” Eur. Clin. Microbiol. Infect. Dis., 14(12):1029-1038 (1995). |
Bucke, W. E. et al., “Surface-modified amikacin-liposomes: organ distribution and interaction with plasma proteins,” Journal of Drug Targeting, 5(2):99-108 (1997). |
Bunderberg de Jong, H. G. et al., Koazevation (Entmischung in Kolloidalen Systemen), Koll, Zeitsch, 50(10):39-48 (1930). |
Cantin, A. M. et al., “Aerosolized prolastin suppresses bacterial proliferation in a model of chronic pseudomonas aeruginosa lung infection,” Am. J. Respir. Crit. Care Med., 160:1130-1135 (1999). |
Carlier, M. B. et al., “Inhibition of lysosomal phospholipases by aminoglycoside antibiotics: in vitro comparative studies,” Antimicrobial Agents and Chemotherapy, 23(3):440-449 (1983). |
Cash, H. A. et al., “A rat model of chronic respiratory infection with Pseudomonas aeruginosa,” American Review of Respiratory Disease, 119(3):453-459 (1979). |
Challoner, P. B. et al., “Gamma Scintigraphy Lung Deposition Comparison of TOBI in the PARI LC PLUS Nebulizer and the Aerodose Inhaler,” American Thoracic Society 97th International Conference, San Francisco, California, Aerogen, Inc. (2001). |
Chambless, J. D. et al., “A three-dimensional computer model of four hypothetical mechanisms protecting biofilms from antimicrobials,” Appl. Environ. Microbiol., 72(3):2005-2013 (2006). |
Chan, C. H. S. et al., “Mycobacteria as a cause of infective exacerbation in bronchiectasis,” Postgrad. Med. J., 68:896-899 (1992). |
Chapman, D., “Physicochemical Properties of Phospholipids and Lipid-Water Systems,” In: Liposome Technology, Chapter 1, vol. I, Preparation of Liposomes, Gregoriadis G. (ed.), CRC Press, Inc., Boca Raton, Florida, pp. 1-18 (1984). |
Chmiel, J. F. et al., “State of the art: why do the lungs of patients with cystic fibrosis become infected and why can't they clear the infection?”, Respiratory Research, 4:8-20 (2003). |
Ciofu, O. et al., “Occurrence of Hypermutable Pseudomonas aeruginosa in Cystic Fibrosis Patients Is Associated with the Oxidative Stress Caused by Chronic Lung Inflammation,” Antimicrobial Agents and Chemotherapy, 49(6):2276-2282 (Jun. 2005). |
Clancy, J. P. et al., “Phase II studies of nebulised Arikace in CF patients with Pseudomonas aeruginosa infection,” Thorax, 68(9):818-825 (2013). |
Clay. M. M. et al., “Assessment of jet nebulisers for lung aerosol therapy,” Lancet, 2:592-594 (1983). |
ClinicalTrials.gov, “Safety and Efficacy Study of Ciprofloxacin for Inhalation in Patients With Non-Cystic Fibrosis Bronchiectasis ‘ORBIT-1’”, Identifier: NCT00889967, First Received: Apr. 27, 2009, 3 pages. |
Colardyn, F., “The efficacy and safety of isepamicin and ceftazidime compared with amikacin and ceftazidime in acute lower respiratory tract infection,” Journal of Chemotherapy, 7(2):129-135 (1995). |
Coleman, L. T. et al., “Bronchiectasis in children,” Journal of Thoracic Imaging, 10(4)268-279 (1995). |
Comis, R. L., “Carboplatin in the treatment of non-small cell lung cancer: a review,” Oncology, 50(2):37-41 (1993). |
Conley et al., “Aerosol Delivery of Liposome-Encapsulated Ciprofloxacin: Aerosol Characterization and Efficacy against Francisella tularensis Infection in Mice,” Antimicrobial Angents, 41(6):1288-1292 (Jun. 1997). |
Cooksey, R. C. et al., “Antimicrobial susceptibility patterns of Streptococcus pneumoniae,” Antimicrobial Agents and Chemotherapy, 13(4):645-648 (1978). |
Costerton, J. W. et al., “Bacterial biofilms: A common cause of persistent infections,” Science, 284:1318-1322 (1999). |
Couvreur, P. et al., “Liposomes and nanoparticles in the treatment of intracellular bacterial infections,” Pharmaceutical Research, 8(9):1079-1085 (1991). |
Cremades, M. J. et al., “Repeated pulmonary infection by Nocardia asteroides complex in a patient with bronchiectasis,” Respiration, 65:211-213 (1998). |
Crowther, N. R. et al., “Inhaled aminoglycoside (gentamicin) in bronchiectasis: Dry powder vs. nebulization vs. intravenous therapy,” Clinical and Investigative Medicine, Annual Meeting of the Canadian Society for Clinical Investigation, The Royal College of Physicians and Surgeons of Canada and Participating Societies, Toronto, Canada, Abstract 530 (Sep. 24-27, 1998). |
Currie, D. C., “Nebulisers for bronchiectasis,” Thorax, 52(Suppl. 2):572-574 (1997). |
Cymbala, A. A. et al., “The Disease-Modifying Effects of Twice-Weekly Oral Azithromycin in Patients with Bronchiectasis,” Treat Respir. Med. 2005;4(2):117-122. |
Cynamon, M. H. et al., “Liposome-Encapsulated-Amikacin Therapy of Mycobacterium avium Complex Infection in Geige Mice,” Antimicrobial Agents and Chemotherapy, 33(8):1179-1183 (1989). |
Dally, M. B. et al., “Ventilatory effects of aerosol gentamicin,” Thorax, 33:54-56 (1978). |
Damaso, D. et al., “Susceptibility of current clinical isolates of Pseudomonas aeruginosa and enteric gram-negative bacilli to amikacin and other aminoglycoside antibiotics,” The Journal of Infectious Diseases, 134:S394-S390 (1976). |
Deamer, D. W. et al., “Liposome Preparation: Methods and Mechanisms,” Chapter 1 in: Liposomes, Ostro, M. J. (ed.), Marcel Dekker, Inc., New York (1983), 27 pages. |
Dees, C. et al., “The mechanism of enhanced intraphagocytic killing of bacteria by liposomes containing antibiotics,” Veterinary Immunology and Immunopathology, 24:135-146 (1990). |
Del Porto, P. et al., “Dysfunctional CFTR alters the bactericidal activity of human macrophages against Pseudomonas aeruginosa,” PLoS One, 6(5):e19970 (2011). |
Demaeyer, P. et al., “Disposition of liposomal gentamicin following intrabronchial administration in rabbits,” Journal Microencapsulation, 10(1):77-88 (1993). |
Deol, P. et al., “Lung specific stealth liposomes: stability, biodistribution and toxicity of liposomal antitubular drugs in mice,” Biochimica et Biophysica Acta, 1334:161-172 (1997). |
Dequin, P. F. et al., “Urinary excretion reflects lung deposition of aminoglycoside aerosols in cystic fibrosis,” Eur. Respir. J., 18(2):316-322 (2001). |
Desai et al., “A facile method of delivery of liposomes by nebulization,” Journal of Controlled Release, 84(1-2):69-78 (2002). |
Desai, T. R. et al., “Determination of surface free energy of interactive dry powder liposome formulations using capillary penetration technique,” Colloids and Surfaces B: Biointerfaces, 22:107-113 (2001). |
Desai, “Delivery of liposomes in dry powder form: aerodynamic dispersion properties,” European Journal of Pharmaceutical Sciences 20:459-467 (2003). |
Di Ninno et al. (1993). Liposome-encapsulated ciprofloxacin is effective in the protection and treatment of BALB/c mice against Francisella tularensis. The Journal of Infectious Diseases 168, pp. 793-794. |
Dickie, K. J. et al., “Ventilatory effects of aerosolized kanamycin and polymyxin,” Chest, 63(5):694-697 (1973). |
Dong, C. et al., “Acacia-gelatin microencapsulated liposomes: preparation, stability and release of acetylsalicylic acid,” Pharmaceutical Research, 10(1):141-146 (1993). |
Doring, G. et al., “Antibiotic therapy against Pseudomonas aeruginosa in cystic fibrosis: a European consensus,” Eur Respir J., 16(4):749-767 (2000). |
Drenkard, E. et al., “Pseudomonas biofilm formation and antibiotic resistance are linked to phenotypic variation,” Nature, 416:740-743 (2002). |
Eboka (2005). Aqueous solubility of ciprofloxacin in the presence of metal cations. Tropical Journal of Pharmaceutical Research, 4(1), pp. 349-354. |
Ehlers, S. et al., “Liposomal amikacin for treatment of M. avium Infections in clinically relevant experimental settings,” Zbl. Bakt., 284:218-231 (1996). |
Eigen (1994). A multicenter study of alternate-day prednisone therapy in patients with cystic fibrosis. The Journal of Pediatrics, 126(4), pp. 515-523. |
El-Din, M. A. T. et al., “Nebulizer therapy with antibiotics in chronic suppurative lung disease,” Journal of Aerosol Medicine, 7(4):345-350 (1994). |
