专利摘要:
According to the present invention there is provided a composition and method useful for the in vivo delivery of a substantially water-insoluble pharmacologically active agent (such as paclitaxel, an anticancer agent), wherein the pharmacologically active agent is suspended in a protein coated (functioning as a stabilizer) protein. It is delivered in the form of particles. In particular, proteins and pharmacologically active agents in biocompatible dispersion media are subjected to high shear forces without any conventional surfactants, and also without any polymeric core material to the particles. In this way, particles having a diameter of less than about 1 μm are obtained. Careful selection of suitable organic phases and phase fractions, using particular compositions and preparation conditions (eg, addition of polar solvents into the organic phases), enables the reproduction of conventionally small nanoparticles with diameters of less than 200 nm that can be sterile-filtered. Will be. The particle system produced according to the invention can be converted into a redispersible dry powder comprising nanoparticles of a water insoluble drug coated with a protein as a free protein, the molecule to which the pharmacologically active agent is bound. This results in a unique delivery system in which the pharmacologically active moiety (in the form of a molecule bound to the protein) is readily available in vivo and the drug moiety is present inside the particle without any polymer matrix.
公开号:KR20010014254A
申请号:KR1019997012361
申请日:1998-06-26
公开日:2001-02-26
发明作者:닐 피. 디사이;패트릭 순-쉬옹;쉴로모 매그다시;데이비드 씨. 사하데반
申请人:패트릭 순 쉬옹;비보륵스 파마슈티칼스, 인크.;
IPC主号:
专利说明:

New Drug Formulations, Methods of Making the Same and Methods of Use thereof {Novel Formulations of Pharmacological Agents, Methods for the Preparation Thereof and Methods for the Use Thereof}
Intravenous drug delivery allows rapid and direct equilibrium of the bloodstream and carries the drug to the rest of the body. To prevent peak serum levels achieved within a short time after endovascular injection, administration of the drug contained in a stable carrier allows gradual release of the drug in the vascular compartment after intravenous intravenous injection of therapeutically active nanoparticles.
Injectable release controlled nanoparticles provide a preprogrammed duration of action from days to weeks to months with a single injection. In addition, they may provide some significant advantages for drugs that are commonly administered, including drugs that are involuntarily certain to adhere to the dosing regimen, and for those that target specific tissues or organs [Tice and Gilley, Journal of Controlled Release 2: 343-352 (1985).
Particulates and foreign substances present in the blood are generally removed from the circulatory system by the "blood filtration organ", ie the spleen, lung and liver. Particulate matter contained in normal whole blood includes erythrocytes (generally 8 μm in diameter), leukocytes (generally 6 to 8 μm in diameter) and platelets (generally 1 to 3 μm in diameter). By virtue of circulation within most organs and tissues, the blood cells move freely. If thrombus (blood clots) larger than 10-15 μm in size are present in the circulatory system, infarction or blockage of capillaries may occur, resulting in ischemia or oxygen deficiency and tissue necrosis potential. Therefore, the injection of particles larger than 10-15 μm in diameter into the circulation system should be avoided. However, suspensions of particles smaller than 7-8 μm are relatively safe and have been used for the delivery of pharmacologically active agents in the form of liposomes and emulsions, nutritional substances and contrast agents for imaging applications.
The size of the particles and their mode of delivery determine the biological behavior of the particles. Strand et al., Microspheres-Biomedical Application, Ed. A. Rembaum, pp 193-227, CRC Press (1988), states that the fate of a particle depends on its size. Particles ranging in size from several nm to 100 nm are introduced into lymphatic capillaries after interstitial injection, and phagocytosis may occur in the lymph nodes. After intravenous / intravenous injection, particles of less than about 2 μm can be rapidly cleared from the blood stream by the reticuloendothelial system (RES), also known as mononuclear phagocyte (MPS). Particles larger than about 7 μm will be trapped in pulmonary capillaries after intravenous injection. After intraarterial injection, the particles are captured in the first capillary layer that arrives. Inhaled particles are captured by macrophages of alveoli.
Water-insoluble or poorly water-soluble pharmaceuticals that are sensitive to the gastric acid environment cannot be administered by conventional methods (eg, intravenous injection or oral administration). Parenteral administration of the pharmaceutical is accomplished by emulsifying the fat soluble drug using an aqueous liquid (eg, conventional saline) in the presence of a surfactant or emulsion stabilizer to produce a stable microemulsion. Such emulsions may be injected intravenously if the emulsion component is pharmacologically inactive. US Pat. No. 4,073,943 describes water-insoluble drugs which are dissolved in oil and emulsified with water in the presence of surfactants such as egg phosphatides, pluronics (copolymers of polypropylene glycol and polyethylene glycol), polyglycerol oleate, and the like. Administration of pharmaceutical actives is described. WO 85/00011 describes pharmaceutical microdroplets of anesthetics coated with phospholipids such as dimyristoyl phosphatidylcholine having dimensions suitable for intradermal or intravenous injection.
An example of a water insoluble drug is paclitaxel, a natural product first isolated from the Pacific yew Taxus brevifolia by Wani et al. [J. Am. Chem. Soc. 93: 2325 (1971). Among antimitotic agents, paclitaxel containing diterpene carbon backbones exhibits a unique mode of action for microtubule proteins that function to form mitotic spindles. Unlike other antimitotic agents, such as vinblastine or colchicine, which inhibit the association of tubulin, paclitaxel is the only plant product known to inhibit the process of cell replication by inhibiting the depolymerization process of tubulin.
Paclitaxel, a natural diterpenoid, has been shown to have significant anti-neoplastic and anticancer effects in drug-resistant ovarian cancer. Paclitaxel has shown excellent antitumor activity in various tumor models such as B16 melanoma, L1210 leukemia, MX-1 breast tumors and CS-1 colon tumor xenografts. Some recent literature called par as a novel anticancer specific drug. In fact, paclitaxel has recently been approved by the FDA as a substance for the treatment of ovarian cancer. However, poor water solubility of paclitaxel has problems for human administration. Indeed, delivery of drugs that are inherently insoluble or poorly soluble in aqueous media are not effective. Thus, currently used paclitaxel formulations (ie Taxol®) require cremaphor to solubilize the drug. The human clinical dosage range is 200 to 500 mg. This dose is dissolved in a 1: 1 solution of ethanol: cremaphore and diluted with about 300 to 1000 ml of saline fluid administered intravenously. The cremaphor currently used is polyethoxylated perm oil. The presence of cremaphore in the formulations is characterized by animals [Lorenz et al., Agents Actions 7, 63-67 (1987)] and humans [Weiss et al. Oncol. 8, 1263-68 (1990), which is associated with severe hypersensitivity reactions, and as a result, it is necessary to prepare patients with corticosteroids (dexamethasone) and antihistamine. Many dilutions require large amounts of infusion (typical dosages of 175 mg / m 2 or less than 1 liter) and infusion times of 3 to 24 hours. Thus, there is a need for a lower toxicity, replaceable paclitaxel formulation.
In phase I clinical trials, paclitaxel itself did not show excessive toxic effects, but a severe allergic reaction was caused by the emulsifier used to solubilize the drug. Current dosing regimens need to treat patients with antihistamines and steroids prior to drug injection to reduce the allergic side effects of cremaphore.
To improve the water solubility of paclitaxel, some researchers altered their chemical structure using functional groups that increase water solubility. Among these, sulfonated derivatives (Kingston et al., US Pat. No. 5,059,699 (1991)) and amino acid esters [Mathew et al., J. Med. Chem. 35: 145-151 (1992). Modifications to produce water soluble derivatives facilitate intravenous delivery of paclitaxel dissolved in nontoxic carriers such as conventional saline. However, such modifications can increase the cost of drug preparation, lead to the induction of unwanted side reactions and / or allergic reactions, and / or a decrease in drug efficacy.
Protein microspheres have been reported in the literature as carriers of pharmacological agents or diagnostic agents. Microspheres of albumin were prepared by thermal denaturation or chemical crosslinking. Thermally modified microspheres are prepared from emulsion mixtures (eg albumin, incorporating substances and suitable oils) at 100 to 150 ° C. The microspheres are then washed and stored with a suitable solvent. Leucuta et al., International Journal of Pharmaceutics 41: 213-217 (1988) describe a method for producing thermally modified microspheres.
Methods of preparing chemically crosslinked microspheres involve treating the emulsion with glutaraldehyde to crosslink the protein, wash and store it. Lee et al., Science 213: 233-235 (1981) and US Pat. No. 4,671,954 teach this preparation method.
The method for preparing protein microspheres as carriers of the pharmacologically active agents is suitable for the delivery of water-soluble substances but cannot capture water-insoluble substances. This limitation is an inevitable problem of the above production techniques that depends on the crosslinking or thermal denaturation of protein components in the aqueous phase of water-in-oil emulsions. Any water soluble material dissolved in the protein containing aqueous phase may be trapped in the resulting crosslinked or heat denatured protein matrix, but poorly water soluble or fat soluble material cannot be incorporated into the protein matrix formed by the above technique.
One common method of making drug-containing nanoparticles is to dissolve polylactic acid (or other biocompatible water insoluble polymer) in a water immiscible solvent (eg methylene chloride or other chloride, aliphatic or aromatic solvent) and in a polymer solution. Dissolving the pharmacologically active agent, adding the surfactant in the oily or aqueous phase, forming the oil-in-water emulsion by suitable means, and slowly evaporating the emulsion under vacuum. If the oil droplets are small enough and stable during evaporation, a polymer suspension in water is obtained. Since the drug is present in the polymer solution from the beginning, the method can obtain a composition in which the drug molecule is trapped in the particles constituting the polymer matrix. The formation of microspheres and nanoparticles using solvent evaporation has been reported by several researchers using various drugs (see, eg, Tice and Gilley, Journal of Controlled Release 2: 343-352 (1985); Bodmeier and McGinity). , Int. J. Pharmaceutics 43: 179 (1988); Cavalier et al., J. Pharm. Pharmacol. 38: 249 (1985); D'Souza et al., WO 94/10980).
Bazile et al., Biomaterials, 13: 1093 (1992) and Spenlehauer et al., French Patent No. 2 660 556 disclose that one polymer (e.g. polylactide) is dissolved in the organic phase together with the active ingredient such as a drug. In addition, other polymers such as albumin have been reported to form nanoparticles using two biocompatible polymers used as surface active agents. After emulsification and removal of the solvent, nanoparticles are formed and the drug is present in the polymer matrix of the polylactide particles.
The nature of the polymer solution in which the polymer matrix is formed is of great importance in obtaining a suitable emulsion in the first step. For example, polylactide (a polymer commonly used in the preparation of injectable nanoparticles) has a surface activity that results in its rapid adsorption at the dichloromethane-water interface, thereby lowering the interfacial tension to improve the emulsification process (eg See Boury et al., Langmuir 11: 1636 (1995). In addition, the same researchers found that bovine serum albumin (BSA) can interact with polylactide and pass through polylactide monolayers present at the oil-water interface. Thus, on the basis of this document, emulsification during conventional solvent evaporation methods is greatly improved by the presence of surface active polymers (polylactides) in the non-aqueous organic phase. Indeed, the presence of polylactide is not only sufficient condition, but also actually necessary for the formation of nanoparticles of suitable size.
Another method based on solvent evaporation dissolves the drug in a hydrophobic solvent (eg toluene or cyclohexane) without dissolving any polymer in the organic solvent, adding a conventional surfactant to the mixture as an emulsifier and sonicating Forming an oil-in-water emulsion using a treatment or high shear device, followed by evaporation of the solvent to obtain anhydrous drug particles (see, eg, Sjostrom et al., J. Dispersion Science and Technology 15: 89- 117 (1994)] Upon removal of the nonpolar solvent, precipitation of the drug in the solvent droplet occurs and particles of less than μm are obtained.
It has been found that the size of the particles is mainly controlled by the initial size of the emulsion droplets. It is also interesting to note that the final particle size is reported to decrease with decreasing drug concentration in the organic phase. This finding is in contrast to the results reported herein, which do not use any conventional surfactants in the manufacture of nanoparticles (in some embodiments of the invention). In addition, the authors of the Sjostrom literature should know that the drug used, cholesteryl acetate, has surface activity in toluene and has a higher drug concentration at the interface towards the oil-water interface, increasing the likelihood of precipitation.
In addition, the formation of particles smaller than μm is described by Calvo et al., J. Pharm. Sci. 85: 530 (1996). This method dissolves the drug (e.g. indomethacin) and polymer (polycaprolactone) in methylene chloride and acetone, and then the solution is poured into an aqueous phase containing a surfactant (Poloxamer 188) to produce particles of sub-μm size ( 216 nm). However, this method is carried out at solvent concentrations where no emulsion is formed.
Paclitaxel is a very promising natural compound as an anticancer drug. See, for example, McGuire et al., "Taxol: A Unique Anti-Neoplastic Agent With Significant Activity Against Advanced Ovarian Epithelial Neoplasms" Ann. Int. Med., 111: 273-279 (1989)]. All patents, scientific and other references mentioned herein are incorporated herein by reference in their entirety.
Unfortunately, paclitaxel has a very low solubility in water, making it difficult to provide a suitable dosage form. Indeed, in phase I clinical trials, severe allergic reactions occurred by emulsifiers administered with paclitaxel to compensate for low water solubility of paclitaxel, and one or more patients died due to allergic reactions caused by the emulsifier. Toxicity limiting doses include neutropenia, peripheral neuropathy and hypersensitivity.
Brown et al., "A Phase I Trial of Taxol Given by A 6-Hour Intravenous Infusion" J of Clin Oncol, 9 (7): 1261-1267 (July 1991), showed that paclitaxel was not administered without preparation. Intravenous infusion was reported for 6 hours every day. Thirty-one patients were administered Taxol in 64 evaluable routes. One patient developed severe (or acute) hypersensitivity and needed immediate treatment to stop the infusion and save the patient's life. Other patients were hypersensitivity, but not severe enough to discontinue infusion. Spinal cord suppression killed 2 patients with sepsis, limiting the dose. Non-blood toxicity was grade 1 and 2 except that one patient had grade 3 mucositis and two patients had grade 3 neuropathy. Neuropathy consists of painful paresthesia that can be ameliorated and it was necessary to discontinue Taxol infusion in two patients. Partial responses were observed in four patients (three non-small cell lung cancer patients and one adenocarcinoma patient). The maximum allowable dose was reported at 275 mg / m 2 and the recommended starting phase II dose was 225 mg / m 2 . The incidence of hypersensitivity is dependent on the dosing schedule, with incidences of hypersensitivity at drug infusion for 6 to 24 hours reported to be from 0 to 8%. In addition, hypersensitivity reaction continued with or without preparation in spite of prolonged infusion time. Since these Phase I studies were conducted on various terminal cancer patients, the efficacy of Taxol treatment could not be determined.
In a study by Kris et al., Paclitaxel formulated with Cremaphor EL in dehydrated alcohol was administered as an intravenous infusion for 3 hours every 21 days at a dosage of 15 to 230 mg / m 2 in increments over 9 steps. It became. Chris et al. Concluded that, due to the depth and unpredictability of the hypersensitivity, the dosing regimen of the drug formulation does not mean the use of taxol in higher doses (Cancer Treat. Rep., Vol. 70 , No. 5, May 1986).
Because initial trials using mass infusions or short (1 to 3 hours) infusions cause anaphylaxis or other hypersensitivity reactions, steroids (eg dexamethasone), antihistamines (eg diphenhydramine) and H 2 -antagonists (eg Taxol was administered only after preparation with cimetidine or ranitidine), and another study was performed by extending the infusion time to 24 hours to eliminate the most severe allergic reaction. Different Phase I and II studies have been published with 24 hour infusions of Taxol using a maximum total dose of 250 mg / m 2 , typically every three weeks. Patients were pretreated with dexamethasone, diphenhydramine and cimetidine to eliminate allergic reactions [Einzig, et al., "Phase II Trial of Taxol in Patients with Metastatic Renal Cell Carcinoma," Cancer Investigation, 9 (2) 133 -136 (1991), and AB Miller et al., "Reporting Results of Cancer Treatment," Cancer, Vol 47, 207-214 (1981).
Koeller et al., “A Phase I Pharmacokinetic Study of Taxol Given By a Prolonged Infusiion Without Premedication,” Proceedings of ASCO, Vol. 8 (March, 1989) recommended routine preparations to prevent many of the allergic reactions that are thought to be caused by the Cremophore (polyethoxylated perme oil) vehicle used for paclitaxel injection. Patients were dosed from 175 mg / m 2 to 275 mg / m 2 .
In addition, Wiernik et al., “Phase I Clinical and Pharmacokinetic Study of Taxol,” Cancer Research, 47: 2486-93 (May 1, 1987), reported that crease by intravenous infusion over 6 hours in phase I studies. The administration of paclitaxel in the mofo vehicle was reported. Hypersensitivity reactions in grades 3-4 occurred in four of the 13 pathways. The starting dose in this study was 15 mg / m 2 (1/3 of the dog's lowest toxic dose). The dose was increased stepwise to treat at least three patients at each dose level until toxicity was confirmed, followed by 4 to 6 patients each at subsequent levels. The study concluded that neurotoxicity and leukopenia limited the dose and the recommended phase II dose was 250 mg / m 2 with predose.
Examples of other studies on paclitaxel are described in Legha et al., “Phase II Trial of Taxol in Metastatic Melanoma,” 65: 2478-81 (June 1990); Rowinsky et al., "Phase I and Pharmacodynamic Study of Taxol in Refractory Acute Leukemias," Cancer Research, 49, 4640-47 (Aug. 15, 1989); Grem et al., "Phase I Study of Taxol Administered as a Short IV Infusion Daily For 5 Days," Cancer Treatment Reports, Vol. 71, No. 12, (December, 1987); Donehower et al., "Phase I Trial of Taxol in Patients With Advanced Cancer," Cancer Treatment Reports, Vol. 71, No. 12, (December, 1987); Holmes et al., "Phase II Study of Taxol in Patients (PT) with Metastatic Breast Cancer (MBC)," Proceedings of the American Society of Clinical Oncology, 10:60 (March, 1991) and Suffness. "Development of Antitumor Natural Products at the National Cancer Institute," Gann Monograph or Cancer Research, 31: 21-44 (1989) (Taxol is recommended for only 24 hour infusion administration).
Weiss et al., "Hypersensitivity Reactions from Taxol," Journal of Clinical Oncology, 8 (7) 1263-1268 (July 1990), are highly variable in paclitaxel dosage and dosing regimens used, and include changes in the infusion plan and It is difficult to determine a reliable overall incidence of hypersensitivity reactions because the extent of the effect of the use of the preparations on the incidence of hypersensitivity reactions is unknown. For example, of five patients who received Taxol by injecting a dose of more than 190 mg / m 2 for three hours without pre-treatment, three of them were hypersensitivity, with a much higher dose of 6 hours without preparation. Only one of the 30 patients who received infusion received hypersensitivity. Thus, this suggests that extended infusion over 6 hours is sufficient to reduce the incidence of hypersensitivity reactions. Nevertheless, Base et al. Found that patients who received Taxol 250 mg / m 2 over 24 hours continued to have a clear hypersensitivity reaction. Thus, prolonging drug infusion to 6 to 24 hours may reduce the risk of acute reactions, but this conclusion suggests that 78% of hypersensitivity reactions occur within 10 minutes of initiation of Taxol infusion, which means that the length of the overall infusion planning time is not significant. Not sure because it indicates no relationship. In addition, the infusion concentration of paclitaxel may not be significant because many patients respond to different small amounts of Taxol dosages. After all, not only is the mechanism of Taxol hypersensitivity known, but it is unclear whether paclitaxel itself causes hypersensitivity or whether the hypersensitivity is due to excipients (Cremaphor EL; BASF, Ludwigshafen, Germany). While it is unclear how predose affects the depth and frequency of hypersensitivity reactions, prophylactic therapy is recommended because the risks of its use are not known.
Cremaphor EL emulsion as an approved cancer treatment protocol due to the conflicting recommendations of the prior art on whether pre-medication should be used to prevent hypersensitivity when using extended infusion periods and the lack of efficacy data for infusions performed over 6 hours A 24-hour infusion of high doses of paclitaxel in excess (170 mg / m 2 ) was used.
Although the use of prolonged infusion periods seems to minimize the side effects of paclitaxel administration in emulsions, long infusion periods are inconvenient for patients and are expensive due to the need to monitor the patient for a total infusion period of 6 to 24 hours. . In addition, long infusion periods require patients to be hospitalized for more than one day in a hospital or treatment clinic.
In addition, higher doses of paclitaxel are described in the literature. Stem et al., “High-dose paclitaxel, cyclophosphamide to determine the maximum tolerated dose (MTD) of paclitaxel in combination with high doses of cyclophosphamide and cisplatin followed by autologous hematopoietic progenitor support (AHPCS). , and cisplatin with autologous hematopoietic progenitor-cell support: A phase I trial, "J. Clin. Oncol. 14: 1463-1472 (1996) was injected over 24 hours in incremental doses of paclitaxel followed by injection of cyclophosphamide (5,625 mg / m 2 ) and cisplatin (165 mg / m 2 ) and AHPCS. Phase I trials were performed on 49 patients with poor prognosis of breast cancer, Non-Hodgkin's lymphoma (NHL) or ovarian cancer. Dose limiting toxicity was seen in two patients at paclitaxel 825 mg / m 2 , one of whom died due to multi-organ dysfunction and the other developed grade 3 respiratory, CNS and nephrotoxicity. Grade 3 multiple neuropathy and grade 4 CNS toxicity were also observed. The maximum tolerated dose (MTD) of this combination was determined to be paclitaxel (775 mg / m 2 ), cyclophosphamide (5,625 mg / m 2 ) and cisplatin (165 mg / m 2 ) followed by AHPCS. Sensory multiple neuropathy and mucositis showed significant toxicity, but both could improve and tolerate. 18 of 33 breast cancer patients (54%) had a partial response. In addition, responses were observed in patients with NHL (4 of 5 patients) and ovarian cancer (2 of 2 patients).
US Pat. No. 5,641,803 reports the use of taxol at doses 175 and 135 mg / m 2 administered as a three hour infusion. The infusion protocol requires the use of preparatory medications and the occurrence of hypersensitivity reactions has been reported in 35% of patients. Neurotoxicity was reported in 51% of patients, 66% in the high dose group and 37% in the low dose group. In addition, 48% of patients experienced neurotoxicity for a longer infusion time of 24 hours and 54% of patients infused for a shorter time of 3 hours experienced neurotoxicity.
It has been demonstrated in the literature that the higher the paclitaxel dose, the higher the response rate. Optimal dosages and dosing regimens for paclitaxel are under study. To assess the possibility that paclitaxel dose intensity is important for the induction of disease response, Reed et al. Analyzed NII phase data in the treatment of ovarian and breast cancers [Reed E, Bitton R, Sarosy G, Kohn E: Paclitaxel dose intensity, “Journal of Infusional Chemotherapy, 6: 59-63 (1996)]. The results indicate that the relationship between objective disease response and paclitaxel dose intensity in recurrent ovarian cancer is a two-sided p-value of 0.022. suggests that large statistically significant relationship in breast cancer is much more strong as a two-side p-value of 0.004. 135 mg / m 2 / objective response rates at 21 days, 13.2% and, 250 mg / m 2/21 days The objective response rate was 35.9% at, and the response rate observed at intermediate doses of 175 mg / m 2 was linear to the results of 135 mg / m 2 and 250 mg / m 2 , and a linear regression analysis of 0.946 for this data. Correlation coefficients were shown (Reed et al. 1996).
Holmes's study [Holmes et al, "Phase II trial of Taxol, an active drug in the treatment of metastatic breast cancer," J. Natkl. Cancer Inst., 83: 1797-1805, (1991)] and MSKCC [Reichman BS et al, "Paclitaxel and recombinant human granulocyte colony-stimulating factor as initial chemotherapy for metastatic breast cancer," J. Clin. Oncol., 11: 1943-1951 (1993), increasing the dose of paclitaxel to 250 mg / m 2 resulted in a response rate (60%) greater than the currently approved dose of 175 mg / m 2 (26%) for paclitaxel. It has been found that However, these results were not reproduced because of the high toxicity at these high doses. However, the study provides evidence for the possibility of an increase in response rate at increasing paclitaxel doses.
Since Taxol requires pre-dosing, which often requires a patient to be hospitalized daily, it is highly desirable to develop a paclitaxel formulation that eliminates the need for pre-dosing.
It is highly desirable to develop paclitaxel preparations that do not cause hypersensitivity because preparations for Taxol are necessary by hypersensitivity related to Taxol administration. It is also desirable to develop paclitaxel formulations that do not cause neurotoxicity.
It is highly desirable to develop paclitaxel formulations that do not require hospitalization of the treated donor because taxol infusions usually require pre-dose and prior to infusion monitoring and recording. Do.
Since higher doses of Taxol have achieved improved clinical response but have been demonstrated to be highly toxic, it is desirable to develop paclitaxel formulations that can achieve these doses without this toxicity.
Since the toxicity limiting dose of Taxol has proven to be brain and neurotoxic, it is desirable to develop paclitaxel formulations that reduce this toxicity.
In addition, it is desirable to eliminate the preparation because it increases patient discomfort and increases the cost and duration of treatment.
In addition, it is desirable to shorten the infusion period of paclitaxel currently administered 3 to 24 hours to minimize the hospitalization period of the patient.
Since taxol is currently approved to be administered at a concentration of 0.6 to 1.2 mg / ml and a typical dosage for humans is about 250 to 350 mg, this requires an injection volume of greater than 300 ml. It is desirable to lower the infusion volume by developing stable paclitaxel formulations at higher concentrations to reduce the duration of administration.
Taxol infusions do not contain cremaphores and therefore leach out toxic substances from conventionally used plastic tubes or bags because they have the limitation of using special intravenous tubes and bags or bottles due to leaching of plasticizers by cremaphores in taxol formulations. It is desirable to develop paclitaxel formulations that do not.
<Overview of invention>
Accordingly, it is an object of the present invention to provide a pharmacologically active agent in an unmodified form (e.g., paclitaxel, taxane, Taxotere, etc.).
It is also an object of the present invention to deliver pharmacologically active agents in compositions of microparticles or nanoparticles that can be suspended in suitable biocompatible liquids.
It is yet another object of the present invention to provide a method for forming ultra-microparticles (nanoparticles) of pharmacologically active agents from oil-in-water emulsions by solvent evaporation techniques. Some methods use proteins as stabilizers. Some methods are performed without any conventional surfactants, and without any polymeric core material.
Objects other than this object of the present invention will become apparent from the specification and claims.
In accordance with the present invention, the inventors have discovered that substantially water-insoluble pharmacologically active agents can be delivered in the form of microparticles or nanoparticles suitable for parenteral administration in aqueous dispersions. This delivery mode requires the administration of a pharmaceutically active agent (eg, paclitaxel) that is substantially water insoluble in an emulsion containing, for example, ethanol diluted with saline and polyethoxylated castor oil (eg, paclitaxel). See Norton et al., Abstracts of the 2nd National Cancer Institute Workshop on Taxol & Taxus, September 23-24, 1992. A disadvantage of such known compositions lies in their properties causing allergic side effects.
