WO2022246055A1 - Procédé d'optimisation d'une recette de remplissage pour un récipient de médicament - Google Patents

Procédé d'optimisation d'une recette de remplissage pour un récipient de médicament Download PDF

Info

Publication number
WO2022246055A1
WO2022246055A1 PCT/US2022/030014 US2022030014W WO2022246055A1 WO 2022246055 A1 WO2022246055 A1 WO 2022246055A1 US 2022030014 W US2022030014 W US 2022030014W WO 2022246055 A1 WO2022246055 A1 WO 2022246055A1
Authority
WO
WIPO (PCT)
Prior art keywords
degrees
vial
pump
fill
setting
Prior art date
Application number
PCT/US2022/030014
Other languages
English (en)
Inventor
Devrishi GOSWAMI
Jun Zhang
Deirdre PIEDMONTE
Vikashni PADMAKUMAR
David Le
Maria Raquel SANTOS
Wei Qi
Original Assignee
Amgen Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amgen Inc. filed Critical Amgen Inc.
Priority to KR1020237038066A priority Critical patent/KR20240011135A/ko
Priority to US18/288,549 priority patent/US20240208680A1/en
Priority to CN202280035596.1A priority patent/CN117320964A/zh
Priority to BR112023024278A priority patent/BR112023024278A2/pt
Priority to MX2023013640A priority patent/MX2023013640A/es
Priority to JP2023569877A priority patent/JP2024523779A/ja
Priority to AU2022279223A priority patent/AU2022279223A1/en
Priority to IL307418A priority patent/IL307418A/en
Priority to CA3217207A priority patent/CA3217207A1/fr
Priority to EP22731368.1A priority patent/EP4341161A1/fr
Publication of WO2022246055A1 publication Critical patent/WO2022246055A1/fr

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B65CONVEYING; PACKING; STORING; HANDLING THIN OR FILAMENTARY MATERIAL
    • B65BMACHINES, APPARATUS OR DEVICES FOR, OR METHODS OF, PACKAGING ARTICLES OR MATERIALS; UNPACKING
    • B65B3/00Packaging plastic material, semiliquids, liquids or mixed solids and liquids, in individual containers or receptacles, e.g. bags, sacks, boxes, cartons, cans, or jars
    • B65B3/003Filling medical containers such as ampoules, vials, syringes or the like
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B65CONVEYING; PACKING; STORING; HANDLING THIN OR FILAMENTARY MATERIAL
    • B65BMACHINES, APPARATUS OR DEVICES FOR, OR METHODS OF, PACKAGING ARTICLES OR MATERIALS; UNPACKING
    • B65B3/00Packaging plastic material, semiliquids, liquids or mixed solids and liquids, in individual containers or receptacles, e.g. bags, sacks, boxes, cartons, cans, or jars
    • B65B3/04Methods of, or means for, filling the material into the containers or receptacles
    • B65B3/10Methods of, or means for, filling the material into the containers or receptacles by application of pressure to material
    • B65B3/12Methods of, or means for, filling the material into the containers or receptacles by application of pressure to material mechanically, e.g. by pistons or pumps
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B65CONVEYING; PACKING; STORING; HANDLING THIN OR FILAMENTARY MATERIAL
    • B65BMACHINES, APPARATUS OR DEVICES FOR, OR METHODS OF, PACKAGING ARTICLES OR MATERIALS; UNPACKING
    • B65B3/00Packaging plastic material, semiliquids, liquids or mixed solids and liquids, in individual containers or receptacles, e.g. bags, sacks, boxes, cartons, cans, or jars
    • B65B3/26Methods or devices for controlling the quantity of the material fed or filled
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B65CONVEYING; PACKING; STORING; HANDLING THIN OR FILAMENTARY MATERIAL
    • B65BMACHINES, APPARATUS OR DEVICES FOR, OR METHODS OF, PACKAGING ARTICLES OR MATERIALS; UNPACKING
    • B65B3/00Packaging plastic material, semiliquids, liquids or mixed solids and liquids, in individual containers or receptacles, e.g. bags, sacks, boxes, cartons, cans, or jars
    • B65B3/26Methods or devices for controlling the quantity of the material fed or filled
    • B65B3/28Methods or devices for controlling the quantity of the material fed or filled by weighing