Eller, J. M. et al., “The therapy of bronchiectasis,” Deutsche Medizinische Wochenschrift, 118(44):1608-1610 (1993). |
Farber, J. E. et al., “The use of aerosol penicillin and Streptomycin in bronchopulmonary infections,” California Medicine, 73(3):214-217 (1950). |
Fielding, R. M. et al., “Pharmacokinetics and Urinary Excretion of Amikacin in Low-Clearance Unilamellar Liposomes after a Single or Repeated Intravenous Administration in the Rhesus Monkey,” Antimicrobial Agents and Chemotherapy, 43(3):503-509 (1999). |
Finke, W., “Long-term antibiotic therapy in chronic bronchitis and infectious asthma. Control and prevention of bronchopulmonary disease.” Antibiotics and Chemotherapy, 4(3):319-329 (1954). |
Finlay, W. H. et al., “Regional lung deposition of nebulized liposome-encapsulated ciprofloxacin,” International Journal of Pharmaceutics (Amsterdam), 167(1-2):121-127 (Jun. 1, 1998). |
Fountain, M. W. et al., “Treatment of Brucella canis and Brucella abortus in vitro and in vivo by stable plurilamellar vesicle-encapsulated aminoolycosides,” The Journal of Infectious Diseases, 152(3):529-535 (1985). |
Garcia, A. T., “Efficacy of amikacin sulfate in lower respiratory infections,” Investigacion Medica Internacional, 9(3):235-240 (1982) (with English Abstract). |
Geller, D. E. et al., “Pharmacokinetics and bioavailability of aerosolized tobramycin in cystic fibrosis,” Chest, 122(1):219-226 (2002). |
Gibson, R. L. et al., “Pathophysiology and management of pulmonary infections in cystic fibrosis,” American Journal of Respiratory and Critical Care Medicine, 168(8):918-951 (2003). |
Gibson, R. L. et al., “Significant microbiological effect of inhaled tobramycin in young children with cystic fibrosis,” American Journal of Respiratory and Critical Care Medicine, 167(6):841-849 (2003). |
Gilbert, B. E. et al., “Tolerance of volunteers to cyclosporine A-dilauroylphosphatidylcholine liposome aerosol,” American Journal of Respiratory and Critical Care Medicine, 156(6):1789-1793 (1997). |
Gleiser, C. A. et al., “Pathology of experimental respiratory anthrax in Macaca mulatta,” Brit. J. Exp. Path., 44:416-426 (1963). |
Goldman, J. M. et al., “Inhaled micronised gentamicin powder: a new delivery system,” Thorax, 45:939-940 (1990). |
Gonzales-Rothi, R. J. et al., “Liposomes and pulmonary alveolar macrophages: functional and morphologic interactions,” Experimental Lung Research, 17:687-705 (1991). |
Goss, C. H. et al., “Update on cystic fibrosis epidemiology,” Current Opinion in Pulmonary Medicine, 10(6):510-514 (2004). |
Graczyk, J. et al., “Staphylococcal pneumonia—analysis of material of patients treated in lung diseases hospital in years 1981-1994,” Pneumonologia I Alergologia Polska, 65(11-12):767-774 (1997) (with English Abstract). |
Greene, K. E. et al., “Radiographic changes in acute exacerbations of cystic fibrosis in adults: A pilot study,” AJR, 163:557-562 (1994). |
Gunther, A. et al., “Surfactant alteration and replacement in acute respiratory distress syndrome,” Respiratory Research, 2(6): 353-364 (2001). |
Gursoy et al. (1997). Characterization of ciprofloxacin liposomes; derivative ultraviolet spectrophotometric determinations. J. Microencapsulation 14(6), pp. 769-776. |
Hagwood, S. et al., “Structure and properties of surfactant protein B,” Biochimica et Biophysica Acta., 1408:150-160 (1998). |
Hansen, C. R. et al., “Long-term azithromycin treatment of cystic fibrosis patients with chronic pseudomonas aeruginosa infection: an observational cohort study,” Journal of Cystic Fibrosis, 4(1):35-40 (2005). |
Helbich, T. et al., “High-resolution computed tomography of the lung in young patients with cystic fibrosis,” Radiologe, 33(3):142-146 (1993) (English Abstract). |
Hess, D. et al., “Medication nebulizer performance. Effects of diluent volume, nebulizer flow, and nebulizer brand,” Chest, 110:498-505 (1996). |
Hess, D. R., “Nebulizers: Principles and Performance,” Respiratory Care, 45(6):609-622 (2000). |
Hewitt, W. L. et al., “Antibiotic therapy of abscess of the lung and bronchiectasis,” California Medicine, 76(5):319-324 (1952). |
Hoffman, L. R. et al., “Aminoglycoside antibiotics induce bacterial biofilm formation,” Nature, 436:1171-1175 (2005). |
Honeybourne, D., “Antibiotic penetration in the respiratory tract and implications for the selection of antimicrobial therapy,” Current Opinion in Pulmonary Medicine 1997, 3(2):170-174. |
Howell, S. B., “Clinical applications of a novel sustained-release injectable drug delivery system: Depofoam Technology,” Cancer Journal, 7:219-227 (2001). |
Hrkach, J. S. et al., “Synthesis of poly(L-lactic acid-co-L-lysine) graft copolymers,” Macromolecules, 28:4736-4739 (1995). |
Hrkach, J. S. et al., “Poly(L-Lactic acid-co-amino acid) graft copolymers: A class of functional biodegradable biomaterials,” In: Hydrogels and Biodegradable Polymers for Bioapplications, Chapter 8, ACS Symposium Series No. 627, Ottenbrite, R. M. et al. (eds.), American Chemical Society, pp. 93-102 (1996). |
Huang, L. et al., “Progress of liposome's applications in biomedicine,” International Journal of Biologicals, 29(3):130-132 and 137 (2006). |
Huang et al. (2006). Pulmonary delivery of insulin by liposomal carriers. Journal of Controlled Release 113, pp. 9-14. |
Hubble, D., “Discussion on respiratory catarrh in children,” Proceedings of the Royal Society of Medicine, 52(9):701-710 (1959). |
Hung, O. R. et al., “Pharmacokinetics of inhaled liposome-encapsulated fentanyl,” Anesthesiology, 83(2): 277-284 (Aug. 1995). |
Hung, J. C. et al., “Evaluation of two commercial jet nebulisers and three compressors for the nebulisation of antibiotics,” Archives of Disease in Childhood, 71(4):335-338 (Oct. 1994). |
Hunt, B. E. et al., “Macromolecular mechanisms of sputum inhibition of tobramycin activity,” Antimicrobial Agents and Chemotherapy, 39(1):34-39 (1995). |
Ikegami, M. et al., “Surfactant protein metabolism in vivo,” Biochimica et Biophysica Acta, 1408:218-225 (1998). |
Ikemoto, H. et al., “Susceptibility of bacteria isolated from the patients with lower respiratory tract infections to antibiotics,” The Japanese Journal of Antibiotics, 42(11):2350-2353 (1989). |
IP, M. S. M. et al., “Bronchiectasis and related disorders,” Respirology, 1:107-114 (1996). |
Ishii, F. et al., “Procedure for Preparation of Lipid Vesicles (Liposomes) Using the Coacervation (Phase Separation) Technique,” Langmuir, 11(2):483-486 (1995). |
Janoff, A. S. et al., “Unusual lipid structures selectively reduce the toxicity of amphotericin B,” Proc. Nat. Acad. Sci. USA, 85:6122-6126 (1988). |
Jayaraman, S. et al., “Noninvasive in vivo fluorescence measurement of airway-surface liquid depth, salt concentration, and pH,” J. Clin. Invest. 107:317-324 (2001). |
Johansson, J., “Structure and properties of surfactant protein C,” Biochimica et Biophysica Acta, 1408:161-172 (1998). |
Johnston, M. J. W. et al., “Therapeutically optimized rates of drug release can be achieved by varying the drug-to-lipid ratio in liposomal vincristine formulations,” Biochimica et Biophysica Acta, 1758:55-64 (2006). |
Katare, O. P. et al., “Enhanced in vivo Performance of Liposomallndomethacin Derived From Effervescent Granule Based Proliposomes,” J. Microencapsulation, 12(5):487-493 (1995). |
Kesavalu, L. et al., “Differential effects of free and liposome encapsulated amikacin on the survival of Mycobacterium avium complex in mouse peritoneal macrophages,” Tubercle, 71:215-218 (1990). |
Kim, E. K. et al., “Pharmacokinetics of intravitreally injected liposomes encapsulated tobramycin in normal rabbits,” Yonsei Medical Journal, 31(4):308-314 (1990). |
Klemens, S. P. et al., “Liposome-encapsulated-gentamicin therapy of Mycobacterium avium complex infection in beige mice,” Antimicrobial Agents and Chemotherapy, 34(6):967-970 (1990). |
