CA2713128A1 - Pharmaceutical dosage form - Google Patents

Pharmaceutical dosage form Download PDF

Info

Publication number
CA2713128A1
CA2713128A1 CA2713128A CA2713128A CA2713128A1 CA 2713128 A1 CA2713128 A1 CA 2713128A1 CA 2713128 A CA2713128 A CA 2713128A CA 2713128 A CA2713128 A CA 2713128A CA 2713128 A1 CA2713128 A1 CA 2713128A1
Authority
CA
Canada
Prior art keywords
dosage form
pharmaceutical dosage
form according
front side
back side
Prior art date
Legal status (The legal status 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 status listed.)
Granted
Application number
CA2713128A
Other languages
French (fr)
Other versions
CA2713128C (en
Inventor
Eugeen Marie Jozef Jans
Filip Rene Irena Kiekens
Jody Firmin Marceline Voorspoels
Anne Faure
Elisabeth Arkenau-Maric
Lutz Barnscheid
Johannes Bartholomaeus
Marc Frevel
Eric Galia
Iris Ziegler
Andrea Schuessele
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Gruenenthal GmbH
Original Assignee
Individual
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=40511977&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2713128(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Publication of CA2713128A1 publication Critical patent/CA2713128A1/en
Application granted granted Critical
Publication of CA2713128C publication Critical patent/CA2713128C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2031Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2059Starch, including chemically or physically modified derivatives; Amylose; Amylopectin; Dextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/24Structurally defined web or sheet [e.g., overall dimension, etc.]
    • Y10T428/24479Structurally defined web or sheet [e.g., overall dimension, etc.] including variation in thickness

Abstract

The invention relates to a pharmaceutical dosage form, preferably with controlled release of a pharmacologically active compound (A) contained therein, the pharmaceutical dosage form very preferably being tamper-resistant and most preferably having a breaking strength B1 of at least 500 N in direction of extension E1 and having a breaking strength B2 of less than 500 N in direction of extension E2.

Description

Pharmaceutical dosage form FIELD OF THE INVENTION

The invention relates to a pharmaceutical dosage form, preferably a tablet for oral administration.

BACKGROUND ART

For many pharmaceutically active compounds it is still preferred to have them orally administered by way of tablets. It is also well known that depending on how a pharmaceutically active ingredient is formulated into a tablet its release pattern can be modified. In this regard, tablets providing a retarded release profile are of primary importance. With retarded release tablets care has to be taken that under no circumstances the pharmaceutically active ingredient will be released completely and instantaneously in an uncontrolled manner ("dose-dumping") since regularly the dosage used for retarded release tablets is much higher than for non-retarded release tablets. This may cause serious adverse effects or even death depending on the active ingredient and potency thereof.

Controlled release (e.g. delayed release, prolonged release, sustained release, and the like) may be based upon various concepts such as coating the pharmaceutical dosage form with a controlled release membrane, embedding the pharmacologically active compound in a matrix, binding the pharmacologically active compound to an ion-exchange resin, forming a complex of the pharmacologically active compound, and the like. In this context it can be referred to, e.g., W.A. Ritschel, Die Tablette, 2. Auflage, Editio Cantor Verlag Aulendorf, 2002.

In WO 01/97783 Al a controlled-release, in particular a retarded release oral drug dosage form for releasing a drug into at least a portion of a region defined by the stomach and the front gastrointestinal tract is obtained by employing a solid monolithic matrix having a non-circular shape wherein the longitudinal axis has a maximum length of 3.0 cm and the transversal axis achieves a minimum length of 1.2 cm within one hour of immersion in water. Furthermore, it is required that the matrix has a shape which when projected onto a plane is either an oval or a parallelogram.

CONFIRMATION COPY
2 US 4,353,887 discloses a divisible tablet which exhibits controlled and delayed release of an active substance. This is achieved by use of a coating on a compacted tablet having an oblong shape in which the ratio of length to width to depth is approximately 2.5 to 5:

oppiroxi.o atel.. A to 2: and the idth constitute = t 2 /n of the I _ -- _I'_ in "Htlvnnllaicly U.a w c. I a1 1U LIIV VVIUUI %U1I Lit L at IIIVJI G/J VI
Lilt,- It:-[1 L dUUIUOfI, one or more relatively deep dividing grooves have to be present which run perpendicular to the length and depth and have a total depth of from approximately 1/3 to approximately 1/2 of the depth of the tablet.

WO 01/08661 is directed to a controlled release formulation, capable of providing sustained, prolonged, repeat and/or delayed release of oxycodone.

WO 03/094812 relates to abuse-resistant opioid-containing solid dosage pharmaceuticals comprising a sustained release carrier and an analgesically effective amount of an opioid analgesic in combination with an opioid euphoria-inhibiting amount of an isolated nontoxic N-methyl-D-aspartate receptor antagonist which is substantially not released when the dosage form is administered intact.

WO 99/48481 discloses a tablet for the controlled release of an active pharmaceutical ingredient. The tablet comprises a core having a donut-like configuration with a cylindrical hole extending through the center of the core. The core is coated with a hydrophobic, water-insoluble material covering all of the core except that which is defined by the cylindrical hole.

GB-A 2 057 878 discloses a divisible tablet having controlled and delayed release of the active substance, consisting of a compact that is formed by at least one active substance in an adjunct composition that effects a delayed and controlled release of the active substance, the compact being of an oblong shape in which the ratio of length to width to depth is approximately 2.5 to 5: approximately 0.9 to 2:1 and the width constitutes at most 2/3 of the length, and in which one or more relatively deep dividing grooves are present which run perpendicularly to the length and depth and have a total depth of from approximately 113 to approximately '12 of the depth of the tablet, but are at least so deep that one fracture surface area multiplied by the number of possible fragments constitutes a maximum of 15% of the surface area of the undivided tablet, the base and top faces independently of one another are planar or are convexly curved about the longitudinal axis or about parallels to this axis, the side faces are planar, the end faces can be of any shape and edges are optionally bevelled or rounded.
3 DE-A 198 56 147 discloses a solid, elongate dosage form with a long axis and with a length which is defined by projection of the ends of the dosage form onto the long axis, where a cross-sectional area oriented perpendicular to the long axis has an area which is variable along the long axis and increases from a cross-sectional area which is located between the ends and has a minimal area essentially continuously toward the two ends up to in each case a cross-sectional area with a maximal area, wherein the distance of the maximal cross-sectional area which is located near one end from the maximal cross-sectional area which is located near the other end is, projected on the long axis, more than half the length of the dosage form.

DE 28 08 505 C2 discloses a tablet which dissolves at essentially constant speed and which contains a water soluble component as well as a water insoluble coating.
It is required that the side of the tablet has to comprise one or more cavities having a breadth of 0.1 to 1.0 mm, a depth of 0.1 to 0.4 mm and a length of more than 0.1 mm, wherein the side of said cavities is less than 1/6 of the total side of the tablet.

In DE 692 29 881 T2 it is proposed to obtain a tablet having a retarded release profile by use of a water soluble gel as well as of a specific coating having a specific thickness. The coating has to contain either ethylcellulose or acetylcellulose and has to be water insoluble as well as insoluble in gastric liquids.

A tablet having a controlled release profile will according to WO 99/48481 Al be obtained by use of a doughnut-shaped core material with a cylindrical hole. A
hydrophobic, water-insoluble coating is applied to the doughnut-shaped core except for the side of said core which surrounds the cylindrical hole. In such a manner an inherent limitation of compressed monolithic tablets for extended release dosages shall be overcome, namely the increase in diffusion length resistance over time due to the insolubility of the polymer.
Apparently, this problem has been solved by insuring that the inner exposed area of the doughnut-shaped configuration is clear of any coating.

In DE 42 39 085 Al it is described to make use of an oblong tablet only the opposed edges of which are in contact with an underlying side but not the intermediate section. In this manner the tablet is easily divisible by use of one hand only. The two parts forming such an oblong tablet may have the form of spherical sectors.

It is well known that a pharmaceutical formulation or its mode of manufacture, e.g. for an oral dosage form, might undergo modifications during clinical testing, for example with
4 respect to the ingredients used or to the relative amounts of the excipients, or with respect to the reaction conditions and reactants used during manufacture. Frequently, such modifications at least to some extent have an impact on the release profile of pharmaceutically active ingredients. This is particularly unpleasant if for a specific formulation an approved optimized release profile has already been found which can not be reproduced with the modified formulation. In such a case, the clinical tests have either to be interrupted or have to be started from the beginning. Given the huge expenditures necessary to bring a new drug formulation up to and through clinical testing the above scenario has indeed proven to be rather unsatisfactory.

Pharmaceutical dosage forms having an increased breaking strength (resistance to crushing) have been recently reported. Dosage forms of this type may also exhibit a certain degree of controlled release of the pharmacologically active compound contained therein.
The major advantage of such pharmaceutical dosage forms is that comminuting, particularly pulverization, by conventional means, such as grinding in a mortar or fracturing by means of a hammer, is impossible or at least substantially impeded.

On the one hand, pharmaceutical dosage forms having an increased breaking strength are useful for avoiding drug abuse of the pharmacologically active compound contained therein.
Many pharmaceutical active compounds, in addition to having excellent activity in their appropriate application, also have abuse potential, i.e., they can be used by an abuser to bring about effects other than those intended. Opiates, for example, which are highly active in combating severe to very severe pain, are frequently used by abusers to induce a state of narcosis or euphoria. In order to make abuse possible, the corresponding pharmaceutical dosage forms, such as tablets or capsules are comminuted, for example ground in a mortar, by the abuser, the active compound is extracted from the resultant powder using a preferably aqueous liquid and the resultant solution, optionally after being filtered through cotton wool or cellulose wadding, and is administered parenterally, in particular intravenously. An additional phenomenon of this kind of administration, in comparison with abusive oral administration, is a further accelerated increase in active compound levels giving the abuser the desired effect, namely the "kick" or "rush". This kick is also obtained if the powdered pharmaceutical dosage form is administered nasally, i.e. is sniffed. Since controlled-release pharmaceutical dosage forms containing active compounds with abuse potential do not give rise to the kick desired by the abuser when taken orally even in abusively high quantities, such pharmaceutical dosage forms are also comminuted and extracted in order to be abused. Pharmaceutical dosage forms exhibiting an increased breaking strength, however, may not be powdered by conventional means and thus, cannot be administered nasally thereby avoiding drug abuse. In the context of such tamper resistant dosage forms, it can be referred to, e.g., WO 2005/016313, WO
2005/016314, WO 2005/ 063214, WO 2005/102286, WO 2006/002883, WO 2006/002884, WO
2006/002886, and WO 2006/082097.

These dosage forms have a breaking strength of at least 500 N in every direction of extension.

On the other hand, pharmaceutical dosage forms having an increased breaking strength are useful for avoiding an (unintentional) overdose of the pharmacologically active compound contained therein, which overdose would otherwise be caused by diminishing the retardant effect due to pulverization. It is known that many patients, particularly older patients frequently have difficulties in taking solid pharmaceutical dosage forms, such as tablets, gelatine capsules, etc. They choke on them and sometimes develop pronounced aversion to such pharmaceutical dosage forms. To counter this problem, various apparatuses have been developed by means of which conventional solid pharmaceutical dosage forms may be comminuted or pulverized ("tablet crushers"). Such apparatuses are used, for example, by the care staff in old people's homes. The pharmaceutical dosage forms are then administered to the people being cared for not as tablets etc.
but rather as powder, for example to get round the difficulties involved in swallowing tablets. However, the comminution of pharmaceutical dosage forms with such apparatuses is problematic if the pharmaceutical dosage forms are prolonged-release formulations. As a rule, comminution results in destruction of the inner structure of the pharmaceutical dosage form, which is responsible for the prolonged release, so doing away with the prolonged-release action. Consequently, after administration, frequently all the physiologically active substance originally contained in the pharmaceutical dosage form is released in a relatively short time, whereby a comparatively very high plasma concentration of the substance is abruptly reached within a relatively short time frame. In this way, the originally prolonged-release formulations become immediate release formulations. Depending on the physiological activity of the substance, this may cause considerable side-effects however, and in extreme cases may even lead to the death of the patient. Pharmaceutical dosage forms having an increased breaking strength, however, cannot be comminuted by tablet crushers and thus, have to be swallowed as a whole thereby avoiding any (unintentional) overdose. In this context, it can be further referred to, e.g., WO
2006/082099.

These dosage forms also have a breaking strength of at least 500 N in every direction of extension.

The release profile of controlled-release formulations depends on a variety of factors, such as properties of the pharmaceutical dosage form per se, nature and content of the matrix, nature of the release medium, nature and content of the active compound, nature and content of further pharmaceutical excipients as well as the interrelationship of these factors.
When the control of the release profile relies on a polymer matrix in which the active compound is embedded, the release rate depends on the properties of the pharmaceutical dosage form as such, e.g. its geometry, method of manufacture, additives and excipients contained therein, and the like. Further, the release rate depends on the properties of the matrix polymer, such as molecular weight, viscosity, particle properties, interaction with other polymers, chain entanglements, degree of cross-linking, chemical nature of monomer units, interaction of the matrix material with the release medium (e.g., swelling and gelling), and the like. Still further, the release rate depends on the properties of the active compound, e.g., its dose, particle size, particle form and its solubility in the release medium, which in turn is a function of various properties, such as molecular size, molecular weight, ionogenicity, acidity, steric hindrance, arrangement of dipols, hydrophilicity, etc.
Furthermore, the release rate depends on the individual interactions of a given matrix material with a given active compound (cf. Ning Wu et at., Journal of Controlled Release 102 (2005) 569-81; V.S. Manthena et at., Am J Drug Deliv. 2004 2(1) 43-57).

The release profile of conventional pharmaceutical dosage forms that do not exhibit an increased breaking strength can usually be adjusted within certain limits, usually by the variation of the content and/or the nature of the pharmaceutical excipients, such as the matrix forming polymer.

In some cases it has also been reported that the release of a drug in the body can be controlled by the surface area to volume ratio of a conventional dosage form which does not exhibit an increased breaking strength. For example, US 5,427,798 discloses film coated tablets containing bupropion hydrochloride and having a surface area to tablet volume of 3:1 to 25:1 cm-1 for tablets of 50, 100 and 150 mg drug content.
Similarly, US
4,940,556 and US 5,198,226 disclose spheroids containing dihydropyridine calcium channel blockers and having area radius to circumference radius ratios in the range of 0.85 to 1Ø

With respect of pharmaceutical dosage forms exhibiting an increased breaking strength, however, the variation of the content, the nature of the pharmaceutical excipients and/or the surface area to volume ratio also affects the mechanical properties. This is because the increased breaking strength of the pharmaceutical dosage form typically relies on the presence of a particular polymer that is processed by a particular method when manufac-turing the pharmaceutical dosage form. It seems that said polymer also serves as a matrix embedding the pharmacologically active compound. In consequence, the polymer matrix that is essential to the breaking strength of the pharmaceutical dosage form simultaneously serves as a controlled release matrix and thus, varying the content, nature and/or spacial distribution of the polymer causes both, a change of the release profile as well as a change of the mechanical properties of the pharmaceutical dosage form.

Particular problems arise when the dose of the pharmacologically active compound and thus, also the total weight of the pharmaceutical dosage form is comparatively high.
Depending upon the content and the nature of the pharmacologically active compound and of the pharmaceutical excipients, the retardant effect of the polymer may be so strong that the pharmaceutical dosage form cannot be adapted to a specific dosing regimen, e.g., twice daily, particularly when the increased breaking strength is to be maintained.

On the one hand, a decrease of the content of the retardant polymer for the purpose of accelerating drug release would substantially affect the mechanical properties of the pharmaceutical dosage form and in a worst case scenario, would completely diminish its specific and unique mechanical properties (breaking strength). Further, a decrease of the content of the matrix polymer beyond a certain limit may cause a deterioration or even loss of other desired properties, such as storage stability. A poor storage stability results, e.g., in a change of the release profile over time.

On the other hand, the addition of non-retardant pharmaceutical excipients (auxiliaries) for the purpose of weakening the retardant effect of the retardant polymer would increase the total weight of the dosage form. As highly dosed pharmaceutical dosage forms have comparatively high total weights anyway, a further increase of the total weight is disadvantageous and could deteriorate patient compliance (e.g.
swallowability).

Thus, there is a demand for pharmaceutical dosage forms, particularly tamper-resistant pharmaceutical dosage forms, the release profile of which may be varied within certain limits without diminishing the tamper resistance and without deteriorating the compliance of the pharmaceutical dosage form.

It is an object of the invention to provide pharmaceutical dosage forms having advantages compared to pharmaceutical dosage forms of the prior art.

It is another object of the present invention to provide an oral dosage form which does not exhibit the shortcomings of the dosage forms of the state of the art and which in particular allows for an improved control of the release profile of the active pharmaceuticai ingredient incorporated in the oral dosage form. It has been another object of the present invention to be able to regain the original release profile of an elaborated formulation for an oral dosage form, the release pattern of which has experienced changes due to modifications to the composition of said formulation.

These objects have been solved by the subject-matter described hereinbelow.
SUMMARY OF THE INVENTION

The present invention relates in a first aspect to a pharmaceutical dosage form, especially an oral dosage form, particularly a tablet, comprising at least one pharmaceutically active ingredient, and having a shape comprising a longitudinal axis and two opposite longitudinal edges, a transversal axis perpendicular to the longitudinal axis and two opposite transversal edges, a front side, an opposite back side and a circumferential rim between said front and back side, wherein the front side and/or the back side comprise a basis area and wherein the front side and/or the back side comprise at least one bulge which extends above said basis area, said at least one bulge being present at and/or adjacent to at least a section of one or both longitudinal edges and/or at and/or adjacent to at least a section of one or both transversal edges and/or between both longitudinal edges and both transversal edges. The front side and/or the back side of the dosage form, in particular the basis area of the front side and/or the basis area of the back side, can further comprise at least one indentation.
The present invention relates in a second aspect to a tamper-resistant pharmaceutical dosage form having a retarded release profile, especially a tamper-resistant oral dosage form having a retarded release profile, particularly a tamper-resistant tablet having a retarded release profile, comprising at least one pharmaceutically active ingredient with potential for abuse, and having a shape comprising a longitudinal axis and two opposite longitudinal edges, a transversal axis perpendicular to the longitudinal axis and two opposite transversal edges, a front side, an opposite back side and a circumferential rim between said front and back side, wherein the front side and/or the back side comprise a basis area and wherein the front side and/or the back side comprise at least one bulge which extends above said basis area, said at least one bulge being present at and/or adjacent to at least a section of one or both longitudinal edges and/or at and/or adjacent to at least a section of one or both transversal edges and/or between both longitudinal edges and both transversal edges. The front side and/or the back side of the dosage form, in particular the basis area of the front side and/or the basis area of the back side, can further comprise at least one indentation.

The present invention relates in a third aspect to a pharmaceutical dosage form with controlled release of a pharmacologically active compound (A) contained therein, the pharmaceutical dosage form having a breaking strength B, of at least 500 N in direction of extension E, and having a breaking strength B2 of less than 500 N in direction of extension E2.

The pharmaceutical dosage forms according to the invention preferably exhibit anisotropic mechanical properties (i.e., different mechanical properties in different directions).

It has been surprisingly found that by modifying the outer shape of the pharmaceutical dosage form the release profile may be modified without simultaneously diminishing the breaking strength of the pharmaceutical dosage form. In particular, it has been surprisingly found that in spite of a modified outer shape of the pharmaceutical dosage form which causes a certain degree of fragility, the overall tamper resistance of the pharmaceutical dosage form can be maintained.

Furthermore, it has been surprisingly found that by modifying the outer shape of the pharmaceutical dosage form the storage stability, e.g. the storage stability of the release profile, can be increased compared to conventional dosage forms having a comparable release profile before storage.

With the pharmaceutical dosage forms of the invention, it is possible to have in general a greater control on the release profile of pharmaceutically active ingredients.
The release profile can be finely adjusted or tailored in a more accurate, predictable and reliable manner; the release profile can be manipulated or tailored so that a variety of retarded release profiles can be provided for the same formulation. Control of the release profile with the pharmaceutical dosage forms of the present invention can be achieved for pharmaceutical dosage forms designed for immediate release or pharmaceutical dosage forms designed for retarded (sustained) release. It is also an advantage that it is no longer necessary to rely on the choice and amount of hydrophilic polymer(s) to modify the dissolution profile of a retarded release formulation which in various circumstances has proven to not even be possible under realistic conditions.

BRIEF DESCRIPTION OF THE DRAWINGS

. r.
4 Al L.....+atic a.... of tablet of th.. invention. r:..--.r-j ij a jv1,aiiiLi~~ LIJ~! vIGVV VI Q IaLIL VI L1IG ILlruyuiC Figure ID) IS
d l.IUSS-sectional view along A-A of the tablet of Figure 1A).

Figure 2A) is a schematic top view of another embodiment of the tablet of the invention.
Figure 2B) is a cross-sectional view along A-A of the tablet of Fig. 2A).

Figure 3A) is a schematic top view of another embodiment of a tablet of the invention.
Figure 3B) is a cross-sectional view along line A-A of the tablet of Figure 3A). Figure 3C) is a cross-sectional side view along B-B of the tablet of Figure 3A).

Figure 4A) is a schematic top view of another embodiment of a tablet of the invention.
Figure 4B) is a cross-sectional view along line A-A of the tablet of Figure 4A). Figure 4C) is a cross-sectional side view along B-B of the tablet of Figure 4A).

Figure 5A) is a schematic top view of another embodiment of a tablet of the invention.
Figure 5B) is a cross-sectional view along line A-A of the tablet of Figure 5A). Figure 5C) is a cross-sectional side view along B-B of the tablet of Figure 5A).

Figure 6 is a schematic top view of another embodiment of the tablet of the invention.
Figure 6B) is a cross-sectional view along A-A of the tablet of Figure 6A).
Figure 6C) is a cross-sectional side view along B-B of the tablet of Figure 6A).

Figure 7A) is a schematic view of a conventional oblong tablet. Figure 7B) is a schematic view of an embodiment of a pharmaceutical dosage form according to the invention having an increased ratio of surface to volume and surface to weight, respectively, achieved by a taper right in the middle of the pharmaceutical dosage form.

Figure 8A) is a schematic view of a preferred pharmaceutical dosage form according to the invention having two recesses on opposing sides ("inner courtyards"). Figure 8B) is a schematic view of the cross-section of the pharmaceutical dosage form depicted in Figure 8A) showing that the face of the cross-section assumes the shape of a H.

Figure 9A) is a schematic view of the cross-sectional face of the pharmaceutical dosage form depicted in Figure 8A). Figure 9B) is a schematic view of the cross-sectional face of a pharmaceutical dosage form according to the invention that is similar to the cross-sectional face of the pharmaceutical dosage form depicted in Figure 9A). Figure 9C) is a schematic view of the cross-sectional face of a pharmaceutical dosage form according to the invention +6..,+ . .I.,r to +L.e +',..,al ---- 4...--- aceua=-_i J----.~ C--.--- J--:-1-J
L1 IcA ii .iI ..iiai w u I cross-sectional ~G~ IQ'...G Of 1 J 1 IQI I I
IQIiGUULiOI U L J Z ) O y . C IUI I I IJ UCPIULUU III
Figures 9A) and 9B).

Figure 10 is a schematic view of a particularly preferred pharmaceutical dosage form according to the invention. Figure 10A) is a top view, Figure 1OB) is a side view.

Figure 11 is a schematic view of a conventional pharmaceutical dosage form comprising a cylindrical central element and two spherical caps.

Figure 12 is a schematic view of a cross-section of a preferred pharmaceutical dosage form (cf. Figure 8A)) divided into voxels of identical volume and surface to roughly estimate the overall surface of the dosage form.

Figure 13 is a schematic view of preferred pharmaceutical dosage forms according to the invention. The pharmaceutical dosage form depicted in Figure 13A) assumes the shape of a cross, the pharmaceutical dosage form depicted in Figure 13B) assumes the shape of a star.

Figure 14 is a schematic view of various pharmaceutical dosage forms according to the invention. The pharmaceutical dosage form depicted in Figure 14A) assumes the shape of an eight, the pharmaceutical dosage form depicted in Figure 14B) assumes the shape of a flattened peanut, the pharmaceutical dosage form depicted in Figure 14C) assumes the shape of a parallel double cylinder, the pharmaceutical dosage form depicted in Figure 14D) assumes the shape of a parallel double tube, the pharmaceutical dosage form depicted in Figure 14E) assumes the shape of a square with a round hole in the middle, and the pharmaceutical dosage form depicted in Figure 14F) assumes the shape of a wavy or corrugated item.

Figure 15 is a schematic view of preferred pharmaceutical dosage forms according to the invention. The pharmaceutical dosage form depicted in Figure 15A) assumes the shape of a short tube, the pharmaceutical dosage form depicted in Figure 15B) assumes the shape of a ring.

Figure 16 is a schematic view of preferred pharmaceutical dosage forms according to the invention. The pharmaceutical dosage form depicted in Figure 16A) assumes the shape of a triangle with two recesses on opposing sides, the pharmaceutical dosage form depicted in Figure 1 6B) assumes the shape of a pentagon with two recesses on opposing sides.

Figure 17 is a schematic view of the cross-sectional face of the preferred pharmaceutical dosage form depicted in Figure 10. In Figure 17A) the maximum extension (56) of the dosage form orthogonal to the main area of extension (57) of the dosage form is spaced from the centre of mass (58) of the dosage form parallel to said main area of extension (57). In Figure 17B) the centre of mass (58) is surrounded by concentric rings (60a) to (60d) indicating increasing distances from the centre of mass.

Figure 18 is a schematic view of the pharmaceutical dosage form shown in Figure 10 and the set-up for measuring the breaking strength in directions of extension E,, E2 and E3, respectively. Figure 18A) shows how the pharmaceutical dosage form should be placed between the two plain jaws (61a) and (61b) of the measuring device in order to measure the breaking strength in direction of extension E,. Figure 18B) shows how the pharmaceu-tical dosage form should be placed between the two plain jaws (61a) and (61b) of the measuring device in order to measure the breaking strength in direction of extension E2.
Figure 18C) shows how the pharmaceutical dosage form should be placed between the two plain jaws (61a) and (61b) of the measuring device in order to measure the breaking strength in direction of extension E3.

Figure 19 is a preferred embodiment of the measuring system that is used in order to measure the breaking strength of the pharmaceutical dosage form according to the invention. The two jaws (61 a) and (61 b) of this measuring system are not plain, but contain an embossment (62) and a cavity (63, 64), respectively.

Figure 20A) shows how the pharmaceutical dosage form shown in Figure 10 should be placed between the two jaws (61a) and (61b) of the measuring device shown in Figure 19 in order to measure the breaking strength in direction of extension El. Figure 20B) shows how the pharmaceutical dosage form should be placed between the two jaws (61a) and (61b) of the measuring device in order to measure the breaking strength in direction of extension E2. Figure 20C) shows how the pharmaceutical dosage form should be placed between the two jaws (61a) and (61b) of the measuring device in order to measure the breaking strength in direction of extension E3.

Figure 21 shows the release profile of the tablet according to inventive example 1-2 (H-shape) and of the tablet according to comparative example C-1 (oblong) immediately after manufacture.

Figure 22 shows the release profile of the tablet according to inventive example I-1 (H-shape) and of the tablet according to inventive example 1-2 (H-shape).

Figure 23 shows the release profile of the tablet according to comparative example C-1 (oblong) before and after storage under various conditions (40 C, 6 months;
25 C, 9 months; and 30 C, 9 months, respectively).

Figure 24 shows that the release profile of the tablet according to inventive example 1-2 (H-shape) before and after storage under various conditions (40 C, 6 months; 25 C, 9 months; and 30 C, 9 months, respectively).

DETAILED DESCRIPTION OF THE INVENTION

Unless expressly stated otherwise, any preferred embodiment of the invention that will be described in connection with a particular aspect of the invention hereinafter shall also apply to the other aspects of the invention. In this regard, embodiments that have been described in terms of key words that are synonymous to or at least partially overlap with similar key words mentioned elsewhere in the specification, such as "pharmaceutically active ingredient" and "pharmacologically active compound (A)", or "pharmaceutical dosage form" and "tablet", shall be understood as being also applicable in terms of said similar key words.

A first aspect of the invention relates to a pharmaceutical dosage form, especially an oral dosage form, particularly a tablet, comprising at least one pharmaceutically active ingredient (pharmacologically active compound), and having a shape comprising a longitudinal axis and two opposite longitudinal edges, a transversal axis perpendicular to the longitudinal axis and two opposite transversal edges, a front side, an opposite back side and a circumferential rim between said front and back side, wherein the front side and/or the back side comprise a basis area and wherein the front side and/or the back side comprise at least one bulge which extends above said basis area, said at least one bulge being present at and/or adjacent to at least a section of one or both longitudinal edges and/or at and/or adjacent to at least a section of one or both transversal edges and/or between both longitudinal edges and both transversal edges. The front side and/or the back side of the dosage form, in particular the basis area of the front side and/or the basis area of the back side, can further comprise at least one indentation.

Although it is also possible that the longitudinal axis and the transversal axis of the tablet have essentially the same length, it is preferred that the tablet of the invention has a longitudinal axis being longer than its transversal axis. That is, preferred embodiments of the tablet of the invention exhibit an oblong shape. The longitudinal axis is typically extending through the middle part of the tablet between both opposing longitudinal edges from one transversal edge to the opposite transversal edge, in particular in such a way that its length is maximized. The transversal axis is typically extending from one longitudinal edge to the opposite longitudinal edge, in particular in such a way that its length is maximized. The transversal axis is oriented perpendicular to the longitudinal axis.

The basis area of the front side and/or the back side of the tablet of the invention does not necessarily have to be flat, but can in one embodiment exhibit an irregular or regular three dimensional pattern, which, however, is not extending to any significant degree towards the dimension of a bulge or an indentation.

The average distance between the front basis area and the back basis area of one embodiment of the tablet of the invention usually is smaller than the length of its transversal axis. Those opposite sides of the tablet which have the smallest average distance are usually comprising the front and the back basis areas.

According to another preferred embodiment, a tablet is provided, wherein the front side and the back side each comprise at least one bulge at least along a section at and/or adjacent to both longitudinal edges and/or at least along a section at and/or adjacent to both transversal edges. In this respect it is even more preferred in certain cases that said front side and said back side comprise an at least essentially continuous bulge at and/or adjacent to at least two third of both opposite longitudinal edges and/or at and/or adjacent to at least two third of both opposite transversal edges.

The bulge may have any geometric cross-section, and can, for example, be rounded or can have a rectangular, triangular or square cross-section. The bulges preferably have a width which is less than half the width, more preferably less than one third of the width of the tablet. The length of the bulges can vary to a great extent. It is preferred that the overall length of an individual bulge is at least one half of the length of the longitudinal edge or of the transversal edge, depending on its location. Typically, the overall length of a bulge is much longer than its width, e.g. several times the width of the bulge, such as more than 2, 3, 4, 5 or 6 times of its width, in particular when oriented in the longitudinal direction, or more than 2, 3 or 4 times of its width, in particular when oriented in the transversal direction. A bulge in the meaning of the present invention shall also comprise a series of adjacent bulge portions. These bulge portions, when viewed from above, can, for example, have the circumferential form of a circle, an oval, a rectangle, a square, a triangle or any other polygonal form, or may come close to these forms, or may even have an irregular form.

A bulge which is located at a longitudinal and/or at a transversal edge regularly passes over from the circumferential rim of the tablet without a significant transition zone or transition step, i.e. without a "land". In such' an embodiment there is a smooth transition from the rim part to the bulge part so that the outer surfaces of the rim and the bulge form a continuous surface at least over a section. A bulge which is positioned adjacent to a longitudinal or adjacent to a transversal edge is in contrast thereto not directly placed at the circumferential rim of the tablet but is separated from the rim in the plane of the basis area by a portion, in particular a minor portion, which can be attributed to be part of the basis area. Said minor portion is known in the field of tablet technology as "the land". This minor area usually has a width being smaller than the average width of the bulge itself. In a preferred embodiment, the land is in the range from about 0.05 mm to about 0.5 mm, e.g. about 0.1 mm.

In a particularly suitable embodiment, the tablet of the invention is provided with bulges at both longitudinal edges and/or both transversal edges of both the front side and the back side of the tablet, wherein these bulges extend at least over one half, more preferably over two thirds of the length of the longitudinal and/or transversal edges, even more preferably over the whole length of the longitudinal and/or transversal edges. In another preferred embodiment, the bulges continuously circumscribe the basis area of the front side and/or the back side, preferably the front and the back side, at and/or adjacent to the respective longitudinal and transversal edges. Most desirable results in terms of an improved release profile can for example be obtained with tablets of the invention having bulges at both longitudinal edges of both sides of the tablet. The cross-section of these tablets can be described to have or come close to an H-shape. By use of the expression H-shape it shall just be indicated that a tablet body having opposite, in particular rather flat, basis areas is provided with opposing bulges at the longitudinal edges on both sides of the tablet body.
For example, in one H-shape embodiment the bulges can protrude above their respective basis areas only to a minor extent compared to the lateral distance between the bulges along opposite longitudinal edges, e.g. up to about 1 or 2 mm.

In one preferred embodiment, a tablet of the invention, in particular its oblong form, comprises at or adjacent to, in particular adjacent to, major portions of both opposite longitudinal edges, in particular at least along two thirds of the iongitudinai edges, ui the front side at least one bulge. In another preferred embodiment, a tablet of the invention, in particular its oblong form, comprises at least one bulge at or adjacent to, in particular adjacent to, major portions of both opposite longitudinal edges, in particular at least along two thirds of the longitudinal edges, of both the front side and the back side of the tablet. In another preferred embodiment, the tablet of the invention, in particular its oblong form, comprises a circumferential bulge at or adjacent to, in particular adjacent to, the circumferential edge of the front side of said tablet. In another preferred embodiment, the tablet of the invention, in particular its oblong form, comprises a circumferential bulge at or adjacent to, in particular adjacent to, the circumferential edge of both the front side and the back side of said tablet.

According to another suitable embodiment of the tablet of the invention, it is provided that one or both longitudinal edges are essentially straight over at least a major part of their length and/or wherein one or both transversal edges are curved over a major part of their length, in particular curved in the form of an essentially circular arc. It is of course also possible that the longitudinal edges exhibit any other irregular or regular shape, for example, having a wave-like edge portion at least over a section. It is also possible that the transversal edge exhibits the shape of a triangle or any other polygonal shape. In general, both longitudinal and transversal edges form the circumference of the front side and the back side of the tablet.

For most applications it is sufficient that the longitudinal length, that is, the length of the longitudinal axis, of the tablet does not exceed 30 mm.

According to another embodiment, the tablet of the invention preferably has an average thickness over the basis areas of the front and the back side of at least about 1 mm, and in particular of no more than about 6 mm, more in particular ranging from about 1 mm to about 3 mm or more in particular ranging from about 2 mm to about 4 mm.

According to one embodiment of the tablet of the invention, the bulge extends perpendicular from the basis area of the front side and/or from the basis area of the back side in average from about 0.5 mm to about 2 mm, in particular from about 0.5 mm to about 1 mm.

Tablets of the invention preferably have a length in the longitudinal direction in the range of about 5 mm to about 30 mm, in particular in the range of about 15 mm to about 25 mm, riiore iri pdriicuidr alUUL i7 iiirii LU a'UUL 23 riirii, eveii iiuUre iii pdriicuidr dbuUL 21 rririi; a width in the range of about 5 mm to about 15 mm, in particular in the range of about 7 mm to about 12 mm, more in particular about 7 mm to about 10 mm, even more in particular 7mm, 9 mm or 10 mm; and a thickness over the basis areas in the range of about 1 mm to about 6 mm, in particular in the range of about 1.5 mm to about 4 mm, even more in particular from 2 mm to about 4 mm, even further in particular from about 2.5 mm to about 3.5 mm.

As indicated above, the front side and/or the back side of the tablet of the invention, in particular the basis area of the front side and/or the basis area of the back side, can in one embodiment further comprise at least one indentation. As has been found, this generally allows for a further improvement of the control of the release profile. The indentation in general in one embodiment represents a hollow space which is provided or embedded in the overall surface of the tablet. For example, the front side, the back side, in particular the basis areas of the front side and/or the back side, the rim and/or at least one bulge can be provided with at least one indentation.

Indentations, when viewed from above, can have any irregular or regular shape, for example, the form of a square, rectangle, triangle, oval or circle. In one embodiment the indentations can take the form of a cylinder, a cube, a cuboid or a half-sphere, that is the walls and the opening forming the indentation come close to describing the form of a cylinder, a cube, a cuboid or a half-sphere. When viewed from above, the silhouette shape of the indentations has essentially the same width and length dimensions. It is also possible that when viewed from above, the silhouette shape of an indentation has a longer length dimension than a width dimension, for example, a length dimension which is at least 2, 3 or 4 times the width dimension. Accordingly, when viewed from above, the silhouette shape can be rather elongate, e.g. a rectangle, and can have a regular silhouette form, e.g.
straight, wave-like, or zig-zag, or can be rather irregular. In another embodiment an array of indentations can be formed, for example on the front side and/or the back side. For many applications it has been found to be sufficient that when viewed from above, the silhouette-shape of the indentation has a length dimension which is essentially identical to its width dimension as, for example, can be found with a circular, square-like or slightly oval or slightly rectangular shape. Said width dimension of the indentations, which is regularly determined parallel to the transversal axis, usually is less than one half of the transversal length of the tablet, in particular less than one third of the transversal length of the tablet. In one embodiment the width dimension is essentially identical to the depth of the indentation or is no more than 2 or 3 times the depth of the indentation. The length dimension of the indentation, which is regularly determined parallel to the iongitudinai axis, usuaiiy is no longer than three quarters of the longitudinal length of the tablet, in particular no longer than one half of the longitudinal length of the tablet, and preferably no longer than one third of the longitudinal length of the tablet. A hole in a tablet is not an indentation in the meaning of the present invention. The silhouette shape and the depth of said indentations can vary depending on the desired release profile. Usually care should be taken that the depths of these indentations does not come too close to the thickness of the tablet in order to prevent that upon handling a hole through the tablet will be formed. Preferably the indentations have a depth which does not go beyond half the thickness of the tablets of the invention.
For most applications it is frequently already sufficient that the maximum depth of said indentations does not go beyond one third of the thickness of the tablet of the invention.
The average thickness of the tablet of the invention in general is determined as the distance between the front and back side of the tablet or preferably between the basis area of the front side and the basis area of the back side.

By using the expressions front side and back side it shall be indicated that the tablet of the invention has two opposite sides which each can be provided with bulges and/or indentations. In consequence, the selection of which is the front side and which is the back side is rather arbitrary. Accordingly, the expressions front side and back side could also be replaced by first side and opposite second side, respectively.

In one embodiment of the invention, there is provided a tablet wherein the front side and/or the back side, in particular the, in particular essentially flat, basis area of the front side and/or the, in particular essentially flat, basis area of the back side, comprise in addition to at least one bulge at least one indentation, in particular between opposite longitudinal and/or transversal bulges.

In one embodiment of the invention it is provided that both the front and the back side comprise at least one indentation.

The indentations on the front side and on the back side of the tablet of the invention can at least once be at least partially off-set or can at least once be positioned in a congruent manner. In one preferred embodiment, all the indentations of the front side and all indentations on the back side are at least partially off-set or are positioned in a congruent manner.

The indentations are regularly positioned in the basis area of the front andior the back side of the tablet of the invention. It is for example possible to place two or more of such indentations adjacent to each other, e.g. in a row located between the longitudinal edges of the front and/or the back side. The indentations are preferably located between opposite longitudinally extending bulges at or adjacent to the longitudinal edges of the front and/or the back side of the tablet of the invention.

In one preferred embodiment, a tablet of the invention, in particular its oblong form, comprises at or adjacent to, in particular adjacent to, major portions of both longitudinal edges, in particular at least along two thirds of the longitudinal edges of the front side at least one bulge, and, in particular between the bulges along opposite longitudinal edges, at least one indentation.

In another preferred embodiment, a tablet of the invention, in particular its oblong form, comprises at least one bulge at or adjacent to, in particular adjacent to, major portions of both opposite longitudinal edges, in particular at least along two thirds of the longitudinal edges of both the front side and the back side of the tablet as well as at least one indentation on the front side and/or the back side, in particular on the basis area of the front side and/or the basis of the back side, of the tablet, in particular between the bulges which are located along opposite longitudinal edges on the front side and/or the back side, respectively. In another preferred embodiment, the tablet of the invention, in particular its oblong form, comprises a circumferential bulge at or adjacent to, in particular adjacent to, the circumferential edge of the front side and/or back side of said tablet, and at least one indentation on the front side and/or back side, in particular on the basis area circumscribed by the circumferential bulge on the front and/or on the back side. In another preferred embodiment, the tablet of the invention, in particular its oblong form, comprises a circumferential bulge at or adjacent to, in particular adjacent to, the circumferential edge of both the front side and the back side of said tablet and at least one indentation on the front side and the back side, in particular on the basis area circumscribed by the circumferential bulge of the front side and on the basis area circumscribed by the circumferential bulge of the back side.

There are generally no limitations as to the pharmaceutically active ingredient(s) (pharmacologically active compound(s)) which can be incorporated into the tablet of the invention.

Suitable active ingredients are those which exert a local physiological effect, as well as those which exert a systemic effect, after oral administration. Examples of suitable active ingredients encompass:
analgesic and anti-inflammatory drugs (NSAIDs, fentanyl, indomethacin, ibuprofen, ketoprofen, nabumetone, paracetamol, piroxicam, tramadol, COX-2 inhibitors such as celecoxib and rofecoxib);
anti-arrhythmic drugs (procainamide, quinidine, verapamil);
antibacterial and antiprotozoal agents (amoxicillin, ampicillin, benzathine penicillin, benzylpenicillin, cefaclor, cefadroxil, cefprozil, cefuroxime axetil, cephalexin, chloramphenicol, chloroquine, ciprofloxacin, clarithromycin, clavulanic acid, clindamycin, doxyxycline, erythromycin, flucloxacillin sodium, halofantrine, isoniazid, kanamycin sulphate, lincomycin, mefloquine, minocycline, nafcillin sodium, nalidixic acid, neomycin, norfloxacin, ofloxacin, oxacillin, phenoxymethyl-penicillin potassium, pyrimethamine-sulfadoxime, streptomycin, TMC207);
anti-coagulants (warfarin);
antidepressants (amitriptyline, amoxapine, butriptyline, clomipramine, desipramine, dothiepin, doxepin, fluoxetine, reboxetine, amineptine, selegiline, gepirone, imipramine, lithium carbonate, mianserin, milnacipran, nortriptyline, paroxetine, sertraline; 3-[2-[3,4-dihydrobenzofuro[3,2-c]pyridin-2(1 H)-yl]ethyl]-2-methyl-4H-pyrido[1,2-a]pyrimidin-4-one);
anti-diabetic drugs (glibenclamide, metformin);
anti-epileptic drugs (carbamazepine, clonazepam, ethosuximide, gabapentin, lamotrigine, levetiracetam, phenobarbitone, phenytoin, primidone, tiagabine, topiramate, valpromide, vigabatrin);
antifungal agents (amphotericin, clotrimazole, econazole, fluconazole, flucytosine, griseofulvin, itraconazole, ketoconazole, miconazole nitrate, nystatin, terbinafine, voriconazole);
antihistamines (astemizole, cinnarizine, cyproheptadine, decarboethoxyloratadine, fexofenadine, flunarizine, levocabastine, loratadine, norastemizole, oxatomide, promethazine, terfenadine);
anti-hypertensive drugs (captopril, enalapril, ketanserin, lisinopril, minoxidil, prazosin, ramipril, reserpine, terazosin);
anti-muscarinic agents (atropine sulphate, hyoscine);

antineoplastic agents and antimetabolites (platinum compounds, such as cisplatin, carboplatin; taxanes, such as paclitaxel, docetaxel; tecans, such as camptothecin, irinotecan, topotecan; vinca alkaloids, such as vinblastine, vindecine, vincristine, vinorelbine; nucleoside derivatives and folic acid antagonists such as 5-f'iuvrvurauii, capecitabine, gemcitabine, mercaptopurine, thioguanine, cladribine, methotrexate;
alkylating agents, such as the nitrogen mustards, e.g. cyclophosphamide, chlorambucil, chlormethine, iphosphamide, melphalan, or the nitrosoureas, e.g. carmustine, lomustine, or other alkylating agents, e.g. busulphan, dacarbazine, procarbazine, thiotepa;
antibiotics, such as daunorubicin, doxorubicin, idarubicin, epirubicin, bleomycin, dactinomycin, mitomycin; HER 2antibody, such as trastuzumab; podophyllotoxin derivatives, such as etoposide, teniposide; farnesyl transferase inhibitors; anthrachinon derivatives, such as mitoxantron);
anti-migraine drugs (alniditan, naratriptan, sumatriptan);
anti-Parkinsonian drugs (bromocryptine mesylate, levodopa, selegiline);
antipsychotic, hypnotic and sedating agents (alprazolam, buspirone, chlordiazepoxide, chlorpromazine, clozapine, diazepam, flupenthixol, fluphenazine, flurazepam, 9-hydroxyrisperidone, lorazepam, mazapertine, olanzapine, oxazepam, pimozide, pipamperone, piracetam, promazine, risperidone, selfotel, seroquel, sertindole, sulpiride, temazepam, thiothixene, triazolam, trifluperidol, ziprasidone, zolpidem);
anti-stroke agents (lubeluzole, lubeluzole oxide, riluzole, aptiganel, eliprodil, remacemide);
antitussive (dextromethorphan, laevodropropizine);
antivirals (acyclovir, ganciclovir, loviride, tivirapine, zidovudine, lamivudine, zidovudine +
lamivudine, didanosine, zalcitabine, stavudine, abacavir, lopinavir, amprenavir, nevirapine, efavirenz, delavirdine, indinavir, nelfinavir, ritonavir, saquinavir, adefovir, hydroxyurea, TMC120, TMC125, TMC278);
beta-adrenoceptor blocking agents (atenolol, carvedilol, metoprolol, nebivolol, propanolol);
cardiac inotropic agents (amrinone, digitoxin, digoxin, milrinone);
corticosteroids (beclomethasone dipropionate, betamethasone, budesonide, dexamethasone, hydrocortisone, methylprednisolone, prednisolone, prednisone, triamcinolone);
disinfectants (chlorhexidine);
diuretics (acetazolamide, frusemide, hydrochlorothiazide, isosorbide);
enzymes;
essential oils (anethole, anise oil, caraway, cardamom, cassia oil, cineole, cinnamon oil, clove oil, coriander oil, dementholised mint oil, dill oil, eucalyptus oil, eugenol, ginger, lemon oil, mustard oil, neroli oil, nutmeg oil, orange oil, peppermint, sage, spearmint, terpineol, thyme);

gastro-intestinal agents (cimetidine, cisapride, clebopride, diphenoxylate, domperidone, famotidine, lansoprazole, loperamide, loperamide oxide, mesalazine, metoclopramide, mosapride, nizatidine, norcisapride, olsalazine, omeprazole, pantoprazole, perprazole, prucalopride, rabeprazole, ranitidine, ridogrei, suiphasaiazine);
haemostatics (aminocaproic acid);
lipid regulating agents (atorvastatin, lovastatin, pravastatin, probucol, simvastatin);
local anaesthetics (benzocaine, lignocaine);
opioid analgesics (buprenorphine, codeine, dextromoramide, dihydrocodeine, hydrocodone, oxycodone, morphine);
parasympathomimetics and anti-dementia drugs (ATT-082, eptastigmine, galanthamine, metrifonate, milameline, neostigmine, physostigmine, tacrine, donepezil, rivastigmine, sabcomeline, talsaclidine, xanomeline, memantine, lazabemide);
peptides and proteins (antibodies, becaplermin, cyclosporine, erythropoietin, immunoglobulins, insuline);
sex hormones (oestrogens; conjugated oestrogens, ethinyloestradiol, mestranol, oestradiol, oestriol, oestrone; progestogens; chiormadinone acetate, cyproterone acetate, 17-deacetyl norgestimate, desogestrel, dienogest, dydrogesterone, ethynodiol diacetate, gestodene, 3-keto desogestrel, levonorgestrel, lynestrenol, medroxy-progesterone acetate, megestrol, norethindrone, norethindrone acetate, norethisterone, norethisterone acetate, norethynodrel, norgestimate, norgestrel, norgestrienone, progesterone, quingestanol acetate);
stimulating agents (sildenafil);
vasodilators (amlodipine, buflomedil, amyl nitrite, diltiazem, dipyridamole, glyceryl trinitrate, isosorbide dinitrate, lidoflazine, molsidomine, nicardipine, nifedipine, oxpentifylline, pentaerythritol tetranitrate); their N-oxides, their pharmaceutically acceptable acid or base addition salts and their stereochemically isomeric forms.