Definitions

  • the present disclosure generally relates to filling recipes for drug containers and, more particularly, to a universal method of optimizing a filling recipe for a drug container.
  • existing fill recipes are individually tailored to a single drug product, each of which has a unique filling process and set of operating parameters.
  • a pump in a manufacturing facility carrying out the filling recipe must be calibrated each time a new filling recipe is needed for a new drug product.
  • the time involved to calibrate the pump or related equipment in a manufacturing facility before beginning the filling process according to a particular filling recipe is often lengthy. For example, there are typically many cycles needed, such as multiple number of strokes of the pump, until the pump is able to operate according to a particular filling recipe.
  • a method of filling a vial comprises providing a pump corresponding to a vial, and setting a drip retraction parameter for the pump to any value equal to or less than 20 degrees.
  • the method further comprises setting a no adjustment limit for a fill weight of the vial to T1 , with T 1 being at or in a range of about 2% more or less than a fill weight of a target fill weight TO, and wherein a process performance index Cpk (Cpk) for the vial throughout a fill cycle exceeds a minimum value.
  • a method of filling a plurality of vials of a nested syringe and vial line comprises providing a plurality of pumps corresponding to a plurality of vials of a nested syringe and vial line, and setting a drip retraction parameter for each pump in the plurality of pumps to any value equal to or less than 20 degrees.
  • the method also includes filling each vial of the plurality of vials with a drug product via a corresponding pump of the plurality of pumps; and wherein a Cpk for each vial of the plurality of vials exceeds a minimum value throughout a fill cycle.
  • a method of optimizing a filling recipe for a nested syringe and vial line comprises setting a drip retraction parameter for at least one pump in an offline manufacturing system corresponding to any value equal to or less than 20 degrees, and monitoring a performance of the at least one pump with the drip retraction parameter of the at least one pump set to any value equal to or less than 20 degrees.
  • the method also includes obtaining at least a minimum value for a Cpk for the at least one container throughout at least one fill cycle and for at least one drug product using the at least one pump in the offline manufacturing system; and finalizing a filling recipe for a nested syringe and vial line using data from fill cycles of the at least one drug product using the at least one pump in the offline manufacturing system.
  • a method of filling a vial may comprise providing a pump corresponding to a vial, setting a drip retraction parameter for the pump to any value equal to or less than 20 degrees, and setting a no adjustment limit for a fill weight of the vial to any value within a range of a target fill weight TO and T1 , with T 1 being at or in a range between the target fill weight TO and T2. So configured, a minimum value for a process performance index Cpk (Cpk) for the vial throughout a fill cycle is exceeded.
  • Cpk process performance index
  • setting a drip retraction parameter for the pump to any value equal to or less than 20 degrees may comprise setting the drip retraction parameter for the pump to one of 10 degrees, 20 degrees, or any value in a range of 10 degrees to 20 degrees.
  • the method may further comprise setting an end drip retraction value to 290 degrees when setting the drip retraction parameter to 20 degrees or setting an end drip retraction value to 280 degrees when setting the drip retraction parameter to 10 degrees.
  • providing a pump corresponding to a vial may comprise providing a pump corresponding to a vial of a nested syringe and vial line.
  • providing a pump corresponding to a vial may comprise providing one or more of a first fill set or a second fill set, the first fill set including a peristaltic pump filling assembly having a needle with an outer diameter of about 2.0mm, and the second fill set including a peristaltic pump filling assembly having a needle with an outer diameter of about 3.0mm.
  • a Cpk for the vial exceeds a minimum value throughout a fill cycle may comprise one or more of: (1) the Cpk for the vial exceeds a value of 1.33; or (2) the Cpk for the vial exceeds the minimum value during a temperature range throughout the fill cycle, the temperature range is one of: (1) 5 (+/-3) degrees Celsius; (2) 20 (+/-5) degrees Celsius; or (3) 10 to 19 degrees Celsius.
  • the method may further comprise filling the vial with a drug product via the pump, wherein the drug product has one or more of the following characteristics: (a) a density in a range of about 1.0-1.2 g/cm 3 ; (b) a viscosity in a range of about 1.0-10.0 cP; and (c) a surface tension in a range of about 40.0-72.7 mN/m.
  • the drug product has a density in a range of 1.0-1.2 g/cm3; a viscosity in a range of about 1.0-10.0 cP; and a surface tension in a range of about 40.0- 72.7 mN/m.
  • the drug product comprises a biologic drug (e.g., peptides, mAb, siRNA) or a small molecule drug.
  • the method may further comprise monitoring a performance of the filling recipe in the nested syringe and vial line and obtaining at least a minimum value for the Cpk for the at least one container for each pump in a plurality of pumps in the nested syringe and vial line.
  • FIG. 1 is a schematic representation of one embodiment of an offline filling system utilizing a filling recipe of the present disclosure
  • FIG. 2A is a perspective view of a filler of the system of FIG. 1 ;
  • FIG. 2B is a portion of the filler of FIG. 2A;
  • FIG. 2C is a perspective view of a fill set of the system of FIG. 1 ;
  • FIG. 2D is a perspective view of another fill set of the system of FIG. 1 ;
  • FIG. 2E is a perspective view of an exemplary fill target of the system of FIG. 1 ;
  • FIG. 3A is a chart depicting parameters of each of the fill sets of FIG. 1;
  • FIG. 3B is a chart depicting in process control parameters of the fill sets of FIG. 1 ;
  • FIG. 3C is a chart depicting product characteristics of drug products for use with the methods of the present disclosure.
  • FIG. 4 is a flow chart depicting a filling recipe parameter optimization strategy relative to the filling recipe of the present disclosure
  • FIG. 5A is a perspective view of fill performance results of a vial after filling at various drip retraction values
  • FIG. 5B is a chart depicting aspects of an exemplary fill recipe used with the fill performance results of FIG. 5A;
  • FIG. 6A depicts an exemplary filling recipe according to one aspect of the present disclosure
  • FIG. 6B is a graph depicting filling performance results of the filling recipe of FIG. 6A;
  • FIG. 6C is a chart depicting the filling performance results of the graph of FIG. 6B;
  • FIG. 7A is a chart depicting needle parameters of a needle used with a filling recipe of the present disclosure
  • FIG. 7B is a chart depicting pump parameters of at least one pump used with a filling recipe of the present disclosure
  • FIG. 8A is a graph depicting filling performance results of the filling recipe of the present disclosure in a temperature range of 10 - 11 degrees Celsius and using a second fill set of the system of FIG. 1;
  • FIG. 8B is a chart depicting the filling performance results of the graph of FIG. 8A;
  • FIG. 9A is a graph depicting the filling performance results of the filling recipe of the present disclosure at a temperature of about 19 degrees Celsius and using the second fill set of the system of FIG. 1;
  • FIG. 9B is a chart depicting the filling performance results of the graph of FIG. 9A;
  • FIG. 10A is a graph depicting the filling performance results of the filling recipe of the present disclosure at a temperature of about 10-11 degrees Celsius and using the first fill set of the system of FIG. 1;
  • FIG. 10B is a chart depicting the filling performance results of the graph of FIG. 10A;
  • FIG. 11A is a graph depicting the filling performance results of the filling recipe of the present disclosure at a temperature of about 19 degrees Celsius and using the first fill set of the system of FIG. 1;
  • FIG. 11 B is a chart depicting the filling performance results of the graph of FIG. 11 A;
  • FIG. 12 is a schematic representation of a manufacturing line in a manufacturing plant using the optimized filling recipe of the present disclosure
  • FIG. 13A is a perspective view of a nested syringe and vial line of the manufacturing line of FIG. 12;
  • FIG. 13B is a schematic view of the nested syringe and vial line of FIG. 13A;
  • FIG. 13C is a perspective view of a plurality of pumps corresponding to a plurality of vials of the nested syringe and vial line of FIGS. 13A-13B;
  • FIG. 14 is a chart depicting filling performance results of the filling recipe of the present disclosure in at least one of the nested syringe and vial line of FIG. 12;
  • FIG. 15 is a chart depicting filling performance results of a previously used filling recipe used in a nested syringe and vial line.
  • an efficient filling recipe for filling formulations comprising therapeutic proteins in a nested syringe and vial line is disclosed.
  • the universal filling recipe include a drip retraction parameter optimization within a specified value, which results in a significantly more efficient fill recipe compared to other existing, known filling recipes.
  • the new filling recipe of the present disclosure may be used with many different drug products and results in 95% reduction in fill weight optimization cycles. This led to an improvement of 10-30% increase utilization of a nested syringe and vial line fill time and potentially saving a significant number of units, such as vials, from rejection.
  • the drug products referred to herein comprise therapeutic proteins, such as monoclonal antibodies, as explained more below.
  • the offline manufacturing system 10 is a small scale bench set-up in a pilot facility, which made various tests related to the new filling recipe easier to assess and update based on test results for various attempted recipes, for example.
  • the offline manufacturing system 10 includes a filler 12, a first fill set 14, and a second fill set 16.
  • the first fill set 14 includes a corresponding first fill target 18, such as a vial
  • the second fill set 16 likewise includes a corresponding second fill target 20, also such as a vial.
  • the filler 12 is a Bausch+Strobel (B&S) scale-down filler in which recipe optimization, including many experiments, were conducted before arriving at the optimal, universal filling recipe of the present disclosure. While the specific B&S scale-down filler was used, it will be appreciated that various other fillers may also or alternatively be used.
  • B&S scale-down filler was used, it will be appreciated that various other fillers may also or alternatively be used.
  • each of the first and second fill targets 18, 20 are referred to as a vial in one example, it will be understood that the fill targets 18, 20 may alternatively and more generally be any other similar drug container and still fall within the scope of the present disclosure.
  • the optimal filling recipe selected from the recipe optimization from the scale-down filler 12 and first and second fill sets 12, 16 is transferable to a manufacturing line, such as a nested syringe and vial line in FIGS. 13A and 13B.
  • FIGS. 2A-2E a perspective view of each of the filler 12, first and second fill sets 14, 16 and first and second fill targets 18, 20 of FIG. 1 is depicted.
  • the filler 12 is a Bausch+Strobel Filler, a bench scale down filler, and more generally a developmental filler to support clinical and commercial manufacturing, as explained more below.
  • Exemplary containers that may be used with this filler include bulk ISO 2R, 6R, 20R, 3cc, 5cc, 10cc, 20cc vials, bulk 1mL glass and 1 mL plastic syringes, and bulk 5cc plastic cartridges.
  • the filler 12 includes a dosing vessel 21, a pump 22, such as a peristaltic pump, and a product bag 23.
  • FIG. 2B depicts a portion of the filler 12 of FIG. 2A.
  • the pump 22 is depicted cooperating with a fill target, such as the first fill target 18 or the second fill target 20.
  • the first and second fill targets 18, 20 are the same vial, but may also be any other container and still fall within the scope of the present disclosure.
  • the first filling set 14 of FIG. 1 is depicted.
  • the first filling set 14 is a peristaltic pump and includes a bag 27, tubing 28A, and a needle 29A.
  • the tubing 28A is coupled to the bag 27 at one end and the fill target 18 at the other end, such that the fluid in the bag is able to be drawn into the tubing and to the fill target 18 by way of the needle 29A, for example.
  • the outer diameter of the needle 29A is 2.0m
  • the inner diameter of the needle is 1 6mm
  • the inner diameter of the pump tubing 28A is 1.6 mm.
  • the tubing 28A branches into two tubing and converges again.
  • FIG. 2D depicts the second fill set 16 of FIG. 1.
  • the second fill set 16 is a peristaltic pump and includes the bag 27, tubing 28B, and a needle 29B, which is different from the needle 29A of the first fill set 14.
  • the tubing 28B is again coupled to the bag 27 at one end and the second fill target 20 at the other end, such that fluid in the bag is able to be drawn through the tubing and into the second fill target 20 by way of the needle 29B.
  • the tubing 28B again branches into two tubing and then converges again, but the two tubing have a different inner diameter.
  • the inner diameter of the tubing 28B is 1 6mm and 3.2 mm, respectively.
  • the outer diameter of the needle 29B is 3.0mm
  • the inner diameter of the needle is 2.6mm
  • the inner diameter of the pump tubing is 1.6 mm.
  • the exemplary fill target may include the first and second fill targets 18, 20 of FIG. 1, for example.
  • the first and second fill targets 18, 20 include a 1.3 mL fil in ISO 2R vial.
  • the target weight is 1.365 grams
  • T2+ is 0.05grams, which is 1.415 grams
  • T2- is 0.05 grams, which is 1.315 grams.
  • T1+ is 0.03 grams, which is 1.395 grams
  • T1- is 0.03 grams, which is 1.335 grams.
  • the net weight no adjustment limit + is 0.02 grams, which is 1.385 grams
  • the net weight no adjustment limit - is 0.02 grams, which is 1.345 grams.
  • the first fill set 14 is a peristaltic pump filling assembly having a needle with an outer diameter of 2.0mm and an inner diameter of 1 6mm.
  • the peristaltic pump filling assembly includes tubing (in FIG. 2C) having an inner diameter of 1 6mm.
  • the chart in FIG. 3A also includes information about the second fill set 16, which in this example also includes a peristaltic pump filling assembly having a needle with an outer diameter of 2.0mm and an inner diameter of 2.6mm.
  • the second fill set 16 also includes tubing having an inner diameter of 1 6mm, and another tubing having an inner diameter of 3.2mm, as mentioned above.
  • a no adjustment limit for a fill weight of the second fill target 20, such as the vial was set to T1 , with T 1 being at or in a range of about 2% more or 2% less than a fill weight of a target fill weight TO.
  • the target fill weight TO includes a volume of 1.3 mL and a mass of 1.365 grams.
  • the no adjustment limit was 80% of T1 , which may be +/- 0.02 grams of T1 +/- 0.03 grams.
  • the fill target mass of a vial in which no adjustment is needed is any value in the range of 1.345 grams to 1.385 grams in this example.
  • the no adjustment limit for a fill weight of the vial, such as the second fill target 20 may be set to any value within a range of a target fill weight TO and T1 , with T 1 being at or in a range between the target fill weight TO and T2 based on process performance, for example. In some examples, T 1 is set at 2%, but may be changed.
  • the filling recipe of the present disclosure includes filling a vial, such as the vials of the first and second filling targets 18, 20, with a drug product via a pump, and the drug product includes a mAb formulation.
  • the mAb formulations used are drug product 1 (DP1) and drug product 1 (DP2) As provided in the chart, at 5 degrees Celsius the density of DP1 is 1.055 g/cm 3 and the viscosity is 4.857.
  • the density of DP1 is 1.05 g/cm 3 , the viscosity is 2.604 cP, and the surface tension is 41.63 mN/m .
  • the density of DP2 is 1.054 g/cm 3 and the viscosity is 4.07 cP.
  • the density of DP2 is 1.049 g/cm 3 , the viscosity is 2.19 cP, and the surface tension is 43.716 mN/m .
  • the drug product used in the filling recipe is a mAb formulation including one or more of: (1) a density in a range of either about 1.054-1.055 g/cm 3 at 5 degrees Celsius or about 1.049-1.05 g/cm 3 at 25 degrees Celsius;
  • the method disclosed herein can be used to fill any liquid drug product, such as drug products comprising a biologic drug (e.g., peptides, mAbs, siRNAs) and a small molecule drug, provided the drug product has a specified physical parameter range.
  • the drug product may include one or more of the following characteristics: (1) a density in a range of about 1.0-1.2 g/cm 3 ; and/or (2) a viscosity in a range of about 1.0-10.0 cP; and/or (3) a surface tension in a range of about 40.0-72.7 mN/m.
  • preferred ranges for the viscosity are one or more of 1.0-8.0 cP, 1.0-6.0 cP,
  • the drug product has a density in a range of 1.0-1.2 g/cm 3 ; a viscosity in a range of about 1.0-10.0 cP; and a surface tension in a range of about 40.0-72.7 mN/m.
  • the drug product has one or more of a density in a range of 1.0-1.2 g/cm 3 and a viscosity in a range of about 1.0-10.0 cP, and any value for a surface tension.
  • the determining factors for the filling recipe are the density and the viscosity of the drug product, and the filling recipe may work with any surface tension. It will be understood that a drug product meeting any of these parameters may be used with the methods and filling recipe of the present disclosure.
  • manufacturing data suggests that a drug product with a viscosity of about 8.0 cP or more worked well with a drip retraction value of 20 degrees.