Knoch, M. et al., “The customised electronic nebuliser: a new category of liquid aerosol drug delivery systems,” Expert Opin. Drug Deliv., 2(2):377-390 (2005). |
Knox, K. et al., “Chronic bronchitis. An attempt to control chronic infection with Haemophilus influenzae by aerosol therapy,” The Lancet, pp. 120-122 (1955). |
Labiris, N. R. et al., “Pulmonary drug delivery. Part II: The role of inhalant delivery devices and drug formulations in Therapeutic effectiveness of aerosolized medications,” Br.J.Clin.Pharmacol., 56(6):600-612 (2003). |
Lagace, J. et al., “Liposome-encapsulated antibiotics: preparation, drug release and antimicrobial activity against Pseudomona aeruginosa,” Journal Microencapsulation, 8(1) 53-61 (1991). |
Landyshev, Y. S. et al., “Clinical and experimental aspects of liposomal hydrocortisone treatment of bronchial asthma,” Ter. Arkh., 74(8):45-48 (2002). |
Lass, J. S. et al., “New advances in aerosolised drug delivery: vibrating membrane nebuliser technology,” Expert Opin Drug Deliv., 3(5):693-702 (2006). |
Le Brun, P. P. H. et al., “A review of the technical aspects of drug nebulization,” Pharmacy World & Science, 22(3):75-81 (2000). |
Le Brun, P. P. H. et al., “Inhalation of tobramycin in cystic fibrosis part 1: The choice of a nebulizer,” International Journal of Pharmaceutics, 189:205-214 (1999). |
Le Brun, P. P. H. et al., “Inhalation of tobramycin in cystic fibrosis part 2: Optimization of the tobramycin solution for a jet and ultrasonic nebulizer,” International Journal of Pharmaceutics, 189:215-225 (1999). |
Le Brun, P. P. H. et al., “Dry powder inhalation of antibiotics in cystic fibrosis therapy: part 2. Inhalation of a novel colistin dry powder formulation: a feasibility study in healthy volunteers and patients,” European Journal of Pharmaceutics and Biopharmaceutics, 54:25-32 (2002). |
Li, Z. et al., “Nebulization of liposomal amikacin formulations: SLIT Amikacin,” Respiratory Drug Delivery, 3:801-804 (2006). |
Li, Z. et al., “Characterization of nebulized liposomal amikacin (Arikace) as a function of droplet size,” Journal of Aerosol Medicine and Pulmonary Drug Delivery, 21(3):245-253 (2008). |
Lin, H.-C. et al., “Inhaled gentamicin reduces airway neutrophil activity and mucus secretion in bronchiectasis,” Am. J. Respir. Crit. Care Med., 155:2024-2029 (1997). |
Lutwyche, P. et al., “Intracellular delivery and antibacterial activity of gentamicin encapsulated in pH-sensitive liposomes,” Antimicrobial Agents and Chemotherapy, 42(10):2511-2520 (1998). |
Magallanes, M. et al., “Liposome-incorporated ciprofloxacin in treatment of murine Salmonellosis,” Antimicrobial Agents and Chemotherapy, Nov. 1993, 37(11):2293-2297. |
Majumdar, S. et al., “Efficacies of Liposome-Encapsulated Streptomycin and Ciprofloxacin against Mycobacterium avium—M. intracellulare Complex Infections in Human Peripheral Blood Monocyte/Macrophages,” Antimicrobial Agents and Chemotherapy, 36(12):2808-2815 (Dec. 1992). |
Marcotte, G. V. et al., “Chronic productive cough and bronchiectasis in a 40-year-old woman,” Annals of Allergy, Asthma & Immunology, 78(6):559-564 (1997). |
Marier, J. F. et al., “Liposomal tobramycin against pulmonary infections of Pseudomonas aeruginosa: a pharmacokinetic and efficacy study following single and multiple intratracheal administrations in rats,” Journal Antimicrobial Chemotherapy, 52:247-252 (2003). |
Marier, J-F. et al., “Pharmacokinetics and efficacies of liposomal and conventional formulations of tobramycin after intratracheal administration in rats with pulmonary burkholderia cepacia infection,” Antimicrobial Agents and Chemotherapy, 46(12):3776-3781 (2002). |
Mariotti, A. B. et al., “Aerosol therapy with tobramycin in exacerbations of chronic obstructive lung disease (7 cases),” 66(2):198-202 (1996) (with English Abstract). |
Martini, W. Z. et al., “Lung surfactant kinetics in conscious pigs,” Am J Physiol., 277(1 Pt 1): E187-E195 (1999). |
Marwah, O. S. et al., “Bronchiectasis. How to identify, treat and prevent,” Postgrad. Med., 97(2):149-150, 153-156, 159 (1995) (Abstract). |
McAllister, S. M. et al., “Antimicrobial properties of liposomal polymyxin B,” Journal of Antimicrobial Chemotherapy, 43:203-210 (1999). |
Meers, P. et al., “Biofilm penetration, triggered release and in vivo activity of inhaled liposomal amikacin in chronic Pseudomonas aeruginosa lung infections,” Journal of Antimicrobial Chemotherapy, 61(4):859-868 (2008). |
Mendelman, P. M. et al., “Aminoglycoside penetration, inactivation, and efficacy in cystic fibrosis sputum,” American Review of Respiratory Disease, 132(4):761-765 (1985). |
Mercer, R. R. et al., “Cell Number and Distribution in Human and Rat Airways,” Am. J. Respir. Cell Mol. Biol., vol. 10, pp. 613-624, 1994. |
Mohanty, B. et al., “Systematic of alcohol-induced simple coacervation in aqueous gelatin solutions,” Biomacromolecules, 4:1080-1086 (2003). |
Mombelli, G. et al., “Anti-pseudomonas activity in bronchial secretions of patients receiving amikacin or tobramycin as a continuous infusion,” Antimicrobial Agents and Chemotherapy, 19(1):72-75 (1981). |
Montero et al. (1998). Fluoroquinolone-biomembrane interactions: monolayer and calorimetric studies. Langmuir 14(9), pp. 2451-2454. |
Morgan, J. R. et al., “Preparation and properties of liposome-associated gentamicin,” Antimicrobial Agents and Chemotherapy, 17(4):544-548 (1980). |
Myers, M. A. et al., “Pulmonary effects of chronic exposure to liposome aerosols in mice,” Experimental Lung Research, 19:1-19 (1993). |
Nakazawa, S. et al., “Studies on a new aminoglycoside antibiotic, amikacin (BB-K8) in pediatrics,” The Japanese Journal of Antibiotics, 27(4):438-445 (1974). |
Nasu, M. et al., “Appropriate use of antimicrobial agents,” Selection of Anti-infective, Clinic in Japan (Special Number) Infection Disease Study in New Era (first volume), 2003, 61st issue, pp. 718-723. |
Newton, D. W. et al., Chapter 4: “Coacervation: Principles and Applications,” In: Polymers for Controlled Drug Delivery, Tarcha, P. J. (ed.), CRC Press, Boca Raton, pp. 67-81 (1991). |
Nightingale, S. D. et al., “Liposome-encapsulated gentamicin treatment of Mycobacterium avium—Mycobacterium intracellulare complex bacteremia in AIDS patients,” Antimicrobial Agents and Chemotherapy, 37(9):1869-1872 (1993). |
Niven, R. W. et al., “Nebulization of liposomes. I. Effects of lipid composition,” Pharmaceutical Research, 7(11):1127-1133 (1990). |
Niven, R. W. et al., “Nebulization of liposomes. II. The effects of size and modeling of solute release profiles,” Pharmaceutical Research, 8(2):217-221 (1991). |
Niven, R. W. et al., “Nebulization of liposomes. III. The effects of operating conditions and local environment,” Pharmaceutical Research, 9(4):515-520 (1992). |
Oh, Y-K et al., “Formulation and Efficacy of Liposome-Encapsulated Antibiotics for Therapy of Intracellular Mycobacterium avium Infection,” Antimicrobial Agents and Chemotherapy, 39(9):2104-2111 (Sep. 1995). |
Oizumi, K. et al., “Therapeutic effect of amikacin for infections with gram-negative bacilli, especially for stubborn respiratory infections,” The Japanese Journal of Antibiotics, 31(1):15-23 (1978). |
Olsen, A. M., “Streptomycin aerosol in the treatment of chronic bronchiectasis: preliminary report,” Staff Meetings of the Mayo Clinic, pp. 53-54 (1946). |
Olsen, A. M., “Nebulization therapy in bronchiectasis: The use of penicillin and Streptomycin aerosols,” In: Collected Papers of The Mayo Clinic and The Mayo Foundation, Hewitt, R. M. et al. (eds.), 38:579-586 (1946). |
Olsen, A. M., “Nebulization therapy in bronchiectasis: The use of penicillin and streptomycin aerosols,” J.A.M.A., 134(11):947-953 (1947). |
Omri, A. et al., “Incorporation, release and in-vitro antibacterial activity of liposomal aminoglycosides against Pseudomonas aeruginosa,” Journal Antimicrobial Chemotherapy, 36(4):631-639 (1995). |