Pharmaceutically acceptable acid addition salts comprise the acid addition salt forms which can conveniently be obtained by treating the base form of the active ingredient with appropriate organic and inorganic acids. Active ingredients containing an acidic proton may be converted into their non-toxic metal or amine addition salt forms by treatment with appropriate organic and inorganic bases. The term addition salt also comprises the hydrates and solvent addition forms which the active ingredients are able to form. Examples of such forms are e.g. hydrates, alcoholates and the like. The N-oxide forms of the active ingredients comprise those active ingredients wherein one or several nitrogen atoms are oxidized to the so-called N-oxide.

The active ingredient(s) is (are) present in the dosage form in a therapeutically effective amount. The amount that constitutes a therapeutically effective amount varies according to the ingredients being used, the condition being treated, the severity of said condition, the patient being treated, and whether the dosage torm is designed for an immediate or retarded release. The amount of active ingredient(s) used in the present invention preferably ranges from about 0.01 % to about 90% (w/w), in particular from about 0.01 % to about 50% (w/w), more in particular from about 20% to about 50% (w/w).

Unless explicitly stated otherwise, in the meaning of the present invention the indication "w/w" shall mean weight of the compound specified per total weight of the composition forming the tablet.

In one embodiment, the one or more active ingredients are incorporated in a tablet for immediate release.

In another embodiment, the one or more active ingredients are incorporated in a tablet for retarded release. In this case, the active ingredient(s) of the tablet of the invention is (are) conventionally embedded in conventional formulating aids and/or one or more hydrophilic polymers. These hydrophilic polymers tend to swell upon contact with aqueous fluids following administration, and result in a viscous, drug release regulating gel layer.
Preferably the viscosity of these polymers ranges from 150 to 100.000 mPa.s (apparent viscosity of a 2 % aqueous solution at 20 C). Examples of suitable hydrophilic polymers include:
- alkylcelluloses, such as, methylcellulose;
- hydroxyalkylcelluloses, for example, hyd roxym ethylcel I u lose, hydroxyethylcellulose, hydroxypropylcellulose and hyd roxyb utylcel I u lose;
- hydroxyalkyl alkylcelluloses, such as, hydroxyethyl methylcellulose and hydroxypropyl methylcellulose;
- carboxyalkylcelluloses, such as, carboxymethylcellulose;
- alkali metal salts of carboxyalkylcelluloses, such as, sodium carboxymethylcellulose;
- carboxyalkylalkylcelluloses, such as, carboxymethylethylcellulose;
- carboxyalkylcellulose esters;
- other natural, semi-synthetic, or synthetic polysaccharides, such as, alginic acid, alkali metal and ammonium salts thereof, carrageenans, galactomannans, tragacanth, agar-agar, gummi arabicum, guar gummi, xanthan gummi, starches, pectins, such as sodium carboxymethylamylopectin, chitin derivates such as chitosan, polyfructans, inulin;
polyacrylic acids and the salts thereof;

- polymethacrylic acids and the salts thereof, methacrylate copolymers;
- polyvinylalcohol;
- polyvinylpyrrolidone, copolymers of polyvinylpyrrolidone with vinyl acetate;
- combinations of polyvinylalcohol and polyvinylpyrrolidone;
- polyalkylene oxides such as polyethylene oxide and polypropylene oxide and copolymers of ethylene oxide and propylene oxide.

Preferably the one or more hydrophilic polymers are cellulose derivatives, in particular cellulose ether derivatives, such as for example alkylcelluloses or hydroxyalkylcelluloses or hydroxyalkyl alkylcelluloses, more in particular hydroxyalkylcelluloses or hydroxyalkyl alkylcelluloses.

Most preferred cellulose ether derivatives are hydroxypropyl methylcellulose (HMPC) and hydroxypropyl cellulose (HPC). Different viscosity grades of hydroxypropyl cellulose and hydroxypropyl methylcellulose are commercially available. Hydroxypropyl methylcellulose preferably used in the present invention has a viscosity grade ranging from about 3,500 mPa.s to about 100,000 mPa.s, in particular ranging from about 4,000 mPa.s to about 20,000 mPa.s and most in particular a viscosity grade of about 6,500 mPa.s to about 15,000 mPa.s (apparent viscosity of a 2% aqueous solution at 20 C), e.g.
hypromellose 2208 (DOW, Antwerp, Belgium). Hydroxypropyl cellulose having a viscosity lower than 1,500 mPa.s (apparent viscosity of a 2% aqueous solution at 20 C) is preferred, in particular hydroxypropyl cellulose having a viscosity in the range from about 150 to about 700 mPa.s, preferably from 200 to 600 mPa.s, e.g. Klucel EF (Hercules, Wilmington, USA).
Preferably, the amount of viscous hydrophilic polymers, in particular HPMC and HPC, in the present formulation ranges from about 0.01 to about 80% (w/w), in particular from about 10 to about 60 % (w/w), more in particular between 30 and 60% (w/w).

In addition to the one or more hydrophilic polymers, the retarded release formulation can in one embodiment comprise pregelatinized starch. The amount of pregelatinized starch preferably is in the range from 5 to 80 % (w/w), in particular from 5 to 15%
(w/w).

In one embodiment, the tablet of the invention comprises at least one pharmaceutically active ingredient (pharmacologically active compound), pregelatinized starch, and HPC
and/or HPMC, in particular at least one pharmaceutically active ingredient (pharmacologically active compound), pregelatinized starch, HPC and HPMC.

The tablets according to the invention are preferably prepared by way of compression using a die and a punch.

The tablets of the invention can optionally be provided, partiaiiy or compieieiy, with a conventional tablet coating. The tablets of the present invention are preferably film coated with art-known film coating compositions. The coating is applied to improve the aesthetic impression and/or the taste of the tablets and the ease with which they can be swallowed.
Coating the tablets of the present invention can also serve other purposes, e.g. improving stability and shelf-life. Suitable coating formulations comprise a film forming polymer such as, for example, hydroxypropyl methylcelIulose, e.g. hypromellose 2910 (5 mPa.s), a plasticizer such as, for example, a glycol, e.g. propylene glycol or polyethylene glycol, an opacifier, such as, for example, titanium dioxide, and a film smoothener, such as, for example, talc. Suitable coating solvents are water as well as organic solvents. Examples of organic solvents are alcohols, e.g. ethanol or isopropanol, ketones, e.g.
acetone, or halogenated hydrocarbons, e.g. methylene chloride. Optionally, the coating can contain a therapeutically effective amount of one or more active ingredients to provide for an immediate release of said active ingredient(s) and thus for an immediate relief of the symptoms treated by said active ingredient(s). Coated tablets of the present invention are prepared by first making the tablet cores in the way as described above and subsequently coating said tablet cores using conventional techniques, such as coating in a coating pan.
For example, PEG, e.g. PEG 20.000, or HPMC can be used for the coating.

Beside active ingredient(s) and optional hydrophilic polymers, the tablet of the present invention can also optionally comprise pharmaceutically acceptable formulation agents such as fillers, glidants, binding agents, granulating agents, anti-caking agents, lubricants, flavors, dyes, and preservatives.

The filler may be selected from soluble fillers, for example, sucrose, lactose, trehalose, maltose, mannitol, sorbitol, inulin, and from insoluble fillers, for example, dicalcium or tricalcium phosphate, talc. An interesting filler is lactose, in particular, lactose monohydrate.
Different grades of lactose can be used. One type of lactose preferably used in the present invention is lactose monohydrate 200 mesh (DMV, Veghel, the Netherlands).
Another lactose monohydrate, lactose monohydrate of the type DCL 11 (DMV, Veghel, the Netherlands), can also preferably be used. The notation DCL refers to "Direct Compression Lactose". The number 11 is a reference number of the manufacturer. This type of lactose is characterised in that 98 % (w/w) of the particles, based on the total amount of lactose employed, have a diameter smaller than 250 pm, 30 % (w/w) to 60 % (w/w) of the particles, based on the total amount of lactose employed, have a diameter of 100 pm, and at maximum 15 % (w/w) of the particles, based on the total amount of lactose employed, have a diameter of smaller than 45 pm. The weight percentage of filler ranges between about 6 % and about 54 % (w/w).

Among the optional formulating agents that further can be comprised in the present formulation there can be mentioned agents such as polyvidone; starch; acacia gum; gelatin;
seaweed derivatives, e.g. alginic acid, sodium and calcium alginate; cellulose derivatives, e.g. ethylcellulose, hyoroxypropyl methylcellulose, having useful binding and granulating properties; glidants such as colloidal silica, starch or talc; lubricants such as magnesium stearate and/or palmitate, calcium stearate, stearic acid, polyethylene glycol, liquid paraffin, sodium or magnesium lauryl sulphate; anti-adherents such as talc and corn starch.

The first aspect of the present invention can be more readily understood by reference to Figures 1-6.

In Figure 1A) a tablet 1 is depicted showing its front side 2 from the top.
The tablet 1 has a longitudinal axis 4 in the longitudinal direction and a transversal axis 6, perpendicular thereto, in the transversal direction. The two opposing longitudinal edges 8 and 10 and the two opposing transversal edges 12 and 14 are forming the circumference of the front side 2 of the tablet 1. In the depicted embodiment the longitudinal edges 8, 10 are essentially straight whereas the transversal edges 12, 14 are rounded. Tablet 1 of Figure 1A) has a circumferential bulge 16, 20 on both the upper side 2 and the lower side 18 (Figure 113)) having reference sign 20. The bulges 16 and 20 are positioned adjacent to the longitudinal and transversal edges and raise above the basis area 22 of the front side 2, and above the basis area 24 of the back side 18, respectively. In the embodiment shown in Figure 1A), 1B) the basis areas 22, 24 have a transversal width "a" which is smaller than the transversal width "b" of the bulges.

In Figure 2A) tablet 1 is depicted which is rather similar to the tablet shown in Figure 1A) with the difference that the lateral width a of the basis area 22 of the front side 2 is larger than the transversal width b of the respective bulges 16, 20 of the front side 2 and the back side 18, respectively. The embodiment of the tablet 1 of Figure 2A) also differs from that of Figure 1A) insofar as the basis areas 22, 24 are no longer completely flat as depicted in Figure 1B) but can be curved inwardly, in particular in a symmetrical manner towards the center, as depicted in Figure 2B).

Both embodiments as shown in Figures 1 and 2 have in common that the circumferential rim 26 which connects the front side 2 and the back side 18 has a rather flat profile as can be derived from Figures 1 B) and 2B).

In Figure 3A) another embodiment of tablet 1 of the invention is shown which differs from the tablet as depicted in Figure 2A) insofar as the basis areas 22 and 24 of the front side 2 and the back side 18, respectively, each are provided with four indentations 28. These indentations 28 have an essentially circular shape and are positioned in a row within the basis areas 22 and 24 (s.a. Figure 3B) and Figure 3C)). As can be derived from Figures 3B) and 3C) the indentations 28 of the front side 2 are placed in a congruent manner with respect to the indentations 30 in the basis area 24 of the back side 18.
Again, the rim 26 has a rather flat profile. The indentations 28, 30 have a trough-like, hollow form in the depicted embodiment, that is, from the circumferential rim of the indentations the depth is slightly increasing up to the center of the indentations, which can, for example be derived from Figure 3B).

The embodiment of tablet 1 according to the invention as depicted in Figure 4A) differs from the embodiment of Figure 3A) in that the bulges 16 and 20 of the front and back sides 2, 18 are no longer present along the entire circumference of the tablet. In the embodiment of Figure 4 sections of the opposite transversal edges 12 and 14 of the front side 2 and back side 18 are no longer provided with a bulge section. Accordingly, the bulge 16 of the front side 2 extends only along the longitudinal edges 8 and 10 of the front side as well as over portions of opposing transversal edges 12 and 14 of the front side leaving on both opposing transversal edges of the front and back side significant portions without any bulge. The same applies to the back side in the present case. The individual bulge portions of the front side of the embodiment of Figure 4 have been assigned reference numbers 17a and 17b for the front side 2 and 21 a and 21 b for the back side 18. The indentations 28 and 30 of the front and back side are lying within the basis areas 22 and 24, respectively, and are placed in a congruent manner as with the embodiment of Figure 3 (s.a. Figure 4C)).
Again, the circumferential rim 26 has an essentially flat configuration.

The embodiment of a tablet 1 of the present invention as depicted in Figure 5A) differs from the tablet as shown in Figure 4 only insofar as the indentations 28 of the front side 2 and the indentations 30 of the back side 18 are no longer located in a congruent manner, but are at least partially off-set as can be best derived from Figure 5C).

Figure 6A) shows another embodiment of a tablet 1 of the invention which is rather similar to the embodiment depicted in Figure 1, for example, as to the circumferential bulge 16 on the front side 2 as well as to the circumferential bulge 20 on the back side 18 (not shown in Figure 6A, but in Figures 6B) and 6C)) and as to its oblong shape and dimensions. Different from the tablet of Figure 1 the embodiment shown in Figure 6 makes use of a circumferential land 32 on the front side 2 of a tablet as well as of such a corresponding circumferential land 34 on back side 18 (not shown in Figure 6A), but in Figures 6B) and 6C)). That is, the circumferential bulges 16 and 20 are not extending up to circumferential rim 26, but are located adjacent to, i.e. spaced apart from said rim 26. As can be derived from Figures 6B) and 6C) the bulge 16 does not smoothly go over into the rim section 26, but terminates adjacent to the rim section thereby furnishing a small portion which is located at a height somewhat similar to that of the basis area 22 of the front side 2. The same applies to the circumferential land 34 and the bulge 20 on the back side 18. In the embodiment shown in Figures 6A) to 6C) the land 32 of the front side 2 is not exactly lying in the plane of the basis area 22, but slightly above. This can be best derived from Figure 6C). The same applies to the land 34 of the back side 18. For certain tablet formulations it has been found advantageous to make use of, in particular circumferential, land sections, for example, in order to alleviate the die punching cycle in mass production.

A second aspect of the invention relates to a tamper-resistant pharmaceutical dosage form having a retarded release profile, especially a tamper-resistant oral dosage form having a retarded release profile, particularly a tamper-resistant tablet having a retarded release profile, comprising at least one pharmaceutically active ingredient (pharmacologically active compound) with potential for abuse, and having a shape comprising a longitudinal axis and two opposite longitudinal edges, a transversal axis perpendicular to the longitudinal axis and two opposite transversal edges, a front side, an opposite back side and a circumferential rim between said front and back side, wherein the front side and/or the back side comprise a basis area and wherein the front side and/or the back side comprise at least one bulge which extends above said basis area, said at least one bulge being present at and/or adjacent to at least a section of one or both longitudinal edges and/or at and/or adjacent to at least a section of one or both transversal edges and/or between both longitudinal edges and both transversal edges. The front side and/or the back side of the dosage form, in particular the basis area of the front side and/or the basis area of the back side, can further comprise at least one indentation.

In general, the foregoing teachings relating to the first aspect of the present invention are also applicable to this second aspect of the present invention. However, there are emphasized the following:

A preferred embodiment of the tablet of the present invention is a tablet with a length (longitudinal axis) of about 21 mm; a width (transversal axis) of about 9 mm;
a thickness from the top of the bulge on the front side to the top of the opposite bulge on the back side of about 5 mm; a thickness over the basis area of about 3 mm and an extension of the top of the bulge from the basis area of about 1 mm, and in particular having a circumferential bulge on the front and/or on the back side of the tablet, and preferably without any indentations, as for example derivable from Fig. 1A.

Active ingredients with potential for abuse are known to the person skilled in the art and comprise tranquillisers, stimulants, barbiturates, narcotics, opioids or opioid derivatives.

A pharmaceutically active ingredient (pharmacologically active compound) which is preferably used with the tamper-resistant tablets of the invention is an analgesic compound or a pain-killing compound, such as for example an opioid or an opioid derivative, in particular tapentadol or salts thereof, more in particular tapentadol.

By tamper-resistant it is meant that the active ingredient can not readily be separated from the tablet in a form suitable for abuse because the tablet is such that it can not easily be grinded. This renders extraction of the active ingredient, which could be used for drug-abuse, from the tablet difficult or this renders it very difficult to transform the tablet into a powder for sniffing.

The tablet of the invention can in one embodiment be rendered tamper-resistant by incorporating at least one synthetic or natural polymer which increases the breaking strength of the tablet so that it is much higher than a conventional tablet.
In this way, pulverization of the tablet by conventional means, such as for example by a pestle and mortar, is rendered almost impossible, and, hence, the conversion of the active ingredient incorporated in the tablet in a form which is suitable for abuse is complicated.

In one embodiment of the present invention, said at least one synthetic or natural polymer is a polyalkylene oxide such as polyethylene oxide and polypropylene oxide and copolymers of ethylene oxide and propylene oxide.

Preferably, the polyalkylene oxide is polyethylene oxide, in particular polyethylene oxide having a molecular weight above 500.000, preferably above 1.000.000, and more preferably in the range of about 2.000.000 to about 7.000.000. The amount of polyethylene oxide can in one embodiment range from about 20 to about oU% (wiw), in particular from about 20 to 50 % (w/w), more in particular from about 30 to about 50% (w/w).

In one embodiment, the tablet of the present invention has a breaking strength of at least 300 N, in particular at least 350 N, more in particular at least 400 N, even more in particular at least 450 N. The breaking strength of the tablet of the invention can be determined by the method for determining breaking strength of tablets described in the European Pharmacopoeia 1997, page 143, 144, method No. 2.9.8. Preferred alternatives for measuring the breaking strength are described in connection with further aspects of the invention below.

Retarded release of an active ingredient from an oral dosage form is known to a person skilled in the art. For a retarded release tablet, it usually is sufficient to administer the tablet once or twice daily. The retarded release profile of the tablet of the present invention can be achieved by embedding the active ingredient in an amount of hydrophilic polymer or of a blend of hydrophilic polymers, optionally also containing conventional formulating agents.
These hydrophilic polymers tend to swell upon contact with aqueous fluids following administration, and result in a viscous, drug release regulating gel layer.
Preferably the viscosity of these polymers ranges from 150 to 100.000 mPa.s (apparent viscosity of a 2 %
aqueous solution at 20 C). Examples of suitable hydrophilic polymers include:
- alkylcelluloses, such as, methylcellulose;
- hydroxyalkylcelluloses, for example, hyd roxymethylcel I u lose, hyd roxyethylce I lu lose, hydroxypropylcellulose and hyd roxybutyl cel I u lose;
- hydroxyalkyl alkylcelluloses, such as, hydroxyethyl methylcellulose and hydroxypropyl methylcellulose;
- carboxyalkylcelluloses, such as, carboxymethylcellulose;
- alkali metal salts of carboxyalkylcelluloses, such as, sodium carboxymethylcellulose;
- carboxyalkylalkylcelluloses, such as, carboxymethylethylcellulose;
- carboxyalkylcellulose esters;
- other natural, semi-synthetic, or synthetic polysaccharides, such as, alginic acid, alkali metal and ammonium salts thereof, carrageenans, galactomannans, tragacanth, agar-agar, gummi arabicum, guar gummi, xanthan gummi, starches, pectins, such as sodium carboxymethylamylopectin, chitin derivates such as chitosan, polyfructans, inulin;
polyacrylic acids and the salts thereof;

- polymethacrylic acids and the salts thereof, methacrylate copolymers;
- polyvinylalcohol;
- polyvinylpyrrolidone, copolymers of polyvinylpyrrolidone with vinyl acetate;
- combinations of polyvinylalcohol and polyvinyipyrroiidone.

In one embodiment of the invention, the hydrophilic polymer in which the active ingredient is embedded is selected from a cellulose derivative. Preferably, the cellulose derivative is a cellulose ether derivative, more preferably the cellulose ether derivative is HPMC. The amount of cellulose derivative can in one embodiment range from about 1 to about 20%
(w/w), in particular from about 10 to about 20% (w/w).

In addition to said synthetic or natural polymer which increases the breaking strength of the tablet, in particular polyalkylene oxide, and to said hydrophilic polymers, the tablet formulation in one embodiment can further comprise a polyalkylene glycol, such as for example PEG 6000. The amount of polyalkylene glycol can for example range from about 1 to about 20% (w/w), in particular from about 1 to about 10% (w/w).

In one embodiment, the tamper-resistant tablet of the invention comprises at least one pharmaceutically active ingredient (pharmacologically active compound) with potential for abuse, in particular a pain-killing drug, more in particular an opioid or an opioid derivative, e.g. tapentadol; at least one polyalkylene oxide, in particular polyethylene oxide, more in particular polyethylene oxide having a molecular weight in the range of about 2.000.000 to 7.000.000; at least one cellulose ether derivative, in particular hydroxypropyl methylcellulose (HMPC); and at least one polyalkylene glycol, in particular polyethylene glycol, such as PEG 6000. In preferred compositions further components can be present such as anti-oxidants, for example vitamin E. In another embodiment, the tablet comprises pharmaceutically active ingredient (pharmacologically active compound) with potential for abuse in an amount of at least 5 weight percent; polyethylene oxide in an amount of at least 15 weight percent; cellulose ether derivatives, in particular HPMC, in an amount of at least weight percent; and polyalkylene glycol, in particular polyethylene glycol, in an amount of at least 5 weight percent.

The tamper-resistant tablets of the present invention are preferably prepared by melt-extruding the tablet formulation. The thus obtained melt-extruded strands are preferably cut into monoliths, which are then compressed into tablets.

It has, for example, been found that in particular when the tablet of the invention is produced from melt extruded monolithic masses, very hard compacted tablets are obtained which can no longer be easily grinded or crushed thereby allowing for a very high degree of tamper-resistance. With these very hard tablets an improved coniroi of the drug release profile can be obtained by the tablet of the present invention.

The tablets according to the invention are preferably prepared by way of compression using a die and a punch, preferably from a monolithic mass obtained by melt extrusion. If obtained via melt extrusion, the compressing step is preferably carried out with a monolithic mass exhibiting ambient temperature, that is, a temperature in the range from 20 to 25 C.
The strands obtained by way of extrusion can either be subjected to the compression step as such or can be cut prior to the compression step. It is of course also possible to subject the extruded strands to the compression step or to the cutting step when still warm, that is more or less immediately after the extrusion step. The extrusion is preferably carried out by way of twin-screw extruder.

The aforementioned polymers used for the preparation of the tablet of the invention, that is, the chains of these polymers are preferably predominantly oriented along the direction of the extrusion. In case the monolith resulting from extrusion has a length greater than its width, the compression of said monolith is preferably performed with a compression direction perpendicular to the length.

A third aspect of the invention relates to a pharmaceutical dosage form with controlled release of a pharmacologically active compound (A) contained therein, the pharmaceutical dosage form having a breaking strength B, of at least 500 N in a direction of extension E,, and having a breaking strength B2 of less than 500 N in a direction of extension E2.

In general, the foregoing teachings relating to the first aspect of the invention and the second aspect of the invention are also applicable to this third aspect of the invention.
However, there are emphasized the following:

Direction of extension E, and direction of extension E2, respectively, can principally be any directions of extension of the pharmaceutical dosage form, i.e., any arbitrarily chosen directions of extension, provided that the corresponding requirement for the breaking strength B, and B2, respectively, is satisfied.

The "breaking strength" (resistance to crushing) of a pharmaceutical dosage form is known to the skilled person. In this regard it can be referred to, e.g., W.A.
Ritschel, Die Tablette, 2.
Auflage, Editio Cantor Verlag Aulendorf, 2002; H Liebermann et al., Pharmaceutical dosage forms: Tablets, Vol. 2, Informa Healthcare; 2 edition, 1990; and Encyclopedia Of Pharma-ceutical Technology, Informa Healthcare; 1 edition.

For the purpose of the specification, the breaking strength is preferably defined as the amount of force that is necessary in order to fracture the pharmaceutical dosage form (=
breaking force). Therefore, for the purpose of the specification the dosage form does preferably not exhibit the desired breaking strength when it breaks, i.e., is fractured into at least two independent parts that are separated from one another. In another preferred embodiment, however, the pharmaceutical dosage form is regarded as being broken if the force decreases by 25% (threshold value) of the highest force measured during the measurement (see below).

The dosage forms according to the invention are preferably distinguished from conventional dosage forms in that, due to their breaking strength, they cannot be pulverized by the application of force with conventional means, such as for example a pestle and mortar, a hammer, a mallet or other usual means for pulverization, in particular devices developed for this purpose (tablet crushers). In this regard "pulverization" means crumbling into small particles that would immediately release the pharmacologically active compound (A) in a suitable medium. Avoidance of pulverization virtually rules out oral or parenteral, in particular intravenous or nasal abuse.

Conventional tablets typically have a breaking strength well below 200 N in any direction of extension. The breaking strength of conventional round tablets may be estimated according to the following empirical formula: Breaking Strength [in N] = 10 x Diameter Of The Tablet [in mm]. Thus, according to said empirical formula, a round tablet having a breaking strength of at least 500 N would require a diameter of at least 50 mm (about 2 inches).
Such a tablet, however, could not be swallowed. The above empirical formula does not apply to the pharmaceutical dosage forms of the invention, which are not conventional but rather special.

Further, the actual mean chewing force is about 220 N (cf., e.g., P.A.
Proeschel et al., J
Dent Res, 2002, 81(7), 464-468, copy attached). This means that conventional tablets having a breaking strength well below 200 N may be crushed upon chewing, whereas the dosage forms according to the invention may not, at least not in direction of extension E,.

Still further, when applying a gravitational acceleration of about 9.81 m/s2, correspond to a gravitational force of more than 50 kg, i.e. the pharmaceutical dosage forms according to the invention can withstand a weight of more than 5u kg, at ieasi iii direction of extension E,.

Methods for measuring the breaking strength of a pharmaceutical dosage form are known to the skilled artisan. Suitable devices are commercially available.

For example, the breaking strength (resistance to crushing) can be measured in accor-dance with the European Pharmacopoeia 5.0, 2.9.8 or 6.0, 2.09.08 "Resistance to Crushing of Tablets". The test is intended to determine, under defined conditions, the resistance to crushing of tablets, measured by the force needed to disrupt them by crushing.
The apparatus consists of 2 jaws facing each other, one of which moves towards the other. The flat surfaces of the jaws are perpendicular to the direction of movement. The crushing surfaces of the jaws are flat and larger than the zone of contact with the tablet. The apparatus is calibrated using a system with a precision of 1 Newton. The tablet is placed between the jaws, taking into account, where applicable, the shape, the break-mark and the inscription; for each measurement the tablet is oriented in the same way with respect to the direction of application of the force (and the direction of extension in which the breaking strength is to be measured). The measurement is carried out on 10 tablets, taking care that all fragments of tablets have been removed before each determination. The result is expressed as the mean, minimum and maximum values of the forces measured, all expressed in Newton.

A similar description of the breaking strength (breaking force) can be found in the US
Pharmacopoeia. The breaking strength can alternatively be measured in accordance with the method described therein where it is stated that the breaking strength is the force required to cause a tablet to fail (i.e., break) in a specific plane. The tablets are generally placed between two platens, one of which moves to apply sufficient force to the tablet to cause fracture. For conventional, round (circular cross-section) tablets, loading occurs across their diameter (sometimes referred to as diametral loading), and fracture occurs in the plane. The breaking force of tablets is commonly called hardness in the pharmaceutical literature; however, the use of this term is misleading. In material science, the term hardness refers to the resistance of a surface to penetration or indentation by a small probe. The term crushing strength is also frequently used to describe the resistance of tablets to the application of a compressive load. Although this term describes the true nature of the test more accurately than does hardness, it implies that tablets are actually crushed during the test, which is often not the case.

Alternatively, the breaking strength (resistance to crushing) can be measured in accordance with WO 2006/082099, which can be regarded as a modification of the method described in the European Pharmacopoeia. The apparatus used for the measurement is preferably a "Zwick Z 2.5" materials tester, Finax = 2.5 kN with a maximum draw of 1150 mm, which should be set up with one column and one spindle, a clearance behind of 100 mm and a test speed adjustable between 0.1 and 800 mm/min together with testControl software.
Measurement is performed using a pressure piston with screw-in inserts and a cylinder (diameter 10 mm), a force transducer, Finax= 1 kN, diameter = 8 mm, class 0.5 from 10 N, class 1 from 2 N to ISO 7500-1, with manufacturer's test certificate M
according to DIN
55350-18 (Zwick gross force Fmax = 1.45 kN) (all apparatus from Zwick GmbH &
Co. KG, Ulm, Germany) with Order No BTC-FR 2.5 TH. D09 for the tester, Order No BTC-LC
0050N. P01 for the force transducer, Order No BO 70000 S06 for the centring device.

In a preferred embodiment of the invention, the breaking strength is measured by means of a breaking strength tester Sotax , type HT100 (Alischwil, Switzerland).
Preferably, the Auto AlignmentTM device of the Sotax HT100 is not used, just to allow placing the dosage form individually between the jaws in order to measure the breaking strength in specific direc-tions of extension. The Sotax HT100 can measure the breaking strength according to two different measurement principles: constant speed (where the test jaw is moved at a constant speed adjustable from 5-200 mm/min) or constant force (where the test jaw increases force linearly adjustable from 5-100 N/sec). In principle, both measurement principles are suitable for measuring the breaking strength of the dosage form according to the invention. Preferably, the breaking strength is measured at constant speed, preferably at a constant speed of 120 mm/min.

In a preferred embodiment, the pharmaceutical dosage form is regarded as being broken if it is fractured into at least two separate pieces.

In another preferred embodiment, the pharmaceutical dosage form is regarded as being broken if the force decreases by 25% (threshold value) of the highest force measured during the measurement. For example, if the highest force measured during the measurement is 144 N, the tablet is regarded as being broken when the force decreases below 108 N (= 75% of 144 N; decrease by 25%). The value of the breaking strength in the respective direction of extension is then 144 N. In a preferred embodiment, said threshold value is 30%, more preferably 35%, still more preferably 40%, most preferably 45% and in particular 50%. Under these circumstances, a dosage form may have to be regarded as being broken although it has not been fractured into at least two separate pieces. For example, a dosage form that has been torn in the middle but that has riot been f age er"t-ed, may have to be regarded as being broken in accordance with this definition of the breaking strength. Thus, in accordance with this definition, failure of the breaking strength test at a particular force may be due to fracture of the dosage form or any other deformation that causes the force to drop below the above threshold value, e.g. rupture, cracking, dunting, cleaving, fissure, and the like.

Figure 18 schematically illustrates the measurement of the breaking strength of the dosage form depicted in Figure 10, in particular the adjustment device for the dosage form used for this purpose before and during the measurement, in three different directions of extension a) to c). This measurement set-up is applicable to the various methods for measuring the breaking strength of the dosage form, including the above method according to the European Pharmacopeia, the variation thereof (according to Zwick) and the preferred method using the Sotax HT100. To this end, the dosage form is held between the plain jaw (61a) and the plain jaw (61b) of the force application apparatus (not shown) with the assistance of two 2-part clamping devices, which are in each case firmly attached (not shown) to the jaws once the spacing necessary for accommodating and centering the tablet to be measured has been established. The spacing may be established by moving the 2-part clamping devices horizontally outwards or inwards in each case on the jaw on which they are mounted. The measurement of the breaking strength in different directions of extensions is illustrated in Figure 18A) to Figure 18C). Figure 18A) illustrates the arrangement for measuring the breaking strength in direction of extension E, that is perpendicular to the main direction of extension E2 of the dosage form. Figure 18B) illustrates the arrangement for measuring the breaking strength in the main direction of extension E2 of the dosage form. This arrangement is the standard arrangement when measuring the breaking strength of conventional oblong tablets. The Auto AlignmentTM
device of the Sotax HT100 serves the purpose of aligning the tablet shapes automatically in order to ensure reproducible results in this direction of extension.
According to the present invention, however, the breaking strength of the dosage form needs to be measured in different directions of extension and the main direction of extension may be among said different directions of extension, but does not have to necessarily. Figure 18C) illustrates the arrangement for measuring the breaking strength in direction of extension E3 that is perpendicular to the main direction of extension E2 of the dosage form as well as perpendicular to direction of extension E,.

In a preferred embodiment, the breaking strength tester, preferably the Sotax HT100, is equipped with two plain jaws (cf. Figure 18).

In another preferred embodiment, the breaking strength tester, preferably the Sotax HT1 00, is equipped with two jaws that are not plain (cf. Figures 19, 20).
Preferably, one jaw contains an embossment and the other jaw contains an indentation. Embossment and indentation can be congruent with one another (like positive and negative), but do not have to.

Preferably, the embossment serves as an arbor, spike or mandrel and can be round of angular (e.g., triangular, rectangular, etc.). In a preferred embodiment, the embossment assumes the shape of a hemisphere. In another preferred embodiment, the embossment assumes the shape of a cone. In yet another preferred embodiment, the embossment assumes the shape of a trigonal or rectangular pyramid. Most preferably, the embossment assumes the shape of a half cylinder, preferably having a radius of curvature of 2.5 mm. In a preferred embodiment, the centre of said half cylinder lies within the surface of the main area of extension of the jaw. In another preferred embodiment, the centre lies about 0.5 mm away from said surface, just in the inside of the jaw.

Preferably, the indentation serves as a recess for the embossment. In a preferred embodiment, the indentation assumes the shape of a hollow hemisphere. In another preferred embodiment, the indentation assumes the shape of a hollow cone. In yet another preferred embodiment, the indentation assumes the shape of a hollow trigonal or rectangular pyramid. Most preferably, the indentation assumes the shape of a chamfering, kerf or notch with an angle of preferably 90 .

Figure 19A) shows a schematic view of jaw (61a) containing embossment (62) and jaw (61 b) containing indentation (63/64). The embossment (62) assumes the shape of a half cylinder that is characterized by radius r. Preferably, r = 2.5 mm. The center of the circular cross-section of the half cylinder may lie within the surface main area of extension of the jaw so that the entire half cylinder forms the embossment (62). Alternatively, the center of the circular cross-section of the half cylinder may lie within the body of the jaw, e.g. at a distance r from the surface of the main area of extension, so that the only a part of the half cylinder forms the embossment (62). Preferably, r' = 0 or 0.5 mm. The indentation is located in a rectangular recess (64) having a side length of 2 x, + 2 x2 and a height of y2. In the centre of said rectangular recess (64) is located a chamfering, kerf or notch (63) having a side length of 2 x, and a height/depth of y,. In a preferred embodiment, x, =
3 mm, x2 = 6 mm, y, = 3 mm and y2 = 2 mm. In another preferred embodiment, x, = 4 mm, x2 =
7 mm, y, = 4 mm and y2 = 2 mm. Preferably, the chamfering, kerf or notch (63) assumes an angle of yu~. Figure 19B) shows a schematic view of jaws (61a) and (bib) on faces (65a) and (65b), respectively.

Figure 20 shows in analogy to Figure 18 how the dosage form depicted in Figure 10 should be placed between the two jaws having embossment and indentation, respectively, in order to measure the breaking strength in directions of extension E,, E2 and E3. The skilled person recognizes that during the measurement, i.e. the movement of the jaws towards one another, it may become necessary to additionally clamp the dosage form, e.g.
by means of suitable guiding shafts or leading tracks (not shown), in order to avoid that the dosage form is tilted or evades to the side.

In general, a measuring set-up with jaws equipped with embossment and indentation (cf.
Figure 20) realizes harsher measuring conditions than a measuring set-up with plain jaws (cf. Figure 18). Thus, it may happen that a given dosage form which passes the measurement in accordance with Figure 18 fails when being measured in the same direction of extension in accordance with Figure 20.

As far as tablet orientation during the measurement of the breaking force is concerned, the US Pharmacopoeia (USP) states that in general, tablets are tested either across the diameter or parallel to the longest axis. Scored tablets have two orientation possibilities.
When they are oriented with their scores perpendicular to the platen faces, the likelihood that tensile failure will occur along the scored line increases. This provides information about the strength of the matrix at the weakest point in the structure. When scored tablets are oriented with their scores parallel to the platen faces, more general information about the strength of the matrix is derived. Capsule-shaped tablets or scored tablets may best be broken in a three-point flexure test. A fitting, which is either installed on the platens or substituted for the platens, supports the tablet at its ends and permits the breaking load to be applied to the opposite face at the unsupported midpoint of the tablet. The fittings are often available from the same source that supplies the hardness tester. Figure 20 is in accordance with this description in the USP.

The pharmaceutical dosage form according to the invention preferably has a breaking strength B, of at least 500 N in direction of extension E,. Preferably, direction of extension E, is perpendicular to the main direction of extension of the dosage form.
Preferably, the breaking strength in direction of extension E, is at least 500 N regardless of whether the measuring device is equipped with plain jaws or with two jaws one of which containing an embossment and the other containing an indentation, as described above.

Further, the pharmaceutical dosage form according to the invention preferably has a breaking strength B2 of less than 500 N in direction of extension E2.
Preferably, direction of extension E2 is the main direction of extension of the dosage form.
Preferably, the breaking strength in direction of extension E2 is less than 500 N when being measured with two plain jaws. However, it is not required (but possible) that the breaking strength in direction of extension E2 is less than 500 N when being measured with two jaws one of which containing an embossment and the other containing an indentation, as described above.

In a preferred embodiment according to the invention, the breaking strength B, of the pharmaceutical dosage form in direction of extension E, is measured in accordance with Figure 20A), i.e. orthogonal to the main direction of extension and by means of embossment and indentation. The breaking strength B2 in direction of extension E2, however, is preferably measured in accordance with Figure 18B), i.e. along the main direction of extension and by means of plain jaws.

The pharmaceutical dosage form according to the invention preferably exhibits mechanical strength over a wide temperature range, in addition to the breaking strength (resistance to crushing) optionally also sufficient hardness, impact resistance, impact elasticity, tensile strength and/or modulus of elasticity, optionally also at low temperatures (e.g. below -24 C, below -40 C or in liquid nitrogen), for it to be virtually impossible to pulverize by spontaneous chewing, grinding in a mortar, pounding, etc. Thus, preferably, in direction of extension E, the comparatively high breaking strength of the pharmaceutical dosage form according to the invention is maintained even at low or very low temperatures, e.g., when the pharmaceutical dosage form is initially chilled to increase its brittleness, for example to temperatures below -25 C, below -40 C or even in liquid nitrogen.

The pharmaceutical dosage form according to the invention is preferably characterized by a certain degree of breaking strength. This does not mean that the pharmaceutical dosage form must also exhibit a certain degree of hardness. Hardness and breaking strength are different physical properties. Therefore, the tamper resistance of the pharmaceutical dosage form does not necessarily depend on the hardness of the pharmaceutical dosage form. For instance, due to its breaking strength, impact strength, elasticity modulus and tensile strength, respectively, the pharmaceutical dosage form can preferably be deformed when exerting an external force, for example using a hammer, but cannot be pulverized, i.e., crumbled into a high number of fragments. In other words, the pharmaceutical dosage form according to the invention is characterized by a certain degree of breaking strength, but not necessarily aiso by a certain degree of form siabiiiiy.

Therefore, in the meaning of the specification, a pharmaceutical dosage form that is deformed when being exposed to a force in a particular direction of extension but that does not break is to be regarded as having the desired breaking strength in said direction of extension.

Due to the anisotropic mechanical properties in directions of extension E, and E2, the application of force with conventional means, e.g. the application of 400 N, may cause disruption of the dosage form to a certain and limited degree, but may not cause pulverization thereof. For example, when the breaking strength in direction of extension E2 is below 400 N, the dosage form is disrupted into pieces by applying 400 N in direction of extension E2. Preferably, however, said pieces may not be disrupted any further by applying 400 N or more.

It has been surprisingly found that the outer shape of the pharmaceutical dosage forms may be varied within wide limits without diminishing their breaking strength.
Although a variation of the outer shape of the pharmaceutical dosage forms may cause a decrease of the breaking strength in a certain direction of extension, it has been found that specific forms and shapes establish a certain degree of fragility at predetermined sites of fracture (weakening points) without completely diminishing the overall breaking strength of the remainder of the pharmaceutical dosage forms (fragments).

The degree of weakening can be controlled and when exerting a force on the pharmaceutical dosage forms, said weakening points can serve as predetermined sites of fracture, provided that the amount of force is sufficiently high.

The degree of fragility may be adjusted to values that are still well above the typical breaking strength of conventional pharmaceutical dosage forms, e.g. well above 100 N or 200 N. In particular, it has been surprisingly found that pharmaceutical dosage forms can be designed which can be fractured into large pieces, e.g. into halves or thirds, by conventional means (e.g. tablet crushers), but not any further. In consequence, the resultant fragments (subunits) in turn exhibit a breaking strength which is far above the breaking strength at the predetermined site of fracture, e.g. well above 500 N, preferably in any direction of extension.

Figure 7A) is a schematic view of a conventionai obiong iabiei. Cross-sectional areas (35) and (35') are smaller than cross-sectional areas (36) and (36') which in turn are smaller than cross-sectional area (37). When exerting external force (F2) in direction of extension (E2), the implied pressure is not constant but varies with varying cross-sectional areas. For example, the pressure implied at the section having cross-sectional area (37) is lower than the pressure at the section having cross-sectional areas (35) and (35'), respectively, because cross-sectional area (37) is larger than cross-sectional areas (35) and (35').

Figure 7B) is a schematic view of an embodiment of a pharmaceutical dosage form according to the invention. An increased surface to volume ratio and surface to weight ratio, respectively, is achieved by taper T right in the middle of the pharmaceutical dosage form.
Under these circumstances, when exerting external force (F2) in direction of extension (E2), the implied pressure at the section having cross-sectional area (42) is higher than the pressure at the section having cross-sectional areas (38) and (38'), respectively, because cross-sectional area (42) is smaller than cross-sectional areas (38) and (38'). The pharmaceutical dosage form depicted in Figure 7B) exhibits a breaking strength in direction of extension (E2) that is substantially lower than the breaking strength of the pharmaceutical dosage form depicted in Figure 7A) in direction of extension (E2).

In Figures 7A) and 7B), direction of extension (E2) is the principal direction of extension of the pharmaceutical dosage forms (main direction of extension, major direction of extension).

The predetermined sites of fracture (weakening points) can improve the patient compliance, as patients having problems in swallowing large pharmaceutical dosage forms can fracture the pharmaceutical dosage forms along the predetermined sites of fracture (weakening points) prior to swallowing. Those patients can limitedly fracture the pharmaceutical dosage form into fragments of a size they can swallow. However, as the resultant fragments in turn cannot be comminuted any further, at least not by conventional means, the pharmaceutical dosage forms are still tamper resistant, i.e. efficiently avoid drug abuse and (unintentional) drug overdose, respectively. In other words, the pharmaceutical dosage forms according to the invention realize both, a very high mechanical resistance in order to avoid misuse as well as a certain degree of a mechanical weakness in order to improve patient compliance.