  • the preferred density of the drug product is about 1.0-1.1 g/cm 3 . It will be appreciated that many other values within the density, viscosity, and surface tension ranges provided above may be used for the drug products used with the methods and filling recipes of the present disclosure and fall within the scope of the present disclosure.
  • step 30 an initial optimization based on an existing filling recipe, which is referred to as existing filling recipe No. 1, was conducted on the second fill set 16.
  • step 32 with this initially optimized DP1 filling recipe for the second fill set 16, a range of drip retraction parameters ranging from 0 degrees to 45 degrees was tested.
  • step 34 another existing filling recipe, referred to as existing filling recipe No. 2, was started and a range of drip retraction parameters from 10 degrees to 20 degrees was tested.
  • step 36 was then developed in step 36 and various drip retraction parameters were also tested, including 5 degrees, 10 degrees and 20 degrees.
  • step 38 the hybrid recipe was finalized for the second fill set 16 with a drip retraction parameter set at 20 degrees.
  • step 40 the same filling recipe was used for the first fill set 14 and drip retraction parameters were set at 10 degrees and 20 degrees. Based on the fill performance, a filling recipe for the first fill set 14 was finalized with a drip retraction parameter set at 20 degrees.
  • fill performance results of the second fill target 20, such as the vial, of the second fill set 16 after utilizing a filling recipe with various drip retraction values is provided.
  • a test was conducted and the fill performance of the vial monitored when the drip retraction parameter of the filling recipe for the pump was set to each of 0 degrees, 10 degrees, 20 degrees, 30 degrees, 40 degrees, and 45 degrees.
  • an air gap AG in the fill target 20 or vial increased with increasing drip retraction parameter leading to an air liquid bilayer, explaining a likely reason for poor fill performance when a higher drip retraction parameter is set in the filling recipe.
  • FIG. 5A an air gap AG in the fill target 20 or vial increased with increasing drip retraction parameter leading to an air liquid bilayer, explaining a likely reason for poor fill performance when a higher drip retraction parameter is set in the filling recipe.
  • the start pump dosing was set at 40 degrees
  • the pump dosing start ramp was set at 90 degrees
  • the pump dosing stop ramp was set at 210 degrees
  • the end pump dosing was set at 260 degrees
  • the end drip retraction was set at 310 degrees
  • the distance run per dose was 766 degrees.
  • the start pump dosing was set at 40 degrees
  • the pump dosing start ramp was set at 90 degrees
  • the pump dosing stop ramp was set at 210 degrees
  • the end pump dosing was set at 260 degrees
  • the end drip retraction was set at 290 degrees
  • the distance run per dose was 766 degrees.
  • the impact of the drip retraction parameter set at 20 degrees on fill performance of DP1 in the second fill set 16 is provided.
  • the minimum filling weight was 1.348 grams and the maximum filling weight was 1.384 grams, making the average fill weight 1.366 grams, with a standard deviation of 0.006.
  • the process performance index Cpk value was 2.69, which is considerably higher than experiments using a filling recipe with the same parameters except for the drip retraction parameter of 45 degrees. Said another way, the process performance index was much higher when the drip retraction parameter was set to 20 degrees compared to higher values, such as 45 degrees, further indicating the impact of the drip retraction parameter on the fill performance.
  • the process performance index Cpk provides a value indicative of the efficiency of a particular process.
  • the process performance index value Cpk relates to how close an actual fill weight of a container, such as a vial, is to a target fill weight.
  • the process performance index value Cpk also relates to how close each subsequent fill rate of the additional containers, e.g., vials, are to each other. If there is a high number of the process performance index value Cpk, a given pump is providing an optimal performance. Likewise, a low number for the process performance index value Cpk indicates the pump is poorly performing.
  • the filling recipe 50 includes specific needle parameters, including setting a needle setting dimension to one of 134.5 mm and 39.0 mm, setting a basic needle position to 7.0 mm, and setting the start needle down to 25 degrees.
  • the needle parameters for the filling recipe 50 also include setting the needle at a dosing start of 10 mm, setting the needle down to one of 60 degrees and 23 degrees, and setting the needle up at 125 degrees.
  • the filling recipe 50 also includes setting the needle at the end of dosing to 13.0 mm and 310 degrees, setting the start needle to cut-off position of 315 degrees, setting the cut off position reached to 13.0 mm and 315 degrees, setting the start needle to a basic position of 315 degrees, and the basic needle position reached to 359 degrees.
  • the finalized filling recipe 50 of the present disclosure also includes setting several parameters for the pump.
  • the filling recipe 50 includes setting the start pump dosing to 40 degrees, setting the pump dosing start ramp to 90 degrees, setting the pump dosing stop ramp to 210 degrees, and setting the end pump dosing to 260 degrees.
  • the filling recipe includes setting the drip retraction parameter to 20 degrees, such as the pump distance run for the drip retraction to 20 degrees, the end drip retraction parameter to 290 degrees, and the distance run per dose parameter to 766 degrees.
  • the filling recipe 50 may include setting the drip retraction parameter to any value equal to or less than 20 degrees and still fall within the scope of the present disclosure.
  • the lowest value of the drip retraction parameter is 0 degrees.
  • the filling recipe 50 may include setting the drip retraction parameter for the pump to one of 10 degrees, 20 degrees, or any value in a range of 10 degrees to 20 degrees.
  • the filling recipe 50 may include setting the end drip retraction to 290 degrees when the drip retraction parameter is set to 20 degrees, or setting the end drip retraction value to 280 when setting the drip retraction parameter to 10 degrees, for example.
  • the finalized filling recipe 50 also includes setting a no adjustment limit for the fill weight of any vial to T1 , with T 1 being at or in a range of about 2% more or 2% less than a fill weight of a target fill weight TO, as explained above relative to FIG. 3B, for example.
  • the same filling recipe 50 was finalized for the exemplary drug product DP1 in the second fill set 16
  • the same filling recipe 50 may also be used for the first fill set 14 using DP1 or other drug products in the mAb formulation programs, for example.
  • the same finalized filling recipe 50 is also effectively used for a nested syringe and vial line of a manufacturing plant.
  • the method of optimizing a filling recipe for a nested syringe and vial line or other manufacturing line in a manufacturing plant includes using the first and second fill sets 14, 16 of the offline manufacturing system 1 of FIG. 1, for example.
  • FIGS. 8A and 8B the impact of temperature on fill performance of the second fill set 16 was evaluated for efficacy and the results are provided in the graph of FIG. 8A and the table of FIG. 8B.
  • the temperature was set at 10-11 degrees Celsius and the process performance parameter Cpk during this temperature range of 10-11 degrees Celsius throughout the fill cycle well exceeded a minimum value of 1.33.
  • the cycle included 103 fills, with a minimum fill weight of 1.340 g, a maximum fill weight of 1.393 g, and an average fill weight of 1.362 g with a standard deviation of 0.008 grams.
  • the process performance parameter Cpk value was 1.95, well above the target minimum value of 1.33, for example. While FIG. 8A shows that occasional filling weights were close to the T 1 limits at this temperature range, subsequent fill weights were able to return close to the target without significantly impacting the process performance index Cpk.
  • FIGS. 9A and 9B the impact of a temperature higher than 10-11 degrees Celsius (as in FIGS. 8A and 8B) on fill performance of the second fill set 16 was evaluated, and the results are provided in the graph of FIG. 9A and the table of FIG. 9B.
  • the temperature was set at about 19 degrees Celsius, and the process performance parameter Cpk during this temperature of about 19 degrees Celsius throughout the fill cycle well exceeded a minimum value of 1.33.
  • the cycle included 103 fills, with a minimum fill weight of 1.348 g, a maximum fill weight of 1.384 g, and an average fill weight of 1.366 g with a standard deviation of 0.006 grams.
  • process performance parameter Cpk against T2 was 2.69, well above both the target minimum value of 1.3, and the process performance parameter Cpk for the fill performance at the lower temperature of 10-11 degrees Celsius, in this example. This indicates an even better fill performance for the finalized filling recipe at slightly higher temperatures.
  • the impact of a temperature on fill performance of the first fill set 14 was evaluated, and the results are provided in the graph of FIG. 10A and the table of FIG. 10B.
  • the temperature was set at about a range of 10-11 degrees Celsius, and the process performance parameter Cpk during this temperature throughout the fill cycle well exceeded a minimum value, such as 1.33.
  • the cycle included 100 fills, with a minimum fill weight of 1.354 g, a maximum fill weight of 1.380 g, and an average fill weight of 1.365 g with a standard deviation of 0.005 grams.
  • the process performance parameter Cpk against T2 was 3.16, well above each of the target minimum value of 1.33, and the process performance parameter Cpk for the fill performance of the second fill set 16 at both the lower temperature of 10-11 degrees Celsius (FIGS. 8A and 8B) and the higher temperature 19 degrees Celsius (FIGS. 9A and 9B), for example.
  • these results further show the universal nature and applicability of the finalized filling recipe on different fill sets, and this filling recipe may also be successfully transferred and used with a nested syringe and vial line of a manufacturing plant, for example.
  • FIGS. 11 A and 11 B the impact of a temperature higher than 10-11 degrees Celsius (as in FIGS. 10A and 10B) on fill performance of the first fill set 14 was evaluated, and the results are provided in the graph of FIG. 11A and the table of FIG. 11 B.
  • the temperature was set at about 19 degrees Celsius and the process performance parameter Cpk during this temperature of about 19 degrees Celsius throughout the fill cycle well exceeded a minimum value of 1.33.
  • the cycle included 120 fills, with a minimum fill weight of 1.352 g, a maximum fill weight of 1.373 g, and an average fill weight of 1.362 g with a standard deviation of 0.004 grams.
  • the process performance parameter Cpk against T2 was 3.61 , well above both the target minimum value of 1.33, and the process performance parameter Cpk of 3.16 for the fill performance at the lower temperature of 10-11 degrees Celsius, for example.
  • the manufacturing line 102 includes at least one nested syringe and vial line 104 in which the efficient universal filling recipe 50 of the present disclosure is effectively used.
  • the nested syringe and vial line 104 is a nested syringe and vial line (NSVL) having a plurality of vials 105, such as ISO 2R RTU vials.
  • NSVL nested syringe and vial line
  • the filling recipe 50 of the present disclosure may be utilized in a variety of other nested syringe and vial lines and still fall within the scope of the present disclosure.
  • the vial 105 may more generally be any container 105, such as a syringe, and still fall within the scope of the present disclosure.
  • the manufacturing line 102 may include a plurality of nested syringe and vial lines 106, each of which includes the at least one nested syringe and vial line 104, for example.
  • the manufacturing line 102 also includes at least one pump 110 that corresponds to and cooperates with at least one vial of the at least one nested syringe and vial line 104.
  • the nested syringe and vial line 104 is a B20 nested syringe and vial line (NSVL) that includes a semi-automated debagger 104a, an automated debagger 104b, a rapid transfer airlock 104c, a nested filler (isolator) 104d, and a capper 104e.
  • NSVL B20 nested syringe and vial line
  • Various other clinical or commercial manufacturing fillers may alternatively be used and still fall within the scope of the present disclosure.
  • FIG. 13B depicts a plurality of pumps, which may be the plurality of pumps 112 that correspond to the plurality of vials 114 of the nested syringe and vial line 104 of FIG. 12, for example.
  • the plurality of pumps 112 include five pumps 110 that cooperate with each vial 105 of the plurality of vials 114.
  • the nested syringe and vial line 104 is a clinical manufacturing filler, such a Bausch & Strobel filler.
  • the containers 105 are referred to generally as vials, it will be understood that the containers 105 may be one or more of vials, syringes or plastic cartridges and still fall within the scope of the present disclosure.
  • the containers 105 may include nested ISO 2R vials, nested 1mL glass and plastic syringes, or nested 5cc plastic cartridges.
  • the plurality of pumps include five pumps 110 in this example, it will be understood that more or fewer pumps may alternatively be used and still fall within the scope of the present disclosure.
  • the plurality of pumps may include 10 pumps or 2 pumps for different filler, or any other number of pumps within this range, for example, and still fall within the scope of the present disclosure.
  • a method of filling the plurality of vials 105 of the nested syringe and vial line 104 comprises providing one of the pump 110 or a plurality of pumps 112 corresponding to one of the vial 105 or the plurality of vials 105 of the nested syringe and vial line 104.
  • the method also includes setting the drip retraction parameter for each pump 110 to any value equal to or less than 20 degrees.
  • the method also includes setting a no adjustment limit for the fill weight of the vial 105 to T 1 , with T 1 being at or in a range of about 2% more or 2% less than the fill weight of the target fill weight TO.
  • the method may still further include filling each vial 105 of the plurality of vials 105 with a drug product, such as a mAb formulation, via a corresponding pump 110 of the plurality of pumps 112.
  • the method may still also include exceeding a minimum value for the process performance index Cpk for each vial 105 of the plurality of vials 105 during a temperature range throughout a fill cycle, the temperature range one of: (1) 5 (+/-3) degrees Celsius; (2) 20 (+/-5) degree Celsius; or (3) 10 to 19 degrees Celsius.
  • the minimum value for the process performance index Cpk is 1.33. In other examples, such as in clinical fills, a minimum value for the process performance index Cpk is 1.0. However, in this example, and as generally understood in commercial fills, the minimum value for the process performance index Cpk is 1.33.
  • filling each vial 105 with the drug product via the corresponding pump 110 of the plurality of pumps 112 comprises filling each vial 105 with a drug product, wherein the drug product has one or more of the following characteristics: (1) a density in a range of about 1.0-1.2 g/cm3; (b) a viscosity in a range of about 1.0-10.0 cP; and (c) a surface tension in a range of about 40.0-72.7 mN/m.
  • the drug product has a density in a range of 1.0-1.2 g/cm3; a viscosity in a range of about 1.0-10.0 cP; and a surface tension in a range of about 40.0-72.7 mN/m.
  • the fill performance of the new filling recipe of the present disclosure in the nested syringe and vial line 104 is provided.
  • the total dose optimization cycle such as the number of strokes of the pump 110 (or pumps 112) needed to teach the pump 110 how to operate using the new filling recipe is minimized.
  • the total dose optimization cycle value is 4, which is significantly reduced compared to values of the total dose optimization cycle of previous filling recipes set forth in FIG. 15, for example.
  • the process performance index Cpk for each nozzle (not shown) of each pump 110 of the plurality of pumps 112 of the nested syringe and vial Iine104 exceeds the minimum value of 1.33 for the process performance index Cpk desired.
  • the average process performance index Cpk value for all nozzles of the pumps 110 is 1.4.
  • FIG. 15 a chart listing the process performance index Cpk of nozzles of the pumps using old filling recipes is set forth. Specifically, when using old filling recipes for a large variety of different drug products, including the mAb formulations, the average process performance index Cpk for all nozzles of the pumps was well below the desired process performance index Cpk value of 1 .33. Said another way, all of the process performance index Cpk values were less than 1.33.
  • the total dose optimization cycle was greater for each drug product using the old filling recipe compared to the dose optimization cycle value listed in FIG. 14 when using the new filling recipe 50.
  • the method of optimizing the filling recipe for the nested syringe and vial line 104 includes setting the drip retraction parameter for at least one pump 14, 16, 22 in the offline manufacturing system 10 corresponding to at least one container 18, 20 to any value equal to or less than 20 degrees.
  • the method further includes monitoring a performance of the at least one pump 14, 16, 20 with the drip retraction parameter of the at least one pump 14, 16, 22 set to any value equal to or less than 20 degrees.
  • the method still further includes obtaining at least a minimum value for the process performance index (Cpk) for the at least one container 18, 20 throughout at least one fill cycle and for at least one drug product using the at least one pump 14, 16, 22 in the offline manufacturing system 10.