Thus, according to the present invention, there is provided a process for the formation of nanoparticles of pharmacologically active agents by solvent evaporation techniques from oil-in-water emulsions prepared under various conditions. For example, high shear forces (eg, sonication, high pressure homogenization, etc.) can be used without any conventional surfactant, and without any polymeric core material, to form a matrix of nanoparticles. Instead, proteins (eg human serum albumin) are used as stabilizers. In a separate method, nanoparticles can be formed by simply selecting a material that spontaneously forms a microemulsion, without any high shear forces.
The present invention also provides a method for the reproducible formation of typically small nanoparticles (less than 200 nm in diameter) that can be sterile filtered through a 0.22 nm filter. This can be achieved by adding a water insoluble solvent (eg ethanol) to the organic phase and carefully selecting the organic phase form, phase fraction, and concentration of drug in the organic phase. Since formulations containing large amounts of protein (eg albumin) cannot be sterilized by conventional methods such as autoclaving due to the thermal coagulation of the protein, the ability to form nanoparticles that are filterable in a 0.22 nm filter is important and meaningful.
According to another embodiment of the present invention, the inventors have developed a composition useful for in vivo delivery of a substantially water insoluble pharmacologically active agent. The composition of the present invention comprises a substantially water insoluble pharmacologically active agent (as solid or liquid) contained within the polymer shell. The polymer shell is a biocompatible crosslinked polymer. Polymeric shells containing substantially water-insoluble pharmacologically active agents can be suspended in a biocompatible liquid for administration.
The present invention provides a drug delivery system in which a portion of the pharmacologically active molecule is bound to a protein (eg, human serum albumin), which is readily available in vivo upon administration to a mammal. Another portion of the pharmacologically active agent is contained within the nanoparticles coated with the protein. Nanoparticles containing pharmacologically active agents exist as pure active ingredients without dilution by any polymeric matrix.
Most of the conventional pharmacologically active agents are bound to a carrier protein such as, for example, serum albumin (via hydrophobic or ionic interactions) and circulate along the blood flow. The methods of the invention and the resulting compositions provide pharmacologically active agents that are "pre-bound" to the protein (via hydrophobic or ionic interactions) prior to administration.
The present specification demonstrates all the above-mentioned methods of in vivo use for paclitaxel, an anticancer agent capable of binding to human serum albumin (see, eg, Kumar et al., Research Communications in Chemical Pathology and Pharmacology, 80: 337 ( 1993). Since the concentration of albumin in the particles of the present invention is higher than that of Taxol® (Bristol-Myers Squibb Company), a large amount of drug in the form of a molecule bound to albumin, a natural carrier of the drug in the blood stream, is provided. .
Another advantage is the binding capacity of human serum albumin to bind paclitaxel as well as other drugs that enhance the ability of paclitaxel to adsorb on the surface of the particles. Albumin is present on the colloidal drug particles (formed by the removal of organic solvents), thus promoting the formation of long-term stable colloidal dispersions due to the combination of electrical repulsion and steric stabilization.
According to the present invention, ultrafine particles in powder form are provided which can be easily reconstituted in water or brine. This powder can be obtained by freeze drying after the water has been removed. Human serum albumin functions as a structural component of some of the nanoparticles of the invention and also functions as cryoprotectants and reconstitution aids. The particles filterable through a 0.22 μm filter with the method according to the process of the invention described above are dried or lyophilized and then produced into sterile solid preparations useful for intravenous infusion.
In another aspect, the present invention provides a composition of anticancer drug, such as paclitaxel, in the form of nanoparticles in a liquid dispersion or as a solid that can be easily reconstituted for administration. Because of the particular nature of certain drugs, such as paclitaxel, such compositions cannot be obtained with conventional solvent evaporation methods that rely on the use of surfactants. In the presence of various surfactants, large drug crystals (eg, sizes from about 5 μm to several hundred μm) are formed during storage for several minutes after the manufacturing process. The size of such crystals is typically much larger than the size that can be administered intravenously.
While it is understood that the particles according to the invention may be crystalline, amorphous, or mixtures thereof, it is usually preferred that the medicament is present in the formulation in an amorphous form. This facilitates dissolution and adsorption resulting in better bioavailability.
Paclitaxel, an anticancer agent, has prominent clinical activity among many human cancers, including ovarian, breast, lung, esophagus, head and neck areas, bladder and lymph cancer. It has recently been demonstrated for metastatic breast cancer that has failed to treat ovarian cancer with cisplatin and one previous combination chemotherapy. Since the main limitation of Taxol® is its poor solubility, the BMS formulation contains 50% cremaphor EL and 50% ethanol as the solubilizing vehicle. Each vial of this formulation contains 30 mg of paclitaxel dissolved at a concentration of 6 mg / ml. Prior to intravenous administration, this preparation should be diluted 1:10 in saline for the final dose solution containing 0.6 mg / ml of paclitaxel. This agent is used in animals [Lorens et al., Agents Actions, 7: 63-67 (1987)] and humans [Weiss et al., J. Clin. Oncol., 8: 1263-68 (1990)], it is necessary to pretreat patients with corticosteroids (eg dexamethasone) and antihistamines. Higher dilution results in a larger injection volume (typical dosage of 175 mg / m 2 or less than 1 L) and an injection time of 3 to 24 hours. Thus, there is a need for a separate, less toxic preparation for paclitaxel.
Capsol® is a cremaphor-free formulation of the novel anticancer drug paclitaxel. We believe that based on animal studies, cremaphor-free formulations significantly reduce toxicity and will not require predose to patients. Preparations are necessary to reduce anaphylaxis and hypersensitivity caused by cremaphore in Paclitaxel's recently approved and commercially available Taxol® formulation. Capsol® is a lyophilized powder for reconstitution and intravenous administration. When reconstituted with a suitable aqueous medium such as 0.9% sodium chloride injection or 5% dextrose injection, the capsol® forms a stable colloidal solution of paclitaxel. The size of the colloidal suspension may range from 20 nm to 8 μm, preferably from about 20 to 400 nm. The two main components of Capsol® are unmodified paclitaxel and human serum albumin (HSA). Since HSA is readily soluble in water, Capsol® can be reconstituted with paclitaxel at any desired concentration, limited only by the solubility limit of HSA. Thus, Capsol® can be reconstituted at a wide range of concentrations from diluent (0.1 mg / ml paclitaxel) to concentrate (20 mg / ml paclitaxel). This significantly reduces the dosage volume.
In accordance with the present invention, there are provided compositions and methods useful for in vivo delivery of biological agents in nanoparticle form suitable for parenteral administration in aqueous suspension. The composition of the present invention is stabilized by a polymer. The polymer is a biocompatible material such as albumin protein. The use of the composition of the present invention for the delivery of a biological agent eliminates the need to administer the biological agent in a vehicle or toxic diluent such as, for example, ethanol and polyethoxylated castor oil, diluted in saline (eg [ Norton et al., Abstracts of the 2nd National Cancer Institute Workshop on Taxol & Taxus, September 23-24, 1992). A disadvantage of these known compositions lies in their properties, which indicate severe allergies and other side effects.
The delivery of biological agents in the form of particle suspensions targets these organs as they are taken up by the cell's reticuloendothelial (RES) system into the organs such as the liver, lungs, spleen, lymph circulation, and the like. Targeting RES containing organs can be controlled by administering different routes using particles of various sizes. However, when administered to rats, Capsol® accumulates in tissues other than RES containing tissues such as prostate, pancreas, testes, lavage tube, bone, etc. at significantly higher levels than similar amounts of Taxol®. I found it surprisingly surprising.
Accordingly, the capsule of the present invention of paclitaxel, a nanoparticle formulation, is a paclitaxel such as Taxol® in tissues such as the prostate, pancreas, testes, lavage, bone, etc., i.e. organs that do not contain RES. Concentrated to significantly higher levels than the non-particle formulation of. Thus, Capsol® can be used to treat cancer of these tissues with higher efficacy than Taxol®. However, because the distribution to many other tissues is similar to Capsol® and Taxol®, it is expected that Capsol® maintains anticancer activity at least at a level similar to that of Taxol® in other tissues. do.
The basic position in the prostate is dependent on the particle size of the formulation (20-400 nm), or the presence of protein albumin in the formulation, thereby positioning in prostate tissue via specific membrane receptors (gp60, gp18, gp13, etc.). Likewise, other biocompatible, in vivo degradable polymers other than albumin exhibit specificity for certain tissues, such as the prostate, by which the above-mentioned properties cause a high local concentration of paclitaxel in these tissues. Such biocompatible materials are within the scope of the present invention. Preferred embodiments of the compositions for achieving high local concentrations of paclitaxel in the prostate are paclitaxel and albumin preparations, free of cremaphor and in the particle size range of 20-400 nm. This embodiment has also been demonstrated to result in higher levels of paclitaxel in the pancreas, kidneys, lungs, heart, bones and spleen compared to administration of equivalent amounts of Taxol®. This property provides new uses of the formulations, including methods of providing high local concentrations into the vasculature for the treatment of lowering testosterone levels, achieving medical testicular stenosis, and treating restenosis.
In addition, paclitaxel, when administered as a capsol®, is converted to its metabolite by metabolism at a lower rate than when administered as a taxol®. This indicates that similar doses of paclotaxel have increased anticancer activity for a long time.
Surprisingly, the dose of capsol® and Taxol® in rats in equivalent amounts to paclitaxel resulted in a much higher degree of myelosuppression in the Taxol® group than the Capsol® group. This can lower the incidence of infection and fever episodes (eg, febrile neutropenia). It can also reduce the inter-treatment cycle time, which is typically 21 days. Thus, the use of Capsol® may provide a substantial advantage over Taxol®.
Surprisingly, it was found that Taxol® vehicle (cremaphor / ethanol diluted with saline) alone caused strong bone marrow suppression and showed severe hypersensitivity, resulting in lethality in several groups of mice. This reaction was not observed in more than equivalent amounts of Capsol® group. Thus, Capsol®, a preparation of paclitaxel that does not contain Taxol® vehicle, is substantially advantageous.
Surprisingly, when capsol (registered trademark) and taxol (registered trademark) are administered to rats in equivalent amounts of paclitaxel, the doses of capsol (registered trademark) are higher than that of taxol (registered trademark), as shown by a significantly higher LD 50 value. Much lower toxicity. This allows more, therapeutically effective amounts of paclitaxel to be administered to the patient. The literature describes an increase in response rate with higher doses of paclitaxel. The lower toxicity allows the capsol® formulation to be administered at higher dosages, allowing for the total potential of the drug.
Surprisingly also, Capsol®, a formulation of substantially water-insoluble drug paclitaxel, is stable when reconstituted at several different concentrations in the aqueous medium, including but not limited to 0.1-20 mg / ml. This provides a substantial advantage over Taxol®, such as a smaller injection volume during drug administration, which overcomes the known instability (eg precipitation) of Taxol® in the infusion line. There is no need to use an in-line filter. Thus, Capsol® significantly simplifies and improves administration of paclitaxel to patients.
In addition, Taxol® exhibits a neurotoxic effect even at low doses, while Capsol® does not show neurotoxicity in rats to be administered in equivalent amounts to paclitaxel, such as Taxol®.
The formulations of the present invention also administer paclitaxel, and other substantially water-insoluble pharmacologically active agents, which use smaller volumes of liquid and require significantly reduced dosing periods compared to the dosage volume and time required by the delivery system of the art. To do it.
In combination with a biocompatible polymer matrix, the formulations of the present invention (Capsol®) allow for localized sustained delivery of paclitaxel with lower toxicity and prolonged activity.
The surprising findings of the capsules® offer the possibility of substantially improving the quality of life of patients receiving paclitaxel.
Capsol® is a lyophilized powder containing only paclitaxel and human serum albumin. Due to the nature of the colloidal solution formed by the reconstitution of the lyophilized powder, toxic emulsifiers such as cremaphor (in the BMS formulation of paclitaxel) or polysorbate 80 (in the Longplan formulation of docetaxel), and solvents such as ethanol, which dissolve the drug No solvent is needed. Removing the toxic emulsifier will reduce the incidence of severe hypersensitivity and anaphylactic reactions known to occur with taxol products.
In addition, steroids and antihistamines are prepared prior to drug administration.
As the toxicity is reduced, the dosage can be increased for higher efficacy, as indicated by the LD 10 / LD 50 study.
Reduction of bone marrow suppression (relative to BMS formulations) is expected to reduce the treatment cycle duration (usually three weeks) and improve the therapeutic outcome.
Capsol® can be administered at higher concentrations (up to 20 mg / ml), allowing for lower infusion volume and intravenous mass, compared to BMS preparations (0.6 mg / ml).
Taxol® has the further disadvantage that it is only injectable with nitroglycerin polyolefin infusion sets due to the leaching of the plasticizer standard injection tubing into the formulation. Capsol® does not show leaching and can be used for any standard infusion. In addition, only glass or polyolefin containers can be used for all cremaphor storage containing solutions. Capsules do not have this limitation.
Another problem with Taxol® is the precipitation of paclitaxel in the underlying catheter. This causes a poorly controlled dosage of immobility. Due to the inherent stability of the colloidal solution of the new formulation, Capsol®, precipitation problems are alleviated. Due to its sedimentation problem, it is necessary to use a line filter to remove precipitates and other particulates when administering Taxol®. Capsol® does not have this problem because of its inherent stability.
The literature states that particles of several hundred nm size are preferentially distributed into cancer through leaky blood vessels in the cancerous site. Thus, the colloidal particles of paclitaxel in Capsol® may have a preferential desired effect that significantly reduces the side effects of paclitaxel administered in the Taxol® formulation.
It is therefore a first object of the present invention to provide novel formulations of paclitaxel that provide the desired features.
Another object of the present invention is to provide paclitaxel in specific tissues, thereby providing higher anticancer activity to such tissues.
Another object of the present invention is to administer paclitaxel at a concentration of about 2 mg / ml or more to reduce the infusion volume.
It is also an object of the present invention to provide a paclitaxel formulation which does not contain Taxol® vehicle.
It is another object of the present invention to provide a paclitaxel formulation which improves the quality of life of a patient receiving paclitaxel for cancer treatment.
The present invention relates to a process for the preparation of particulate vehicles for intravenous administration of pharmacologically active agents and to novel compositions prepared thereby. In certain aspects, the present invention relates to methods of in vivo delivery of substantially water-insoluble pharmacologically active agents (eg anticancer drug paclitaxel). In another aspect, a dispersible colloidal system containing a water insoluble pharmacologically active agent is provided. Suspended particles may be formed of 100% active agent or may be coated with a polymer shell formulated with a biocompatible polymer, the diameter of which is less than about 1 μm. The colloidal system of the present invention can be prepared without the use of conventional surfactants or any polymeric core matrix. In a preferred aspect of the present invention, a process for producing very small particles that can be sterile filtered is provided. The polymer shell contains particles of the pharmacologically active agent, and optionally a biocompatible dispersant into which the pharmacologically active agent can be dissolved or dispersed. Accordingly, the present invention provides a drug delivery system in liquid form or in redispersible powder form. Either form provides both readily bioavailable drug molecules (ie, drug molecules molecularly bound to the protein) and pure drug particles coated with the protein.
The present invention also relates to methods of use and preparation of the compositions (formulations) of drugs such as the anticancer agent paclitaxel. In one aspect, the paclitaxel formulation known as Capxol® is significantly less toxic and has greater efficacy than the commercially available formulation of paclitaxel, Taxol®. In another aspect, the new formulation capsules® are ubiquitous in certain tissues after parenteral administration to increase the therapeutic efficacy of cancers associated with the tissues.
1 shows the results of intravenous administration of paclitaxel nanoparticles to tumor-bearing mice (n = 5 per group), showing complete regression of tumors in treatment group (■) compared to control group (•) receiving saline. Substantially unregulated tumor proliferation is seen in the control group. The dose of the treatment group is paclitaxel 20 mg / kg administered as an intravenous mass for 5 consecutive days.
Figure 2 shows the results of intradermal collagen injection after intraperitoneal administration of paclitaxel nanoparticles to the rat with advanced arthritis in the forefoot. The volume of the forefoot was measured and the severity of the disease described. The forefoot volume was 100% at the start of treatment. Day 0 represents initiation of treatment. Control group receiving three group-saline (n = 2, denoted by solid line and “untreated” in the figure); A first treatment group receiving paclitaxel nanoparticles at a dose of 1 mg / kg (n = 4, denoted by bold lines and shown in the figure as “1.0 paclitaxel nanoparticles 1.0 mg / kg); and 0.5 mg / kg paclitaxel nanoparticles A second treatment group receiving a combination of a dose and a dose of prednisone 0.2 mg / kg (n = 4, shown in bold lines and shown in the figure as “prednisone 0.2 mg / kg + paclitaxel nanoparticle 0.5 mg / kg) have. While the control group increased forefoot volume over the same period, the two treatment groups showed a dramatic decrease in forefoot volume over the same period as arthritis regressed.
3 shows the results of bone marrow suppression studies in rats. Capsol® is administered to the first group of three rats, Taxol® is administered to the second group of three rats, both at a dosage of 7 ml of formulation per kg of body weight. , Each formulation contains 5 mg / kg paclitaxel. All doses are given as IV masses through the tail vein. 3 shows the percent change in white blood cells (WBC) that function as bone marrow suppression indicators.
4 shows a preliminary study using Capsol® to determine target dosage range and efficacy. Mice (n = 10) were implanted subcutaneously into MX-1 breast cancer and treatment started when the size of the cancer reached about 150-300 mg. This occurred on day 12 and treatment commenced on day 13 after initial seeding. Capsol® was reconstituted in brine to obtain a colloidal solution of nanoparticles of paclitaxel. Tumored mice (n = 5) were treated with a dose of 20 mg / kg (denoted as VIV-1) with a reconstituted capsol® given as a tail vascular infusion mass daily for 5 days. Tumor control group (n = 5) received saline only according to the same scheme. Tumor size was monitored by time function. The control group showed a significant increase in tumor weight above the mean of 4500 mg, and animals in this group were sacrificed between Days 28 and 39. On the other hand, the treatment group showed significant efficacy, and tumors were not detected in all animals until day 25. Animals in this group were sacrificed on day 39, which showed no tumor recurrence and development.
According to the present invention, paclitaxel in a patient receiving paclitaxel treatment comprising systemic administration of the paclitaxel in a pharmaceutically acceptable formulation to a patient receiving paclitaxel at a dose of at least 175 mg / m 2 over a period of up to 2 hours A method of reducing toxicity is provided.
In accordance with the present invention, a pretreatment prior to paclitaxel administration is required, comprising systemically administering paclitaxel in a pharmaceutically acceptable formulation to a patient receiving paclitaxel at a dose of at least 135 mg / m 2 over a period of up to 2 hours. Also provided is a method of administering paclitaxel to a patient in need thereof without paclitaxel treatment.
According to a further embodiment of the present invention, systemic administration of said paclitaxel in a pharmaceutically acceptable formulation to a patient in need of paclitaxel at a treatment cycle of less than 3 weeks over a period of up to 2 hours at a dose of at least 135 mg / m 2 It involves doing.
According to a further embodiment of the invention, a patient in need of paclitaxel treatment comprising systemic administration of said paclitaxel in a dosage of at least 250 mg / m 2 in a pharmaceutically acceptable formulation to a patient in need of paclitaxel treatment Provided are paclitaxel administration methods.
According to a further embodiment of the invention there is a need for paclitaxel, which comprises systemically administering said paclitaxel in a formulation that can be safely administered to a patient in need thereof using medical hardware made of extractable ingredient-containing material. A method of administering paclitaxel to a patient is provided.
According to a further embodiment of the invention, a method of administering paclitaxel to a patient in need of paclitaxel comprising systemic administration of the paclitaxel in a formulation which can be safely administered to a patient in need of paclitaxel without the use of an inline filter. To provide.
According to a further embodiment of the present invention there is provided a method of paclitaxel administration to a patient in need of paclitaxel, comprising systemically administering the full dose of paclitaxel to a patient in need of paclitaxel in a volume of less than 250 ml.
According to a further embodiment of the present invention there is provided a method of paclitaxel administration to a patient in need of paclitaxel comprising systemic administration of the paclitaxel to a patient in need thereof at a rate of 50 mg / m 2 / hour or more.
According to a further embodiment of the present invention, paclitaxel hemotoxicity of a patient receiving paclitaxel treatment comprising paclitaxel in a pharmaceutically acceptable formulation suitable for systemic administration at a dose of 175 mg / m 2 over a period of up to 2 hours. Reduced paclitaxel formulations are provided.
In accordance with a further embodiment of the present invention, paclitaxel, which does not require preparatory dosing before paclitaxel, comprises paclitaxel in a pharmaceutically acceptable formulation suitable for systemic administration at a dosage of 135 mg / m 2 over a period of up to 2 hours. Paclitaxel formulations to patients in need are provided.
According to a further embodiment of the invention, paclitaxel is required in less than 3 weeks of treatment cycle, comprising paclitaxel in a pharmaceutically acceptable formulation suitable for systemic administration at a dose of 135 mg / m 2 over a period of up to 2 hours. Paclitaxel formulations suitable for administration to a patient,
According to a further embodiment of the present invention there is provided a paclitaxel formulation suitable for administration to a patient in need of paclitaxel, comprising paclitaxel in a pharmaceutically acceptable formulation that does not contain cremaphor.
According to a further embodiment of the invention, there is provided a paclitaxel lyophilized formulation suitable for reconstitution to administration to a patient in need of paclitaxel.
According to a further embodiment of the present invention there is provided a frozen formulation of paclitaxel suitable for thawing to administration to a patient in need of paclitaxel.
According to a further embodiment of the present invention there is provided a liquid formulation of paclitaxel comprising paclitaxel and water at a concentration of at least 2.0 mg / ml.
According to a further embodiment of the present invention, there is provided a drug delivery system comprising solid or liquid particles which are substantially water-insoluble pharmaceutically acceptable active agents.
The protein coating has a free protein associated therewith, the pharmacologically active agent is contained within the protein coating, the pharmacologically active agent is associated with the organic protein, and the average diameter of the particle size is about 1 μm or less.
According to a further embodiment of the invention, there is provided a drug formulation suitable for drug administration by inhalation into a patient in need of paclitaxel, the formulation optionally together with excipients, protein microparticles of about 1-10 μm in size Wherein the microparticles comprise drug nanoparticles having a size of about 50 to 1,000 nm.
According to the present invention, there is also provided a process for the preparation of a substantially water-insoluble pharmacologically active agent for in vivo delivery.
a) spontaneously forming a microemulsion
i) an organic solvent having the active agent dissolved therein,
ii) water or aqueous solutions,
iii) surfactants, and
iv) combining the cosurfactant,
b) removing the organic solvent to obtain a nanoparticle suspension of the active agent in water.
According to a further embodiment of the present invention, there is provided a process for preparing nanoparticles containing an active agent, the method comprising combining a nonvolatile phase, a volatile phase, and a surfactant which spontaneously form a microemulsion, wherein The volatile phase comprises the active agent, and removal of the volatile phase yields a suspension of solid nanoparticles in the nonvolatile phase, the nanoparticles containing the active agent and an average diameter of less than 100 nm.
According to a further embodiment of the present invention, there is provided a process for preparing nanoparticles containing an active agent, the method comprising combining a volatile phase with a non-volatile phase that spontaneously forms a microemulsion, wherein the non-volatile phase is And removing the nonvolatile phase yields solid nanoparticles in the nonvolatile phase, the nanoparticles containing the active agent and having an average diameter of less than 100 nm.
It is particularly advantageous to observe that the compositions produced by the above-mentioned methods provide various forms of pharmacologically active agents with very low toxicity. Also described herein are methods for the preparation of low toxicity pharmacologically active agents, such as paclitaxel.
In a preferred embodiment, the above-mentioned particles have an average diameter of about 200 nm or less. Such particles can be injected into sterile filters, which is particularly advantageous as there is no need for more potent treatment to achieve sterility of the solution containing the desired pharmacologically active agent.
"Paclitaxel" as used herein encompasses all forms, variants, and derivatives of paclitaxel, such as taxotere, unless otherwise noted.
Capsol® is a registered trademark of the paclitaxel formulation marketed by the applicant's assignee. Capsol® as used herein is a shorthand for the protein-coated paclitaxel nanoparticles prepared by the method of Example 1. Capsol® is a novel cremaphor-free formulation of the anticancer drug paclitaxel. We believe that based on animal studies, cremaphor-free formulations are significantly less toxic and do not require predose to patients. Pre-dose is necessary to reduce anaphylaxis and hypersensitivity caused by cremaphore, which has been recently demonstrated and designated as a Taxol® formulation of paclitaxel. Capsol® is a lyophilized powder for reconstitution and intravenous administration. Each container of Capsol® contains palm oil 30 mg paclitaxel and about 400 mg human serum albumin. When reconstituted with a suitable aqueous medium such as 0.9% Sodium Chloride Injection or 5% Extrose Injection, Capsol® forms a stable colloidal solution of paclitaxel. Colloidal nanoparticles are typically less than 400 nm in size. Nanoparticles are prepared by high pressure homogenization of USP human serum albumin solution and paclitaxel solution in organic solvents. This solvent is then removed to produce a colloidal suspension or solution of paclitaxel in human serum albumin. This suspension is sterile filtered and lyophilized to yield Capsule. This formulation does not contain other additional excipients or stabilizers. Sterility of the product is confirmed by aseptic manufacturing methods and / or sterile filtration. The two main components of Capsol® are unmodified paclitaxel and human serum albumin (HSA). Since HSA is readily soluble in water, Capsol® can be reconstituted to any desired concentration of paclitaxel, only limited to the solubility limit of HSA. Thus, Capsol® can be extensively reconstituted from diluent (0.1 mg / ml paclitaxel) to concentrate (20 mg / ml paclitaxel). This makes the dose volume quite small.
As used herein, the term “in vivo delivery” refers to oral, intravenous, subcutaneous, intraperitoneal, intradermal, intramuscular, inhalation, topical, transdermal, suppository (intestinal), vaginal suppository (vaginal), urethra, intraportal Means delivery of the pharmacologically active agent by the route of administration, such as intrahepatic, intraarterial, or body fluid.
The term "smile" as used herein is a unit of measurement of 1/1000 of a millimeter.
As used herein, the term "biocompatible material" refers to a material that does not alter or affect the biosystem in any reversible manner so that it can be detected in the direction of introduction.