In a preferred embodiment, the pharmaceutical dosage form according to the invention provides fragments when exerting a force higher than B2 in direction of extension E2, preferably under the standard conditions for measuring the breaking strength set forth above, said fragments in turn having a breaking strength of preferabiy ai ieasi 500 N, at least 550 N or at least 600 N; more preferably at least 650 N, at least 700 N
or at least 750 N; still more preferably at least 800 N, at least 850 N or at least 900 N; yet more preferably at least 950 N, at least 1000 N or at least 1100 N; and in particular at least 1200 N, at least 1300 N, at least 1400 N or at least 1500 N; preferably in any (each and every) of their directions of extension.

Preferably, the dosage form provides at most 10, more preferably at most 8, still more preferably at most 6, yet more preferably at most 5, most preferably at most 4 and in particular at most 3 fragments when exerting a force that is higher than B2 in direction of extension E2.

Preferably the volume of each fragment is at least 5%, more preferably at least 10%, still more preferably at least 15%, yet more preferably at least 20%, most preferably at least 25% and in particular at least 30% of the volume of the pharmaceutical dosage form.

It has been surprisingly found that the release profile of the pharmaceutical dosage form may be varied within certain limits by varying the outer shape of the pharmaceutical dosage form without diminishing its tamper resistance. Therefore, particularly at a high drug load, the pharmaceutical dosage forms according to the invention allow to realize release profiles that may not be achieved with conventional pharmaceutical dosage forms having an increased breaking strength (e.g. oblong tablets).

Typically, the pharmaceutical dosage form according to the invention assumes the form of a tablet. The pharmaceutical dosage form is preferably not in film form.

The pharmaceutical dosage form (tablet) according to the invention may assume various shapes. Preferably, from top view, the shape of the pharmaceutical dosage form can be substantially hexagonal, elliptic, cyclic, oblong, rectangular, squared, triangular, and the like. Preferably, from side view, the shape of the pharmaceutical dosage form can be substantially flat-convex, biconvex, flat with facet, flat without facet, cyclic, and the like.

For example, the pharmaceutical dosage form (tablet) according to the invention may assume simple geometries, such as spherical, planar, cubic, hexagonal and cylindrical, or complex geometries, such as convex, hollow cylindrical, doughnut-shaped, hemispheric, cruciform, astral (cf. Figures 13, 14 and 15).

Particularly preferred shapes of the dosage form (tabiei) according to Lilt!
i11veIMo die already been described in connection with the first and second aspects of the invention above.

In a particularly preferred embodiment, the pharmaceutical dosage form (tablet) according to the invention can be described as a body having a recess or cavity on at least one side, preferably two recesses or two cavities on two sides, preferably on opposing sides.
Alternatively, said cavities and recesses, respectively, may be regarded as bulges, indentations, troughs, hollows, depressions, synclines, deepenings, and the like. Examples of such embodiments are depicted in Figures 8-10, 16A) and 16B).

The pharmaceutical dosage form is preferably adapted for oral administration, i.e., should be capable of being swallowed. Thus, obscure geometrical forms which are obviously harmful cannot be regarded as pharmaceutical dosage forms according to the invention.
According to a preferred embodiment, the pharmaceutical dosage form is characterized by a specific aspect ratio. For the purpose of the specification, the aspect ratio is defined as the ratio of the main direction of extension of the dosage form to the maximum extension of the pharmaceutical dosage form orthogonal to said main direction of extension, e.g.
maximum length to maximum height (and maximum length to maximum width, respectively). Preferably, said aspect ratio is within the range of 2.4 1.3 :
1, more preferably 2.4 1.0 : 1, still more preferably 2.4 0.8 : 1, yet more preferably 2.4 0.6 :
1, most preferably 2.4 0.4 : 1 and in particular 2.4 0.2 : 1.

According to a preferred embodiment, the pharmaceutical dosage form is characterized by a specific length to height to width ratio, where length > height >_ width.
For the purpose of the specification, in this embodiment the length corresponds to the main direction of extension of the dosage form, the height corresponds to the maximum extension of the pharmaceutical dosage form orthogonal to the length, and the width corresponds to the maximum extension orthogonal to the length and orthogonal to the width (Cartesian space).
Preferably, the length to height to width ratio is within the range of 4.7 2.0 : 2.0 1.0 : 1, more preferably 4.7 1.6 : 2.0 0.8 : 1, still more preferably 4.7 1.2 : 2.0 0.6 : 1, yet more preferably 4.7 0.8 : 2.0 0.4 : 1, most preferably 4.7 0.6 : 2.0 0.3 : 1, and in particular 4.7 0.4: 2.0 0.2 : 1.

Preferably, a portion of the surface of the pharmaceutical dosage form is convex, i.e.
curved out or bulged outward, and another portion of its surface is concave, i.e. curved in or hollowed inward. For the purpose of the specification, the radius of curvature is 1101 critical.
Preferably, the overall surface of the pharmaceutical dosage form can be divided into concave portions, convex portions and planar portions. Typically, the sum of the concave portions, convex portions and planar portions corresponds to the overall surface of the dosage form. However, at least theoretically, a given portion can be convex and concave simultaneously (saddle). Under these circumstances, the sum of the concave portions, convex portions and planar portions exceeds the overall surface of the dosage form.

In a preferred embodiment, the convex portion of the surface of the dosage form is at most 95%, more preferably at most 90% or at most 85%, still more preferably at most 80% or at most 75%, yet more preferably at most 70% or at most 65%, most preferably at most 60%
or at most 55% and on particular at most 50% or at most 45%, based on the sum of concave portions, convex portions and planar portions.

In another preferred embodiment, the concave portion of the surface of the dosage form is at least 5%, more preferably at least 10% or at least 15%, still more preferably at least 20%
or at least 25%, yet more preferably at least 30% or at least 35%, most preferably at least 40% or at least 45% and in particular at least 50% or at least 55%, based on the sum of concave portions, convex portions and planar portions.

In a preferred embodiment of the pharmaceutical dosage form according to the invention, the maximum extension of the dosage form orthogonal to the main area of extension of the dosage form is spaced from the centre of mass of the dosage form parallel to said main area of extension. For the purpose of the specification, the main area of extension of the dosage form is preferably the largest plain area that can be placed along a cut of the body of the dosage form. This embodiment is further illustrated in Figure 17A) which shows a schematic view of the cross-sectional face of the preferred pharmaceutical dosage form depicted in Figure 10. The maximum extension (56) of the dosage form orthogonal to the main area of extension (57) of the dosage form is spaced from the centre of mass (58) of the dosage form parallel to said main area of extension (57).

Preferably, the closest distance (59) from the maximum extension of the dosage form orthogonal to the main area of extension of the dosage form to the centre of mass of the dosage form is at least 0.5 mm, more preferably at least 1.0 mm, still more preferably at least 1.5 mm, yet more preferably at least 2.0 mm, most preferably at least 2.5 mm and in particular at least 3.0 mm.

Preferably, the pharmaceutical dosage form according to the invention has a shape so that when exerting an increasing amount of force in direction of extension E2, the dosage form is firstly deformed and, when the amount of force reaches the breaking strength B2, defor-mation causes tractive forces that lead to disruption of the dosage form.

In a preferred embodiment, the cross sectional area of the pharmaceutical dosage form that is orthogonal to the main direction of extension and that contains the centre of mass of the dosage form has a shape so that at least 50%, more preferably at least 60% and in particular at least 75% of its area is spaced at least 0.2 mm, at least 0.3 mm, at least 0.4 mm or at least 0.5 mm, more preferably at least 0.6 mm, at least 0.7 mm, at least 0.8 mm or at least 0.9 mm, still more preferably at least 1.0 mm, at least 1.1 mm, at least 1.2 mm or at least 1.3 mm, yet more preferably at least 1.4 mm, at least 1.5 mm, at least 1.6 mm or at least 1.7 mm, most preferably at least 1.8 mm, at least 1.9 mm, at least 2.0 mm or at least 2.1 mm and in particular at least 2.2 mm, at least 2.3 mm, at least 2.4 mm or at least 2.5 mm from the centre of mass. Preferably, said cross sectional area contains the centre of mass as well as direction of extension E,, or the centre of mass as well as direction of extension E2. This embodiment is further illustrated in Figure 17B) in which the centre of mass (58) is surrounded by concentric rings (60a) to (60d) indicating increasing distances from the centre of mass (58). The portions of the cross sectional area which do not overlap with any of the concentric rings are spaced from the centre of mass by more than the radius of ring (60d).

In a preferred embodiment of the pharmaceutical dosage form according to the invention, the closest distance of each and every geometrical point within the dosage form to the surface of the dosage form is at most 10 mm, at most 9 mm, at most 8 mm or at most 7.5 mm; more preferably at most 7.0 mm, at most 6.5 mm or at most 6.0 mm; still more preferably at most 5.8 mm, at most 5.6 mm, at most 5.4 mm, at most 5.2 mm or at most 5.0 mm; yet more preferably at most 4.8 mm, at most 4.6 mm, at most 4.4 mm, at most 4.2 mm or at most 4.0 mm; yet more preferably at most 3.8 mm, at most 3.6 mm, at most 3.4 mm, at most 3.2 mm or at most 3.0 mm; most preferably at most 2.8 mm, at most 2.6 mm, at most 2.4 mm, at most 2.2 mm or at most 2.0 mm; and in particular at most 1.9 mm, at most 1.8 mm, at most 1.7 mm, at most 1.6 mm, at most 1.5 mm, at most 1.4 mm, at most 1.3 mm, at most 1.2 mm, at most 1.1 mm, or at most 1.0 mm.

Preferably, the pharmaceutical dosage form according to the invention is not radial symmetric about its main direction of extension (principal direction of extension), preferably the pharmaceutical dosage form is not radial symmetric at all.

In a preferred embodiment, the symmetry of the pharmaceutical dosage form is selected from the group consisting of Ci, C, C1, C2, C3, C4, C5, C6, C7, C8, C.o, C2h, C3h, CO, C5h, C6h, C7h, C8h, Cmh, C2v, C3v, C4,, C5v, C6v. C7v, C8v, Cmv, C3i, D2, D3, D4, D5, D6, D7, D8, D-, D2h, D3h, D4h, D5h, D6h, D7h, D8h, Dmh, D2d, D3d, D4d, D5d, D6d, D7d, D8d, D-d, S2, S4, S61 S8, T, Th, Td, 0, Oh and Ih according to Schoenflies notation. D2h and D4h are particularly preferred.

In a preferred embodiment, the centre of mass of the pharmaceutical dosage form lies within the main area of extension of the dosage form. Preferably, the pharmaceutical dosage form is symmetric about its main area of extension.

The pharmaceutical dosage form according to the invention preferably has a breaking strength B, of at least 500 N in at least one of its directions of extension, namely E,, preferably, however, in more than one of its directions of extension, more preferably in a plurality of directions of extension.

In direction of extension E, the pharmaceutical dosage form according to the invention preferably has a breaking strength B, of at least 500 N, preferably of at least 510 N, at least 520 N, at least 530 N, at least 540 N or at least 550 N; more preferably at least 560 N, at least 570 N, at least 580 N, at least 590 N or at least 600 N; still more preferably at least 620 N, at least 640 N, at least 660 N, at least 680 N or at least 700 N; yet more preferably at least 720 N, at least 740 N, at least 760 N, at least 780 N or at least 800 N; most preferably at least 850 N, at least 900 N, at least 950 N, at least 1000 N or at least 1050 N;
and in particular at least 1100 N, at least 1200 N, at least 1300 N, at least 1400 N, at least 1500 N, at least 1600 N, at least 1700 N, at least 1800 N, at least 1900 N or at least 2000 N.

E, may be any direction of extension of the pharmaceutical dosage form, i.e.
any straight line connecting any first point on the surface of the pharmaceutical dosage form with any second point on the surface of the pharmaceutical dosage form. Preferably, said straight line lies completely within the body of the pharmaceutical dosage form, i.e., preferably does not "leave" the pharmaceutical dosage form somewhere and "reenters" the pharmaceutical dosage form elsewhere. Preferably, the distance between said first point and said second point of direction of extension E1 is at least 50%, more preferably at least 75% of the main direction of extension of the pharmaceutical dosage form. The latter is defined as the maximal distance between two points on the surface of the pharmaceutical dosage form (principal direction of extension), regardless of whether it compieteiy lies within the body of the dosage form or not. Preferably, the main direction of extension of the dosage form goes through the centre of mass of the dosage form.

In a preferred embodiment, the main direction of extension of the pharmaceutical dosage form according to the invention is not longer than 32 mm, more preferably not longer than 30 mm, still more preferably not longer than 28 mm, yet more preferably not longer than 26 mm, most preferably not longer than 24 mm, and in particular not longer than 22 mm.

In a preferred embodiment, the pharmaceutical dosage form according to the invention is deformed in the breaking strength tester when the measurement of the breaking strength is conducted. Deformation means typically compression, although bending, turning and other modes of deformation are also possible. When exerting a force of 100 N in direction of extension E1, the dosage form is preferably deformed by at least 0.1 mm but does not break. When exerting a force of 200 N in direction of extension E1, the dosage form is preferably deformed by at least 0.2 mm, more preferably at least 0.3 mm, but does not break. When exerting a force of 300 N in direction of extension E1, the dosage form is preferably deformed by at least 0.5 mm, more preferably at least 0.7 mm, but does not break. When exerting a force of 400 N in direction of extension E1, the dosage form is preferably deformed by at least 1.0 mm, more preferably at least 1.2 mm, but does not break. When exerting a force of 500 N in direction of extension E1, the dosage form is preferably deformed by at least 1.5 mm, more preferably at least 2.0 mm, but does not break. When exerting a force of 1000 N in direction of extension E1, the dosage form is preferably deformed by at least 3.0 mm, more preferably at least 4.0 mm, but does not break.

For theoretical reasons, every pharmaceutical dosage form comprises an unlimited number of directions of extension.

Some typical directions of extension of a preferred pharmaceutical dosage form according to the invention are illustrated in Figure 8. Figure 8A) shows different directions of extension. Directions of extension E9 and E10 are parallel to one another, directions of extension E10, Eõ and E12 (and E9, respectively) are orthogonal to one another. Directions of extension E9, E10, E11 and E12 enter the body of the pharmaceutical dosage form at sites (43), (44), (45) and (46), respectively. Direction of extension E9 lies completely within the body of the pharmaceutical dosage form; it enters the body of the pharmaceutical dosage form at site (43 and leaves the body of the pharmaceutical dosage form at its rear side (not shown). Direction of extension E10, however, enters the body of the pharmaceutical dosage form at site (44), transiently leaves the body of the pharmaceutical dosage form at the rear side of the upper front edge (not shown) and re-enters the body of the pharmaceutical dosage form at site (44'), before it finally leaves the body of the pharmaceutical dosage form at its rear side (not shown). Direction of extension E13 enters the body of the pharmaceutical dosage form at corner (47) and leaves the pharmaceutical dosage form at the diagonally opposite corner. The dotted line of direction of extension E13 represents the maximal distance between two points on the surface of the pharmaceutical dosage form, i.e., direction of extension E13 is the main direction of extension of the pharmaceutical dosage form. In total, the pharmaceutical dosage form depicted in Figure 8A) has four of such main directions of extension.

Figure 8B) shows a cross-sectional view of the pharmaceutical dosage form according to Figure 8A). The cross-sectional face (48, hatched) assumes the shape of an H.
The pharmaceutical dosage form assumes a rectangular shape with two recesses (50, 50') on opposing sides. In other words, the pharmaceutical dosage form has the size of a comparatively flat rectangle with an edge (49) circumventing the upper side and an edge (49') circumventing the opposite side. Pharmaceutical dosage forms of this type may be manufactured by a tabletting tool that is equipped with a so-called H-plunger.

Figure 9A) is a schematic view of the cross-sectional face (48) of the pharmaceutical dosage form depicted in Figure 8A). Figure 9B) is a schematic view of the cross-sectional face of a pharmaceutical dosage form according to the invention that is similar to the cross-sectional face of the pharmaceutical dosage form depicted in Figure 9A). The rim (49) of the cross-sectional face, however, is rounded (51) thereby forming a convex surface area.
Figure 9C is a schematic view of the cross-sectional face of a pharmaceutical dosage form according to the invention that is similar to the cross-sectional face of pharmaceutical dosage forms depicted in Figures 9A) and 9B). The edge at the bottom of the recess is rounded (52) thereby forming a concave surface area.

Figure 10 is a schematic view of a particularly preferred pharmaceutical dosage form according to the invention. Figure 10A) is a top view which is identical from both opposing sides and Figure 1 0B) is a side-view which is also identical from both opposing sides.

In the Cartesian space, the principal dimensions of the pharmaceutical dosage form schematically illustrated in Figure 10 can be defined as a, b and c, where a =
a,+a2+a3, b =
b,+b2+b3 and c=c,+c2+c3. Preferred relative dimensions D1 to D6 of the pharmaceutical dosage form depicted in Figure 10 can be defined in relative relations of a, b and c; a,, a2 and a3; b,, b2 and b3; and c,, c2 and c3, respectively:

D1: c > a >_b; c > a > b;
D2: c > 1.5 a; c > 2 a; c > 2.5 a; c > 3 a;
D3: a2>a,a3;a2>1.1 a,1.1 a3;a2>1.2a,1.2a3;a2>1.3a,1.3a3;
D4: b2 _ b,b3;b2?1.1 b,1.1 b3;b2>_1.2b,1.2b3;b2 _1.3b,1.3b3;
D5: b25 b,b3;b2:5 0.9b,0.9b3;b2<0.8b,0.8b3;b2:5 0.7b,0.7 b3; and/or D6: c2>c1 c3;c2>1.1 c,-1.1 c3;c2>1.2c,1.2c3;c2>1.3c,-1.3c3.

Preferred embodiments D7 to D18 regarding the absolute dimensions of the pharmaceutical dosage form depicted in Figure 10 are displayed in the table here below:
mm D7 D8 D9 D10 D11 D12 a 8.6 4.3 8.6 2.1 8.6 1.0 9.0 4.5 9.0 2.2 9.0 1.1 b 4.9 2.5 4.9 1.3 4.9 0.7 4.3 2.1 4.3 1.0 4.3 0.6 C 21.9 11.0 21.9 5.5 21.9 2.7 20.4 10.2 20.4 5.1 20.4 2.5 mm D13 D14 D15 D16 D17 D18 a 9.0 4.3 9.0 2.1 9.0 1.0 9.1 4.5 9.1 2.2 9.1 1.1 b 4.1 2.5 4.1 1.3 4.1 0.7 4.5 2.1 4.5 1.0 4.5 0.6 c 20.5 11.0 20.5 5.5 20.5 2.7 20.5 10.2 20.5 5.1 20.5 2.5 Preferred embodiments D19 to D30 regarding the absolute dimensions of the pharmaceutical dosage form depicted in Figure 10 are displayed in the table here below:

a 8.6 4.3 8.6 2.1 8.6 1.0 9.0 4.5 9.0 2.2 9.0 1.1 a, 3.3 1.6 3.3 0.8 3.3 0.4 3.5 1.8 3.5 0.9 3.5 0.5 a2 2.1 1.0 2.1 0.5 2.1 0.3 2.1 1.1 2.1 0.6 2.1 0.3 a3 3.3 1.6 3.3 0.8 3.3 0.4 3.5 1.8 3.5 0.9 3.5 0.5 b 4.9 2.5 4.9 1.3 4.9 0.7 4.3 2.1 4.3 1.0 4.3 0.6 b, 0.9 0.5 0.9 0.3 0.9 0.2 0.9 0.4 0.9 0.2 0.9 0.1 b2 3.1 1.5 3.1 0.7 3.1 0.4 2.6 1.3 2.6 0.6 2.6 0.3 b3 0.9 0.5 0.9 0.3 0.9 0.2 0.9 0.4 0.9 0.2 0.9 0.1 C 21.9 11.0 21.9 5.5 21.9 2.7 20.4 10.2 20.4 5.1 20.4 2.5 c, 3.2 1.6 3.2 0.8 3.2 0.4 3.3 1.7 3.3 0.9 3.3 0.4 C2 15.6 7.8 15.6 3.9 15.6 2.0 13.8 6.9 13.8 3.5 13.8 1.7 c3 3.2 1.6 3.2 0.8 3.2 0.4 3.3 1.7 3.3 0.9 3.3 0.4 a 9.0 4.3 9.0 2.1 9.0 1.0 9.1 4.5 9.1 2.2 9.1 1.1 a, 3.2 1.6 3.2 0.8 3.2 0.4 3.2 1.8 3.2 0.9 3.2 0.5 a2 2.6 1.0 2.6 0.5 2.6 0.3 2.7 1.1 2.7 0.6 2.7 0.3 a3 3.2 16 32 08 32+na 32 18 32 09 3:2 0.5 b 4.1 2.5 4.1 1.3 4.1 0.7 4.5 2.1 4.5 1.0 4.5 0.6 b, 1.0 0.5 1.0 0.3 1.0 0.2 1.0 0.4 1.0 0.2 1.0 0.1 b2 2.1 1.5 2.1 0.7 2.1 0.4 2.5 1.3 2.5 0.6 2.5 0.3 b3 1.0 0.5 1.0 0.3 1.0 0.2 1.0 0.4 1.0 0.2 1.0 0.1 c 20.5 11.0 20.5 5.5 20.5 2.7 20.5 10.2 20.5 5.1 20.5 2.5 c, 3.3 1.6 3.3 0.8 3.3 0.4 3.3 1.7 3.3 0.9 3.3 0.4 C2 13.9 7.8 13.9 3.9 13.9 2.0 13.9 6.9 13.9 3.5 13.9 1.7 C3 3.3 1.6 3.3 0.8 3.3 0.4 3.3 1.7 3.3 0.9 3.3 0.4 The pharmaceutical dosage form according to the invention preferably has a breaking strength B2 in a second direction of extension E2, wherein B2 < 500 N.

Direction of extension E, differs from direction of extension E2. E2 may be any direction of extension of the pharmaceutical dosage form, i.e. any straight line connecting any first point on the surface of the pharmaceutical dosage form with any second point on the surface of the pharmaceutical dosage form. Preferably, said straight line lies completely within the body of the pharmaceutical dosage form, i.e., preferably does not "leave" the pharmaceutical dosage form somewhere and "re-enters" the pharmaceutical dosage form elsewhere. Preferably, the distance between said first point and said second point is at least 50%, more preferably at least 75% of the main direction of extension of the pharmaceutical dosage form, the latter being defined as the maximal distance between two points on the surface of the pharmaceutical dosage form.

Preferably, E, and E2 assume an angle with one another of from 10 to 170 , more preferably 20 to 160 , still more preferably 30 to 150 , yet more preferably 40 to 140 , most preferably 50 to 130 , and in particular 60 to 120 . In a particularly preferred embodiment, E, is orthogonal to E2.

Preferably, E2 is the main direction (principal direction) of extension of the pharmaceutical dosage form, i.e., B2 can preferably be measured by placing the pharmaceutical dosage form between the jaws of the measuring device so that the two jaws have the maximum distance from one another but are each in contact the pharmaceutical dosage form.

In a preferred embodiment, E, and E2, which both are preferably orthogonal to each another, lie within the main area of extension of the dosage form, which main area of extension preferably also contains the centre of mass of the dosage form.

In a preferred embodiment, B2 is below 490 N, below 480 N, below 460 N, below 440 N, or below 420 N; more preferably below 400 N, below 380 N, below 360 N, below 340 N, or L.nln... 7']/1 AI= =~II f..... l.l.. L...1..... 7/1/1 1I L...I..... 11O(1 AI
L...1..... '11'/1 AI L...I..... '1A/1 AI
UGIVVV ..lLV 114; still! IIIVIG 1JI GIGI QUIy UGIVVV JVV IV, UGIVVV LUV IV, UGIVVV '_UV 1V, UGIVVV LYV IV VI
below 220 N.

In another preferred embodiment, B2 is at least 200 N, at least 220 N, at least 240 N, at least 260 N, or at least 280 N; more preferably at least 300 N, at least 320 N, at least 340 N, at least 360 N, or at least 380 N; still more preferably at least 420 N, at least 440 N, at least 460 N, or at least 480 N.

Preferably, the ratio of B, to B2 is within the range of from 100: 1 to 1.1 :
1, more preferably 75 : 1 to 1.2 : 1, still more preferably 50 : 1 to 1.3 : 1, yet more preferably 25 : 1 to 1.4 : 1, most preferably 10 : 1 to 1.5 : 1 and in particular 5 : 1 to 1.6 : 1.

Preferably, the difference B, - B2 is at least 10 N, at least 20 N, at least 30 N, at least 40 N
or at least 50 N, more preferably at least 60 N, at least 70 N, at least 80 N
or at least 90 N, still more preferably at least 100 N, at least 125 N, at least 150 N, at least 175 N or at least 200 N, most preferably at least 250 N, at least 300 N, at least 350 N, at least 400 or at least 450 N, and in particular at least 500 N, at least 600 N, at least 750 or at least 1000 N.

In a preferred embodiment of the pharmaceutical dosage form according to the invention, B, and B2 satisfy one of the following conditions C 1 to C 72:

C B2 B, C B2 B, C B2 B, 1 100 N < B2 < 500 N 500 N < B, 33 100 N < B2 < 400 N 500 N < B, 57 100 N < B2 < 300 N 500 N < B, 2 100N<B2<500N 600N<B, 34 100N<B2<400N 600N<B, 58 100N<B2<300N 600N<B, 3 100N<B2<500N 700N<B, 35 100N<B2<400N 700N<B, 59 100N<B2<300N 700N<B, 4 100N<B2<500N 800N<B, 36 100N<B2<400N 800N<B, 60 100N<B2<300N 800N<B, 100N<B2<500N 900N<B, 37 100N<B2<400N 900N<B, 61 100N<B2<300N 900N<B, 6 100N<B2<500N 1000N<B, 38 100N<B2<400N 1000N<B, 62 100N<B2<300N 1000N<B, 7 100N<B2<500N 1500N<B, 39 100N<B2<400N 1500N<B, 63 100N<B2<300N 1500N<B, 8 100 N < B2 < 500 N 2000 N < B, 40 100 N < B2 < 400 N 2000 N < B, 64 100 N <
B2 < 300 N 2000 N < B, 9 200 N < B2 < 500 N 500 N < B, 41 200 N < B2 < 400 N 500 N < B, 65 200 N < B2 < 300 N 500 N < B, 200 N < B2 < 500 N 600 N < B, 42 200 N < B2 < 400 N 600 N < B, 66 200 N < B2 <
300 N 600 N < B, 11 200 N < B2 < 500 N 700 N < B, 43 200 N < B2 < 400 N 700 N < B, 67 200 N <
B2 < 300 N 700 N < B, 12 200N<82<500N 800N<B, 44 200N<B2<400N 800N<B, 68 200N<B2<300N 800N<B, 13 200 N < B2 < 500 N 900 N < B, 45 200 N < B2 < 400 N 900 N < B, 69 200 N <
B2 < 300 N 900 N < B, 14 200 N < B2 < 500 N 1000 N < B, 46 200 N < B2 < 400 N 1000 N < B, 70 200 N <
B2 < 300 N 1000 N < B, 200 N < B2 < 500 N 1500 N < B, 47 200 N < B2 < 400 N 1500 N < B, 71 200 N < B2 < 300 N 1500 N < B, 16 200 N < B2 < 500 N 2000 N < B, 48 200 N < B2 < 400 N 2000 N < B, 72 200 N <
B2 < 300 N 2000 N < B, 17 300 N < B2 < 500 N 500 N < B, 49 300 N < B2 < 400 N 500 N < B, 18 300N<B2<500N 600N<B, 50 300N<B2<400N 600N<B, 19 300 N < B2 < 500 N 700 N < B, 51 300 N < B2 < 400 N 700 N < B, 300 N < B2 < 500 N 800 N < B, 52 300 N < B2 < 400 N 800 N < B, 21 300N<B2<500N 900N<B1 53 300N<B2<400N 900N<Bi 22 300 N < B2 < 500 N 1000 N < B1 54 300 N < B2 < 400 N 1000 N < Bi 23 300 N < B2 < 500 N 1500 N < Bi 55 300 N < B2 < 400 N 1500 N < Bi 24 300N<B2<500N 2000N<Bi 56 300N<B2<400N 2000N<Bi 25 400 N < B2 < 500 N 500 N < Bt 26 400 N < B2 < 500 N 600 N < Bi 27 400N <B2<500N 700N <Bi 28 400 N < B2 < 500 N 800 N < B, 29 400 N < B2 < 500 N 900 N < B, 30 400 N < B2 < 500 N 1000 N < B, 31 400N<B2 5500N 1500N<Bi 32 400 N < B2 < 500 N 2000 N < Bi Further preferred embodiments C 73 to C 96 are summarized in the table here below:

C B2 Bi C B2 Bi C B2 Bi 73 320N<B2<420N 500N<Bi 81 330N<B2<420N 500N<Bi 89 340N<B2<420N 500N<Bi 74 320N<B2<420N 600N<Bi 82 330N<B2<420N 600N<Bi 90 340N<B2<420N 600N<Bi 75 320 N < B2 < 420 N 700 N < Bi 83 330 N < B2 < 420 N 700 N < Bi 91 340 N <
B2 < 420 N 700 N < Bi 76 320 N < B2 < 420 N 800 N < Bi 84 330 N < B2 < 420 N 800 N < Bi 92 340 N <
B2 < 420 N 800 N < Bi 77 320 N < B2 < 420 N 900 N < B, 85 330 N < B2 < 420 N 900 N < B, 93 340 N <
B2 < 420 N 900 N < Bi 78 320 N < B2 < 420 N 1000 N < B, 86 330 N < B2 < 420 N 1000 N < B, 94 340 N <
B2 < 420 N 1000 N < Bi 79 320 N < B2 < 420 N 1500 N < B, 87 330 N < B2 < 420 N 1500 N < B, 95 340 N <
B2 < 420 N 1500 N < Bi 80 320 N < B2 < 420 N 2000 N < B, 88 330 N < B2 < 420 N 2000 N < B, 96 340 N <
B2 < 420 N 2000 N < Bi Due to the property B2 < B1, the pharmaceutical dosage form according to the invention preferably has anisotropic mechanical properties, i.e., is mechanically weakened with respect of at least one of its directions of extension (E2) compared to its mechanical strength with respect of at least another of its directions of extension (E,).
In a preferred embodiment, the quality of the mechanical weakening is such that the pharmaceutical dosage form may be fractured once by exerting a sufficient amount of force in direction of extension E2, but the thus obtained fragments are substantially break resistant in any (each and every) of their directions of extension, i.e., may not be fractured any further by exerting the same amount of force in any (each and every) of their directions of extension.
Preferably, said amount of force is 400 N, 500 N, 600 N, 700 N, 800 N, 900 N, 1000 N, 1100 N, 1200 N, 1300 N, 1400 N, or 1500 N. In consequence, according to this embodiment the pharmaceutical dosage form may be fractured once by exerting a sufficient amount of force in direction of extension E2, but may not be fractured any further unless the exerted force is increased. Preferably, the number of fragments that are obtained when the pharmaceutical dosage form is fractured once is limited, preferably only two, three, four, five or six fragments, preferably of substantially identical or different size and/or weight, are obtained.

In a preferred embodiment, the surface S [mm2] to weight W [mg] ratio S / W of the pharmaceutical dosage form according to the invention is at least 0.50 mm2/mg.
Preferably, S / W is at least 0.51, at least 0.52, at least 0.53, at least 0.54 or at least 0.55; more preferably at least 0.56, at least 0.57, at least 0.58, at least 0.59 or at least 0.60; still more preferably at least 0.61, at least 0.62, at least 0.63, at least 0.64 or at least 0.65; yet more preferably at least 0.66, at least 0.67, at least 0.68, at least 0.69 or at ieasi 0.70; most preferably at least 0.705, at least 0.710, at least 0.715, at least 0.720, at least 0.725, at least 0.730, at least 0.735, at least 0.740, at least 0.745 or at least 0.750;
and in particular at least 0.755, at least 0.760, at least 0.765, at least 0.770, at least 0.775, at least 0.780, at least 0.785, at least 0.790, at least 0.795 or at least 0.80 mm2/mg.

In a preferred embodiment, the pharmaceutical dosage form according to the invention has a total surface S defined by the formula S?A.W2'3, wherein A is at least 4.5, i.e. S 2: 4.5 = W2'3.

For example, when the pharmaceutical dosage form according to the invention has a total weight of 623 mg, its total surface S is preferably at least 328 mm2 (4.5-623 2/3 ) and when the pharmaceutical dosage form according to the invention has a total weight of 983 mg, its total surface S is preferably at least 445 mm2 (4.5.983213).

Methods for measuring the total surface of a pharmaceutical dosage form are known to the skilled artisan. For example, the total surface may be calculated from the three dimensional extension of the pharmaceutical dosage form based on simple geometrical considerations (cf., e.g., Eudragit Application Guidelines, 10th edition, 07/2007, Rohm GmbH, Darmstadt).

For example, the geometry of the conventional tablet that is depicted in Figure 11 may be described by a cylindrical element (53) that is located between two spherical caps (54a) and (54b). The volumes V(53), V(54a) and V(54b) as well as the surfaces S(53), S(54a) and S(54b) can be calculated according to the following formulas:

V(54a) = V(54b) = 1 7t h1(3r22 +h12) V(53) _ ir r22 h2 6 ` Vt = V(53) + V(54a) + V(54b) S(53) = 2 it r2 h2 S(54a) = S(54b) = 2 it r1 h1 St = S(53) + S(54a) + S(54b) In approximation, the pharmaceutical dosage form may also be mentally divided into a plurality of identical cubic volume elements of suitable size (voxels) and the total surface may be determined by counting the squared area elements (pixels) being located at the surface. This approximation is further illustrated in Figure 12 where the pharmaceutical dosage form according to Figure 8B) is divided into a plurality of voxels (55).

Preferably, when measuring the total surface of the pharmaceutical dosage form, the micro-fine structure of the pharmacologically active compound (A) and of all other constituents of the dosage form including polymers and pharmaceutical excipients, e.g. their porosity, is not taken into account. For the purpose of the specification, the term "surface" of the pharmaceutical dosage form preferably refers to the macroscopic surface (outer dimensions, silhouette). In other words, for the purpose of determining the surface of the pharmaceutical dosage form, the surface structure is preferably considered perfectly smooth.

In a preferred embodiment of the pharmaceutical dosage form according to the invention, A
is 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9 or 6.0; more preferably 6.05, 6.1, 6.15, 6.2, 6.25, 6.3, 6.35, 6.4, 6.45, 6.5, 6.55, 6.6, 6.65, 6.7, 6.75, 6.8, 6.85, 6.9, 6.95, 7.0, 7.05, 7.1, 7.15, 7.2, 7.25, 7.3, 7.35, 7.4, 7.45 or 7.5.

In another preferred embodiment of the pharmaceutical dosage form according to the invention, A is 7.6, 7.7, 7.8, 7.9, 8.0, 8.1, 8.2, 8.3, 8.4, 8.5, 8.6, 8.7, 8.8, 8.9 or 9.0; more preferably 9.1, 9.2, 9.3, 9.4, 9.5, 9.6, 9.7, 9.8, 9.9, 10.0, 10.1, 10.2, 10.3, 10.4 or 10.5; most preferably 10.6, 10.7, 10.8, 10.9, 11.0, 11.1, 11.2, 11.3, 11.4, 11.5, 11.6, 11.7, 11.8, 11.9 or 12.0; and in particular 12.1, 12.2, 12.3, 12.4, 12.5, 12.6, 12.7, 12.8, 12.9, 13.0, 13.1, 13.2, 13.3, 13.4 or 13.5.

In a preferred embodiment, the total surface S of the pharmaceutical dosage form according to the invention satisfies the following requirement B W3 >S>A.W3 where A and W are defined as above and B is at most 20, more preferably at most 19, still more preferably at most 18, yet more preferably at most 17, most preferably at most 16 and in particular at most 15.

In a preferred embodiment, the total surface S of the pharmaceutical dosage form according to the invention is at least 50 mm2, at least 75 mm2, at least 100 mm2, at least 125 mm2, at least 150 mm2, at least 175 mm` or at least 200 mm2; more preferably at ieasi 225 mm2, at least 250 mm2, at least 275 mm2, at least 300 mm2, at least 325 mm2, at least 350 mm2, at least 375 mm2 or at least 400 mm2; still more preferably at least 425 mm2, at least 450 mm2, at least 475 mm2, at least 500 mm2, at least 525 mm2; at least 550 mm2, at least 575 mm2 or at least 600 mm2; yet more preferably at least 625 mm2, at least 650 mm2, at least 675 mm2, at least 700 mm2, at least 725 mm2, at least 750 mm2, at least 775 mm2 or at least 800 mm2; most preferably at least 825 mm2, at least 850 mm2, at least 875 mm2, at least 900 mm2, at least 925 mm2, at least 950 mm2, at least 975 mm2 or at least 1000 mm2; and in particular at least 1025 mm2, at least 1050 mm2, at least 1075 mm2, at least 1100 mm2, at least 1125 mm2, at least 1150 mm2, at least 1175 mm2 or at least 1200 mm2.
In a preferred embodiment, the total surface S of the pharmaceutical dosage form according to the invention is at most 1500 mm2, more preferably at most 1400 mm2, still more preferably at most 1300 mm2, yet more preferably at most 1200 mm2, most preferably at most 1100 mm2, and in particular at most 1000 mm2.

In a preferred embodiment the pharmaceutical dosage form according to the invention is manufactured, particularly shaped, by means of a so-called H-plunger. The silhouette of a dosage form obtainable by means of such a H-plunger is schematically illustrated in Figure 10. H-plungers of suitable size and shape are commercially available.
Typically, the volume and the surface of the dosage forms that are obtainable by a given H-plunger can be calculated with a formula usually provided by the manufacturer of the H-plunger.

For example, Notter GmbH, Germany offers a H-plunger forming a volume of 94.3 + 171.6 h [mm3] and a surface of 382 + 52.3 h [mm2], where h is the height of the dosage form (corresponding to distance b2 in Figure 10). Therefore, for example, when shaping 650 mg of a compacted composition having an overall density of 1.000 mg/mm3 with such H-plunger, a dosage form is obtained having a height of h = (650-94.3)/171.6 =
3.24 mm.
Thus, said dosage form has a surface of 382 + 52.3 , 3.24 = 551 mm2. When A =
4.5, the requirement of 551 mm2 >- 4.5 6502/3 (= 337.6 mm2) is satisfied. When A is about 7.3, the requirement of 551 mm2 ? 7.3 65023 (= 547 mm2) is still satisfied, but when A
is 7.4, the requirement 551 mm2 ? 7.4 65023 (= 555 mm2) is not satisfied.

In a preferred embodiment, the pharmaceutical dosage form according to the invention has a total weight W of at least 50 mg, at least 75 mg, at least 100 mg, at least 125 mg or at least 150 mg; more preferably at least 175 mg, at least 200 mg, at least 225 mg, at least 250 mg or at least 275 mg; still more preferably at least 300 mg, at ieast 325 mg, ai ieasi.
350 mg, at least 375 mg or at least 400 mg; yet more preferably at least 425 mg, at least 450 mg, at least 475 mg, at least 500 mg or at least 525 mg; most preferably at least 550 mg, at least 575 mg, at least 600 mg, at least 625 mg or at least 650 mg; and in particular at least 675 mg, at least 700 mg, at least 725 mg, at least 750 mg or at least 775 mg.
Preferably, the total weight of the pharmaceutical dosage form according to the invention is within the range from 0.01 g to 1.5 g, more preferably 0.05 g to 1.2 g, still more preferably 0.1 g to 1.0 g, most preferably 0.2 g to 0.9 g and in particular 0.25 g to 0.8 g.

Once the breaking strength of the pharmaceutical dosage form has been measured in a particular direction of extension, its tensile strength in said direction of extension may be calculated taking into account the outer shape of the dosage form. For example, the tensile strength of conventional tablets can be calculated according to the following equation: T S =
2 x P / rr x D x t, where TS is the tensile strength (N=cm"2, MPa), P is the breaking strength of the tablet (N), D is the tablet diameter (cm), and t is the tablet thickness (cm). The skilled person knows how to modify the above formula when modifying the outer shape of the dosage form. In this regard it can be referred to, e.g., J.T. Fell et al., J
Pharm Sci. 1970, 59, 688-691; M.P. Summers et al., J Pharm Sci., 1977, 66, 1172-1175; and P.N.
Davies et al., Eur J Pharm Biopharm. 2007, 67(1), 268-76.

Preferably, the pharmaceutical dosage form according to the invention has a tensile strength of at least 2.5 N/cm2, at least 3.0 N/cm2, at least 3.5 N/cm2, at least 4.0 N/cm2, at least 4.5 N/cm2, at least 5.0 N/cm2, at least 6.0 N/cm2, at least 7.5 N/cm2, at least 10.0 N/cm2, at least 12.5 N/cm2 or at least 15.0 N/cm2; more preferably at least 17.5 N/cm2, at least 20.0 N/cm2, at least 22.5 N/cm2 or at least 25.0 N/cm2; still more preferably at least 27.5 N/cm2, at least 30.0 N/cm2, at least 32.5 N/cm2 or at least 35.0 N/cm2;
yet more preferably at least 37.5 N/cm2, at least 40.0 N/cm2, at least 42.5 N/cm2 or at least 45.0 N/cm2; most preferably at least 47.5 N/cm2, at least 50.0 N/cm2, at least 52.5 N/cm2 or at least 55.0 N/cm2; and in particular at least 57.5 N/cm2, at least 60.0 N/cm2, at least 62.5 N/cm2 or at least 65.0 N/cm2; preferably at least in direction of extension E,.

The pharmaceutical dosage forms according to the invention preferably exhibit high impact strength.

For example, the falling impact strength of the pharmaceutical dosage forms is preferably about 0%. The falling impact strength is a breakage ratio obtained when a tablet is allowed to fall from the height of 50 cm onto a stainless steel plate and defined by:
{(broken tabiets)i(tesiea tabieis)1 100(%).

Preferably, the impact strength of the pharmaceutical dosage form according to the invention is sufficiently high so that it cannot be comminuted by means of a hammer.
Preferably, when applying five manual hammer strokes by means of a hammer having a weight of 500 g, the pharmaceutical dosage form cannot be comminuted. In a preferred embodiment, the pharmaceutical dosage form does not only exhibit this impact strength at ambient temperature, but also below +4 C (refrigerator), more preferably below (deep freezer), most preferably below -77 C (dry ice) and in particular below -190 C (liquid nitrogen).

Preferably, the pharmaceutical dosage form according to the invention exhibits a cutting resistance of at least 75 N, more preferably at least 100 N, still more preferably at least 125 N, yet more preferably at least 140 N, most preferably at least 150 N and in particular at least 160 N, in at least one direction of extension, preferably in direction of extension E,.
Preferably, the cutting test is performed according to DIN EN ISO 604, preferably at a testing speed of 30 mm/min and by means of a universal glass cleaning blade having a thickness of 0.30 mm.

The friability of the pharmaceutical dosage form according to the invention can be measured, e.g., by means of a Pharmatest PTF-E apparatus (Hainburg, Germany) following, e.g., the European Pharmacopeia (Ph. Eur.) specifications.
Preferably, the friability of the pharmaceutical dosage form according to the invention is at most 0.50%, more preferably at most 0.40%, still more preferably at most 0.30%, yet more preferably at most 0.20%, most preferably at most 0.10% and in particular at most 0.05%.

In a preferred embodiment the pharmaceutical dosage form according to the invention has an overall density of at least 0.80 or at least 0.85 g/cm3, more preferably at least 0.90 or at least 0.95 g/cm3, still more preferably at least 1.00, at least 1.05 or at least 1.10 g/cm3, most preferably in the range from 0.80 to 1.35 g/cm3, and in particular in the range from 0.95 to 1.25 g/cm3.

In a preferred embodiment, the pharmaceutical dosage form according to the invention has an overall density within the range of 1.00 0.30 g/cm3, more preferably 1.00 0.25 g/cm3, still more preferably 1.00 0.20 g/cm3, yet more preferably 1.00 0.15 g/cm3, most preferably 1.00 0.10 g/cm3, and in particular 1.00 0.05 g/cm3. In another preferred embodiment, the pharmaceutical dosage form according to the invention has an overall density within the range of 1.10 0.30 g/cm3, more preferably 1.10 0.25 gicm3, stiii more preferabiy 1.10+-0.20 g/cm3, yet more preferably 1.10 0.15 g/cm3, most preferably 1.10 0.10 g/cm3, and in particular 1.10 0.05 g/cm3. In still another preferred embodiment, the pharmaceutical dosage form according to the invention has an overall density within the range of 1.20 0.30 g/cm3, more preferably 1.20 0.25 g/cm3, still more preferably 1.20 0.20 g/cm3, yet more preferably 1.20 0.15 g/cm3, most preferably 1.20 0.10 g/cm3, and in particular 1.20 0.05 g/cm3. Preferably, the overall density of the pharmaceutical dosage form according to the invention is 1.00 0.02 g/cm3, 1.02 0.02 g/cm3, 1.04 0.02 g/cm3, 1.06 0.02 g/cm3, 1.08 0.02 g/cm3, 1.10 0.02 g/cm3, 1.12 0.02 g/cm3, 1.14 0.02 g/cm3, 1.16 0.02 g/cm3, 1.18 0.02 g/cm3, 1.20 0.02 g/cm3, 1.22 0.02 g/cm3, 1.24 0.02 g/cm3, 1.26 0.02 g/cm3, 1.28 0.02 g/cm3, 1.30 0.02 g/cm3, 1.32 0.02 g/cm3, 1.34 0.02 g/cm3, 1.36 0.02 g/cm3, 1.38 0.02 g/cm3, or 1.40 0.02 g/cm3.

Preferably, the pharmaceutical dosage form according to the invention is characterized by a comparatively homogeneous distribution of density. Preferably, the densities of two segments of the pharmaceutical dosage form having a volume of 1.0 mm3 each, deviate from one another by not more than 10%, more preferably not more than more than 7.5%, still more preferably not more than 5.0%, most preferably not more than 2.5%, and in particular not more than 1.0%. When the pharmaceutical dosage form is film coated, said two segments of the pharmaceutical dosage form having a volume of 1.0 mm3 each are preferably segments of the core, i.e. do not contain any coating material.