  • the method also includes finalizing a filling recipe for the nested syringe and vial line 104 using data from fill cycles of the at least one drug product using the at least one pump 14, 16, 22 in the offline manufacturing system 10.
  • the above description describes various systems and methods of filling a vial of a nested syringe and vial line. It should be clear that the system or methods can further comprise use of a medicament listed below with the caveat that the following list should neither be considered to be all inclusive nor limiting.
  • the medicament will be contained in a reservoir.
  • the reservoir is a primary container that is either filled for treatment with the medicament.
  • the primary container can be a vial, a cartridge or a syringe.
  • drug products that may be used with the methods disclosed herein may include colony stimulating factors, such as granulocyte colony-stimulating factor (G-CSF).
  • G-CSF agents include, but are not limited to, Neupogen® (filgrastim) and Neulasta® (pegfilgrastim).
  • the methods may use various pharmaceutical products, such as an erythropoiesis stimulating agent (ESA), which may be in a liquid or a lyophilized form.
  • ESA erythropoiesis stimulating agent
  • An ESA is any molecule that stimulates erythropoiesis, such as Epogen® (epoetin alfa), Aranesp® (darbepoetin alfa), Dynepo® (epoetin delta), Mircera® (methyoxy polyethylene glycol-epoetin beta), Flematide®, MRK-2578, INS-22, Retacrit® (epoetin zeta), Neorecormon® (epoetin beta), Silapo® (epoetin zeta), Binocrit® (epoetin alfa), epoetin alfa Hexal, Abseamed® (epoetin alfa), Ratioepo®
  • An ESA can be an erythropoiesis stimulating protein.
  • erythropoiesis stimulating protein means any protein that directly or indirectly causes activation of the erythropoietin receptor, for example, by binding to and causing dimerization of the receptor.
  • Erythropoiesis stimulating proteins include erythropoietin and variants, analogs, or derivatives thereof that bind to and activate erythropoietin receptor; antibodies that bind to erythropoietin receptor and activate the receptor; or peptides that bind to and activate erythropoietin receptor.
  • Erythropoiesis stimulating proteins include, but are not limited to, epoetin alfa, epoetin beta, epoetin delta, epoetin omega, epoetin iota, epoetin zeta, and analogs thereof, pegylated erythropoietin, carbamylated erythropoietin, mimetic peptides (including EMP1/hematide), and mimetic antibodies.
  • Exemplary erythropoiesis stimulating proteins include erythropoietin, darbepoetin, erythropoietin agonist variants, and peptides or antibodies that bind and activate erythropoietin receptor (and include compounds reported in U.S. Publication Nos. 2003/0215444 and 2006/0040858, the disclosures of each of which is incorporated herein by reference in its entirety) as well as erythropoietin molecules or variants or analogs thereof as disclosed in the following patents or patent applications, which are each herein incorporated by reference in its entirety: U.S. Patent Nos.
  • Examples of other pharmaceutical products that may be used with the methods disclosed herein may include, but are not limited to, antibodies such as Vectibix® (panitumumab), XgevaTM (denosumab) and ProliaTM (denosamab); other biological agents such as Enbrel® (etanercept, TNF-receptor / Fc fusion protein, TNF blocker), Neulasta® (pegfilgrastim, pegylated filgastrim, pegylated G-CSF, pegylated hu-Met-G-CSF), Neupogen® (filgrastim , G-CSF, hu-MetG-CSF), and Nplate® (romiplostim); small molecule drugs such as Sensipar® (cinacalcet).
  • antibodies such as Vectibix® (panitumumab), XgevaTM (denosumab) and ProliaTM (denosamab)
  • other biological agents such
  • the methods may also be used with a therapeutic antibody, a polypeptide, a protein or other chemical, such as an iron, for example, ferumoxytol, iron dextrans, ferric glyconate, and iron sucrose.
  • a therapeutic antibody for example, a polypeptide, a protein or other chemical, such as an iron, for example, ferumoxytol, iron dextrans, ferric glyconate, and iron sucrose.
  • the pharmaceutical product may be in liquid form, or reconstituted from lyophilized form.
  • proteins are the specific proteins set forth below, including fusions, fragments, analogs, variants or derivatives thereof: OPGL specific antibodies, peptibodies, and related proteins, and the like (also referred to as RANKL specific antibodies, peptibodies and the like), including fully humanized and human OPGL specific antibodies, particularly fully humanized monoclonal antibodies, including but not limited to the antibodies described in PCT Publication No.
  • WO 03/002713 which is incorporated herein in its entirety as to OPGL specific antibodies and antibody related proteins, particularly those having the sequences set forth therein, particularly, but not limited to, those denoted therein: 9H7; 18B2; 2D8; 2E11; 16E1; and 22B3, including the OPGL specific antibodies having either the light chain of SEQ ID NO:2 as set forth therein in Figure 2 and/or the heavy chain of SEQ ID NO:4, as set forth therein in Figure 4, each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing publication;
  • WO 2004/058988 which are incorporated by reference herein in their entirety particularly in parts pertinent to myostatin specific peptibodies, including but not limited to peptibodies of the mTN8-19 family, including those of SEQ ID NOS:305-351, including TN8-19-1 through TN8-19-40, TN8-19 coni and TN8-19 con2; peptibodies of the mL2 family of SEQ ID NOS:357-383; the mL15 family of SEQ ID NOS:384- 409; the mL17 family of SEQ ID NOS:410-438; the mL20 family of SEQ ID NOS:439-446; the mL21 family of SEQ ID NOS:447- 452; the mL24 family of SEQ ID NOS:453-454; and those of SEQ ID NOS:615-631, each of which is individually and specifically incorporated by reference herein in their entirety fully as disclosed in the foregoing publication; [0085
  • Interleukin 1-receptor 1 (“IL1-R1”) specific antibodies, peptibodies, and related proteins, and the like, including but not limited to those described in U.S. Publication No. 2004/097712, which is incorporated herein by reference in its entirety in parts pertinent to IL1-R1 specific binding proteins, monoclonal antibodies in particular, especially, without limitation, those designated therein: 15CA, 26F5, 27F2, 24E12, and 10H7, each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the aforementioned publication;
  • Ang2 specific antibodies, peptibodies, and related proteins, and the like including but not limited to those described in PCT Publication No. WO 03/057134 and U.S. Publication No. 2003/0229023, each of which is incorporated herein by reference in its entirety particularly in parts pertinent to Ang2 specific antibodies and peptibodies and the like, especially those of sequences described therein and including but not limited to: L1(N); L1(N) WT; L1(N) 1 K WT; 2xL1(N); 2xL1(N) WT; Con4 (N), Con4 (N) 1 K WT, 2xCon4 (N) 1 K; L1C; L1C 1 K; 2xL1C; Con4C; Con4C 1 K; 2xCon4C 1 K; Con4-L1 (N); Con4-L1C; TN-12-9 (N); C17 (N); TN8-8(N); TN8-14 (N); Con 1 (N),
  • WO 2003/030833 which is incorporated herein by reference in its entirety as to the same, particularly Ab526; Ab528; Ab531; Ab533; Ab535; Ab536; Ab537; Ab540; Ab543; Ab544; Ab545; Ab546; A551; Ab553; Ab555; Ab558; Ab559; Ab565; AbFIAbFD; AbFE; AbFJ; AbFK; AbG1D4; AbGC1E8; AbH1C12; AblA1 ; AblF; AbIK, AblP; and AblP, in their various permutations as described therein, each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing publication;
  • NGF specific antibodies, peptibodies, and related proteins, and the like including, in particular, but not limited to those described in U.S. Publication No. 2005/0074821 and U.S. Patent No. 6,919,426, which are incorporated herein by reference in their entirety particularly as to NGF-specific antibodies and related proteins in this regard, including in particular, but not limited to, the NGF-specific antibodies therein designated 4D4, 4G6, 6H9, 7H2, 14D10 and 14D11, each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing publication;
  • CD22 specific antibodies, peptibodies, and related proteins, and the like such as those described in U.S. Patent No. 5,789,554, which is incorporated herein by reference in its entirety as to CD22 specific antibodies and related proteins, particularly human CD22 specific antibodies, such as but not limited to humanized and fully human antibodies, including but not limited to humanized and fully human monoclonal antibodies, particularly including but not limited to human CD22 specific IgG antibodies, such as, for instance, a dimer of a human-mouse monoclonal hLL2 gamma-chain disulfide linked to a human-mouse monoclonal hLL2 kappa-chain, including, but limited to, for example, the human CD22 specific fully humanized antibody in Epratuzumab, CAS registry number 501423-23-0;
  • IGF-1 receptor specific antibodies such as those described in PCT Publication No. WO 06/069202, which is incorporated herein by reference in its entirety as to IGF-1 receptor specific antibodies and related proteins, including but not limited to the IGF-1 specific antibodies therein designated L1 H1, L2H2, L3H3, L4H4, L5H5, L6H6, L7H7, L8H8, L9H9, L10H10, L11H11, L12H12, L13H13, L14H14, L15H15, L16H16, L17H17, L18H18, L19H19, L20H20, L21H21, L22H22, L23H23, L24H24, L25H25, L26H26, L27H27, L28H28, L29H29, L30H30, L31H31, L32H32, L33H33, L34H34, L35H35, L36H36, L
  • anti-IGF-1 R antibodies for use in the methods and compositions of the present invention are each and all of those described in:
  • IL-15 specific antibodies, peptibodies, and related proteins, and the like such as, in particular, humanized monoclonal antibodies, particularly antibodies such as those disclosed in U.S. Publication Nos. 2003/0138421; 2003/023586; and 2004/0071702; and U.S. Patent No. 7,153,507, each of which is incorporated herein by reference in its entirety as to IL-15 specific antibodies and related proteins, including peptibodies, including particularly, for instance, but not limited to, HuMax IL-15 antibodies and related proteins, such as, for instance, 146B7;
  • IFN gamma specific antibodies peptibodies, and related proteins and the like, especially human IFN gamma specific antibodies, particularly fully human anti-IFN gamma antibodies, such as, for instance, those described in U.S. Publication No. 2005/0004353, which is incorporated herein by reference in its entirety as to IFN gamma specific antibodies, particularly, for example, the antibodies therein designated 1118; 1118*; 1119; 1121; and 1121*.
  • Specific antibodies include those having the heavy chain of SEQ ID NO:17 and the light chain of SEQ ID NO:18; those having the heavy chain variable region of SEQ ID NO:6 and the light chain variable region of SEQ ID NO:8; those having the heavy chain of SEQ ID NO:19 and the light chain of SEQ ID NO:20; those having the heavy chain variable region of SEQ ID NO:10 and the light chain variable region of SEQ ID NO:12; those having the heavy chain of SEQ ID NO:32 and the light chain of SEQ ID NO:20; those having the heavy chain variable region of SEQ ID NO:30 and the light chain variable region of SEQ ID NO:12; those having the heavy chain sequence of SEQ ID NO:21 and the light chain sequence of SEQ ID NO:22; those having the heavy chain variable region of SEQ ID NO:14 and the light chain variable region of SEQ ID NO:16; those having the heavy chain of SEQ ID NO:21 and the light chain of SEQ ID NO:33; and those having the heavy chain variable region of SEQ ID NO:14 and the
  • TALL-1 specific antibodies such as those described in U.S. Publication Nos. 2003/0195156 and 2006/0135431, each of which is incorporated herein by reference in its entirety as to TALL-1 binding proteins, particularly the molecules of Tables 4 and 5B, each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing publications;
  • PTH Parathyroid hormone
  • TPO-R Thrombopoietin receptor
  • TRAIL-R2 specific antibodies, peptibodies, related proteins and the like such as those described in U.S. Patent No. 7,521,048, which is herein incorporated by reference in its entirety, particularly in parts pertinent to proteins that bind TRAIL-R2;
  • Activin A specific antibodies, peptibodies, related proteins, and the like, including but not limited to those described in U.S. Publication No. 2009/0234106, which is herein incorporated by reference in its entirety, particularly in parts pertinent to proteins that bind Activin A;
  • TGF-beta specific antibodies, peptibodies, related proteins, and the like including but not limited to those described in U.S. Patent No. 6,803,453 and U.S. Publication No. 2007/0110747, each of which is herein incorporated by reference in its entirety, particularly in parts pertinent to proteins that bind TGF-beta;
  • Amyloid-beta protein specific antibodies including but not limited to those described in PCT Publication No. WO 2006/081171, which is herein incorporated by reference in its entirety, particularly in parts pertinent to proteins that bind amyloid-beta proteins.
  • One antibody contemplated is an antibody having a heavy chain variable region comprising SEQ ID NO:8 and a light chain variable region having SEQ ID NO:6 as disclosed in the foregoing publication;
  • c-Kit specific antibodies including but not limited to those described in U.S. Publication No. 2007/0253951, which is incorporated herein by reference in its entirety, particularly in parts pertinent to proteins that bind c-Kit and/or other stem cell factor receptors;
  • OX40L specific antibodies, peptibodies, related proteins, and the like including but not limited to those described in U.S. Publication No. 2006/0002929, which is incorporated herein by reference in its entirety, particularly in parts pertinent to proteins that bind OX40L and/or other ligands of the 0X40 receptor; and
  • Activase® (alteplase, tPA); Aranesp® (darbepoetin alfa); Epogen® (epoetin alfa, or erythropoietin); GLP-1, Avonex® (interferon beta-1a); Bexxar® (tositumomab, anti-CD22 monoclonal antibody); Betaseron® (interferon-beta); Campath® (alemtuzumab, anti-CD52 monoclonal antibody); Dynepo® (epoetin delta); Velcade® (bortezomib); MLN0002 (anti- a4b7 mAb); MLN1202 (anti-CCR2 chemokine receptor mAb); Enbrel® (etanercept, TNF-receptor /Fc fusion protein, TNF blocker); Eprex® (epoetin alfa);
  • LymphoCide® epratuzumab, anti-CD22 mAb
  • BenlystaTM lymphostat B, belimumab, anti-BlyS mAb
  • Metalyse® tenecteplase, t-PA analog
  • Mircera® methoxy polyethylene glycol-epoetin beta
  • Mylotarg® gemtuzumab ozogamicin
  • efalizumab Cimzia® (certolizumab pegol, CDP 870); SolirisTM (eculizumab); pexelizumab (anti-C5 complement); Numax® (MEDI-524); Lucentis® (ranibizumab); Panorex® (17-1 A, edrecolomab); Trabio® (lerdelimumab); TheraCim hR3 (nimotuzumab); Omnitarg (pertuzumab, 2C4); Osidem® (IDM-1); OvaRex® (B43.13); Nuvion® (visilizumab); cantuzumab mertansine (huC242- DM1); NeoRecormon® (epoetin beta); Neumega® (oprelvekin, human interleukin-11); Neulasta® (pegylated filgastrim, pegylated G-CSF, pegylated hu-Met-G
  • Tysabri® natalizumab, anti-a4integrin mAb
  • Valortim® MDX-1303, anti-B. anthracis protective antigen mAb
  • ABthraxTM Vectibix® (panitumumab); Xolair® (omalizumab); ETI211 (anti-MRSA mAb)
  • IL-1 trap the Fc portion of human lgG1 and the extracellular domains of both IL-1 receptor components (the Type I receptor and receptor accessory protein)
  • VEGF trap Ig domains of VEGFR1 fused to lgG1 Fc
  • Zenapax® diaclizumab
  • Zenapax® diaclizumab, anti-IL-2Ra mAb
  • Zevalin® ibritumomab tiuxetan
  • Zetia® ezetimibe
  • Orencia® atacicept, TACI-lg
  • sclerostin antibody such as but not limited to romosozumab, blosozumab, or BPS 804 (Novartis).
  • therapeutics such as rilotumumab, bixalomer, trebananib, ganitumab, conatumumab, motesanib diphosphate, brodalumab, vidupiprant, panitumumab, denosumab, NPLATE, PROLIA, VECTIBIX or XGEVA.
  • a monoclonal antibody that binds human Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9), e.g. U.S. Patent No. 8,030,547, U.S. Publication No.
  • talimogene laherparepvec or another oncolytic HSV for the treatment of melanoma or other cancers.
  • oncolytic HSV include, but are not limited to talimogene laherparepvec (U.S. Patent Nos. 7,223,593 and 7,537,924); OncoVEXGALV/CD (U.S. Pat. No. 7,981,669); OrienXOIO (Lei et al. (2013), World J. Gastroenterol., 19:5138-5143); G207, 1716; NV1020; NV12023; NV1034 and NV1042 (Vargehes et al. (2002), Cancer Gene Ther., 9(12):967-978).
  • TIMPs are endogenous tissue inhibitors of metal loproteinases (TIMPs) and are important in many natural processes.
  • TIMP-3 is expressed by various cells or and is present in the extracellular matrix; it inhibits all the major cartilage-degrading metalloproteases, and may play a role in role in many degradative diseases of connective tissue, including rheumatoid arthritis and osteoarthritis, as well as in cancer and cardiovascular conditions.
  • the amino acid sequence of TIMP-3, and the nucleic acid sequence of a DNA that encodes TIMP-3 are disclosed in U.S. Patent No. 6,562,596, issued May 13, 2003, the disclosure of which is incorporated by reference herein. Description of TIMP mutations can be found in U.S. Publication No. 2014/0274874 and PCT Publication No. WO 2014/152012.
  • CGRP human calcitonin gene-related peptide
  • bispecific antibody molecule that target the CGRP receptor and other headache targets. Further information concerning these molecules can be found in PCT Application No. WO 2010/075238.
  • bispecific T cell engager (BiTE®) molecules e.g. BLINCYTO® (blinatumomab)
  • BLINCYTO® blindatumomab
  • APJ large molecule agonist e.g., apelin or analogues thereof in the device.
  • Information relating to such molecules can be found in PCT Publication No. WO 2014/099984.
  • the medicament comprises a therapeutically effective amount of an anti-thymic stromal lymphopoietin (TSLP) or TSLP receptor antibody.
  • TSLP anti-thymic stromal lymphopoietin
  • anti-TSLP antibodies that may be used in such embodiments include, but are not limited to, those described in U.S. Patent Nos. 7,982,016, and 8,232,372, and U.S. Publication No. 2009/0186022.
  • anti-TSLP receptor antibodies include, but are not limited to, those described in U.S. Patent No. 8,101,182.
  • the medicament comprises a therapeutically effective amount of the anti-TSLP antibody designated as A5 within U.S. Patent No. 7,982,016.