Substantially water-insoluble pharmacologically active agents used in the practice of the present invention include pharmacologically active agents, diagnostic agents, nutrients, and the like. Pharmacologically active agents include the following:
Analgesics / antipyretic agents (e.g., aspirin, acetaminophen, ibuprofen, naproxen sodium, buprenorphine hydrochloride, propoxyphene hydrochloride, naphsyl proppropene pene, meperidine hydrochloride, hydromorphone hydrochloride, morphine sulfate, oxycodone hydrochloride , Codeine phosphate, dihydrocodene of heavy tartaric acid, pentazosin hydrochloride, heavy tartaric acid hydrocodone, tartaric acid leborpanol, diflunisal, salicylic acid trolamine, nalbuphine hydrochloride, mephenamic acid, tartaric acid butyrolpanol, salicylic acid sheet, butalbital, citrate Phenyltoloxamine, diphenhydramine citrate, metotrimeprazine, cinnamicrine hydrochloride and meprobamate);
Anesthetics (eg, cyclopropane, efluran, halotan, isoflurane, methoxyflurane, nitrus oxide, propofol, etc.);
Anti-asthmatic drugs (e.g. azelastine, ketotifen, traxanox, ampexanox, chromoline, ibudilast, montelukast, nedocromyl, oxatomide, franlukast, cerastast, supra Tast tosylate, tiaramid, kafirlukast, ileurton, beclomethasone, budesonide, dexamethasone, flunisolide, trimkinolone acetonide and the like);
Antibiotics (eg, neomycin, streptomycin, chloramphenicol, cecalosporin, ampicillin, penicillin, tetracycline, etc.);
Antidepressant drugs (e.g. neopopam, oxyfertin, doxepin hydrochloride, amoxapine, trazodone hydrochloride, amitriptyline hydrochloride, aprotriline hydrochloride, phenelzin sulfate, decipramine hydrochloride, Nortriptyline hydrochloride, Tranylcyclominate sulfate, Fluoxetine hydrochloride, Doxepine hydrochloride, Imipramine hydrochloride, Imipramine pamoate, Portriftyline, Amitriphylline hydrochloride, Isocar Copying zide, desipramine hydrochloride, trimimipramine maleate, protriphthyl hydrochloride, etc.);
Antidiabetic drugs (eg biguanides, hormones, sulfonylurea derivatives, etc.)
Antifungal agents (eg, griseofulvin, keloconazole, amphotericin B, nistatin, candicidin, etc.);
Antihypertensive agents (e.g., propanolol, propapenone, oxyprenol, nifedipine, reserpin, trimetaphan campylate, phenoxybenzamine hydrochloride, pargiline hydrochloride, decefidine, diazooxide, guar Netidine monosulfate-, minoxidil, nescinnamin, sodium nitrofusid, laupulia serpentina, alseroxylon, phentolamine mesylate, reserpin, etc.);
Anti-inflammatory drugs (e.g. (sterile) indomethacin, naproxen, ibuprofen, ramipenazone, pyricampam, (steroidal) cortisone, dexamethasone, fluazacort, hydrocortisone, prenisolone, prednisone, etc.);
Antitumor agents (e.g., adriamycin, cyclophosphamide, actinomycin, bleomycin, duanorubicin, doxorubicin, epirubicin, mitomycin, methotrexate, fluorouracil, carboplatin, carmustine (BUNU), methyl-CCNU, cisplatin, etosides, interferon, camptothecins and derivatives thereof, penesterin, paclitaxel and derivatives thereof, taxotere and derivatives thereof, vinblastine, vincristine, carmoxifene, etoposide, Skin patching and the like);
Anti-anxiety agents (e.g. lorazepam, buspirone hydrochloride, prazepam, chlodiazepoxide hydrochloride, oxazepam, chlorazate dipotassium, diazepam, hydroxyzine pamoate, piroxyzin hydrochloride, alprazolam , Dropperidol, palazepam, chlormezanone, dantrolene and the like);
Immunosuppressive agents (eg, cyclosporin, azathioprine, myzoribin, FK506 (tacrolimus), etc.);
Antimigraine drugs (eg, ergotamine tartrate, propanolol hydrochloride, isomeptane mucate, dichloralfenazone, etc.);
Sedatives / hypnotics (e.g. barbiturates (e.g. pentobarbital, pentobarbital sodium, secobarbital sodium), benzodiazepines (e.g. fluzepam hydrochloride, triazolam, tomazepam, midazolam hydrochloride), Etc);
Antianginal agents (eg beta-adrenergic blockers, calcium channel blockers (eg nifedipine, diltiazem hydrochloride, etc.), nitrates (eg nitroglycerin, isosorbide dinitrate, pentaerythritol tetra) Nitrate, erythryl tetranitrate), and the like);
Antipsychotics (e.g. haloperidol, roxapsin succinate, roxaphine hydrochloride, thiolidazine, thiolidazine hydrochloride, thiotixene, flufenazine, hydrofenazine decanoate, flufenazine deanthate, trifluchloride hydrochloride Operazine, chlorpromazine hydrochloride, perphenazine, lithium citrate and prochlorperazine;
Antihypertensives (eg lithium carbonate);
Antiarrhythmic medicines (e.g., brethlium tosylate, esmoldol hydrochloride, verapamil hydrochloride, amiodarone, encaine hydrochloride, digoxin, digitoxin, mesylenetin hydrochloride, disopyramid hydrochloride, procaineamide hydrochloride, quinidine sulfate, quiniconate gluconate) Dine, polygalacturonic acid quinidine, plecaine acetate, tocaine hydrochloride and lidocaine hydrochloride;
Anti-arthritis drugs (e.g., phenylbutazone, sulindac, penicillamine, salsalate, pyroxicam, azathioprine, indomethacin, meclofenamate sodium, gold sodium thiomalate, ketoprofen, auranopine , Aurothioglucose, tolmethin sodium, etc.);
Antigout agents (eg, colchicine, allopurinol, etc.);
Anticoagulants (eg, heparin, heparin sodium, warfarin sodium, etc.);
Thrombolytics (eg, urokinase, streptokinase, altoplase, etc.);
Antifibrinolytic agents (eg aminocaproic acid);
Blood flow agents (eg, pentoxifylline);
Hemoplatelets (eg, aspirin, empyrin, ascriptin, etc.);
Anticonvulsants (e.g. valproic acid, divalproate sodium, phenytoin, phenytoin sodium, clonazepam, pyrimidonedon, phenobarbitol, phenobarbitol sodium, carbamazepine, amobarbital sodium, metsuccimid, Metharbital, mepobarbital, mefenitoin, pensuccimid, paramethadione, etotoin, phenacemid, secobarbitol sodium, chlorazate dipotassium, trimetadione and the like);
Anti-Parkinson's agents (eg, etosuccimid, etc.);
Antihistamines / antipruritic agents (e.g., hydroxyzin hydrochloride, diphenhydramine hydrochloride, chlorpheniamine maleate, brompheniramine maleate, cipropeptadine hydrochloride, terfenadine, clemastine fumarate, tripletlori Dean hydrochloride, carbinoxamine maleate, diphenylpyraline hydrochloride, phenanthamine tartrate, azatadine maleate, tripelinamine hydrochloride, dexchlorpheniamine maleate, metdilazine hydrochloride, trimrazine tart Rate, etc.);
Drugs useful for calcium regulation (eg, calcitonin and parathyroid hormone, etc.);
Antibacterial agents (e.g. amikacin sulfate, aztreonam, chloramphenicol, chloramphenicol, palmitic acid chloramphenicol, chloramphenicol sodium succinate, ciprofloxacin hydrochloride, klindamycin hydrochloride, palmitic acid klindamycin, phosphalicinmycin, metronidazole, metronidazole hydrochloride, gentamicin hydrochloride, gentamicin hydrochloride Mycin, tobramycin sulfate, vancomycin hydrochloride, polymyxin sulfate B, colistetate sodium and colistin sulfate);
Antiviral agents (eg, interferon gamma, zidobudine, amantadine hydrochloride, ribavirin and acyclovir, etc.);
Antimicrobial agents (e.g., cephalosporins (e.g., cefazoline sodium, cepradine, cefacller, cefapirine sodium, ceftioxime sodium, cephaperazole sodium, cetethetan disodium, ceputoxime azotyl, cefotaxime sodium, Sepaderoxyl monohydrate, ceftazidime, cephalexin, cephalotin sodium, cephalexin monohydrate, cefamandol naphate, cefacithin sodium, cenisidide sodium, celaridide, ceftriaxone sodium, ceftazidim , Cephadoxyl, cepradine and cefuroxime sodium, etc., penicillin (e.g., ampicillin, amoxicillin, penicillin G benzatin, cyclacillin, ampicillin sodium, penicillin G potassium, penicillin V potassium, piperacillin sodium, oxacillin Sodium, Bacampicillin, Kreaxacillin Sodium, Ticarcillin Disodium, Azolocillin Sodium, Carbenicillin Indanyl Sodium, Penicillin G Potassium, Penicillin G Procaine, Methicillin Sodium and naphcillin sodium, etc.), erythromycin (e.g., erythromycin ethyl succinate, erythromycin, erythromycin estoleate, erythromycin lactobionate, erythromycin sierate and erythromycin ethyl succinate, etc.) and tetra Cyclins (eg, tetracycline hydrochloride, doxycycline cyclate and minocycline hydrochloride, etc.);
Anti-infectives (eg, GM-CSF);
Bronchodilators (e.g., sympathetic stimulants (e.g. epinephrine hydrochloride, methaproterenol sulfate, terbutalene sulfate, isotarin, isotarin mesylate, isoetherin hydrochloride, albuterol sulfate, albuterol, bitol) Terrol mesylate, isoproterenol hydrochloride, terbutaline sulfate, epinephrine bitartarate, methaproterenol sulfate, epinephrine, epinephrine bitartarate), anticholinergic agents (e.g., ipratropium bromide), xanthine (e.g., aminophylline, Diphylline, metaproterenol sulfate, aminophylline), mast cell stabilizer (e.g. chromoline sodium), inhaled corticosteroids (e.g. fluolisolid, beclomethasone dipropionate, beclomethasone dipropio Nate monohydrate), salbutamol, beclomethasone dipropionate (BDP), ipratropium bromide, budesonide, ketotifen, salmeterol, xinapoate, terbuta sulfate , Triamcinolone, theophylline, nedo croissant wheat sodium sulfate meth Pro terephthalate play, albuterol and flu you solid and the like);
Hormones (e.g. androgens (e.g., danazole, testosterone cypionate, fluoxymesterone, ethyl testosterone, testosterone enaniate, methyltestosterone, fluoxymesterone, testosterone cypionate), estrogen (e.g. estradiol) , Estrophytate, complex estrogens), progestins (e.g. methoxyprogesterone acetate, noethynedrone acetate), corticosteroids (e.g. triamcinolone, betamethasone, betamethasone sodium phosphate, dexamethasone, dexamethasone sodium, dexamethasone acetate, prednisone, methyl Prednisolone Acetate Suspension, Triamcinolone Acetonide, Methylprednisolone, Prednisolone Sodium Phosphate, Methylprednisolone Sodium Succinate, Hydrocortisone Sodium Succinate, Methylprednisolone Sodium Succinate, Triamcinolone Hexacato Hydrocortisone, hydrocortisone cypionate, prednisolone, fluorocortizone acetate, paramethasone acetate, prednisolone tebulate, prednisolone acetate, prednisolone sodium phosphate and succinic acid hydrocortisone sodium ) And thyroid hormones (eg, levothyroxine sodium) and the like;
Hypoglycemic agents (eg, human insulin, purified bovine insulin, purified porcine insulin, glyburide, chlorpropamide, glypide, tolbutamide and tolazamide, etc.);
Hypolipidemic agents (eg, clofibrate, dextrothyroxine sodium, probucol, lovastatin and niacin, etc.);
Proteins (eg, DNases, alginases, superoxide dismutases and lipases, etc.);
Nucleic acids (eg, sense or antisense nucleic acids encoding any therapeutically active protein, including proteins described herein);
Erythropoietic stimulants (eg, erythropoietin);
Ulcer therapy / antireflective agents (eg, famotidine, cimetidine and ranitidine hydrochloride, etc.);
Anti-nausea medications (eg, mecrizine hydrochloride, nabilone, prochlorperazine, dimenhydrinate, promethazine hydrochloride, thiethylperazine and scopolamine, etc.);
Fat-soluble vitamins (eg, vitamins A, D, E, K, etc.);
Other drugs such as mitotans, bisadins, halonitrossoureas, anthracyclines, ellipsine and the like.
Examples of diagnostic agents for use in the practice of the present invention include ultrasound contrast agents, radiographic agents (e.g., iodo-octane, halocarbons, lenographene, etc.), magnetic contrast agents (e.g., fluorocarbons, fat-soluble paramagnetic compounds, etc.). In addition, other diagnostics are also included that are not readily delivered without some physical and / or chemical modifications to accommodate their substantially water insoluble properties.
Examples of nutrients for use in the practice of the present invention include amino acids, sugars, proteins, carbohydrates, fat soluble vitamins (eg, vitamins A, D, E, K, etc.), fats or any combination of two or more thereof.
<A. Nanoparticle Formation Using High Shear Homogenization>
An important difference between the pharmacologically active agents contained in the polymerization shells according to the present invention and the protein microspheres of the prior art lies in the nature of the formation and the final state of the protein after particle formation and the ability to transport water-soluble or substantially water-insoluble formulations. According to the invention, the polymer (eg protein) can be crosslinked by exposure to high shear conditions in a high pressure homogenizer. High shear is a biocompatible polymer (e.g., containing a disulfide containing dissolved or suspended pharmacologically active agent, optionally containing sulfhydryl or disulfide groups, which allows the shell of the crosslinked polymer to form around fine droplets of a non-aqueous medium). , Albumin). High shear conditions can cause tremendous local exotherms, e.g. oxidizing sulfhydryl moieties and / or interfering disulfide bonds present to cross-link the polymer to form new crosslinked disulfide bonds. Cavitation in the liquid to form is produced.
Unlike the methods of the present invention, the prior art glutaraldehyde crosslinking methods react mainly with all nucleophilic groups (eg, amine and hydroxyl groups) that are nonspecific and present in the protein structure. As taught in the prior art, thermal denaturation significantly and irreversibly alters protein structure. On the other hand, disulfide formation contemplated by the present invention does not substantially denature the protein. In addition, the particles of the substantially water-insoluble pharmacologically active agent contained in the shell are different from the crosslinked or thermally denatured protein microspheres of the prior art, because the polymer shells produced by the present invention are compared to the diameter of the coated particles. This is because it is relatively thin. The “shell thickness” of the polymer coating was determined to be about 25 nm for coating particles having a diameter of 1 μm (1000 nm) (by transmission transcription microscope). On the other hand, the microspheres of the prior art do not have a protein shell, but rather the protein is dispersed throughout the volume of the microspheres.
Thus, the pharmacologically active agents according to the invention may be suitable solvents, for example chloroform, methylene chloride, ethyl acetate, ethanol, tetrahydrofuran, dioxane, butanol, butyl acetate, acetonitrile, acetone, dimethylsulfoxide, dimethylformamide , Methyl pyrrolidinone and the like and a mixture of two or more thereof. Additional solvents contemplated for use in the practice of the present invention include soybean oil, coconut oil, olive oil, safflower oil, cottonseed oil, sesame seed oil, orange oil, limonene oil, C 1 -C 20 alcohol, C 1 -C 20 ester , C 3 -C 20 ketones, polyethylene glycols, aliphatic hydrocarbons, aromatic hydrocarbons, halogenated hydrocarbons and combinations thereof.
Unlike conventional methods of nanoparticle formation, polymers (eg polylactic acid) do not dissolve in solvents. The oil phase used in the preparation of the compositions of the invention typically contains only pharmacologically active agents dissolved in a solvent.
Next, the protein (eg, human serum albumin) is added to the aqueous phase to act as a stabilizer for the formation of stable nanoparticles. The protein is added at a concentration in the range of about 0.05-25% (w / v), more specifically in the range of about 0.5-5% (w / v). Unlike conventional methods of nanoparticle formation, surfactants (eg sodium laurylsulfate, lecithin, Tween 80, Pluronic F-68, etc.) are not added to the mixture.
Next, the emulsion is formed by homogenization under high pressure and high shear force. Such homogenization is conveniently carried out in a high pressure homogenizer, which is typically operated at a pressure in the range of about 3,000 to 60,000 psi. Preferably this method is carried out at a pressure in the range of about 6,000 to 40,000 psi. The resulting emulsion comprises very small nano sized drops of non-aqueous solvent (containing dissolved pharmaceutically active agent) and very small nano sized drops of protein stabilizer. Acceptable homogenization methods include methods for providing high shear and cavitation, such as high pressure homogenization, high shear mixers, ultrasonic digestion, high shear propellers, and the like.
Finally, the solvent is evaporated under reduced pressure to produce a colloidal system comprising protein coated nanoparticles and protein of the pharmacologically active agent and protein. Acceptable evaporation methods include the use of rotary evaporators, drop membrane evaporators, spray dryers, freeze dryers and the like. Ultrafiltration is also used for solvent removal.
Following evaporation of the solvent, the liquid suspension can be dried to obtain a powder containing the pharmacologically active agent and protein. The powder obtained can be administered to a mammal at any convenient time for redispersion in a suitable aqueous medium such as saline, buffered saline, water, buffered aqueous medium, amino acid solutions, vitamin solutions, carbohydrate solutions, and the like, or mixtures of two or more thereof. A suspension can be obtained. Methods contemplated for obtaining this powder include freeze drying, spray drying, and the like.
Another embodiment of the present invention provides an alternative for the formation of very small μm or smaller particles (nanoparticles), ie particles having a diameter of less than 200 nm. Such particles may be sterile filtered before use in the form of a liquid suspension. The ability to sterilize the final product (i.e. drug particles) of the production process of the present invention is difficult to sterilize dispersions containing high concentrations of protein (e.g. serum albumin) by conventional means such as autoclaves. very important.
In order to obtain sterile filterable particles (particles less than 200 nm), the pharmacologically active agent is first subjected to a substantially water immiscible organic solvent (eg, a solvent having a solubility of less than about 5% in water, for example chloroform) at high concentrations. ) To form an oil phase containing the pharmacologically active agent. Suitable solvents are disclosed above. Unlike conventional methods for forming nanoparticles, polymers (eg polylactic acid) do not dissolve in solvents. The oil phase used in the process of the invention contains only pharmacologically active agents dissolved in a solvent.
Next, a water miscible organic solvent (e.g., a solvent having a solubility in excess of about 10% in water, such as ethanol) is added to the range of about 1 to 99% (v / v), more preferably Is added to the oil phase at a final concentration ranging from about 5 to 25% (v / v). The water miscible organic solvent can be selected from solvents such as ethyl acetate, ethanol, tetrahydrofuran, dioxane, acetonitrile, butanol, acetone, propylene glycol, glycerol, dimethyl sulfoxide, dimethyl formamide, methyl pyrrolidinone and the like. Alternatively, a mixture of a water immiscible solvent and a water miscible solvent is first prepared and then the pharmacologically active agent is dissolved in this mixture.
Next, human serum albumin or any other suitable stabilizer as described above is dissolved in the aqueous medium. This component acts as a stabilizer for the formation of stable nano-sized droplets. Optionally, a sufficient amount of the first organic solvent (eg chloroform) is dissolved in the aqueous phase to bring it closer to the saturation concentration. A separate metered amount of organic phase (now containing pharmacologically active agent, first organic solvent and second organic solvent) is added to the saturated aqueous phase so that the phase fraction of the organic phase is about 0.5-15% (v / v). ), More preferably between 1 and 8% (v / v).
Next, a mixture comprising micro and nano sized droplets is formed by homogenization at low shear. This can be accomplished in a variety of ways that can be readily identified by one skilled in the art using, for example, conventional laboratory homogenizers operating in the range of about 2,000 to about 15,000 rpm. The homogenization is then carried out at high pressure (eg, in the range of about 3,000 to 60,000 psi). The resulting mixture comprises an aqueous protein solution (eg human serum albumin), a water insoluble pharmacologically active agent, a first solvent and a second solvent. Finally, the solvent produces colloidal dispersion systems (pharmacologically active agents and proteins) in the form of extremely small nanoparticles (ie particles ranging in diameter from about 10 nm to 200 nm) that can be easily evaporated under vacuum to be sterile filtered. The preferred size range of the particles is between about 50 and 170 nm, depending on the formulation and operating parameters.
Colloidal systems prepared according to the invention can also be converted to powder form by removing water, for example, by lyophilization or spray drying in a suitable temperature-time profile. The protein (eg, human serum albumin) itself acts as a cryoprotectant or cryoprotectant and the powder is easily added by adding water, saline or buffer without the need for conventional cryoprotectants such as mannitol, sucrose, glycine and the like. It is reconstituted. While not required, it is, of course, understood that conventional cryoprotectants can be added to the formulations of the present invention as needed.
Colloidal systems of pharmacologically active agents can deliver high doses of pharmacologically active agents in relatively small volumes. This minimizes patient discomfort and minimizes hospital stay for receiving large volumes of fluid. In addition, since the walls of the polymer shell or coating are generally fully degradable in vivo by proteolytic enzymes (for example when the polymer is a protein), there are substantially no side effects in the delivery system, which leads to linear formulation. This is in stark contrast to the serious side effects caused by this.
Numerous biocompatible polymers can be used in the practice of the present invention to form polymer shells substantially surrounding the water insoluble pharmacologically active agent. Basically all natural or synthetic polymers optionally containing sulfhydryl groups or disulfide bonds in the structure can be used to prepare disulfide crosslinked shells for particles of substantially water-insoluble pharmacologically active agents. Sulfhydryl groups or disulfide bonds may be present in the polymer structure in advance, or they may be introduced by appropriate chemical modification. For example, natural polymers such as proteins, peptides, poly nucleic acids, polysaccharides (e.g., starch, cellulose, dextran, alginate, chitosan, pectin, hihaluronic acid, etc.), proteoglycans, lipoproteins, etc. I'm a candidate.
Proteins contemplated for use as stabilizers according to the invention include albumin (containing 35 cysteine residues), immunoglobulins, casein, insulin (containing 6 cysteines), hemoglobin (containing 6 cysteine residues per unit of a 2 β 2 ), immunoglobulins , Alpha-2-polymer globulin, fibronectin, vitronectin, hybridogen, lipase and the like. Proteins, peptides, enzymes, antibodies and combinations thereof are a group of common stabilizers contemplated for use in the present invention.
Currently, the preferred protein for use as a stabilizer is silver albumin. Optionally, proteins such as known opsonins, which are alpha-2-polymer globulins, are used to increase the uptake of shell encapsulated particles of substantially water-insoluble pharmacologically active agents by macrophage cells, or to liver and spleen. The absorption of shell encapsulated particles may be increased. Specific antibodies can also be used to target nanoparticles to specific sites. In addition, other functional proteins such as antibodies or enzymes may be used as components of the stabilizer, which may facilitate the targeting of the biomass to the desired site.
Similarly, synthetic polymers are also good candidates for the formation of particles containing polymer shells. In addition, polyalkylene glycols (e.g., straight or branched chains), polyvinyl alcohols, polyacrylates, polyhydroxyethyl methacrylates, polyacrylic acids, polyethyloxazolines, polyacrylamides, polyisopropyl acrylamides , Polyvinyl pyrrolidinone, polylactide / glycolide and the like and combinations thereof are good candidates for biocompatible polymers of the formulations of the present invention.
Similarly, synthetic polypeptides are also good candidates for stabilizers for substantially water-insoluble pharmacologically active agents. Also contemplated for use in the practice of the present invention are synthetic polyamino acids containing cysteine residues and / or disulfide groups, polyvinyl alcohols modified to contain free sulfhydryl groups and / or disulfide groups, glass Polyhydroxyethyl methacrylate modified to contain sulfhydryl and / or disulfide groups, polyacrylic acid, free sulfhydryl groups and / or modified to contain free sulfhydryl and / or disulfide groups ) Polyethyloxazoline modified to contain disulfide groups, free sulfhydryl groups and / or polyacrylamide modified to contain disulfide groups, free sulfhydryl groups and / or polysulfide modified to contain disulfide groups Polyalkylene glycols, polylactides, polyglycols modified to contain vinyl pyrrolidinone, free sulfhydryl and / or disulfide groups There may be mentioned the call leads, polycaprolactone, or free sulfhydryl group, and (or) disulfide-modified to contain these groups copolymers and their mixture of two or more.
In the preparation of the compositions of the present invention, a wide variety of organic media can be used to suspend or dissolve substantially water-insoluble pharmacologically active agents. Organic media contemplated for use in the practice of the present invention include any non-aqueous liquid capable of suspending or dissolving pharmacologically active agents that have not been chemically reacted with the polymer or pharmacologically active agent itself used to prepare the shell. do. Examples include vegetable oils (e.g., soybean oil, olive oil, etc.), coconut oil, safflower oil, cottonseed oil, sesame seed oil, orange oil, limonene oil, aliphatic, alicyclic or aromatic hydrocarbons having 4 to 30 carbon atoms (e.g. For example, n-dodecane, n-decane, n-hexane, cyclohexane, toluene, benzene, etc.), aliphatic or aromatic alcohols having 2 to 30 carbon atoms (for example, octanol, etc.), 2 carbon atoms Aliphatic or aromatic esters of 30 to 30 (eg, ethyl caprylate (octanoate), etc.), alkyl, aryl or cyclic ethers of 2 to 30 carbon atoms (eg, diethyl ether, tetrahydrofuran) Etc.), alkyl or aryl halides having 1 to 30 carbon atoms (and optionally having one or more halogen substituents) (e.g., CH 3 Cl, CH 2 Cl 2 , CH 2 Cl-CH 2 Cl, etc.), carbon Ketones having 3 to 30 atoms (eg, acetone, methyl ethyl ketone, etc.), polyal Or ethylene glycol (eg, polyethylene glycol, etc.) or a combination of two or more thereof.
Combinations of particularly preferred organic media contemplated for use in the practice of the present invention typically have a boiling point of about 200 ° C. or less and, together with high molecular weight (less volatile) organic media, volatile liquids such as dichloromethane, chloroform, ethyl acetate , Benzene, ethanol, butanol, butyl acetate and the like (ie, solvents soluble in other organic media used with high solubility for pharmacologically active agents). When added to other organic media, these volatile additives assist in dissolving the pharmacologically active agent in the organic medium. This is preferable because this step usually consumes time. Following dissolution, the volatile components can be removed by evaporation (optionally under vacuum).
Particles of pharmacologically active agent comprising a polymer shell prepared as described above are delivered as a suspension in a biocompatible aqueous liquid. The liquid can be selected from water, saline, solutions containing appropriate buffers, solutions containing nutrients such as amino acids, sugars, proteins, carbohydrates, vitamins or fats.