In a preferred embodiment, the pharmaceutical dosage form according to the invention is thermoformed, preferably by extrusion, although also other methods of thermoforming may be used in order to manufacture the dosage form according to the invention such as press-molding at elevated temperature.

The pharmaceutical dosage form according to the invention contains a pharmacologically active compound (A), for the purpose of the specification also referred to as "component (A)".

Preferred pharmacologically active compounds (A) have already been mentioned as pharmaceutically active ingredients and pharmaceutically active ingredients with potential for abuse in connection with the first and second aspects of the invention.

In a preferred embodiment, under ambient conditions, the solubility of component (A) in pure water is at least 1.0 g/L, more preferably at least 5.0 g/L, still more preferably at least g/L, yet more preferably at least 25 g/L, most preferably at least 50 giL and in particuiar at least 100 g/L.

In another preferred embodiment, under ambient conditions, the solubility of component (A) in pure water is at most 1.0 g/L, more preferably at most 0.5 g/L, still more preferably at most 0.1 g/L, yet more preferably at most 0.05 g/L, most preferably at most 0.01 g/L and in particular at most 0.005 g/L.

In a preferred embodiment, the pharmaceutical dosage form according to the invention contains a pharmaceutically effective amount of a pharmacologically active compound (A), which justifies use of the pharmaceutical dosage form as a pharmaceutical preparation and is the cause of the activity thereof. Pharmacologically active compounds (A) which may in principle be considered in the pharmaceutical dosage form according to the invention are any known pharmaceutical substances, wherein these substances may be present in the pharmaceutical dosage form according to the invention as such, in the form the derivatives thereof, in particular esters or ethers, or in each case in the form of corresponding physiologically acceptable compounds, in particular in the form of the corresponding salts or solvates thereof, as racemates or in a form enriched in one or more stereoisomers (enantiomers or diastereomers).

The pharmaceutical dosage form according to the invention is suitable for the administration of a number of pharmacologically active compounds (A) in a single pharmaceutical dosage form. Preferably, the pharmaceutical dosage form contains only one particular pharmacologically active compound (A).

The amount of the pharmacologically active compound (A), based on the total amount of the pharmaceutical dosage form, is preferably within the range from 0.01 to 95 wt.-%, more preferably from 0.5 to 80 wt.-%, still more preferably 1.0 to 70 wt.-%, most preferably 5.0 to 60 wt.-% and in particular 10 to 50 wt.-%. In a preferred embodiment it is more than 20 wt.-In a preferred embodiment the pharmaceutical dosage form according to the invention contains a psychotropically acting substance as the pharmacologically active compound (A).

The person skilled in the art knows which substances have a psychotropic action.
Substances which influence psychological processes commonly have a psychotropic action, i.e. they act specifically on psychological functions. Substances with a psychotropic action may thus influence mood, either raising or lowering it. For the purpose of the description, substances with a psychotropic action include in particular opioids, stimulants, tranquillisers (e.g. barbiturates and benzodiazepines) and other narcotics.
Substances with a psychotropic action preferably comprise substances which, in particular when improperly administered (in particular with the intention of abuse), cause an accelerated increase in active compound levels relative to proper oral administration, giving the abuser the desired effect, namely the "kick" or "rush". This kick is also obtained if the powdered pharmaceutical dosage form is administered nasally, i.e. is sniffed. Substances with a psychotropic action are preferably substances which (in the appropriate dose and pharmaceutical dosage form and when administered appropriately) influence human mental activity and/or sensory perception in such a way that they are fundamentally suited to abuse.

In particular, the pharmaceutical dosage form according to the invention preferably contains a psychotropically acting substance selected from the group consisting of opioids [A07DA, N01AH, N02A, R05DA, R05FA,]; barbiturates [N01AF, N01AG, N03AA];
benzodiazepine derivatives [N03AE]; agents for treating opiate dependency [NO7BC];
anxiolytics [N05B];
hypnotics and sedatives [N05C]; psychostimulants, agents for treating attention-deficit/
hyperactivity disorder (ADHD) and nootropics [N06B]; antiemetics [A04A];
antiobesity preparations excluding diet products [A08A]; centrally acting muscle relaxants [M03B]; and antidotes [V03AB]. The abbreviations stated in square brackets here correspond to the ATC
Index ("Gelbe Liste"), as used by the WHO for classifying pharmaceutical substances (preferred version: 2007 or 2008).

The pharmaceutical dosage form according to the invention preferably contains a psychotropically acting substance selected from the group consisting of opioids, vanilloid receptor modulators, serotonin/norepinephrine/dopamine modulators, GABA
modulators, NMDA antagonists, ion channel blockers/modulators, cannabionoids, and other NSAIDS.
The following opiates, opioids, tranquillisers or other narcotics are substances with a psychotropic action, i.e. have a potential of abuse, and hence are preferably contained in the pharmaceutical dosage form according to the invention: alfentanil, allobarbital, allylprodine, alphaprodine, alprazolam, amfepramone, amphetamine, amphetaminil, amobarbital, anileridine, apocodeine, axomadol, barbital, bemidone, benzylmorphine, bezitramide, bromazepam, brotizolam, buprenorphine, butobarbital, butorphanol, camazepam, carfentanil, cathine/D-norpseudoephedrine, chlordiazepoxide, clobazam clofedanol, clonazepam, clonitazene, clorazepate, clotiazepam, cloxazolam, cocaine, codeine, cyciobarbitai, cyciorphan, cypr a for pl III le, delor azepal I I, desoimor phii e, dextromoramide, dextropropoxyphene, dezocine, diampromide, diamorphone, diazepam, dihydrocodeine, dihydromorphine, dihydromorphone, dimenoxadol, dimephetamol, dimethylthiambutene, dioxaphetylbutyrate, dipipanone, dronabinol, eptazocine, estazolam, ethoheptazine, ethylmethylthiambutene, ethyl loflazepate, ethylmorphine, etonitazene, etorphine, fencamfamine, fenethylline, fenpipramide, fenproporex, fentanyl, fludiazepam, flunitrazepam, flurazepam, halazepam, haloxazolam, heroin, hydrocodone, hydromorphone, hydroxypethidine, isomethadone, hydroxymethylmorphinan, ketazolam, ketobemidone, levacetylmethadol (LAAM), levomethadone, levorphanol, levophenacyl-morphane, levoxemacin, lisdexamfetamine dimesylate, lofentanil, loprazolam, lorazepam, lormetazepam, mazindol, medazepam, mefenorex, meperidine, meprobamate, metapon, meptazinol, metazocine, methylmorphine, metamphetamine, methadone, methaqualone, 3-methylfentanyl, 4-methylfentanyl, methylphenidate, methylphenobarbital, methyprylon, metopon, midazolam, modafinil, morphine, myrophine, nabilone, nalbuphene, nalorphine, narceine, nicomorphine, nimetazepam, nitrazepam, nordazepam, norlevorphanol, normethadone, normorphine, norpipanone, opium, oxazepam, oxazolam, oxycodone, oxymorphone, Papaver somniferum, papaveretum, pernoline, pentazocine, pentobarbital, pethidine, phenadoxone, phenomorphane, phenazocine, phenoperidine, piminodine, pholcodeine, phenmetrazine, phenobarbital, phentermine, pinazepam, pipradrol, piritramide, prazepam, profadol, proheptazine, promedol, properidine, propoxyphene, remifentanil, secbutabarbital, secobarbital, sufentanil, tapentadol, temazepam, tetrazepam, tilidine (cis and trans), tramadol, triazolam, vinylbital, N-(1-methyl-2-piperidinoethyl)-N-(2-pyridyl)propionamide, (1R,2R)-3-(3-dimethylamino-1-ethyl-2-methyl-propyl)phenol, (1R,2R,-4S)-2-(dimethylamino)methyl-4-(p-fluorobenzyloxy)-1-(m-methoxyphenyl)cyclohexanol, (1 R,2R)-3-(2-dimethylaminomethyl-cyclohexyl)phenol, (1 S,2S)-3-(3-dimethylamino-1-ethyl-2-methyl -propyl)phenol, (2R,3R)-1-dimethylamino-3(3-methoxyphenyl)-2-methyl-pentan-3-ol, (1 RS,3RS,6RS)-6-dimethylaminomethyl-1 -(3-methoxyphenyl)-cyclohexane-1,3-diol, preferably as racemate, 3-(2-dimethylaminomethyl-1-hydroxy-cyclohexyl)phenyl 2-(4-isobutyl-phenyl)propionate, 3-(2-dimethylaminomethyl-1-hydroxy-cyclohexyl)phenyl 2-(6-methoxy-naphthalen-2-yl)propionate, 3-(2-dimethylaminomethyl-cyclohex-1-enyl)-phenyl 2-(4-isobutyl-phenyl)propionate, 3-(2-dimethylaminomethyl-cyclohex-1 -enyl)-phenyl 2-(6-methoxy-naphthalen-2-yl)propionate, (RR-SS)-2-acetoxy-4-trifluoromethyl-benzoic acid 3-(2-dimethylaminomethyl- 1-hydroxy-cyclohexyl)-phenyl ester, (RR-SS)-2-hydroxy-4-trifluoro-methyl-benzoic acid 3-(2-dimethylaminomethyl-1-hydroxy-cyclohexyl)-phenyl ester, (RR-SS)-4-chloro-2-hydroxy-benzoic acid 3-(2-dimethylaminomethyl-1-hydroxy-cyclohexyl)-phenyl ester, (RR-SS)-2-hydroxy-4-methyl-benzoic acid 3-(2-dimethylaminomethyl-hydroxy-cyclohexyl)-phenyl ester, (RR-SS)-2-hydroxy-4-methoxy-benzoic acid 3-(2-dimethylaminomethyl-1-hydroxy-cyclohexyl)-phenyl ester, (RR-SS)-2-hydroxy-5-niiru-benzoic acid 3-(2-dimethyl aminom ethyl- 1 -hyd roxy-cyclohexyl)-phenyl ester, (RR-SS)-2',4'-difluoro-3-hydroxy-biphenyl-4-carboxylic acid 3-(2-dimethylaminom ethyl- 1 -hyd roxy-cyclo-hexyl)-phenyl ester, and corresponding stereoisomeric compounds, in each case the corresponding derivatives thereof, physiologically acceptable enantiomers, stereoisomers, diastereomers and racemates and the physiologically acceptable derivatives thereof, e.g.
ethers, esters or amides, and in each case the physiologically acceptable compounds thereof, in particular the salts thereof and solvates, e.g. hydrochlorides.

In a preferred embodiment the pharmaceutical dosage form according to the invention contains an opioid selected from the group consisting of DPI-125, M6G (CE-04-410), ADL-5859, CR-665, NRP290 and sebacoyl dinalbuphine ester.

In a preferred embodiment the pharmaceutical dosage form according to the invention contains one pharmacologically active compound (A) or more pharmacologically active compounds (A) selected from the group consisting of oxymorphone, hydromorphone and morphine.

In another preferred embodiment, the pharmacologically active compound (A) is selected from the group consisting of tapentadol, faxeladol and axomadol.

For the purposes of the description, the pharmacokinetic parameters, which may be determined from the blood plasma concentrations of the pharmacologically active compound (A), are defined as follows:

Cmax maximum measured plasma concentration of the active ingredient after single administration average peak plasma leveo t_M interval of time from administration of the active ingredient until Cm, is reached t1/2 half-life AUCao total area under the curve The above parameters are in each case stated as mean values of the individual values for all investigated patients/test subjects.

A person skilled in the art knows how the pharmacokinetic parameters of the active ingredient may be calculated from the measured concentrations of the active ingredient in the blood plasma. In this connection, reference may be made, for example, to Willi Cawello (ed.) Parameters for Compartment-free Pharmacokinetics, Shaker Verlag Aachen (1999).

In a preferred embodiment, after preferably orai administration of the dosage form according to the invention, in vivo the average peak plasma level (Cmax) is on average reached after tmax 4.0 2.5 h, more preferably after tmax 4.0 2.0 h, still more preferably after tmax 4.0 1.5 h, most preferably after tmax 4.0 1.0 h and in particular after tmax 4.0 0.5 h. In another preferred embodiment, after preferably oral administration of the dosage form according to the invention, in vivo the average peak plasma level (Cmax) is on average reached after tmax 5.0 2.5 h, more preferably after tmax 5.0 2.0 h, still more preferably after tmax 5.0 1.5 h, most preferably after tmax 5.0 1.0 h and in particular after tmax 5.0 0.5 h. In still another preferred embodiment, after preferably oral administration of the dosage form according to the invention, in vivo the average peak plasma level (Cmax) is on average reached after tmax 6.0 2.5 h, more preferably after tmax 6.0 2.0 h, still more preferably after tmax 6.0 1.5 h, most preferably after tmax 6.0 1.0 h and in particular after tmax 6.0 0.5 h.

In a preferred embodiment, the average value for t12 after preferably oral administration of the dosage form according to the invention in vivo is 4.3 2.5 h, more preferably 4.3 2.0 h, still more preferably 4.3 1.5 h, most preferably 4.3 1.0 h, and in particular 4.3 0.5 h. In another preferred embodiment, the average value for t112 after preferably oral administration of the dosage form according to the invention in vivo is preferably 5.3 2.5 h, more preferably 5.3 2.0 h, still more preferably 5.3 1.5 h, most preferably 5.3 1.0 h, and in particular 5.3 0.5 h. In still another preferred embodiment, the average value for t12 after preferably oral administration of the dosage form according to the invention in vivo is preferably 6.3 2.5 h, more preferably 6.3 2.0 h, still more preferably 6.3 1.5 h, most preferably 6.3 1.0 h, and in particular 6.3 0.5 h.

In a preferred embodiment, the pharmacologically active compound (A) is tapentadol or a physiologically acceptable salt thereof, and after preferably oral administration of the dosage form according to the invention, in vivo the average value for the total area under the curve AUC0_b is 825 600 ng=h/mL, more preferably 825 500 ng=h/mL, still more preferably 825 400 ng=h/mL, yet more preferably 825 300 ng=h/mL, most preferably 825 200 ng=h/mL, and in particular 825 100 ng=h/mL. In another preferred embodiment, the pharmacologically active compound (A) is tapentadol or a physiologically acceptable salt thereof, and after preferably oral administration of the dosage form according to the invention, in vivo the average value for the total area under the curve AUC0-is 1100 600 ng=h/mL, more preferably 1100 500 ng=h/mL, still more preferably 1100 400 ng=h/mL, yet more preferably 1100 300 ng=h/mL, most preferably 1100 200 ng=h/mL, and in particular 1100 100 ng=h/mL.

In a preferred embodiment, the pharmacologically active compound (A) is tapentadoi or a physiologically acceptable salt thereof, and after preferably oral administration of the dosage form according to the invention, in vivo the average value of Cmax is 63 40 ng/mL, more preferably 63 30 ng/mL, still more preferably 63 20 ng/mL, yet more preferably 63 15 ng/mL, most perefrably 63 10 ng/mL and in particular 63 5 ng/mL. In another preferred embodiment, the pharmacologically active compound (A) is tapentadol or a physiologically acceptable salt thereof, and after preferably oral administration of the dosage form according to the invention, in vivo the average value of Cmax is 89 40 ng/mL, more preferably 89 30 ng/mL, still more preferably 89 20 ng/mL, yet more preferably 89 15 ng/mL, most perefrably 89 10 ng/mL and in particular 89 5 ng/mL.

In a particularly preferred embodiment the pharmacologically active compound (A) is tapentadol or a physiologically acceptable salt thereof and the pharmaceutical dosage form according to the invention is bioequivalent to a formulation that contains tapentadol or a physiologically acceptable salt thereof in a dosage of 200 mg and 250 mg, respectively, and is characterized by the following pharmacokinetic data:

Parameter dosage 200 dosage 250 mg mg AUCao 825 ng=h/mL 1096 ng=h/mL
Cmax 62.5 ng/mL 89.3 ng/mL
tmax 5.00 h 5.00 h tii2 5.2 h 5.4 h The skilled person is aware what requirements have to be satisfied in order to achieve bioequivalence. In this regard it can be referred e.g. to "Note for Guidance on the Investigation of Bioavailability and Bioequivalence", EMEA, London, 26 July (CPMP/EWP/QWP/1401/98); "Guidance for Industry - Bioavailability and Bioequivalence -Studies for Orally Administered Drug Products - General Considerations", FDA, BP, Announced in the Federal Register: Volume 68, Number 53/March 19, 2003; and "Guidance for Industry - Statistical Approaches to Establishing Bioequivalence", FDA, BP, January 2001.

In general, two medicinal products are bioequivalent if they are pharmaceutically equivalent or pharmaceutical alternatives and if their bioavailabilities after administration in the same molar dose are similar to such degree that their effects, with respect to both efficacy and safety, will be essentially the same. Preferably, statistical data should be analyzed using ANOVA based on a 90% confidence interval. For example, as regards AUC-ratio, the 90%
confidence interval for this measure of relative bioavailability should lie within an acceptance interval of 0.80-1.25, and as regards Cmax-ratio, the 90%
confidence interval for this measure of relative bioavailability should lie within an acceptance interval of 0.80-1.25.

In a preferred embodiment the pharmaceutical dosage form according to the invention contains one pharmacologically active compound (A) or more pharmacologically active compounds (A) selected from the group consisting of 1,1-(3-dimethylamino-3-phenyl-pentamethylen)-6-fluor-1,3,4,9-tetrahydropyrano[3,4-b]indole, in particular its hemicitrate;
1,1-[3-dimethylamino-3-(2-thienyl)pentamethylen]-1,3,4,9-tetrahydropyrano[3,4-b]indole, in particular its citrate; and 1,1-[3-dimethylamino-3-(2-thienyl)pentamethylen]-1,3,4,9-tetrahydropyrano[3,4-b]-6-fluoro-indole, in particular its hemicitrate. These compounds are known, for example, from WO 2004/043967 or WO 2005/066183. The corresponding descriptions are hereby introduced as a reference and are deemed to be part of the disclosure.

In a preferred embodiment, the pharmaceutical dosage form according to the invention contains no substances which irritate the nasal passages and/or pharynx, i.e.
substances which, when administered via the nasal passages and/or pharynx, bring about a physical reaction which is either so unpleasant for the patient that he/she does not wish to or cannot continue administration, for example burning, or physiologically counteracts taking of the corresponding active compound, for example due to increased nasal secretion or sneezing.
Further examples of substances which irritate the nasal passages and/or pharynx are those which cause burning, itching, an urge to sneeze, increased formation of secretions or a combination of at least two of these stimuli. Corresponding substances and the quantities thereof which are conventionally to be used are known to the person skilled in the art.
Some of the substances which irritate the nasal passages and/or pharynx are accordingly based on one or more constituents or one or more plant parts of a hot substance drug.
Corresponding hot substance drugs are known per se to the person skilled in the art and are described, for example, in "Pharmazeutische Biologie - Drogen and ihre Inhaltsstoffe"
by Prof. Dr. Hildebert Wagner, 2nd., revised edition, Gustav Fischer Verlag, Stuttgart-New York, 1982, pages 82 et seq.. The corresponding description is hereby introduced as a reference and is deemed to be part of the disclosure.

The pharmaceutical dosage form according to the invention furthermore preferably contains no antagonists for the pharmacologically active compound (A), preferably no antagonists against psychotropic substances, in particular no antagonists against opioids.
Antagonists suitable for a given pharmacologically active compound (A) are known to the person skilled in the art and may be present as such or in the form of corresponding derivatives, in particular esters or ethers, or in each case in the form of corresponding physiologically acceptable compounds, in particular in the form of the salts or solvates thereof. The pharmaceutical dosage form according to the invention preferably contains no antagonists selected from among the group comprising naloxone, naltrexone, nalmefene, nalide, nalmexone, nalorphine or naluphine, in each case optionally in the form of a corresponding physiologically acceptable compound, in particular in the form of a base, a salt or solvate;
and no neuroleptics, for example a compound selected from among the group comprising haloperidol, promethacine, fluphenazine, perphenazine, levomepromazine, thioridazine, perazine, chlorpromazine, chlorprothixine, zuclopenthixol, flupentixol, prothipendyl, zotepine, benperidol, pipamperone, melperone and bromperidol.

The pharmaceutical dosage form according to the invention furthermore preferably contains no emetic. Emetics are known to the person skilled in the art and may be present as such or in the form of corresponding derivatives, in particular esters or ethers, or in each case in the form of corresponding physiologically acceptable compounds, in particular in the form of the salts or solvates thereof. The pharmaceutical dosage form according to the invention preferably contains no emetic based on one or more constituents of ipecacuanha (ipecac) root, for example based on the constituent emetine, as are, for example, described in "Pharmazeutische Biologie - Drogen and ihre Inhaltsstoffe" by Prof. Dr.
Hildebert Wagner, 2nd, revised edition, Gustav Fischer Verlag, Stuttgart, New York, 1982. The corresponding literature description is hereby introduced as a reference and is deemed to be part of the disclosure. The pharmaceutical dosage form according to the invention preferably also contains no apomorphine as an emetic.

Finally, the pharmaceutical dosage form according to the invention preferably also contains no bitter substance. Bitter substances and the quantities effective for use may be found in US-2003/0064099 Al, the corresponding disclosure of which should be deemed to be the disclosure of the present application and is hereby introduced as a reference.
Examples of bitter substances are aromatic oils, such as peppermint oil, eucalyptus oil, bitter almond oil, menthol, fruit aroma substances, aroma substances from lemons, oranges, limes, grapefruit or mixtures thereof, and/or denatonium benzoate.

The pharmaceutical dosage form according to the invention accordingly preferably contains neither substances which irritate the nasal passages and/or pharynx, nor antagonists for the pharmacologically active compound (A), nor emetics, nor bitter substances.

In a preferred embodiment the pharmaceutical dosage form according to the invention contains a non-psychotropically acting substance as the pharmacologically active compound (A).

Particularly preferably the pharmaceutical dosage form according to the invention contains a pharmacologically active compound (A) or two or more such compounds selected from the group consisting of - agents for the treatment and prevention of diseases of the alimentary system and metabolism [A]; in particular stomatological preparations [A01], agents for the treatment and prevention of acid-related disorders [A02], agents for the treatment and prevention of functional gastrointestinal tract disorders [A03], serotonin 5HT3 antagonists [A04 a)A], antihistamine preparations [A04 a)B], agents for bile and liver therapy [A05], laxatives [A06], intestinal antiinfectives [A07A], intestinal adsorbents [A07B], electrolytes with carbohydrates [A07C], intestinal anti inflammatory agents [A07E], microbial antidiarrhoeals [A07F], digestives including enzymes [A09], drugs used in diabetes [A10], vitamins [All], minerals [A12], anabolic agents for systemic applications [A14] and appetite stimulants [A15];

- agents for the treatment and prevention of diseases of the blood and the blood forming organs [B]; in particular antithrombotic agents [B01], antihaemorrhagics [B02], antianaemic preparations [B03] and other haematological agents [B06];

- agents for the treatment and prevention of diseases of the cardiovascular system [C];
in particular agents for cardiac therapy [COl], antihypertensives [C02], diuretics [C03], peripheral vasodilatators [C04], vasoprotectives [C05], antihypotensives [C06A], R-adrenoceptor antagonists [C07], calcium channel blockers [C08], agents acting on the renin-angiotensin system [C09] and lipid reducing agents [C10];

- dermatologicals [D]; in particular antifungals for systemic use [D01 B], antipsoriatics for systemic use [D05B], antiacne preparations for systemic use [D1 0B];

agents for the treatment and prevention of diseases of the genitourinary system and sex hormones [G]; in particular gynaecological antiinfectives and antiseptics [G01], oxytocics [G02A], sympathomimetic labour repressants [G02CA], prolactin inhibitors [G02CB], hormonal contraceptives for systemic use [G03] and uruiogicals [G04];

systemic hormone preparations excluding sex hormones and insulins [H]; in particular pituitary and hypothalamic hormones and analogue [H01], corticosteroids for systemic use [H02], thyroid preparations [H03], pancreatic hormones [H04], and agents for regulating calcium homeostatis [H05];

antiinfectives for systemic use [J]; in particular antibiotics for systemic use [J01], antimycotics for systemic use [J02], antimycobacterials [J04], antivirals for systemic use [J05], immune sera and immunoglobulins [J06], and vaccines [J07]);

antineoplastic and immunomodulating agents [L] (in particular antineoplastistic agents [L01], agents for endocrine therapy [L02], immunostimulants [L03] and immunosuppressive agents [L04];

agents for the treatment and prevention of diseases of the musculo-skeletal system [M]; in particular antiinflammatory and antirheumatic agents [M01], peripherally acting muscle relaxants [M03A], directly acting muscle relaxants [M03C], antigout preparations [M04] and agents for the treatment of bone diseases [M05];

agents for the treatment and prevention of diseases of the nervous system [N];
in particular salicylic acid the derivatives thereof [N02 b)A], pyrazolones [N02 b)B], anilides [N02 b)E], ergot alkaloids [N02CA], corticosteroid derivatives [NO2CB], selective serotonin-5HT, agonists [NO2CC], hydantoin derivatives [N03 a)B], oxazolidine derivatives [N03 a)C], succinimide derivatives [N03 a)D], carboxamide derivatives [N03 a)F], fatty acid derivatives [N03 a)G], antiparkinson drugs [N04]), antipsychotics [N05A], antidepressants [N06A], antidementia drugs [N06D], parasympathomimetics [N07A] and antivertigo preparations [N07C];

antiparasitic products, insecticides and repellents [P]; in particular antiprotozoals [P01], anthelmintics [P02] and ectoparasiticides, including scabicides, insecticides and repellents [P03];

agents for the treatment and prevention of diseases of the respiratory system [R]; in particular nasal preparations [R01], throat preparations [R02], drugs for obstructive airways diseases [R03], expectorants, excluding combinations with cough suppressants [R05Cj and antihistamines for systemic use [R06];

agents for the treatment and prevention of diseases of the sensory organs [S];
in particular otologicals [S02]; and general diet products [V06] and therapeutic radiopharmaceuticals [V10], wherein the abbreviations stated in square brackets here correspond to the ATC
Index, as used by the WHO for classifying pharmaceutical substances (preferred version:
2007 or 2008).

The pharmaceutical dosage form according to the invention preferably contains one, two or more pharmacologically active compounds (A) selected from the group consisting of 4-aminomethylbenzoic acid, abacavir, abamectin, abciximab, abibendan, abrin, acamprosat, acarbose, acebutolol, aceclidine, aceclofenac, acediasulfone, acemetacin, acenocoumarol, acetazolamide, acetoacetic acid, acetyldigoxin, acetylandromedol, acetylcysteine, (3-acetyldigoxin, acetylhistamine, acetylsalicylic acid, acetylthiocholine, aciclovir, acipimox, acitretin, aclarubicin, aconitine, acriflavinium chloride, acrivastine, actinoquinol, acylaminopenicillin, adalimumab, adapalene, adefovir, adefovir dipivoxil, adenosine, adenosine phosphate, adenosine triphosphate, adipiodone, adrenalin, aescin, agalsidase alfa, agalsidase beta, agaricic acid, ajmaline, alanine, albendazole, alcuronium, aldesleukin, aldosterone, alemtuzumab, alendronic acid, alfacalcidol, alfuzosin, algeldrate F, alitretinoin, alizapride, allantoin F, allopurinol, allyl isorhodanate, almasilate F, almotriptan, a-acetyldigoxin, alprenolol, aiprostadil, alteplase, aluminium glycinate F, aluminium hydroxide F, aluminium phosphate F, aluminium triformate, amantadine, ambazone, ambroxol, ambutonium bromide, formic acid, amicacin, amidephrine, amidotrizoic acid, amifostine, amikacin, amiloride, aminoacetic acid, aminoglutethimide, aminophylline, aminoquinuride, amiodarone, amisulpride, amitriptyline, amitryptiline, amlodipine, amorolfine, amoxicillin, amphotericin B, ampicillin, amprenavir, amylmetacresol, amyl nitrite, anagrelide, anakinra, anastrozole, ancrod, anistreplase, antazoline, antithrombin III, apomorphine, apraclonidine, aprepitant, aprindine, aprotinin, arcitumomab, arginine, aripiprazole, arsenic trioxide, artemether, articaine, ascorbic acid, asparagine, L-asparaginase, aspartic acid, atazanavir, atenolol, atomoxetine, atorvastatin, atosiban, atovaquone, atracurium, atracurium besylate, atropine, auranofin, azapropazone, azathioprine, azelaic acid, azelastine, azidothymidine, azithromycin, azlocillin, aztreonam, N2 alanyl levoglutamide, p-aminosalicylic acid, bacampicillin, bacitracin, bacloten, baisaiazide, bambuteroi, bameihan, bamipine, barbexaclone, barium sulfate F, barnidipine, basiliximab, batroxobin, becaplermin, beclomethasone, bendamustine, befunolol, bemiparin, benactyzine, benazepril, bencyclane, bendazac, bendroflumethiazide, benproperine, benserazide, benzaseride, benzathine, benzatropine, benzbromarone, benzocaine, benzoyl peroxide, benzyclane, benzydamine, benzylpenicillin, benzylphenyl glycolate, betacarotene, betahistidine, betahistine, betamethasone, bethanechol, betaxolol, bethanechol chloride, betiatide, bevacizumab, bexarotene, bezafibrate, bibenzonium bromide, bicalutamide, bicisate, bifonazole, bimatoprost, biperiden, bisoprolol, bivalirudin, bleomycin, blood clotting factor VII, VIII, IX, X, XIII, bornapine, bornaprine, bortezomib, bosentan, botulinum toxin type B, brimonidine, brinzolamide, brivudin, bromhexine, bromocriptine, bromperidol, brompheniramine, brotizolam, budesonide, budipine, bufexamac, buflomedil, bumetanide, bunazosin, buphenine, bupivacaine, bupranolol, bupropion, buserelin, buspirone, busulfan, butalamine, butanilicaine, butenafine, butethamate, butinoline, butizide, butylscopolaminium,
5-chlorcarvacrol, C1 esterase inhibitor, cabergoline, cadexomer iodine, cafedrine, calcipotriol, calcitonin, calcitriol, camylofine, candesartan cilexetil, canrenoic acid, capecitabine, capreomycin, capsaicin, captopril, carazolol, carbaldrate F, carbamazepine, carbasalate calcium, carbenoxolone, carbidopa, carbimazole, carbinoxamine, carboplatin, carglumic acid, carmustine, caroverine, carteolol, carvedilol, caspofungin, cefaclor, cefadroxil, cefalexin, cefaloridine, cefamandole, cefazolin, cefdinir, cefepime, cefetamet-pivotil, cefixime, cefodizime, cefoperazone, cefotaxime, cefotiam, cefoxitin, cefpirome, cefpodoxime, cefpodoxime-proxetil, cefprozil, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefuroxime, celecoxib, celiprolol, certoparin, cetirizine, cetrimide, cetrimonium bromide, cetrorelix, cetuximab, cetylpyridinium, chenodeoxycholic acid, quinidine, quinine, quinine iron citrate F, quinine tannate F, chlorambucil, chloramphenicol, chlorobutynol, chlorhexidine, chlormidazole, chlorobutanol, chloroquine, chloroxylenol, chlorphenamine, chlorphenesin, chlorphenoxamine, chlorpromazine, chlorprotheaxine, chlorprothixine, chlortalidone, chlortetracycline, chlorzoxazone, choline, chondroitin sulfate, choriogonadotropin alfa, chorionic gonadotropin, chrysarobin, chymotrypsin, ciclesonide, cicletanine, ciclopirox, ciclosporin, cidofovir, cilastatin, cilazapril, cimetidine, cinacalcet, cinchocaine, cinnarizine, cinolazepam, ciprofloxacin, cisapride, cisatracurium besylate, cisplatin, citalopram, citicoline, cladribine, clarithromycin, clavulanic acid, clemastine, clenbuterol, clindamycin, clioquinol, clobetasol, clobetasone, clobutinol, clocortolone, clodronic acid, Clofibrate, clomifene, clomipramine, clonazepam, clonidine, clopamide, clopidogrel, clostebol acetate, clostridium botulinum, clotrimazole, cloxiquine, clozapine, cocarboxylase, colchicine, colecalciferol, coleseveiam, coiestipoi, coiestyrai-nine, colfosceril palmitate, colistin, zinc eyewash F, corticorelin, corticotrophin, cortisone, cresol, croconazole, cromoglicic acid, crotamiton, cryofluorane, coumarin, cyanamide, cyanocobalamin, cyclizine, cyclobutyrol, cyclopentolate, cyclophosphamide, cycloserine, cyproheptadine, cyproterone, cysteine, cytarabine, cytarabine, 2,4-dichlorobenzyl alcohol, 2-diethylaminoethanol, dacarbazine, daclizumab, dactinomycin, dalfopristin, dalteparin, danaparoid, danazol, dantrolene, dapiprazole, dapsone, darbepoetin alfa, darifenacin, Daunorubicin, deanol, deanolace, decarbazine, dectaflur F, deferiprone, deferoxamine, delapril, demeclocycline, denaverine, depreotide, dequalinium, desflurane, desipramine, desirudin, deslanoside, desloratadine, desmeninol, desmopressin, desogestrel, desoximetasone, deoxyribonuclease, detajmium, dexamethasone, dexchlorpheniramine, dexibuprofen, dexketoprofen, dexrazoxane, dextran, dextromethorphan, diacerein, diacetyl morphine, dibenzepin, dibotermin alfa, diclofenac, diclofenamide, didanosine, dienestrol, dienogest, diethylstilbestrol, difloxacin, diflucortolone, diflunisal, digitoxin, digoxin, dihydralazine, dihydroergocornine, dihydroergocristine, dihydroergocryptine, dihydroergotamine, dihydroergotoxine, dihydrotachysterol, diisopropylamine, dipotassium clorazepate, diltiazem, dimenhydrinate, dimepranol, dimercaprol, dimethyl sulfoxide, dimethindene, disodium selenite, dinoprost, dinoprostone, diosmin, diphenhydramine, diphenoxylate, diphenylpyraline, dipivefrine, diprophylline, dipyridamole, disopyramide, dinitrogen monoxide, distigmine, disulfiram, dithranol, dixyrazine, D-norpseudoephedrine, dobesilate calcium, dobutamine, docetaxel, dofetilide, dolasetron, domperidone, donepezil, dopamine, dopexamine, dornase alfa, dorzolamide, dosulepin, doxapram, doxazosin, doxepin, doxorubicin, doxycycline, doxylamine, drofenine, droperidol, drospirenone, drotrecogin alfa, duloxetine, dutasteride, dydrogesterone, N,N'-dihydroxymethyl urea, ebastine, econazole, ecothiopate iodide, efalizumab, efavirenz, eflornithine, iron(Ill) ammonium citrate F, superparamagnetic iron oxide, elcatonin, eletriptan, emedastine, emepronium, emepronium carrageenate, emetine, emtricitabine, enalapril, enalaprilat, enflurane, enfuvirtide, enoxacin, enoxaparin, entacapone, ephedrine, ephedrine racephedrine, epinastine, epinephrine, epirubicin, eplerenone, epoetin alfa, epoetin beta, epoetin delta, epoprostenol, eprazinone, eprosartan, eptacog alfa, eptifibatide, eptotermin alfa, erdosteine, ergocalciferol, ergometrine, ergotamide, ertapenem, erythromycin, escitalopram, esmolol, esomeprazole, estradiol, estramustine, estriol, estrone, etacrynic acid, etamivan, etanercept, ethacridine, ethambutol, ethaverine, ethinylestradiol, ethisterone, ethosuximide, etidronic acid, etilefrine, etodolac, etofenamate, etofibrate, etofylline, etomidate, etonogestrel, etoposide, etoricoxib, everoiimus, exametazimmme, exemestane, ezetimibe, 3-fluorotyrosine, famciclovir, famotidine, felbamate, felbinac, felodipine, fenbufene, fendiline, fenofibrate, fenoterol, fenticonazole, fexofenadine, fibrinogen, fibrinolysin, filgrastim, finasteride, flavoxate, flecainide, flucloxacillin, fluconazole, fludarabine, fludeoxyglucose [18F], fludrocortisone, flufenamic acid, flumazenil, flumetasone, flunarizine, flunisolide, fluocinolone acetonide, fluocinonide, fluocortolone, fluophenozine, fluorescein dilaurate, fluorescein sodium, fluorometholone, fluorouracil, fluorophosphoric acid, fluorosilane, fluoxetil, fluoxetine, flupentixol, fluphenazine, flupirtine, fluprednidene, flurbiprofen, flutamide, fluticasone, flutrimazole, fluvastatin, fluvoxamine, folic acid, follitropin alfa, follitropin beta, folic acid, fomepizole, fomivirsen, fondaparinux, formestane, formoterol, fosamprenavir, foscarnet, fosfestrol, fosfomycin, fosinopril, fosphenytoin, fotemustine, framycetin, framycetin, frovatriptan, fulvestrant, furosemide, fusafungine, fusidic acid, fytic acid, gabapentin, gadobenic acid, gadobutrol, gadodiamide, gadopentetic acid, gadoteridol, gadoteric acid, gadoteric acid-meglumine, gadoxetic acid, galantamine, gallopamil, ganciclovir, ganirelix, gatifloxacin, gemcitabine, gemfibrozil, gentamicin, gepefrine, gestodene, glatiramer, glibenclamide, glibornuride, gliclazide, glimepiride, glipizide, gliquidone, glisoxepide, glucagon, glutamine, glutamic acid, glycopyrronium, glycopyrronium bromide, glycyrrhetinic acid, gonadorelin, goserelin, gramicidin, granisetron, grepafloxacin, griseofulvin, g-strophanthin, guajacol, guanethidine, guanfacine, 13C urea, 4-hydroxybutyric acid, halcinonide, halofantrine, halometasone, haloperidol, halothane, haem, haematoporphyrin, heparin, hepatitis B vaccine, heptaminol, hexobarbital, hexobendine, hexoprenaline, histamine, histidine, homatropine, homofenazine, human albumin, hyaluronidase, hydralazine, hydrastinine, hydroquinone, hydrochlorothiazide, hydrocortisone, hydrotalcite F, hydroxocobalamin, hydroxycarbamide, hydroxychloroquine, hydroxycine, hydroxylamine, hydroxyprogesterone, hydroxyzine, hymecromone, ibandronic acid, ibopamine, ibritumomab tiuxetan, ibuprofen, ibutilide, idarubicin, ifosfamide, iloprost, imatinib, imatinib mesylate, imidapril, imiglucerase, imipenem, imipramine, imiquimod, immunocyanin, indanazoline, indapamide, indinavir, indium chloride ["'In], indobufen, indometacin, indoramin, infliximab, inosine, insulin, insulin aspart, insulin detemir, insulin glargine, insulin glulisine, insulin lispro, interferon alfa, interferon alfa-2 b), interferon alfacon-1, interferon beta, interferon beta-1 a), interferon beta-1 b), interferon L:l.: J-1 J:.-- J-J:..-._-1 n..._-.-- r123111 ,_____1 ganni-na, IUUILIIUUI, IUUIIIC, IUUCIIIIIU IUUIXCIIIUI, IUIIU~.JC111C j, IUIICXUI, IUIIIC~JIUI, IUI,Jc11111UUI, iopentol, iopromide, iosarcol, iotrolan, iotroxic acid, ioversol, ioxaglic acid, ioxitalamic acid, ipatropium, irbesartan, irinotecan, irinotecan, isepamicin, isoaminile, isoconazole, isoflurane, isoleucine, isoniazid, isonicotinic acid, isoprenaline, isosorbide, isospaglumic acid, isotretinoin, isoxsuprine, isradipine, itraconazole, josamycin, potassium permanganate, kallidinogenase, kanamycin, kawain, kebuzone, ketamine, ketoconazole, ketoprofen, ketorolac, ketotifen, collagenase, creosote, labetalol, lacidipine, lactitol, lamivudine, lamotrigine, lanreotide, lansoprazole, laronidase, latanoprost, leflunomide, lenograstim, lepirudin, lercanidipine, letrozole, leucine, leuprorelin, levallorphan, levamisole, levetiracetam, levobunolol, levobupivacaine, levocabastine, levocetirizine, levodopa, levofloxacin, levofolinate calcium, levomepromazine, levomethadyl, levonorgestrel, levopropylhexedrine, levosimendan, levothyroxine, lidocaine, lincomycin, lindane, linezolid, liothyronine, lisinopril, lisuride, lobeline, lodoxamide, lofepramine, lomefloxacin, lomustine, lonazolac, loperamide, lopinavir, loratadine, lorazepam oxide, lornoxicam, losartan, loteprednole, lovastatin, lumefantrine, lutropin alfa, lymecycline, lynestrenol, lypressin, lysine, magaldrate F, magnesium pidolate, magnesium L-aspartate, mangafodipir, manidipine, maprotiline, mebendazole, mebeverine, meclofenoxate, mecloxamine, meclozine, medrogestone, medroxyprogesterone, mefenamic acid, mefloquine, megestrol, melagatrane, melitracen, melperol, melperone, melphalan, memantine, menadione, mepacrine, mepartricin, mephenytoin, mepindolol, mepivacaine, mepyramine, mequinol, mercaptamine, mercaptopurine, meropenem, mesalazine, mesna, mesterolone, mesuximide, metaclazepam, metamizole, metamphetamine, metenolone, metenolone acetate, metformin, methanthelinium, methazolamide, methenamine, methionine, methohexital, methotrexate, 5-methoxypsoralen, 8-methoxypsoralen, methyl 5-aminolevulinate, methylbenactyzium bromide, methyldopa, methylergometrine, methylprednisolone, methylrosanilinium, methyltestosterone, methylthionium chloride, methysergide, metildigoxin, metipranolol, metoclopramide, metoprolol, methixene, metronidazole, mexiletine, mezlocillin, mianserine, miconazole, midodrine, mifepristone, miglitol, miglustat, milnacipran, milrinone, miltefosine, minocycline, minoxidil, mirtazapine, misoprostol, mitobronitol, mitomycin, mitotane, mitoxantrone, mivacurium chloride, mivacuronium, mizolastine, moclobemide, moexipril, molgramostim, molsidomine, mometasone, monochloroacetic acid, monteiukast, moroctocog aifa, moxaverirle, moxifloxacin, moxonidine, mupirocin, mycophenolate mofetil, nadifloxacin, nadrolon decanonate, nadroparin calcium, naftidrofuryl, naftifine, nalbuphine, nalide, nalmefene, nalmexone, naloxone, naltrexone, naluphine, naphazoline, 2-naphthol, naproxen, naratriptan, naratriptan, nateglinide, sodium aurothiomalate, sodium phenylbutyrate, sodium fluoride, sodium hyaluronate, sodium iodide ['31 1], sodium molybdate [99Mo], sodium phenylbutyrate, n-butyl-p-aminobenzoate, N-butylscopolaminium bromide, nebivolol, nedocromil, nefazodone, nefopam, nelfinavir, neomycin, neostigmine, neostigmine methylsulfate, netilmicin, nevirapine, n-heptyl-2-phenyl glycinate, nicardipine, nicergoline, nicethamide, niclosamine, nicoboxil, nicorandil, nicotine, nicotine aldehyde, nicotinamide, nicotine resinate, nicotinic acid, nicotinic acid ester, nicotinyl alcohol, nifedipine, niflumic acid, nifuratel, nilvadipine, nimesulide, nimodipine, nimorazole, nimustine, nisoldipine, nitisinone, nitrendipine, nitric oxide, nitrofurantoin, nitroglycerine, nizatidine, N-methylephedrine, nonacog alfa, nonivamide, noradrenalin, norelgestromin, norepinephrine, norethisterone, norfenefrine, norfloxacin, norgestimate, norgestrel, nortriptyline, noscapine, nystatin, obidoxime chloride, octafluoropropane, octocog alfa, octodrine, octreotide, odansetron, ofloxacin, olaflur F, olanzapine, olmesartan medoxomil, olopatadine, olsalazine, omeprazole, omoconazole, ondansetron, opipramol, oral cholera vaccine, orciprenaline, orlistat, ornipressin, orphenadrine, oseltamivir, osteogenic protein-1: BMP-7, oxaprozin, oxatomide, oxcarbazepine, oxedrine tartrate, oxetacaine, oxiconazole, oxilofrine, oxitropium, 2-oxo-3-methylbutyric acid, 2-oxo-3-methylvaleric acid, 2-oxo-3-phenylpropionic acid, 2-oxo-4-methylvaleric acid, oxprenolol, oxybuprocaine, oxybuprocaine, oxybutynin, oxybutynin, oxyfedrine, oxymetazoline, oxytetracycline, oxytocin, paclitaxel, palinavir, palivizumab, palonosetrone, pamidronic acid, pancuronium, pantoprazole, papaverine, paracetamol, paraldehyde, parecoxib, paricalcitol, parnaparin, paromomycin, paroxetine, pefloxacin, pegfilgrastim, peginterferon alfa, pegvisomant, pemetrexed, penbutolol, penciclovir, penfluridol, penicillamine, benperidol, pentaerithrityl tetranitrate, pentamidine, pentetrazol, pentetreotide, pentosan polysulfate sodium, pentoxifylline, pentoxyverine, perazine, perchloric acid, perflenapent, perflisopent, perflutren, pergolide, perindopril, perphenazine, phenacetin, phenamazid, phenazone, phenazopyridine, pheniramine, phenol, phenolphthalein, phenoxybenzamine, phenoxymethylpenicillin, phenprocoumon, phentolamine, phenylalanine, phenylbutazone, phenylephrine, phenyipropanolamine, phenyltoloxamine, phenytoin, phloroglucinol, pholedrine, phthalylsultathiazole, physostigmine, phytomenadione, phytosteroi, picric acid, pilocarpine, pimecrolimus, pimozide, pinaverium bromide, pindolol, pioglitazone, pipamperone, pipazetate, pipecuronium bromide, pipemidic acid, pipenzolate, piperacillin, piprinhydrinate, piracetam, pirarubicin, pirbuterol, pirenzepine, piritramide, piroxicam, pivmecillinam, pizotifen, podophyllotoxin, polidocanol, polycarbophil, polyestradiol phosphate, polymyxin B, polymyxin-B, polystyrenesulfonic acid, porfimer, prajmaline, prajmalium bitartrate, pramipexole, pranoprofen, prasterone, pravastatin, prazepam, prazosin, prednicarbate, prednisolone, prednisone, pregabalin, proglumetacin, pridinol, prilocaine, primaquine, primidone, prithipendyl, procaine, procainamide, procarbazil, procarbazine, procyclidin, progesterone, proglumetacin, proglumide, proguanil, proline, promethazine, propacetamol, propafenon, propanolol, propicillin, propiverine, propofol, propranolol, propylthiouracil, propyphenazone, protamine, protamine sulfate, protein C, prothipendyl, prothrombin, protionamide, protirelin, proxymetacaine, proxyphylline, pseudoephedrine, Pulmonal, pyrantel, pyrazinamide, pyridostigmine, pyridostigmine bromide, pyridoxine, 3-pyridylmethanol, pyrimethamine, pyrithione zinc, pyritinol, pyrogallol, pyrvinium, pyrvinium embonate, mercury amide chloride, quetiapine, quinagolide, quinapril, quinupristin, rabeprazole, racephedrine, racecadotrile, raloxifene, raltitrexed, ramipril, ranitidine, rasagiline, rasburicase, raubasine, reboxetine, repaglinide, reproterol, reserpine, resorcinol, reteplase, retinol, reviparin, ribavirin, riboflavin, rifabutin, rifampicin, rifamycin, rifaximin, rilmenidine, riluzole, rimexolone, risedronic acid, risperidone, ritonavir, rituximab, rivastigmine, rizatriptan, rocuronium bromide, rofecoxib, ropinirole, ropivacaine, ropivacaine, rosiglitazone, red mercuric sulfide F, roxatidine, roxithromycin, salbutamol, salicylic acid, salmeterol, nitric acid, nitrous acid, salverine, samarium [153Sm]
lexidronam, saquinavir, sulfur hexafluoride, scopolamine, selegiline, selenium sulfide, serine, sermorelin, sertaconazole, sertindole, sertraline, sevelamer, sevoflurane, sibutramine, silver chloride F, sildenafil, silibinin, simvastatin, sirolimus, formaldehyde solution, solifenacine, somatostatin, somatropin, sotalol, spaglumic acid, sparteine, spectinomycin, spiramycin, spirapril, spironolactone, stavudine, streptodornase, streptokinase, streptomycin, strontium ranelate, strontium chloride, strychnine, sucralfate F, sulbactam, sulesomab, sulfacetamide, sulfadiazine, sulfadimethoxine, sulfaguanidine, sulfamerazine, sulfamethoxazole, sulfamethoxydiazine, sulfametrole, sulfanilamide, sulfasalazine, sulfathiazole, sulfisomidine, sulindac, sulodexide, sulfur hexafluoride, sulpiride, sulprostone, sultamicillin, sultiame, sumatriptan, suxamethonium, tacalcitol, tacrolimus, tadalafil, tamoxifen, tamsulosin, tasonermin, taurolidine, tazarotene, tazobactam, tegafur, teicoplanin, telithromycin, telmisartan, temoporfin, temozolomide, tenecteplase, teniposide, tenofovir, tenofovir disoproxil, tenoxicam, terazosin, terbinafine, terbutaline, terfenadine, teriparatide, terizidone, terlipressin, testosterone, testosterone propionate, testosterone undecanoate, tetracaine, tetracosactide, tetracycline, tetrafluoroborate-1+, tetrofosmin, tetryzoline, thallium chloride [201T1], theobromine, theodrenaline, theodrenaline, theophylline, thiamazole, thiamine, thiethylperazine, thiocolchicoside, thiopental, thioridazine, thiotepa, threonine, thrombin, thrombokinase, thymol, thyrotropin alfa, tiagabine, tianeptine, tiapride, tibolone, ticlopidine, tiludronic acid, timolol, tinzaparin, tioconazole, tioguanine, tiotropium bromide, tirilazad, tirofiban, tisopurine, tizamidine, tizanidine, tobramycin, tocainide, tolazoline, tolbutamide, tolcapone, tolfenamic acid, tolmetin, tolperisone, tolterodine, topiramate, topotecan, torasemide, toremifene, tramazoline, trandolapril, tranexamic acid, tranylcypromine, trapidil, trastuzumab, travoprost, trazodone, tretinoin, triamcinolone, triamcinolone acetonide, triamterene, trichioroacetic acid, triethylperazine, trifluoperazine, triflupromazine, trihexyphenidyl, trimebutine, trimecaine, trimegestone, trimetazidine, trimethoprim, trimipramine, tripelennamine, triprolidine, triptorelin, tritoqualine, trofosfamide, tromantadine, trometamol, tropicamide, tropisetron, trospium, tryptophan, tubocurarine chloride, tulobuterol, tyloxapol, tyrosine, tyrothricin, unoprostone, urapid, urapidil, urokinase, ursodeoxycholic acid, valaciclovir, valdecoxib, valganciclovir, valine, valproic acid, valsartan, vancomycin, vardenafil, vecuronium, vecuronium bromide, venlafaxine, verapamil, verteporfin, vigabatrin, viloxazine, vinblastine, vincamine, vincristine, vindesine, vinorelbine, vinpocetine, viquidil, voriconazole, votumumab, hydrogen peroxide, xantinol nicotinate, ximelagatrane, xipamide, xylometazoline, yohimbine, yttrium 90Y chloride, zalcitabine, zaleplon, zanamivir, zidovudine, zinc acetate dihydrate, zinc chloride, zinc citrate, zinc sulfate, ziprasidone, zofenopril, zoledronic acid, zolmitriptan, zolpidem, zolpidem tartrate, zonisamide, zopiclone, zotepine, zucklopantexol, and zuclopenthixol.