Omri, A. et al., “Comparison of the bactericidal action of amikacin, netilmicin and tobramtcin in free and liposomal formulation against pseudomonas aeruginosa,” Chemotherapy, 42:170-176 (1996). |
Omri, A. et al., “Pulmonary retention of free and liposome-encapsulated tobramycin after intratracheal administration in uninfected rats and rats infected with Pseudomonas aeruginosa,” Antimicrobial Agents and Chemotherapy, 38(5):1090-1095 (1994). |
Pai, V. B. et al., “Efficacy and safety of aerosolized tobramycin in cystic fibrosis,” Pediatric Pulmonology, 32(4):314-327 (2001). |
Papahadjopoulos, D. et al., “Phospholipid model membranes. I. Structural characteristics of hydrated liquid crystals,” Biochimica et Biophysica Acta., 135:624-638 (1967). |
Paradisi, F. et al, “Acute and chronic bronchopulmonary infections and aminoglycoside antibiotics,” Chemioterapia Antimicrobica, 1(2):224-227 (1978). |
Parsek, M. R. et al., “Acyl-homoserine lactone quorum sensing gram-negative bacteria: a signaling mechanism involved in associations with higher organisms,” Proc. Nat. Acad. Sci., 97(16):6789-6793 (2000). |
Patton, J. S. et al., “The lungs as a portal of entry for systemic drug delivery,” Proc. Am. Thor. Soc., 1:338-344 (2004). |
Perkins, W. R. et al., “Aerosolization of liposomal amikacin (Arikace) using different nebulizers: Selection of the eflow nebulizer,” Poster and Oral Presentation at North American Cystic Fibrosis Conference (Oct. 2007), Pediatric Pulmonology, 42(30):356-357, abs. 434, 12 pages. |
Perkins, W. R. et al., “Role of lipid polymorphism in pulmonary surfactant,” Science, 273:330-332 (Jul. 1996). |
Petersen, E. A. et al., “Liposomal amikacin: improved treatment of Mycibacterium avium complex infection in the beige mouse model,” Journal Antimicrobial Chemotherapy, 38:819-828 (1996). |
Petkowicz, J. et al., “Hypoglycemic Effect of Liposome-Entrapped Insulin Administered by Various Routes into Normal Rats,” Pol. J. Pharmacol. Pharm., 41:299-304 (1989). |
Pilewski, J. M. et al., “Role of CFTR in airway disease,” Physiological Reviews, 79M:S215-S255 (1999). |
Pines, A. et al., “Treatment of severe pseudomonas infections of the bronchi,” British Medical Journal, 1:663-665 (1970). |
Pines, A. et al., “Gentamicin and colistin in chronic purulent bronchial infections,” British Medical Journal, 2:543-545 (1967). |
Potter, B. P., “Aerosol antibiotic therapy in suppurative diseases of the lung and bronchi,” Aerosol Antibiotic Therapy, 25:436-448 (1949). |
Poyner, E. A. et al., “A comparative study on the pulmonary delivery of tobramycin encapsulated into liposomes and PLA microspheres following intravenous and endotracheal delivery,” Journal of Controlled Release, 35(1):41-48 (1995). |
Poyner, E. A. et al., “Preparation, properties and the effects of free and liposomal tobramycin on siderophore production by Pseudomonas aeruginosa,” Journal of Antimicrobial Chemotherapy, 34:43-52 (1993). |
Press Release, “Transave Announces Positive Phase II Results for Once-Daily Arikace in the Treatment of Cystic Fibrosis Patients Who Have Pseudomonas Lung Infections,” Presented at the European Cystic Fibrosis Society Conference, Monmouth Junction, NJ, Jun. 13, 2008, 3 pages. |
Price, C. I. et al., “Liposome delivery of aminoglycosides in burn wounds,” Surgery, Gynecology & Obstetrics, 174:414-418 (1992). |
Price, C. I. et al., “Liposome encapsulation: a method for enhancing the effectiveness of local antibiotics,” Surgery, 115(4):480-487 (1994). |
Price, C. I. et al., “Enhanced effectiveness of intraperitoneal antibiotics administered via liposomal carrier,” Arch Surgery, 124:1411-1415 (1989). |
Price, K. E. et al., “Amikacin, an aminoglycoside with marked activity against antibiotic-resistant clinical isolates,” The Journal of Infectious Diseases, 134:S249-S261 (1976). |
Ramsammy, L. S. et al., “The effect of gentamicin on the biophysical properties of phosphatidic acid liposomes is influenced by the O—C=0 group of the lipid,” Biochemistry, 27:8249-8254 (1988). |
Ramsey, B. W. et al., “Intermittent administration of inhaled tobramycin in patients with cystic fibrosis. Cystic Fibrosis Inhaled Tobramycin Study Group,” The New England Journal of Medicine, 340(1):23-30 (1999). |
Ramsey, B. W. et al., “Efficacy of aerosolized tobramycin in patients with cystic fibrosis,” The New England Journal of Medicine, 328:1740-1746 (1993). |
Rastogi et al. (2006). Particulate and vesicular drug carriers in the management of tuberculosis. Current Drug Delivery 3(1), pp. 121-128. |
Rau, J. L. et al., “Performance Comparison of Nebulizer Designs: Constant-Output, Breath-Enhanced, and Dosimetric,” Respir. Care 2004;49(2):174-179. |
Roehrborn, A. A. et al., “Lipid-based slow-release formulation of amikacin sulfate reduces foreign body-associated infections in mice,” Antimicrobial Agents and Chemotherapy, 39(8):1752-1755 (1995). |
Sabra, W. et al., “Physiological responses of pseudomonas aeruginosa PAO1 to oxidative stress in controlled microaerobic and aerobic cultures,” Microbiology, 148:3195-3202 (2002). |
Schaad, U. B. et al., “Efficacy of inhaled amikacin as adjunct to intravenous combination therapy (ceftazidime and amikacin) in cystic fibrosis,” Journal of Pediatrics, 111(4):599-605 (Oct. 1987). |
Schentag, J. J., Antimicrobial action and pharmacokinetics/pharmacodynamics: the use of AUIC to improve efficacy and avoid resistance, Journal of Chemotherapy, 11(6):426-439 (1999). |
Schiffelers, R. et al., “Liposome-encapsulated aminoglycosides in pre-clinical and clinical studies,” Journal of Antimicrobial Chemotherapy, 48:333-344 (2001). |
Schiffelers, R. M. et al., “Therapeutic efficacy of liposomal gentamicin in clinically relevant rat models,” International Journal of Pharmaceutics, 214:103-105 (2001). |
Schiffelers, R. M. et al., “In vivo synergistic interaction of liposomecoencapsulated gentamicin and ceftazidime,” Journal Pharmacology Experimental Therapeutics, 298(1):369-375 (2001). |
Schlegel, L. et al., “In-vitro killing activity of combinations of beta-lactam agents with aminoglycosides against penicillin-resistant pneumococci,” The Journal of Antimicrobial Chemotherapy, 39(1):95-98 (1997). |
Schreier, H. et al., “Pulmonary delivery of amikacin liposomes and acute liposome toxicity in the sheep,” International Journal of Pharmaceutics, 87(1-3):183-193 (1992). |
Schreier, H. et al., “Pulmonary delivery of liposomes,” Journal of Controlled Release, 24(1):209-223 (1993). |
Sermet-Gaudelus, I. et al., “Nebulized antibiotics in cystic fibrosis,” Pediatric Drugs, 4(7):455-467 (2002). |
Shah, S. P. et al., “Liposomal amikacin dry powder inhaler: effect of fines on in vitro performance,” AAPS PharmSciTech, 5(4):e65:1-7 (2004). |
Shima, K. et al., “A study of amikacin (BB-K8) on the clinical effects on the respiratory infection,” Chemotherapy, 23(6):2128-2130 (1975) (with English Abstract). |
Singh, P. K. et al., “Quorum-sensing signals indicate that cystic fibrosis lungs are infected with bacterial biofilms,” Nature, 407:762-764 (2000). |
Skubitz, K. M. et al., “Inhalational interleukin-2 liposomes for pulmonary metastases: a phase I clinical trial,” Anti-Cancer Drugs, 11(7): 555-563 (2000). |
Smith, A. L. et al., “Safety of aerosol tobramycin administration for 3 months to patients with cystic fibrosis,” Pediatric Pulmonology, 7(4):265-271 (1989). |
Smith et al. (1986). Pharmacokinetics and sputum penetration of ciprofloxacin in patients with cystic fibrosis. Antimicrobial Agents and Chemotherapy 30(4), pp. 614-616. |
Stott, P. W. et al., “Characterization of complex coacervates of some tricyclic antidepressants and evaluation of their potential for enhancing transdermal flux,” Journal of Controlled Release, 41(3):215-227 (1996). |