In addition, these biocompatible materials can be used in some physical form, such as (crosslinked or uncrosslinked) gels, to provide a matrix from which pharmacologically active ingredients, such as paclitaxel, can be released by diffusion and / or degradation of the matrix. Can be. In addition, temperature sensitive materials can be used as the dispersion matrix for the formulations of the invention. Thus, for example, capsol® is a temperature sensitive substance (eg, a copolymer of polyacrylamide or polyalkylene glycol and polylactide / glycolide) that gels at the tumor site and provides a sustained release capsol Copolymer)) into the liquid formulation. The capsol formulation is dispersed in a matrix of the biocompatible polymer to provide a controlled release formulation of paclitaxel, which, through the properties of the capsol formulation (albumin bound to paclitaxel), lowers systemic toxicity as follows, Lower toxicity Other chemotherapeutic agents or combinations of capsols formulated similarly to capsols with biocompatible polymer matrices may be useful for regulating local delivery of chemotherapeutic agents to treat solid tumors in the brain and peritoneum and for topical application to other solid tumors. have. These formulations are not limited to the use of paclitaxel and can be used with various pharmacologically active ingredients including anti-infectives, immunosuppressants, other chemotherapeutic agents, and the like.
Colloidal particles substantially completely contained or bound within the polymer stabilization layer, prepared as described herein, may be delivered in pure form or, optionally, in suspension in a biocompatible medium. This medium comprises water, buffered aqueous medium, saline, buffered saline, optionally buffered solutions of amino acids, optionally buffered solutions of proteins, optionally buffered solutions of sugars, optionally buffered solutions of carbohydrates, optionally buffered solutions of vitamins, Optionally buffered solutions of synthetic polymers, lipid containing emulsions and the like.
In addition, the colloidal particles may optionally be modified with a suitable formulation, wherein the formulation is bound to the polymer layer via any covalent bond. Covalent bonds contemplated for such bonding include esters, ethers, urethanes, diesters, amides, secondary or tertiary amines, phosphate esters, sulfate esters and the like. Suitable formulations contemplated for any modification of the polymer shell include synthetic polymers (polyalkylene glycols such as straight or branched polyethylene glycols), polyvinyl alcohol, polyhydroxyethyl methacrylate, polyacrylic acid, polyethyloxazoline , Polyacrylamide, polyvinyl pyrrolidinone, etc.), phospholipids (e.g., phosphatidyl choline (PC), phosphatidyl ethanolamine (PE), phosphatidyl inositol (PI), spingomyelin, etc.), proteins (e.g., Enzymes, antibodies, etc.), polysaccharides (eg, starch, cellulose, dextran, alginate, chitosan, pectin, hyaluronic acid, etc.), chemical modifiers (eg, pyridoxal 5'-phosphate, pyridoxal Derivatives, dialdehydes, diaspirin esters, etc.) or combinations of two or more thereof.
Modifications to the general theme of stabilized colloidal particles are possible. A suspension of fine particles of the drug in the biocompatible dispersant may be used to prepare a polymer shell containing particles suspended in the biomaterial's dispersant (instead of the biocompatible dispersant containing the dissolved biomass). That is, the polymer shell may contain a saturated solution of biomass in the dispersant. Another variant is first by dissolving the biomass in a volatile organic solvent (eg benzene) to form a polymer shell and evaporating the volatile solvent under vacuum, for example by an evaporator, spray dryer or by freeze drying the entire suspension. It is a polymer shell containing the biosolid solid core produced. This produces a structure containing biosolid solid cores surrounded by a polymer coating. This method is particularly advantageous for delivering high doses of biomass in relatively small volumes. In some cases, the biocompatible material that forms the shell for cores may itself be a therapeutic or diagnostic agent, for example in the case of insulin, which may be delivered as part of the polymer shell formed by the methods described above. In other cases, the polymer forming the shell may participate in the delivery of the biomass, for example in the case of antibodies used for targeting or in the case of hemoglobin, which is delivered as part of the polymer shell formed by the ultrasonic irradiation described above and with oxygen. It is possible to provide blood replacement with high binding force.
Those skilled in the art will appreciate that several variations are possible within the scope and spirit of one aspect of the invention. The organic medium in the polymer shell can be modified, various pharmacologically active agents can be used, and a wide range of proteins and other natural and synthetic polymers can be used to form the walls of the polymer shell. It is also equally widely applied. In addition to the delivery of other biopharmaceutical applications such as drugs, diagnostics (in imaging applications), artificial blood and parenteral nutritional agents, the polymer shell structures of the present invention can be applied to cosmetics, perfume products and pressure sensitive materials such as skin creams or hair protectants. It may be incorporated in the ink or the like.
One aspect of the invention will be described in more detail with reference to the following non-limiting examples.
<Example 1>
<Production of Nanoparticles by High Pressure Homogenization>
30 mg of paclitaxel was dissolved in 3.0 ml of methylene chloride. The solution was added to 27.0 ml of human serum albumin solution (1% (w / v)). The mixture was homogenized at low RPM (Vitris Homogenizer, Model: Tempest I.Q.) for 5 minutes to form a crude emulsion, which was then transferred to a high pressure homogenizer (Avestin). The emulsion was emulsified at 9,000-40,000 psi with recycling of the emulsion for at least 5 times. The resulting system was transferred to a rotary evaporator and the methylene chloride was quickly removed at 40 ° C. for 20-30 minutes under reduced pressure (30 mmHg).
The dispersion obtained was translucent and the usual diameter of the paclitaxel particles obtained was 160-220 nm (Z-mean, Malvern Zetasizer).
The dispersion was also lyophilized for 48 hours without adding any cryoprotectant. The resulting cake could easily be reconstituted into the original dispersion by addition of sterile water or saline. The particle size after recomposition was the same as before lyophilization.
<Example 2>
Large crystals were formed when using conventional surfactants and proteins
The following examples demonstrate the effect of adding surfactants used in conventional solvent evaporation methods. A series of experiments were performed using a procedure similar to that described in Example 1, except that a surfactant such as Tween 80 (1-10%) was added to the organic solvent. After removal of methylene chloride, it was found that a number of paclitaxel crystals having an average size of 1 to 2 µm were obtained when observed by light microscopy and polarization. The crystals grew in a few hours to form very large needle-like crystals of about 5 to 15 μm in size. Similar phenomena were observed with other commonly used surfactants such as Pluronic F-68, Pluronic F-127, Cremaphor EL and Brij 58.
From these results, conventional solvent evaporation methods using conventional surfactants in combination with proteins such as albumin have been found that drug particles (eg, paclitaxel) up to μm without polymer cores using polar solvents (eg methylene chloride). It can be seen that it is not suitable for the formation of).
<Example 3>
Large crystals were formed when conventional surfactants were used alone.
This example has demonstrated that nanoparticles cannot be formed when using conventional surfactants without polymeric core materials, with pharmacologically active agents soluble in polar water immiscible solvents (eg chloroform).
30 mg of paclitaxel was dissolved in 0.55 ml of chloroform and 0.05 ml of ethanol. The solution was added to 29.4 mL of Tween 80 solution (1% (w / v)) presaturated with 1% chloroform. The mixture was homogenized for 6 minutes at low RPM (Vitris homogenizer, model: Tempest I.Q.) to form a crude emulsion, which was then transferred to a high pressure homogenizer (Avestin). The emulsion was emulsified at 9,000-40,000 psi with recycling of the emulsion for at least 5 times.
The resulting system was transferred to a rotary evaporator and chloroform was quickly removed at 40 ° C. for 15-30 minutes under reduced pressure (30 mmHg). The resulting dispersion was opaque and contained drugs of large needle crystals. The initial size of the crystals was 0.7-5 μm. Storing the dispersion for several hours at room temperature further increased the crystal size, eventually forming a precipitate.
<Example 4>
Preparation of Sterile Filterable Nanoparticles <200 nm
This example describes how to obtain sterile filterable drug particles. 30 mg of paclitaxel was dissolved in 0.55 ml of chloroform and 0.05 ml of ethanol. The solution was added to 29.4 mL of human serum albumin solution (1% (w / v)) previously saturated with 1% chloroform. The mixture was homogenized at low RPM (Vitris Homogenizer, Model: Tempest I.Q.) for 5 minutes to form a crude emulsion, which was then transferred to a high pressure homogenizer (Avestin). The emulsion was emulsified at 9,000-40,000 psi with recycling of the emulsion for at least six times. The resulting system was transferred to a rotary evaporator and chloroform was quickly removed at 40 ° C. for 15-30 minutes under reduced pressure (30 mmHg). The dispersion obtained was translucent and the usual diameter of the obtained particles was 140-160 nm (Z-mean, Malvern Zetasizer). The dispersion was filtered through a 0.22 μm filter (Millipore) without causing any significant change in turbidity or particle size.
HPLC analysis of the paclitaxel content showed that more than 97% of paclitaxel was recovered after filtration and thus a sterile paclitaxel dispersion was obtained.
In addition, the sterile dispersion was lyophilized for 48 hours without the addition of any cryoprotectant. The resulting cake could easily be reconstituted into the original dispersion by addition of sterile water or saline. The particle size after recomposition was the same as before lyophilization.
Example 5
Preparation of Sterile Filterable Nanoparticles <200 nm
This example describes how to obtain sterile filterable drug particles. 225 mg of paclitaxel was dissolved in 2.7 ml of chloroform and 0.3 ml of ethanol. The solution was added to 97 ml human serum albumin solution (3% (w / v)). The mixture was homogenized at low RPM (Vitris Homogenizer, Model: Tempest I.Q.) for 5 minutes to form a crude emulsion, which was then transferred to a high pressure homogenizer (Avestin). The emulsion was emulsified at 9,000-40,000 psi with recycling of the emulsion for at least six times. The resulting system was transferred to a rotary evaporator and chloroform was quickly removed at 40 ° C. for 15-30 minutes under reduced pressure (30 mmHg). The dispersion obtained was translucent and the usual diameter of the obtained particles was 140-160 nm (Z-mean, Malvern Zetasizer). The dispersion was filtered through a 0.22 μm filter (Sartorius, sartobran 300) without causing any significant change in turbidity or particle size. HPLC analysis of the paclitaxel content showed that after filtration, 70-100% of paclitaxel was recovered depending on the conditions used. Thus, a sterile paclitaxel dispersion was obtained.
Sterile dispersions were aseptically filled with sterile glass vials and lyophilized without the addition of any lyoprotectant. The resulting cake could easily be reconstituted into the original dispersion by addition of sterile water or saline. The particle size after recomposition was the same as before lyophilization.
<Example 6>
Influence of Phase Fraction of Organic Solvents on Particle Size
The following examples demonstrate the importance of having a very low phase fraction of organic solvents in the system.
A series of experiments were performed following a procedure similar to that described in Example 4 except that the phase fraction of the organic solvent was changed and the ethanol content was maintained at 10% (v / v) in the organic phase. It was found that increasing the phase fraction significantly increased the particle size. That is, particles obtained at 4% (v / v) phase fractions (above 5% v / v total chloroform concentration or saturation concentration) had a diameter of 250 nm, and at 3% (v / v) phase fractions the particles had a diameter of 200 nm, and the particle diameter was 150 nm in the 2% (v / v) phase fraction.
Clearly, only particles prepared in very low phase fractions could be sterile filtered.
<Example 7>
Effect of Drug Concentration on Particle Size
The role of drug concentration in the organic phase is demonstrated in the examples below. In both experiments the paclitaxel concentration in the organic phase was 50 mg / ml or 75 mg / ml and all other variables were the same as described in Example 2. Low drug concentrations resulted in particles of about 150 nm in diameter, while particles produced at high drug concentrations were found to be smaller, 130-138 nm in diameter. Similar trends were observed when similar experiments were performed with an ethanol concentration of about 50% in the organic phase. That is, when the drug concentration was 25 mg / ml and 50 mg / ml, the diameters of the particles were 210 nm and 156 nm, respectively.
These findings were in contrast to those reported in the literature on the formation of nanoparticles in the presence of surfactants (Sjostrom et al., Supra).
<Example 8>
Formation of Nanoparticles in Model Drugs
30 mg of isopresserpin (model drug) was dissolved in 3.0 ml of methylene chloride. The solution was added to 27.0 ml of human serum albumin solution (1% (w / v)). The mixture was homogenized at low RPM (Vitris Homogenizer, Model: Tempest I.Q.) for 5 minutes to form a crude emulsion, which was then transferred to a high pressure homogenizer (Avestin). The emulsion was emulsified at 9,000-18,000 psi with recycling of the emulsion for at least 5 times. The resulting system was transferred to a rotary evaporator and the methylene chloride was quickly removed at 40 ° C. for 20-30 minutes under reduced pressure (30 mmHg). The dispersion obtained was translucent and the usual diameter of the obtained particles was 120-140 nm (Z-average, Malvern Zetasizer). The dispersion was filtered through 0.22 μm pores.
In addition, the sterile dispersion was lyophilized for 48 hours without the addition of any cryoprotectant. The resulting cake could easily be reconstituted into the original dispersion by addition of sterile water or saline. The particle size after recomposition was the same as before lyophilization.
Example 9
Very small particle formation of model drug
The effect of ethanol addition on reducing particle size is demonstrated for isopresserpin. 30 mg of isopresserpin were dissolved in 2.7 ml of methylene chloride and 0.3 ml of ethanol. The solution was added to 27.0 ml of human serum albumin solution (1% (w / v)). The mixture was homogenized at low RPM (Vitris Homogenizer, Model: Tempest I.Q.) for 5 minutes to form a crude emulsion, which was then transferred to a high pressure homogenizer (Avestin). The emulsion was emulsified at 9,000-40,000 psi with recycling of the emulsion for at least 5 times. The resulting system was transferred to a rotary evaporator and the methylene chloride was quickly removed at 40 ° C. for 20-30 minutes under reduced pressure (30 mmHg). The dispersion obtained was translucent and the usual diameter of the obtained particles was 90-110 nm (Z-mean, Malvern Zetasizer). The dispersion was filtered through a 0.22 μm filter (Millipore).
In addition, the sterile dispersion was lyophilized for 48 hours without the addition of any cryoprotectant. The resulting cake could easily be reconstituted into the original dispersion by addition of sterile water or saline. The particle size after recomposition was the same as before lyophilization.
<Example 10>
Use of a water-miscible solvent, supersaturated with a drug, not suitable for the method of the present invention>
30 mg of paclitaxel was dispersed in 0.6 ml of ethanol. At this concentration (50 mg / ml), paclitaxel did not dissolve completely, forming a supersaturated dispersion. This dispersion was added to 29.4 mL of human serum albumin solution (1% (w / v)). The mixture was homogenized at low RPM (Vitris Homogenizer, Model: Tempest I.Q.) for 5 minutes to form a crude dispersion, which was then transferred to a high pressure homogenizer (Avestin). The emulsion was emulsified at 9,000 to 40,000 psi with recycling for at least six times. The resulting system was transferred to a rotary evaporator and ethanol was quickly removed at 40 ° C. for 15-30 minutes under reduced pressure (30 mm Hg). The dispersion particle size obtained was very wide ranging from about 250 nm to several microns.
Microscopy showed large particles of paclitaxel and typical needle-like crystals. These particles were so large that intravenous injection was impossible. The above experiments demonstrate that the use of a solvent such as ethanol, which is freely miscible with water in the process of the present invention, results in the formation of large particles with a very large particle size distribution and cannot be used in the process of the present invention alone. . Thus, the method of the present invention excludes water miscible solvents from use when solvents are used alone to dissolve or disperse the drug components. The method of the present invention requires mixing the solvent with the essentially water immiscible solvent which produces the nanoparticles of the present invention in use.
<Example 11>
<Use of a water miscible solvent containing a dissolved drug alone, which is not suitable for the method of the present invention>
30 mg of paclitaxel was dispersed in 1.3 ml of ethanol. At this concentration (about 24.5 mg / ml), paclitaxel was completely dissolved in ethanol. This solution was added to 28.7 mL of human serum albumin solution (1% (w / v)). The mixture was homogenized at low RPM (Vitris homogenizer, model: Tempest I.Q.) for 5 minutes to form a crude dispersion, which was then transferred to a high pressure homogenizer (Avestin). The emulsion was emulsified at 9,000 to 18,000 psi with recycling for at least six times. The resulting system was introduced into a rotary evaporator and ethanol was quickly removed at 40 ° C. for 15-30 minutes under reduced pressure (30 mm Hg). The resulting dispersion particle size was very wide ranging from about 250 nm to several microns. Microscopy showed large particles of paclitaxel and typical needle-like crystals. These particles were so large that intravenous injection was impossible.
In addition to the above Example 10, this example uses a solvent such as ethanol that can be freely miscible with water in the method of the present invention, so that large particles having a very large particle size distribution are formed, and are thus used alone in the method of the present invention. Demonstrates that it cannot be used. Thus, the method of the present invention excludes water miscible solvents from use when solvents are used alone to dissolve or disperse the drug components. The method of the present invention requires mixing the solvent with the essentially water immiscible solvent which produces the nanoparticles of the present invention in use.
<Example 12>
Measurement of the physical state of paclitaxel in nanoparticle form by X-ray powder diffraction
In general, paclitaxel raw materials are present in acicular crystals of various sizes, typically 5 to 500 μm. If the size of the crystals exceeds several micrometers, it is clearly harmful that the crystals are present in the intravenous drug formulation because of the potential for clogging capillaries. In addition, since the solubility of drug crystals is generally lower than amorphous drugs, it lowers the bioavailability of drugs after intravenous administration. It is also known that the tendency of crystallization also increases as the loading of the drug in the formulation increases. Thus, it is advantageous for the formulation to contain the drug in essentially amorphous form.
X-ray powder diffraction was used to determine the crystalline or amorphous nature of paclitaxel in the lyophilized powder formulation. Sample 1-paclitaxel powder; Sample 2-lyophilized serum albumin; Physical mixture of sample 3-paclitaxel and albumin; And sample 4-samples of formulated paclitaxel. Each sample was x-rayed at 2 ° to 70 ° 2-theta angle using CuKa radiation, an acceleration voltage of 40 KeV / 30 mA, a step size of 0.05 ° 2-theta and a data acquisition time of 2.0 seconds per step. . Sample 1 showed a strong strong peak typical of crystalline samples. The strongest paclitaxel peak was at 5.1 ° 2-Theta. Sample 2 showed a typical wide hump of amorphous material. Sample 3 mainly exhibited the wide hump of Sample 2, but in addition, a peak at 5.1 ° 2-theta of paclitaxel was seen. Sample 4, formulated paclitaxel, showed no basis for crystallinity of paclitaxel and looked the same as sample 2, indicating that there was a substantially amorphous pharmacologically active agent in the formulated sample.
The amorphousness of nanoparticles prepared according to the present invention is directly different from the products produced by other methods conventionally used for nanoparticle preparation. For example, the use of the grinding techniques described in US Pat. No. 5,145,684 to Liversidge et al. And Liversidge-Merisko et al., Pharmaceutical Research 13 (2): 272-278 (1996) substantially produces crystalline products.
Example 13
Preparation of Cyclosporine Nanoparticles (Intravenous Capsulin) by High Pressure Homogenization
30 mg of cyclosporin was dissolved in 3.0 ml of methylene chloride. This solution was then added to 27.0 ml of human serum albumin solution (1% w / v). This mixture was homogenized at low RPM (Bitless Homogenizer Model: Tempest I.Q.) for 5 minutes to form a crude emulsion, which was then transferred to a high pressure homogenizer (Avestin). Emulsification was carried out at 9000 to 40,000 psi while recycling the emulsion at least five times. The resulting system was transferred to a rotary evaporator to quickly remove methylene chloride for 20-30 minutes at 40 ° C. under reduced pressure (30 mmHg). The resulting dispersion was translucent and the typical diameter of the resulting particles was 160-220 nm (Z-mean, Malvern Zetasizer).
This dispersion was further lyophilized for 48 hours with no cryoprotectant added. The resulting cake could easily be reconstituted into the original dispersion with the addition of sterile water or brine. The particle size after reconstitution was the same as before lyophilization.
<Example 14>
Preparation of Cyclosporine Idol Droplets (Oral Capsulin) by High Pressure Homogenization
30 mg of cyclosporin was dissolved in 3.0 ml of a suitable oil (sesame oil containing 10% orange oil). The solution was then added to 27.0 ml of human serum albumin solution (1% w / v). This mixture was homogenized at low RPM (Bitless Homogenizer Model: Tempest I.Q.) for 5 minutes to form a crude emulsion, which was then transferred to a high pressure homogenizer (Avestin). Emulsification was carried out at 9000 to 40,000 psi while recycling the emulsion at least five times. The resulting dispersion had a typical diameter (Z-average, Malvern Zetasizer) of 160-220 nm.
This dispersion was lyophilized for 48 hours either directly or optionally by adding the appropriate cryoprotectant. The resulting cake could easily be reconstituted into the original dispersion with the addition of sterile water or brine.
<Example 15>
Inhalation preparations of anti-asthma
Anti-asthma drugs were prepared using microparticle technology to make effective formulations for dry powder inhalants (DPI). Dry preparations with suitable particle size and release properties for effective delivery to the respiratory system starting from steroidal drugs (e.g. beclomethaneson, beclomethasone dipropionate, budesonide, dexamethasone, flunisolidide, triamcinolone acetonide, etc.) Was prepared.
This formulation was prepared using a sonic grinding technique or a homogenization method in which an active drug dissolved in a solvent was dispersed in an aqueous protein solution to form an emulsion of nanoparticles. This emulsion was then evaporated to remove the solvent and the active drug applied with the protein remained in solution. Liquid samples containing colloidal drug particles were measured by Malvern Zetasizer and had a Z-average size of 260 nm. In a preferred embodiment, the size range of such colloidal particles is about 20 to 1,000 nm, more preferably about 70 to 400 nm.
Other excipients can also be dissolved in this liquid. Such excipients include, but are not limited to, 0.5-15% mannitol, 0.1-5% lactose and maltodextrin. In this step, the resulting solution of the active drug, protein and excipient can be spray dried or lyophilized and ground to obtain a dry powder. After spray drying, the size of the dry particles is measured by Malvern mastersizer to a D (v. 0.5) of about 1 to 10 μm. The preferred size range of the particles is 0.5-15 μm, more preferably 0.7-8 μm.
This spray dried powder was then mixed with excipient carrier powder. In addition, several carriers may be used including lactose, trehalose, Pharmatose 325 M, sucrose, mannitol, and the like. The size of the carrier powder is considerably larger than the formulated drug particles (lactose is about 63-90 μm, and pharmatos are 40-100 μm).
The efficacy of dry powder formulations is demonstrated by testing with an Andersen 8 stage continuous paddle. The impact test results showed that the fine particle fraction (FPF) was about 60%. This suggests that particles properly tuned to the respiratory material are released very effectively. This FPF is quite large because of formulation compositions containing colloidal nanoparticles of the drug in larger formulation particles.
Such formulations indicate that microparticle and spray-drying techniques are suitable for processing and adjusting dry powder formulations for aerosol delivery via DPI.
<Example 16>
Overview of Current Preferred Manufacturing Processes: Starting with 1 Gram of Paclitaxel as BDS
A 3% HSA solution was prepared. To 51.7 ml of 25% albutane was added 379.3 ml of infusion water. Mix thoroughly and filter the solution through a sterile 0.22 μm Nalgen disposable filter ware. Store at 4 ° C. until use.
1.0 g of paclitaxel in the glass bottle was weighed. The vials were mixed well by mixing CHCl 3 and ethyl alcohol in the appropriate fractions. 13.33 ml of chloroform / ethyl alcohol mixture were added to paclitaxel. The paclitaxel was stirred to dissolve all in solution. This solution was filtered through a 0.22 μm sterile Teflon filter and collected in sterile glass bottles.
HSA solution was added to the dissolved paclitaxel solution in the vial. Paclitaxel / HSA solution was mixed using a Centri Microprocessor mixer. When mixing this solution, the components were poured into the chamber of the homogenizer. The mixture was circulated under pressure to a homogenizer until the desired particle size was obtained. Homogenized samples in Kontes round bottom flasks were collected.
The flask with the final sample was fixed on a rotary evaporator. The organic solvent was evaporated by applying vacuum and rotating the rotary evaporator to the maximum. As a result, a colloidal solution of paclitaxel in human albumin was obtained. Approximately 3 ml of rotary evaporated sample was placed for particle size analysis.
Under a sterile hood, the colloidal solution was filtered using a sterile 0.45 / 0.2 μm filter and collected in a sterile recovery container. About 3 ml of the filtered sample was placed for analysis by HPLC for paclitaxel concentration.
Fill volume was measured to obtain 30 mg (or other amount) of paclitaxel per vial. Sterile filtered samples were filled with about 17 ml each of 30 ml vials of autoclaved Heaton (based on analysis). The vial was closed with autoclaved hyton serum vial stopper. Each vial should contain about 30 mg of paclitaxel.
Samples were lyophilized using the predetermined lyophilization cycle in an FTS system stopper tray lyophilizer. After the sample was lyophilized, the vial was closed and pleated with 20 mm Hiton aluminum cut caps to seal the vial. Samples were marked properly. All procedures were performed in a clean indoor environment under sterile conditions.
Lyophilized samples contain less than 1000 ppm, more preferably less than 500 ppm, or even less than 100 ppm residual solvent.
Final product sterilization filtration: Following removal of the solvent by evaporation, the colloidal solution of paclitaxel in the flask was filtered through a 0.45 / 0.2 μm combination sterilization filter. The filtrate solution was collected in a sterile beaker and filled into 30 ml vials. The vial was then placed in the lyophilizer. Following completion of the lyophilization cycle, the vial is filled with dry sterile nitrogen gas and capped under nitrogen.
It is noteworthy to use a high pressure homogenization process to catch, kill and remove bacteria and other cells.
<Example 17>
Formation of Nanoparticles Using Acoustic Wave Crushing
Preparation of Oil-Containing Protein Shells
Similar to using high shear homogenization, it is believed that the formation of protein-coated nanoparticles of water-insoluble pharmaceutically active agents using sonic disruption is accomplished by crosslinking proteins through the formation of intermolecular disulfide bonds. The various advantages of the prior art by the high shear uniformity technique described above apply equally to the sonic wave grinding method described below.
With regard to organic solvents, proteins and nano-protein polymers that can be used in sonic grinding, reference is made to the components described above for high shear homogenization. The same components are expected to work the same in both methods.
This feature of the invention will now be described in more detail with reference to the following non-limiting examples.
3 ml of USP (United Stated Pharmacopia) 5% human serum albumin solution (from Alpha Therapeutic Corporation) was placed in a cylindrical container that could be fixed to a sonicating probe (heating system, model XL2020). The albumin solution was covered with 6.5 ml USP grade soybean oil (soy milk). The tip of the sonicator probe was placed at the interface between the two solutions and the assembly was maintained in a 20 ° C. cold bath. The system was equilibrated and the sonicator was run for 30 seconds. A vigorous mixing took place and a white milky suspension was obtained. The suspension was diluted 1: 5 with normal brine. The particle counter (Particle Data System, Elzone, Model 280 PC) was used to measure particle size distribution and concentration of oil containing protein shells. The resulting protein shell was determined to have a maximum cross-sectional area of about 1.35 ± 0.73 μm and the total concentration was measured to about 10 9 shells per ml of the original suspension.