The above-stated compounds are predominantly stated by their international nonproprietary name (INN) and are known to the person skilled in the art. Further details may be found, for example, by referring to International Nonproprietary Names (INN) for Pharmaceutical Substances, World Health Organization (WHO).

In a preferred embodiment, the pharmaceutical dosage form is monolithic.

The pharmaceutical dosage form according to the invention is characterized by a comparatively homogeneous distribution of the pharmacologically active compound (A).
Preferably, the content of the pharmacologically active compound (A) in two segments of the pharmaceutical dosage form having a volume of 1.0 mm3 each, deviates from one another by not more than 10%, more preferably not more than more than 7.5%, still more preferably not more than 5.0%, most preferably not more than 2.5%, and in particular not more than 1.0%. When the pharmaceutical dosage form is film coated, said two segments of the pharmaceutical dosage form having a volume of 1.0 mm3 each are preferably segments of the core, i.e. do not contain any coating material.

Preferably, all components of the pharmaceutical dosage form according to the invention have a comparatively homogeneous distribution within the pharmaceutical dosage form.
Preferably, the content of each component in two segments of the pharmaceutical dosage form having a volume of 1.0 mm3 each, deviates from one another by not more than 10%, more preferably not more than more than 7.5%, still more preferably not more than 5.0%, most preferably not more than 2.5%, and in particular not more than 1.0%.
When the pharmaceutical dosage form is film coated, said two segments of the pharmaceutical dosage form having a volume of 1.0 mm3 each are preferably segments of the core, i.e. do not contain any coating material.

Preferably, the pharmaceutical dosage form according to the invention is adapted for oral administration. It is also possible, however, to administer the pharmaceutical dosage form via different routes and thus, the pharmaceutical dosage form may alternatively be adapted for buccal, lingual, rectal or vaginal administration. Implants are also possible.

In a preferred embodiment, the pharmaceutical dosage form according to the invention is adapted for administration once daily. In another preferred embodiment, the pharmaceutical dosage form according to the invention is adapted for administration twice daily. In still another preferred embodiment, the pharmaceutical dosage form according to the invention is adapted for administration thrice daily.

For the purpose of the specification, "twice daily" means equal time intervals, i.e., every 12 hours, or different time intervals, e.g., 8 and 16 hours or 10 and 14 hours, between the individual administrations.

For the purpose of the specification, "thrice daily" means equal time intervals, i.e., every 8 hours, or different time intervals, e.g., 6, 6 and 12 hours; or 7, 7 and 10 hours, between the individual administrations.

Preferably, the pharmaceutical dosage form according to the invention effects an at least partially delayed release of the pharmacologically active compounds (A).

Delayed release is understood according to the invention preferably to mean a release profile in which the pharmacologically active compound (A) is released over a relatively long period with reduced intake frequency with the purpose of extended therapeutic action. This is achieved in particular with peroral administration. The expression "at least partially delayed release" covers according to the invention any pharmaceutical dosage forms which ensure modified release of the pharmacologically active compounds (A) contained therein.
The pharmaceutical dosage forms preferably comprise coated or uncoated pharmaceutical dosage forms, which are produced with specific auxiliary substances, by particular processes or by a combination of the two possible options in order purposefully to change the release rate or location of release.

In the case of the pharmaceutical dosage forms according to the invention, the release time profile may be modified e.g. as follows: extended release, repeat action release, prolonged release and sustained release.

For the purpose of the specification "extended release" preferably means a product in which the release of active compound is delayed for a finite lag time, after which release is unhindered. For the purpose of the specification "repeat action release"
preferably means a product in which a first portion of active compound is released initially, followed by at least one further portion of active compound being released subsequently. For the purpose of the specification "prolonged release" preferably means a product in which the rate of release of active compound from the formulation after administration has been reduced, in order to maintain therapeutic activity, to reduce toxic effects, or for some other therapeutic purpose.
For the purpose of the specification "sustained release" preferably means a way of formulating a medicine so that it is released into the body steadily, over a long period of time, thus reducing the dosing frequency. For further details, reference may be made, for example, to K.H. Bauer, Lehrbuch der Pharmazeutischen Technologie, 6th edition, WVG
Stuttgart, 1999; and European Pharmacopoeia.

The pharmaceutical dosage form according to the invention may comprise one or more pharmacologically active compounds (A) at least in part in a further delayed-release form, wherein delayed release may be achieved with the assistance of conventional materials and processes known to the person skilled in the art, for example by embedding the substance in a delayed-release matrix or by applying one or more delayed-release coatings. Substance release must, however, be controlled such that addition of delayed-release materials does not impair the necessary breaking strength. Controlled release from the pharmaceutical dosage form according to the invention is preferably achieved by embedding the substance in a matrix. Component (C) may serve as such a matrix.
The auxiliary substances acting as matrix materials control release. Matrix materials may, for example, be hydrophilic, gel-forming materials, from which release proceeds mainly by diffusion, or hydrophobic materials, from which release proceeds mainly by diffusion from the pores in the matrix.

Preferably, the release profile is substantially matrix controlled, preferably by embedding component (A) in a matrix comprising component (C) and optionally, further matrix materials. Preferably, the release profile is not osmotically driven.
Preferably, release kinetics is not zero order.

Preferably, under physiological conditions the pharmaceutical dosage form according to the invention has released after 30 minutes 0.1 to 75%, after 240 minutes 0.5 to 95%, after 480 minutes 1.0 to 100% and after 720 minutes 2.5 to 100% of the pharmacologically active compound (A). Further preferred release profiles R, to R5 are summarized in the table here below [all data in wt.-% of released pharmacologically active compound (A)]:

time R, R2 R3 R4 R5 60 min 0-30 0-50 0-50 15-25 20-50 120 min 0-40 0-75 0-75 25-40 40-75 240 min 3-55 3-95 10-95 40-70 60-95 480 min 10-65 10-100 35-100 60-90 80-100 720 min 20-75 20-100 55-100 70-100 90-100 960 min 30-88 30-100 70-100 >80 1440 min 50-100 50-100 >90 21'60....:., 00 o vvn 21 '60 nun - >vv - I I I

Preferably, under in vitro conditions the pharmaceutical dosage form has released after 0.5 h 1.0 to 35 wt.-%, after 1 h 5.0 to 45 wt.-%, after 2 h 10 to 60 wt.-%, after 4 h at least 15 wt.-%, after 6 h at least 20 wt.-%, after 8 h at least 25 wt.-% and after 12 h at least 30 wt.-% of the pharmacologically active compound (A) that was originally contained in the pharmaceutical dosage form.

Suitable in vitro conditions are known to the skilled artisan. In this regard it can be referred to, e.g., the European Pharmacopoeia and to the experimental section.
Preferably, the release profile is measured under the following conditions: Paddle apparatus equipped with sinker, 50 rpm, 37 5 C, 900 mL simulated intestinal fluid pH 6.8 (phosphate buffer). In a preferred embodiment, to rotational speed of the paddle is increased to 100 rpm.

Preferred release profiles R6 to R11 of the pharmacologically active compound (A) contained in the pharmaceutical dosage form are summarized in the following table:

wt.% R6 R7 R8 R9 Rio R11 after 30 min 16.3 7.5 16.3 5.0 16.3 2.5 15.8 7.5 15.8 5.0 15.8 2.5 after 60 min 27.5 7.5 25.0 5.0 25.0 2.5 24.3 7.5 24.3 5.0 24.3 2.5 after 90 min 32.1 7.5 32.1 5.0 32.1 2.5 31.2 7.5 31.2 5.0 31.2 2.5 after 120 min 38.2 7.5 38.2 5.0 38.2 2.5 37.4 7.5 37.4 5.0 37.4 2.5 after 150 min 43.4 7.5 43.4 5.0 43.4 2.5 42.7 7.5 42.7 5.0 42.7 2.5 after 180 min 48.2 7.5 48.2 5.0 48.2 2.5 47.6 7.5 47.6 5.0 47.6 2.5 after 210 min 52.7 7.5 52.7 5.0 52.7 2.5 52.2 7.5 52.2 5.0 52.2 2.5 after 240 min 56.8 7.5 56.8 5.0 56.8 2.5 56.5 7.5 56.5 5.0 56.5 2.5 after 270 min 60.7 7.5 60.7 5.0 60.7 2.5 60.4 7.5 60.4 5.0 60.4 2.5 after 300 min 64.5 7.5 64.5 5.0 64.5 2.5 64.2 7.5 64.2 5.0 64.2 2.5 after 330 min 67.9 7.5 67.9 5.0 67.9 2.5 67.7 7.5 67.7 5.0 67.7 2.5 after 360 min 71.1 7.5 71.1 5.0 71.1 2.5 71.0 7.5 71.0 5.0 71.0 2.5 after 390 min 74.2 7.5 74.2 5.0 74.2 2.5 74.0 7.5 74.0 5.0 74.0 2.5 after 420 min 77.0 7.5 77.0 5.0 77.0 2.5 76.9 7.5 76.9 5.0 76.9 2.5 after 450 min 79.5 7.5 79.5 5.0 79.5 2.5 79.5 7.5 79.5 5.0 79.5 2.5 after 480 min 81.9 7.5 81.9 5.0 81.9 2.5 82.0 7.5 82.0 5.0 82.0 2.5 after 510 min 84.2 7.5 84.2 5.0 84.2 2.5 84.2 7.5 84.2 5.0 84.2 2.5 after 540 min 86.3 7.5 86.3 5.0 86.3 2.5 86.3 7.5 86.3 5.0 86.3 2.5 after 570 min 88.3 7.5 88.3 5.0 88.3 2.5 88.1 7.5 88.1 5.0 88.1 2.5 after 600 min 90.1 7.5 90.1 5.0 90.1 2.5 89.8 7.5 89.8 5.0 89.8 2.5 after 630 min 91.9 7.5 91.9 5.0 91.9 2.5 91.4 7.5 91.4 5.0 91.4 2.5 after 660 min 93.3 7.5 93.3 5.0 93.3 2.5 92.7 7.5 92.7 5.0 92.7 2.5 after 690 min 94.3 7.5 94.3 5.0 94.3 2.5 94.0 7.5 94.0 5.0 94.0 2.5 after 720 min 95.8 7.5 95.8 5.0 95.8 2.5 95.1 7.5 95.1 5.0 95.1 2.5 Preferably, the release properties of the pharmaceutical dosage form according to the invention are substantially independent from the pH value of the release medium, i.e.
preferably the release profile in artificial intestinal juice substantially corresponds to the release profile in artificial gastric juice. Preferably, at any given time point the release profiles deviate from one another by not more than 20%, more preferably not more than 15%, still more preferably not more than 10%, yet more preferably not more than 7.5%, most preferably not more than 5.0% and in particular not more than 2.5%.

Preferably, the pharmaceutical dosage form according to the invention exhibits a uniform release profile. Preferably, the release profile of the pharmacologically active compound (A) is interindividually uniform (i.e. when comparing pharmaceutical dosage forms obtained from the same process) (c.f. Figure 22) and/or uniform within a single pharmaceutical dosage form (i.e. when comparing segments of the same pharmaceutical dosage form).
Preferably, when comparing two probes each having a mass of preferably 500 mg, the total amount of the released active compound for any given time point of the measurement does not deviate by more than 20%, more preferably not more than 15%, still more preferably not more than 10%, yet more preferably not more than 7,5%, most preferably not more than 5,0% and in particular not more than 2,5%.

Preferably, the entire outer surface of the dosage form, preferably tablet, according to the invention is permeable for water and for component (A), i.e., preferably the water penetration characteristics of the surface are identical everywhere. This means that preferably, the dosage form, preferably tablet, is not partially coated with a material having a barrier effect which would impede the penetration of water at the location of the surface where it is applied.

Preferably, the release profile of the pharmaceutical dosage form according to the present invention is stable upon storage, preferably upon storage at elevated temperature, e.g.
37 C, for 3 months in sealed containers. In this regard "stable" means that when comparing the initial release profile with the release profile after storage, at any given time point the release profiles deviate from one another by not more than 20%, more preferably not more than 15%, still more preferably not more than 10%, yet more preferably not more than 7.5%, most preferably not more than 5.0% and in particular not more than 2.5%
(cf. Figure 24).

It has been surprisingly found that by modifying the outer shape of the pharmaceutical dosage form the storage stability, e.g. the storage stability of the release profile, can be increased compared to conventional dosage forms having a comparable release profile before storage.

Preferably, the pharmaceutical dosage form according to the invention contains at least one polymer (C), for the purpose of the specification also referred to as "component (C)".
Preferably, the pharmaceutical dosage form contains at least one synthetic, semi-synthetic or natural polymer (C), which contributes considerably to the elevated breaking strength (resistance to crushing) of the pharmaceutical dosage form. For the purpose of the specification a "semi-synthetic" product has been produced by chemical manipulation of naturally occurring substances.

Preferably, the mechanical properties of the pharmaceutical dosage form according to the invention, particularly its breaking strength, substantially rely on the presence of polymer (C), although its mere presence does not suffice in order to achieve said properties. The advantageous properties of the pharmaceutical dosage form according to the invention, in particular also its mechanical properties, may not automatically be achieved by simply processing the pharmacologically active compound (A), polymer (C), and optionally further excipients by means of conventional methods for the preparation of pharmaceutical dosage forms. In fact, usually suitable apparatuses must be selected for the preparation and critical processing parameters must be adjusted, particularly pressure/force, temperature and time.
Thus, even if conventional apparatuses are used, the process protocols usually must be adapted in order to meet the required criteria.

Preferably, polymer (C) is water-soluble. Preferably, polymer (C) is substantially unbranched.

Polymer (C) may comprise a single type of polymer having a particular average molecular weight, or a mixture (blend) of different polymers, such as two, three, four or five polymers, e.g., polymers of the same chemical nature but different average molecular weight, polymers of different chemical nature but same average molecular weight, or polymers of different chemical nature as well as different molecular weight.

Individual or combinations of polymers may be selected from the group comprising poly-alkylene oxide, preferably polymethylene oxide, polyethylene oxide, polypropylene oxide;
polyethylene, polypropylene, polyvinyl chloride, polycarbonate, polystyrene, polyvinyl-pyrrolidone, poly(alk)acrylate, poly(hydroxy fatty acids), such as for example poly(3-hyd roxybutyrate-co-3-hyd roxyva I e rate) (Biopol ), poly(hydroxyvaleric acid); polycapro-lactone, polyvinyl alcohol, polyesteramide, polyethylene succinate, polylactone, polyglycolide, polyurethane, polyamide, poiyiactide, poiyacetsi (for examiple polysaccharides optionally with modified side chains), polylactide/glycolide, polylactone, polyglycolide, polyorthoester, polyanhydride, block polymers of polyethylene glycol and polybutylene terephthalate (Polyactive ), polyanhydride (Polifeprosan), copolymers thereof, block-copolymers thereof, and mixtures of at least two of the stated polymers, or other polymers with the above characteristics.

Preferably, polymer (C) comprises a polyalkylene oxide, more preferably a polyethylene oxide, a polypropylene oxide, an ethylene oxide-propylene oxide copolymerisate, which may be e.g. a random copolymer, alternating copolymer or block copolymer, or a mixture of any of the foregoing.

Particularly preferred are high molecular weight polymers with a preferably weight average molecular weight (Mw) or viscosity average molecular weight (M1) of at least of at least 0.1 106 g/mol, of at least 0.2 = 106 g/mol, of at least 0.5 = 106 g/mol, of at least 1.0 = 106 g/mol, of at least 2.5 = 106 g/mol, of at least 5.0 = 106 g/mol, of at least 7.5 = 106 g/mol or of at least 10 = 106 g/mol, preferably 1.0 = 106 g/mol to 15 = 106 g/mol. Suitable methods for determining MW or M. are known to the person skilled in the art. Preferably, M. is determined using rheological measurements and MW is determined using gel permeation chromatography (GPC) on suitable phases.

Preferably, the molecular weight dispersity M,/Mn of polymer (C) is within the range of 2.5 2.0, more preferably 2.5 1.5, still more preferably 2.5 1.0, yet more preferably 2.5 0.8, most prefrably 2.5 0.6, and in particular 2.5 0.4.

The polymers preferably have a viscosity at 25 C of 4,500 to 17,600 cP, measured in a 5 wt.-% aqueous solution using a model RVF Brookfield viscosimeter (spindle no.

rotational speed 2 rpm), of 400 to 4,000 cP, measured on a 2 wt.-% aqueous solution using the stated viscosimeter (spindle no. 1 or 3 / rotational speed 10 rpm) or of 1,650 to 10,000 cP, measured on a 1 wt.-% aqueous solution using the stated viscosimeter (spindle no. 2 /
rotational speed 2 rpm).

Most preferred are thermoplastic polyalkylene oxides having a weight average molecular weight (M,w) or a viscosity average molecular weight (Mn) of at least 0.2 =
106 g/mol, more preferably at least 0.3 = 106 g/mol, still more preferably at least 0.4 = 106 g/mol, yet more preferably at least 0.5 = 106 g/mol, most preferably at least 1.0 = 106 g/mol and in particular within the range of 1.0 = 106 to 15 = 106 g/mol are preferred, e.g.
polyethylene oxides, polypropylene oxides or the (block-)copolymers thereot.

In a preferred embodiment according to the invention the polymer (C) comprises - a polyalkylene oxide having a weight average molecular weight (Mw) or viscosity average molecular weight (M,,) of at least 0.2 = 106 g/mol in combination with - at least one further polymer, preferably but not necessarily also having a weight average molecular weight (Mw) or viscosity average molecular weight (Mn) of at least 0.2 = 106 g/mol, selected from the group consisting of polyethylene, polypropylene, polyvinyl chloride, polycarbonate, polystyrene, polyacrylate, poly(hydroxy fatty acids), polycaprolactone, polyvinyl alcohol, polyesteramide, polyethylene succinate, polylactone, polyglycolide, polyurethane, polyvinylpyrrolidone, polyamide, polylactide, polylactide/glycolide, polylactone, polyglycolide, polyorthoester, polyanhydride, block polymers of polyethylene glycol and polybutylene terephthalate, polyanhydride, polyacetal, cellulose esters, cellulose ethers and copolymers thereof.
Cellulose esters and cellulose ethers are particularly preferred, e.g. methylcelIulose, ethylcellulose, hyd roxymethylcel I u lose, hydroxypropylmethylcellulose, ca rboxymethylcel I
u lose, and the like.

In a preferred embodiment, said further polymer is neither a polyalkylene oxide nor a polyalkylene glycol. Nonetheless, the pharmaceutical dosage form may contain polyalkylene glycol, e.g. as plasticizer, but then, the pharmaceutical dosage form preferably is a ternary mixture of polymers: component (C) + further polymer +
plasticizer.

In a particularly preferred embodiment, said further polymer is a hydrophilic cellulose ester or cellulose ether, preferably hydroxypropylmethylcellulose, preferably having an average viscosity of 100,000 50,000 mPas, more preferably 100,000 20,000 mPas.

Preferably, the content of said further polymer amounts to 0.5 to 25 wt.-%, more preferably 1.0 to 20 wt.-%, still more preferably 2.0 to 17.5 wt.-%, yet more preferably 3.0 to 15 wt.-%

and most preferably 4.0 to 12.5 wt.-% and in particular 5.0 to 10 wt.-%, based on the total weight of the polyalkylene oxide.

in a preferred embodiment the reiaiive weight ratio of said poiyaikyiene oxide and said further polymer is within the range of from 20:1 to 1:20, more preferably 10:1 to 1:10, still more preferably 7:1 to 1:5, yet more preferably 5:1 to 1:1, most preferably 4:1 to 1,5:1 and in particular 3:1 to 2:1.

Preferably, the content of said further polymer amounts to 0.5 to 25 wt.-%, more preferably 1.0 to 20 wt.-%, still more preferably 2.0 to 22.5 wt.-%, yet more preferably 3.0 to 20 wt.-%
and most preferably 4.0 to 17.5 wt.-% and in particular 5.0 to 15 wt.-%, based on the total weight of the pharmaceutical dosage form.

It is not intended to be bound by any theory, but it is believed that the further polymer may serve as a supplementary matrix material that guarantees a minimal retardant effect on the release of the pharmacologically active compound (A) even if the molecular chains of the polyalkylene oxide have been partially damaged in the course of the manufacture of the pharmaceutical dosage form, e.g. by extrusion, thereby decreasing the average molecular weight. Furthermore, it seems that the further polymer contributes to the storage stability of the dosage form, particularly with respect to its release profile.

Physiologically acceptable, hydrophobic materials which are known to the person skilled in the art may be used as supplementary matrix materials. Polymers, particularly preferably cellulose ethers, cellulose esters and/or acrylic resins are preferably used as hydrophilic matrix materials. Ethylcellulose, hydroxypropylmethylcellulose, hydroxypropylcelIulose, hyd roxymethylcel I u lose, poly(meth)acrylic acid and/or the derivatives thereof, such as the salts, amides or esters thereof are very particularly preferably used as matrix materials.
Matrix materials prepared from hydrophobic materials, such as hydrophobic polymers, waxes, fats, long-chain fatty acids, fatty alcohols or corresponding esters or ethers or mixtures thereof are also preferred. Mono- or diglycerides of C12-C30 fatty acids and/or C12-C30 fatty alcohols and/or waxes or mixtures thereof are particularly preferably used as hydrophobic materials. It is also possible to use mixtures of the above-stated hydrophilic and hydrophobic materials as matrix materials.

Preferably, the overall content of polymer (C) is preferably at least 5 wt.-%, at least 10 wt.-%, at least 15 wt.-% or at least 20 wt.-%, more preferably at least 30 wt.-%, still more preferably at least 40 wt.-%, most preferably at least 50 wt.-% and in particular at least 60 wt.-%, of the total weight of the pharmaceutical dosage form. In a preferred embodiment the content of the polymer (C) is within the range of from about 20 to about 49 wt.-% of the total weight of the pharmaceutical dosage form.

In a preferred embodiment, the overall content of polymer (C) is within the range of 25 20 wt.-%, more preferably 25 15 wt.-%, most preferably 25 10 wt.-%, and in particular 25 5 wt.-%. In another preferred embodiment, the overall content of polymer (C) is within the range of 35 20 wt.-%, more preferably 35 15 wt.-%, most preferably 35 10 wt.-%, and in particular 35 5 wt.-%. In still another preferred embodiment, the overall content of polymer (C) is within the range of 45 20 wt.-%, more preferably 45 15 wt.-%, most preferably 45 10 wt.-%, and in particular 45 5 wt.-%. In yet another preferred embodiment, the overall content of polymer (C) is within the range of 55 20 wt.-%, more preferably 55 15 wt.-%, most preferably 55 10 wt.-%, and in particular 55 5 wt.-%. In a further preferred embodiment, the overall content of polymer (C) is within the range of 65 20 wt.-%, more preferably 65 15 wt.-%, most preferably 65 10 wt.-%, and in particular 65 5 wt.-%. In still a further a preferred embodiment, the overall content of polymer (C) is within the range of 75 20 wt.-%, more preferably 75 15 wt.-%, most preferably 75 10 wt.-%, and in particular 75 5 wt.-%.

In a preferred embodiment, polymer (C) is homogeneously distributed in the pharmaceutical dosage form according to the invention. Preferably, polymer (C) forms a matrix in which the pharmacologically active compound (A) is embedded. In a particularly preferred embodiment, the pharmacologically active compound (A) and polymer (C) are intimately homogeneously distributed in the pharmaceutical dosage form so that the pharmaceutical dosage form does not contain any segments where either pharmacologically active compound (A) is present in the absence of polymer (C) or where polymer (C) is present in the absence of pharmacologically active compound (A).

When the pharmaceutical dosage form is film coated, the polymer (C) is preferably homogeneously distributed in the core of the pharmaceutical dosage form, i.e.
the film coating preferably does not contain polymer (C). Nonetheless, the film coating as such may of course contain one or more polymers, which however, preferably differ from the polymer (C) contained in the core.

Preferably, the pharmaceutical dosage form according to the invention contains a coating, preferably a film-coating. Suitable coating materials are known to the skilled person.

Suitable coating materials are commercially available, e.g. under the trademarks Opadry and Eudragit .

Examples of suitable materials include cellulose esters and cellulose ethers, such as methylcellulose (MC), hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), hydroxyethylcellulose (HEC), sodium carboxymethylcellulose (Na-CMC), ethylcellulose (EC), cellulose acetate phthalate (CAP), hydroxypropylmethylcellulose phthalate (HPMCP); poly(meth)acrylates, such as aminoalkylmethacrylate copolymers, ethylacrylate methylmethacrylate copolymers, methacrylic acid methylmethacrylate copolymers, methacrylic acid methylmethacrylate copolymers; vinyl polymers, such as polyvinylpyrrolidone, polyvinylacetatephthalate, polyvinyl alcohol, polyvinylacetate; and natural film formers, such as shellack.

In a particularly preferred embodiment, the coating is water-soluble.
Preferably, the coating is based on polyvinyl alcohol, such as polyvinyl alcohol-part. hydrolyzed, and may additionally contain polyethylene glycol, such as macrogol 3350, and/or pigments.

The coating of the pharmaceutical dosage form can increase its storage stability.

The coating can be resistant to gastric juices and dissolve as a function of the pH value of the release environment. By means of this coating, it is possible to ensure that the pharmaceutical dosage form according to the invention passes through the stomach undissolved and the active compound is only released in the intestines. The coating which is resistant to gastric juices preferably dissolves at a pH value of between 5 and 7.5.
Corresponding materials and methods for the delayed release of active compounds and for the application of coatings which are resistant to gastric juices are known to the person skilled in the art, for example from "Coated Pharmaceutical dosage forms -Fundamentals, Manufacturing Techniques, Biopharmaceutical Aspects, Test Methods and Raw Materials"
by Kurt H. Bauer, K. Lehmann, Hermann P. Osterwald, Rothgang, Gerhart, 1st edition, 1998, Medpharm Scientific Publishers.

It has been surprisingly found that the pharmaceutical dosage forms can be easily film coated, although the outer shape of the pharmaceutical dosage forms may deviate from conventional shapes of dosage forms. For example, when the pharmaceutical dosage forms according to the invention are shaped by means of an H-plunger, a skilled person would expect that substantial stapling or stacking and pair formation occurs when spraying the coating material on the dosage form in suitable devices. Such stacking and agglomeration of the dosage form would deteriorate the coating performance and hence, the film quality. However, it has been surprisingly found that even pharmaceutical dosage forms having irregular shapes may be smoothly coated.

In a particularly preferred embodiment, - the pharmaceutical dosage form is thermoformed, preferably by extrusion;
and/or - a portion of the surface of the pharmaceutical dosage form is convex, another portion of its surface is concave; and/or - the pharmacologically active compound (A) is a psychotropically acting substance, preferably an analgesic, more preferably a drug selected from the group consisting of opioids, stimulants, tranquillisers (e.g. barbiturates and benzodiazepines) and other narcotics; and/or - the content of the pharmacologically active compound (A) is at least 0.5 wt.-%, based on the total weight of the dosage form; and/or - polymer (C) is an polyalkylene oxide having a weight average molecular weight of at least 200,000 g/mol, preferably at least 500,000 g/mol, more preferably within the range of from 1,000,000 to 10,000,000 g/mol; and/or - the content of the polyalkylene oxide is at least 15 wt.-%, based on the total weight of the dosage form; and/or - the opioid is embedded in the polyalkylene oxide, i.e., in a matrix formed by the polyalkylene oxide; and/or - the pharmaceutical dosage form contains a plasticizer, preferably polyethylene glycol;
and/or - the content of said plasticizer is at least 5 wt.-%, based on the total weight of the dosage form; and/or - besides polymer (C), the pharmaceutical dosage form contains a further polymer selected from the group consisting of cellulose ethers, cellulose esters and acrylates, preferably HPMC; and/or - the content of said further polymer is at least 1 wt.-%, based on the total weight of the dosage form; and/or - the pharmaceutical dosage form contains an antioxidant, preferably a-tocopherol;
and/or - the content of said antioxidant is at least 0.05 wt.-%, based on the total weight of the dosage form; and/or - the pharmaceutical dosage form is adapted for oral administration once daily or twice daily; and/or - the pharmaceutical dosage form contains a coating, preferably a film-coating.

Besides the pharmacologically active compound (A) and polymer (C) the pharmaceutical dosage form according to the invention may contain further constituents, such as conventional pharmaceutical excipients.

In a preferred embodiment, the pharmaceutical dosage form contains at least one natural, semi-synthetic or synthetic wax (D), for the purpose of the specification also referred to as "component (D)". Preferred waxes are those with a softening point of at least 50 C, more preferably of at least 55 C, still more preferably of at least 60 C, most preferably of at least 65 C and in particular at least 70 C.

Carnauba wax and beeswax are particularly preferred. Carnauba wax is very particularly preferred. Carnauba wax is a natural wax which is obtained from the leaves of the carnauba palm and has a softening point of at least 80 C. When the wax component is additionally contained, its content is sufficiently high so that the desired mechanical properties of the pharmaceutical dosage form are achieved.

Auxiliary substances (B), further purpose of the specification also referred to as "component (B)", which may be contained in the pharmaceutical dosage form according to the invention are those known auxiliary substances which are conventional for the formulation of solid pharmaceutical dosage forms.

Examples of auxiliary substances (B) are plasticizers, (further) matrix materials, antioxidants and the like.

Suitable plasticizers include triacetin and polyethylene glycol, preferably a low molecular weight polyethylene glycol (e.g. macrogol 6000).

Matrix materials are auxiliary substances which influence active compound release, preferably hydrophobic or hydrophilic, preferably hydrophilic polymers, very particularly preferably hydroxypropylmethylcellulose, and/or antioxidants. Polymers, particularly preferably cellulose ethers, cellulose esters and/or acrylic resins are preferably contained as hydrophilic matrix materials. Ethylcellulose, hydroxypropylmethylcellulose, hyd roxypropyl cel I u lose, hydroxymethylcellulose, poly(meth)acrylic acid and/or the derivatives thereof, such as the copolymers, salts, amides or esters thereof are very particularly preferably contained as matrix materials.

Suitable antioxidants are ascorbic acid, butylhydroxyanisole (BHA), butylhydroxytoluene (BHT), salts of ascorbic acid, monothioglycerol, phosphorous acid, vitamin C, vitamin E and the derivatives thereof, coniferyl benzoate, nordihydroguajaretic acid, gallus acid esters, :. ..... :_..Ira.. :_..I__~.. L_-_1-bi 1.. L bu..L_.IL. '---'--sou.,1iui i i bisunna, par..1tcuiai iy pre to atp ryd oxytoluene or bulyihydroxyariisoie and a-tocopherol. The antioxidant is preferably used in quantities of 0.01 to 10 wt.-%, preferably of 0.03 to 5 wt.-%, relative to the total weight of the pharmaceutical dosage form.

Preferred compositions X, to X4 of the pharmaceutical dosage form according to the invention are summarized in the table here below:

wt.-% X1 X2 X3 X4 component (A) 26.5 25 26.5 20 26.5 15 26.5 13 polyalkylene oxide (e.g. PEO) 46.5 25 46.5 17 46.5 12 46.5 10 cellulose ester or ether (e.g. HPMC) 14 7 14 5 14 2.5 14 0.5 plasticizer (e.g. PEG) 12.5 10 12.5 7 12.5 5 12.5 3 antioxidant (e.g. a-toco hero) 0.125 0.12 0.125 0.1 0.125 0.05 0.125 0.03 The pharmaceutical dosage form according to the invention may be produced by different processes, which are explained in greater detail below; the present invention also relates to pharmaceutical dosage forms that are obtainable by any of the processes described here below:

In general, the, process for the production of the pharmaceutical dosage form according to the invention preferably comprises the following steps:

(a) mixing of components (A), (C), optionally (B) and/or (D);

(b) optionally pre-forming the mixture obtained from step (a), preferably by applying heat and/or force to the mixture obtained from step (a), the quantity of heat supplied preferably not being sufficient to heat component (C) up to its softening point;

(c) hardening the mixture by applying heat and force, it being possible to supply the heat during and/or before the application of force and the quantity of heat supplied being sufficient to heat component (C) at least up to its softening point;

(d) optionally singulating the hardened mixture;

(e) optionally shaping the pharmaceutical dosage form; and (f) optionally providing a film coating.

Heat may be supplied directly or with the assistance of ultrasound. Force may be applied and/or the pharmaceutical dosage form may be shaped ioi exampie by direct iabieiiing or with the assistance of a suitable extruder, particularly by means of a screw extruder equipped with two screws (twin-screw-extruder) or by means of a planetary gear extruder.
The shaping of the pharmaceutical dosage form according to the invention is of particular importance. The final shape of the pharmaceutical dosage form may either be provided during the hardening of the mixture by applying heat and force (step (c)) or in a subsequent step (step (e)). In both cases, the mixture of all components is preferably in the plastified state, i.e. preferably, shaping is performed at a temperature at least above the softening point of component (C).

Shaping can be performed, e.g., by means of a tabletting press comprising die and plunger of appropriate shape. Preferably, the plunger is a H-plunger so that the cross section of the pharmaceutical dosage form assumes the form of a H.

The following process variants are particularly preferred:
Process variant 1:

In this embodiment, the pharmaceutical dosage form according to the invention is preferably produced without using an extruder by preferably mixing components (A), (C), optionally (B) and/or (D) and, optionally after granulation, shaping the resultant mixture by application of force to yield the pharmaceutical dosage form with preceding and/or simultaneous exposure to heat.

This heating and application of force for the production of the pharmaceutical dosage form proceeds without using an extruder.

Components (A), (C), optionally (B) and/or (D) are mixed in a mixer known to the person skilled in the art. The mixer may, for example, be a roll mixer, shaking mixer, shear mixer or compulsory mixer.

The resultant mixture is preferably directly shaped into the pharmaceutical dosage form according to the invention by application of force with preceding and/or simultaneous exposure to heat. The mixture may, for example, be formed into tablets by direct tabletting.
In direct tabletting with preceding exposure to heat, the material to be pressed is heated immediately prior to tabletting at least to the softening temperature of component (C), preferably to its melting temperature, and then pressed. in the case of direct iabietting with simultaneous application of heat, the mixture to be press-formed is heated at least to the softening point of polymeric component (C) with the assistance of the tabletting tool, i.e. the bottom punch, top punch and the die, and is so press-formed.

This method may also be regarded as a sintering method and the dosage form thus obtained may be regarded as a sintered dosage form. In this regard, sintering means the solidification of crystalline, granular or powdered material by growing together of the crystallites when heated appropriately. The growing together can take place, e.g., by diffusion (solid/solid-reaction). The sintered structure can be analyzed by methods known in the art, e.g., by (electron)microscopy. As the dosage form contains several components, the sintering method is preferably a so-called "fusion sintering" where one of the components involved melts, wets and coats the higher-melting components. When cooling down, the melted component (re)solidifies. In a preferred embodiment, component (C) is the component that melts while the other components do not melt under the given conditions. Nonetheless, the other components may - but do not have to - at least partially dissolve in the melted component (C).

By such process using a tabletting tool with bottom punch, top punch and die, e.g. a powder mixture of all components may be compressed at a temperature of e.g. 80 C, the pressure caused by a force of e.g. 2 kN or 4 kN being maintained for e.g. 15 seconds.

The resultant mixture of components (A), (C), optionally (B) and/or (D) may also first be granulated and then, with preceding and/or simultaneous exposure to heat, be shaped into the pharmaceutical dosage form according to the invention by application of force.

Granulation may be performed in known granulators by wet granulation or melt granulation.
Each of the above-mentioned process steps, in particular the heating steps and simultaneous or subsequent application of force for production of the pharmaceutical dosage form according to the invention proceeds without using an extruder.

Process variant 2:

In this process variant, the pharmaceutical dosage form according to the invention is produced by thermoforming with the assistance of an extruder, preferably without there being any observable consequent discoloration of the extrudate.

In order to investigate the extent of discoloration due to this thermoforming, the colour of the mixture of starting components of which the pharmaceutical dosage form consists is first determined without addition of a color-imparting component, such as for example a coloring pigment or an intrinsically colored component (for example a-tocopherol). This composition is then thermoformed according to the invention, wherein all process steps, including cooling of the extrudate, are performed under an inert gas atmosphere. By way of comparison, the same composition is produced by the same process, but without an inert gas atmosphere. The color of the pharmaceutical dosage form produced according to the invention from the starting composition and of the pharmaceutical dosage form produced by way of comparison is determined. The determination is performed with the assistance of "Munsell Book of Color" from Munsell Color Company Baltimore, Maryland, USA, edition. If the color of the pharmaceutical dosage form thermoformed according to the invention has a color with identification no. N 9.5, but at most a color with the identification no. 5Y 9/1, thermoforming is classed as being "without discoloration". If the pharmaceutical dosage form has a color with the identification no. 5Y 9/2 or greater, as determined according to the Munsell Book of Color, the thermoforming is classed as being "with discoloration".

Surprisingly, the pharmaceutical dosage forms according to the invention exhibit no discoloration classed in accordance with the above classification, if the entire production process is performed under an inert gas atmosphere, preferably under a nitrogen atmosphere with the assistance of an extruder for thermoforming.

This variant according to the invention for the production of pharmaceutical dosage forms according to the invention is characterized in that z) components (A), (C), optionally (B) and/or (D) are mixed, y) the resultant mixture is heated in the extruder at least up to the softening point of component (C) and extruded through the outlet orifice of the extruder by application of force, x) the still plastic extrudate is singulated and formed into the pharmaceutical dosage form or w) the cooled and optionally reheated singulated extrudate is formed into the pharmaceutical dosage form, wherein process steps y) and x) and optionally process steps z) and w) are performed under an inert gas atmosphere, preferably a nitrogen atmosphere.

Mixing of the components according to process step z) may also proceed in the extruder.
Components (A), (C), optionally (B) and/or (D) may also be mixed in a mixer known to the person skilled in the art. The mixer may, for example, be a roll mixer, shaking mixer, shear mixer or compulsory mixer.

Before blending with the remaining components, component (C) and/or (D) is preferably provided according to the invention with an antioxidant. This may proceed by mixing the two components, (C) and the antioxidant, preferably by dissolving or suspending the antioxidant in a highly volatile solvent and homogeneously mixing this solution or suspension with component (C) and the optionally present component (D) and removing the solvent by drying, preferably under an inert gas atmosphere.

The, preferably molten, mixture which has been heated in the extruder at least up to the softening point of component (C) is extruded from the extruder through a die with at least one bore.

The process according to the invention requires the use of suitable extruders, preferably screw extruders. Screw extruders which are equipped with two screws (twin-screw-extruders) are particularly preferred.

The extrusion is preferably performed so that the expansion of the strand due to extrusion is not more than 50%, i.e. that when using a die with a bore having a diameter of e.g. 6 mm, the extruded strand should have a diameter of not more than 9 mm. More preferably, the expansion of the strand is not more than 40%, still more preferably not more than 35%, most preferably not more than 30% and in particular not more than 25%. It has been surprisingly found that if the extruded material in the extruder is exposed to a mechanical stress exceeding a certain limit, a significant expansion of the strand occurs thereby resulting in undesirable irregularities of the properties of the extruded strand, particularly its mechanical properties.

Preferably, extrusion is performed in the absence of water, i.e., no wafer is added.
However, traces of water (e.g., caused by atmospheric humidity) may be present.

The extruder preferably comprises at least two temperature zones, with heating of the mixture at least up to the softening point of component (C) proceeding in the first zone, which is downstream from a feed zone and optionally mixing zone. The throughput of the mixture is preferably from 2.0 kg to 8.0 kg/hour.

After heating at least up to the softening point of component (C), the molten mixture is conveyed with the assistance of the screws, further homogenised, compressed or compacted such that, immediately before emerging from the extruder die, it exhibits a minimum pressure of 5 bar, preferably of at least 7.5 bar, more preferably at least 10 bar, still more preferably at least 12.5 bar, yet more preferably at least 15 bar, most preferably at least 17.5 bar and in particular at least 20 bar, and is extruded through the die as an extruded strand or strands, depending on the number of bores which the die comprises.