Landscapes

  • Engineering & Computer Science (AREA)
  • Mechanical Engineering (AREA)
  • Medical Preparation Storing Or Oral Administration Devices (AREA)
  • Basic Packing Technique (AREA)
  • Filling Of Jars Or Cans And Processes For Cleaning And Sealing Jars (AREA)

Abstract

La présente invention concerne un procédé de remplissage d'un flacon. Le procédé comprend la fourniture d'une pompe correspondant à un flacon et le réglage d'un paramètre de rétraction de goutte-à-goutte pour la pompe à une valeur quelconque égale ou inférieure à 20 degrés. Le procédé comprend en outre le réglage d'une limite de non-réglage pour un poids de remplissage du flacon à T1, T1 étant égal à un poids de remplissage cible T0 ou dans une plage de plus ou moins environ 2 % autour de celui-ci, un indice de performance de traitement Cpk (Cpk) pour le flacon tout au long d'un cycle de remplissage dépasse une valeur minimale.
PCT/US2022/030014 2021-05-21 2022-05-19 Procédé d'optimisation d'une recette de remplissage pour un récipient de médicament WO2022246055A1 (fr)

Priority Applications (10)

Application Number Priority Date Filing Date Title
KR1020237038066A KR20240011135A (ko) 2021-05-21 2022-05-19 약물 용기를 위한 충전 레시피를 최적화하는 방법
US18/288,549 US20240208680A1 (en) 2021-05-21 2022-05-19 Method of optimizing a filling recipe for a drug container
CN202280035596.1A CN117320964A (zh) 2021-05-21 2022-05-19 优化药物容器的灌装方案的方法
BR112023024278A BR112023024278A2 (pt) 2021-05-21 2022-05-19 Método de otimizar uma receita de enchimento para um recipiente de fármacos
MX2023013640A MX2023013640A (es) 2021-05-21 2022-05-19 Metodo de optimizacion de una receta de llenado para un contenedor de medicamento.
JP2023569877A JP2024523779A (ja) 2021-05-21 2022-05-19 薬物容器用の充填レシピを最適化する方法
AU2022279223A AU2022279223A1 (en) 2021-05-21 2022-05-19 Method of optimizing a filling recipe for a drug container
IL307418A IL307418A (en) 2021-05-21 2022-05-19 A method for optimizing a filling recipe for a drug container
CA3217207A CA3217207A1 (fr) 2021-05-21 2022-05-19 Procede d'optimisation d'une recette de remplissage pour un recipient de medicament
EP22731368.1A EP4341161A1 (fr) 2021-05-21 2022-05-19 Procédé d?optimisation d?une recette de remplissage pour un récipient de médicament

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163191797P 2021-05-21 2021-05-21
US63/191,797 2021-05-21

Publications (1)

Publication Number Publication Date
WO2022246055A1 true WO2022246055A1 (fr) 2022-11-24

Family

ID=82100162

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/030014 WO2022246055A1 (fr) 2021-05-21 2022-05-19 Procédé d'optimisation d'une recette de remplissage pour un récipient de médicament

Country Status (12)

Country Link
US (1) US20240208680A1 (fr)
EP (1) EP4341161A1 (fr)
JP (1) JP2024523779A (fr)
KR (1) KR20240011135A (fr)
CN (1) CN117320964A (fr)
AU (1) AU2022279223A1 (fr)
BR (1) BR112023024278A2 (fr)
CA (1) CA3217207A1 (fr)
CL (1) CL2023003444A1 (fr)
IL (1) IL307418A (fr)
MX (1) MX2023013640A (fr)
WO (1) WO2022246055A1 (fr)

Citations (155)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4703008A (en) 1983-12-13 1987-10-27 Kiren-Amgen, Inc. DNA sequences encoding erythropoietin
WO1991005867A1 (fr) 1989-10-13 1991-05-02 Amgen Inc. Isoformes d'erythropoietine
WO1995005465A1 (fr) 1993-08-17 1995-02-23 Amgen Inc. Analogues d'erytropoietine
US5441868A (en) 1983-12-13 1995-08-15 Kirin-Amgen, Inc. Production of recombinant erythropoietin
US5547933A (en) 1983-12-13 1996-08-20 Kirin-Amgen, Inc. Production of erythropoietin
WO1996038557A1 (fr) 1995-06-02 1996-12-05 Genentech, Inc. Antagonistes du recepteur du facteur de croissance des hepatocytes et leurs utilisations
WO1996040772A2 (fr) 1995-06-07 1996-12-19 Ortho Pharmaceutical Corporation Dimeres peptidiques d'agonistes
US5773569A (en) 1993-11-19 1998-06-30 Affymax Technologies N.V. Compounds and peptides that bind to the erythropoietin receptor
US5789554A (en) 1994-08-12 1998-08-04 Immunomedics, Inc. Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US5830851A (en) 1993-11-19 1998-11-03 Affymax Technologies N.V. Methods of administering peptides that bind to the erythropoietin receptor
US5856298A (en) 1989-10-13 1999-01-05 Amgen Inc. Erythropoietin isoforms
WO1999066054A2 (fr) 1998-06-15 1999-12-23 Genzyme Transgenics Corp. Fusion analogue d'erythropoietine-albumine serique humaine
US6030086A (en) 1998-03-02 2000-02-29 Becton, Dickinson And Company Flash tube reflector with arc guide
WO2000024893A2 (fr) 1998-10-23 2000-05-04 Amgen Inc. Methodes et compositions permettant de prevenir et de traiter l'anemie
WO2000061637A1 (fr) 1999-04-14 2000-10-19 Smithkline Beecham Corporation Anticorps du recepteur d'erythropoietine
WO2001031007A2 (fr) 1999-10-22 2001-05-03 Millennium Pharmaceuticals, Inc. Molecules d'acide nucleique derivees d'un cerveau de rat et modeles de mort cellulaire programmee
WO2001036489A2 (fr) 1999-11-12 2001-05-25 Merck Patent Gmbh Formes d'erythropoietine dotees de proprietes ameliorees
US6310078B1 (en) 1998-04-20 2001-10-30 Ortho-Mcneil Pharmaceutical, Inc. Substituted amino acids as erythropoietin mimetics
WO2001081405A2 (fr) 2000-04-21 2001-11-01 Amgen Inc. Methodes et compositions destinees a la prevention et au traitement de l'anemie
WO2002014356A2 (fr) 2000-08-11 2002-02-21 Baxter Healthcare Sa Methodes therapeutiques de traitement de sujet avec une erytrhopoietine recombinee presentant une activite elevee et peu d'effets secondaires
WO2002020034A1 (fr) 2000-09-08 2002-03-14 Gryphon Therapeutics, Inc. Ligation chimique 'pseudo'-native
US6391633B1 (en) 1997-07-23 2002-05-21 Roche Diagnostics Gmbh Production of erythropoietin by endogenous gene activation
WO2002049673A2 (fr) 2000-12-20 2002-06-27 F. Hoffmann-La Roche Ag Conjugues d'erythropoietine
WO2002085940A2 (fr) 2001-04-04 2002-10-31 Genodyssee Polynucleotides et polypeptides du gene de l'erythropoietine (epo)
WO2003002713A2 (fr) 2001-06-26 2003-01-09 Abgenix, Inc. Anticorps opgl
US20030023586A1 (en) 2000-03-03 2003-01-30 Super Internet Site System Pty Ltd. On-line geographical directory
WO2003029291A2 (fr) 2001-09-25 2003-04-10 F. Hoffmann-La Roche Ag Erythropoietine pegylee et diglycosylee
WO2003030833A2 (fr) 2001-10-11 2003-04-17 Amgen Inc. Agents de liaison spécifiques de l'angiopoïétine-2
US20030082749A1 (en) 2001-08-17 2003-05-01 Sun Lee-Hwei K. Fc fusion proteins of human erythropoietin with increased biological activities
US6562596B1 (en) 1993-10-06 2003-05-13 Amgen Inc. Tissue inhibitor of metalloproteinase type three (TIMP-3) composition and methods
US6583272B1 (en) 1999-07-02 2003-06-24 Hoffmann-La Roche Inc. Erythropoietin conjugates
US6586398B1 (en) 2000-04-07 2003-07-01 Amgen, Inc. Chemically modified novel erythropoietin stimulating protein compositions and methods
WO2003055526A2 (fr) 2001-12-21 2003-07-10 Maxygen Aps Conjugues d'erythropoietine
WO2003057134A2 (fr) 2001-10-11 2003-07-17 Amgen, Inc. Agents de liaison specifiques de l'angiopoietine-2 humaine
US20030138421A1 (en) 2001-08-23 2003-07-24 Van De Winkel Jan G.J. Human Antibodies specific for interleukin 15 (IL-15)
WO2003059951A2 (fr) 2002-01-18 2003-07-24 Pierre Fabre Medicament Anticorps anti-igf-ir et leurs applications
US20030143202A1 (en) 2002-01-31 2003-07-31 Binley Katie (Mary) Anemia
US20030195156A1 (en) 2001-05-11 2003-10-16 Amgen Inc. Peptides and related molecules that bind to TALL-1
WO2003084477A2 (fr) 2002-03-29 2003-10-16 Centocor, Inc. Corps mimetiques de cdr de mammifere, compositions, procedes et utilisations
US20030215444A1 (en) 1994-07-26 2003-11-20 Amgen Inc. Antibodies which activate an erythropoietin receptor
WO2003094858A2 (fr) 2002-05-13 2003-11-20 Modigenetech Ltd. Érythropoïétine comportant une extension ctp
US20030235582A1 (en) 2002-06-14 2003-12-25 Immunogen, Inc. Anti-IGF-I receptor antibody
WO2004002417A2 (fr) 2002-06-28 2004-01-08 Centocor, Inc. Corps mimetiques mammaliens a deletion ch1, compositions, procedes et utilisations
WO2004002424A2 (fr) 2002-06-28 2004-01-08 Centocor, Inc. Corps mimetiques d'epo de mammifere a deletion ch1, compositions, methodes et utilisations associees
WO2004009627A1 (fr) 2002-07-19 2004-01-29 Cangene Corporation Composes erythropoietiques pegyles
US20040018191A1 (en) 2002-05-24 2004-01-29 Schering Corporation Neutralizing human anti-IGFR antibody
WO2004018667A1 (fr) 2002-08-26 2004-03-04 Kirin Beer Kabushiki Kaisha Peptides et medicaments les contenant
WO2004024761A1 (fr) 2002-09-11 2004-03-25 Fresenius Kabi Deutschland Gmbh Polypeptides-has, notamment, erythropoietine-has ayant subi une acylation
US20040071702A1 (en) 2001-08-23 2004-04-15 Genmab, Inc. Human antibodies specific for interleukin 15 (IL-15)
US20040071694A1 (en) 2002-10-14 2004-04-15 Devries Peter J. Erythropoietin receptor binding antibodies
WO2004033651A2 (fr) 2002-10-09 2004-04-22 Neose Technologies, Inc. Erythropoietine: remodelage et glycoconjugaison d'erythropoietine
WO2004035603A2 (fr) 2002-10-14 2004-04-29 Abbott Laboratories Anticorps se liant au recepteur de l'erythropoietine
US20040086503A1 (en) 2001-01-05 2004-05-06 Cohen Bruce D. Antibodies to insulin-like growth factor I receptor
US20040091961A1 (en) 2002-11-08 2004-05-13 Evans Glen A. Enhanced variants of erythropoietin and methods of use
US20040097712A1 (en) 2002-09-06 2004-05-20 Amgen, Inc. A Corporation Of The State Of Delaware Therapeutic human anti-IL1-R1 monoclonal antibody
US6756480B2 (en) 2000-04-27 2004-06-29 Amgen Inc. Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein
WO2004058988A2 (fr) 2002-12-20 2004-07-15 Amgen, Inc. Agents de liaison inhibant la myostatine
US20040175379A1 (en) 2002-10-14 2004-09-09 Devries Peter J. Erythropoietin receptor binding antibodies
US6803453B1 (en) 1998-11-27 2004-10-12 Darwin Discovery Ltd. Antibodies associated with alterations in bone density
US20040202655A1 (en) 2003-03-14 2004-10-14 Morton Phillip A. Antibodies to IGF-I receptor for the treatment of cancers
US20040228859A1 (en) 2003-04-02 2004-11-18 Yvo Graus Antibodies against insulin-like growth factor 1 receptor and uses thereof
US20040229318A1 (en) 2003-05-17 2004-11-18 George Heavner Erythropoietin conjugate compounds with extended half-lives
WO2004101611A2 (fr) 2003-05-12 2004-11-25 Affymax, Inc. Nouveaux peptides se fixant au recepteur de l'erythropoietine
WO2004101606A2 (fr) 2003-05-12 2004-11-25 Affymax, Inc. Nouveaux peptides se fixant au recepteur de l'erythropoietine
WO2004101600A2 (fr) 2003-05-12 2004-11-25 Affymax, Inc. Nouveaux composes modifies par du poly(ethylene glycol) et leurs utilisations
US6835809B1 (en) 1998-10-23 2004-12-28 Amgen Inc. Thrombopoietic compounds
US20040265307A1 (en) 2002-06-14 2004-12-30 Immunogen Inc. Anti-IGF-I receptor antibody
US20040266690A1 (en) 2003-05-30 2004-12-30 Chadler Pool Formation of novel erythropoietin conjugates using transglutaminase
WO2005001136A1 (fr) 2003-06-04 2005-01-06 Irm Llc Methodes et compositions pour la modulation de l'expression de l'erythropoietine
US20050004353A1 (en) 2002-10-16 2005-01-06 Amgen, Inc., A Corporation Of The State Of Delaware Human anti-IFN-gamma neutralizing antibodies as selective IFN-gamma pathway inhibitors
WO2005001025A2 (fr) 2003-05-06 2005-01-06 Syntonix Pharmaceuticals, Inc. Hybrides monomeres/dimeres chimeriques d'immunoglobuline
US20050008642A1 (en) 2003-07-10 2005-01-13 Yvo Graus Antibodies against insulin-like growth factor 1 receptor and uses thereof
US20050019914A1 (en) 2003-07-24 2005-01-27 Aventis Pharma Deutschland Gmbh Perfusion process for producing erythropoietin
US20050026834A1 (en) 1999-01-14 2005-02-03 Bolder Biotechnology, Inc. Methods for making proteins containing free cysteine residues
WO2005016970A2 (fr) 2003-05-01 2005-02-24 Imclone Systems Incorporated Anticorps entierement humains diriges contre le recepteur du facteur de croissance 1 de type insuline
WO2005017107A2 (fr) 2003-07-18 2005-02-24 Amgen Inc. Agents de liaison specifiques se liant a un facteur de croissance hepatocyte
WO2005021579A2 (fr) 2003-08-28 2005-03-10 Biorexis Pharmaceutical Corporation Peptides mimetiques epo et proteines de fusion
WO2005025606A1 (fr) 2003-09-09 2005-03-24 Warren Pharmaceuticals, Inc. Erythropoietines a action prolongee pouvant maintenir une activite de protection tissulaire d'une erythropoietine endogene
US20050074821A1 (en) 2003-07-15 2005-04-07 Wild Kenneth D. Human anti-NGF neutralizing antibodies as selective NGF pathway inhibitors
WO2005032460A2 (fr) 2003-09-30 2005-04-14 Centocor, Inc. Mimeticorps de noyau charniere mimetiques de l'epo humaine, compositions, procedes et applications correspondantes
US20050084906A1 (en) 2002-01-18 2005-04-21 Liliane Goetsch Novel anti-IGF-IR antibodies and uses thereof
US20050096461A1 (en) 1997-07-14 2005-05-05 Bolder Biotechnology, Inc. Cysteine variants of erythropoietin
WO2005047331A2 (fr) 2003-11-07 2005-05-26 Immunex Corporation Anticorps liant un recepteur de l'interleucine 4
WO2005051327A2 (fr) 2003-11-24 2005-06-09 Neose Technologies, Inc. Erythropoietine glycopegylee
US20050124564A1 (en) 2002-01-31 2005-06-09 Binley Katie M. Anemia
US20050136063A1 (en) 2003-11-21 2005-06-23 Schering Corporation Anti-IGFR antibody therapeutic combinations
WO2005058967A2 (fr) 2003-12-16 2005-06-30 Pierre Fabre Medicament Nouveau recepteur hybride anti-insuline/igf-i ou recepteur hybride anti-insuline/igf-i et anticorps igf-ir et applications
US20050153879A1 (en) 2002-03-26 2005-07-14 Monica Svetina Process for the preparation of a desired erythropoietin glyco-isoform profile
WO2005063809A1 (fr) 2003-12-22 2005-07-14 Dubai Genetics Fz-Llc Erythopoietine identique a la forme naturelle
WO2005063808A1 (fr) 2003-12-31 2005-07-14 Merck Patent Gmbh Proteine hybride fc-erythropoietine a pharmacocinetique amelioree
US6919426B2 (en) 2002-09-19 2005-07-19 Amgen Inc. Peptides and related molecules that modulate nerve growth factor activity
US20050158822A1 (en) 2004-01-20 2005-07-21 Insight Biopharmaceuticals Ltd. High level expression of recombinant human erythropoietin having a modified 5'-UTR
US20050170457A1 (en) 2003-12-31 2005-08-04 Chadler Pool Novel recombinant proteins with N-terminal free thiol
WO2005070451A1 (fr) 2004-01-22 2005-08-04 Zafena Aktiebolag Composition pharmaceutique comprenant une erythropoietine non glycosylee
US20050181359A1 (en) 1999-04-15 2005-08-18 Crucell Holland B.V. Compositions of erythropoietin isoforms comprising Lewis-X structures and high sialic acid content
US20050181482A1 (en) 2004-02-12 2005-08-18 Meade Harry M. Method for the production of an erythropoietin analog-human IgG fusion proteins in transgenic mammal milk
WO2005081687A2 (fr) 2003-09-30 2005-09-09 Centocor, Inc. Mimeticorps de noyau-charniere humain, compositions, procedes et applications correspondantes
WO2005084711A1 (fr) 2004-03-02 2005-09-15 Chengdu Institute Of Biological Products Erythropoietine recombinante pegylee a activite in vivo
US20050202538A1 (en) 1999-11-12 2005-09-15 Merck Patent Gmbh Fc-erythropoietin fusion protein with improved pharmacokinetics
WO2005092369A2 (fr) 2004-03-11 2005-10-06 Fresenius Kabi Deutschland Gmbh Conjugues d'hydroxy-ethyl-amidon et d'erythropoietine
US20050227289A1 (en) 2004-04-09 2005-10-13 Reilly Edward B Antibodies to erythropoietin receptor and uses thereof
WO2005103076A2 (fr) 2004-04-23 2005-11-03 Cambridge Antibody Technology Limited Variants d'erythropoietine
US20060002929A1 (en) 2004-03-23 2006-01-05 Khare Sanjay D Monoclonal antibodies
WO2006002646A2 (fr) 2004-07-07 2006-01-12 H. Lundbeck A/S Nouvelle erythropoietine carbamylee et son procede de production
WO2006013472A2 (fr) 2004-07-29 2006-02-09 Pierre Fabre Medicament Nouveaux anticorps anti-igf-ir et utilisations
US20060040358A1 (en) 1998-12-03 2006-02-23 Tanja Ligensa IGF-1 receptor interacting proteins
WO2006029094A2 (fr) 2004-09-02 2006-03-16 Xencor, Inc. Derives d'erythropoietine a antigenicite modifiee
WO2006050959A2 (fr) 2004-11-10 2006-05-18 Aplagen Gmbh Molecules favorisant l'hematopoiese
US20060111279A1 (en) 2003-11-24 2006-05-25 Neose Technologies, Inc. Glycopegylated erythropoietin
WO2006069202A2 (fr) 2004-12-22 2006-06-29 Amgen Inc. Compositions et procedes impliquant des anticorps diriges contre le recepteur igf-1r
WO2006081171A1 (fr) 2005-01-24 2006-08-03 Amgen Inc. Anticorps anti-amyloide humanise
WO2006138729A2 (fr) 2005-06-17 2006-12-28 Imclone Systems Incorporated Antagonistes de recepteur pour le traitement de cancer osseux metastatique
WO2007000328A1 (fr) 2005-06-27 2007-01-04 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Anticorps se fixant à un épitope sur un récepteur de facteur de croissance insulinomimétique de type 1 et leurs utilisations
WO2007011941A2 (fr) 2005-07-18 2007-01-25 Amgen Inc. Anticorps neutralisants anti-b7rp1 humains
WO2007012614A2 (fr) 2005-07-22 2007-02-01 Pierre Fabre Medicament Nouveaux anticorps anti igf-ir et leur utilisation
US7217689B1 (en) 1989-10-13 2007-05-15 Amgen Inc. Glycosylation analogs of erythropoietin
US20070110747A1 (en) 2005-05-03 2007-05-17 Ucb S.A. Binding agents
US7220410B2 (en) 2003-04-18 2007-05-22 Galaxy Biotech, Llc Monoclonal antibodies to hepatocyte growth factor
US7223593B2 (en) 2000-01-21 2007-05-29 Biovex Limited Herpes virus strains for gene therapy
US7271689B1 (en) 2000-11-22 2007-09-18 Fonar Corporation Magnet structure
US20070253951A1 (en) 2006-04-24 2007-11-01 Gordon Ng Humanized c-Kit antibody
WO2007136752A2 (fr) 2006-05-19 2007-11-29 Glycofi, Inc. Compositions d'érythropoïétine
WO2008057458A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
WO2008057459A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
WO2008057457A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
WO2008125623A2 (fr) 2007-04-13 2008-10-23 Novartis Ag Molécules et procédés de modulation de proprotéine convertase subtilisine/kexine de type 9 (pcsk9)
WO2008133647A2 (fr) 2006-11-07 2008-11-06 Merck & Co., Inc. Antagonistes de pcsk9
US7521048B2 (en) 2005-08-31 2009-04-21 Amgen Inc. TRAIL receptor-2 polypeptides and antibodies
WO2009055783A2 (fr) 2007-10-26 2009-04-30 Schering Corporation Anti-pcsk9 et méthodes de traitement de troubles du métabolisme lipidique et du cholestérol
US20090186022A1 (en) 2006-02-23 2009-07-23 Novartis Ag Organic Compounds
WO2009100297A1 (fr) 2008-02-07 2009-08-13 Merck & Co., Inc. Antagonistes de pcsk9 1d05
WO2009100318A1 (fr) 2008-02-07 2009-08-13 Merck & Co., Inc. Antagonistes de 1b20 pcsk9
US20090234106A1 (en) 2006-09-08 2009-09-17 Amgen Inc. Anti-activin a antibodies and uses thereof
WO2010029513A2 (fr) 2008-09-12 2010-03-18 Rinat Neuroscience Corporation Antagonistes de pcsk9
WO2010075238A1 (fr) 2008-12-23 2010-07-01 Amgen Inc. Protéines de liaison au récepteur cgrp humain
WO2010077854A1 (fr) 2008-12-15 2010-07-08 Regeneron Pharamaceuticals, Inc. Anticorps humains à grande affinité contre pcsk9
WO2011037791A1 (fr) 2009-09-25 2011-03-31 Merck Sharp & Dohme Corp. Antagonistes de pcsk9
WO2011053759A1 (fr) 2009-10-30 2011-05-05 Merck Sharp & Dohme Corp. Antagonistes de la pcsk9 avec anticorps fab ax189 et ax1, et variantes afférentes
WO2011053783A2 (fr) 2009-10-30 2011-05-05 Merck Sharp & Dohme Corp. Antagonistes et variants ax213 et ax132 pcsk9
WO2011072263A1 (fr) 2009-12-11 2011-06-16 Irm Llc Antagonistes de pcsk9
US7982016B2 (en) 2007-09-10 2011-07-19 Amgen Inc. Antigen binding proteins capable of binding thymic stromal lymphopoietin
US7981669B2 (en) 2003-07-25 2011-07-19 Biovex Limited Viral vectors
WO2011111007A2 (fr) 2010-03-11 2011-09-15 Rinat Neuroscience Corporation Anticorps présentant une liaison à l'antigène dépendante du ph
US8030547B2 (en) 2002-03-29 2011-10-04 Kumiai Chemical Industry Co., Ltd. Gene coding for acetolactate synthase
US8101182B2 (en) 2007-06-20 2012-01-24 Novartis Ag Methods and compositions for treating allergic diseases
WO2012054438A1 (fr) 2010-10-22 2012-04-26 Schering Corporation Anti-pcsk9
WO2012088313A1 (fr) 2010-12-22 2012-06-28 Genentech, Inc. Anticorps anti-pcsk9 et procédés d'utilisation
US8232372B2 (en) 2006-12-14 2012-07-31 Schering Corp. Engineered anti-TSLP antibody
WO2012101253A1 (fr) 2011-01-28 2012-08-02 Sanofi Compositions pharmaceutiques comprenant des anticorps humains contre pcsk9
WO2012109530A1 (fr) 2011-02-11 2012-08-16 Irm Llc Antagonistes de pcsk9
US20130064825A1 (en) 2011-05-10 2013-03-14 Amgen Inc. Methods of treating or preventing cholesterol related disorders
WO2014099984A1 (fr) 2012-12-20 2014-06-26 Amgen Inc. Agonistes du récepteur apj et leurs utilisations
US20140274874A1 (en) 2013-03-14 2014-09-18 Amgen Inc. Variants of tissue inhibitor of metalloproteinase type three (timp-3), compositions and methods
WO2014152012A2 (fr) 2013-03-14 2014-09-25 Amgen Inc. Variants d'inhibiteur tissulaire de la métalloprotéinase type iii (timp-3), compositions et procédés
US20190344257A1 (en) * 2011-07-22 2019-11-14 Vanrx Pharmasystems Inc. Fill needle system