As a control, these components without protein did not form stable microemulsions upon ultrasound irradiation. This result suggests that proteins are essential for the formation of centrosomes. This was confirmed by the scanning electron microscopy and transmission electron microscopy studies described below.
Example 18
Preparation of a Polymerizable Shell Containing Dissolved Paclitaxel
Paclitaxel was dissolved at a concentration of 2 mg / ml in USP grade soybean oil. 3 ml of USP 5% human serum albumin solution was taken in a cylindrical container that could be fixed to the sonicating probe. The albumin solution was covered with 6.5 ml soybean oil / paclitaxel solution. The sonicator probe tip was placed at the interface of the two solutions, the assembly was in equilibrium and the sonicator was run for 30 seconds. Vigorous mixing occurred and a stable white milky suspension was obtained, containing a protein-coated polymerizable shell wrapped in an oil / paclitaxel solution.
To load more drug into the crosslinked protein shell, the amphoteric solvents for the oil and the drug (the drug's solubility is significantly higher) can be mixed with the oil. If this solvent is relatively non-toxic (eg ethyl acetate), it can be injected with the original carrier. In other cases, the liquid may be removed by evaporation under vacuum following the preparation of the polymerizable shell.
It is known that several different methods can be used to obtain the physical properties of the formulations of the invention. Biologic characteristics associated with agents with high local concentrations and low toxicity (increased LD50, reduced spinal cord suppression, reduced brain toxicity) at specific organ sites (prostate, lung, pancreas, bone, kidney, heart), which are associated with high potency Is independent of the method of preparation.
Example 19
Preparation of Nanoparticles by Acoustic Wave Grinding
20 mg of paclitaxel was dissolved in 1.0 ml of methylene chloride. This solution was added to 4.0 ml of human serum albumin solution (5% w / v). This mixture was homogenized at low RPM (Bitless Homogenizer, Model: Tempest I.Q.) for 5 minutes to form a crude emulsion which was then transferred to a 40 kHz sonicator cell. The sonicator was performed at a power of 60 to 90% at grade 0 for 1 minute (550 Sonic Dosmembrator). The mixture was transferred to a rotary evaporator and methylene chloride was rapidly removed at 40 ° C. for 20-30 minutes under reduced pressure (30 mmHg). Typical diameters of the resulting paclitaxel particles were 350-420 nm (Z-means, Balman zetasizer).
This dispersion was further lyophilized for 48 hours with no cryoprotectant added. The resulting cake could easily be reconstituted into the original dispersion with the addition of sterile water or brine. The particle size after reconstitution was the same as before lyophilization.
Example 20
In vivo biodistribution of crosslinked protein shells containing fluorophores
To measure the absorption and biodistribution of the liquid trapped in the protein polymerizable shell after intravenous injection, fluorescent dyes (obtained from Rudren, Aldrich) were trapped in human serum albumin (HSA) protein polymerizable shells and It was used as a labeling substance. Thus, rubrene was dissolved in toluene and an albumin shell containing toluene / rubrene was prepared by ultrasonic irradiation as described above. The resulting milky suspension was diluted five times with normal saline. Subsequently, 2 ml of the diluted suspension was injected into the tail vein of the rat over 10 minutes. One animal was sacrificed 1 hour after the injection and 24 hours after the injection.
100 μm of frozen portions of lung, liver, kidney, spleen, and bone marrow were tested by fluorescence microscopy for the presence of fluorescent or released dye trapped in the polymerizable shell. After 1 hour, most of the polymerizable shells appeared to be intact (ie, fluorescent particles of about 1 μm diameter appeared bright) and were located in the lungs and liver. After 24 hours, dyes were observed in the liver, lungs, spleen and bone marrow. General staining of the tissue was also observed, indicating that the shell wall of the polymerizable shell was digested and the dye was released therein. This result is in line with expectations and demonstrates the possibility of using the compositions of the present invention for delayed or controlled release of trapped drugs such as paclitaxel.
Example 21
Toxicity of Polymerizable Shells Containing Soybean Oil (SBO)
A polymerizable shell containing soybean oil was prepared as described in Example 15. The resulting suspension was diluted with normal brine to prepare two different solutions, one containing 20% SBO and the other containing 30% SBO.
Intralipid, a commercial TPN agent, contains 20% SBO. The LD 50 for intralipid in mice when injected at 1 cc / min is 4 ml, or 120 ml / kg, for 30 g of mouse.
Two groups of mice (each group having three mice; each mouse about 30 g) were treated with a composition of the present invention containing SBO as follows. Each mouse was injected with 4 ml of the preparation suspension of SBO-containing polymerizable shell. Each group of mice received a suspension containing 20% SBO and each group of mice received a suspension containing 30% SBO.
All three mice in the group injected with a suspension containing 20% SBO withstood this treatment, and no severe toxicity was observed in any tissues or organs when observed one week after SBO treatment. Only one of the three mice in the group injected with a suspension containing 30% SBO died after injection. This result clearly demonstrates that the oil contained in the polymerizable shell according to the invention is not toxic at LD 50 doses compared to commercially available SBO preparations (Intralipids). This effect may be due to the delayed release of oil from the polymerizable shell (ie, used in vivo at a controlled rate). This delayed release can prevent the oil lethality from being reached, as opposed to reaching a high oil dose by commercially available emulsions.
<Example 22>
In vivo bioavailability of soybean oil released from the polymerizable shell
Tests were performed to determine the slow release or delayed release of the material surrounding the polymeric shell after injecting a suspension of the polymeric shell into the bloodstream of the rat. Polymerizable shells containing cross-linked protein (albumin) containing soybean oil (SBO) were prepared by the sonic grinding method described above. The resulting suspension of the polymerizable shell containing oil was diluted in brine to obtain a final suspension containing 20% oil. 5 ml of this suspension was injected into the external jugular vein into which the rat cannula was inserted over 10 minutes. After injection, blood was collected from this rat several times, and the amount of triglyceride in blood (soybean oil is triglyceride as a main component) was measured by a conventional assay.
5 ml of a commercial fat emulsion (intralipid, 20% soybean oil, 1.2% egg yolk phospholipid, and an aqueous parenteral nutrition containing 2.25% glycerin) was used as a control. The control stabilized the emulsion using the egg's phospholipid as an emulsifier. The comparison of serum amounts of triglycerides in both cases will directly compare the bioavailability of the oil as a function of time. In addition to a suspension of the polymerizable shell containing 20% oil, 5 ml of a sample of the polymerizable shell containing oil in brine was also injected at a final oil concentration of 30%. Two animals from each of the three groups were used. In each case, the amount of triglyceride in the blood is summarized in Table 1 in mg / dl.
group Serum Triglycerides (mg / dl) Pre 1 hour 4 hours 24 hours 48 hours 72 hours Intralipid Control (20% SBO) 11.4 941.9 382.9 15.0 8.8 23.8 Polymerizable Shell (20% SBO) 24.8 46.7 43.8 29.3 24.2 43.4 Polymerizable Shell (30% SBO) 33.4 56.1 134.5 83.2 34.3 33.9
The amount of blood before injection is shown in a column labeled 'Pre'. Clearly, very high amounts of triglycerides were observed after injection for the intralipid control. Thereafter, it was shown that it took 24 hours for the triglyceride amount to fall to the level before injection. Thus, it can be seen that the oil is immediately available for metabolism after injection.
Suspensions of polymerizable shells containing oil having the same total oil amount as intralipid (20%) showed significantly different bioavailability of triglycerides detectable in serum. The amount rises to about two times its normal level and triglycerides remain at this level for a long time, indicating that they are slowly delayed release into the bloodstream at levels very close to normal. Groups ingested with oil containing polymerizable shells with 30% oil showed very high triglyceride amounts (higher with dose) and fell to normal values within 48 hours. In addition, in this group, the amount of triglycerides in the blood does not increase significantly compared to the control group receiving the intralipids. This also indicates that the oil is slow and consistently used in the compositions of the present invention, which avoids dangerously high blood levels of the substance contained in the polymerizable shell and allows the use of acceptable amounts over time. Has Clearly, the drug delivered in the polymerizable shell of the present invention will have this same advantage.
Polymerizable shells containing soybean oil can be suspended in aqueous solutions of amino acids, essential electrolytes, vitamins, and sugars to form parenteral comprehensive nutritional agents (TPN). Such TPNs cannot be formulated into the fat emulsions currently used (eg, intralipids) due to the instability of the emulsions in the presence of electrolytes.
<Example 23>
Preparation of Polymeric Shells Covered with Proteins Containing Solid Cores of Pharmaceutically Active Agents
Another method of delivering poorly water-soluble drugs such as paclitaxel in a polymerizable shell is to prepare a shell of polymerizable material around the solid drug core. 'Protein coated drug particles can be obtained as follows. The method described in Example 16 was repeated using an organic solvent to dissolve relatively high concentrations of paclitaxel. Commonly used solvents are organic solvents such as benzene, toluene, hexane, ethyl ether, chloroform, alcohols and the like. The polymerizable shell was prepared as described in Example 15. 5 ml of the milky suspension of the polymerizable shell containing dissolved paclitaxel was diluted to 10 ml in normal saline. This suspension was placed on a rotary evaporator and the volatile organics were removed by vacuum. Microscopic examination of the resulting suspension revealed an opaque core, suggesting that all organic solvents were substantially removed and that solid paclitaxel was present. The suspension can be frozen and stored for a long time, can be used directly or later lyophilized.
Alternatively, the polymerizable shell having a core containing the organic solvent in which the drug is dissolved is lyophilized to obtain a dry crumbly powder that can be resuspended in brine (or other suitable liquid) upon use. Preferred proteins for use in forming polymerizable shells are albumin at present, but other proteins such as α-2-macroglobulin, known opsonin, may be used to increase the amount of polymerizable shell uptake by cells such as macrophages. Can be. Alternatively, molecules such as PEG can be introduced into the particles to prepare polymeric shells with increased in vivo circulation time.
<Example 24>
Formation of Nanoparticles by Spontaneous Microemulsions
It is also possible to form nanoparticles without using sonic grinding, high shear homogenization, or any other high energy technique. Thus, it is possible to form suspensions (or dry powders) of essentially pure drugs, if necessary.
Microemulsions are thermodynamically stable emulsion systems that spontaneously form when all components are contacted without the use of high shear devices or other practical stirrers. Microemulsions are not substantially opaque, ie transparent or translucent. Microemulsions have optical transparency because they include a dispersion layer with a typical droplet size of less than 1000 mm 3. Droplets in microemulsions are typically spherical, but other structures, such as long cylinders, are weak (for further explanation see, for example, Rosof, Progress in Surface and Membrane Science, 12: 405, Academic Press (1975), Friberg, Dispersion Science and Technology, 6: 317 (1985)).
As shown below, the present invention utilizes the unique properties of the microemulsion as the first step to obtain very small nanoparticles after removing the oil layer.
As described earlier, the microparticles and nanoparticles can be formed by various methods, among which solvent evaporation. This method is in principle based on the formation of a simple oil-in-water emulsion in the presence of a surfactant, but high shear forces can be used by various devices such as rotor-stator mixers, sonicators, high pressure homogenizers, colloidal mixers and the like. After forming an emulsion containing the drug and the drug dissolved in the dispersion oil droplets, the oil layer is removed by evaporation, typically at reduced pressure and elevated temperature, to form microparticles or nanoparticles of the dissolved drug and polymer. Clearly, the size of the particles depends on the emulsion droplet size, and the smaller the droplet, the smaller the resulting particles. Small emulsion droplets can only be achieved by using very high energy, even later using the most advanced high pressure homogenizers such as Microfluidizers, and achieving emulsion droplets of less than 75 nm is feasible. Not. Since the emulsion is originally an unstable system and undergoes processes such as aggregation and droplet fusion, larger particles are formed as a result of the solvent evaporation process for such emulsions.
A new method to overcome the problems associated with using solvent evaporation in conventional emulsions consists of the following steps:
a. The water insoluble drug is dissolved in a solvent having low solubility in water and having a higher water vapor pressure than water. A binder may exist in principle but the drug dissolves without additional polymeric binder.
b. The solvent is mixed with a suitable surfactant and a water soluble cosurfactant.
c. Appropriate amount of water or aqueous solution is added to the mixture so that an oil-in-water emulsion is spontaneously formed without using any high shear device. The aqueous solution may contain electrolytes, amino acids, or any other additives that may affect the formation of the microemulsion during the first preparation step.
d. The protein solution is optionally added to the microemulsion.
e. The solvent is removed by evaporation under reduced pressure to precipitate the drug in the form of very small amorphous nanoparticles having a conventional size of less than 1000 mm 3. In this step the particles are dispersed and stabilized in an aqueous medium containing surfactants, cosurfactants, and optionally protective agents such as proteins, sugars and the like. Acceptable evaporation methods include rotary evaporators, polling film evaporators, spray dryers, freeze dryers, and methods using standard evaporators commonly used in industry.
f. The nanoparticles stabilized by the protein can be obtained by optionally removing the surfactant and the co-surfactant by dialysis, ultrafiltration, adsorption and the like.
g. Following evaporation of the solvent, the liquid dispersion of the nanoparticles can be dried to obtain a powder containing the drug and optionally protein, which is then redispersed in a suitable aqueous medium such as saline, buffer, water, etc. to a life-size having a particle size of less than 1000 mm 3 A suspension that can be administered can be obtained. Acceptable methods for obtaining such powders are freeze drying, spray drying and the like. If the conversion to solid by lyophilization is carried out, lyophilization, various cryoprotectants such as mannitol, lactose, albumin, carboxymethyl cellulose, polyvinylpyrrolidone, maltodextrin and / or polyethylene glycol may be added. Can be.
Such nanoparticles can be further mixed with additional excipients or matrix-forming materials to obtain drug delivery systems with high bioavailability, controlled release properties, and protection in gastric juice. The final product may be administered to the mammal in tablets, capsules, reconstituted solutions, and the like.
The formulations of the present invention have significant advantages over previously used methods for preparing nanoparticles and microparticles and for using microemulsions or “pre-microemulsion concentrates”.
Various advantages can be realized using the method of the present invention. If the proper ingredients are selected, the microemulsion is spontaneously formed without the need for expensive equipment and energy investment. The droplet size is about one order smaller than the minimum emulsion droplets obtained by the high shear device, so very small nanoparticles can be obtained. Since microemulsions are thermodynamically stable, general problems associated with emulsion instability (eg, time dependence of the resulting particle size) will be avoided. The whole process is much simpler than conventional emulsion solvent evaporation methods and is less sensitive to several parameters. Since this process involves only simple mixing, scaling up over large production volumes is very simple compared to emulsifying with a device such as a high shear homogenizer. Since the particle size obtained by the novel method is one order smaller than the pore size of the membrane used for sterile filtration, it is very effective without problems associated with membrane clogging such as increased filtration pressure and many drug losses during the filtration process. Since no high shear force is used in the emulsification process, the temperature does not increase at all during the emulsification process and thus even the temperature sensitive drug can be processed by the novel method of the present invention. Since the drug in the liquid formulation of the present invention contains dispersed nanoparticles as compared to conventional microemulsions containing dispersed nanodroplets, that is, more chemical reactions occur in the liquid state (microdroplet) versus the solid state (nanoparticle). Therefore, the chemical stability was increased. The present invention has increased chemical stability as a dry formulation over conventional microemulsions that are liquid as continuous microemulsion layers. Solid formulations may include the drug in a variety of solid dosage forms such as tablets, granules and capsules as compared to conventional microemulsions or “pre-microemulsion concentrates” that are in liquid form. The very narrow particle size distribution with very small average particle size increases the adsorption of the drug in a more homogeneous way than the microparticles and nanoparticles prepared by conventional methods, thus increasing bioavailability.
Although the examples presented below refer to water insoluble molecules, drugs that are now considered useful for nanoparticle preparation include, but are not limited to, water soluble or water insoluble drugs, diagnostics, therapeutics, nutrients, and the like. Non-limiting listing of drug categories and compounds is not limited to all of the compounds listed above for use in the high shear homogenization aspect of the present invention.
Solvents mentioned in the examples below include toluene and butyl acetate, but any solvent or solvent mixture capable of dissolving the required drug will be suitable for use in the process of the present invention, provided that suitable microemulsions Can be formed before removal. Such solvents include chloroform, methylene chloride, ethyl acetate, butyl acetate, isobutyl acetate, propyl acetate, tert-butylmethyl ether, butanol, propylene glycol, heptane, anisole, cumene, ethyl formate ethanol, propanol, tetrahydrofuran , Dioxane, acetonitrile, acetone, dimethyl sulfoxide, dimethyl formamide, methyl pyrrolidinone, soybean oil, coconut oil, castor oil, olive oil, safflower oil, cottonseed oil, C1-C20 alcohol, C2-C20 ester, C3-C20 Ketones, polyethylene glycols, aliphatic hydrocarbons, aromatic hydrocarbons, halogenated hydrocarbons, d-limonene, combinations thereof, and the like.
The protein (or mixture of some proteins) used in this method should not precipitate during the initial mixing or during the evaporation step. There are several proteins including albumin (eg, BSA, HSA, eggs), gelatin, collagen, IgG, various enzymes, lactoglobulin, casein, soy protein and the like.
The surfactants used in the present invention should be able to spontaneously form oil-in-water microemulsions in the presence of suitable co-surfactants and solvents without causing precipitation of drugs or proteins (if any). Surfactants may be nonionic (twin, span, triton, pluronic, polyglycerol esters, etc.), anionic (SDS, cholate and dioxycholate, fatty acid soaps, etc.), cationic (cetyltrimethyl ammonium chloride, etc.) or both Warmth (lecithin, amino acids, etc.).
Cosurfactants should have the ability to spontaneously form microemulsions with selected surfactants without inducing precipitation of dissolved drug molecules (or proteins, if present) and inducing formation of large crystalline materials. Cosurfactants may be water soluble or oil soluble, such as butanol, propylene glycol, benzyl alcohol, propanol, and the like.
Conversion of the liquid dispersion of nanoparticles by lyophilization may require the addition of cryoprotectants such as mannitol, lactose, amino acids, proteins, polysaccharides and the like.
It is clear that the principles described in the present invention can be applied in several variations of the following methods.
1. Formation of drug particles may be induced by dilution of the microemulsion in a suitable solvent in which the solvent may be mixed. For example, when the solvent has low water solubility, the microemulsion can be diluted to such an extent that the solvent is less than its water solubility.
2. Solvents and optionally surfactants and cosurfactants can be removed using selective extractants that do not dissolve the drug.
3. Surfactants and cosurfactants use filters with cut-offs less than the size of the protein molecular weight and can be removed by ultrafiltration. Simple dialysis may also be chosen.
4. The formulation may contain only ingredients which are acceptable for the intended use of the final formulation (oral, intravenous, topical, etc.), so no removal thereof is necessary.
5. Similarly, cosurfactants may be used which may remain in the final product, such as glycerol, benzyl alcohol and the like.
6. It is possible to add various water-soluble molecules that can affect the state of the microemulsion (electrolyte, ethanol, etc.), so the optimum drug load is obtained by adjusting the ratio between the various components.
7. The spontaneous emulsification step can be carried out at a temperature other than room temperature in order to influence the degree of state (and the component ratios leading to the formation of microemulsions). In particular, temperature effects (in ethoxylated surfactants) can be used to change the system from oil in water to water microemulsion in oil.
8. Other ingredients may be added on the solvent to affect the bioavailability of the drug. In particular, it is desirable to add oils such as soybean oil to improve oral absorption and to prevent the drug from chemical and enzymatic degradation.
9. Similarly, addition of the matrix forming polymer (eg PVP) to the solvent can be carried out with the drug.
10. Stabilization and solid phase formation properties can be changed by adding water soluble polymers other than proteins (carboxymethyl cellulose, rubber, etc.) to the external aqueous phase of the microemulsion.
11. The flowability of the obtained solid form powder can be varied by the addition of colloidal particles (eg silica) as a filter, or by the addition of reconstitution / anti-aggregation aids.
12. The same principle described in the present invention can be applied to form water-soluble particles, and the emulsifying step is carried out in a composition range in which a water microemulsion in oil is formed. For example, the method can be used to form very few protein nanoparticles.
<Example 25>
Preparation of Nanoparticles of Cyclosporin A
115 mg of cyclosporin A was dissolved in 1 ml of butyl acetate and mixed with 2 grams of Triton X-100: n-butanol 4: 1 solution. A transparent system was obtained. 10 g of water was added dropwise with slight shaking. An oil microemulsion in clear water was obtained. 10 g of 1% casein solution was added with slight shaking. The system became somewhat cloudy. Butyl acetate was removed in Rotovap at 80 ° C. under 80 mm Hg. The system became completely transparent.
Particle size was measured by a photon correlation spectrometer. It was found that the Z mean is 25 to 33 nm and the size of the number or volume distribution criterion is 9 nm. No particles were observed under an optical microscope or polarized light. The results indicate that no crystalline particles are present.
The liquid dispersion of the nanoparticles was lyophilized after the addition of lactose (2% w / w).
A white solid material was obtained and a clear system similar to that before lyophilization upon reconstitution in water was obtained. The particle size of the reconstituted sample was very similar to that of the original formulation, with a Z-average of about 40 nm and diameters based on volume and number distribution of 10-12 nm.
Example 26
Preparation of Nanoparticles of Cyclosporin A
119 mg of cyclosporin A was dissolved in butyl acetate and mixed with 2 grams of Triton X-100: propylene glycol 4: 1 solution. A transparent system was obtained. 7 g of water was added dropwise with slight shaking. An oil microemulsion in clear water was obtained. 7 g of 1% casein solution was added with slight shaking. The system became somewhat cloudy. The sample was diluted 1: 1 with water and then the solvent was evaporated. Butyl acetate was removed in rotochet at 40 ° C. under 80 mm Hg. The system became completely transparent. In addition, very small nanoparticles were obtained by the above method, the Z-average was 45 nm and the diameters based on volume and number distribution were 11 nm.
The liquid dispersion of the nanoparticles was lyophilized after the addition of lactose (2% w / w).
A white solid material was obtained which obtained a transparency similar to that before lyophilization upon reconstitution in water. The particle size of the reconstituted sample was very similar to that of the original formulation, with a Z-average of about 25 nm and diameters of 9 to 11 nm based on volume and number distribution.
Example 27
Cyclosporin nanoparticles
Microemulsions were prepared with a composition, 50 mg of cyclosporine, 0.5 g of butyl acetate, 3.04 g of Tween 80: propylene glycol (1: 1) and 6.8 g of water. The microemulsion was evaporated to give a clear liquid containing 5 mg / ml cyclosporine. In the comparative experiment, the above components were carried out by simple mixing except for butyl acetate, but after 17 hours, cyclosporin was not dissolved.
Polysorbate (twin), sorbitan ester (span), sucrose ester, lecithin, monodiglyceride, polyethylene-polypropylene block copolymer (fluromix), soap (sodium stearate, etc.), sodium glycolate bile salt, There are several possibilities for surfactants including ethoxylated castor oil, sodium stearoyl-lactylate, ethoxylated fatty acids (myrj), ethoxylated fatty alcohols (Brij), sodium dodecyl sulfate (SDS) and the like. In addition, generally biopolymers such as starch, gelatin, cellulose derivatives and the like can be used. In addition, all acceptable food grade surfactants, as well as those present in the McCutcheon Handbook of Surfactants or the CTFA Index, can be used for oral administration. Cosolvents or co-surfactants possible for microemulsions include propylene glycol, ethanol, glycerol, butanol, oleic acid and the like.
<Example 28>
Preparation of BHT Nanoparticles
110 mg of butylated hydroxy toluene (BHT) was dissolved in 1 ml of toluene and mixed with 2 ml of Triton X-100: n-butanol 4: 1 solution. 32 g of 1% casein solution was added and the microemulsion formed spontaneously. The microemulsion was evaporated under reduced pressure 80 mmHg until it became clear at 40 ° C. The particle size obtained had a Z-average of 30 nm and diameters of the volume and number distribution criteria were 16 and 15 nm, respectively.
<Example 29>
Preparation of BHT Nanoparticles
A similar method as described in Example 27 was performed using water instead of casein solution. After evaporation at 40 ° C. under 80 mmHg the system became clear and the Z-average size was about 10 nm.
<Example 30>
Preparation of Paclitaxel Nanoparticles
30 mg of paclitaxel was dissolved in 2 ml of butyl acetate and added to 4 g of 4: 1 triton x-100: propylene glycol. 40 ml of water was added and the system became slightly cloudy. After evaporation the system became completely clear. The Z-average size was 6 nm and the size of the volume and number distribution criteria was 7-9 nm. The same size was obtained at 4 ° C. after 1 day.
<Example 31>
Confirmation of microemulsion state diagram
It has been found that the composition can be used to provide microemulsions and obtain nanoparticles by solvent evaporation. The composition should contain a hydrophobic molecule, an aqueous solution as a continuous medium, a surfactant and possibly a water miscible solvent that can dissolve the cosurfactant.
Microemulsions of butyl acetate in water may be formed in various compositions described by state diagrams (butyl acetate is classified as a solvent having an acceptable residual high concentration in the final product). In addition, surfactants and cosurfactants are used in food and pharmaceutical applications: Tween 80 (ethoxylated sorbitan monooleate) and propylene glycol. Preliminary experiments were performed by using BHT as a model hydrophobic molecule to obtain a dispersion of particles of 20-50 nm in size. About 100% of BHT passed through the membrane after filtration with a 0.2 μm filter.
The state diagrams of the various compositions of surfactant / cosurfactant were obtained by vortexing a mixture of surfactant / cosurfactant (prepared prior to mixing with the solvent in various proportions) and solvent followed by dropwise addition of water. The turbidity of various compositions was observed along the "water line" and the compositions providing the translucency were further analyzed by light scattering. By using various ratios of solvent / surfactant / cosurfactant the region of the state diagram providing the microemulsion was identified (only a few selected components gave the microemulsion). The same method was used for the system in which the BHT was dissolved in butyl acetate and then the state diagram experiment was performed.
The microfiltration and the "filterability" of the nanoparticles containing BHT were evaluated by comparing the UV absorption spectra before and after 0.2 μm filtration. Nanoparticles were obtained by vacuum evaporation of butyl acetate (60 mmHg, 40 ° C.). It should be stressed that no high shear device is used throughout the entire method.
It has been found that microemulsion systems can be used for oral delivery. n-butyl acetate was selected as solvent. The following surfactants and cosurfactants were evaluated at various ratios.