In a preferred embodiment, the die head pressure is within the range of from 25 to 85 bar.
The die head pressure can be adjusted inter alia by die geometry, temperature profile and extrusion speed.

The die geometry or the geometry of the bores is freely selectable. The die or the bores may accordingly exhibit a round, oblong or oval cross-section, wherein the round cross-section preferably has a diameter of 0.1 mm to 15 mm and the oblong cross-section preferably has a maximum lengthwise extension of 21 mm and a crosswise extension of 10 mm. Preferably, the die or the bores have a round cross-section. The casing of the extruder used according to the invention may be heated or cooled. The corresponding temperature control, i.e. heating or cooling, is so arranged that the mixture to be extruded exhibits at least an average temperature (product temperature) corresponding to the softening temperature of component (C) and does not rise above a temperature at which the pharmacologically active compound (A) to be processed may be damaged.
Preferably, the temperature of the mixture to be extruded is adjusted to below 180 C, preferably below 150 C, but at least to the softening temperature of component (C). Typical extrusion temperatures are 120 C and 130 C.

In a preferred embodiment, the extruder torque is within the range of from 25 to 55 N/m.
Extruder torque can be adjusted inter alia by die geometry, temperature profile and extrusion speed.

After extrusion of the molten mixture and optional cooling of the extruded strand or extruded strands, the extrudates are preferably singulated. This singulation may preferably be performed by cutting up the extrudates by means of revolving or rotating knives, water jet cutters, wires, blades or with the assistance of laser cutters.

An inert gas atmosphere is not necessary for intermediate or final storage of the optionally singulated extrudate or the final shape of the pharmaceutical dosage form according to the invention.

The singulated extrudate may be pelletized with conventional methods or be press-formed into tablets in order to impart the final shape to the pharmaceutical dosage form. It is, however, also possible not to singulate the extruded strands and, with the assistance of contrarotating calender rolls comprising opposing recesses in their outer sleeve, to form them into the final shape, preferably a tablet, and to singulate these by conventional methods.

Should the optionally singulated extrudate not immediately be formed into the final shape, but instead cooled for storage, after the period of storage an inert gas atmosphere, preferably a nitrogen atmosphere, should be provided and must be maintained during heating of the stored extrudate up until plasticization and definitive shaping to yield the pharmaceutical dosage form.

The application of force in the extruder onto the at least plasticized mixture is adjusted by controlling the rotational speed of the conveying device in the extruder and the geometry thereof and by dimensioning the outlet orifice in such a manner that the pressure necessary for extruding the plasticized mixture is built up in the extruder, preferably immediately prior to extrusion. The extrusion parameters which, for each particular composition, are necessary to give rise to a pharmaceutical dosage form with a resistance to crushing of at least 400 N, preferably of at least 500 N, may be established by simple preliminary testing.
For example, extrusion may be performed by means of a twin-screw-extruder type Micro 27 GL 40 D (Leistritz, Nurnberg, Germany), screw diameter 27 mm. Screws having eccentric ends may be used. A heatable die with a round bore having a diameter of 8 mm may be used. The entire extrusion process should be performed under nitrogen atmosphere. The extrusion parameters may be adjusted e.g. to the following values: rotational speed of the screws: 100 Upm; delivery rate: 4 kg/h; product temperature: 125 C; and jacket temperature: 120 OC.

Process variant 3:

In this process variant for the production of the pharmaceutical dosage form according to the invention energy is applied to a mixture of the components by means of ultrasonication.
First of all a homogeneous mixture of at least component (A) and component (C) (= binder) is produced. Further auxiliary substances, such as for example fillers, plasticisers, slip agents or dyes, may also be incorporated into this mixture. A low molecular weight polyethylene glycol is preferably used as plasticizser.

Mixing may be performed with the assistance of conventional mixers. Examples of suitable mixers are roll mixers, which are also known as tumbler, drum or rotary mixers, container mixers, barrel mixers (drum hoop mixers or tumbling mixers) or shaking mixers, shear mixers, compulsory mixers, plough bar mixers, planetary kneader-mixers, Z
kneaders, sigma kneaders, fluid mixers or high-intensity mixers.

Selection of the suitable mixer is determined inter alia by the flowability and cohesiveness of the material to be mixed.

The mixture is then subjected to shaping. The mixture is preferably shaped during or after ultrasonication, preferably by compaction.

It is particularly preferred during ultrasonication that there is direct contact between the mixture and the sonotrode of the ultrasound device.

A frequency of 1 kHz to 2 MHz, preferably of 15 to 40 kHz, should be maintained during ultrasonication. Ultrasonication should be performed until softening of the polymer (C) is achieved. This is preferably achieved within a few seconds, particularly preferably within 0.1 to 5 seconds, preferably 0.5 to 3 seconds.

Ultrasonication and the application of force ensure uniform energy transfer, so bringing about rapid and homogeneous sintering of the mixture. In this manner, pharmaceutical dosage forms are obtained which have a resistance to crushing of at least 400 N, preferably of at least 500 N, and thus cannot be pulverized.

Before shaping is performed, the mixture may be granulated after the mixing operaiiori, after which the resultant granules are shaped into the pharmaceutical dosage form with ultrasonication and application of force.

Granulation may be performed in machinery and apparatus known to the person skilled in the art.

If granulation is performed as wet granulation, water or aqueous solutions, such as for example ethanol/water or isopropanol/water, may be used as the granulation liquid.

The mixture or the granules produced therefrom may also be subjected to melt extrusion for further shaping, wherein the mixture is converted into a melt by ultrasonication and exposure to force and then extruded through a dies. The strands or strand obtained in this manner may be singulated to the desired length using known apparatus. The formed articles singulated in this manner may optionally furthermore be converted into the final shape with ultrasonication and application of force.

Final shaping to yield the pharmaceutical dosage form preferably proceeds with application of force in appropriate moulds.

The above-described formed articles may also be produced with a calendering process by initially plasticising the mixture or the granules produced therefrom by means of ultrasonication and application of force and performing extrusion through an appropriate die. These extrudates are then shaped into the final shape between two contrarotating shaping rolls, preferably with application of force.

As already mentioned, shaping to yield the final shape of the pharmaceutical dosage form by using a mixture comprising pharmacologically active compound (A) and polymer (C) proceeds preferably in powder form by direct compression with application of force, wherein ultrasonication of this mixture is provided before or during the application of force. The force is at most the force which is conventionally used for shaping pharmaceutical dosage forms, such as tablets, or for press-forming granules into the corresponding final shape.

The tablets produced according to the invention may also be multilayer tablets.

In multilayer tablets, at least the layer which contains pharmacologically active compound (A) should be ultrasonicated and exposed to force.

The corresponding necessary application of force may also be applied to the mixture with the assistance of extruder rolls or calender rolls. Shaping of the pharmaceutical dosage forms preferably proceeds by direct press-forming of a pulverulent mixture of the components of the pharmaceutical dosage form or corresponding granules formed therefrom, wherein ultrasonication preferably proceeds during or before shaping. Such exposure continues until the polymer (C) has softened, which is conventionally achieved in less than 1 second to at most 5 seconds.

A suitable press is e.g. a Branson WPS, 94-003-A, pneumatical (Branson Ultraschall, Dietzenbach, Germany) having a plain press surface. A suitable generator (2000 W) is e.g.
a Branson PG-220A, 94-001-A analogue (Branson Ultraschall) with a sonotrode having a diameter of 12 mm. A die having a diameter of 12 mm may be used, the bottom of the die being formed by a bottom punch having a plain press-surface and a diameter of 12 mm.
Suitable parameters for plastification are frequency: 20 kHz; amplitude: 50%;
force: 250 N.
The effect of ultrasound and force by means of the sonotrode may be maintained for e.g.
0,5 seconds, and preferably both effects take place simultaneously.

Process variant 4:

In this process variant for the production of the pharmaceutical dosage form according to the invention, components (A), (C), optionally (B), such as antioxidants, plasticizers and/or delayed-release auxiliary substances, and optionally component (D), are processed with the assistance of a planetary-gear extruder to yield the pharmaceutical dosage form according to the invention.

Planetary-gear extruders are known and described inter alia in detail in Handbuch der Kunststoff-Extrusionstechnik I (1989) "Grundlagen" in Chapter 1.2 "Klassifizierung von Extrudern", pages 4 to 6. The corresponding description is hereby introduced as a reference and is deemed to be part of the disclosure.

The mixture is conveyed into the feed zone of the planetary-gear extruder. By heating at least to the softening point of component (C), the mixture is melted and the molten mixture is conveyed into the area of the central spindle, i.e. the extrusion zone, by the interaction of the central spindle 3 and the planetary spindles 4, further homogenized, compressed or compacted and extruded through the die 8 as an extruded strand or extruded strands, depending on how many bores the die comprises. The die geometry or the geometry of the bores is freely selectable. Thus, the die or the bores may exhibit a round, oblong or ovai cross-section, wherein the round cross-section preferably has a diameter of 0.1 mm to 15 mm and the oblong cross-section preferably has a maximum lengthwise extension of 21 mm and a crosswise extension of 10 mm. The extrusion die may also take the form of a slot die. Preferably, the die or the bores have a round, oval or oblong cross-section. Both the casing 6 of the planetary-gear extruder used according to the invention and the central spindle may be heated or cooled. The corresponding temperature control, i.e.
heating or cooling, is so arranged that the mixture to be extruded exhibits an average temperature corresponding to the softening temperature of component (C) and does not rise above a temperature at which component (A) to be processed may be damaged. Preferably, the temperature of the mixture to be extruded is adjusted to below 180 C, preferably below 150 C, but at least to the softening temperature of component (C).

After extrusion of the molten mixture and optional cooling of the extruded strand or extruded strands, the extrudates are singulated. This singulation may preferably be performed by cutting up the extrudates by means of revolving or rotating knives, water jet cutters, wires, blades or with the assistance of laser cutters.

Optionally after further cooling of the singulated extrudates, which are preferably present in the form of disks, they are optionally re-shaped into the final shape of the pharmaceutical dosage form, wherein they may be exposed to heat again if necessary.

This shaping for example into tablets may proceed in that the plastic extrudate is shaped with press-forming with the assistance of two contrarotating rolls preferably with mutually opposing recesses for plastification in the roll sleeve, the construction of which recesses determines the tablet shape.

However, it is also possible to form the tablets from the singulated extrudates in each case with the assistance of an optionally heated die and at least one shaping punch. To this end, the cylindrical granules obtained after singulation of the extruded strand may preferably be used. Apart from being press-formed into tablets, these granules or other multiparticulate shapes obtained, such as pellets or spheroids, may also be packaged into capsules in order to be used as a pharmaceutical dosage form produced according to the invention.

In a further preferred embodiment, the extruded strands extruded through a plurality of bores in the extrusion die may, after cooling thereof, optionally be brought together by interlacing or wrapping in the manner of rope production to yield a thicker strand than the individual extruded strands. This strand may optionally be further processed by soiveril.
attack with a suitable solvent or by heating to the softening point of the polymer (C) and optionally removing the solvent in accordance with the above-stated singulation and shaping of an individual strand.

If necessary, the planetary-gear extruder used may comprise not only an extrusion zone but also at least one further zone, so that the mixture to be extruded may optionally also be degassed.

The process according to the invention may be performed discontinuously or continuously, preferably continuously.

A suitable extruder, for example, is a planetary gear extruder type BCG 10 (LBB Bohle, Ennigerloh, Germany) having four planetary spindles and an extrusion die with bores having a diameter of 8 mm. A gravimetrical dosing of 3.0 kg/h is suitable. The extrusion may be performed, for example, at a rotational speed of 28,6 rmp and a product temperature of about 88 C.

Process variant 5:

This variant for the production of the pharmaceutical dosage form according to the invention is performed by processing at least the components (A), (C), optionally (B), such as antioxidants, plasticizers and/or delayed-release auxiliary substances, and optionally component (D), with addition of a solvent for component (C), i.e. for the polymer or polymers (C), to yield the pharmaceutical dosage form.

To this end, components (A), (C), optionally (B) and/or (D) are mixed and, after addition of the solvent and optionally after granulation, the resultant formulation mixture is shaped to yield the pharmaceutical dosage form.

Components (A), (C), optionally (B) and/or (D) are mixed in a mixer known to the person skilled in the art. The mixer may, for example, be a roll mixer, shaking mixer, shear mixer or compulsory mixer.

The solvent for the polymer (C) is added at least in such quantities that the formulation mixture is uniformly moistened.

Solvents which are suitable for the polymer (C) are preferably aqueous solvents, such as water, mixtures of water and aliphatic alcohols, preferably C, to C6 alcohols, esters, ethers, hydrocarbons, particularly preferably distilled water, short-chain alcohols, such as methanol, ethanol, isopropanol, butanol or aqueous alcohol solutions.

The solvent is preferably added with stirring. The uniformly moistened composition is then dried. Drying preferably proceeds with exposure to heat at temperatures at which it is possible to rule out any discoloration of the composition. This temperature may be established by simple preliminary testing.

Before or after drying, the composition may be divided into sub-portions which preferably in each case correspond to the mass of a unit of the pharmaceutical dosage form.
The corresponding dried portions are then shaped to yield the pharmaceutical dosage form.
This is preferably achieved by using tablet presses.

The formulation mixture may also be moistened in such a manner that, before addition of the solvent, the formulation mixture is divided, preferably in moulds, into sub-portions, is dispersed in a liquid dispersant with stirring and then the solvent is added.
Component (C) is not soluble in the dispersant, which must be miscible with the solvent.

Suitable dispersants are preferably hydrophilic solvents, such as aliphatic alcohols, ketones, esters. Short-chain alcohols are preferably used.

Alternatively, the formulation mixture may also be moistened in such a manner that the solvent is incorporated into the formulation mixture as a foam. Such a foam of the solvent is preferably produced with the assistance of a high-speed mixer, preferably with the addition of conventional foam stabilizers. Suitable stabilizers are, for example, hydrophilic polymers such as for example hydroxypropylmethylcellulose.

The foam is also preferably incorporated into the formulation mixture with stirring, a granulated composition so preferably being obtained.

Before or after being divided into sub-portions, which preferably correspond to the mass of a unit of the pharmaceutical dosage form, the granulated composition is dried and then shaped into the pharmaceutical dosage form.

Drying and shaping may preferably proceed as described above. The process according to the invention may also be performed in such a manner that solvent is added to the formulation mixture in such a quantity that a shapeable paste is obtained.

Before or after being dried, which may proceed as explained above, such a paste may be divided into sub-portions and the dried portions, after further division in each case into a portion corresponding to the mass of a unit of the pharmaceutical dosage form, are shaped or converted to yield the pharmaceutical dosage form.

It is here possible to form the sub-portions in the form of strands, which may be produced with the assistance of a screen or a strand former. The dried strands are preferably singulated and shaped to yield the pharmaceutical dosage form. This shaping preferably proceeds with the assistance of a tablet press, using shaping rollers or shaping belts equipped with rollers.

It is also possible to convert the paste into a planar structure and to stamp the pharmaceutical dosage form out of it once it has dried.

The paste is advantageously processed with an extruder, wherein, depending on the configuration of the extrusion, strands or planar structures articles are produced, which are singulated by chopping, cutting or stamping. The singulated sub-portions may be shaped, formed or stamped as described above to yield the pharmaceutical dosage form.
Corresponding apparatuses are known to the person skilled in the art.

The process according to the invention may here be performed continuously or discontinuously.

It is also possible to add solvent to the formulation mixture in such a quantity that at least the polymer component (C) is dissolved. Such a solution or dispersion/
suspension is preferably converted into a planar structure, an extruder with a flat die preferably being used or the solution being cast onto a planar support.

As stated above, after drying, the pharmaceutical dosage forms may be obtained from the planar structures by stamping or calendering. It is also possible, as stated above, to convert the solution into strands and to singulate these, preferably after they have been dried, and shape them to yield the pharmaceutical dosage form.

Alternatively, the solution may also be divided into portions such that, after drying, they each correspond to the mass of a unit of the pharmaceutical dosage form, with moulds which already correspond to the shape of the unit of the pharmaceutical dosage form preferably being used for this purpose.

If the solution is divided into any desired portions, the portions may, after drying, optionally be combined again and be shaped to form the pharmaceutical dosage form, being for example packaged in a capsule or press-formed to form a tablet.

The formulation mixtures combined with solvent are preferably processed at temperatures of 20 C to 40 C, wherein, apart from during drying to remove the solvent and the optionally present dispersant, no higher temperatures are used. The drying temperature must be selected below the decomposition temperature of the components. After shaping to yield the pharmaceutical dosage form, further drying corresponding to the above-described drying may optionally be performed.

Combinations of individual process steps of the above process variants are also possible in order to produce the pharmaceutical dosage form according to the invention.

Process variants 2 and 4 as described above involve the extrusion of a composition comprising components (A), (C), optionally (B) and/or (D). Preferably, extrusion is performed by means of twin-screw-extruders or planetary-gear-extruders, twin-screw extruders being particularly preferred.

The process for the preparation of the pharmaceutical dosage form according to the invention is preferably performed continuously. Preferably, the process involves the extrusion of a homogeneous mixture of components (A), (C), optionally (B) and/or (D). It is particularly advantageous if the obtained intermediate, e.g. the strand obtained by extrusion, exhibits uniform properties. Particularly desirable are uniform density, uniform distribution of the active compound, uniform mechanical properties, uniform porosity, uniform appearance of the surface, etc. Only under these circumstances the uniformity of the pharmacological properties, such as the stability of the release profile, may be ensured and the amount of rejects can be kept low.

Preferably, the process according to the present invention may be performed with less than 25% rejects, more preferably less than 20%, most preferably less than 15% and in particular less than 10% rejects, wherein the criteria for rejection are the FDA standards regarding the intervariability of the content of component (A), its release profile and/or the density of the pharmaceutical dosage form when comparing two pharmaceutical dosage forms, preferably taken from the same batch.

It has been surprisingly found that the above properties may be obtained by means of twin-screw-extruders and planetary-gear-extruders, twin-screw-extruders being particularly preferred.

Further, it has been surprisingly found that extrudates exhibiting an advantageous morphology are obtainable by means of planetary-gear-extruders and twin-screw-extruders.
It has been found that under suitable conditions the extrudate is surrounded by a shell which may be denoted as "extrusion skin". Said extrusion skin can be regarded as a collar-like or tubular structure forming a circumferential section of the extrudate about its longitudinal extrusion axis so that the outer surface of said collar-like or tubular structure forms the closed shell of the extrudate. Usually, only the front faces of the extrudate are not covered by said extrusion skin.

The extrusion skin surrounds the core of the extrudate in a collar-like or tubular arrangement and preferably is connected therewith in a seamless manner. The extrusion skin differs from said core in its morphology. Usually, the extrusion skin is visible with the naked eye in the cross-section of the extrudate, optionally by means of a microscope, since due to the different morphology of the material forming the extrusion skin and the material forming the core, the optical properties differ as well. It seems that during extrusion the material forming the extrusion skin is exposed to mechanical and thermal conditions differing from the conditions the core of the extrudate is exposed to. In consequence, a heterogeneous morphology of the extruded strand is obtained, which e.g.
assumes radial symmetry when an extrusion die having circular shape is used. The material forming the extrusion skin and the material forming the core are usually distinguished by their morphology, preferably, however, not by their composition, particularly not by the relative content of components (A), (C), optionally (B) and/or (D).

Usually the extrusion skin covers the entire shell of the extrudate like a one-piece collar, independently of what geometry has been chosen for the extrusion die.
Therefore, the extrudate may assume circular, elliptic or other cross-sections.

The extrusion skin is preferably characterized by a unitary thickness.
Preferably, the thickness of the extrusion skin is within the range from 0.1 to 4.0 mm, more preferably 0.15 to 3.5 mm, still more preferably 0.2 to 3.0 mm, most preferably 0.2 to 2.5 mm and in particular 0.2 to 2.0 mm. In a preferred embodiment the thickness of the extrusion skin in the sum over both opposing sides amounts to 0.5 to 50%, more preferably 1.0 to 40%, still more preferably 1.5 to 35%, most preferably 2.0 to 30% and in particular 2.5 to 25% of the diameter of the extrudate.

When the pharmaceutical dosage forms according to the invention are prepared by means of extrusion processes which lead to intermediates having an extrusion skin as described above, the pharmaceutical dosage forms obtained therefrom are preferably also characterized by a particular morphology.

In a preferred embodiment those regions, which have formed the extrusion skin in the extruded intermediate, are still visible with the naked eye, optionally by means of a microscope, in the cross-section of the pharmaceutical dosage form. This is because usually by further processing the extrudate, particularly by singulating and/or shaping, the different nature and thereby also the different optical properties of the material forming the extrusion skin and the material forming the core are maintained. In the following, that domain of the pharmaceutical dosage forms which has emerged from the extrusion skin in the course of further processing the extruded intermediate, will be denoted as "skin domain".

Preferably, the pharmaceutical dosage form according to the invention comprises a skin domain and a core located therein. Preferably, the skin domain is connected with the core in a seamless manner. Preferably the skin domain as well as the core have substantially the same chemical composition, i.e. substantially the same relative content of components (A), (C), optionally (B) and/or (D). The material forming the skin domain has a morphology differing from the material forming the core. Usually, this different morphology is also expressed in terms of different optical properties, so that the skin domain and the core are visible with the naked eye in the cross-section of the pharmaceutical dosage form.

In case that the pharmaceutical dosage form has been coated, e.g. by a film coating, the skin domain is located between the film coating and the core.

Since the pharmaceutical dosage form according to the invention may be obtained in different ways from the extrudate containing the extrusion skin (intermediate), the skin domain may take different arrangements and extensions within the pharmaceutical dosage form according to the invention. All arrangements have in common, however, that the skin domain partially covers the surface of the core, but usually not its entire surface. Preferably, two opposing surfaces of the core are not, or at least not fully covered by the skin domain.
In other words, preferably the skin domain has two openings/blanks on opposing sides.

The thickness of the skin domain may be uniform. It is also possible, however, that in the course of the processing, i.e. due to the subsequent shaping (e.g. press-forming) of the extrudate, various sections of the extrusion skin are expanded or compressed differently thereby leading to a variation of the thickness of the skin domain within the pharmaceutical dosage form.

Preferably the thickness of the skin domain is within the range from 0.1 to 4.0 mm, more preferably 0.15 to 3.5 mm, still more preferably 0.2 to 3.0 mm, most preferably 0.2 to 2.5 mm and in particular 0.2 to 2.0 mm.

The process according to the invention preferably involves the extrusion of a mixture of components (A), (C), optionally (B) and/or (D), preferably by means of a planetary-gear-extruder or a twin-screw-extruder. After extrusion the extrudate is preferably singulated, shaped and optionally coated in order to obtain the final pharmaceutical dosage form.

In a preferred embodiment of the process according to the invention, shaping is performed in the plasticized state of the mixture of components (A), (C), optionally (B) and/or (D). It has been surprisingly found that the extrusion of certain polymers (C), particular of high molecular weight polyethylene oxides, yields intermediates exhibiting some kind of memory effect: when the singulated extrudates are shaped at ambient temperature, e.g.
by press-forming, pharmaceutical dosage forms are obtained which tend to regain their original outer form upon storage under stressed storage conditions, i.e. they return to the form they had prior to shaping.

The shape of the pharmaceutical dosage form upon storage at stressed conditions, e.g. at 40 C/75% RH, may also be unstable for other reasons.

Said memory effect significantly deteriorates the storage stability of the pharmaceutical dosage form, as by regaining its outer form several properties of the pharmaceutical dosage form are changed. The same applies to any changes of the outer form due to other reasons.

It has been found that, for example, depending on the extrusion conditions a significant expansion of the strand may occur thereby resulting in an increase of the volume of the extrudate, i.e. a decrease of its density. Said expansion may be compensated by subsequently press-forming the singulated extrudate at a sufficient pressure, since under these conditions the expansion of the material may be reversed.

However, if press-forming has been performed at ambient temperature, the memory effect of the compressed extrudate will cause it to swell and to expand upon storage, thereby significantly increasing the volume of the pharmaceutical dosage form.

It has been surprisingly found that such memory effect may be suppressed if shaping of the singulated extrudate is performed at increased temperature, i.e. in the plasticized state of the mixture of components (A), (C), optionally (B) and/or (D). Preferably, shaping is performed at a pressure of at least 1 kN, more preferably within the range from 2 kN to 50 kN, e.g. by means of a tablet press. Preferably, shaping is performed at a temperature which preferably is about 40 C, more preferably about 30 C and in particular about 25 C
below the melting range of the mixture of components (A), (C), optionally (B) and/or (D).
The melting range of a given mixture may be determined by conventional methods, preferably by DSC (e.g. with a DSC model 2920 (TA Instruments, New Castle) and ultrahigh pure nitrogen as purge gas at a flow rate of 150 ml/min; approximate sample weight of 10-20 mg, sealed in nonhermetic aluminium pans; temperature ramp speed 10 C/min).

In a preferred embodiment the outer shape of the pharmaceutical dosage form according to the invention does not substantially change when being stored for at least 12 h, preferably for at least 24 h, at 40 C and 75% RH, preferably in an open container.

In a preferred embodiment the volume of the pharmaceutical dosage form according to the invention increases by not more than 20% or 17.5%, more preferably not more than 15% or 12.5%, still more preferably not more than 10% or 7.5%, most preferably not more than
6.0%, 5.0% or 4.0% and in particular not more than 3.0%, 2.0% or 1.0% when being stored for at least 12 h, preferably for at least 24 h, at a temperature of 20 C
below the melting range of the mixture of components (A), (C), optionally (B) and/or (D), optionally at a temperature of 40 C and 75% RH.

A further aspect of the invention relates to a packaging containing the pharmaceutical dosage form according to the invention and an oxygen scavenger. Suitable packages include blister packages and bottles, such as glass bottles or bottles made from thermoplastic polymers.

Oxygen scavengers and the application thereof in pharmaceutical packaging are known to the skilled artisan. In a preferred embodiment, the oxygen scavenger is selected from the group consisting of metal-catalyzed oxidizable organic polymers and anti-oxidants. It has been surprisingly found that the storage stability of the pharmaceutical dosage form can be increased when keeping the oxygen content of the atmosphere within the packaging low.
Methods for packaging pharmaceutical dosage forms and the application of suitable oxygen scavengers are known to the skilled artisan. In this regard it can be referred to e.g. D.A.
Dean, Pharmaceutical Packaging Technology, Taylor & Francis, 1st ed.; F.A.
Paine et al., Packaging Pharmaceutical and Healthcare Products, Springer, 1st ed.; and O.G.
Piringer et al., Plastic Packaging: Interactions with Food and Pharmaceuticals, Wiley-VCH, 2nd ed.
The pharmaceutical dosage form according to the invention is suitable to avoid various misuses, particularly - accidental misuse (e.g. unintentional);
- recreational misuse; and - experienced drug misuse.

A further aspect of the invention relates to the use of an opioid for the manufacture of the pharmaceutical dosage form as described avove for the treatment of pain.

A further aspect of the invention relates to the use of a pharmaceutical dosage form as described above for avoiding or hindering the abuse of the pharmacologically active compound (A) contained therein.

A further aspect of the invention relates to the use of a pharmaceutical dosage form as described above for avoiding or hindering the unintentional overdose of the pharmacologically active compound (A) contained therein.

In this regard, the invention also relates to the use of a pharmacologically active compound (A) as described above and/or a synthetic or natural polymer (C) as described above for the manufacture of the pharmaceutical dosage form according to the invention for the prophylaxis andior the treatment of a disorder, thereby preventing an overdose of the pharmacologically active compound (A), particularly due to comminution of the pharmaceutical dosage form by mechanical action.

Further, the invention relates to a method for the prophylaxis and/or the treatment of a disorder comprising the administration of the pharmaceutical dosage form according to the invention, thereby preventing an overdose of the pharmacologically active compound (A), particularly due to comminution of the pharmaceutical dosage form by mechanical action.
Preferably, the mechanical action is selected from the group consisting of chewing, grinding in a mortar, pounding, and using apparatuses for pulverizing conventional pharmaceutical dosage forms.

Preferred embodiments emb-1 to emb-81 according to the invention are displayed here below:

emb-1. A pharmaceutical dosage form, preferably a tablet, comprising at least one pharmaceutically active ingredient (pharmacologically active compound), and having a shape comprising a longitudinal axis and two opposite longitudinal edges, a transversal axis perpendicular to the longitudinal axis and two opposite transversal edges, a front side, an opposite back side and a circumferential rim between said front and back side, wherein the front side and/or the back side comprise a basis area and wherein the front side and/or the back side comprise at least one bulge which extends above said basis area, said at least one bulge being present at and/or adjacent to at least a section of one or both longitudinal edges and/or at and/or adjacent to at least a section of one or both transversal edges and/or between both longitudinal edges and both transversal edges.

emb-2. The pharmaceutical dosage form according to emb-1, which is a tablet having an oblong shape.

emb-3. The pharmaceutical dosage form according to emb-1 or 2, wherein the front side and the back side each comprise at least one bulge at least along a section at and/or adjacent to both longitudinal edges and/or at least along a section at and/or adjacent to both transversal edges.

emb-4. The pharmaceutical dosage form according to any of emb-1 to 3, wherein said front side and said back side comprise an at least essentially continuous bulge at an(i/nr adjacent to at least two third of both opposite longitudinal .ages vrrvvw .iynuu~ ucuycJ.

emb-5. The pharmaceutical dosage form according to any of emb-1 to 4, wherein one or both longitudinal edges are essentially straight over at least a major part of their length and/or wherein one or both transversal edges are curved over a major part of their length.

emb-6. The pharmaceutical dosage form according to any of emb-1 to 5, wherein the front side and/or the back side comprise an essentially circumferential bulge at and/or adjacent to the circumferential edge of the front side and/or the back side of said pharmaceutical dosage form.

emb-7. The pharmaceutical dosage form according to any of emb-1 to 6, wherein said pharmaceutical dosage form in its oblong form comprises at and/or adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of the front side at least one bulge, or wherein said pharmaceutical dosage form in its oblong form comprises at least one bulge at and/or adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of both the front side and the back side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form comprises in its oblong form a circumferential bulge at and/or adjacent to the circumferential edge of the front side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form in its oblong form comprises a circumferential bulge at and/or adjacent to the circumferential edge of both the front side and the back side of said pharmaceutical dosage form.

emb-8. The pharmaceutical dosage form according to emb-7, wherein said pharmaceutical dosage form in its oblong form comprises at major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of the front side at least one bulge, or wherein said pharmaceutical dosage form in its oblong form comprises at least one bulge at major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of both the front side and the back side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form comprises in its oblong form a circumferential bulge at the circumferential edge of the front side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form in its obiong form comprises a cir Cum i i erai ntial bulge at the circumferential edge of both the front side and the back side of said pharmaceutical dosage form.

emb-9. The pharmaceutical dosage form according to emb-7 or 8, wherein said pharmaceutical dosage form in its oblong form comprises adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of the front side at least one bulge, or wherein said pharmaceutical dosage form in its oblong form comprises at least one bulge adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of both the front side and the back side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form comprises in its oblong form a circumferential bulge adjacent to the circumferential edge of the front side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form in its oblong form comprises a circumferential bulge adjacent to the circumferential edge of both the front side and the back side of said pharmaceutical dosage form.

emb-10. The pharmaceutical dosage form according to any of emb-7 to 9, wherein said pharmaceutical dosage form in its oblong form comprises a circumferential bulge adjacent to the circumferential edge of both the front side and the back side of said pharmaceutical dosage form.

emb-11. The pharmaceutical dosage form according to any of emb-1 to 10, wherein the front side and/or the back side, in particular the basis area of the front side and/or the basis area of the back side comprise, in addition to at least one bulge on the front side and/or the back side, at least one indentation, in particular between opposite longitudinal and/or transversal bulges.

emb-12. The pharmaceutical dosage form according to emb-11, wherein the front and the back side comprise at least one indentation.

emb-13. The pharmaceutical dosage form according to emb-12, wherein the indentation(s) in the front side and the back side are at least once at least partially off-set.

emb-14. The pharmaceutical dosage form according to emb-12 or 13, wherein all the indentations of the front side and all the indentations of the back side are at least partially off-set.

emb-15. The pharmaceutical dosage form according to emb-12, wherein at least one indentation of the front side and at least one indentation of the back side are positioned in a congruent manner.

emb-16. The pharmaceutical dosage form according to emb-15, wherein all indentations of the front side and all indentations of the back side are positioned in a congruent manner.

emb-17. The pharmaceutical dosage form according to any of emb-11 to 16, wherein the indentations have a cylindrical or cube-like or cuboid-like shape or the shape of a half-sphere.

emb-18. The pharmaceutical dosage form according to any of emb-1 to 17, comprising one or more hydrophilic polymers.

emb-19. The pharmaceutical dosage form according to emb-18, wherein the one or more hydrophilic polymers are hydroxypropyl methyl cellulose and/or hydroxypropyl cellulose.

emb-20. The pharmaceutical dosage form according to emb-18 or 19 further comprising pregelatinized starch.

emb-21. The pharmaceutical dosage form according to any of emb-1 to 20, wherein the longitudinal length of the, in particular oblong, pharmaceutical dosage form does not exceed 30 mm.

emb-22. The pharmaceutical dosage form according to any of emb-1 to 21, having an average thickness over the basis areas of the front and the back side of about at least 1 mm, and in particular of no more than about 6 mm.

emb-23. The pharmaceutical dosage form according to any of emb-1 to 22, wherein the bulge extends perpendicular from the basis area of the front side and/or from the basis area of the hark aisle in average from about O .X; mm to ahoy it 2 mm emb-24. The pharmaceutical dosage form according to any of emb-1 to 23, having a length in the longitudinal direction in the range of about 5 mm to about 30 mm, in particular in the range of about 15 mm to about 25 mm, a width in the range of about 5 mm to about 15 mm, in particular in the range of about 7 mm to about mm, and a thickness over the basis areas in the range of about 1 mm to about 6 mm, in particular in the range of about 2 mm to about 4 mm.

emb-25. The pharmaceutical dosage form according to any of emb-1 to 24, further comprising at least partially a coating.

emb-26. Tamper-resistant pharmaceutical dosage form which has a retarded release profile for pharmaceutical application as an oral dosage form comprising at least one pharmaceutically active ingredient (pharmacologically active compound) with potential for abuse, and having a shape comprising a longitudinal axis and two opposite longitudinal edges, a transversal axis perpendicular to the longitudinal axis and two opposite transversal edges, a front side, an opposite back side and a circumferential rim between said front and back side, wherein the front side and/or the back side comprise a basis area and wherein the front side and/or the back side comprise at least one bulge which extends above said basis area, said at least one bulge being present at and/or adjacent to at least a section of one or both longitudinal edges and/or at and/or adjacent to at least a section of one or both transversal edges and/or between both longitudinal edges and both transversal edges.

emb-27. The pharmaceutical dosage form according to emb-26, which is a tablet having an oblong shape.

emb-28. The pharmaceutical dosage form according to emb-26 or 27, wherein the front side and the back side each comprise at least one bulge at least along a section at and/or adjacent to both longitudinal edges and/or at least along a section at and/or adjacent to both transversal edges.

emb-29. The pharmaceutical dosage form according to any of emb-26 to 28, wherein said front side and said back side comprise an at least essentially continuous bulge at and/or adjacent to at least two third of both opposite longitudinal edges.

emb-30. The pharmaceutical dosage form according to any of emb-26 to 29, wherein one or both longitudinal edges are essentially straight over at least a major part of their length and/or wherein one or both transversal edges are curved over a major part of their length.

emb-31. The pharmaceutical dosage form according to any of emb-26 to 30, wherein the front side and/or the back side comprise an essentially circumferential bulge at and/or adjacent to the circumferential edge of the front side and/or the back side of said pharmaceutical dosage form.

emb-32. The pharmaceutical dosage form according to any of emb-26 to 31;
wherein said pharmaceutical dosage form in its oblong form comprises at and/or adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of the front side at least one bulge, or wherein said pharmaceutical dosage form in its oblong form comprises at least one bulge at and/or adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of both the front side and the back side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form comprises in its oblong form a circumferential bulge at and/or adjacent to the circumferential edge of the front side of' said pharmaceutical dosage form, or wherein said pharmaceutical dosage form in its oblong form comprises a circumferential bulge at and/or adjacent to the circumferential edge of both the front side and the back side of said pharmaceutical dosage form.

emb-33. The pharmaceutical dosage form according to emb-32, wherein said pharmaceutical dosage form in its oblong form comprises at major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of the front side at least one bulge, or wherein said pharmaceutical dosage form in its oblong form comprises at least one bulge at major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of both the front side and the back side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form comprises in its oblong form a circumferential bulge at the circumferential edge of the front side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form in its oblong form comprises a circumferential bulge a-__ -----.-1]-1 -J-- Of Loth the u IG J: a-_ui...n J ..:.J .. ai...w .J t ui.iG ... L LJac . side .. vi sa ..
at 1110 VII 1:U1111G1 CI 1ui11 Cult'. C ul both JiuG d iu pharmaceutical dosage form.

emb-34. The pharmaceutical dosage form according to emb-32, wherein said pharmaceutical dosage form in its oblong form comprises adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of the front side at least one bulge, or wherein said pharmaceutical dosage form in its oblong form comprises at least one bulge adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of both the front side and the back side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form comprises in its oblong form a circumferential bulge adjacent to the circumferential edge of the front side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form in its oblong form comprises a circumferential bulge adjacent to the circumferential edge of both the front side and the back side of said pharmaceutical dosage form.

emb-35. The pharmaceutical dosage form according to emb-32, wherein said pharmaceutical dosage form in its oblong form comprises a circumferential bulge adjacent to the circumferential edge of both the front side and the back side of said pharmaceutical dosage form.

emb-36. The pharmaceutical dosage form according to any of emb-26 to 35, wherein the front side and/or the back side, in particular the basis area of the front side and/or the basis area of the back side comprise, in addition to at least one bulge on the front side and/or the back side, at least one indentation, in particular between opposite longitudinal and/or transversal bulges.

emb-37. The pharmaceutical dosage form according to emb-36, wherein the front and the back side comprise at least one indentation.

emb-38. The pharmaceutical dosage form according to emb-37, wherein the indentation(s) in the front side and the back side are at least once at least partially off-set.

emb-39. The pharmaceutical dosage form according to emb-37 or 38, wherein all the indentations of the front side and all the indentations of the back side are at least partially off-set.

emb-40. The pharmaceutical dosage form according to emb-36, wherein at least one indentation of the front side and at least one indentation of the back side are positioned in a congruent manner.

emb-41. The pharmaceutical dosage form according to emb-40, wherein all indentations of the front side and all indentations of the back side are positioned in a congruent manner.

emb-42. The pharmaceutical dosage form according to any of emb-36 to 41, wherein the indentations have a cylindrical or cube-like or cuboid-like shape or the shape of a half-sphere.

emb-43. The pharmaceutical dosage form according to any of emb-26 to 42 comprising a polyalkylene oxide.

emb-44. The pharmaceutical dosage form according to any of emb-26 to 43 comprising at least one cellulose ether derivative.

emb-45. The pharmaceutical dosage form according to emb-44, wherein the cellulose ether derivative is HPMC.

emb-46. The pharmaceutical dosage form according to any of emb-43 to 45, wherein the polyalkylene oxide is polyethylene oxide.

emb-47. The pharmaceutical dosage form according to emb-46, wherein the polyethylene oxide has a molecular weight in the range of about 2.000.000 to about
7.000.000.
emb-48. The pharmaceutical dosage form according to any of emb-26 to 47, wherein the pharmaceutical dosage form further comprises a polyalkylene glycol.

emb-49. The pharmaceutical dosage form according to emb-48, wherein the polyalkylene glycol is polyethylene glycol.

emb-50. The pharmaceutical dosage form according to any of emb-26 to 49, wherein the pharmaceutically active ingredient (pharmacologically active compound) with nntantial fnr 2hi igg is n nnin-killinn rin in in nnrtini filar nn nniniri nr nn nniniri derivative.

emb-51. The pharmaceutical 'dosage form according to emb-50, wherein the pharmaceutically active ingredient (pharmacologically active compound) is tapentadol.

emb-52. The pharmaceutical dosage form according to any of emb-26 to 51, comprising at least one pharmaceutically active ingredient (pharmacologically active compound) with potential for abuse, in particular an analgesic compound; at least one poly(alkylene oxide), in particular poly(ethylene oxide); at least one cellulose ether derivative, in particular hydroxypropyl methylcellulose (HPMC); and at least one polyalkylene glycol, in particular polyethylene glycol; and optionally an additive.

emb-53. The pharmaceutical dosage form according to emb-52, wherein the active ingredient with potential for abuse is present in an amount of at least 5 weight percent; poly(alkylene oxide), in particular poly(ethylene oxide) is present in an amount of at least 15 weight percent; cellulose ether derivative, in particular HPMC, is present in an amount of at least 5 weight percent; and polyalkylene glycol, in particular polyethylene glycol, is present in an amount of at least weight percent.

emb-54. The pharmaceutical dosage form according to any of emb-26 to 53, wherein the longitudinal length of the, in particular oblong, pharmaceutical dosage form does not exceed 30 mm.

emb-55. The pharmaceutical dosage form according to any of emb-26 to 54, having an average thickness over the basis areas of the front and the back side of about at least 1 mm, and in particular of no more than about 6 mm.

emb-56. The pharmaceutical dosage form according to any of emb-26 to 55, wherein the bulge extends perpendicular from the basis area of the front side and/or from the basis area of the back side in average from about 0.5 mm to about 2 mm.

emb-57. The pharmaceutical dosage form according to any of emb-26 to 56, having a length in the longitudinal direction in the range of about 5 mm to about 30 mm, in =cuiGr I F about i ~ 15j + abo + 2r, mm u width in the range of t.1QI.1lIl.III the IG rQilyc of about ~ini~ to uvvua w IInII , U-about 5 mm to about 15 mm, in particular in the range of about 7 mm to about mm, and a thickness over the basis areas in the range of about 1 mm to about 6 mm, in particular in the range of about 2 mm to about 4 mm.

emb-58. The pharmaceutical dosage form according to any of emb-26 to 57, wherein said pharmaceutical dosage form is a monolith, in particular formed from an extruded monolithic mass.

emb-59. The pharmaceutical dosage form according to any of emb-26 to 58, which has been prepared via compression using a die and a punch from a monolithic mass obtained by melt extrusion.

emb-60. The pharmaceutical dosage form according to emb-58 or 59, wherein said pharmaceutical dosage form has been obtained by compressing a melt extruded monolithic mass which exhibits ambient temperature.

emb-61. The pharmaceutical dosage form according to any of emb-58 to 60, wherein the extruded monolithic mass has been cut prior to compression.

emb-62. The pharmaceutical dosage form according to any of emb-58 to 61, wherein melt extrusion has been conducted with a twin-screw extruder.
emb-63. The pharmaceutical dosage form according to any of emb-1 to 62, further comprising at least partially a coating.

emb-64. A pharmaceutical dosage form with controlled release of a pharmacologically active compound (A) contained therein, the pharmaceutical dosage form having a breaking strength B1 of at least 500 N in direction of extension El and having a breaking strength B2 of less than 500 N in direction of extension E2.

emb-65. The pharmaceutical dosage form according to emb-64, wherein E, is orthogonal to E2.

emb-66. The pharmaceutical dosage form according to emb-64 or 65, wherein E2 is the main direction of extension of the pharmaceutical dosage form.

emb-67. The pharmaceuiicai dosage form according to any of emb-64 to 66, ::'hick provides fragments when exerting a force higher than B2 in direction of extension E2, said fragments in turn having a breaking strength of at least 500 N in any of their directions of extension.

emb-68. The pharmaceutical dosage form according to emb-67, wherein the volume of each fragment is at least 5% of the volume of the pharmaceutical dosage form.
emb-69. The pharmaceutical dosage form according to any of emb-64 to 68, wherein the total surface S of the pharmaceutical dosage form in mm2 satisfies the following requirement:

S>A=W3, wherein A is 4.5 and W is the total weight of the pharmaceutical dosage form in mg.

emb-70. The pharmaceutical dosage form according to emb-69, wherein A is 6.2.

emb-71. The pharmaceutical dosage form according to any of emb-64 to 70, which is deformed when exerting a force in direction of extension E2 so that, when the amount of force reaches the breaking strength B2, deformation causes tractive forces that lead to disruption of the dosage form.

emb-72. The pharmaceutical dosage form according to any of emb-64 to 71, which has released under in vitro conditions after 0.5 h 1.0 to 35 wt.-%, after 1 h 5.0 to 45 wt.-%, after 2 h 10 to 60 wt.-%, after 4 h at least 15 wt.-%, after 6 h at least 20 wt.-%, after 8 h at least 25 wt.-% and after 12 h at least 30 wt.-%

of the pharmacologically active compound (A) that was originally contained in the pharmaceutical dosage form.

emb-73. The pharmaceutical dosage form according io any of emb-64 to 72, which is not radial symmetric about its principal direction of extension.

emb-74. The pharmaceutical dosage form according to any of emb-64 to 73, wherein the maximum extension of the dosage form orthogonal to the main area of extension of the dosage form is spaced from the centre of mass of the dosage form parallel to said main area of extension.

emb-75. The pharmaceutical dosage form according to any of emb-64 to 74, wherein a portion of its surface is convex, another portion of its surface is concave.

emb-76. The pharmaceutical dosage form according to any of emb-64 to 75, which contains a polymer (C) having a weight average molecular weight of at least 100.000 g mol-1.

emb-77. The pharmaceutical dosage form according to emb-76, wherein the polymer (C) comprises a polyalkylene oxide.

emb-78. The pharmaceutical dosage form according to any of emb-64 to 77, wherein - a portion of the surface of the pharmaceutical dosage form is convex, another portion of its surface is concave; and - the pharmacologically active compound (A) is a psychotropically acting substance; and - the content of the pharmacologically active compound (A) is at least 0.5 wt.-%, based on the total weight of the dosage form; and - polymer (C) is an polyalkylene oxide having a weight average molecular weight of at least 200,000 g/mol; and - the content of polymer (C) is at least 15 wt.-%, based on the total weight of the dosage form; and - the pharmacologically active compound (A) is embedded in the polymer (C);
and - the pharmaceutical dosage form is adapted for oral administration once daily or twice daily; and - the pharmaceutical dosage form is thermoformed.

emb-79. Use of a pharmaceutical dosage form according to any one of emb-1 to 25 as an immediate release or a retarded release oral dosage form.

emb-80. Use of a pharmaceutical dosage form according to any one of emb-26 to 78 as a tamper-resistant or break-resistant retarded release oral dosage form.

emb-81. A method of treating pain in a patient in need of such treatment, said method comprising administering to said patient an effective amount therefor of a pharmaceutical dosage form according to any one of emb-1 to 78, wherein the pharmaceutically active ingredient, the pharmaceutically active ingredient for abuse and the pharmacologically active compound (A) are each selected from the group consisting of analgesics.