Sweeney et al. (2005). Spray-freeze-dried liposomal ciprofloxacin powder for inhaled aerosol drug delivery. International Journal of Pharmaceutics 305, pp. 180-185. |
Swenson, K. A. et al., “Pharmacokinetics and in vivo activity of liposome-encapsulated gentamicin,” Antimicrobial Agents and Chemotherapy, 34(2)235-240 (1990). |
Swenson, C. E. et al., “Liposomal aminoglycosides and TLC G-65,” Aids Patient Care, pp. 290-296 (1991). |
Szoka, F. Jr. et al., “Comparative properties and methods of preparation of lipid vesicles (liposomes),” Ann. Rev. Biophys. Bioeng., 9:467-508 (1980). |
Tarran, R., “Regulation of Airway Surface Liquid Volume and Mucus Transport by Active Ion Transport,” Proc. Am. Thorac. Soc., vol. 1, pp. 42-46, 2004. |
Takamoto, M. et al., “Imipenem/cilastatin sodium alone or combined with amikacin sulfate in respiratory infections,” The Japanese Journal of Antibiotics, 47(9):1131-1144 (1994) (with English Abstract). |
Tateda, K. et al., “Efficacy of beta-lactam antibiotics combined with gentamicin against penicillin-resistant pneumococcal pneumonia in CBA/J mice,” The Journal of Antimicrobial Chemotherapy, 43(3):367-371 (1999). |
Taylor, K. M. G. et al., “The influence of liposomal encapsulation on sodium cromoglycate pharmacokinetics in man,” Pharmaceutical Research, 6(7):633-636 (1989). |
Ten, R. M. et al., “Interleukin-2 liposomes for primary immune deficiency using the aerosol route,” International Immunopharmacology, 2(2-3):333-344 (2002). |
Terzano, C. et al., “Tobramycin aerosol: could the delivery system influence the particle size and deposition in the lower airways?” Recenti. Prog. Med., 89(5):245-249 (1998) (English Abstract). |
Thomas, D. A. et al., “Acute effects of liposome aerosol inhalation on pulmonary function in healthy human volunteers,” Chest, 99(5):1268-1270 (1991). |
Thomasin, C. et al., “Drug microencapsulation by PLA/PLGA coacervation in the light of thermodynamics. 2. Parameters determining microsphere formation,” Journal of Pharmaceutical Sciences, 87(3):269-275 (1998). |
Trafny, E. A. et al., “Effects of free and liposome-encapsulated antibiotics on adherence of Pseudomonas aeruginosa to collagen type I,” Antimicrobial Agents and Chemotherapy, 39(12):2645-2649 (1995). |
Ulrich, A. S., “Biophysical aspects of using liposomes as delivery vehicles,” Bioscience Reports, 22(2):129-150 (Apr. 2002). |
Van Der Straeten, M. et al., “Amikacin in the treatment of gram-negative bronchopulmonary infections,” The Journal of Infectious Diseases, 134:S391-S393 (1976). |
Vecellio, L., “The mesh nebuliser: a recent technical innovation for aerosol delivery,” Breathe, 2(3):253-260 (2006). |
Veldhuizen, R. et al., “The role of lipids in pulmonary surfactant,” Biochimica et Biophysica Acta, 1408:90-108 (1998). |
Vidgren, M. et al., “A study of 99m technetium-labelled beclomethasone dipropionate dilauroylphosphatidylcholine liposome aerosol in normal volunteers,” International Journal of Pharmaceutics, 115:209-216 (1995). |
Vitas, A. I. et al., “Effect of composition and method of preparation of liposomes on their stability and interaction with murine monocytes infected with Brucella abortus,” Antimicrobial Agents and Chemotherapy, 40(1):146-151 (1996). |
Wang, W. et al., “Research progress in pulmonary administration of liposome,” Journal of Shenyang Pharmaceutical University, 17(3):226-229 (2000). |
Webb, M. S. et al., “Antibacterial Efficacy against an in Vivo Salmonella typhimurium Infection Model and Pharmacokinetics of a Liposomal Ciprofloxacin Formulation,” Antimicrobial Agents and Chemotherapy, 42(1):45-52 (Jan. 1998). |
Weber et al. (1997). Effect of nebulizer type and antibiotic concentration on device performance. Pediatric Pulmonology 23, pp. 249-260. |
Wise et al. (1983). In vitro activity of Bay 09867, a new quinolone derivate compared with those of other antimicrobial agents. Antimicrobial Agents and Chemotherapy 23(4), pp. 559-564. |
Westerman, E. M. et al., “Effect of nebulized colistin sulphate and colistin sulphomethate on lung function in patients with cystic fibrosis: a pilot study,” Journal of Cystic Fibrosis, 3(1):23-28 (2004). |
Whitehead, T. C. et al., “Kinetics and Toxicity of Liposomal and Conventional Amikacin in a Patient with Multidrug-Resistant Tuberculosis,” Eur J Clin Microbiol. Infect. Dis., 17:794-797 (1998). |
Wolff, R. K. et al., “Toxicologic testing of inhaled pharmaceutical aerosols,” Critical Reviews in Toxicology, 23(4):343-369 (1993). |
Worlitzsch, D. et al., “Effects of reduced mucus oxygen concentration in airway pseudomonas infections of cystic fibrosis patients,” J. Clin. Invest., 109:317-325 (2002). |
Xiu, L. et al., “Drug Resistant Analysis of Pseudomonas Aeruginosa in Patients with Mechanical Ventilation,” Med. J. Chin. PLA, 27(6):544-545 (2002) (with English Abstract). |
Yanagihara, K. et al., “Design of anti-bacterial drug and anti-Mycobacterial drug for drug delivery system,” Current Pharmaceutical Design, 8:475-482 (2002). |
Yim, D. et al., “The Development of Inhaled Liposome-Encapsulated Ciprofloxacin to Treat Cystic Fibrosis,” Respiratory Drug Delivery, pp. 425-428 (2006). |
Zeng, S. et al., “Intravitreal Pharmacokinetics of Liposome-encapsulated Amikacin in a Rabbit Model,” Ophthamology, 100:1640-1644 (1993). |
Zhang, J. H. et al., “A Novel Method to Prepare Liposomes Containing Amikacin,” Journal Microencapsulation, 16(4):511-516 (1999). |
Zhang, X. et al., “Antibacterial drug treatment of community acquired pneumonia,” Chinese Journal of Respiratory and Critical Care Medicine, 4(4):258-260 (2005). |
Zlatanov, Z. et al., “Gentamycin-pharmachim. Aerosol inhalation treatment of patients with chronic bronchitis,” Medico Biologic Information 2, pp. 5-8 (1976). |
Bakker-Woudenberg, I. A. J. M. et al., “Long-Circulating Sterically Stabilized Liposomes in the Treatment of Infections,” Method in Enzymology, Available online Feb. 21, 2005, 391:228-260 (2005). |
Office Action for European Application No. 09821103.0, dated Jan. 27, 2017, 10 pages. |
Office Action for Philippine Application No. 12011500726, dated May 29, 2015, 2 pages. |
Office Action for Israel Application No. 168279, dated Aug. 25, 2014, 2 pages. |
Office Action for Israel Application No. 188406, dated Aug. 25, 2014, 2 pages. |
Office Action for Canadian Application No. 2,853,611, dated Jul. 9, 2014, 3 pages. |
Office Action for U.S. Appl. No. 14/080,922, dated Sep. 18, 2014, 10 pages. |
Office Action for European Application No. 07754936.8, dated Sep. 2, 2014, 7 pages. |
Office Action for U.S. Appl. No. 11/696,343, dated Jan. 19, 2017, 13 pages. |
Office Action for Japanese Application No. 2014-093375, dated Apr. 7, 2015, 2 pages. |
Office Action for European Application No. 13793204.2, dated Jan. 18, 2017, 3 pages. |
Office Action for U.S. Appl. No. 13/899,457, dated Jun. 19, 2015, 24 pages. |
Office Action for Israel Application No. 236190, dated Dec. 22, 2016, 3 pages. |
Office Action for Israel Application No. 212268, dated Dec. 26, 2016, 3 pages. |
Second Office Action for Chinese Application No. 201380030763.4, dated Sep. 27, 2016, 8 pages. |
British Thoracic Society Nebuliser Project Group, Thorax, 1997, vol. 52 (Suppl. 2), S1-S24. |
U.S. Appl. No. 60/748,468, filed Dec. 8, 2005, Nicholson et al. |
Supplementary European Search Report for European Application No. 08840993.3, dated Aug. 22, 2013. |
International Preliminary Report on Patentability for International Application No. PCT/US2008/080954, dated Apr. 27, 2010, 6 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2008/080954, dated Jul. 17, 2009. |
Supplementary European Search Report for European Application No. 13858844.7, dated Jun. 15, 2016, 4 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2013/072136, dated Feb. 12, 2014, 15 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2015/031079, dated Aug. 5, 2015, 9 pages. |