Tween 80: glycerol 5: 1
Tween 80: glycerol 4: 1
Tween 80: glycerol 3: 1
Tween 80: glycerol 2: 1
Tween 80: Glycerol 1: 1
Span 80: Glycerol 4: 1
Span 80: Glycerol 3: 1
Tween 80: Propylene Glycol 4: 1
Tween 80: Propylene Glycol 3: 1
Tween 80: Propylene Glycol 2.5: 1
Tween 80: Propylene glycol 1.5: 1
Tween 80: Propylene glycol 1: 1
Tween 80: Propylene Glycol 1: 2
((Twin 80 + span 80) 7: 1): Propylene glycol 3.5: 1
((Twin 80 + span 80) 7: 1): Propylene glycol 1: 1
((Twin 80 + span 80) 8: 1): Propylene glycol 4: 1
((Twin 80 + span 80) 5: 1): Propylene glycol 1: 1
Tween 80: ((propylene glycol + glycerol) 1: 1.2) 2: 1
Suitable compositions are Tween 80 as surfactant and propylene glycol 1: 1 as cosurfactant. The overall state diagram was evaluated for n-butyl acetate based, tween 80: propylene glycol 1: 1, water. Two other solvents were tested, s-butyl acetate and t-butyl acetate. The state diagram for the system was the same as that of n-butyl acetate. n-butyl acetate system, Tween 80: propylene glycol 1: 1, water was further evaluated.
Measurement of particle size for sample 7% butyl acetate, 30% surfactant / PG, 63% water was performed. The Z mean was about 20 nm. The nanoparticle formation method was carried out at a concentration of about 10 mg (5% butyl acetate, 23% surfactant / PG, 72% water) in 1 g of butyl acetate for the water insoluble dye, Sudan III. The particle size was about 17 nm. The nanoparticle formation method was also performed on BHT at a concentration of 100 mg in 1 g butyl acetate. The state diagram for the system was determined. Depending on the composition, the particle size was about 20-50 nm.
Comparative experiments with means III and BHT were performed. 14.4 g of water were added to 10 mg of Sudan III and 4.6 g of surfactant / PG were added to the mixture. Samples were stirred for 24 hours in a magnetic stirrer. The degradation of means III was observed. However, no degradation of BHT was observed when the same experiment was performed with BHT (100 mg of BHT in 9 g of water and 4.3 g of surfactant / PG). Evaporation was carried out in this step (temperature 40 ° C., pressure about 60 mmHg). Particle size measurements on the samples were performed before and after evaporation. Z-averages of about 20-50 nm and 30 nm were obtained for samples before and after evaporation, respectively.
The sample after evaporation was filtered through a 0.2 μm filter and the BHT concentration before and after filtration was measured by UV absorption. There was no difference between the two samples. The results clearly suggest very few BHT nanoparticles.
Two samples were prepared (composition for this sample: Sample No. 1: 4% Butyl Acetate; 14% Surfactant / PG; 80% Water; Sample No. 2: BHT 123 mg / Butyl Acetate g; 5% Butyl Acetate; 18% surfactant / PG; 77% water).
<Example 32>
Substitute in method device selection
The method apparatus used to make current batches is one that increases in scale for clinical manufacturing. Capsol There are several alternatives in the selection of larger scale eye devices for manufacture. Some substitutes are as follows.
Device category Select device Premixer Blade Mixer, Rotostato Mixer High pressure device High Pressure Homogenizer (Avestin, Microfluidics, Stansted), Acoustic Wave Crusher (Heating System) Solvent removal device Rotary Evaporators, Continuous Flow Evaporators, Wiped Membrane Evaporators, Flash Evaporators, Recirculating Concentrators, Ultrafilters Dewatering device Freeze Dryer, Spray Dryer
<Example 33>
Formulated Intravenous Delivery System from Various Materials
The material used for the preparation of the intravenous delivery system may be a polymer (eg, polyethylene, polyvinyl, polypropylene perfusion, etc.) or glass. Standard medical grade perfusion is known to contain hydrophobic residues on its inner surface. The residue thus comes into contact with the injection solution. In fact, such perfusion is specifically designed to bring hydrophobic moieties into contact with the treatment solution, such as catheters, to reduce the adsorption of aqueous materials to the perfusion. However, any hydrophobic moiety in the treatment solution will attempt to bind to catheter perfusion and other components of the delivery system. Thus, a substantial portion of the hydrophobic pharmacologically active agent is sequestered on the inner wall of the perfusion catheter and delivery tube. Thus, the dosage of the hydrophobic pharmacologically active agent can vary because a substantial portion of the active agent can be absorbed into the perfusion wall. In severe treatment, when hydrophobic pharmacologically active agents are used to treat a disease, a significant reduction in the effective dose of the active agent can lead to treatment failure. Failure occurs especially when using therapeutic moieties, which require the active agent to be present above a certain level but with a narrow therapeutic window.
New methods of intravenous introduction of hydrophobic pharmacologically active agents have been developed. By protecting the hydrophobic moiety of the active agent, the tendency of the active agent to attach to perfusion through association with the hydrophobic moiety (eg albumin) of the biocompatible coating is drastically reduced. Thus, the present invention makes it possible to use very hydrophobic drugs with standard medical grade polymers and hydrophobic glasses, which are protected and therefore not absorbed by the surface. The method of the present invention involves placing a protective coating of a biocompatible polymer (eg, albumin) around the hydrophobic drug and placing the resulting composition in a hydrophobic polymer delivery system. The method of the present invention may thus improve the delivery of various hydrophobic therapies.
<Example 34>
HPLC Analysis of Paclitaxel
Chromatography
HPLC: Shimadzu LC-10AS Solvent Delivery System
Shimatsu SIL-10A Automatic Injector
Shimatsu SCL-10A type regulator
Shimatsu SPD-M10AV Diode Array Detector
Shimatsu CTO-10A Column Oven
Column: Kurosil-PPP, 5 μm, 4.6 mm × 25 cm, Phenomenex or C-18
Mobile phase: water / acetonitrile 65:45
Flow rate: isocratic, 1.0 ml / min
Detection: 228 nm
Identification of Paclitaxel Bulk Drug Substance (BDS)
Paclitaxel BDS and Paclitaxel Standard (99.9%, Hauser Chemical Research, Inc., Lot 1782-105-5) were quantitatively dissolved in acetonitrile and injected separately by HPLC. 10 μl of 1.00 mg / ml paclitaxel BDS and 10 μl of 2.07 mg / ml standard paclitaxel were injected. The retention time of the main peak of paclitaxel BDS was consistent with the retention time of paclitaxel standard (Hauser).
<Efficacy of Paclitaxel BDS>
Paclitaxel BDS and standard paclitaxel were injected into HPLC as described above. The efficacy of paclitaxel was derived based on the peak area ratio of paclitaxel BDS over standard paclitaxel and the known efficacy of standard paclitaxel.
Impurity Profile of Paclitaxel BDS
Chromatography systems as described above were able to provide high resolution of taxanes. 10-20 μl of 1.0 mg / ml paclitaxel BDS in acetonitrile corresponding to the linear reaction range of the HPLC system was injected into the HPLC. Impurity profiles were measured by relative peak areas.
<Efficacy analysis of paclitaxel in capsol®
Standard solutions (60, 100, 120, 140 and 160 μg / ml) were prepared by quantitative dissolution of paclitaxel BDS in 3% HSA. Capsol The sample was diluted in saline to a paclitaxel concentration of about 100 μg / ml. Standard Solution and Capsule The sample was fixed with cepharomannin as an internal standard, followed by solid phase extraction and liquid phase extraction (see below). Individually standard formulations and capsules 20-30 μl of the same volume of the sample preparation was injected into HPLC to determine the peak reaction ratio between paclitaxel and internal standard cepharomanmanine. Calibration curves were obtained by conventional least squares methods for results from standard injection solutions. Capsol The efficacy of paclitaxel in (registered trademark) was determined by comparing the peak reaction ratio of the sample injection and the standard injection.
<Capsule Impurity Profile of Paclitaxel in
Capsol (R) was subjected to solid phase extraction or liquid phase extraction (see below) followed by injection into HPLC. Capsol The impurity profile was examined as above by injecting 30 µl of approximately 1 mg / ml paclitaxel extracted from the trademark.
<Solid phase extraction>
Capsol The sample was reconstituted to about 100 μg / ml in saline. Solid phase extraction column, Bond-Elut (C-18) was conditioned with water. The column was loaded with a sample and passed through the column using vacuum. The column was then washed with water and then paclitaxel eluted with acetonitrile. Paclitaxel containing eluate extracted in acetonitrile was injected into HPLC.
<Liquid extraction>
Capsol The sample was reconstituted to about 100 μg / ml in saline. To about 200 μl of the sample, 800 μl of acetonitrile was added. The mixture was vortexed for 30 seconds and then centrifuged at 3000 g for 5 minutes. The supernatant was removed and collected. The pellet was resuspended in 200 μl brine and the extraction step was repeated. The second supernatant was combined with the first supernatant. The combined extracts were concentrated by evaporation and then injected into HPLC.
<Example 35>
Particle Size Distribution by Photon Correlation Spectrometer (PCS)
Reconstituted Capsule The particle size distribution of (registered trademark) was analyzed by a photon correlation spectrometer (PCS) on Malvern Zetasizer, Malvern Instruments Ltd. Jetta Sizer NIST Records Nanosphere Size Standard (Nanosphere Size Standards) was calibrated by Duke Scientific Corporation. Capsules on Malvern Zetasizer (Registered trademark) The method for measuring the particle size includes adjusting the following factors.
Temperature: 20.70 ℃,
Scattering angle: 90。
Index of Dispersion: 1.33
Wavelength: 633 nm
Viscosity (Auto): 0.99
Real refractive index: 1.59
Virtual refractive index: 0
The dilution of the sample required for good size measurements was determined from the kcts / sec recorder after preparation of the zetaser (200 μL of the sample was aliquoted into a cuvette and then diluted with about 2 mL of 0.22 μm filtered filtered distilled water). The cuvette was placed in a cuvette holder in the zetasizer and the measurement started. After the measurement began, the correlator adjustment display appeared. Display speedometer was selected from the menu. The speed should be in the mid range 100-250 kcts / sec. If the rate is too large or too small, another sample was prepared in thick or dilute diluent. Reconstituted Capsule The size of (registered trademark) was automatically performed three times by multimode analysis, analyzed, averaged and recorded. Average particle size is capsule It was 155 nm +/- 23 nm with respect to 25 batches of (trademark).
<Example 36>
Polymer shells as carriers for polynucleotide structures, enzymes and vaccines
As gene therapy is more widely accepted as an available treatment option (currently more than 40 human gene transfer proposals are approved by NIH and / or FDA reviewers), one should overcome in implementing the treatment method The barrier is resistance to the use of viral vectors to incorporate genetic material into the genome of human cells. Viruses are inherently toxic. Thus, the risks inherent in the use of viral vectors in gene therapy, particularly in nonfatal and nongenic diseases, are not tolerated. Disadvantageously, plasmids delivered without the use of viral vectors are generally not incorporated into the genome of the target cell. In addition, as in the case of conventional drugs, the plasmid has a half-life limited to the human body. Thus, general limitations in the implementation of gene therapy (and anti-sensitization therapy in which it is a reverse form of gene therapy and in which nucleic acids or oligonucleotides are introduced to inhibit gene expression) are large in size and therefore efficient for nucleic acids or oligonucleotides that can penetrate the cell membrane It cannot be delivered to.
Encapsulating DNA, RNA, plasmids, oligonucleotides, enzymes, and the like into protein microcapsule shells as described above may facilitate their targeted delivery to the liver, lung, spleen, lymph and bone marrow. Thus, the biologic according to the present invention can be delivered to an intercellular location without the associated risks associated with the use of the viral vector. Agents of this type facilitate nonspecific or cellular foreign uptake of the polymer shell directly from the blood stream into the cells of the RES, into the muscle cells by intramuscular injection, or into the tumor by direct injection. In addition, monoclonal antibodies against nuclear receptors can be used to target products encapsulated in the nucleus of certain cell types.
Diseases that may be targeted by the construct include diabetes, hepatitis, hemophilia, cystic fibrosis, multiple sclerosis, general cancer, colds, AIDS, and the like. For example, the gene for insulin type growth factor (IGF-1) can be encapsulated into protein cells for delivery for the treatment of diabetic peripheral neuropathy and poor health. Genes encoding Factor IX and Factor VIII (useful for treating hemophilia) can be targeted to the liver by encapsulation in the protein microcapsule shell of the invention. Similarly, genes for low density lipoprotein (LDL) receptors can be targeted to the liver for the treatment of atherosclerosis by encapsulation in the protein microcapsule shell of the invention.
Other genes useful in the practice of the present invention are genes that restimulate the body's immune response to tumor cells. Antigens such as, for example, HLA-B7 encoded by DNA contained in plasmids can be incorporated into the protein shells of the invention for direct injection into tumors (eg skin cancer). Within a tumor, antigens are recruited to tumor specific cells to increase the level of cytokines (eg, IL-2) that make the tumor a target for immune system attack.
As another example, plasmids containing portions of the adeno-associated viral genome are believed to be encapsulated in the protein microcapsule shell of the present invention. In addition, the protein microcapsule shell of the present invention can deliver the therapeutic genes to CD8 + T cells for immunotherapy adopted against various tumors and infectious diseases.
The protein shells of the invention can also be used as delivery systems to combat infectious diseases, for example, via targeted delivery of anti-sensitizing nucleotides to hepatitis B virus. An example of such antisensitizing oligonucleotides is 21-merphosphothioate for the polyadenylation signal of hepatitis B virus.
The protein shells of the invention can also be used for the delivery of cystic fibrosis transmembrane regulatory (CFTR) genes. Humans lacking these genes develop cystic fibrosis, which can be treated by spraying the protein microcapsule shell of the present invention containing the CFTR gene and inhaling it directly into the lungs.
Enzymes can also be delivered using the protein shells of the invention. For example, the enzyme, DNAse, can be encapsulated and delivered to the lungs. Similarly, ribozymes can be targeted to virus envelope proteins or virus infected cells by encapsulating and attaching suitable antibodies to the exterior of the polymer shell.
<Example 37>
Local treatment of brain tumors and intraperitoneal tumors
Local delivery of chemotherapeutic agents to tumors is an effective method of limiting side effects by minimizing the dose during prolonged exposure to the drug. The biocompatible materials discussed above are also used in several physical forms, such as gels (crosslinked or uncrosslinked), to provide a substrate, which releases the pharmacologically active component, eg, paclitaxel, by diffusion and / or degradation of the substrate. Can be. Capsol® can be dispersed in a matrix of biocompatible material to continuously release the formulation of paclitaxel during treatment of brain tumors and tumors in the peritoneal cavity (ovarian cancer and metastatic disease). Temperature sensitive materials can also be used as the dispersion substrate for the formulations of the present invention. That is, for example, Capsol® can be injected into liquid formulations of temperature sensitive materials (e.g., copolymers of polyacrylamide or copolymers of polyalkylene glycol, and polyactide / glycolide, etc.) It gels at the tumor site and slowly releases Capsol®. Capsol® formulations can be dispersed into the substrates of the biocompatible polymers described above to control the release of paclitaxel formulations, thereby lowering the above-mentioned systemic toxicity in terms of the properties of the capsules (albumin combined with paclitaxel). In addition, the toxicity to brain tissue is reduced. This combination of a capsol or similarly formulated chemotherapeutic agent with a biocompatible polymer substrate is used to locally deliver chemotherapeutic agents for treatment of solid tumors (ovarian cancer) in the brain and peritoneum to other solid tumors and It may be useful to regulate local use for solid tumors. This combination preparation is not limited to the use of paclitaxel and can be used with a wide range of pharmacologically active ingredients including anti-infectives, immunosuppressants and other chemotherapeutic agents.
<Example 38>
Stability of Capsol® after Reconstitution
Lyophilized capsol® in glass vials were reconstituted at sterile standard saline to concentrations of 1, 5, 10 and 15 mg / ml and stored at room temperature and refrigerated conditions. This suspension was found to be homogeneous for at least 3 days under these conditions. The particle size was measured at various time points, but the size distribution did not change. No precipitation was found under these conditions. This stability was unexpected and overcomes the problems associated with Taxol® which precipitates within about 24 hours after reconstitution at a recommended concentration of 0.6-1.2 mg / ml.
In addition, the reconstituted capsol® was stable in the presence of different polymeric tubing materials such as Teflon, plastic, polyethylene, Tygon and other standard infusion tubing materials. This is a major advantage over Taxol® limited to polyethylene infusion sets and glass infusion bottles.
<Example 39>
Unit dosage form for CAPSOL (registered trademark)
Capsol® was prepared as a lyophilized powder in a vial of the appropriate size. Subsequently, the desired dosage was filled into a suitable container and lyophilized to obtain a powder mainly containing the desired amount of albumin and paclitaxel. This vessel was then reconstituted to an appropriate volume from the point of view of use using sterile standard saline or other aqueous dilution to obtain a homogeneous suspension of paclitaxel in the diluent. The reconstituted solution can be administered directly to the patient by injection or infusion using a standard intravenous infusion set.
In addition, Capsol® was prepared as a frozen solution readily available for immediate use in a bottle or bag that can be thawed upon use and simply administered to a patient. As such, the lyophilization step was omitted in the manufacturing process.
When capsol® and Taxol® were administered to rats at equal doses of paclitaxel, the Taxol® group showed higher myelosuppressive effects than the Capsol® group. It was amazing. This could reduce the incidence of infection and fever episodes (eg, neutropenia due to fever). This may also reduce the interval between treatments, which is currently 21 days. When using a pharmaceutical composition prepared according to the present invention, the cycle may be reduced to less than two weeks, which may be more effective in treating cancer. Thus, the use of pharmaceutical compositions prepared according to the present invention may provide substantial advantages over Taxol®.
<Example 40>
Oral delivery of the drug
Taxol® is very poorly absorbed by the oral route. Particulate agents, such as Capsol®, can greatly improve the absorption of drugs such as paclitaxel. In addition, the paclitaxel formulations of the invention prepared via the microemulsion / deposition method are useful for absorbing drugs through the oral cavity. Combining such agents with surfactants greatly improves the oral biocompatibility of the drug. The use of lipids, surfactants, enzyme inhibitors, osmotic agents, ion pairing agents, metabolic inhibitors and the like has surprisingly been found to increase the oral absorption of paclitaxel formulations of the invention. Examples of ion pairing agents include, but are not limited to, trichloroacetate, trichloroacetate salicylate, naphthalene sulfonic acid, glycine, bis-N, N-dibutylaminoethylene carbonate, n-alkyl sulfonate and n-alkyl sulfate It doesn't happen. Examples of membrane osmosis improvers include sodium caprate, acyl glycerides, polyoxyethylene alkyl ether saxyl carnitine, sodium cholate, sodium taurocholate, sodium taurodihydrofucidate, EDTA, sodium salicylate and sodium methoxysalicylate Include, but are not limited to. A non-limiting list of lipids and surfactants that can be used in the formulations of the present invention is described below.
<Example 41>
Mode of Administration of the Inventive Formulations and Capsules of Other Drugs
Formulations of the present invention may be administered by intravenous injection, intravenous infusion, intraperitoneal injection, intraarterial injection, intraportal injection, liver coloration, intratumoral injection or subcutaneous injection, urethral injection or iontophoresis, intramuscular injection, subcutaneous injection , Intrathecal injection, inhalation of dry powder or spray, and the like.
<Example 42>
Use of Capsol® for targeting angiogenic vasculature
Angiogenesis has been implicated as a factor that causes and / or augments disease progression such as cancer, rheumatoid arthritis and retinopathy. The inventors have surprisingly found that capsol® can not only treat tumors in animal models, but can also relieve or reverse the pain of rheumatoid arthritis. Thus, it is possible for the capsol® to have antiangiogenic activity. In order to make Capsol® more effective, it is possible to bind a suitable peptide to Capsol® to target the angiogenic vasculature. An example of such a peptide is RGD (arginine-glycine-aspartic acid). For targeting therapies, many other peptides with similar activity can be bound to capsol® or other drugs made by the methods of the invention. Peptide / Capsol® may be administered to a patient by conventional means as needed.
<Example 43>
Use of capsol® for the treatment of liver disease
Terminal hepatocellular carcinoma and other cancers of the liver can be treated by intracapsular administration of Capsol®. By directly developing the liver, the dosage reached by the liver can be greatly improved. In addition, higher dosages than conventional Taxol® may be used to treat the disease more efficiently. In addition, for greater therapeutic efficacy, suitable targeting agents such as peptides or proteins located in hepatic tissue may be used in combination with Capsol®.
<Example 44>
Toxicity / myelosuppression studies of paclitaxel-a comparative study of Taxol® and Capsol® for single dose administration to rats
An overview of preclinical studies is shown below:
Schedule: 1X, single dose intravenous infusion (1 day)
Animals: Sprague Dawley rats, 40 males, 40 females, sex per 5 rats / group
Weight: 300 ± 50 g
Study period: 15 days
Treatment group: Taxol® (vehicle 1 + treatment group 3)
CAPSOL® (Vehicle * 1 + Treatment Group 3)
Dosage: Taxol® (0, 3, 6 and 9 mg / kg)
Capsol® (0, 6, 9 and 12 mg / kg)
Dose concentration: 0.6 mg / ml (all rats)
Dosing volume: Taxol® (15, 5, 10, 15 mL / kg)
Capsol® (20, 10, 15 and 20 ml / kg)
Infusion rate: approximately 0.75 ml / hour (all rats)
Route of administration: intravenous infusion, tail vein
Clin obs: 1X / day
Clin Path: 0 (before treatment), 1, 3, 7, 11, 15 days, Do standard list for NCI Tox Branch
Weight: -1, 1, 3, 8 and 15 days
(* Vehicles were prepared in the same manner as described in the Preparation section, except that the addition of paclitaxel was omitted.)
<Example 45>
Pilot myelosuppression (hematologic toxicity) study
Prior to beginning a formal study, a pilot study using three rats of the Capsol® group and three rats of the Taxol® group was performed to determine the results. The dosage used was 5 mg / kg, with a dosing volume of 7 ml / kg. Doses were administered intravenously via the tail vein. The results of this study are summarized in the graph of FIG. 3 showing the percentage change in WBC count (indicator of myelosuppression) for each formulation as a function of time.
Conclusion of Pilot Myelosuppression Study
The data showed that the WBC count (mean value + SD) was significantly lower in the Taxol® group compared to the Capsol® group, indicating that the degree of bone marrow suppression for Taxol® was greater (Taxol (registered) Maximum WBC inhibition for>) was> 70%, whereas maximum WBC inhibition for Capsol® was <30%). Analysis of this data indicated that there was a statistically significant difference (p <0.05) between the two groups for all data points except 0, 13 and 14 days. In addition, normal levels of WBCs recovered within 6 days in the group injected with Capsol®, whereas Taxol® group recovered WBC normal levels on day 14. This indicates that the hematologic toxicity was significantly reduced for Capsol®. If similar results are found in human clinical trials, this data can significantly reduce the interval between treatment cycles (currently 3 weeks for Taxol®) (perhaps 2 weeks with Capsol®). Or 1 week or less).
<Example 46>
Pilot Study of Antitumor Efficacy
Prior to starting the study, a pilot study using Capsol® was performed to determine the target dose range and efficacy. MX-1 breast tumors were implanted subcutaneously in mice (n = 10) and treatment started when the tumor size reached about 150-300 mg. This tumor size reached day 12 and started treatment 13 days after implantation. Capsol® was reconstituted with saline to obtain a nanoparticle colloidal solution of paclitaxel. Tumor containing mice (n = 5) were treated with a reconstituted capsol® at a dose of 20 mg / kg (denoted as VIV-1) via tail vein mass injection daily for 5 days. Only saline was administered to the control tumor-containing group (n = 5) on the same schedule. Tumor size was monitored as a function of time. Tumor weight was found to increase significantly above average 4500 mg and all animals in this group were sacrificed between 28 and 39 days. On the other hand, the treatment group showed significant therapeutic efficacy and all animals had no measurable tumor at 25 days. Animals in this group were all sacrificed on day 39, when there were no signs of recurrence and no evidence of tumor. The result is shown in FIG.
Conclusion
This study showed significant antitumor activity against Capsol®. In other words, the antitumor activity of paclitaxel was preserved in the Capsol® formulation. This study showed that intravenous administration of paclitaxel nanoparticles can be as effective as administration of soluble forms of the drug. That is, Capsol® exhibited potent and potent anti-tumor activity without the toxic effects seen in approved and commercially available cremaphor-containing Taxol® formulations.
Note: Based on the literature data and experience of Southern Research Institute (SRI) scholars, the maximum tolerated dose (MTD) of paclitaxel dissolved in diluent 12 (the same diluent used for Cremaphor / Ethanol, Taxol®) Was established to be 22.5 mg / kg for this particular strain of athymic mice. This result was obtained by dissolving paclitaxel at a much higher concentration in dilution 12 compared to Taxol® (6 mg / ml in cremaphor / ethanol). This minimized the amount of cremaphor / ethanol administered to the mice to avoid vehicle toxicity. At the dose of 22.5 mg / kg, the efficacy of paclitaxel in dilution 12 was similar to that of the capsule.
<Example 47>
Treatment with Paclitaxel Nanoparticles for Rheumatoid Arthritis in Animal Models
A collagen-induced arthritis model in Lauuvain rats was used to test the therapeutic effect of paclitaxel nanoparticles on arthritis. The paw size of the test animals was monitored to assess the severity of arthritis.
After sufficient development of arthritis (generally 9 to 10 days after collagen injection), the experimental animals were treated with 1 mg / kg of paclitaxel nanoparticles or 0.5 mg / kg of paclitaxel nanoparticles and 0.2 mg / kg of prednisone, respectively (complex treatment). ) Was divided into different groups administered 6 times intraperitoneally with 1 dose per week for 3 weeks. Forefoot size was measured at the beginning of treatment (day 0) and measured at each drug infusion. One group to which only normal saline was administered was used as a control. At the end of the experiment, the forefoot size of the group receiving paclitaxel nanoparticles decreased by 42%, and the forefoot size of the combined treatment group decreased by 33%, while the forefoot size of the control group increased by about 20%. The original forefoot size before arthritis was 50%. The results are shown in FIG. In conclusion, the therapeutic effect of paclitaxel-containing nanoparticles against arthritis has been demonstrated. To avoid the side effects of long-term use of both paclitaxel and steroids, it would probably be better to choose a combination therapy that achieves similar effects but administers each drug in half the dose.
<Example 48>
Effect of Capsol® on Arterial Stenosis
Abnormal vascular smooth muscle division (VSMP) is associated with cardiovascular diseases such as atherosclerosis, hypertensin and most internal vascular progression. Abnormal VSMP is a common complication of percutaneous transcutaneous coronary angiogenesis (PTCA). The incidence of chronic restenosis caused by VSMP after PTCA has been reported to be 40-50% higher in 3-6 months.
As the in vivo model of restenosis develops due to the high incidence of vascular reclosure associated with PTCA, an agent must be found to prevent this. The following study demonstrates the use of Capsol® in preventing restenosis after endovascular trauma to the artery.