Patent Citations (204)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5618698A (en) 1983-12-13 1997-04-08 Kirin-Amgen, Inc. Production of erythropoietin
US4703008A (en) 1983-12-13 1987-10-27 Kiren-Amgen, Inc. DNA sequences encoding erythropoietin
US5955422A (en) 1983-12-13 1999-09-21 Kirin-Amgen, Inc. Production of erthropoietin
US5441868A (en) 1983-12-13 1995-08-15 Kirin-Amgen, Inc. Production of recombinant erythropoietin
US5547933A (en) 1983-12-13 1996-08-20 Kirin-Amgen, Inc. Production of erythropoietin
US5756349A (en) 1983-12-13 1998-05-26 Amgen Inc. Production of erythropoietin
US5621080A (en) 1983-12-13 1997-04-15 Kirin-Amgen, Inc. Production of erythropoietin
US7217689B1 (en) 1989-10-13 2007-05-15 Amgen Inc. Glycosylation analogs of erythropoietin
WO1991005867A1 (fr) 1989-10-13 1991-05-02 Amgen Inc. Isoformes d'erythropoietine
US5856298A (en) 1989-10-13 1999-01-05 Amgen Inc. Erythropoietin isoforms
WO1995005465A1 (fr) 1993-08-17 1995-02-23 Amgen Inc. Analogues d'erytropoietine
US6562596B1 (en) 1993-10-06 2003-05-13 Amgen Inc. Tissue inhibitor of metalloproteinase type three (TIMP-3) composition and methods
US5773569A (en) 1993-11-19 1998-06-30 Affymax Technologies N.V. Compounds and peptides that bind to the erythropoietin receptor
US5986047A (en) 1993-11-19 1999-11-16 Affymax Technologies N.V. Peptides that bind to the erythropoietin receptor
US5830851A (en) 1993-11-19 1998-11-03 Affymax Technologies N.V. Methods of administering peptides that bind to the erythropoietin receptor
US20030215444A1 (en) 1994-07-26 2003-11-20 Amgen Inc. Antibodies which activate an erythropoietin receptor
US5789554A (en) 1994-08-12 1998-08-04 Immunomedics, Inc. Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
WO1996038557A1 (fr) 1995-06-02 1996-12-05 Genentech, Inc. Antagonistes du recepteur du facteur de croissance des hepatocytes et leurs utilisations
US5686292A (en) 1995-06-02 1997-11-11 Genentech, Inc. Hepatocyte growth factor receptor antagonist antibodies and uses thereof
US6468529B1 (en) 1995-06-02 2002-10-22 Genentech, Inc. Hepatocyte growth factor receptor antagonists and uses thereof
WO1996040772A2 (fr) 1995-06-07 1996-12-19 Ortho Pharmaceutical Corporation Dimeres peptidiques d'agonistes
US5767078A (en) 1995-06-07 1998-06-16 Johnson; Dana L. Agonist peptide dimers
US20050107591A1 (en) 1997-07-14 2005-05-19 Bolder Biotechnology, Inc. Cysteine variants of erythropoietin
US20050096461A1 (en) 1997-07-14 2005-05-05 Bolder Biotechnology, Inc. Cysteine variants of erythropoietin
US6391633B1 (en) 1997-07-23 2002-05-21 Roche Diagnostics Gmbh Production of erythropoietin by endogenous gene activation
US6030086A (en) 1998-03-02 2000-02-29 Becton, Dickinson And Company Flash tube reflector with arc guide
US6310078B1 (en) 1998-04-20 2001-10-30 Ortho-Mcneil Pharmaceutical, Inc. Substituted amino acids as erythropoietin mimetics
US20040248815A1 (en) 1998-04-20 2004-12-09 Ortho Mcneil Pharmaceutical, Inc. Substituted amino acids as erythropoietin mimetics
US6750369B2 (en) 1998-04-20 2004-06-15 Ortho Mcneil Pharmaceutical, Inc. Substituted amino acids as erythropoietin mimetics
WO1999066054A2 (fr) 1998-06-15 1999-12-23 Genzyme Transgenics Corp. Fusion analogue d'erythropoietine-albumine serique humaine
US20040143857A1 (en) 1998-06-15 2004-07-22 Michael Young Erythropoietin analog-human serum albumin fusion
US20020155998A1 (en) 1998-06-15 2002-10-24 Genzyme Transgenics Corporation, A Massachusetts Corporation Erythropoietin analog-human serum albumin fusion
US20050158832A1 (en) 1998-06-15 2005-07-21 Michael Young Erythropoietin analog-human serum albumin fusion
US6835809B1 (en) 1998-10-23 2004-12-28 Amgen Inc. Thrombopoietic compounds
WO2000024893A2 (fr) 1998-10-23 2000-05-04 Amgen Inc. Methodes et compositions permettant de prevenir et de traiter l'anemie
US6803453B1 (en) 1998-11-27 2004-10-12 Darwin Discovery Ltd. Antibodies associated with alterations in bone density
US20060040358A1 (en) 1998-12-03 2006-02-23 Tanja Ligensa IGF-1 receptor interacting proteins
US20050026834A1 (en) 1999-01-14 2005-02-03 Bolder Biotechnology, Inc. Methods for making proteins containing free cysteine residues
US20050244409A1 (en) 1999-04-14 2005-11-03 Smithkline Beecham Corporation Erythropoietin receptor antibodies
WO2000061637A1 (fr) 1999-04-14 2000-10-19 Smithkline Beecham Corporation Anticorps du recepteur d'erythropoietine
US20050181359A1 (en) 1999-04-15 2005-08-18 Crucell Holland B.V. Compositions of erythropoietin isoforms comprising Lewis-X structures and high sialic acid content
US6583272B1 (en) 1999-07-02 2003-06-24 Hoffmann-La Roche Inc. Erythropoietin conjugates
WO2001031007A2 (fr) 1999-10-22 2001-05-03 Millennium Pharmaceuticals, Inc. Molecules d'acide nucleique derivees d'un cerveau de rat et modeles de mort cellulaire programmee
US20050202538A1 (en) 1999-11-12 2005-09-15 Merck Patent Gmbh Fc-erythropoietin fusion protein with improved pharmacokinetics
WO2001036489A2 (fr) 1999-11-12 2001-05-25 Merck Patent Gmbh Formes d'erythropoietine dotees de proprietes ameliorees
US7537924B2 (en) 2000-01-21 2009-05-26 Biovex Limited Virus strains
US7223593B2 (en) 2000-01-21 2007-05-29 Biovex Limited Herpes virus strains for gene therapy
US20030023586A1 (en) 2000-03-03 2003-01-30 Super Internet Site System Pty Ltd. On-line geographical directory
US6586398B1 (en) 2000-04-07 2003-07-01 Amgen, Inc. Chemically modified novel erythropoietin stimulating protein compositions and methods
WO2001081405A2 (fr) 2000-04-21 2001-11-01 Amgen Inc. Methodes et compositions destinees a la prevention et au traitement de l'anemie
US6756480B2 (en) 2000-04-27 2004-06-29 Amgen Inc. Modulators of receptors for parathyroid hormone and parathyroid hormone-related protein
WO2002014356A2 (fr) 2000-08-11 2002-02-21 Baxter Healthcare Sa Methodes therapeutiques de traitement de sujet avec une erytrhopoietine recombinee presentant une activite elevee et peu d'effets secondaires
WO2002020034A1 (fr) 2000-09-08 2002-03-14 Gryphon Therapeutics, Inc. Ligation chimique 'pseudo'-native
WO2002019963A2 (fr) 2000-09-08 2002-03-14 Gryphon Therapeutics, Inc. Protéines de synthèse stimulant l'érythropoïèse
US7271689B1 (en) 2000-11-22 2007-09-18 Fonar Corporation Magnet structure
WO2002049673A2 (fr) 2000-12-20 2002-06-27 F. Hoffmann-La Roche Ag Conjugues d'erythropoietine
US7037498B2 (en) 2001-01-05 2006-05-02 Abgenix, Inc. Antibodies to insulin-like growth factor I receptor
US20050244408A1 (en) 2001-01-05 2005-11-03 Cohen Bruce D Antibodies to insulin-like growth factor I receptor
US20040086503A1 (en) 2001-01-05 2004-05-06 Cohen Bruce D. Antibodies to insulin-like growth factor I receptor
WO2002085940A2 (fr) 2001-04-04 2002-10-31 Genodyssee Polynucleotides et polypeptides du gene de l'erythropoietine (epo)
US20060135431A1 (en) 2001-05-11 2006-06-22 Amgen Inc. Peptides and related molecules that bind to TALL-1
US20030195156A1 (en) 2001-05-11 2003-10-16 Amgen Inc. Peptides and related molecules that bind to TALL-1
WO2003002713A2 (fr) 2001-06-26 2003-01-09 Abgenix, Inc. Anticorps opgl
US20040175824A1 (en) 2001-08-17 2004-09-09 Sun Lee-Hwei K. Fc fusion proteins of human erythropoietin with high biological activities
US6900292B2 (en) 2001-08-17 2005-05-31 Lee-Hwei K. Sun Fc fusion proteins of human erythropoietin with increased biological activities
US20050124045A1 (en) 2001-08-17 2005-06-09 Sun Lee-Hwei K. Fc fusion proteins of human erythropoietin with increased biological activities
US20050142642A1 (en) 2001-08-17 2005-06-30 Sun Lee-Hwei K. Fc fusion proteins of human erythropoietin with increased biological activities
US20030082749A1 (en) 2001-08-17 2003-05-01 Sun Lee-Hwei K. Fc fusion proteins of human erythropoietin with increased biological activities
US7030226B2 (en) 2001-08-17 2006-04-18 Sun Lee-Hwei K Fc fusion proteins of human erythropoietin with increased biological activities
US20040071702A1 (en) 2001-08-23 2004-04-15 Genmab, Inc. Human antibodies specific for interleukin 15 (IL-15)
US20030138421A1 (en) 2001-08-23 2003-07-24 Van De Winkel Jan G.J. Human Antibodies specific for interleukin 15 (IL-15)
US7153507B2 (en) 2001-08-23 2006-12-26 Genmab A/S Human antibodies specific for interleukin 15 (IL-15)
WO2003029291A2 (fr) 2001-09-25 2003-04-10 F. Hoffmann-La Roche Ag Erythropoietine pegylee et diglycosylee
US20030077753A1 (en) 2001-09-25 2003-04-24 Wilhelm Tischer Diglycosylated erythropoietin
US20060088906A1 (en) 2001-10-10 2006-04-27 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
US20030229023A1 (en) 2001-10-11 2003-12-11 Oliner Jonathan Daniel Specific binding agents of human angiopoietin-2
WO2003057134A2 (fr) 2001-10-11 2003-07-17 Amgen, Inc. Agents de liaison specifiques de l'angiopoietine-2 humaine
WO2003030833A2 (fr) 2001-10-11 2003-04-17 Amgen Inc. Agents de liaison spécifiques de l'angiopoïétine-2
WO2003055526A2 (fr) 2001-12-21 2003-07-10 Maxygen Aps Conjugues d'erythropoietine
US20050084906A1 (en) 2002-01-18 2005-04-21 Liliane Goetsch Novel anti-IGF-IR antibodies and uses thereof
WO2003059951A2 (fr) 2002-01-18 2003-07-24 Pierre Fabre Medicament Anticorps anti-igf-ir et leurs applications
US20050124564A1 (en) 2002-01-31 2005-06-09 Binley Katie M. Anemia
US20030143202A1 (en) 2002-01-31 2003-07-31 Binley Katie (Mary) Anemia
US20050153879A1 (en) 2002-03-26 2005-07-14 Monica Svetina Process for the preparation of a desired erythropoietin glyco-isoform profile
US8030547B2 (en) 2002-03-29 2011-10-04 Kumiai Chemical Industry Co., Ltd. Gene coding for acetolactate synthase
WO2003084477A2 (fr) 2002-03-29 2003-10-16 Centocor, Inc. Corps mimetiques de cdr de mammifere, compositions, procedes et utilisations
WO2003094858A2 (fr) 2002-05-13 2003-11-20 Modigenetech Ltd. Érythropoïétine comportant une extension ctp
US20040009902A1 (en) 2002-05-13 2004-01-15 Irving Boime CTP extended erythropoietin
US20040018191A1 (en) 2002-05-24 2004-01-29 Schering Corporation Neutralizing human anti-IGFR antibody
US20040265307A1 (en) 2002-06-14 2004-12-30 Immunogen Inc. Anti-IGF-I receptor antibody
US20050249728A1 (en) 2002-06-14 2005-11-10 Immunogen Inc. Anti-IGF-I receptor antibody
US20030235582A1 (en) 2002-06-14 2003-12-25 Immunogen, Inc. Anti-IGF-I receptor antibody
US20050186203A1 (en) 2002-06-14 2005-08-25 Immunogen Inc. Anti-IGF-I receptor antibody
WO2004002417A2 (fr) 2002-06-28 2004-01-08 Centocor, Inc. Corps mimetiques mammaliens a deletion ch1, compositions, procedes et utilisations
WO2004002424A2 (fr) 2002-06-28 2004-01-08 Centocor, Inc. Corps mimetiques d'epo de mammifere a deletion ch1, compositions, methodes et utilisations associees
WO2004009627A1 (fr) 2002-07-19 2004-01-29 Cangene Corporation Composes erythropoietiques pegyles
WO2004018667A1 (fr) 2002-08-26 2004-03-04 Kirin Beer Kabushiki Kaisha Peptides et medicaments les contenant
US20040097712A1 (en) 2002-09-06 2004-05-20 Amgen, Inc. A Corporation Of The State Of Delaware Therapeutic human anti-IL1-R1 monoclonal antibody
WO2004024761A1 (fr) 2002-09-11 2004-03-25 Fresenius Kabi Deutschland Gmbh Polypeptides-has, notamment, erythropoietine-has ayant subi une acylation
US6919426B2 (en) 2002-09-19 2005-07-19 Amgen Inc. Peptides and related molecules that modulate nerve growth factor activity
WO2004033651A2 (fr) 2002-10-09 2004-04-22 Neose Technologies, Inc. Erythropoietine: remodelage et glycoconjugaison d'erythropoietine
US20040071694A1 (en) 2002-10-14 2004-04-15 Devries Peter J. Erythropoietin receptor binding antibodies
US20040175379A1 (en) 2002-10-14 2004-09-09 Devries Peter J. Erythropoietin receptor binding antibodies
WO2004035603A2 (fr) 2002-10-14 2004-04-29 Abbott Laboratories Anticorps se liant au recepteur de l'erythropoietine