Extended European Search Report for European Application No. 15791964.8, dated Dec. 11, 2017, 10 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2016/041776, dated Sep. 16, 2016, 9 pages. |
International Search Report and Written Opinion for International Application No. PCT/US2016/062894, dated Jan. 31, 2017, 10 pages. |
Biller, J. A. et al., “Efficacy of Liposomal Amikacin for Inhalation (LAI) in Achieving Nontuberculous Mycobacteria (NTM) Culture Negativity in Patients Whose Lung Infection Is Refractory to Guideline-Based Therapy,” Poster presented at the ATS 2015 International Conference, May 15-20, 2015, Denver, CO, USA, 1 page. |
Biller, J. A. et al., “Efficacy of Liposomal Amikacin for Inhalation (LAI) in Achieving Nontuberculous Mycobacteria (NTM) Culture Negativity in Patients Whose Lung Infection Is Refractory to Guideline-Based Therapy,” Abstract, D108 Diagnosis and Management of Nontuberculous Mycobacteria Infections, Poster Discussion Session, May 20, 2015, Colorado Convention Center, Am J Respir Crit Care Med 191;2015:A6295, Online Abstracts Issue, 1 page. |
Bolotin, E. M. et al., “Ammonium Sulfate Gradients for Efficient and Stable Remote Loading of Amphipathic Weak Bases into Liposomes and Ligandoliposomes,” Journal of Liposome Research, vol. 4(1), 1994, pp. 455-479. |
Chuchalin et al., “A formulation of aerosolized tobramycin (Bramitob) in the treatment of patients with cystic fibrosis and Pseudomonas aeruginosa infection: a double-blind, placebo-controlled, multicenter study,” Paediatric Drugs, 9(Suppl. 1), pp. 21-31, 2007. |
Desai et al., “A Novel Approach to the Pulmonary Delivery of Liposomes in Dry Powder Form to Eliminate the Deleterious Effects of Milling,” Journal of Pharmaceutical Sciences, 91(2):482-491 (Feb. 2002). |
Dupont et al., “A randomized placebo-controlled study of nebulized liposomal amikacin (Arikace) in the treatment of cystic fibrosis patients with chronic Pseudomonas aeruginosa lung infection,” Journal of Cystic Fibrosis, 1(7):S26, Abstract 102, Jan. 2008. |
Duzgunes, N. et al., “Treatment of intracellular Mycobacterium avium complex infection by free and liposome-encapsulated sparfloxacin,” Antimicrobial Agents and Chemotherapy, 40(11):2618-2621 (Nov. 1996). |
Gubernator, J., “Active methods of drug loading into liposomes: recent strategies for stable drug entrapment and increased in vivo activity,” Expert Opinion in Drug Delivery, vol. 8(5), 2011, pp. 565-580. |
Maurer, N. et al., “Anomalous solubility behavior of the antibiotic ciprofloxacin encapsulated in liposomes: a 1H-NMR study,” Biochimica et Biophysica Acta, 1374:9-20 (1998). |
Nikolaizik et al., “A pilot study to compare tobramycin 80 mg injectable preparation with 300 mg solution for inhalation in cystic fibrosis patients,” Canadian Respiratory Journal, 15(5)259-262, Jul./Aug. 2008. |
Szoka, F. Jr. et al., “Procedure for preparation of liposomes with large internal aqueous space and high capture by reverse-phase evaporation,” PNAS USA, 75(9):4194-4198 (Sep. 1978). |
Winthrop, K. L. et al., “Subgroup Analyses of Baseline Demographics and Efficacy in Patients With Refractory Nontuberculous Mycobacteria (NTM) Lung Infection Treated With Liposomal Amikacin for Inhalation (LAI),” Poster presented at the ATS 2015 International Conference, May 15-20, 2015, Denver, CO, USA, 1 page. |
Winthrop, K. L. et al., “Subgroup Analyses of Baseline Demographics and Efficacy in Patients With Refractory Nontuberculous Mycobacteria (NTM) Lung Infection Treated With Liposomal Amikacin for Inhalation (LAI),” Abstract, Diagnosis and Management of Nontuberculous Mycobacteria Infections, Poster Discussion Session, May 20, 2015, Colorado Convention Center, Am J Respir Crit Care Med 191;2015:A6294, Online Abstracts Issue, 2 pages. |
Xie, C., Respiratory Diseases, Scientific and Technological Documentation Press, Jun. 2000, pp. 79-81, Chapter II Section XI Pseudomonas aerugiosa Pneumonia. |
Abranches, J. et al., “Invasion of human coronary artery endothelial cells by Streptococcus mutans OMZ175,” Oral Microbiol Immunol. Apr. 2009; 24(2):141-145. doi:10.1111/j.1399-302X.2008.00487.x. |
Ahmad, S. et al., “Azithromycin effectiveness against intracellular infections of Francisella,” BMC Microbiology 2010, 10:123. |
Bahar, A. A. et al., “Antimicrobial peptides,” Pharmaceuticals 2013, 6:1543-1575; doi:10.3390/ph6121543. |
Chi, F. et al., “Vimentin-mediated signalling is required for IbeA+E. coli K1 invasion of human brain microvascular endothelial cells,” Biochem. J. (2010) 427, 79-90 (Printed in Great Britain) doi:10.1042/BJ20091097. |
Cordeiro, C. et al., “Antibacterial Efficacy of Gentamicin Encapsulated in pH-Sensitive Liposomes against an in Vivo Salmonella enterica Serovar Typhimurium Intracellular Infection Model,” Antimicrobial Agents and Chemotherapy, Mar. 2000, vol. 44, No. 3, p. 533-539. |
Deshpande, R. G. et al., “Invasion of Aortic and Heart Endothelial Cells by Porphyromonas gingivalis,” Infection and Immunity, Nov. 1998, vol. 66, No. 11, p. 5337-5343. |
Domingue, G. J. et al., “Bacterial Persistence and Expression of Disease,” Clinical Microbiology Reviews, Apr. 1997, vol. 10, No. 2, p. 320-344. |
Dorn, B. R. et al., “Invasion of Human Coronary Artery Cells by Periodontal Pathogens,” Infection and Immunity, Nov. 1999, vol. 67, No. 11, p. 5792-5798. |
Samoshina, N. M. et al., “Fliposomes: pH-Sensitive Liposomes Containing a trans-2-morpholinocyclohexanol-Based Lipid That Performs a Conformational Flip and Triggers an Instant Cargo Release in Acidic Medium,” Pharmaceutics 2011, 3, 379-405; doi:10.3390/pharmaceutics3030379. |
Helguera-Repetto, A. C. et al., (May 2014) “Differential Macrophage Response to Slow- and Fast-Growing Pathogenic Mycobacteria,” Hindawi Publishing Corporation, BioMed Research International, vol. 2014, Article ID 916521, 10 pages, http://dx.doi.org/10.1155/2014/916521. |
Jo, E-K., “Innate immunity to mycobacteria: vitamin D and autophagy,” Cellular Microbiology (2010) 12(8):1026-1035, doi:10.1111/j.1462-5822.2010.01491.x, First published online Jun. 15, 2010. |
Kozarov, E., “Bacterial invasion of vascular cell types: vascular infectology and atherogenesis,” Future Cardiol. Jan. 2012; 8(1):123-138. doi:10.2217/fca.11.75. |
Leite, E. A. et al., “Encapsulation of cisplatin in long-circulating and pH-sensitive liposomes improves its antitumor effect and reduces acute toxicity,” International Journal of Nanomedicine 2012:7 5259-5269. |
Lutwyche, P. et al., “Intracellular Delivery and Antibacterial Activity of Gentamicin Encapsulated in pH-Sensitive Liposomes,” Antimicrobial Agents and Chemotherapy, Oct. 1998, vol. 42, No. 10, p. 2511-2520. |
Martin, D. W. et al., “Invasion and Intracellular Survival of Burkholderia cepacia,” Infection and Immunity, Jan. 2000, vol. 68, No. 1, p. 24-29. |
Nahire, R. et al., “pH-Triggered Echogenicity and Contents Release from Liposomes,” Mol. Pharmaceutics 2014, 11, 4059-4068. |
Nakano, K. et al., “Detection of Cariogenic Streptococcus mutans in Extirpated Heart Valve and Atheromatous Plaque Specimens,” Journal of Clinical Microbiology, Sep. 2006, vol. 44, No. 9, p. 3313-3317. |
Nightingale, S. D. et al., “Liposome-Encapsulated Gentamicin Treatment of Mycobacterium avium—Mycobacterium intracellulare Complex Bacteremia in AIDS patients,” Antimicrobial Agents and Chemotherapy, Sep. 1993, vol. 37, No. 9, p. 1869-1872. |
Niu, J. et al., “Role of MCP-I in cardiovascular disease: molecular mechanisms and clinical implications,” Clinical Science (2009) 117:95-109 (Printed in Great Britain) doi:10.1042/CS20080581. |
Oswald-Richter, K. A. et al., “Multiple Mycobacterial antigens are targets of the adaptive immune response in pulmonary sarcoidosis,” Respiratory Research 2010, 11:161. |
Pierce, E. S., “Where Are All the Mycobacterium avium Subspecies paratuberculosis in Patients with Crohn's Disease?,” Mar. 2009, PLoS Pathogens 5(3):e1000234. doi:10.1371/journal.ppat.1000234. |