Male Sprague-Dawley rats weighing 350-400 gm were anesthetized with ketamine and rompon and the normal right carotid artery was exposed at 3.0 cm intervals. The attached tissue was washed so that the two DIETRICH micro bulldog clamps were located about 2 cm around the carotid artery without crushing the vagus nerve or related upper cervical ganglion and sympathetic cords. There were no branch tubes along this segment of the blood vessel. A 30-gauge needle attached to a three-way stopcock was first inserted, then the lower end of the isolated segment was pulled out to make a hole in the wall of the vessel and then inserted at the upper end for injection. All blood inside the isolated segment was rinsed by injecting 2-3 mL of saline buffered with phosphate, and then the three way stopcock was switched to another connection to a controlled source of compressed air. A gentle stream of air (25 ml / min) was passed through the lumen of the vessel for 3 minutes to dry the endothelial wound. The segment was then refilled with saline and the needle removed from the vessel. Prior to removing the clamp, the needle hole on the vessel wall was cauterized carefully to prevent bleeding. You can also use a cotton swab drenched in saline to block the needle hole and stop bleeding. The skin was closed with 7.5 mm metal clips and washed with betadine.
All animals who received the procedure described above were sacrificed on day 14 after the procedure. Carotid arteries on each side recovered during pathological examination. The unoperated side will serve as a self control. The experimental groups treated differently were as follows:
Group 1: High Dose Capsol® Treatment Group:
Paclitaxel 5 mg (human albumin w / 100 mg) / kg / week, intravenously.
Group 2: Low Dose Capsol® Treatment Group:
Paclitaxel 1 mg (human albumin w / 20 mg) / kg / week, intravenously.
Group 3: Drug Vehicle Controls:
Human albumin 100 mg / kg / week, intravenously.
After carotid biopsy samples were preserved with formalin, the cross section (8 μm) was excised from paraffin blocks and stained with hematoxylin and eosin. The cross sectional area of the vascular layer (intima, mesentery and epithelium) was quantified.
Injured carotid arteries of the control group showed significant accumulation of endothelial smooth muscle cells and invasion of VSMC into the lowermost membrane. The total thickness of the carotid wall was doubled. The treatment group showed a satisfactory reduction in the intima wall thickness compared to the control group.
<Example 49>
In vivo targeting of nanoparticles
By incorporating certain targeting moieties such as proteins, antibodies, enzymes, peptides, oligonucleotides, sugars, polysaccharides and the like into the protein coating of the nanoparticles, it is possible to target specific sites in the body. Such targeting ability can be used for therapeutic or diagnostic purposes.
<Example 50>
Antibody Targeting of Polymer Shells
Due to the nature of the polymer shell of certain aspects of the invention it was possible to bind monoclonal or polyclonal antibodies to the polymer shell or to incorporate the antibodies into the polymer shell. The antibody could be incorporated into the polymer shell as the polymer microcapsule shell formed or the antibody could be bound to the polymer shell after preparation of the polymer shell. Standard protein immobilization techniques could be used for this purpose. For example, for protein microcapsules made of proteins such as albumin, many amino groups on albumin lysine residues were available for binding of suitably modified antibodies. As an example, the anti-tumor agent can be delivered to the tumor by incorporating an antibody against the tumor into the polymer shell, or the antibody against the tumor can be bound to the polymer shell after the polymer shell is made. As another example, the gene product may be delivered to a specific cell by incorporating an antibody against a receptor on the target cell into a polymer shell or the antibody against a receptor on the target cell may be bound to a polymer shell after preparation of the polymer shell. have. In addition, single cell antibodies against the core receptor can be used to target the encapsulated product against the core of a particular cell type.
<Example 51>
Targeting Immunosuppressors to Implanted Tissues Using Intravenous Delivery of Such Polymer Shell-Containing Formulations
Immunosuppressants are widely used after tissue transplantation for the prevention of rejection episodes. In particular, cyclosporin, an effective immunosuppressive agent, extends the survival rate of allogeneic grafts including the skin, heart, kidney, pancreas, bone marrow, small intestine and liver of animals. Cyclosporin inhibits some humoral immunity and in many animal species cell intermediary responses to many organs, such as allograft rejection, delayed hypersensitivity, empirical allergic encephalomyelitis, Freund's adjuvant arthritis and transplant organ versus individual response It has been demonstrated to suppress the disease to a greater extent. Renal, hepatic and cardiac allogeneic transplantation in humans with cyclosporin has been successfully performed.
Cyclosporines are currently delivered through the oral cavity in the form of capsules containing a solution of cyclosporin in alcohol, and emulsions such as corn oil, polyoxyethylated glycerides, or the like, or solutions such as olive oil, polyoxyethylated glycerides. It is also administered by intravenous injection, in which case it is dissolved in a solution of ethanol (about 30%) and cremaphor (polyoxyethylated castor oil) (1: 20 in conventional saline or 5% dextrose prior to injection). To 1: 100). The absolute biocompatibility of the oral solution is about 30% compared to intravenous infusion (Sandoz Pharmaceutical Corporation, Publication SDI-Z10 (A4), 1990). In general, due to intravenous delivery of cyclosporin, problems associated with current intravenous delivery of Taxol®, namely anaphylactic reactions believed to be due to cremaphor, the transport vehicle used for intravenous preparations, and Suffer from an allergic reaction. In addition, intravenous delivery of an encapsulated drug (eg cyclosporin) as described herein will avoid dangerous peak blood levels immediately after administration of the drug. For example, comparing the currently available formulations for cyclosporin with the above-mentioned encapsulated form of cyclosporin showed a five-fold reduction in the peak blood levels of cyclosporin immediately after injection.
To avoid the problems associated with cremaphor, the cyclosporin contained in the polymer shell described above can be delivered by intravenous injection. It can be dissolved in a biocompatible emulsion or a number of other solvents, which can then be dispersed in the polymer shell via sonication as described above. In addition, an important advantage in delivering cyclosporin (or other immunosuppressive agents) in the polymer shell is local targeting due to absorption by the RES system in the liver of the injected material. This may to some extent avoid systemic toxicity and reduce the effective dosage due to local targeting.
<Example 52>
Use of Capsol® for Antibody Targeting
Monoclonal antibodies against various tumors or tissues may be bound to Capsol® to target Capsol® or other drugs made by the methods of the present invention to the affected area of the disease. For example, binding of antibodies against ovarian cancer to Capsol® and intraperitoneal administration would provide a significant benefit to ovarian cancer patients.
<Example 53>
Intravenous Administration of Therapeutic Agents
Intravenous administration of a therapeutic agent, such as a drug, imaging agent, etc., pre-places the therapeutic agent in at least one route through the liver. If the therapeutic agent is filtered through the liver, a significant portion of the therapeutic agent is absorbed and isolated by the liver and therefore cannot be used for systemic distribution. In addition, once absorbed by the liver, they are easily metabolized and the resulting metabolic byproducts often have general systemic toxicity. Isolation by the liver resulting from intravenous administration is alleviated by encapsulating the drug or other therapeutic agent with a coating according to the invention (eg using a protein such as albumin). Albumin is known to be generally distributed throughout the patient, for example through the liver. Thus, isolation of albumin by the liver does not occur to the same extent as drugs or toxic compounds with hepatic receptors (or other mechanisms), which initiates the process of removing them from the bloodstream. By using a biocompatible polymer coating (eg, human albumin coating) to protect the therapeutic, the drug bypasses the liver and is generally distributed throughout all organ systems. According to one aspect of the present invention, a novel method for bypassing the liver is provided, which includes encapsulating the drug with human hepatic albumin, which is in fact a physiological component. In this way, the drug becomes more available for systemic treatment. In addition to increasing drug availability, the production of metabolic byproducts of hepatocellular drug degradation is reduced. Both increased hepatic bypass passage and decreased byproducts of drug metabolism synergistically improve overall drug efficacy. Improved efficacy is extended for all drugs and substances encapsulated with human albumin.
<Example 54>
Myelosuppression (hematologic toxicity) effect of drugs and reduction of general toxicity
Some chemotherapeutic drugs have dose limiting toxicity due to their myelosuppressive effects. Taxol® (Paclitaxel) is a classic example of such a drug. When administered with the current approved formulation of cremaphor / ethanol, Taxol® inhibits the myelosuppressive effect of limiting repeated administration of the drug and eliminating the patient's retreat for at least 3 weeks to restore the patient's blood levels to normal Indicates. It has been evident that the nontoxic biocompatibility of certain aspects of the drug carrier of the present invention, namely human albumin, can greatly reduce the toxic side effects of myelosuppression.
Commercial formulations (Taxol®) or formulations prepared by the methods of the present invention in Sprague Dolay rats were administered as nanoparticles with albumin. Both formulations were administered by tail vein injection. A single dose level of 5 mg / kg was administered for Taxol® while two doses of 5 mg / kg and 12 mg / kg were administered for the formulation of the present invention. After administration, the number of white blood cells in rats was monitored daily as an indicator of myelosuppression.
In the case of Taxol® (5 mg / kg), the number of WBCs decreased by 47.6% and 63.5%, respectively, 1 and 2 days after administration, whereas in the case of 5 mg / kg of the formulation of the present invention, the WBC number was administered. 14.7% and 2.4% increase after 1 day and 2 days of piglet respectively. When the dose of the formulation of the present invention was increased to 12 mg / kg, the number of WBCs increased by 6.5% and 3.6%, respectively, 1 and 2 days after administration.
This result indicates that short-term myelosuppression is greatly reduced by administering the drug in the formulation of the present invention.
Another indicator of general toxicity is the weight of the animal. After paclitaxel was administered, the body weight of the rat was also monitored. When 5 mg / kg of Taxol® was administered, the body weight decreased by 10.4% within 3 days of administration, whereas when the same amount of paclitaxel was administered with the formulation of the present invention, the body weight only decreased by 3.9%. The toxicity of the formulation is greatly reduced.
When the formulations of the present invention and Taxol® were administered to rats equally with the dosage of paclitaxel, a much higher degree of myelosuppressive effect was observed in the Taxol® group than in the formulation group of the present invention. It was amazing. This may reduce the incidence of infection and fever episodes (eg, neutropenia due to fever). This may also reduce the inter-treatment cycle, which is currently 21 days for Taxol®. Using pharmaceutical compositions prepared according to the present invention, this cycle can be reduced to two weeks, one week or less to allow for more effective treatment for cancer. Thus, pharmaceutical compositions prepared according to the present invention can be used to provide substantial benefits to Taxol®.
<Example 55>
Mass Dose Administration of Nanoparticle Formulations
Paclitaxel, an anticancer drug in commercial BMS formulations consisting of cremaphor / ethanol, cannot be administered intravenously in large quantities. This is because the widespread toxicity of the vehicle results in severe hypersensitivity reactions and the patient has to be pre-formulated with steroids, antihistamines and the like. Taxol® formulations are administered by in situ infusion and last from 1 hour to 24 hours. In contrast, the other formulations in the present invention use non-toxic carriers to facilitate large doses intravenously (ie, injection times of less than one hour) without the toxicity problems seen in today's clinically used Taxol®. May be administered to the patient.
Effective dosages of paclitaxel for a patient are typically between 200 and 500 mg depending on the weight of the patient or the patient's appearance. Taxol® must be administered at a final dosage concentration of 0.6 mg / ml, which requires a large volume of infusion (typically in the range of about 300 to 1000 ml). In contrast, the formulations of the present invention do not have this limitation and can be administered at a desired concentration. This allows clinicians to treat patients with rapid intravenous large doses that can be administered in as little as a few minutes. For example, when the formulation of the present invention is reconstituted at a dosage concentration of 20 mg / ml, the injection volume for a total dosage of 200 to 500 mg is only 10 to 25 ml each. This is a great advantage in clinical practice.
<Example 56>
Reduction of Paclitaxel Toxicity in Nanoparticle Formulations Compared to Taxol®
It is known that paclitaxel, an anticancer agent, has a wide range of toxicities and hypersensitivity in its commercial formulation (i.e. Taxol®), and that the patient is required to accept a drug in which preliminary drugs such as steroids and antihistamines are mixed. have. The toxicity of Taxol® is compared to the nanoparticle formulations of the present invention.
Therefore, different doses of the formulation were injected intravenously through the tail vein of C57BL mice, and mice were generally observed after injection to examine the toxic effects.
In the case of Taxol®, a dose of 30 mg / kg led to constant mortality within 5 minutes of intravenous administration. At the same dose, the nanoparticle formulations of the present invention did not show an apparent toxic effect. At the 103 mg / kg dose, the nanoparticle formulation showed some reduction in body weight in mice, but even at these high doses, it did not result in lethality.
The doses of approximately 1000 mg / kg, 800 mg / kg and 550 mg / kg all led to mortality, but differed in the range of several hours to 24 hours in terms of time to mortality. Thus, the lethal dose of the formulation of the present invention was greater than 103 mg / kg but less than 550 mg / kg.
Therefore, the lethal dose of the paclitaxel formulation of the present invention is substantially taxol It is obvious that it is higher than that of. This is of great significance in clinical practice, where higher doses of chemotherapeutic drugs have significantly less toxicity and can be administered for more effective tumor cell disruption activity.
<Example 57>
Determination of LD 50 in mice for Taxol® prepared by the method of the invention and for Taxol after one intravenous administration
The capsol®, Taxol®, and LD 50 of their carrier vehicle were compared after one intravenous administration. A total of 48 CD1 mice were used. Paclitaxol doses of 30, 103, 367. 548 and 822 mg / kg were tested for Capsol® and paclitaxol doses of 4, 6, 9, 13.4 and 20.1 mg / kg were tested for Taxol®. ). Human doses for albumin, vehicle for Capsol® were only tested at 4.94 g / kg (corresponding to 548 mg / ml Capsol® dose) It is not considered to be toxic. The doses tested for Taxol® vehicle (Cremophor EL®) were individually 1.5, 1.9, 2.8, corresponding to paclitaxel doses of 9, 11.3, 16.6 and 20.1 mg / kg. And 3.4 ml / kg. Three to four mice were dosed at each concentration. The results showed that paclitaxel administered with Capsol® was less toxic than that administered with Taxol® or Taxol® vehicle alone. LD 10 for LD 50 and Capsol® is paclitaxel in 447.4 and 371.5 mg / kg, paclitaxel in 7.53 and 5.13 mg / kg Taxol®, and Taxol® at 1325 and 794 mg / kg Vehicle (corresponding to 15.06 and 9.06 mg / kg paclitaxel doses). In this study, LD 50 for Capsol® was 59 times greater than Taxol® and 29 times greater than Taxol® vehicle alone. LD 10 for paclitaxel in Capsol® was 72 times greater than paclitaxel in Taxol®. A review of all data for this study suggests that Taxol® vehicles are responsible for the toxicity of Taxol®. Mice receiving Taxol® and Taxol® vehicles exhibited typical signs of severe hypersensitivity, manifested temporarily as bright pink skin pigmentation after administration. No such reaction was seen for the Capsol® and Capsol® vehicle groups.
The results are shown in Table 2.
Single intravenous administration group Dose (mg / kg) Number of animals (n) Fatalities survival% LD 50 (mg / kg) MTD or LD 10 (mg / kg) CAPSOL (registered trademark) 82254836710330 34333 34000 00100100100 447.4 371.5 Taxol (registered trademark) 20.113.4964 44343 44210 003375100 7.53 5.13
This high dose of Capsol® was administered as a concentrate injection and is equivalent to a dose of approximately 80-2000 mg / m 2 in humans. The maximum tolerated dose of LD 50 or Capsol® in this study corresponds to approximately 1000 mg / m 2 in humans. This is significantly higher than the accepted human dose of 175 mg / m 2 for Taxol®.
Surprisingly, the vehicle, cremophor / ethanol alone, caused severe hypersensitivity reactions and lethality in groups of mice at various doses. LD 50 data for Taxol® vehicle alone shows that it is significantly more toxic than Capsol® and significantly contributes to the toxicity of Taxol®. Although the cause of hypersensitivity in the literature is unclear, based on these data we believe that HSR can contribute to Taxol® vehicles.
<Example 58>
Capsules after Intravenous Multiple Dosing in Mice And LD of Taxol®50Measurement of
LD 50 of Capsol® and Taxol® and their carriers were compared by the following intravenous multiple doses. A total of 32 CD1 mice were used. Capsules containing paclitaxol doses of 30, 69 and 103 mg / kg were administered daily for five consecutive days. Taxol®, including paclitaxol doses of 4, 6, 9, 13.4 and 20.1 mg / kg, was administered daily for five consecutive days. Four mice were dosed at each concentration. The results are shown in Table 3.
Intravenous Multiple Administration group Dose (mg / kg) Number of animals Fatalities Can survive LD 50 (mg / kg) MTD or LD 10CAPSOL (registered trademark) 1036930 444 410 075100 76 64 Taxol (registered trademark) 20.113.4964 44444 44210 005075100 8.0 4.3
The results indicate that Capsol® is less toxic than Taxol®. LD 50 and LD 10 of kapsol (R) is individually paclitaxel of 76.2 and 64.5 ㎎ / ㎏, the Taxol (R) in the individually paclitaxel of 8.07 ㎎ / ㎏ and 4.3 ㎎ / ㎏ than this. In this study, LD 50 for Capsol® was 9.4 times higher than Taxol®. LD 10 for Capsol® was 15 times higher for Capsol® than for Taxol®. The results of this study suggest that Capsol® is less toxic than Taxol® when administered in multiple doses at daily intervals.
<Example 59>
Toxicity and Efficacy of Two Formulations of Capsol® and Taxol®
This study was performed to determine the efficacy of Capsol®, Taxol® and Taxol® vehicles in female athymic NCR-nu mice implanted with MX-1 human breast tumor fraction.
Each group of 5 mice was injected intravenously with Capsol® formulation VR-3 or VR-4 at doses of 13.4, 20, 30, 45 mg / kg / day for 5 days. Five groups of mice were also injected intravenously with Taxol® at doses of 13.4, 20 and 30 mg / kg / day for 5 days. Ten mouse standard groups were treated by intravenous injection of Capsol® vehicle standard (human albumin, 600 mg / kg / day) for 5 days. Evaluation parameters were the number of complete tumor regressions, the average duration of complete regression, tumor-free survivors, and tumor recurrence.
Treatment of Capsol® formulation VR-3 resulted in complete tumor regression at all dose concentrations. 100% survival was achieved after 103 days even at the two highest doses. Capsol® formulation VR-4 resulted in complete tumor regression in the highest three dose groups and 60% regression at 13.4 mg / kg / day. Survival after 103 days was somewhat lower for Formulation VR-4. Taxol® treatment at 30, 20 and 13.4 mg / kg / day showed survival rates of 40%, 20% and 20% after 103 days, respectively. Treatment of the standard vehicle did not show any effect on tumor growth and sacrificed animals after 33-47 days. The results are shown in Table 4.
Dosage (mg / ka / day) CR / gun TSF / TR DCR (days) Unspecified lethal / gun VR- VR- TAX VR-3 VR-4 TAX VR-3 VR-4 TAX VR- VR-4 TAX 45 5/5 5/5 NA 5/0 3/2 NA > 88 > 73 NA 0/5 0/5 NA 30 5/5 5/5 4/4 5/0 5/0 2/2 > 88 > 88 > 56 0/5 0/5 1/5 20 5/5 5/5 4/4 1/4 2/3 1/3 > 51 > 51 > 57 0/5 0/5 1/5 13 4/5 3/5 4/5 0/5 0/5 1/4 10 10 > 29 0/5 0/5 0/5
CR = complete tumor regression
TFS = Survivor Without Tumor
TR = tumor recurrence
DCR = Complete Regression Days
These unexpected and surprising results show increased potency for two capsules® formulations compared to Taxol. In addition, higher doses of paclitaxel are achieved in the Capsol® group due to the lower toxicity of the formulation. This high dose was administered as a concentrate injection.
<Example 60>
Hemodynamics and tissue distribution for 3 H-taxol® and capsol® after single intravenous dosing in rats
Formulated in Capsule3Two studies were performed to compare the pharmacokinetics and tissue distribution of H-paclitaxel and Taxol injection concentrations. 10 mg / kg to 14 male mice3H-taxol And 10 rats were injected intravenously with 4.9 mg / kg. 10 male rats above 5.1 mg / kg3ㅗㅗ H-Capsol® was injected intravenously.
The total concentration of both radioactivity and paclitaxel decreases by abnormalities after administration 5 ㎎ / ㎏ IV bolus of 3 H- Taxol (R) or 3 kapsol H- (R). However, the concentrations of both total radioactivity and paclitaxel are significantly lower after administration of similar 3 H-taxol® and after administration of 3 H-capsol®. These lower concentrations are more quickly distributed out of the blood.
HPLC profiles of the blood shows a similar pattern of metabolism of the 3 H- kapsol (R) and 3 highly polar metabolites for both H- taxol. However, the metabolic rate is markedly lower for 3 H-capsol as there is 44.2% of blood radioactivity 24 hours after paclitaxel dosing compared to 27.7% of 3 H-taxol. Table 5 shows the total radioactivity and hemodynamics for paclitaxel after IV administration of 3 H-Capsol® or 3 H-Taxol at 5 mg / kg.
cure AUC 0-24 (mg eq.hr/ml) Extrapolated C 0 (mg eq / ml) Observed C max (mg eq / ml) Observed T max (hr) t 1/2 β (hr) The total radioactive 3 H- kapsol (TM) 3 H- Taxol (TM) paclitaxel kapsol 3 H- (R) 3 H- Taxol 6.110.23.75.4 7.619.77.017.1 4.213.54.011.8 0.030.030.030.03 19.019.711.47.2
Tissue radioactivity concentrations are higher after 3 H-Capsol® administration than after 3 H-taxol administration to 12 of 14 tissues. The tissue / blood ppm ratio is higher in all tissues for animals dosed with 3 H-Capsol®, but the blood concentration is lower. This aids in the rapid distribution of 3 H-Capsol® from blood to tissues presented by hemodynamic data.
Formulated in Capsol®3H-paclitaxel is formulated in Taxol® for injection concentration3It shows a similar pharmacokinetic profile as H-paclitaxel, but the tissue / blood ppm ratio and metabolic rate are significantly different. Two minutes after administration of the blood sample, Taxol Total radioactivity for animals treated with significantly lower concentrations of Capsol® than for treated animals3It indicates a faster distribution of H-capsol out of the blood. However, metabolic rate3As there is 44% blood reactivity after 24 hours of paclitaxel dosing compared to 28% of H-taxol3H-capsol Significantly lower for (registered trademark).
This finding for Capsol® is surprising and provides new formulations for achieving the sustained activity of paclitaxel as compared to Taxol. Together with high local concentrations, such enhanced activity should exhibit increased efficacy in the treatment of metastases in primary tumors or organs with high local concentrations. The tissue distribution is shown in Table 6 below. The data represent the mean and standard deviation (Capsol® and Taxol®) of 10 mice in each group.
Tissue radioactivity residues of male rats expressed in ppm after one intravenous dose of 3 H-Capsol® and 3 H-Taxol® at 5 mg / kg Sample CAPSOL (registered trademark) Taxol (registered trademark) medium ± SD medium ± SD Brain Heart Lung Kidney Muscle Muscle Gastrointestinal Testis Pancreas Body Bone Spleen Prostate Semen Blood Serum Plasma 0.1060.3681.0061.1920.6700.4220.8020.2650.9630.5960.5310.9121.7281.1420.1310.131 0.0080.0630.1400.1280.1100.1200.2740.0230.3570.0700.1080.1310.3560.2530.0100.012 0.1450.2620.6941.370.4730.3860.8980.3260.4680.4410.2970.4931.101.200.1810.196 0.0200.0370.070.2040.0680.0350.2430.0470.0700.0650.0510.0700.1610.2370.0200.026
The data indicate that significantly higher concentrations of capsol® accumulate in the various organs compared to Taxol®. These organs include the prostate, pancreas, kidneys, lungs, heart, bones and spleen. Thus, Capsol® may be more effective than Taxol® at the same concentration of paclitaxel in the treatment of cancer of these organs.
Concentrations in prostate tissue are of particular interest in the treatment of prostate cancer. This surprising and unexpected result is closely related to the treatment of prostate cancer. Table 7 shows Taxol Data is shown for individual rats (10 in each group) showing increased accumulation of paclitaxel for capsol® in the prostate as compared to trademark. The basis for localization in the prostate is that preparations of protein albumin that can cause localization into prostate tissue via particle size (20-400 nm) or specific membrane receptors (gp 60, gp 18, gp 13, etc.) of the preparation. It may be the result of my presence. In addition, other biologically compatible, biodegradable polymers other than albumin may exhibit specificity for certain tissues that exhibit high local concentrations of paclitaxel in these tissues as a result of the properties described above, such as the prostate. Such biologically compatible materials may be included within the scope of the present invention. A preferred embodiment of the composition for achieving high local concentrations of paclitaxel in the prostate is a formulation comprising paclitaxel and albumin having a particle size of 20 to 400 nm and free of cremophores. This embodiment has also been demonstrated to show higher concentrations of paclitaxel in the pancreas, kidneys, lungs, heart, bones and spleen when compared to the same amount of Taxol®.
Data for 10 rats in each group dosed with 5 mg / kg paclitaxel CAPSOL (registered trademark) Taxol (registered trademark) 1.2282.4631.9041.8501.6601.2461.8951.5631.7981.576 1.131.040.9521.420.311.081.030.950.941.18 Average 1.728 Average 1.103 SD 0.36 SD 0.16
This data shows that the localization of Capsol® to prostate is about 150% compared to Taxol®.
This unexpected localization of paclitaxel to the prostate gland in a capsol® formulation may serve as a similar agent for the treatment of other disease states affecting such organs, for example, for prostatitis (inflammation and infection of the prostate gland). Therapeutic agents that can facilitate the delivery of other pharmacologically active agents to the prostate, such as antibiotics, will be formulated in a similar form to achieve high local delivery, which is effective for the treatment of benign prostatic hypertrophy. Similarly, the surprising finding that Capsol® provides high local concentrations in the heart can facilitate the treatment of atherosclerosis as well as restenosis. Paclitaxel has been demonstrated to be therapeutically effective in restenosis and prevention of atherosclerosis, and therefore Capsol® is the ideal vehicle in this condition. It has also been demonstrated that polymerized albumin selectively binds to the inflammatory endothelial tract, possibly via the gp 60, gp 18 and gp 13 receptors.
<Example 61>
Hemodynamics and Tissue Distribution of Paclitaxel after Intravenous Multiple Dosing in Mice
Studies with 3 H-Capsol® were supplemented by treating four additional groups of mice with concentrated mass doses of paclitaxel in capsules of 9.1, 26.4, 116.7 and 148.1 mg / kg. Blood was collected from the tail vein and AUC 0-24 calculated. Blood samples were collected and extracted at 24 hours and the extracts were injected on HPLC to determine the concentration of parent compound in the blood.
Table 8 shows the total radioactivity and hemodynamics for paclitaxel after IV administration of 3 H-Capsol®.