The invention is explained below with reference to examples. These explanations are given merely by way of example and do not limit the general concept and scope of the invention.
Example 1:

Formulation examples A. Formulation examples for retarded release formulations Active ingredient(s) 0.01-50% (w/w) Viscous hydrophilic polymer(s) 0.01-80% (w/w) Pregelatinized starch 5-80% (w/w) Pharmaceutically acceptable formulating agents ad 100% (w/w).
Active ingredient(s) 0.01-50% (w/w) Viscous hydrophilic polymer(s) comprising 0.01-80% (w/w) hydroxypropyl cellulose Pregelatinized starch 5-80% (w/w) Pharmaceutically acceptable formulating agents ad 100% (w/w).
Active ingredient(s) 0.01-50% (w/w) Viscous hydrophilic polymer(s) 0.01-80% (w/w) Pregelatinized starch 5-15% (w/w) Pharmaceutically acceptable formulating agents ad 100% (w/w).

Active ingredient(s) 0.01-50% (w/w) Viscous hydrophilic polymer(s) comprising 0.01-80% (w/w) hydroxypropyl cellulose C COI /===/...) Pregelatinized starch 0 1 J /O IVY, VV, Pharmaceutically acceptable formulating agents ad 100% (w/w).
Active ingredient(s) 0.01-50% (w/w) Viscous hydrophilic polymer(s) 0.01-80% (w/w) Pregelatinized starch 5% (w/w) Pharmaceutically acceptable formulating agents ad 100% (w/w).
Active ingredient(s) 0.01-50% (w/w) Viscous hydrophilic polymer(s) comprising 0.01-80% (w/w) hydroxypropyl cellulose Pregelatinized starch 5% (w/w) Pharmaceutically acceptable formulating agents ad 100% (w/w).
Active ingredient(s) 0.01-50% (w/w) Hydroxypropyl cellulose 25-62% (w/w) Hydroxypropyl methylcellulose 0-16% (w/w) Pregelatinized starch 5-15% (w/w) Pharmaceutically acceptable formulating agents ad 100% (w/w).

The above formulations can generally be prepared according to the following process:
(1.a) one or more active ingredients, pregelatinized starch, one or more viscous hydrophilic polymers and optionally some or all of the pharmaceutically acceptable formulating agents are mixed;
(1.b) the powder mixture prepared under (1.a) is run through a compactor, thus yielding plates;
(1.c) the resulting plates are broken down and sieved, thus yielding granules;
(1.d) the resulting granules are optionally mixed with all or the remainder of the pharmaceutically acceptable formulating agents, preferably until homogeneous.

In case the active ingredient(s) is a sparingly water soluble, slightly water soluble, very slightly water soluble, practically water insoluble or water insoluble drug or a drug with a pH
dependent solubility, in particular an alkaline drug, the active ingredient(s) can in one embodiment be incorporated in the controlled release formulation as an intimate mixture with a cyclodextrin or derivatives thereof or another water soluble polymer, as described hereinabove. In said case, the preparation of the present controlled release formulation comprises an additional first step, namely (2.a) one or more active ingredients and the wafer soiubie poiyiiier are iiitir ately mixed;
(2.b) the intimate mixture prepared under (2.a) is mixed with pregelatinized starch, one or more viscous hydrophilic polymers and optionally some or all of the pharmaceutically acceptable formulating agents;
(2.c) the powder mixture prepared under (2.b) is run through a compactor, thus yielding plates;
(2.d) the resulting plates are broken down and sieved, thus yielding granules;
(2.e) the resulting granules are optionally mixed with all or the remainder of the pharmaceutically acceptable formulating agents, preferably until homogeneous.

The formulation obtained by the processes as described hereinabove in one embodiment can then be compressed in a tablet according to the invention by tabletting in a tabletting machine with punches and dies adapted for the tablet of the invention the final blend resulting from the above described processes, i.e. the blend resulting under (1.d) or (2.e).

A compactor as mentioned in step (1.b) or (2.c) of the above described processes is an apparatus wherein the powdery mixture is run between two rollers exerting pressure on the powdery mixture. In this way the mixture is compacted and sheets or plates are formed.
Compactors are commercially available, for instance, from the company Gerteis (Jona, Swiss), e.g. a Polygran 3W compactor.

The above general route of preparation of the controlled release formulation may be modified by a person skilled in the art by for instance adding certain ingredients at other stages than indicated above.

As an alternative to the above described route of preparation involving a compaction step, the above described mixture can also be tabletted using direct compression.
When using the technique of direct compression, dies or matrices in the form of the desired tablets of the invention are filled with a powdery mixture having the tablet composition and then are punched with punches adapted for the desired tablet. The advantage of this way of tabletting is that it usually requires less steps. Apparatuses for direct compression tabletting are known to the skilled parson. These apparatuses require forced feeding systems whenever the rheological properties of the mixture are not appropriate to fill the dies or matrices without forced feeding.

Example of a retarded release formulation according to the invention Cisapride-(L)-tartrate 52.92 mg Lactose monohydrate 200 mesh 149.43 mg Hydroxypropyl methylcellulose 2208 74.i mg Hydroxypropyl cellulose 228.00 mg Drum dried waxy maize starch 28.5 mg Magnesium stearate 2.85 mg Colloidal anhydrous silica 5.7 mg Talc 28.5 mg Preparation of the above powder mixture Cisapride-(L)-tartrate, hydroxypropyl methylcellulose, hydroxypropyl cellulose, drum dried waxy maize starch, the lactose filler, are mixed in a planetary mixer, and than compacted using a dry compactor. The compact is broken down, sieved and mixed in a planetary mixer with colloidal anhydrous silica. Magnesium stearate is added and mixed.

The above mixture can then be compressed into a tablet according to the invention.
B. Formulation examples for immediate release formulations A mixture of 100 g of active ingredient(s), 570 g lactose and 200 g starch is mixed well and thereafter humidified with a solution of 5 g sodium dodecyl sulfate and 10 g polyvinyl-pyrrolidone (Kollidon-K 90 ) in about 200 ml of water. The wet powder mixture is sieved, dried and sieved again. Then there are added 100 g microcrystalline cellulose (Avicel ) and 15 g hydrogenated vegetable oil (Sterotex ). The whole is mixed well.

The above mixture can then be compressed into tablets according to the invention, giving 10.000 tablets, each comprising 10 mg of the active ingredient.

Example 2:

Pharmaceutical dosage forms were manufactured from the following compositions:
1-1 (#1) 1-2 (#2) C-1 tapentadol HCI 291.20 291.20 291.20 PEO Mw 7 Mio g/mol 245.00 245.00 PEO Mw 5 Mio g/mol 247.70 polyethylene glycole 6000 65.10 65.10 65.00 HPMC 100,000 mPas 98.00 98.00 45.50 a-tocopherole 0.70 0.70 0.65 SUM 700.00 700.00 650.00 tablet format 9x21 HO 9x21 HO 9x21 oblong 291.20 mg tapentadol HCI correspond to 250 mg tapentadol free base.

General procedure:

The polyethylene oxide was melted at 90 C and the total amount of a -tocopherol was dissolved therein. Then, tapentadol and hydroxypropyl methyl cellulose (HPMC) were mixed in a fast mixer for 5 min at a temperature of 70 C. The melt of polyethylene oxide and a-tocopherole was added to the mixture dropwise within 10 minutes. The thus obtained granulate was sieved on a taper sieving machine and then mixed in a free fall mixer for 15 min. The powder mixture was dosed gravimetrically to an extruder. Extrusion was performed by means of a twin screw extruder of type Micro 27 GL 40 D
(Leistritz, Nurnberg, Germany) that was equipped with a heatable round die having a diameter of 8 mm.

The following extrusion conditions were adjusted:
Number of revolutions of screws: 100 Upm Throughput: 4 kg/h Product temperature: 120 C
Shell temperature: 100 C

The hot extrudate was cooled on a conveyor belt and the cooled extrusion strand was comminuted to cut pieces weighing 700 mg each. The cut pieces were shaped by means of an excenter press. The tablets of inventive examples I-1 and 1-2 were shaped by means of an H-plunger (9*21 HO, type 21x9 WR 1.9 with "inner courtyard", Notter, (5lbronn-Durrn, Germany) and the tablets of comparative example C-1 were shaped by means of a conventional oblong plunger.

The tablets of inventive examples I-1 and 1-2 had the following dimensions (base height H
according to Figure 9B); width, height and length a, b and c according to Figure 10) (average values n = 10):

a 9.06 a, 3.17 The manufacturer of the HO-plunger provides the following formula for the a2 2.72 surface and the volume of the shaped tablets as a function of the base height a3 3.17 (H):
b 4.52 b, 0.99 volume = 94.3 + 171.6 x H [mm3] = 638.3 mm3 b2 2.54 b3 0 99 surface = 382 + 52.3 x H [mm2] = 547.8 mm2 c 20.49 c, 3.26 c2 13.97 c3 3.26 H 3.17 In comparison, the surface of the pharmaceutical dosage form according to comparative example C-1 was estimated to be about 459 mm2 (estimation in accordance with Eudragit Application Guidelines, 10th edition, 07/2007, Rohm GmbH, Darn istadt, page 25).

The correlation of the pharmaceutical dosage forms according to inventive examples I-1 and 1-2 and according to comparative example C-1 with the requirement S > A =
W213 are summarized in the following table:

weight W 700 m 650 mg surface S 548 mm2 459 mm2 S / W 0.783 0.706 A [for S=A=W2' 6.95 6.12 Example 3:

The dissolution profile of the tablets was investigated under the following conditions: Paddle apparatus equipped with sinker, 50 rpm, 37 5 C, 900 mL simulated intestinal fluid pH 6.8 (phosphate buffer).

The results are displayed in Figures 21 to 24.

Figure 21 shows that immediately after manufacture the release profile of the tablet according to inventive example 1-2 (H-shape) is comparable to the release profile of the tablet according to comparative example C-1 (oblong).

Figure 22 shows that the release profile of the tablet according to inventive example I-1 (H-shape) is comparable to the release profile of the tablet according to inventive example 1-2 (H-shape), i.e. that different batches provide reproducible results.

Figure 23 shows that the release profile of the tablet according to comparative example C-1 (oblong) changes upon storage (40 C, 6 months; 25 C 9 months; and 30 C 9 months, respectively).

Figure 24 shows that the release profile of the tablet according to inventive example 1-2 (H-shape) is stable, i.e. does not change upon storage (40 C, 6 months; 25 C 9 months; and 30 C 9 months, respectively).

These experimental findings demonstrate that the pharmaceutical dosage forms according to the invention have a storage stability in terms of the release profile that is better than the storage stability of comparative pharmaceutical dosage forms.

Example 4 In accordance with example 2, pharmaceutical dosage forms were manufactured from the following compositions:

m C-2 C-3 C-4 1-3 1-4 tapentadol HCI 58.24 116.48 174.72 232.96 291.20 PEO Mw 7 Mio g/mol 225.16 187.12 166.83 260.39 245.00 polyethylene I cote 6000 60.00 40.00 45.00 65.00 65.10 HPMC 100,000 mPas 56.00 56.00 63.00 91.00 98.00 a-tocopherole 0.60 0.40 0.45 0.65 0.70 tablet format oblong oblong oblong H-shape H-shape 7x17 mm 7x17 mm 7x17 mm 9x21 mm 9x21 mm Furthermore, two comparative H-shaped tablets (9x21 mm) containing 232.96 mg tapentadol HCI (C-5) and 291.20 mg tapentadol HCI (C-6) were manufactured without extrusion from conventional excipients not comprising polyalkylene oxide of high molecular weight.

The breaking strength of the pharmaceutical dosage forms was measured by means of a Sotax HT100 at a constant speed of 120 mm/min. A tablet was regarded as failing the breaking strength test when during the measurement the force dropped below the threshold value of 25% of the maximum force that was observed during the measurement, regardless of whether the dosage form was fractured into separate pieces or not.

All values are given as mean of 10 measurements (n = 10).

The results of the breaking strength measurements are summarized in the table here below:
form type of jaws direction breaking according to strength C-2 oblong plain Figure 1813) > 500 N
C-3 oblong plain Figure 1813) > 500 N
C-4 oblong plain Figure 18B) > 500 N
C-5 H-shape plain Figure 1813) 188 N
C-6 H-shape plain Figure 1813) 188 N
1-3 H-shape plain Figure 1813) 349 N

1-4 H-shape plain Fiore 18B) 399 N
1-3 H-shape embossment/ indentation Figure 20A) > 500 N
1-4 H-shape embossment/ indentation Figure 20A) > 500 N
1-3 H-shape embossment/ indentation Figure 20C) > 500 N
1-4 H-shape embossment/ indentation Figure 20C) > 500 N

The above breaking strength data demonstrate that the dosage forms according to the invention 1-3 and 1-4 exhibit a breaking strength of more than 500 N (B1) when measured in accordance with Figure 20A) and 20C) (E,), even when equipping the tester jaws with embossment and indentation, whereas they exhibit a breaking strength of less than 500 N
(B2) when measured in accordance with Figure 18B) (E2).

Nonetheless, the breaking strength (B2) of the dosage forms according to the invention 1-3 and 1-4 when measured in accordance with Figure 18B) (E2) is still much higher than that of H-shaped dosage forms that are manufactured from conventional excipients without extrusion (C-5 and C-6).

Still further, comparative oblong tablets having an increased breaking strength, such as described in W02005/016314 or W02005/016313 (C-2, C-3, and C-4), exhibit a breaking strength of more than 500 N when measured in accordance with Figure 18B). Such oblong tablets exhibit a breaking strength of at least 500 N in each and every direction of extension and thus, are distinguished from the pharmaceutical dosage forms according to the invention.

Example 5 Tablets eq200mg Tapentadol were prepared by extrusion (3.5 kg/h) in two different tablet shapes. The compositions were practically identical (minimal variation in a tocopherol), and had the same weight.

Composition of the tablets eq200mg Tapentadol (in mg):

tablets 5.1 5.2 Tapentadol HCL 232.96 mg 34.9% 232.96 mg 34.9%
PEO Mw 5 Mio 300.38 mg 45% 300.38 mg 45%
HPMC 100 000 66.75 mg 10% 66.76 mg 10%
PEG 66.58 mg 9.975% 66.76 mg 10%
a tocopherol 0.83 mg 0.125% 0.66 mg 0.1%
tablet weight 667.5 mg 667.5 mg I

Percentage drug released for the Tapentadol eq200mg tested in dissolution at 50 rpm paddle speed, USP apparatus II, USP buffer pH 6.8:

5.1 5.2 standard shape HO shape average average 30 min 10.8 14.9 120 min 29.8 34.5 240 min 44.3 51.3 600 min 77.3 84.8 Example 6 Tablets eq250mg Tapentadol were prepared by extrusion (3.5 kg/h) in different tablet shapes. The composition is shown in the next table.

Composition of the eg250mg Tapentadol tablets (in mg):
Experiment 6 Tapentadol HCL 291.20 mg 34.9%
PEO Mw 5 Mio 375.47 mg 45%
HPMC 100 000 83.44 mg 10%
PEG 83.44 mg 10%
a tocopherol 0.83 mg 0.1%
tablet weight 834.38 mg A first set of comparative tablets was shaped by means of a conventional oblong plunger, 9x21 mm and a second set of inventive tablets was shaped by means of an H-plunger (22.6*8.6 H1, type CPS Batonett, C7084-1, Elisabeth EPMO, France).

Percentage drug released for the Tapentadol eg250mg tested in dissolution at 50 rpm paddle speed, USP apparatus II, USP buffer pH 6.8:

standard shape 9x21 mm) 11-sha average avers e 30 min 12.0 14.1 120 min 29.3 32.2 240 min 46.2 48.2 600 min 78.7 79.

Claims (59)

1. Pharmaceutical dosage form with controlled release of a pharmacologically active compound (A) contained therein, the pharmaceutical dosage from having a breaking strength B1 of at least 500 N in direction of extension E1 and having a breaking strength B2 of less than 500 N in direction of extension E2.
2. Pharmaceutical dosage form according to claim 1, wherein E1 is orthogonal to E2.
3. Pharmaceutical dosage form according to claim 1 or 2, wherein E2 is the main direction of extension of the pharmaceutical dosage form.
4. Pharmaceutical dosage form according to any of the preceding claims, which is a tablet.
5. Pharmaceutical dosage form according to any of the preceding claims, which has an oblong shape.
6. Pharmaceutical dosage form according to any of the preceding claims, which provides fragments when exerting a force higher than B2 in direction of extension E2, said fragments in turn having a breaking strength of at least 500 N in any of their directions of extension.
7. Pharmaceutical dosage form according to claim 6, wherein the volume of each fragment is at least 5% of the volume of the pharmaceutical dosage form.
8. Pharmaceutical dosage form according to any of the preceding claims, wherein the total surface S of the pharmaceutical dosage form in mm2 satisfies the following requirement:
wherein A is 4.5 and W is the total weight of the pharmaceutical dosage form in mg.
9. Pharmaceutical dosage form according to claim 8, wherein A is 6.2.
10. Pharmaceutical dosage form according to any of the preceding claims, which is deformed when exerting a force in direction of extension E2 so that, when the amount of force reaches the breaking strength B2, deformation causes tractive forces that lead to disruption of the dosage form.
11. Pharmaceutical dosage form according to any of the preceding claims, which has released under in vitro conditions after 0.5 h 1.0 to 35 wt.-%, after 1 h 5.0 to 45 wt.-%, after 2 h 10 to 60 wt.-%, after 4 h at least 15 wt.-%, after 6 h at least 20 wt.-%, after 8 h at least 25 wt.-% and after 12 h at least 30 wt.-%

of the pharmacologically active compound (A) that was originally contained in the pharmaceutical dosage form.
12. Pharmaceutical dosage form according to any of the preceding claims, which is not radial symmetric about its principal direction of extension.
13. Pharmaceutical dosage form according to any of the preceding claims, wherein the maximum extension of the dosage form orthogonal to the main area of extension of the dosage form is spaced from the centre of mass of the dosage form parallel to said main area of extension.
14. Pharmaceutical dosage form according to any of the preceding claims, further comprising at least partially a coating.
15. Pharmaceutical dosage form according to any of the preceding claims, which contains a polymer (C).
16. Pharmaceutical dosage form according to claim 15, wherein polymer (C) has a weight average molecular weight of at least 100.000 g mol-1.
17. Pharmaceutical dosage form according to claim 15 or 16, wherein polymer (C) is hydrophilic.
18. Pharmaceutical dosage form according to any of claims 15 to 17, wherein the polymer (C) comprises a polyalkylene oxide and/or at least one cellulose ether derivative.
19. Pharmaceutical dosage form according to claim 18, wherein the cellulose ether derivative is selected from the group consisting of hydroxypropyl methyl cellulose and/or hydroxypropyl cellulose.
20. Pharmaceutical dosage form according to claim 18, wherein the polyalkylene oxide is polyethylene oxide.
21. Pharmaceutical dosage form according to claim 20, wherein the polyethylene oxide has a molecular weight in the range of about 500.000 g/mol to about 15.000.000 g/mol.
22. Pharmaceutical dosage form according to any of claims 15 to 21 further comprising pregelatinized starch.
23. Pharmaceutical dosage form according to any of claims 15 to 22, which further comprises a polyalkylene glycol.
24. Pharmaceutical dosage form according to claim 23, wherein the polyalkylene glycol is polyethylene glycol.
25. Pharmaceutical dosage form according to any of the preceding claims, which is tamper resistant and wherein the pharmacologically active compound (A) has potential for abuse.
26. Pharmaceutical dosage form according to claim 25, wherein the pharmacologically active compound (A) with potential for abuse is an opioid or an opioid derivative.
27. Pharmaceutical dosage form according to claim 26, wherein the pharmacologically active compound (A) is tapentadol.
28. Pharmaceutical dosage form according to any of the preceding claims, comprising at least one pharmacologiaclly active compound (A) with potential for abuse, in particular an analgesic compound; at least one poly(alkylene oxide), in particular poly(ethylene oxide); at least one cellulose ether derivative, in particular hydroxypropyl methylcellulose (HPMC); and at least one polyalkylene glycol, in particular polyethylene glycol; and optionally an additive.
29. Pharmaceutical dosage form according to claim 28, wherein the active ingredient with potential for abuse is present in an amount of at least 5 weight percent;
poly(alkylene oxide), in particular poly(ethylene oxide) is present in an amount of at least 15 weight percent; cellulose ether derivative, in particular HPMC, is present in an amount of at least 5 weight percent; and polyalkylene glycol, in particular polyethylene glycol, is present in an amount of at least 5 weight percent.
30. Pharmaceutical dosage form according to any of the preceding claims, wherein a portion of its surface is convex, another portion of its surface is concave.
31. Pharmaceutical dosage form according to any of the preceding claims, wherein - a portion of the surface of the pharmaceutical dosage form is convex, another portion of its surface is concave; and - the pharmacologically active compound (A) is a psychotropically acting substance; and - the content of the pharmacologically active compound (A) is at least 0.5 wt.-%, based on the total weight of the dosage form; and - polymer (C) is an polyalkylene oxide having a weight average molecular weight of at least 200,000 g/mol; and - the content of polymer (C) is at least 15 wt.-%, based on the total weight of the dosage form; and - the pharmacologically active compound (A) is embedded in the polymer (C);
and - the pharmaceutical dosage form is adapted for oral administration once daily or twice daily; and - the pharmaceutical dosage form is thermoformed.
32. Pharmaceutical dosage form according to any of the preceding claims, which is a monolith, in particular formed from an extruded monolithic mass.
33. Pharmaceutical dosage form according to any of the preceding claims, which has been prepared via compression using a die and a punch from a monolithic mass obtained by melt extrusion.
34. Pharmaceutical dosage form according to claim 32 or 33, which has been obtained by compressing a melt extruded monolithic mass which exhibits ambient temperature.
35. Pharmaceutical dosage form according to any of claims 32 to 34, wherein the extruded monolithic mass has been cut prior to compression.
36. Pharmaceutical dosage form according to any of claims 32 to 35, wherein melt extrusion has been conducted with a twin-screw extruder.
37. Pharmaceutical dosage form, preferably according to any of the preceding claims, comprising at least one pharmacologically active compound (A), and having a shape comprising a longitudinal axis and two opposite longitudinal edges, a transversal axis perpendicular to the longitudinal axis and two opposite transversal edges, a front side, an opposite back side and a circumferential rim between said front and back side, wherein the front side and/or the back side comprise a basis area and wherein the front side and/or the back side comprise at least one bulge which extends above said basis area, said at least one bulge being present at and/or adjacent to at least a section of one or both longitudinal edges and/or at and/or adjacent to at least a section of one or both transversal edges and/or between both longitudinal edges and both transversal edges.
38. Pharmaceutical dosage form according to any claim 37, wherein the front side and the back side each comprise at least one bulge at least along a section at and/or adjacent to both longitudinal edges and/or at least along a section at and/or adjacent to both transversal edges.
39. Pharmaceutical dosage form according to claim 37 or 38, wherein said front side and said back side comprise an at least essentially continuous bulge at and/or adjacent to at least two third of both opposite longitudinal edges.
40. Pharmaceutical dosage form according to any of claims 37 to 39, wherein one or both longitudinal edges are essentially straight over at least a major part of their length and/or wherein one or both transversal edges are curved over a major part of their length.
41. Pharmaceutical dosage form according to any of claims 37 to 40, wherein the front side and/or the back side comprise an essentially circumferential bulge at and/or adjacent to the circumferential edge of the front side and/or the back side of said pharmaceutical dosage form.
42. Pharmaceutical dosage form according to any of claims 37 to 41, wherein said pharmaceutical dosage form in its oblong form comprises at and/or adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of the front side at least one bulge, or wherein said pharmaceutical dosage form in its oblong form comprises at least one bulge at and/or adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of both the front side and the back side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form comprises in its oblong form a circumferential bulge at and/or adjacent to the circumferential edge of the front side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form in its oblong form comprises a circumferential bulge at and/or adjacent to the circumferential edge of both the front side and the back side of said pharmaceutical dosage form.
43. Pharmaceutical dosage form according to any of claims 37 to 42, wherein said pharmaceutical dosage form in its oblong form comprises at major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of the front side at least one bulge, or wherein said pharmaceutical dosage form in its oblong form comprises at least one bulge at major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of both the front side and the back side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form comprises in its oblong form a circumferential bulge at the circumferential edge of the front side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form in its oblong form comprises a circumferential bulge at the circumferential edge of both the front side and the back side of said pharmaceutical dosage form.
44. Pharmaceutical dosage form according to any of claims 37 to 42, wherein said pharmaceutical dosage form in its oblong form comprises adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of the front side at least one bulge, or wherein said pharmaceutical dosage form in its oblong form comprises at least one bulge adjacent to major portions of both opposite longitudinal edges, in particular at least along two thirds of said longitudinal edges, of both the front side and the back side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form comprises in its oblong form a circumferential bulge adjacent to the circumferential edge of the front side of said pharmaceutical dosage form, or wherein said pharmaceutical dosage form in its oblong form comprises a circumferential bulge adjacent to the circumferential edge of both the front side and the back side of said pharmaceutical dosage form.
45. Pharmaceutical dosage form according to any of claims 37 to 42, wherein said pharmaceutical dosage form in its oblong form comprises a circumferential bulge adjacent to the circumferential edge of both the front side and the back side of said pharmaceutical dosage form.
46. Pharmaceutical dosage form according to any of claims 37 to 45, wherein the front side and/or the back side, in particular the basis area of the front side and/or the basis area of the back side comprise, in addition to at least one bulge on the front side and/or the back side, at least one indentation, in particular between opposite longitudinal and/or transversal bulges.
47. Pharmaceutical dosage form according to claim 46, wherein the front and the back side comprise at least one indentation.
48. Pharmaceutical dosage form according to claim 47, wherein the indentation(s) in the front side and the back side are at least once at least partially off-set.
49. Pharmaceutical dosage form according to claim 47 or 48, wherein all the indentations of the front side and all the indentations of the back side are at least partially off-set.
50. Pharmaceutical dosage form according to claim 47, wherein at least one indentation of the front side and at least one indentation of the back side are positioned in a congruent manner.
51. Pharmaceutical dosage form according to claim 50, wherein all indentations of the front side and all indentations of the back side are positioned in a congruent manner.
52. Pharmaceutical dosage form according to any of claims 47 to 51, wherein the indentations have a cylindrical or cube-like or cuboid-like shape or the shape of a half-sphere.
53. Pharmaceutical dosage form according to any of claims 37 to 52, wherein the longitudinal length of the, in particular oblong, pharmaceutical dosage form does not exceed 30 mm.
54. Pharmaceutical dosage form according to any of claims 37 to 53, having an average thickness over the basis areas of the front and the back side of about at least 1 mm, and in particular of no more than about 6 mm.
55. Pharmaceutical dosage form according to any of claims 37 to 54, wherein the bulge extends perpendicular from the basis area of the front side and/or from the basis area of the back side in average from about 0.5 mm to about 2 mm.
56. Pharmaceutical dosage form according to any of claims 37 to 55, having a length in the longitudinal direction in the range of about 5 mm to about 30 mm, in particular in the range of about 15 mm to about 25 mm, a width in the range of about 5 mm to about 15 mm, in particular in the range of about 7 mm to about 12 mm, and a thickness over the basis areas in the range of about 1 mm to about 6 mm, in particular in the range of about 2 mm to about 4 mm.
57. Use of a pharmaceutical dosage form according to any one of claims 1 to 56 as an immediate release or a retarded release oral dosage form.
58. Use of a pharmaceutical dosage form according to any one of claims 1 to 56 as a tamper-resistant or break-resistant retarded release oral dosage form.
59. A method of treating pain in a patient in need of such treatment, said method comprising administering to said patient a pharmaceutical dosage form according to any one of claims 1 to 56, wherein the pharmacologically active compound (A) is selected from the group consisting of analgesics.
CA2713128A 2008-01-25 2009-01-23 Pharmaceutical dosage form Active CA2713128C (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
EP08001416.0 2008-01-25
EP08001416 2008-01-25
EP08001415 2008-01-25
EP08001415.2 2008-01-25
EP08018221.5 2008-10-17
EP08018221 2008-10-17
PCT/EP2009/000438 WO2009092601A1 (en) 2008-01-25 2009-01-23 Pharmaceutical dosage form

Publications (2)

Publication Number Publication Date
CA2713128A1 true CA2713128A1 (en) 2009-07-30
CA2713128C CA2713128C (en) 2016-04-05

Family

ID=40511977

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2713128A Active CA2713128C (en) 2008-01-25 2009-01-23 Pharmaceutical dosage form

Country Status (17)

Country Link
US (4) US8383152B2 (en)
EP (1) EP2249811A1 (en)
JP (1) JP5774853B2 (en)
KR (1) KR101616246B1 (en)
CN (1) CN102014877B (en)
AU (1) AU2009207796B2 (en)
BR (1) BRPI0906467C1 (en)
CA (1) CA2713128C (en)
CO (1) CO6280466A2 (en)
EC (1) ECSP10010416A (en)
IL (1) IL207128A (en)
MX (1) MX2010008138A (en)
NZ (1) NZ586792A (en)
RU (1) RU2493830C2 (en)
TW (1) TWI454288B (en)
WO (1) WO2009092601A1 (en)
ZA (1) ZA201006029B (en)

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8075872B2 (en) 2003-08-06 2011-12-13 Gruenenthal Gmbh Abuse-proofed dosage form
US8114384B2 (en) 2004-07-01 2012-02-14 Gruenenthal Gmbh Process for the production of an abuse-proofed solid dosage form
US8114383B2 (en) 2003-08-06 2012-02-14 Gruenenthal Gmbh Abuse-proofed dosage form
US8192722B2 (en) 2003-08-06 2012-06-05 Grunenthal Gmbh Abuse-proof dosage form
US8383152B2 (en) 2008-01-25 2013-02-26 Gruenenthal Gmbh Pharmaceutical dosage form
US8722086B2 (en) 2007-03-07 2014-05-13 Gruenenthal Gmbh Dosage form with impeded abuse
US9161917B2 (en) 2008-05-09 2015-10-20 Grünenthal GmbH Process for the preparation of a solid dosage form, in particular a tablet, for pharmaceutical use and process for the preparation of a precursor for a solid dosage form, in particular a tablet
US9636303B2 (en) 2010-09-02 2017-05-02 Gruenenthal Gmbh Tamper resistant dosage form comprising an anionic polymer
US9655853B2 (en) 2012-02-28 2017-05-23 Grünenthal GmbH Tamper-resistant dosage form comprising pharmacologically active compound and anionic polymer
US9675610B2 (en) 2002-06-17 2017-06-13 Grünenthal GmbH Abuse-proofed dosage form
US9737490B2 (en) 2013-05-29 2017-08-22 Grünenthal GmbH Tamper resistant dosage form with bimodal release profile
US9855263B2 (en) 2015-04-24 2018-01-02 Grünenthal GmbH Tamper-resistant dosage form with immediate release and resistance against solvent extraction
US9872835B2 (en) 2014-05-26 2018-01-23 Grünenthal GmbH Multiparticles safeguarded against ethanolic dose-dumping
US9913814B2 (en) 2014-05-12 2018-03-13 Grünenthal GmbH Tamper resistant immediate release capsule formulation comprising tapentadol
US9925146B2 (en) 2009-07-22 2018-03-27 Grünenthal GmbH Oxidation-stabilized tamper-resistant dosage form
US10058548B2 (en) 2003-08-06 2018-08-28 Grünenthal GmbH Abuse-proofed dosage form
US10064945B2 (en) 2012-05-11 2018-09-04 Gruenenthal Gmbh Thermoformed, tamper-resistant pharmaceutical dosage form containing zinc
US10080721B2 (en) 2009-07-22 2018-09-25 Gruenenthal Gmbh Hot-melt extruded pharmaceutical dosage form
US10154966B2 (en) 2013-05-29 2018-12-18 Grünenthal GmbH Tamper-resistant dosage form containing one or more particles
US10201502B2 (en) 2011-07-29 2019-02-12 Gruenenthal Gmbh Tamper-resistant tablet providing immediate drug release
US10300141B2 (en) 2010-09-02 2019-05-28 Grünenthal GmbH Tamper resistant dosage form comprising inorganic salt
US10335373B2 (en) 2012-04-18 2019-07-02 Grunenthal Gmbh Tamper resistant and dose-dumping resistant pharmaceutical dosage form
US10449547B2 (en) 2013-11-26 2019-10-22 Grünenthal GmbH Preparation of a powdery pharmaceutical composition by means of cryo-milling
US10624862B2 (en) 2013-07-12 2020-04-21 Grünenthal GmbH Tamper-resistant dosage form containing ethylene-vinyl acetate polymer
AU2020100442B4 (en) * 2020-03-02 2020-06-18 Grünenthal GmbH Dosage form providing prolonged release of tapentadol phosphoric acid salt
AU2020100441B4 (en) * 2020-03-02 2020-06-18 Grünenthal GmbH Dosage form providing prolonged release of tapentadol phosphoric acid salt
US10695297B2 (en) 2011-07-29 2020-06-30 Grünenthal GmbH Tamper-resistant tablet providing immediate drug release
US10729658B2 (en) 2005-02-04 2020-08-04 Grünenthal GmbH Process for the production of an abuse-proofed dosage form
AU2020202055B2 (en) * 2020-03-02 2020-10-01 Grünenthal GmbH Dosage form providing prolonged release of tapentadol phosphoric acid salt
AU2020202056B2 (en) * 2020-03-02 2020-10-01 Grünenthal GmbH Dosage form providing prolonged release of tapentadol phosphoric acid salt
US10842750B2 (en) 2015-09-10 2020-11-24 Grünenthal GmbH Protecting oral overdose with abuse deterrent immediate release formulations
US11224576B2 (en) 2003-12-24 2022-01-18 Grünenthal GmbH Process for the production of an abuse-proofed dosage form
US11844865B2 (en) 2004-07-01 2023-12-19 Grünenthal GmbH Abuse-proofed oral dosage form

Families Citing this family (132)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1429744A1 (en) 2001-09-21 2004-06-23 Egalet A/S Morphine polymer release system
EP1429739A1 (en) 2001-09-21 2004-06-23 Egalet A/S Polymer release system
DE602004031096D1 (en) 2003-03-26 2011-03-03 Egalet As MORPHINE SYSTEM WITH CONTROLLED RELEASE
US7201920B2 (en) 2003-11-26 2007-04-10 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of opioid containing dosage forms
CN101188999B (en) * 2005-06-03 2012-07-18 尹格莱特股份有限公司 A pharmaceutical delivery system for delivering active component dispersed in dispersion medium
US8445018B2 (en) 2006-09-15 2013-05-21 Cima Labs Inc. Abuse resistant drug formulation
WO2008148798A2 (en) 2007-06-04 2008-12-11 Egalet A/S Controlled release pharmaceutical compositions for prolonged effect
US20090052818A1 (en) * 2007-07-10 2009-02-26 Jason Matthew Mitmesser Hybrid bearing
EP3964243A1 (en) * 2008-01-28 2022-03-09 Implantica Patent Ltd Blood clot removal device, system, and method
EP2700400A1 (en) * 2009-01-26 2014-02-26 Egalet Ltd. Controlled release formulation with continuous efficacy
US8603526B2 (en) 2009-02-06 2013-12-10 Egalet Ltd. Pharmaceutical compositions resistant to abuse
WO2010089132A1 (en) 2009-02-06 2010-08-12 Egalet A/S Immediate release composition resistant to abuse by intake of alcohol
EP2445487A2 (en) 2009-06-24 2012-05-02 Egalet Ltd. Controlled release formulations
KR20110001103A (en) * 2009-06-29 2011-01-06 에스케이케미칼주식회사 Transdermal composition containing tolterodine
CA2804215C (en) 2009-07-22 2019-10-01 Eric Elenko Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
US10265311B2 (en) 2009-07-22 2019-04-23 PureTech Health LLC Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
CA2770165A1 (en) * 2009-08-17 2011-02-24 Basf Se Use of thermoplastic elastomer to increase oxygen scavenger activity of functionalized polyolefin films
EP2488029B1 (en) 2009-09-30 2016-03-23 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse
US9579285B2 (en) 2010-02-03 2017-02-28 Gruenenthal Gmbh Preparation of a powdery pharmaceutical composition by means of an extruder
CA2792046C (en) * 2010-03-04 2014-12-02 Wockhardt Limited Modified release dosage form
EP2555756B1 (en) * 2010-04-07 2018-08-22 Lupin Limited Controlled release pharmaceutical compositions of tapentadol
EP2566461A2 (en) * 2010-05-05 2013-03-13 Ratiopharm GmbH Solid tapentadol in non-crystalline form
CA2798884C (en) 2010-05-10 2016-09-13 Euro-Celtique S.A. Manufacturing of active-free granules and tablets comprising the same
EP2581065B1 (en) * 2010-06-09 2019-12-25 Kao Corporation Water-vapor-generating heating tool
BR112012031836A2 (en) 2010-06-15 2016-11-08 Gruenenthal Gmbh pharmaceutical combination for pain treatment
US9119878B2 (en) * 2010-07-08 2015-09-01 Wellesley Pharmaceuticals, Llc Extended-release formulation for reducing the frequency of urination and method of use thereof
KR101220829B1 (en) * 2010-07-16 2013-01-10 안국약품 주식회사 Sustained-release tablet containing trimebutine
WO2012028317A1 (en) 2010-09-02 2012-03-08 Grünenthal GmbH Tamper resistant dosage form comprising an anionic polymer
EP3685827A1 (en) 2011-03-04 2020-07-29 Grünenthal GmbH Aqueous pharmaceutical formulation of tapentadol for oral administration
ES2573414T3 (en) 2011-03-04 2016-06-07 Grünenthal GmbH Parenteral administration of tapentadol
JP6027549B2 (en) 2011-03-04 2016-11-16 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング Semi-solid aqueous pharmaceutical composition containing tapentadol
US9603809B2 (en) 2011-03-23 2017-03-28 Ironshore Pharmaceuticals & Development, Inc. Methods of treatment of attention deficit hyperactivity disorder
US11241391B2 (en) 2011-03-23 2022-02-08 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
WO2012129551A1 (en) * 2011-03-23 2012-09-27 Ironshore Pharmaceuticals & Development, Inc. Methods and compositions for treatment of attention deficit disorder
US10905652B2 (en) 2011-03-23 2021-02-02 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
US8916588B2 (en) 2011-03-23 2014-12-23 Ironshore Pharmaceuticals & Development, Inc. Methods for treatment of attention deficit hyperactivity disorder
US9283214B2 (en) 2011-03-23 2016-03-15 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
US9119809B2 (en) 2011-03-23 2015-09-01 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
US8927010B2 (en) 2011-03-23 2015-01-06 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
US9498447B2 (en) 2011-03-23 2016-11-22 Ironshore Pharmaceuticals & Development, Inc. Compositions for treatment of attention deficit hyperactivity disorder
US10292937B2 (en) 2011-03-23 2019-05-21 Ironshore Pharmaceuticals & Development, Inc. Methods of treatment of attention deficit hyperactivity disorder
US9650395B2 (en) * 2011-08-29 2017-05-16 Ptc Therapeutics, Inc. Antibacterial compounds and methods for use
US9409905B2 (en) 2011-08-29 2016-08-09 Ptc Therapeutics, Inc. Antibacterial compounds and methods for use
RU2481124C1 (en) * 2011-10-27 2013-05-10 Общество с ограниченной ответственностью "ВАЛЕНТА ИНТЕЛЛЕКТ" Pharmaceutical combination of atorvastatin and nicergolin for preventing or treating cerebrovascular disease
EP2838536B1 (en) * 2012-04-18 2017-08-09 Mallinckrodt LLC Immediate release pharmaceutical compositions with abuse deterrent properties
BR112014024382B1 (en) 2012-04-18 2022-08-09 SpecGx LLC IMMEDIATE RELEASE ABUSE CONTAINMENT PHARMACEUTICAL COMPOSITIONS AND THEIR PREPARATION PROCESS
WO2013165481A1 (en) * 2012-05-01 2013-11-07 Amberwing Solutions Inc. Method and product for headache relief
CA2870012A1 (en) 2012-05-11 2013-11-14 Grunenthal Gmbh Thermoformed, tamper-resistant pharmaceutical dosage form containing zinc
CN104684548A (en) * 2012-07-06 2015-06-03 埃格勒特有限责任公司 Abuse deterrent pharmaceutical compositions for controlled release
US9730885B2 (en) 2012-07-12 2017-08-15 Mallinckrodt Llc Extended release, abuse deterrent pharmaceutical compositions
ES2691982T3 (en) 2012-11-30 2018-11-29 Acura Pharmaceuticals, Inc. Self-regulated release of an active pharmaceutical ingredient
GB2499496B (en) * 2012-12-19 2014-11-19 Kraft Foods R & D Inc A method of dispensing a beverage, a beverage preparation machine, and a system
GB2510826B (en) * 2013-02-13 2015-09-02 Animax Ltd Improvements in or relating to release of a beneficial substance from a bolus
US20140234415A1 (en) * 2013-02-20 2014-08-21 Abbvie Inc. Tablet Dosage Forms
US10596127B2 (en) * 2013-03-14 2020-03-24 Wellesley Pharmaceuticals, Llc Composition for reducing the frequency of urination, method of making and use thereof
US10064826B2 (en) * 2013-03-15 2018-09-04 Navinta, Llc Direct compression and dry granulation processes for preparing carglumic acid tablets having less impurities than those produced by wet granulation process
EP2808319A1 (en) 2013-05-31 2014-12-03 Arevipharma GmbH 3-[3-(Dimethylamino)-1-ethyl-2-methylpropyl]phenol resin complex
CN105579018A (en) * 2013-07-15 2016-05-11 阿尔卡米亚肿瘤学股份有限公司 Method of pre-preparing medications for therapeutic uses
ES2670619T3 (en) 2013-07-22 2018-05-31 Duchesnay Inc. Composition for the management of nausea and vomiting
US9452181B2 (en) 2013-07-22 2016-09-27 Duchesnay Inc. Composition for the management of nausea and vomiting
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US9770514B2 (en) 2013-09-03 2017-09-26 ExxPharma Therapeutics LLC Tamper-resistant pharmaceutical dosage forms
WO2015065547A1 (en) 2013-10-31 2015-05-07 Cima Labs Inc. Immediate release abuse-deterrent granulated dosage forms
WO2015071248A1 (en) 2013-11-15 2015-05-21 Synthon B.V. Abuse-proofed extended release pharmaceutical composition comprising tapentadol
MX2016007848A (en) 2013-12-16 2016-09-07 Gruenenthal Gmbh Tamper resistant dosage form with bimodal release profile manufactured by co-extrusion.
WO2015095391A1 (en) 2013-12-17 2015-06-25 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
WO2015145461A1 (en) 2014-03-26 2015-10-01 Sun Pharma Advanced Research Company Ltd. Abuse deterrent immediate release biphasic matrix solid dosage form
WO2015152433A1 (en) * 2014-03-31 2015-10-08 Hanmi Pharm. Co., Ltd. Amorphous solid dispersion comprising paclitaxel, tablet comprising the same, and method for preparing the same
AU2015284078B2 (en) 2014-07-03 2020-01-30 SpecGx LLC Abuse deterrent immediate release formulations comprising non-cellulose polysaccharides
DK3169315T3 (en) 2014-07-17 2020-08-10 Pharmaceutical Manufacturing Res Services In Liquid-filled dosage form to prevent immediate release abuse
CN104146976B (en) * 2014-08-06 2017-02-15 沈阳药科大学 Heavy-load valproic acid drug sustained release tablet and preparation method thereof
US9132096B1 (en) 2014-09-12 2015-09-15 Alkermes Pharma Ireland Limited Abuse resistant pharmaceutical compositions
US20160106737A1 (en) 2014-10-20 2016-04-21 Pharmaceutical Manufacturing Research Services, Inc. Extended Release Abuse Deterrent Liquid Fill Dosage Form
EP3017809A1 (en) * 2014-11-07 2016-05-11 Ferring B.V. Drug-device unit containing quinagolide
JP2018503693A (en) 2015-02-03 2018-02-08 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング Anti-modified dosage form containing polyethylene glycol graft copolymer
MX2017012312A (en) 2015-03-27 2018-01-18 Gruenenthal Gmbh Stable formulation for parenteral administration of tapentadol.
JP6445923B2 (en) * 2015-04-22 2018-12-26 ダイト株式会社 Preparation of irbesartan-containing tablets
CA2983640A1 (en) 2015-04-24 2016-10-27 Grunenthal Gmbh Tamper-resistant fixed dose combination providing fast release of two drugs from different particles
US20170112766A1 (en) * 2015-04-24 2017-04-27 Grünenthal GmbH Tamper-resistant dosage form with immediate release and resistance against solvent extraction
WO2016170093A1 (en) 2015-04-24 2016-10-27 Grünenthal GmbH Tamper-resistant fixed dose combination providing fast release of two drugs from particles and a matrix
MX2017013633A (en) 2015-04-24 2018-03-08 Gruenenthal Gmbh Tamper-resistant fixed dose combination providing fast release of two drugs from particles.
US20160339015A1 (en) * 2015-05-20 2016-11-24 Lupin Inc. Oral pharmaceutical composition of methylergonovine
US11234974B2 (en) 2015-05-28 2022-02-01 Lumosa Therapeutics Co., Ltd. Pharmaceutical formulations for sustained release of sebacoyl dinalbuphine ester
US10183018B2 (en) 2015-05-28 2019-01-22 Lumosa Therapeutics Co., Ltd. Pharmaceutical formulations for sustained release of sebacoyl dinalbuphine ester
WO2017040607A1 (en) 2015-08-31 2017-03-09 Acura Pharmaceuticals, Inc. Methods and compositions for self-regulated release of active pharmaceutical ingredient
US9943513B1 (en) 2015-10-07 2018-04-17 Banner Life Sciences Llc Opioid abuse deterrent dosage forms
BR112018069785A2 (en) 2016-03-31 2019-01-29 SpecGx LLC dissuasive prolonged release abuse dosage forms
US20170296476A1 (en) 2016-04-15 2017-10-19 Grünenthal GmbH Modified release abuse deterrent dosage forms
US10335405B1 (en) 2016-05-04 2019-07-02 Patheon Softgels, Inc. Non-burst releasing pharmaceutical composition
RU2630064C1 (en) * 2016-05-27 2017-09-05 Общество с ограниченной ответственностью "ДИАМЕД-фарма" Pharmaceutical composition forlocomotor disorders treatment
AU2017294524A1 (en) 2016-07-06 2018-12-20 Grünenthal GmbH Reinforced pharmaceutical dosage form
EP3490537A1 (en) 2016-08-01 2019-06-05 Grünenthal GmbH Tamper resistant dosage form comprising an anionic polysaccharide
AU2017310006A1 (en) 2016-08-12 2019-01-31 Grünenthal GmbH Tamper resistant formulation of ephedrine and its derivatives
US10736905B1 (en) 2016-09-09 2020-08-11 Shahin Fatholahi Nefopam dosage forms and methods of treatment
CN109996533A (en) 2016-09-23 2019-07-09 格吕伦塔尔有限公司 Stabilization formulations for parenteral administration tapentadol hydrochloride
WO2018153947A1 (en) 2017-02-23 2018-08-30 Grünenthal GmbH Tapentadol as local anesthetic
CN107095070A (en) * 2017-03-25 2017-08-29 年付柱 It is a kind of quickly to improve feed of duckling muscular stomach digestion power and preparation method thereof
TR201707764A2 (en) * 2017-05-29 2018-12-21 Biofarma Ilac Sanayi Ve Ticaret Anonim Sirketi Split tablet forms of deferasirox.
US10335375B2 (en) 2017-05-30 2019-07-02 Patheon Softgels, Inc. Anti-overingestion abuse deterrent compositions
US11192139B2 (en) 2017-06-22 2021-12-07 The Procter & Gamble Company Films including a water-soluble layer and a vapor-deposited organic coating
CN110719968A (en) 2017-06-22 2020-01-21 宝洁公司 Film comprising a water-soluble layer and a vapor-deposited inorganic coating
RU2711624C2 (en) * 2017-06-30 2020-01-17 Общество с ограниченной ответственностью "ФБК" Combined agent based on nsiad, muscle relaxant and vitamin
US10736874B1 (en) 2017-09-08 2020-08-11 Shahin Fatholahi Methods for treating pain associated with sickle cell disease
US11446311B2 (en) 2017-09-08 2022-09-20 Shahin Fatholahi Methods for treating pain associated with sickle cell disease
US10441544B2 (en) 2017-10-10 2019-10-15 Douglas Pharmaceuticals, Ltd. Extended release pharmaceutical formulation
US10869838B2 (en) * 2017-10-10 2020-12-22 Douglas Pharmaceuticals, Ltd. Extended release pharmaceutical formulation
WO2020194087A2 (en) * 2019-03-25 2020-10-01 Douglas Pharmaceuticals Ltd. Extended release pharmaceutical formulation
US11471415B2 (en) 2017-10-10 2022-10-18 Douglas Pharmaceuticals, Ltd. Extended release pharmaceutical formulation and methods of treatment
CA3078272A1 (en) 2017-10-13 2019-04-18 Grunenthal Gmbh Modified release abuse deterrent dosage forms
US20190209517A1 (en) * 2018-01-10 2019-07-11 Insys Development Company, Inc. Methods of stabilizing dronabinol
TW202002957A (en) 2018-02-09 2020-01-16 德商歌林達有限公司 Tamper resistant formulation of ephedrine and its derivatives comprising a conversion inhibitor
HRP20230971T1 (en) * 2018-04-16 2023-12-08 Vianex S.A. Sustained release pyridostigmine compositions
WO2020021422A1 (en) 2018-07-21 2020-01-30 Berlia Sushma Paul Controlled release formulation comprising flavoxate
CN110742872B (en) * 2018-08-22 2021-02-23 深圳善康医疗健康产业有限公司 Preparation method of naltrexone implant
WO2020068510A1 (en) 2018-09-25 2020-04-02 SpecGx LLC Abuse deterrent immediate release capsule dosage forms
AU2019346626B2 (en) 2018-09-28 2022-06-02 Karuna Therapeutics, Inc. Compositions and methods for treating disorders ameliorated by muscarinic receptor activation
US11180442B2 (en) 2019-01-30 2021-11-23 Balchem Corporation Storage stable choline chloride compositions
EP3698776A1 (en) 2019-02-19 2020-08-26 Grünenthal GmbH Tamper-resistant dosage form with immediate release and resistance against solvent extraction
CN110327498B (en) * 2019-04-15 2021-02-02 北京大学口腔医学院 Active small molecule controlled release method based on liposome and application thereof
EP3965733A4 (en) 2019-05-07 2023-01-11 Clexio Biosciences Ltd. Abuse-deterrent dosage forms containing esketamine
RU2736713C1 (en) * 2019-12-02 2020-11-19 Общество С Ограниченной Ответственностью "Валента - Интеллект" Combination of mirtazapine and tizanidine for use in pain disorders
DE202021003994U1 (en) 2020-03-02 2022-04-12 Grünenthal GmbH Extended release dosage form of tapentadol phosphoric acid salt
WO2021185804A1 (en) 2020-03-16 2021-09-23 Grünenthal GmbH Scored tablet
WO2021219576A1 (en) 2020-04-27 2021-11-04 Grünenthal GmbH Multiparticulate dosage form containing eva copolymer and additional excipient
DE202020104285U1 (en) 2020-07-24 2020-12-18 Grünenthal GmbH Ethyl cellulose-coated particles containing a salt of tapentadol and phosphoric acid
PL3995135T3 (en) 2020-11-10 2022-10-03 Grünenthal GmbH Prolonged release dosage form of tapentadol l-(+)- tartaric acid salt
DE202020005470U1 (en) 2020-11-10 2022-01-25 Grünenthal GmbH Extended-release dosage forms of a salt of tapentadol with L-(+)-tartaric acid
WO2022101247A1 (en) 2020-11-10 2022-05-19 Grünenthal GmbH Sustained release dosage forms of a salt of tapentadol with l-(+)-tartaric acid
CN115400145A (en) * 2021-05-26 2022-11-29 魏世峰 Method for treating and preventing osteoporosis
US20230330076A1 (en) 2021-11-19 2023-10-19 Sushma Paul BERLIA Controlled release formulations of flavoxate and process for preparation thereof
CN114042148B (en) * 2022-01-13 2022-04-15 浙江湃肽生物有限公司深圳分公司 Etecatide hydrochloride stabilizer