Pujol, C. et al., “Yersinia pestis Can Reside in Autophagosomes and Avoid Xenophagy in Murine Macrophages by Preventing Vacuole Acidification,” Infection and Immunity, Jun. 2009, vol. 77, No. 6, p. 2251-2261. |
Pollock, S. et al., “Uptake and trafficking of liposomes to the endoplasmic reticulum,” FASEB J. 24, 1866-1878 (2010). |
Rahman, S. A. et al., “Comparative Analyses of Nonpathogenic, Opportunistic, and Totally Pathogenic Mycobacteria Reveal Genomic and Biochemical Variabilities and Highlight the Survival Attributes of Mycobacterium tuberculosis,” mBio, Nov./Dec. 2014, 5(6):e02020-14. doi:10.1128/mBio.02020. |
Rose, S. J. et al., “Delivery of Aerosolized Liposomal Amikacin as a Novel Approach for the Treatment of Nontuberculous Mycobacteria in an Experimental Model of Pulmonary Infection,” Sep. 2014, PLoS One 9(9): e108703. doi:10.1371/journal.pone.0108703. |
Savage, P. B. et al., “Antibacterial properties of cationic steroid antibiotics,” FEMS Microbiology Letters 217 (2002) 1-7. |
Simoes, S. et al., “On the formulation of pH-sensitive liposomes with long circulation times,” Advanced Drug Delivery Reviews 56 (2004) 947-965. |
Sudimack, J. J. et al., “A novel pH-sensitive liposome formulation containing oleyl alcohol,” Biochimica et Biophysica Acta 1564 (2002) 31-37. |
Gerasimov, O. V. et al., “Cytosolic drug delivery using pH- and light-sensitive liposomes,” Advanced Drug Delivery Reviews 38 (1999) 317-338. |
Zeituni, A. E. et al., “Porphyromonas gingivalis-dendritic cell interactions: consequences for coronary artery disease,” Journal of Oral Microbiology 2010, 2: 5782. doi: 10.3402/jom.v2i0.5782. |
Anderson, K. E. et al., “Formulation and Evaluation of a Folic Acid Receptor—Targeted Oral Vancomycin Liposomal Dosage Form,” Pharmaceutical Research, 18(3):316-322 (2001). |
Vancomycin (Systemic), VA Classification Primary: AM900, Drugs.com [online], Retrieved from the Internet on Apr. 7, 2011: <URL: http://www.drugs.com/mmx/vancomycin-hydrochloride.html?printable=1>, dated Jun. 15, 1999, 15 pages. |
Harris, C. M. et al., “The stabilization of vancomycin by peptidoglycan analogs,” J Antibiot (Tokyo). Jan. 1985;38(1):51-57. |
Jones, M. N., “Use of Liposomes to Deliver Bactericides to Bacterial Biofilms,” Methods of Enzymology, 391:211-228 (2005). |
Kadry, A. A. et al., “Treatment of experimental osteomyelitis by liposomal antibiotics,” Journal of Antimicrobial Chemotheraphy, 54(6):1103-1108 (2004). |
Levy, D. E. et al., “PEGylated iminodiacetic acid zinc complex stabilizes cationic RNA-bearing nanoparticles,” Bioorganic & Medicinal Chemistry Letters, 20:5499-5501 (Jul. 2010). |
Maiz, L. et al., “Aerosolized vancomycin for the treatment of methicillin-resistant Staphylococcus aureus infection in cystic fibrosis,” Pediatric Pulmonology, 26(4):287-289 (1998). |
Onyeji, C. O. et al., “Enhanced killing of methicillin-resistant Staphylococcus aureus in human macrophages by liposome-entrapped vancomycin and teicoplanin,” Infection, 22(5):338-342 (1994). |
Sanderson, N. M. et al., “Encapsulation of vancomycin and gentamicin within cationic liposomes for inhibition of growth of Staphylococcus epidermidis,” Journal of Drug Targeting, 4(3):181-189 (1996). |
Shek, P. N. et al., “Liposomes in pulmonary applications: Physicochemical considerations, pulmonary distribution and antioxidant delivery,” J. Drug Target, 2:431-442 (1994). |
Takeuchi, Y. et al., “Stabilizing effects of some amino acids on membranes of rabbit erythrocytes perturbed by chlorpromazine,” J Pharm Sci. Jan. 1989;78(1):3-7. |
Weiner, A. L., “Liposomes as carriers for polypeptides,” Advanced Drug Delivery Review, 3(3):307-341 (May-Jun. 1989). |
Wichert, B. V. et al., “Amikacin liposomes: characterization, aerosolization, and in vitro activity against Mycobacterium avium—intracellulare in alveolar macrophages,” International Journal of Pharmaceutics, 78(1-3):227-235 (1992). |
United States Patent and Trademark Office, Before the Patent Trial and Appeal Board, Aradigm Corporation v. Insmed Incorporated, Case PGR2017-00021; U.S. Pat. No. 9,402,845, Petition for Post Grant Review, filed May 1, 2017, 111 pages. |
United States Patent and Trademark Office, Before the Patent Trial and Appeal Board, Aradigm Corporation v. Insmed Incorporated, Case PGR2017-00021; U.S. Pat. No. 9,402,845, Declaration of A. Bruce Montgomery, M.D. dated May 1, 2017, Aradigm Exhibit 1020, 146 pages. |
United States Patent and Trademark Office, Before the Patent Trial and Appeal Board, Aradigm Corporation v. Insmed Incorporated, Case PGR2017-00021; U.S. Pat. No. 9,402,845, Patent Owner's Preliminary Response, filed Aug. 16, 2017, 84 pages. |
United States Patent and Trademark Office, Before the Patent Trial and Appeal Board, Aradigm Corporation v. Insmed Incorporated, Case PGR2017-00021; U.S. Pat. No. 9,402,845, Declaration of Robert J. Lee, Ph.D. In Support of Patent Owner lnsmed's Preliminary Response, dated Aug. 16, 2017, 92 pages. |
Amikacin—DrugBank Accession No. DB00479 (APRD00550) [online], <https://www.drugbank.ca/drugs/DB00479>. Retrieved on Apr. 14, 2017, 10 pages. |
The Asthma Center Education and Research Fund, Nebulizer Instructions [online], <http://www.theasthmacenter.org/index.php/disease information/asthma/using_special_devices/nebulizer_instructions/>. Retrieved on Apr. 14, 2017, 1 page. |
Ciprofloxacin—DrugBank, Accession No. DB00537 (APRD00424, EXPT00999) [online], <https://www.drugbank.ca/drugs/DB00537>. Retrieved on Apr. 14, 2017, 19 pages. |
Prosecution history for U.S. Pat. No. 9,402,845, dated Aug. 2, 2016 (excerpted), 430 pages. |
Anacona et al., “Synthesis and antibacterial activity of metal complexes of ciprofloxacin,” Transition Metal Chemistry (2001) 26:228-231. |
Betageri et al., Liposome Drug Delivery Systems, (Technomic Publishing Co. ed., 1993) (excerpted). |
Bruinenberg, P. et al., “Inhaled Liposomal Ciprofloxacin: Once a Day Management of Respiratory Infections,” Respiratory Drug Delivery, 1:73-82 (2010). |
Bruinenberg, P., “Safety, tolerability and pharmacokinetics of novel liposomal ciprofloxacin of novel liposomal ciprofloxacin formulations for inhalation in healthy volunteers and in non-cystic bronchiectasis patients,” Am. J. Respir. Crit. Care Med. (2010) 181:A3192. |
Cabanes et al., “Sustained release of liposome-encapsulated enrofloxacin after intramuscular administration in rabbits,” American Journal of Veterinary Research, 56(11):1498-501 (1995). |
Carter, G., “Characterization of biofilm formation by Mycobacterium avium strains,” J. Med. Microbial. (2003) 52:747-52. |
Chono, S, et al., “Influence of particle size on drug delivery to rat alveolar macrophages following pulmonary administration of ciprofloxacin incorporated into liposomes,” Journal of Drug Targeting, 14(8):557-566 (2006). |
Cipro® Products FDA Approval Letter (Mar. 2004), 4 pages. |
Cipro® I.V. Label (Jan. 2005), 26 pages. |
Cipolla, D., “Development and Characterization of an in Vitro Release Assay for Liposomal Ciproftoxacin for Inhalation,” J. Pharm. Sci., 103(1):314-327 (2014). |
Cipolla, D., “Liposomal Formulations for Inhalation,” Ther. Deliv., 4(8):1047-1072 (2013). |
Cipolla, D. et al., “Development of Liposomal Ciprofloxacin to Treat Lung Infections,” Pharmaceutics 2016, vol. 8, No. 1, doi:10.3390/pharmaceutics 8010006. |
Cipolla et al., “Assessment of aerosol delivery systems for recombinant human deoxyribonuclease,” S.T.P. Pharma Sciences, 4(1), pp. 50-62 (1994). |
Cullis et al., “Liposomes as Pharmaceuticals,” Liposomes From Biophysics to Therapeutics, pp. 39-72 (M. Ostro ed., 1987). |
Cullis et al., “Generating and loading of liposomal systems for drug delivery applications,” Advanced Drug Delivery Reviews, 3, pp. 267-282 (1989). |