Group / dosage (mg / kg) AUC 0-24 (μg eq.hr/ml) Extrapolated C 0 (μg eq / ml) Observed C max (μg eq / mL) Observed T max (hr) t 1/2 β (hr) A / 9.1 11.5 10.2 7.19 0.03 22.3 B / 26.4 43.5 44.8 29.5 0.03 16.0 C / 116.7 248.9 644.6 283.3 0.03 8.48 D / 148.1 355.3 1009.8 414.2 0.03 9.34
As the dose of paclitaxel increased, the area under the curve increased proportionally. After 24 hours the concentration of the parent compound increased from 9 mg / kg to 148 mg / kg by a factor of 8.5 (0.04 ppm to 0.34 ppm).
<Example 62>
Toxicity of Capsol® and Taxol® after Intravenous Administration of Rats
The purpose of this study was to provide I.V. Toxicity of Capsol® after administration was measured. Capsol® was administered to 6 male and 6 female mice at doses of 5, 9, 30, 90 and 120 mg / kg. Half of the animals in each dose group were euthanized on day 8 and necropsied. The remaining animals were autopsied on day 31. The results of the Capsol® treated animals were compared with the results of the normal saline and vehicle standard groups as well as the results of animals treated with Taxol® of 5, 9 and 30 mg / kg.
Immediately after dosing, animals were examined 1 to 4 hours and 1 day after administration. Blood was collected from each animal for hematological and serum measurements prior to euthanasia.
Thirteen deaths occurred during the 30-day observation period. All 12 animals treated with Taxol® at a dose of 30 mg / kg paclitaxel were all killed on day 4. Only one animal treated with Capsol® was killed. Animals treated with Capsol® received 90 mg / kg and were killed on Day 15. No other animals treated with Capsol® are lethal at a dose of 90 kg or 120 mg / kg and therefore lethal is not considered to be related to treatment.
During the first four hours of observation, nipples and torsion were observed in the majority of animals treated with Taxol®, possibly due to the alcohol content of the drug. Nipples appeared in a small number of animals treated with Capsol®. Animals treated with Taxol® at a paclitaxel dose of 30 mg / kg observed hair drowsiness and lethality on day 4. Capsol® treated animals did not show any obvious signs of toxicity except several occurrences of hair growth at 90 mg / ml and 120 mg / ml dose concentrations.
No abnormalities were reported in capsol® treated animals. Overall autopsy results for 8 and 13 days were normal. Significant changes related to dosage were seen in the male reproductive organs of animals treated with Capsol®. Degeneration and phobia of epithelial cells of the epididymal ducts, often accompanied by invasion of multipathic interstitial lymphocytes, has been observed. As the dose of Capsol® increased, severe atrophy of the fertilized tubules increased in the testicles. In the opinion of the pathologist, significant damage was observed in the male reproductive organs of animals treated with 9, 30, 90 and 120 mg / kg capsules. These changes are associated with diffuse degeneration and necrosis of the testicles. This change was most prevalent in animals that received higher doses of Capsol®. Untreated standard animals, vehicle standard animals, or animals treated with Taxol® did not show any changes in the testes.
This finding is unexpected and has a significant therapeutic relevance for the treatment of hormone dependent cancers such as prostate cancer. Removal of the testicles (orchiectomy) is a therapeutic approach to the treatment of prostate cancer. Capsol® represents a novel formulation for achieving high local concentrations of paclitaxel at such sites, maintaining the activity of active raw materials, reducing the function of the testicles and treating these diseases without toxic cremophorous cysts. Thus, treatment with Capsol® causes a decrease in the concentrations of testosterone and other male hormones.
Cerebral cortex necrosis was present at median dose concentrations of Taxol® treated animals. This may explain the lethality of animals treated with higher doses of Taxol. No cerebral damage was seen in the animals treated with Capsol®.
This lack of cerebral or neurological toxicity is surprising and achieves high tissue doses of 5 to 120 mg / kg (equivalent to 30 to 700 mg / m 2 in humans) in the treatment of brain tumors and in rats. There is a significant association in both capabilities.
In summary, Capsol® was significantly less toxic than Taxol®. None of the animals treated with Taxol® at doses higher than 9 mg / kg survived. All animals that received Capsol® survived at doses up to 120 mg / kg, with the exception of accidental lethality at 90 mg / kg of Capsol®. There was an effect related to high doses of Capsol® on male reproductive organs and weight inhibition. Female rats did not demonstrate any toxic effects by administering Capsol® at 120 mg / kg or less. This high dose was administered in a concentrated mass dose and was equivalent to a dose of 30 to 700 mg / m 2 in humans.
<Example 63>
Pharmacokinetic (PK) data for cyclosporin nanoparticles (Capsorine I.V.) following intravenous administration (compared to Sandimune I.V. formulations available from Sandoz)
Cyclosporin nanoparticles (capsoline I.V.) prepared as described above (Examples 13 and 14) were restored in saline and administered to the first group of three Sprague Dawley mice as intravenous concentrates. The second group of three rats was administered after diluting sandimbium I.V. containing cremaphor / ethanol in saline. Each group received the same dose of cyclosporin of 2.5 mg / kg. Blood samples were taken at 0, 5, 15, 30 (minutes) and 1, 2, 4, 8, 24, 36 and 48 (hours). The concentration of cyclosporin in the blood was analyzed by HPLC and typical PK parameters were measured. The PK curves showed a typical decline for the following times.
Decline over time AUC, mg-hr / ml Cmax, ng / ml Capsorine I.V. 12,228 2,853 Sandim 뮨 I.V. 7,791 2,606
Also, Sandimeng I.V. Due to the toxicity of the formulation, two of the three rats in the herd were killed within 4 hours of dosing. Thus, the nanoparticle formulations according to the invention (capsorine I.V.) show greater AUC and no toxicity compared to commercially available formulations (Sandimmeop I.V.).
<Example 64>
Pharmacokinetic (PK) data for cyclosporin nano mucus (Capsorin Oral) compared to Neoral (commercially available from Sandoz) after oral administration
Cyclosporine nanomucus prepared as above was administered to orange juice to the first group of three Sprague Dawley rats by oral gavage. A microemulsion formulation containing a commercial emulsifier in the second group of three mice was also administered after dilution with orange juice by gavage. Each group received the same dose of cyclosporin of 12 mg / kg in the volume of the same orange juice. Blood samples were taken at 0, 5, 15, 30 (minutes) and 1, 2, 4, 8, 24, 36 and 48 (hours). The concentration of cyclosporin in the blood was analyzed by HPLC and typical PK parameters were measured. The PK curves showed a typical decline for the following times.
Decline over time AUC, mg-hr / ml Cmax, ng / ml Capsorine Oral 3,195 887 Neoral 3,213 690
Thus, the nanoslime formulation (Capsorine Oral) of the present invention exhibits similar PK behavior as the commercially available formulation (Neoral).
<Example 65>
Clinical investigation using capsol®: purpose and advantages
The rationale for selecting an initial dose for Phase I / II will be based on very low preliminary clinical toxicity data for the Capsol® formulation compared to Taxol®. The preliminary clinical data indicate that the initial dosage concentration of Capsol® for Phase I / II studies will use the MTD (maximum tolerated dose) established for paclitaxel for Taxol®.
Based on the current preliminary clinical data, the clinical purpose to be accepted in the market is to eliminate the need for preliminary drugs prior to the administration of paclitaxel, and to dose the same capsol® as the Taxol®, i.e. Determine the dosage at which the tumor response can be obtained, followed by continuous IV for paclitaxel administration The need for infusion (3-4 hours) is eliminated and replaced by administration over a shorter period (less than 1 hour or concentrated mass).
Capsol® formulations for paclitaxel have a number of effective advantages. Capsol® is a lyophilized powder containing only paclitaxel and human serum albumin. Due to the nature of the colloidal solution formed upon restoration of the lyophilized powder, toxic emulsifiers such as cremaphor (in the paclitaxel formulation of BMS) or polysorbate 80 (in the docetaxel formulation of Long Soul Lang), and ethanol to dissolve the drug No solvent, such as Removing toxic emulsifiers will reduce the occurrence of severe hypersensitivity reactions and anaphylaxis, which are known to occur from products such as Taxol®.
In addition, no preliminary medication with steroids and antihistamine prior to administration of the drug is preceded.
Due to the reduced toxicity demonstrated by the LD 10 / LD 50 study, higher doses are used to achieve greater efficacy.
A decrease in myelosuppression (compared to Taxol®) is expected to reduce treatment cycle duration (currently three weeks) and improve therapeutic outcomes.
Capsol® can be administered at higher concentrations (up to 20 mg / ml) compared to Taxol® (0.6 mg / ml), allowing for lower injection volumes and possibly intravenous concentrates It can be administered as.
A recognized problem with Taxol® is that paclitaxel precipitates in the underlying catheter tube. This is caused by a contradictory and incompletely controlled dosing in the upper leg. Due to the inherent stability of the colloidal solution of the new formulation, Capsol®, the problem of precipitation is alleviated.
The literature suggests that particles in the low hundreds of nm size range are selectively distributed to tumors through ducts that tend to leak at the location of the tumor. Thus, the colloidal particles of paclitaxel in the Capsol® formulations significantly reduce the side effects of paclitaxel administered in the BMS formulations and exhibit selective drug targeting effects.
Example 66
Overview of Clinical Trial Design of Capsol®
Indications: Metastatic Breast Cancer
Dosage Scheme: The rationale for selecting the initial dose for the Phase I / II trial is based on significantly lower preliminary clinical toxicity data for the Capsol® formulation compared to Taxol®. The single dose LD 10 in mice is measured at 398.1 mg / kg. Conversion of these doses to surface area criteria (three times the mg / kg value) is estimated at 1194.3 or about 1200 mg / m 2 . For humans, start with a dose of 120 mg / m 2 , which is a 1/10 dose of this value. However, paclitaxel is kapsol a dosage of 175 ㎎ / m, and safety at the dosage of 2, based on the pilot study with kapsol (R) showing a lower bone marrow suppression in mice, 175 ㎎ / m 2 (R It is already well established that the formulation should be stable. The Capsol® solution is delivered in about 15-30 minutes or less.
<Example 67>
Overview of the clinical development program of Capsol®:
Phase I / II Formulation Discovery Study / Limited Efficacy Test
Patient / Purpose: Patients with advanced breast metastases that are unresponsive to standard therapies. The purpose of this test is to establish the rate of response to Capsol® as a single agent in patients with metastatic breast cancer.
Dosing-Stage I Component: The initial dosage used for the Phase I component of the test is the maximum known dosage (MTD, 135 mg / m 2 ) known for paclitaxel. The dosage is then increased in 25% steps until the MTD is reached. There are three patients in each of the initial capsol® dose concentrations and six in MTD. The ability to move to the next dose concentration is based on the reaction pattern. That is, the study is discontinued whenever two or more of six patients at a particular dose concentration exhibit grade 3 non-myelosuppression or grade 4 myelosuppression (for WHO toxicity grades). Doses for Capsol® are designated as the dose immediately preceding the dose at which the test is discontinued. Separate schedules of drug administration, such as daily five-hour or 24-hour infusions, may be investigated as needed based on the results of the initial, single-dose concentrate mass plan.
Pharmacokinetics: For selected patients, complete pharmacokinetic studies are performed using serum collected at appropriately designated time points. Parameters such as t 1/2 (α, β steps), AUC, C max , intelligibility and partition volume are measured.
Patients-Stage II Components: After defining the MTD, patients with breast cancer similar to those used in the prototype paclitaxel test are selected as stage II components. The nominal number is based on the requirement to establish the rate of tumor response with acceptable accuracy in the 95% confidence interval. In such cases, the study is given a single purpose of establishing identity with standard paclitaxel by indicating a confidence interval that includes the expected rate of reaction for the capsol®. The size of the patient sample used is 30, which is common as the Phase II component of the Phase I / II study.
Measurement: The primary outcome is tumor response rate (CR / PR) for enrolled patients. In addition, the time, reaction duration and survival time for the reaction are checked. The stability of the treatment is also assessed from changes in reaction rates and standard experimental parameters.
权利要求:
Claims (65)
[1" claim-type="Currently amended] A method of reducing paclitaxel toxicity in a patient receiving paclitaxel treatment comprising systemic administration of the paclitaxel in a pharmaceutically acceptable formulation to a patient receiving paclitaxel at a dose of at least 175 mg / m 2 over a period of up to 2 hours.
[2" claim-type="Currently amended] The method of claim 1, wherein the dose is at least 250 mg / m 2.
[3" claim-type="Currently amended] The method of claim 1, wherein the dose is at least 325 mg / m 2.
[4" claim-type="Currently amended] The method of claim 1, wherein the administration period is 1 hour or less.
[5" claim-type="Currently amended] The method of claim 1, wherein the administration period is 30 minutes or less.
[6" claim-type="Currently amended] The method of claim 1, wherein the paclitaxel is administered orally, intramuscularly, intravenously, intraperitoneally or by inhalation.
[7" claim-type="Currently amended] The method of claim 1, wherein the treatment is for prostate cancer, testicular spinal surgery, pancreatic cancer or brain tumor.
[8" claim-type="Currently amended] The method of claim 1, wherein the hematological or neurological toxicity of paclitaxel is reduced.
[9" claim-type="Currently amended] Paclitaxel-free, pre-paclitaxel treatment, including systemic administration of paclitaxel in a pharmaceutically acceptable formulation to a patient in need of paclitaxel at a dose of at least 135 mg / m 2 over a period of up to 2 hours Paclitaxel administration to a patient in need thereof.
[10" claim-type="Currently amended] The method of claim 9, wherein the dose is at least 250 mg / m 2.
[11" claim-type="Currently amended] The method of claim 9, wherein the dose is at least 325 mg / m 2.
[12" claim-type="Currently amended] The method of claim 9, wherein the administration period is 1 hour or less.
[13" claim-type="Currently amended] The method of claim 9, wherein the administration period is 5 minutes or less.
[14" claim-type="Currently amended] 10. The method of claim 9, wherein the paclitaxel is administered orally, intramuscularly, intravenously, intraperitoneally, intraarterally, in urethra or in seconds.
[15" claim-type="Currently amended] 10. The method of claim 9, wherein the treatment is for prostate cancer, testicular spinal surgery, pancreatic cancer or brain tumor.
[16" claim-type="Currently amended] Paclitaxel treatment, comprising systemic administration of the paclitaxel in a pharmaceutically acceptable formulation to a patient in need of paclitaxel at a dosage cycle of no less than 3 weeks over a period of up to 2 hours at a dose of at least 135 mg / m 2 Paclitaxel administration method to patient with.
[17" claim-type="Currently amended] The method of claim 16, wherein the dose is at least 250 mg / m 2.
[18" claim-type="Currently amended] The method of claim 16, wherein said dose is at least 325 mg / m 2.
[19" claim-type="Currently amended] The method of claim 16, wherein the treatment cycle is less than two weeks.
[20" claim-type="Currently amended] The method of claim 16, wherein the treatment cycle is less than one week.
[21" claim-type="Currently amended] The method of claim 16, wherein the paclitaxel is administered orally, intramuscularly, intravenously or intraperitoneally.
[22" claim-type="Currently amended] 17. The method of claim 16, wherein said treatment is for prostate cancer, testicular spinal surgery, pancreatic cancer or brain tumor.
[23" claim-type="Currently amended] A method of administering paclitaxel to a patient in need of paclitaxel treatment comprising systemic administration of said paclitaxel in a dosage of at least 250 mg / m 2 in a pharmaceutically acceptable formulation to a patient in need of paclitaxel treatment.
[24" claim-type="Currently amended] The method of claim 23, wherein said dose is at least 325 mg / m 2.
[25" claim-type="Currently amended] The method of claim 23, wherein the treatment cycle is less than two weeks.
[26" claim-type="Currently amended] The method of claim 23, wherein the treatment cycle is less than one week.
[27" claim-type="Currently amended] The method of claim 23, wherein the paclitaxel is administered orally, intramuscularly, intravenously or intraperitoneally.
[28" claim-type="Currently amended] 24. The method of claim 23, wherein the treatment is for prostate cancer, testicular spinal surgery, pancreatic cancer or brain tumor.
[29" claim-type="Currently amended] A method of administering paclitaxel to a patient in need of paclitaxel comprising systemic administration of said paclitaxel in a formulation that can be safely administered to a patient in need of paclitaxel using medical hardware made from extractable ingredient-containing materials.
[30" claim-type="Currently amended] The method of claim 29, wherein the medical hardware is selected from the group consisting of tubes, catheters, infusion bags, bottles and syringes.
[31" claim-type="Currently amended] A method of administering paclitaxel to a patient in need of paclitaxel, comprising systemically administering the paclitaxel in a formulation that can be safely administered to a patient in need of paclitaxel without using an inline filter.
[32" claim-type="Currently amended] A method of administering paclitaxel to a patient in need of paclitaxel, comprising systemically administering the full dose of paclitaxel to a patient in need of paclitaxel in a volume of less than 250 ml.
[33" claim-type="Currently amended] 33. The method of claim 32, wherein said volume is less than 150 ml.
[34" claim-type="Currently amended] 33. The method of claim 32, wherein said volume is less than 60 ml.
[35" claim-type="Currently amended] A method of administering paclitaxel to a patient in need of paclitaxel, comprising systemically administering the paclitaxel to a patient in need of paclitaxel at a rate of 50 mg / m 2 / hour or more.
[36" claim-type="Currently amended] Paclitaxel formulation with reduced paclitaxel toxicity in a patient receiving paclitaxel treatment comprising paclitaxel in a pharmaceutically acceptable formulation suitable for systemic administration at a dose of at least 175 mg / m 2 over a period of up to 2 hours.
[37" claim-type="Currently amended] The formulation of claim 36, wherein said dosage is at least 250 mg / m 2.
[38" claim-type="Currently amended] The formulation of claim 36, wherein said dosage is at least 325 mg / m 2.
[39" claim-type="Currently amended] Suitable for administration to patients in need of paclitaxel that does not require pre-paclitaxel preparation, comprising paclitaxel in a dosage of 135 mg / m 2 or more over a period of up to 2 hours in a pharmaceutically acceptable formulation suitable for systemic administration. Paclitaxel formulation.
[40" claim-type="Currently amended] The formulation of claim 39, wherein said dosage is at least 250 mg / m 2.
[41" claim-type="Currently amended] The formulation of claim 39, wherein said dosage is at least 325 mg / m 2.
[42" claim-type="Currently amended] Paclitaxel formulations suitable for administration to patients in need of paclitaxel in less than 3 weeks of treatment, comprising paclitaxel in a pharmaceutically acceptable formulation suitable for systemic administration in a dosage of at least 135 mg / m 2 over a period of up to 2 hours .
[43" claim-type="Currently amended] The formulation of claim 42, wherein said dosage is at least 250 mg / m 2.
[44" claim-type="Currently amended] The formulation of claim 42, wherein said dosage is at least 325 mg / m 2.
[45" claim-type="Currently amended] A paclitaxel formulation suitable for administration to a patient in need of paclitaxel, comprising paclitaxel in a pharmaceutically acceptable formulation that does not contain cremaphore.
[46" claim-type="Currently amended] A lyophilized formulation of paclitaxel suitable for administration to a patient reconstituted and in need of paclitaxel.
[47" claim-type="Currently amended] A reconstituted formulation of paclitaxel suitable for administration to a patient in need of paclitaxel, comprising the lyophilized formulation of claim 41 and water or an aqueous solution.
[48" claim-type="Currently amended] A frozen formulation of paclitaxel suitable for administration to a patient in need of thawing and paclitaxel.
[49" claim-type="Currently amended] A liquid formulation of paclitaxel comprising paclitaxel and water having a concentration of at least 2.0 mg / ml.
[50" claim-type="Currently amended] The liquid formulation of paclitaxel according to claim 49, wherein said paclitaxel concentration is at least 5.0 mg / ml.
[51" claim-type="Currently amended] The liquid formulation of paclitaxel according to claim 49, wherein said paclitaxel concentration is at least 10.0 mg / ml.
[52" claim-type="Currently amended] A drug formulation suitable for administration by inhalation to a patient in need thereof comprising a drug nanoparticle of about 50 to 1,000 nm size and protein microparticles of about 1 to 10 μm size, optionally comprising an excipient.
[53" claim-type="Currently amended] a) combining the nonvolatile phase, the volatile phase containing the active agent and the surfactant which spontaneously forms a microemulsion, and
b) removing said volatile phase to obtain a suspension of solid nanoparticles in said non-volatile phase, wherein said active agent-containing method comprises a nanoparticle having an average diameter of less than 100 nm.
[54" claim-type="Currently amended] The method of claim 53, wherein the average diameter of the nanoparticles is less than 50 nm.
[55" claim-type="Currently amended] The method of claim 53, wherein the microemulsion further comprises a cosurfactant.
[56" claim-type="Currently amended] 54. The method of claim 53, further comprising c) removing the surfactant and / or cosurfactant by dialysis, ultrafiltration or adsorption.
[57" claim-type="Currently amended] 54. The method of claim 53, further comprising c) removing essentially all of the residual nonvolatile phase by freeze drying, spray drying, or lyophilization to obtain a dry powder of nanoparticles.
[58" claim-type="Currently amended] 59. The method of claim 57, further comprising d) resuspending the dry powder of nanoparticles in a pharmaceutically acceptable carrier.
[59" claim-type="Currently amended] 59. The method of claim 58, further comprising e) administering the resuspended nanoparticles to the patient.
[60" claim-type="Currently amended] 54. The method of claim 53, further comprising c) sterilizing the suspension by filtering the suspension of solid nanoparticles through a filter of sufficiently small pore size.
[61" claim-type="Currently amended] a) combining the non-volatile phase containing the active agent and the volatile phase spontaneously forming a microemulsion, and
b) removing said non-volatile phase to obtain a suspension of solid nanoparticles in said volatile phase, wherein said active agent-containing nanoparticles having an average diameter of less than 100 nm.
[62" claim-type="Currently amended] A suspension of nanoparticles prepared by the method of claim 53.
[63" claim-type="Currently amended] Dry nanoparticles prepared by the method of claim 57.
[64" claim-type="Currently amended] A suspension of nanoparticles prepared by the method of claim 58.
[65" claim-type="Currently amended] A suspension of nanoparticles prepared by the method of claim 61.
类似技术:
公开号 | 公开日 | 专利标题
Feng et al.2013|A critical review of lipid-based nanoparticles for taxane delivery
Dolatabadi et al.2015|Solid lipid nanoparticles as efficient drug and gene delivery systems: recent breakthroughs
Hou et al.2013|Phytosomes loaded with mitomycin C–soybean phosphatidylcholine complex developed for drug delivery
Mattheolabakis et al.2012|Nanodelivery strategies in cancer chemotherapy: biological rationale and pharmaceutical perspectives
Tarhini et al.2017|Protein-based nanoparticles: From preparation to encapsulation of active molecules
Rivas et al.2017|Nanoprecipitation process: From encapsulation to drug delivery
Li et al.2017|A review of the structure, preparation, and application of NLCs, PNPs, and PLNs
Wais et al.2016|Nanoformulation and encapsulation approaches for poorly water-soluble drug nanoparticles
Fang et al.2013|Nanostructured lipid carriers | for drug delivery and targeting
Marianecci et al.2014|Niosomes from 80s to present: the state of the art
Soukasene et al.2011|Antitumor activity of peptide amphiphile nanofiber-encapsulated camptothecin
Luo et al.2015|Nanomedical engineering: shaping future nanomedicines
Mehnert et al.2012|Solid lipid nanoparticles: production, characterization and applications
Pawar et al.2014|Engineered nanocrystal technology: in-vivo fate, targeting and applications in drug delivery
US20170007569A1|2017-01-12|Compositions and methods for preparation of poorly water soluble drugs with increased stability
Fahr et al.2007|Drug delivery strategies for poorly water-soluble drugs
US9993454B2|2018-06-12|Solid nanoparticle formulation of water insoluble pharmaceutical substances with reduced Ostwald ripening
Battaglia et al.2012|Lipid nanoparticles: state of the art, new preparation methods and challenges in drug delivery
ES2685436T3|2018-10-09|Compositions and procedures for administration of pharmacological agents
CN101485629B|2013-01-23|Drug delivery system and preparation method thereof
Souto et al.2010|Lipid nanoparticles: effect on bioavailability and pharmacokinetic changes
Poonia et al.2016|Nanostructured lipid carriers: versatile oral delivery vehicle
AU2006241145B2|2011-04-28|Surface-modified microparticles and methods of forming and using the same
US7179484B2|2007-02-20|Protein-stabilized liposomal formulations of pharmaceutical agents
EP0693924B2|2008-04-09|Methods for | delivery of biologics and compositions useful therefor
同族专利:
公开号 | 公开日
ES2435944T3|2013-12-26|
CN1515246A|2004-07-28|
DK1023050T3|2013-10-14|
US20080161382A1|2008-07-03|
WO1999000113A1|1999-01-07|
SG113402A1|2005-08-29|
IL222002A|2018-02-28|
PT1023050E|2013-12-04|
EP1023050A4|2006-08-30|
NO996433L|2000-02-14|
CN1267214A|2000-09-20|
NZ525580A|2004-08-27|
CA2294981C|2012-04-03|
EP1023050A1|2000-08-02|
JP5405527B2|2014-02-05|
CN100462066C|2009-02-18|
JP2002507976A|2002-03-12|
CA2294981A1|1999-01-07|
KR100923172B1|2009-10-22|
AU8266298A|1999-01-19|
BRPI9810945B1|2016-12-27|
HK1030543A1|2014-04-17|
CN100525748C|2009-08-12|
NO340319B1|2017-03-27|
NO332166B1|2012-07-09|
KR100789008B1|2007-12-26|
JP2011219482A|2011-11-04|
HU0003972A2|2001-08-28|
KR100904931B1|2009-06-29|
HU230338B1|2016-02-29|
IL133672D0|2001-04-30|
KR20050042507A|2005-05-09|
HU0003972A3|2002-10-28|
NO996433D0|1999-12-23|
NO20120338A1|2000-02-14|
EP1023050B1|2013-09-25|
NZ502500A|2002-03-28|
US20080160095A1|2008-07-03|
WO1999000113A9|1999-04-08|
BR9810945A|2002-02-05|
JP4865937B2|2012-02-01|
KR20070091051A|2007-09-06|
MX337149B|2016-02-15|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题
法律状态:
1997-06-27|Priority to US5102197P
1997-06-27|Priority to US60/051,021
1997-09-09|Priority to US08/926,155
1997-09-09|Priority to US08/926,155
1998-06-26|Application filed by 패트릭 순 쉬옹, 비보륵스 파마슈티칼스, 인크.
1998-06-26|Priority to PCT/US1998/013272
2001-02-26|Publication of KR20010014254A
2007-12-26|Application granted
2007-12-26|Publication of KR100789008B1
优先权:
申请号 | 申请日 | 专利标题
US5102197P| true| 1997-06-27|1997-06-27|
US60/051,021|1997-06-27|
US08/926,155|US6096331A|1993-02-22|1997-09-09|Methods and compositions useful for administration of chemotherapeutic agents|
US08/926,155|1997-09-09|
PCT/US1998/013272|WO1999000113A1|1997-06-27|1998-06-26|Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof|
[返回顶部]