US20050004353A1 (en) 2002-10-16 2005-01-06 Amgen, Inc., A Corporation Of The State Of Delaware Human anti-IFN-gamma neutralizing antibodies as selective IFN-gamma pathway inhibitors
US20040091961A1 (en) 2002-11-08 2004-05-13 Evans Glen A. Enhanced variants of erythropoietin and methods of use
US20040157293A1 (en) 2002-11-08 2004-08-12 Evans Glen A. Enhanced variants of erythropoietin and methods of use
WO2004043382A2 (fr) 2002-11-08 2004-05-27 Egea Biosciences, Inc. Variants ameliores de l'erythropoietine et methodes d'utilisation
US20040181033A1 (en) 2002-12-20 2004-09-16 Hq Han Binding agents which inhibit myostatin
WO2004058988A2 (fr) 2002-12-20 2004-07-15 Amgen, Inc. Agents de liaison inhibant la myostatine
US20040202655A1 (en) 2003-03-14 2004-10-14 Morton Phillip A. Antibodies to IGF-I receptor for the treatment of cancers
US20040228859A1 (en) 2003-04-02 2004-11-18 Yvo Graus Antibodies against insulin-like growth factor 1 receptor and uses thereof
US7220410B2 (en) 2003-04-18 2007-05-22 Galaxy Biotech, Llc Monoclonal antibodies to hepatocyte growth factor
WO2005016970A2 (fr) 2003-05-01 2005-02-24 Imclone Systems Incorporated Anticorps entierement humains diriges contre le recepteur du facteur de croissance 1 de type insuline
WO2005001025A2 (fr) 2003-05-06 2005-01-06 Syntonix Pharmaceuticals, Inc. Hybrides monomeres/dimeres chimeriques d'immunoglobuline
WO2004101600A2 (fr) 2003-05-12 2004-11-25 Affymax, Inc. Nouveaux composes modifies par du poly(ethylene glycol) et leurs utilisations
US20060040858A1 (en) 2003-05-12 2006-02-23 Affymax, Inc. Novel peptides that bind to the erythropoietin receptor
US7084245B2 (en) 2003-05-12 2006-08-01 Affymax, Inc. Peptides that bind to the erythropoietin receptor
WO2004101606A2 (fr) 2003-05-12 2004-11-25 Affymax, Inc. Nouveaux peptides se fixant au recepteur de l'erythropoietine
US20050137329A1 (en) 2003-05-12 2005-06-23 Affymax, Inc. Novel peptides that bind to the erythropoietin receptor
WO2004101611A2 (fr) 2003-05-12 2004-11-25 Affymax, Inc. Nouveaux peptides se fixant au recepteur de l'erythropoietine
US20050107297A1 (en) 2003-05-12 2005-05-19 Holmes Christopher P. Novel poly(ethylene glycol) modified compounds and uses thereof
WO2004106373A1 (fr) 2003-05-17 2004-12-09 Centocor, Inc. Composes conjugues d'erythropoietine a demi-vies rallongees
US20040229318A1 (en) 2003-05-17 2004-11-18 George Heavner Erythropoietin conjugate compounds with extended half-lives
US20040266690A1 (en) 2003-05-30 2004-12-30 Chadler Pool Formation of novel erythropoietin conjugates using transglutaminase
WO2005001136A1 (fr) 2003-06-04 2005-01-06 Irm Llc Methodes et compositions pour la modulation de l'expression de l'erythropoietine
US20050008642A1 (en) 2003-07-10 2005-01-13 Yvo Graus Antibodies against insulin-like growth factor 1 receptor and uses thereof
US20050074821A1 (en) 2003-07-15 2005-04-07 Wild Kenneth D. Human anti-NGF neutralizing antibodies as selective NGF pathway inhibitors
WO2005017107A2 (fr) 2003-07-18 2005-02-24 Amgen Inc. Agents de liaison specifiques se liant a un facteur de croissance hepatocyte
US20050118643A1 (en) 2003-07-18 2005-06-02 Burgess Teresa L. Specific binding agents to hepatocyte growth factor
US20050019914A1 (en) 2003-07-24 2005-01-27 Aventis Pharma Deutschland Gmbh Perfusion process for producing erythropoietin
US7981669B2 (en) 2003-07-25 2011-07-19 Biovex Limited Viral vectors
WO2005021579A2 (fr) 2003-08-28 2005-03-10 Biorexis Pharmaceutical Corporation Peptides mimetiques epo et proteines de fusion
WO2005025606A1 (fr) 2003-09-09 2005-03-24 Warren Pharmaceuticals, Inc. Erythropoietines a action prolongee pouvant maintenir une activite de protection tissulaire d'une erythropoietine endogene
WO2005032460A2 (fr) 2003-09-30 2005-04-14 Centocor, Inc. Mimeticorps de noyau charniere mimetiques de l'epo humaine, compositions, procedes et applications correspondantes
WO2005081687A2 (fr) 2003-09-30 2005-09-09 Centocor, Inc. Mimeticorps de noyau-charniere humain, compositions, procedes et applications correspondantes
US20050112694A1 (en) 2003-11-07 2005-05-26 Carter Paul J. Antibodies that bind interleukin-4 receptor
WO2005047331A2 (fr) 2003-11-07 2005-05-26 Immunex Corporation Anticorps liant un recepteur de l'interleucine 4
US20050136063A1 (en) 2003-11-21 2005-06-23 Schering Corporation Anti-IGFR antibody therapeutic combinations
US20060111279A1 (en) 2003-11-24 2006-05-25 Neose Technologies, Inc. Glycopegylated erythropoietin
WO2005051327A2 (fr) 2003-11-24 2005-06-09 Neose Technologies, Inc. Erythropoietine glycopegylee
US20050143292A1 (en) 2003-11-24 2005-06-30 Defrees Shawn Glycopegylated erythropoietin
WO2005058967A2 (fr) 2003-12-16 2005-06-30 Pierre Fabre Medicament Nouveau recepteur hybride anti-insuline/igf-i ou recepteur hybride anti-insuline/igf-i et anticorps igf-ir et applications
WO2005063809A1 (fr) 2003-12-22 2005-07-14 Dubai Genetics Fz-Llc Erythopoietine identique a la forme naturelle
WO2005063808A1 (fr) 2003-12-31 2005-07-14 Merck Patent Gmbh Proteine hybride fc-erythropoietine a pharmacocinetique amelioree
US20050170457A1 (en) 2003-12-31 2005-08-04 Chadler Pool Novel recombinant proteins with N-terminal free thiol
US20050192211A1 (en) 2003-12-31 2005-09-01 Emd Lexigen Research Center Corp. Fc-erythropoietin fusion protein with improved pharmacokinetics
US20050158822A1 (en) 2004-01-20 2005-07-21 Insight Biopharmaceuticals Ltd. High level expression of recombinant human erythropoietin having a modified 5'-UTR
WO2005070451A1 (fr) 2004-01-22 2005-08-04 Zafena Aktiebolag Composition pharmaceutique comprenant une erythropoietine non glycosylee
US20050181482A1 (en) 2004-02-12 2005-08-18 Meade Harry M. Method for the production of an erythropoietin analog-human IgG fusion proteins in transgenic mammal milk
WO2005084711A1 (fr) 2004-03-02 2005-09-15 Chengdu Institute Of Biological Products Erythropoietine recombinante pegylee a activite in vivo
WO2005092369A2 (fr) 2004-03-11 2005-10-06 Fresenius Kabi Deutschland Gmbh Conjugues d'hydroxy-ethyl-amidon et d'erythropoietine
US20060002929A1 (en) 2004-03-23 2006-01-05 Khare Sanjay D Monoclonal antibodies
WO2005100403A2 (fr) 2004-04-09 2005-10-27 Abbott Laboratories Anticorps diriges contre le recepteur de l'erythropoietine et utilisations associees
US20050227289A1 (en) 2004-04-09 2005-10-13 Reilly Edward B Antibodies to erythropoietin receptor and uses thereof
WO2005103076A2 (fr) 2004-04-23 2005-11-03 Cambridge Antibody Technology Limited Variants d'erythropoietine
WO2006002646A2 (fr) 2004-07-07 2006-01-12 H. Lundbeck A/S Nouvelle erythropoietine carbamylee et son procede de production
WO2006013472A2 (fr) 2004-07-29 2006-02-09 Pierre Fabre Medicament Nouveaux anticorps anti-igf-ir et utilisations
WO2006029094A2 (fr) 2004-09-02 2006-03-16 Xencor, Inc. Derives d'erythropoietine a antigenicite modifiee
WO2006050959A2 (fr) 2004-11-10 2006-05-18 Aplagen Gmbh Molecules favorisant l'hematopoiese
WO2006069202A2 (fr) 2004-12-22 2006-06-29 Amgen Inc. Compositions et procedes impliquant des anticorps diriges contre le recepteur igf-1r
WO2006081171A1 (fr) 2005-01-24 2006-08-03 Amgen Inc. Anticorps anti-amyloide humanise
US20070110747A1 (en) 2005-05-03 2007-05-17 Ucb S.A. Binding agents
WO2006138729A2 (fr) 2005-06-17 2006-12-28 Imclone Systems Incorporated Antagonistes de recepteur pour le traitement de cancer osseux metastatique
WO2007000328A1 (fr) 2005-06-27 2007-01-04 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Anticorps se fixant à un épitope sur un récepteur de facteur de croissance insulinomimétique de type 1 et leurs utilisations
WO2007011941A2 (fr) 2005-07-18 2007-01-25 Amgen Inc. Anticorps neutralisants anti-b7rp1 humains
US20080166352A1 (en) 2005-07-18 2008-07-10 Amgen Inc. Human anti-B7RP1 Neutralizing Antibodies
WO2007012614A2 (fr) 2005-07-22 2007-02-01 Pierre Fabre Medicament Nouveaux anticorps anti igf-ir et leur utilisation
US7521048B2 (en) 2005-08-31 2009-04-21 Amgen Inc. TRAIL receptor-2 polypeptides and antibodies
US20090186022A1 (en) 2006-02-23 2009-07-23 Novartis Ag Organic Compounds
US20070253951A1 (en) 2006-04-24 2007-11-01 Gordon Ng Humanized c-Kit antibody
WO2007136752A2 (fr) 2006-05-19 2007-11-29 Glycofi, Inc. Compositions d'érythropoïétine
US20090234106A1 (en) 2006-09-08 2009-09-17 Amgen Inc. Anti-activin a antibodies and uses thereof
WO2008133647A2 (fr) 2006-11-07 2008-11-06 Merck & Co., Inc. Antagonistes de pcsk9
WO2008063382A2 (fr) 2006-11-07 2008-05-29 Merck & Co., Inc. Antagonistes de pcsk9
WO2008057458A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
WO2008057457A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
WO2008057459A2 (fr) 2006-11-07 2008-05-15 Merck & Co., Inc. Antagonistes de pcsk9
US8232372B2 (en) 2006-12-14 2012-07-31 Schering Corp. Engineered anti-TSLP antibody
WO2008125623A2 (fr) 2007-04-13 2008-10-23 Novartis Ag Molécules et procédés de modulation de proprotéine convertase subtilisine/kexine de type 9 (pcsk9)
US8101182B2 (en) 2007-06-20 2012-01-24 Novartis Ag Methods and compositions for treating allergic diseases
US7982016B2 (en) 2007-09-10 2011-07-19 Amgen Inc. Antigen binding proteins capable of binding thymic stromal lymphopoietin
WO2009055783A2 (fr) 2007-10-26 2009-04-30 Schering Corporation Anti-pcsk9 et méthodes de traitement de troubles du métabolisme lipidique et du cholestérol
WO2009100318A1 (fr) 2008-02-07 2009-08-13 Merck & Co., Inc. Antagonistes de 1b20 pcsk9
WO2009100297A1 (fr) 2008-02-07 2009-08-13 Merck & Co., Inc. Antagonistes de pcsk9 1d05
WO2010029513A2 (fr) 2008-09-12 2010-03-18 Rinat Neuroscience Corporation Antagonistes de pcsk9
WO2010077854A1 (fr) 2008-12-15 2010-07-08 Regeneron Pharamaceuticals, Inc. Anticorps humains à grande affinité contre pcsk9
WO2010075238A1 (fr) 2008-12-23 2010-07-01 Amgen Inc. Protéines de liaison au récepteur cgrp humain
WO2011037791A1 (fr) 2009-09-25 2011-03-31 Merck Sharp & Dohme Corp. Antagonistes de pcsk9
WO2011053759A1 (fr) 2009-10-30 2011-05-05 Merck Sharp & Dohme Corp. Antagonistes de la pcsk9 avec anticorps fab ax189 et ax1, et variantes afférentes
WO2011053783A2 (fr) 2009-10-30 2011-05-05 Merck Sharp & Dohme Corp. Antagonistes et variants ax213 et ax132 pcsk9
WO2011072263A1 (fr) 2009-12-11 2011-06-16 Irm Llc Antagonistes de pcsk9
WO2011111007A2 (fr) 2010-03-11 2011-09-15 Rinat Neuroscience Corporation Anticorps présentant une liaison à l'antigène dépendante du ph
WO2012054438A1 (fr) 2010-10-22 2012-04-26 Schering Corporation Anti-pcsk9
WO2012088313A1 (fr) 2010-12-22 2012-06-28 Genentech, Inc. Anticorps anti-pcsk9 et procédés d'utilisation
WO2012101253A1 (fr) 2011-01-28 2012-08-02 Sanofi Compositions pharmaceutiques comprenant des anticorps humains contre pcsk9
WO2012101251A1 (fr) 2011-01-28 2012-08-02 Sanofi Anticorps humains dirigés contre la pcsk9 destinés à être utilisés dans des procédés de traitement basés sur des schémas posologiques particuliers
WO2012101252A2 (fr) 2011-01-28 2012-08-02 Sanofi Anticorps humains contre pcsk9 pour utilisation dans des procédés de traitement de groupes particuliers de sujets
WO2012109530A1 (fr) 2011-02-11 2012-08-16 Irm Llc Antagonistes de pcsk9
US20130064825A1 (en) 2011-05-10 2013-03-14 Amgen Inc. Methods of treating or preventing cholesterol related disorders
US20190344257A1 (en) * 2011-07-22 2019-11-14 Vanrx Pharmasystems Inc. Fill needle system
WO2014099984A1 (fr) 2012-12-20 2014-06-26 Amgen Inc. Agonistes du récepteur apj et leurs utilisations
US20140274874A1 (en) 2013-03-14 2014-09-18 Amgen Inc. Variants of tissue inhibitor of metalloproteinase type three (timp-3), compositions and methods
WO2014152012A2 (fr) 2013-03-14 2014-09-25 Amgen Inc. Variants d'inhibiteur tissulaire de la métalloprotéinase type iii (timp-3), compositions et procédés