Driscoll et al., “Intratracheal Instillation as an Exposure Technique for the Evaluation of Respiratory Tract Toxicity: Uses and Limitations,” Toxicological Sciences, 55, pp. 24-35 (2000). |
Elhissi et al., “Formulations generated from ethanol-based proliposomes for delivery via medical nebulizers,” Journal of Pharmacy and Pharmacology, 58:887-894 (Jul. 2006). |
Fenske et al., “Encapsulation of weakly-basic drugs, antisense oligonucleotides, and plasmid DNA within large unilamellar vesicles for drug delivery applications,” Liposomes Second Edition A Practical Approach, pp. 167-191 (V. Torchilin et al. eds., 2003). |
Furneri et al., “Ofloxacin-Loaded Liposomes: In Vitro Activity and Drug Accumulation in Bacteria,” Antimicrobial Agents Chemotherapy, 44(9):2458-2464 (2000). |
Gay et al., “In Vitro Activities of Norfloxacin and Ciprofloxacin Against Mycobacterium tuberculosis, M. avium Complex, M. chelonei, M. fortuitum, and M. kansaii,” Antimicrobial Agents and Chemotherapy, vol. 26, No. 1, pp. 94-96 (Jul. 1984). |
Hyde et al., “Anatomy, pathology, and physiology of the treacheobronchial tree: Emphasis on the distal airways,” J. Allergy Clin. Immunol., vol. 124, No. 6, pp. S72-S77 (2009). |
Kensil et al., “Alkaline Hydrolysis of Phospholipids in Model Membranes and the Dependence on Their State of Aggregation,” Biochemistry, 20:6079-6085 (1981). |
Kyriacos et al., “In Vitro Testing of Ciprofloxacin Formulations and Preliminary Study on BCS Biowaiver,” Journal of Food and Drug Analysis, (2009) 17(2): 78-84. |
Lasic et al., “Transmembrane gradient driven phase transitions within vesicles: lessons for drug delivery,” Biochemica et Biophysica Acta, 1239:145-156 (1995). |
Lasic, D. D., “Gelation of liposome interior: A novel method for drug encapsulation,” FEBS Letters, 312(2.3):255-258 (Nov. 1992). |
Lipuma, J. J., “Microbiological and immunologic considerations with aerosolized drug delivery,” Chest. Sep. 2001;120(3 Suppl):118S-123S. |
Lowry et al., “Effects of pH and osmolarity on aerosol-induced cough in normal volunteers,” Clinical Science, 74:373-376 (1988). |
Moss, R. B., “Administration of aerosolized antibiotics in cystic fibrosis patients,” Chest, 120(3 Suppl):107S-113S (Sep. 2001). |
National Jewish Health, “Third sputum smear test negative for XDR TB patient Andrew Speaker,” [Online], Retrieved from the Internet: <URL: https://www.nationaljewish.org/about/news/press-releases/2007/smear-test-3>, Jun. 5, 2007, 2 pages. |
Fresenius Kabi USA, New Drug Application (NDA): 019887, NebuPent® on Drugs@FDA [online], <https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=BasicSearch.process>, Retrieved on Apr. 24, 2017. |
U.S. Department of Health and Human Services, “Nonclinical Safety Evaluation of Reformulated Drug Products and Products Intended for Administration by an Alternate Route, Guidance for Industry and Review Staff, Good Review Practice,” Oct. 2015, 12 pages. |
Piersimoni et al., “Pulmonary infections associated with non-tuberculous Mycobacteria in immunocompetent patients,” Lancet Infect Dis, 8: 323-334 (2008). |
Novartis Pharmaceuticals Corporation, TOBI, Tobramycin Inhalation Solution, USP, Nebulizer Solution, Prescribing Information, Oct. 2015, 14 pages. |
Gilead Sciences, Inc., CAYSTON (aztreonam for inhalation solution) Highlights of Prescribing Information (2014), 19 pages. |
Ross et al., “Aqueous solubilities of some variously substituted quinolone antimicrobials,” International Journal of Pharmaceutics, 63(3): 237-250 (1990). |
Saiman et al., “Antibiotic Susceptibility of Multiply Resistant Pseudomonas aeruginosa Isolated from Patients with Cystic Fibrosis, Including Candidates for Transplantation,” Clinical Infectious Diseases, 23:532-537 (Sep. 1996). |
Sangwan et al., “Aerosolized Protein Delivery in Asthma: Gamma Camera Analysis of Regional Deposition and Perfusion,” Journal of Aerosol Medicine, vol. 14, No. 2, pp. 185-195 (2001). |
Sezer et al., “Encapsulation of Enrofloxacin in Liposomes I: Preparation and in Vitro Characterization of LUV,” Journal of Liposome Research, 14(1-2):77-86 (2004). |
Shek et al., “Liposomes in Pulmonary Applications: Physiochemical Considerations, Pulmonary Distribution and Antioxidant Delivery,” Journal of Drug Targeting, 2:431-442 (1994). |
Stark, B., “Long-term stability of sterically stabilized liposomes by freezing and freeze-drying: Effects of cryoprotectants on structure,” Eur. J. Pharm. Sci. 41:546-555 (2010). |
Strauss, G., “Stabilization of lipid bilayer by sucrose during freezing,” PNAS (1986) 83:2422-2426. |
Sunamoto et al., “Unexpected Tissue Distribution of Liposomes Coated With Amylopectin Derivatives and Successful Use in the Treatment of Experimental Legionnaires' Diseases,” Receptor-Mediated Targeting of Drugs, vol. 82, pp. 359-371 (G. Gregoriadis et al. eds., 1984). |
Sunamoto et al., “Improved drug delivery directed to specific tissue using polysaccharide-coated liposomes,” Multiphase Biomedical Materials, pp. 167-190 (T. Tsuruta et al. eds., 1989). |
Van Heeckeren, A et al., “Effects of bronchopulmonary inflammation induced by Pseudomonas aeruginosa on adenovirus-mediated gene transfer to airway epithelial cells in mice,” Gene Ther., 5(3):345-351 (Mar. 1998). |
Van Heeckeren, A. et al., “Delivery of CFTR by adenoviral vector to cystic fibrosis mouse lung in a model of chronic Pseudomonas aeruginosa lung infection,” Am J Physiol Lung Cell Mol Physiol. Apr. 2004;286(4):L717-26. Epub Sep. 26, 2003. |
Van Heeckeren, A. et al., “Effect of Pseudomonas infection on weight loss, lung mechanics, and cytokines in mice,” Am J Respir Crit Care Med. Jan. 2000;161(1):271. |
Van Heeckeren, A. et al., “Murine models of chronic Pseudomonas aeruginosa lung infection,” Lab Anim., 36(3):291-312 (Jul. 2002). |
Van Heeckeren, A. et al., “Role of CFTR genotype in the response to chronic Pseudomonas aeruginosa lung infection in mice,” Am J Physiol Lung Cell Mol Physiol. Nov. 2004;287(5):L944-52. Epub Jul. 9, 2004. |
Wang, Z. et al., “Improved drug delivery: Spray freeze dried nano-liposomal inhaled aerosols,” Proceedings of the 2004 International Conference on MEMS, NANO and Smart Systems (ICMENS 2004), Badawy W. et al. (eds.), (University of Calgary), 1 page. |
Weber, A. et al., “Nebulizer delivery of tobramycin to the lower respiratory tract,” Pediatr Pulmonol., 17(5):331-339 (May 1994). |
Wolkers, W. F. et al., “Preservation of dried liposomes in the presence of sugar and phosphate,” Biochimica et Biophysica Acta, 1661:125-134 (2004). |
Wong et al., “Liposome delivery of ciprofloxacin against intracellular Francisella tularensis infection,” Journal of Controlled Release, 92(3):265-273 (2003). |
Yamazaki, Y. et al., “The ability to form biofilm influences Mycobacterium avium invasion and translocation of bronchial epithelial cells,” Cellular Microbiology, 8(5):806-814 (2006). |
Yu et al., “The Effect of Temperature and pH on the Solubility of Quinolone Compounds: Estimation of Heat of Fusion,” Pharmaceutical Research, vol. 11, No. 4, pp. 522-527 (1994). |
Zhanel et al., “A Critical Review of the Fluoroquinolones Focus on Respiratory Tract Infections,” Drugs, 62(1):13-59 (2002). |
Zhigaltsev, I. V. et al., “Formation of drug-arylsulfonate complexes inside liposomes: A novel approach to improve drug retention,” Journal of Controlled Release, 110:378-386 (2006). Available online Nov. 28, 2005. |
Number | Date | Country | |
---|---|---|---|
20170360818 A1 | Dec 2017 | US |
Number | Date | Country | |
---|---|---|---|
61489940 | May 2011 | US | |
60916342 | May 2007 | US |
Number | Date | Country | |
---|---|---|---|
Parent | 14809128 | Jul 2015 | US |
Child | 15638548 | US | |
Parent | 13480246 | May 2012 | US |
Child | 14809128 | US |
Number | Date | Country | |
---|---|---|---|
Parent | 12250412 | Oct 2008 | US |
Child | 13480246 | US | |
Parent | PCT/US2008/062868 | May 2008 | US |
Child | 12250412 | US |