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
CAS, no. 501423-23-0
COHEN ET AL., CLINICAL CANCER RES., vol. 11, 2005, pages 2063 - 2073
LEI ET AL., WORLD J. GASTROENTEROL., vol. 19, 2013, pages 5138 - 5143
LU ET AL., J. BIOL. CHEM., vol. 279, 2004, pages 2856 - 2865
MALONEY ET AL., CANCER RES., vol. 63, 2003, pages 5073 - 5083
THAKUR ET AL., MOL. IMMUNOL., vol. 36, 1999, pages 1107 - 1115
VARGEHES ET AL., CANCER GENE THER., vol. 9, no. 12, 2002, pages 967 - 978

Also Published As

Publication number Publication date
JP2024523779A (ja) 2024-07-02
US20240208680A1 (en) 2024-06-27
KR20240011135A (ko) 2024-01-25
IL307418A (en) 2023-12-01
CN117320964A (zh) 2023-12-29
CL2023003444A1 (es) 2024-07-12
AU2022279223A1 (en) 2023-10-19
BR112023024278A2 (pt) 2024-01-30
MX2023013640A (es) 2023-11-30
CA3217207A1 (fr) 2022-11-24
EP4341161A1 (fr) 2024-03-27

Similar Documents

Publication Publication Date Title
US20210361869A1 (en) Injector and method of assembly
US11305056B2 (en) Needle insertion-retraction system having dual torsion spring system
US11957883B2 (en) Plunger rod and syringe assembly system and method
US11285266B2 (en) Drug delivery device having minimized risk of component fracture upon impact events
AU2015284463B2 (en) Autoinjector with low energy plunger loading
TWI846741B (zh) 用於藥物遞送裝置之平台組裝方法
US12109389B2 (en) Fluid path assembly for a drug delivery device
JP7510952B2 (ja) シリンジ滅菌確認アセンブリ及び方法
US20240208680A1 (en) Method of optimizing a filling recipe for a drug container
US20220031939A1 (en) Drug delivery devices with partial drug delivery member retraction
CA3170808A1 (fr) Seringue a distribution controlee
US11213620B2 (en) Drug delivery devices with partial drug delivery member retraction

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22731368

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: AU2022279223

Country of ref document: AU

Ref document number: 2022279223

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 307418

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 3217207

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2022279223

Country of ref document: AU

Date of ref document: 20220519

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 18288549

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2023569877

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 202280035596.1

Country of ref document: CN

Ref document number: MX/A/2023/013640

Country of ref document: MX

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112023024278

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 202393224

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 2022731368

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022731368

Country of ref document: EP

Effective date: 20231221

WWE Wipo information: entry into national phase

Ref document number: 11202308830Y

Country of ref document: SG

ENP Entry into the national phase

Ref document number: 112023024278

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20231121