Family Cites Families (503)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2524855A (en) 1950-10-10 Process for the manufacture of
CA722109A (en) 1965-11-23 W. Mock Henry Extrusion of ethylene oxide polymers
US2806033A (en) 1955-08-03 1957-09-10 Lewenstein Morphine derivative
US2987445A (en) 1958-10-10 1961-06-06 Rohm & Haas Drug composition
US3370035A (en) 1961-06-23 1968-02-20 Takeda Chemical Industries Ltd Stabilization of polyalkylene oxide
US3332950A (en) 1963-03-23 1967-07-25 Endo Lab 14-hydroxydihydronormorphinone derivatives
GB1147210A (en) 1965-06-30 1969-04-02 Eastman Kodak Co Improvements in or relating to vitamins
US3652589A (en) 1967-07-27 1972-03-28 Gruenenthal Chemie 1-(m-substituted phenyl)-2-aminomethyl cyclohexanols
US3806603A (en) * 1969-10-13 1974-04-23 W Gaunt Pharmaceutical carriers of plasticized dried milled particles of hydrated cooked rice endosperm
DE2210071A1 (en) * 1971-03-09 1972-09-14 PPG Industries Inc., Pittsburgh, Pa. (V.StA.) Process for applying and curing a wide variety of coatings
US3865108A (en) * 1971-05-17 1975-02-11 Ortho Pharma Corp Expandable drug delivery device
US3966747A (en) 1972-10-26 1976-06-29 Bristol-Myers Company 9-Hydroxy-6,7-benzomorphans
US4014965A (en) * 1972-11-24 1977-03-29 The Dow Chemical Company Process for scrapless forming of plastic articles
US3980766A (en) 1973-08-13 1976-09-14 West Laboratories, Inc. Orally administered drug composition for therapy in the treatment of narcotic drug addiction
US3941865A (en) 1973-12-10 1976-03-02 Union Carbide Corporation Extrusion of ethylene oxide resins
US4002173A (en) * 1974-07-23 1977-01-11 International Paper Company Diester crosslinked polyglucan hydrogels and reticulated sponges thereof
DE2530563C2 (en) * 1975-07-09 1986-07-24 Bayer Ag, 5090 Leverkusen Analgesic drugs with reduced potential for abuse
JPS603286B2 (en) 1977-03-03 1985-01-26 日本化薬株式会社 Constant-dissolution formulation
US4207893A (en) 1977-08-29 1980-06-17 Alza Corporation Device using hydrophilic polymer for delivering drug to biological environment
US4175119A (en) 1978-01-11 1979-11-20 Porter Garry L Composition and method to prevent accidental and intentional overdosage with psychoactive drugs
DE2822324C3 (en) * 1978-05-22 1981-02-26 Basf Ag, 6700 Ludwigshafen Manufacture of vitamin E dry powder
US4211681A (en) 1978-08-16 1980-07-08 Union Carbide Corporation Poly(ethylene oxide) compositions
US4200704A (en) * 1978-09-28 1980-04-29 Union Carbide Corporation Controlled degradation of poly(ethylene oxide)
NO793297L (en) 1978-10-19 1980-04-22 Mallinckrodt Inc PROCEDURE FOR THE MANUFACTURE OF OXYMORPHONE
US4258027A (en) 1979-03-26 1981-03-24 Mead Johnson & Company Multi-fractionable tablet structure
US4215104A (en) * 1979-03-26 1980-07-29 Mead Johnson & Company Multi-fractionable tablet structure
CA1146866A (en) 1979-07-05 1983-05-24 Yamanouchi Pharmaceutical Co. Ltd. Process for the production of sustained release pharmaceutical composition of solid medical material
US4353887A (en) 1979-08-16 1982-10-12 Ciba-Geigy Corporation Divisible tablet having controlled and delayed release of the active substance
CH648754A5 (en) * 1979-08-16 1985-04-15 Ciba Geigy Ag Pharmaceutical slow release tablet
US4457933A (en) 1980-01-24 1984-07-03 Bristol-Myers Company Prevention of analgesic abuse
JPS56169622A (en) 1980-06-03 1981-12-26 Kissei Pharmaceut Co Ltd Method of making solid preparation from oily substance
DE3024416C2 (en) 1980-06-28 1982-04-15 Gödecke AG, 1000 Berlin Process for the production of medicaments with sustained release of active substances
US4473640A (en) 1982-06-03 1984-09-25 Combie Joan D Detection of morphine and its analogues using enzymatic hydrolysis
US4462941A (en) 1982-06-10 1984-07-31 The Regents Of The University Of California Dynorphin amide analogs
US4427778A (en) 1982-06-29 1984-01-24 Biochem Technology, Inc. Enzymatic preparation of particulate cellulose for tablet making
US4485211A (en) 1982-09-15 1984-11-27 The B. F. Goodrich Company Poly(glycidyl ether)block copolymers and process for their preparation
US4427681A (en) * 1982-09-16 1984-01-24 Richardson-Vicks, Inc. Thixotropic compositions easily convertible to pourable liquids
US4529583A (en) 1983-03-07 1985-07-16 Clear Lake Development Group Composition and method of immobilizing emetics and method of treating human beings with emetics
US4603143A (en) 1983-05-02 1986-07-29 Basf Corporation Free-flowing, high density, fat soluble vitamin powders with improved stability
US4765989A (en) 1983-05-11 1988-08-23 Alza Corporation Osmotic device for administering certain drugs
US5082668A (en) * 1983-05-11 1992-01-21 Alza Corporation Controlled-release system with constant pushing source
US4783337A (en) 1983-05-11 1988-11-08 Alza Corporation Osmotic system comprising plurality of members for dispensing drug
US4612008A (en) 1983-05-11 1986-09-16 Alza Corporation Osmotic device with dual thermodynamic activity
US4599342A (en) 1984-01-16 1986-07-08 The Procter & Gamble Company Pharmaceutical products providing enhanced analgesia
US4629621A (en) * 1984-07-23 1986-12-16 Zetachron, Inc. Erodible matrix for sustained release bioactive composition
AU592065B2 (en) 1984-10-09 1990-01-04 Dow Chemical Company, The Sustained release dosage form based on highly plasticized cellulose ether gels
GB8507779D0 (en) 1985-03-26 1985-05-01 Fujisawa Pharmaceutical Co Drug carrier
ZA864681B (en) 1985-06-24 1987-02-25 Ici Australia Ltd Ingestible capsules
DE3665262D1 (en) 1985-06-28 1989-10-05 Carrington Lab Inc Processes for preparation of aloe products, products produced thereby and compositions thereof
US4992279A (en) * 1985-07-03 1991-02-12 Kraft General Foods, Inc. Sweetness inhibitor
US4851521A (en) 1985-07-08 1989-07-25 Fidia, S.P.A. Esters of hyaluronic acid
DE3689650T2 (en) 1985-12-17 1994-05-26 United States Surgical Corp High molecular weight bioabsorbable polymers and implants thereof.
US4711894A (en) 1986-01-16 1987-12-08 Henkel Corporation Stabilized tocopherol in dry, particulate, free-flowing form
US5198226A (en) * 1986-01-30 1993-03-30 Syntex (U.S.A.) Inc. Long acting nicardipine hydrochloride formulation
US4940556A (en) 1986-01-30 1990-07-10 Syntex (U.S.A.) Inc. Method of preparing long acting formulation
US4764378A (en) 1986-02-10 1988-08-16 Zetachron, Inc. Buccal drug dosage form
EP0239973A3 (en) 1986-03-31 1989-11-08 Union Carbide Corporation Catalyst and process for alkylene oxide polymerization
DE3612211A1 (en) 1986-04-11 1987-10-15 Basf Ag CONTINUOUS TABLET METHOD
US4667013A (en) * 1986-05-02 1987-05-19 Union Carbide Corporation Process for alkylene oxide polymerization
US4713243A (en) 1986-06-16 1987-12-15 Johnson & Johnson Products, Inc. Bioadhesive extruded film for intra-oral drug delivery and process
USRE33093E (en) 1986-06-16 1989-10-17 Johnson & Johnson Consumer Products, Inc. Bioadhesive extruded film for intra-oral drug delivery and process
USRE34990E (en) 1986-08-07 1995-07-04 Ciba-Geigy Corporation Oral therapeutic system having systemic action
CA1335748C (en) 1986-09-25 1995-05-30 Jeffrey Lawrence Finnan Crosslinked gelatins
US5227157A (en) 1986-10-14 1993-07-13 Board Of Regents, The University Of Texas System Delivery of therapeutic agents
ES2032802T5 (en) 1986-11-10 2004-01-16 Biopure Corporation SUCEDANEO OF EXTRAPUR SEMISINTETIC BLOOD.
US4892889A (en) * 1986-11-18 1990-01-09 Basf Corporation Process for making a spray-dried, directly-compressible vitamin powder comprising unhydrolyzed gelatin
JPH0831303B2 (en) 1986-12-01 1996-03-27 オムロン株式会社 Chip type fuse
ATE72111T1 (en) * 1987-01-14 1992-02-15 Ciba Geigy Ag THERAPEUTIC SYSTEM FOR POORLY SOLUBLE ACTIVE INGREDIENTS.
US4892778A (en) 1987-05-27 1990-01-09 Alza Corporation Juxtaposed laminated arrangement
US5051261A (en) 1987-11-24 1991-09-24 Fmc Corporation Method for preparing a solid sustained release form of a functionally active composition
EP0391959B1 (en) 1987-12-17 1993-01-27 The Upjohn Company Tri-scored drug tablet
DE3812567A1 (en) 1988-04-15 1989-10-26 Basf Ag METHOD FOR PRODUCING PHARMACEUTICAL MIXTURES
US4960814A (en) 1988-06-13 1990-10-02 Eastman Kodak Company Water-dispersible polymeric compositions
US5350741A (en) 1988-07-30 1994-09-27 Kanji Takada Enteric formulations of physiologically active peptides and proteins
JPH0249719A (en) 1988-08-11 1990-02-20 Dai Ichi Kogyo Seiyaku Co Ltd Oil soluble-vitamin powder having readily water-dispersible and soluble performance
GB8820327D0 (en) 1988-08-26 1988-09-28 May & Baker Ltd New compositions of matter
DE3830353A1 (en) 1988-09-07 1990-03-15 Basf Ag METHOD FOR THE CONTINUOUS PRODUCTION OF SOLID PHARMACEUTICAL FORMS
US5004601A (en) * 1988-10-14 1991-04-02 Zetachron, Inc. Low-melting moldable pharmaceutical excipient and dosage forms prepared therewith
US5139790A (en) 1988-10-14 1992-08-18 Zetachron, Inc. Low-melting moldable pharmaceutical excipient and dosage forms prepared therewith
US4957668A (en) 1988-12-07 1990-09-18 General Motors Corporation Ultrasonic compacting and bonding particles
US5190760A (en) 1989-07-08 1993-03-02 Coopers Animal Health Limited Solid pharmaceutical composition
US5169645A (en) 1989-10-31 1992-12-08 Duquesne University Of The Holy Ghost Directly compressible granules having improved flow properties
US5200197A (en) * 1989-11-16 1993-04-06 Alza Corporation Contraceptive pill
GB8926612D0 (en) 1989-11-24 1990-01-17 Erba Farmitalia Pharmaceutical compositions
EP0449775A3 (en) 1990-03-29 1992-09-02 Ciba-Geigy Ag Polyether-polyester block copolymers and their use as dispersing agents
SU1759445A1 (en) 1990-06-15 1992-09-07 Ленинградский Технологический Институт Им.Ленсовета Method of producing encapsulated hydrophobic substances
FR2664851B1 (en) 1990-07-20 1992-10-16 Oreal METHOD OF COMPACTING A POWDER MIXTURE FOR OBTAINING A COMPACT ABSORBENT OR PARTIALLY DELITABLE PRODUCT AND PRODUCT OBTAINED BY THIS PROCESS.
EP0477135A1 (en) 1990-09-07 1992-03-25 Warner-Lambert Company Chewable spheroidal coated microcapsules and methods for preparing same
US5126151A (en) 1991-01-24 1992-06-30 Warner-Lambert Company Encapsulation matrix
US5273758A (en) 1991-03-18 1993-12-28 Sandoz Ltd. Directly compressible polyethylene oxide vehicle for preparing therapeutic dosage forms
US5149538A (en) 1991-06-14 1992-09-22 Warner-Lambert Company Misuse-resistive transdermal opioid dosage form
JP3073054B2 (en) 1991-07-11 2000-08-07 住友精化株式会社 Method for producing alkylene oxide polymer
JP3126384B2 (en) 1991-08-30 2001-01-22 昭和薬品化工株式会社 Dry gel composition
ATE183642T1 (en) * 1991-10-04 1999-09-15 Yoshitomi Pharmaceutical DELAYED-RELEASE TABLET
WO1993006723A1 (en) 1991-10-04 1993-04-15 Olin Corporation Fungicide tablet
DE4138513A1 (en) 1991-11-23 1993-05-27 Basf Ag SOLID PHARMACEUTICAL RETARD FORM
US5266331A (en) 1991-11-27 1993-11-30 Euroceltique, S.A. Controlled release oxycodone compositions
CA2125148C (en) 1991-12-05 1999-05-11 Siva N. Raman A carbohydrate glass matrix for the sustained release of a therapeutic agent
ES2109377T3 (en) 1991-12-18 1998-01-16 Warner Lambert Co PROCESS FOR THE PREPARATION OF A SOLID DISPERSION.
US5225417A (en) 1992-01-21 1993-07-06 G. D. Searle & Co. Opioid agonist compounds
IL105553A (en) 1992-05-06 1998-01-04 Janssen Pharmaceutica Inc Solid dosage form comprising a porous network of matrix forming material which disperses rapidly in water
EP0641195B1 (en) 1992-05-22 1996-04-10 Gödecke Aktiengesellschaft Process for preparing delayed-action medicinal compositions
GB9217295D0 (en) 1992-08-14 1992-09-30 Wellcome Found Controlled released tablets
DE4227385A1 (en) * 1992-08-19 1994-02-24 Kali Chemie Pharma Gmbh Pancreatin micropellets
DE4229085C2 (en) 1992-09-01 1996-07-11 Boehringer Mannheim Gmbh Elongated, divisible tablet
PL175026B1 (en) 1992-09-18 1998-10-30 Yamanouchi Pharma Co Ltd Prolonged action preparation of hydrogel type
US5472943A (en) 1992-09-21 1995-12-05 Albert Einstein College Of Medicine Of Yeshiva University, Method of simultaneously enhancing analgesic potency and attenuating dependence liability caused by morphine and other opioid agonists
FI101039B (en) 1992-10-09 1998-04-15 Eeva Kristoffersson Method for preparing medicated pellets
AU679937B2 (en) 1992-11-18 1997-07-17 Johnson & Johnson Consumer Products, Inc. Extrudable compositions for topical or transdermal drug delivery
JP3459421B2 (en) 1992-12-23 2003-10-20 サイテック ソチエタ レスポンサビリタ リミテ Process for the preparation of controlled open pharmaceutical forms and the pharmaceutical forms thus obtained
GB2273874A (en) * 1992-12-31 1994-07-06 Pertti Olavi Toermaelae Preparation of pharmaceuticals in a polymer matrix
US6071970A (en) 1993-02-08 2000-06-06 Nps Pharmaceuticals, Inc. Compounds active at a novel site on receptor-operated calcium channels useful for treatment of neurological disorders and diseases
DE4309528C2 (en) 1993-03-24 1998-05-20 Doxa Gmbh Casein film or film tube, process for their production and their use
IL119660A (en) 1993-05-10 2002-09-12 Euro Celtique Sa Controlled release formulation comprising tramadol
IL109944A (en) 1993-07-01 1998-12-06 Euro Celtique Sa Sustained release dosage unit forms containing morphine and a method of preparing these sustained release dosage unit forms
DE4329794C2 (en) * 1993-09-03 1997-09-18 Gruenenthal Gmbh Tramadol salt-containing drugs with delayed release
EP1442745A1 (en) 1993-10-07 2004-08-04 Euro-Celtique Orally administrable opioid formulations having extended duration of effect
DE69429710T2 (en) 1993-11-23 2002-08-08 Euro Celtique Sa Process for the preparation of a drug composition with delayed drug delivery
KR100354702B1 (en) 1993-11-23 2002-12-28 유로-셀티크 소시에떼 아노뉨 Manufacturing method and sustained release composition of pharmaceutical composition
AU1266895A (en) 1993-12-20 1995-07-10 Procter & Gamble Company, The Process for making laxatives containing dioctyl sulfosuccinate
GB9401894D0 (en) 1994-02-01 1994-03-30 Rhone Poulenc Rorer Ltd New compositions of matter
CA2182282C (en) 1994-02-16 2006-04-18 Jacqueline E. Briskin Process for preparing fine particle pharmaceutical formulations
SE9503924D0 (en) 1995-08-18 1995-11-07 Astra Ab Novel opioid peptides
US5458887A (en) 1994-03-02 1995-10-17 Andrx Pharmaceuticals, Inc. Controlled release tablet formulation
DE4413350A1 (en) 1994-04-18 1995-10-19 Basf Ag Retard matrix pellets and process for their production
RO114740B1 (en) 1994-05-06 1999-07-30 Pfizer Controlled release composition, process for preparing the same and method of treatment
DE19509807A1 (en) 1995-03-21 1996-09-26 Basf Ag Process for the preparation of active substance preparations in the form of a solid solution of the active substance in a polymer matrix, and active substance preparations produced using this method
AT403988B (en) 1994-05-18 1998-07-27 Lannacher Heilmittel SOLID ORAL RETARDED PREPARATION
US5460826A (en) 1994-06-27 1995-10-24 Alza Corporation Morphine therapy
DE4426245A1 (en) 1994-07-23 1996-02-22 Gruenenthal Gmbh 1-phenyl-3-dimethylamino-propane compounds with pharmacological activity
IT1274879B (en) 1994-08-03 1997-07-25 Saitec Srl APPARATUS AND METHOD FOR PREPARING SOLID PHARMACEUTICAL FORMS WITH CONTROLLED RELEASE OF THE ACTIVE INGREDIENT.
JP3285452B2 (en) 1994-08-11 2002-05-27 サンスター株式会社 Toothpaste composition
US5837790A (en) 1994-10-24 1998-11-17 Amcol International Corporation Precipitation polymerization process for producing an oil adsorbent polymer capable of entrapping solid particles and liquids and the product thereof
AUPM897594A0 (en) 1994-10-25 1994-11-17 Daratech Pty Ltd Controlled release container
US5965161A (en) 1994-11-04 1999-10-12 Euro-Celtique, S.A. Extruded multi-particulates
DE4446470A1 (en) * 1994-12-23 1996-06-27 Basf Ag Process for the production of dividable tablets
DE19504832A1 (en) 1995-02-14 1996-08-22 Basf Ag Solid drug preparations
US5945125A (en) 1995-02-28 1999-08-31 Temple University Controlled release tablet
US6348469B1 (en) * 1995-04-14 2002-02-19 Pharma Pass Llc Solid compositions containing glipizide and polyethylene oxide
US6117453A (en) 1995-04-14 2000-09-12 Pharma Pass Solid compositions containing polyethylene oxide and an active ingredient
US5900425A (en) 1995-05-02 1999-05-04 Bayer Aktiengesellschaft Pharmaceutical preparations having controlled release of active compound and processes for their preparation
DE19522899C1 (en) 1995-06-23 1996-12-19 Hexal Pharmaforschung Gmbh Process for the continuous sintering of a granulate
US5759583A (en) 1995-08-30 1998-06-02 Syntex (U.S.A.) Inc. Sustained release poly (lactic/glycolic) matrices
US6063405A (en) 1995-09-29 2000-05-16 L.A.M. Pharmaceuticals, Llc Sustained release delivery system
US5811126A (en) 1995-10-02 1998-09-22 Euro-Celtique, S.A. Controlled release matrix for pharmaceuticals
DE19539361A1 (en) 1995-10-23 1997-04-24 Basf Ag Process for the preparation of multilayer, solid pharmaceutical forms for oral or rectal administration
US5908850A (en) 1995-12-04 1999-06-01 Celgene Corporation Method of treating attention deficit disorders with d-threo methylphenidate
US6355656B1 (en) * 1995-12-04 2002-03-12 Celgene Corporation Phenidate drug formulations having diminished abuse potential
DE19547766A1 (en) 1995-12-20 1997-06-26 Gruenenthal Gmbh 1-phenyl-2-dimethylaminomethyl-cyclohexan-1-ol compounds as active pharmaceutical ingredients
EP0914097B1 (en) 1996-03-12 2002-01-16 Alza Corporation Composition and dosage form comprising opioid antagonist
US6461644B1 (en) 1996-03-25 2002-10-08 Richard R. Jackson Anesthetizing plastics, drug delivery plastics, and related medical products, systems and methods
US6096339A (en) 1997-04-04 2000-08-01 Alza Corporation Dosage form, process of making and using same
US20020114838A1 (en) 1996-04-05 2002-08-22 Ayer Atul D. Uniform drug delivery therapy
EP2253327A1 (en) 1996-04-05 2010-11-24 Takeda Pharmaceutical Company Limited Pharmaceutical composition containing a compound having angiotensin II antagonistic activity in combination with another compound
US5817343A (en) 1996-05-14 1998-10-06 Alkermes, Inc. Method for fabricating polymer-based controlled-release devices
EP1014941B2 (en) 1996-06-26 2017-05-17 The Board Of Regents, The University Of Texas System Hot-melt extrudable pharmaceutical formulation
EP0859603B1 (en) 1996-07-08 2008-12-17 Penwest Pharmaceuticals Co. Sustained release matrix for high-dose insoluble drugs
DE19629753A1 (en) 1996-07-23 1998-01-29 Basf Ag Process for the production of solid dosage forms
NL1003684C2 (en) 1996-07-25 1998-01-28 Weterings B V H Device for dispensing a liquid.
DE19630236A1 (en) 1996-07-26 1998-01-29 Wolff Walsrode Ag Biaxially stretched, biodegradable and compostable sausage casing
BE1010353A5 (en) 1996-08-14 1998-06-02 Boss Pharmaceuticals Ag Method for manufacture of pharmaceutical products, device for such a method and pharmaceutical products obtained.
JP4034357B2 (en) 1996-11-05 2008-01-16 ノバモント・ソシエタ・ペル・アチオニ Biodegradable polymer composition comprising starch and thermoplastic polymer
US5991799A (en) 1996-12-20 1999-11-23 Liberate Technologies Information retrieval system using an internet multiplexer to focus user selection
DE19705538C1 (en) 1997-02-14 1998-08-27 Goedecke Ag Process for the separation of active substances in solid pharmaceutical preparations
US5948787A (en) 1997-02-28 1999-09-07 Alza Corporation Compositions containing opiate analgesics
DE19710009A1 (en) 1997-03-12 1998-09-24 Knoll Ag Multi-phase preparation forms containing active ingredients
DE19710213A1 (en) 1997-03-12 1998-09-17 Basf Ag Process for the manufacture of solid combination dosage forms
US6139770A (en) 1997-05-16 2000-10-31 Chevron Chemical Company Llc Photoinitiators and oxygen scavenging compositions
DE19721467A1 (en) 1997-05-22 1998-11-26 Basf Ag Process for the preparation of small-scale preparations of biologically active substances
ES2248908T7 (en) * 1997-06-06 2014-11-24 Depomed, Inc. Dosage forms of drugs orally and gastric retention for continued release of highly soluble drugs
US6635280B2 (en) 1997-06-06 2003-10-21 Depomed, Inc. Extending the duration of drug release within the stomach during the fed mode
PT1009387E (en) 1997-07-02 2006-08-31 Euro Celtique Sa STABILIZED CONTROLLED FREQUENCY FORMULATIONS OF TRAMADOL
IE970588A1 (en) 1997-08-01 2000-08-23 Elan Corp Plc Controlled release pharmaceutical compositions containing tiagabine
CN1290239A (en) 1997-09-10 2001-04-04 联合讯号公司 Injection molding of structure zirconia-based materials by aqueous process
US6009390A (en) 1997-09-11 1999-12-28 Lucent Technologies Inc. Technique for selective use of Gaussian kernels and mixture component weights of tied-mixture hidden Markov models for speech recognition
WO1999027891A2 (en) * 1997-11-28 1999-06-10 Knoll Aktiengesellschaft Method for producing solvent-free non-crystalline biologically active substances
DE19753534A1 (en) 1997-12-03 1999-06-10 Bayer Ag Biodegradable thermoplastic polyester-amides with good mechanical properties for molding, film and fiber, useful for e.g. compostable refuse bag
CA2312479A1 (en) * 1997-12-03 1999-06-10 Bayer Aktiengesellschaft Polyether ester amides
AU755790B2 (en) 1997-12-22 2002-12-19 Euro-Celtique S.A. A method of preventing abuse of opioid dosage forms
US6375957B1 (en) * 1997-12-22 2002-04-23 Euro-Celtique, S.A. Opioid agonist/opioid antagonist/acetaminophen combinations
DE19800698A1 (en) 1998-01-10 1999-07-15 Bayer Ag Biodegradable polyester amides with block-like polyester and polyamide segments
DE19800689C1 (en) 1998-01-10 1999-07-15 Deloro Stellite Gmbh Shaped body made of a wear-resistant material
EP0980894B1 (en) 1998-03-05 2004-06-23 Mitsui Chemicals, Inc. Polylactic acid composition and film thereof
US6245357B1 (en) 1998-03-06 2001-06-12 Alza Corporation Extended release dosage form
US6090411A (en) 1998-03-09 2000-07-18 Temple University Monolithic tablet for controlled drug release
US6110500A (en) 1998-03-25 2000-08-29 Temple University Coated tablet with long term parabolic and zero-order release kinetics
WO1999052135A1 (en) 1998-04-02 1999-10-14 Applied Materials, Inc. Method for etching low k dielectrics
ES2221370T3 (en) 1998-04-03 2004-12-16 Egalet A/S COMPOSITION OF CONTROLLED RELEASE.
US5962488A (en) 1998-04-08 1999-10-05 Roberts Laboratories, Inc. Stable pharmaceutical formulations for treating internal bowel syndrome containing isoxazole derivatives
DE19822979A1 (en) 1998-05-25 1999-12-02 Kalle Nalo Gmbh & Co Kg Film with starch or starch derivatives and polyester urethanes and process for their production
US6333087B1 (en) 1998-08-27 2001-12-25 Chevron Chemical Company Llc Oxygen scavenging packaging
DE19841244A1 (en) 1998-09-09 2000-03-16 Knoll Ag Method and device for making tablets
US6268177B1 (en) * 1998-09-22 2001-07-31 Smithkline Beecham Corporation Isolated nucleic acid encoding nucleotide pyrophosphorylase
WO2000023073A1 (en) 1998-10-20 2000-04-27 Korea Institute Of Science And Technology Bioflavonoids as plasma high density lipoprotein level increasing agent
US20060240105A1 (en) 1998-11-02 2006-10-26 Elan Corporation, Plc Multiparticulate modified release composition
ES2141688B1 (en) 1998-11-06 2001-02-01 Vita Invest Sa NEW ESTERS DERIVED FROM SUBSTITUTED FENIL-CICLOHEXIL COMPOUNDS.
DE19855440A1 (en) 1998-12-01 2000-06-08 Basf Ag Process for the production of solid dosage forms by melt extrusion
DE19856147A1 (en) 1998-12-04 2000-06-08 Knoll Ag Divisible solid dosage forms and methods for their preparation
EP1005863A1 (en) 1998-12-04 2000-06-07 Synthelabo Controlled-release dosage forms comprising a short acting hypnotic or a salt thereof
US6238697B1 (en) * 1998-12-21 2001-05-29 Pharmalogix, Inc. Methods and formulations for making bupropion hydrochloride tablets using direct compression
WO2000040205A2 (en) 1999-01-05 2000-07-13 Copley Pharmaceutical Inc. Sustained release formulation with reduced moisture sensitivity
EP1070504A4 (en) 1999-02-04 2004-03-10 Nichimo Kk Materials for preventing arteriosclerosis, immunopotentiating materials, vertebrates fed with these materials and eggs thereof
US7374779B2 (en) 1999-02-26 2008-05-20 Lipocine, Inc. Pharmaceutical formulations and systems for improved absorption and multistage release of active agents
US6375963B1 (en) * 1999-06-16 2002-04-23 Michael A. Repka Bioadhesive hot-melt extruded film for topical and mucosal adhesion applications and drug delivery and process for preparation thereof
MXPA02000725A (en) 1999-07-29 2003-07-14 Roxane Lab Inc Opioid sustained released formulation.
US20030118641A1 (en) 2000-07-27 2003-06-26 Roxane Laboratories, Inc. Abuse-resistant sustained-release opioid formulation
US6562375B1 (en) 1999-08-04 2003-05-13 Yamanouchi Pharmaceuticals, Co., Ltd. Stable pharmaceutical composition for oral use
KR100345214B1 (en) 1999-08-17 2002-07-25 이강춘 The nasal transmucosal delivery of peptides conjugated with biocompatible polymers
DE19940740A1 (en) 1999-08-31 2001-03-01 Gruenenthal Gmbh Pharmaceutical salts
DE19940944B4 (en) * 1999-08-31 2006-10-12 Grünenthal GmbH Retarded, oral, pharmaceutical dosage forms
NZ517559A (en) 1999-08-31 2004-08-27 Gruenenthal Chemie Sustained release pharmaceutical composition containing tramadol saccharinate
DE19960494A1 (en) * 1999-12-15 2001-06-21 Knoll Ag Device and method for producing solid active substance-containing forms
ES2160534B1 (en) 1999-12-30 2002-04-16 Vita Invest Sa NEW ESTERS DERIVED FROM (RR, SS) -2-HYDROXIBENZOATE 3- (2-DIMETHYLMINOME-1-HYDROXICICLOHEXIL) PHENYL.
US6680070B1 (en) 2000-01-18 2004-01-20 Albemarle Corporation Particulate blends and compacted products formed therefrom, and the preparation thereof
EP2517710B1 (en) 2000-02-08 2015-03-25 Euro-Celtique S.A. Tamper-resistant oral opioid agonist formulations
US20020015730A1 (en) * 2000-03-09 2002-02-07 Torsten Hoffmann Pharmaceutical formulations and method for making
DE10015479A1 (en) 2000-03-29 2001-10-11 Basf Ag Solid oral dosage forms with delayed release of active ingredient and high mechanical stability
US8012504B2 (en) 2000-04-28 2011-09-06 Reckitt Benckiser Inc. Sustained release of guaifenesin combination drugs
US6572887B2 (en) 2000-05-01 2003-06-03 National Starch And Chemical Investment Holding Corporation Polysaccharide material for direct compression
US6419954B1 (en) 2000-05-19 2002-07-16 Yamanouchi Pharmaceutical Co., Ltd. Tablets and methods for modified release of hydrophilic and other active agents
IL153052A0 (en) 2000-05-23 2003-06-24 Cenes Pharmaceuticals Inc Nrg-2 nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods
DE10029201A1 (en) * 2000-06-19 2001-12-20 Basf Ag Retarded release oral dosage form, obtained by granulating mixture containing active agent and polyvinyl acetate-polyvinyl pyrrolidone mixture below the melting temperature
US6488962B1 (en) 2000-06-20 2002-12-03 Depomed, Inc. Tablet shapes to enhance gastric retention of swellable controlled-release oral dosage forms
US6607748B1 (en) 2000-06-29 2003-08-19 Vincent Lenaerts Cross-linked high amylose starch for use in controlled-release pharmaceutical formulations and processes for its manufacture
DE10036400A1 (en) 2000-07-26 2002-06-06 Mitsubishi Polyester Film Gmbh White, biaxially oriented polyester film
WO2002026262A2 (en) 2000-09-25 2002-04-04 Pro-Pharmaceuticals, Inc. Compositions for reducing side effects in chemotherapeutic treatments
WO2002026061A1 (en) 2000-09-27 2002-04-04 Danisco A/S Antimicrobial agent
AU2001294902A1 (en) 2000-09-28 2002-04-08 The Dow Chemical Company Polymer composite structures useful for controlled release systems
GB0026137D0 (en) 2000-10-25 2000-12-13 Euro Celtique Sa Transdermal dosage form
US6344215B1 (en) 2000-10-27 2002-02-05 Eurand America, Inc. Methylphenidate modified release formulations
EP2283829A1 (en) 2000-10-30 2011-02-16 Euro-Celtique S.A. Controlled release hydrocodone formulations
AU2002226098A1 (en) 2000-10-30 2002-05-15 The Board Of Regents, The University Of Texas System Spherical particles produced by a hot-melt extrusion/spheronization process
DE10109763A1 (en) 2001-02-28 2002-09-05 Gruenenthal Gmbh Pharmaceutical salts
JP2002265592A (en) 2001-03-07 2002-09-18 Sumitomo Seika Chem Co Ltd Process for producing alkylene oxide polymer
WO2002071860A1 (en) 2001-03-13 2002-09-19 L.A. Dreyfus Co. Gum base and gum manufacturing using particulated gum base ingredients
JP3967554B2 (en) 2001-03-15 2007-08-29 株式会社ポッカコーポレーション Flavonoid compound and method for producing the same
EP1241110A1 (en) 2001-03-16 2002-09-18 Pfizer Products Inc. Dispensing unit for oxygen-sensitive drugs
US20020132395A1 (en) 2001-03-16 2002-09-19 International Business Machines Corporation Body contact in SOI devices by electrically weakening the oxide under the body
US20020187192A1 (en) 2001-04-30 2002-12-12 Yatindra Joshi Pharmaceutical composition which reduces or eliminates drug abuse potential
DE60211885T2 (en) 2001-05-01 2006-11-02 Union Carbide Chemicals & Plastics Technology Corp., Danbury PHARMACEUTICAL COMPOSITION CONTAINING POLYALKYLENE OXIDES WITH REDUCED AMOIC ACID AND ANTIC ACID DERIVATIVES
CN1525851A (en) 2001-05-11 2004-09-01 ������ҩ�����޹�˾ Abuse-resistant controlled-release opioid dosage form
US6623754B2 (en) 2001-05-21 2003-09-23 Noveon Ip Holdings Corp. Dosage form of N-acetyl cysteine
AU2002339378A1 (en) 2001-05-22 2002-12-03 Euro-Celtique Compartmentalized dosage form
US20030064122A1 (en) 2001-05-23 2003-04-03 Endo Pharmaceuticals, Inc. Abuse resistant pharmaceutical composition containing capsaicin
WO2003002100A1 (en) 2001-06-26 2003-01-09 Farrell John J Tamper-proof narcotic delivery system
US20030008409A1 (en) * 2001-07-03 2003-01-09 Spearman Steven R. Method and apparatus for determining sunlight exposure
US8329216B2 (en) 2001-07-06 2012-12-11 Endo Pharmaceuticals Inc. Oxymorphone controlled release formulations
EP2311460A1 (en) 2001-07-06 2011-04-20 Endo Pharmaceuticals Inc. Oxymorphone controlled release formulations
ES2292775T3 (en) 2001-07-06 2008-03-16 Penwest Pharmaceuticals Co. FORMULATIONS OF PROLONGED RELEASE OF OXIMORPHONE.
JP2003020517A (en) 2001-07-10 2003-01-24 Calp Corp Resin composition for compound fiber
ATE419039T1 (en) * 2001-07-18 2009-01-15 Euro Celtique Sa PHARMACEUTICAL COMBINATIONS OF OXYCODONE AND NALOXONE
US6883976B2 (en) 2001-07-30 2005-04-26 Seikoh Giken Co., Ltd. Optical fiber ferrule assembly and optical module and optical connector using the same
WO2003013433A2 (en) 2001-08-06 2003-02-20 Euro-Celtique S.A. Sequestered antagonist formulations
IL160217A0 (en) 2001-08-06 2004-07-25 Euro Celtique Sa Compositions and methods to prevent abuse of opioids
WO2003013479A1 (en) 2001-08-06 2003-02-20 Euro-Celtique S.A. Compositions and methods to prevent abuse of opioids
US7141250B2 (en) * 2001-08-06 2006-11-28 Euro-Celtique S.A. Pharmaceutical formulation containing bittering agent
US20030068375A1 (en) 2001-08-06 2003-04-10 Curtis Wright Pharmaceutical formulation containing gelling agent
AU2002321879A1 (en) * 2001-08-06 2003-03-03 Thomas Gruber Pharmaceutical formulation containing dye
US7144587B2 (en) 2001-08-06 2006-12-05 Euro-Celtique S.A. Pharmaceutical formulation containing opioid agonist, opioid antagonist and bittering agent
US20030044458A1 (en) 2001-08-06 2003-03-06 Curtis Wright Oral dosage form comprising a therapeutic agent and an adverse-effect agent
US7842307B2 (en) * 2001-08-06 2010-11-30 Purdue Pharma L.P. Pharmaceutical formulation containing opioid agonist, opioid antagonist and gelling agent
US7157103B2 (en) * 2001-08-06 2007-01-02 Euro-Celtique S.A. Pharmaceutical formulation containing irritant
US7332182B2 (en) * 2001-08-06 2008-02-19 Purdue Pharma L.P. Pharmaceutical formulation containing opioid agonist, opioid antagonist and irritant
JP3474870B2 (en) * 2001-08-08 2003-12-08 菱計装株式会社 Elevator
US20030049272A1 (en) 2001-08-30 2003-03-13 Yatindra Joshi Pharmaceutical composition which produces irritation
US20030059467A1 (en) 2001-09-14 2003-03-27 Pawan Seth Pharmaceutical composition comprising doxasozin
US6691698B2 (en) 2001-09-14 2004-02-17 Fmc Technologies Inc. Cooking oven having curved heat exchanger
US20030068276A1 (en) * 2001-09-17 2003-04-10 Lyn Hughes Dosage forms
US20030092724A1 (en) 2001-09-18 2003-05-15 Huaihung Kao Combination sustained release-immediate release oral dosage forms with an opioid analgesic and a non-opioid analgesic
EP1429744A1 (en) 2001-09-21 2004-06-23 Egalet A/S Morphine polymer release system
US20050019399A1 (en) 2001-09-21 2005-01-27 Gina Fischer Controlled release solid dispersions
JP2005523876A (en) 2001-09-26 2005-08-11 ペンウェスト ファーマシューティカルズ カンパニー Opioid formulations with reduced potential for abuse
AU2002342755A1 (en) 2001-09-26 2003-04-14 Klaus-Jurgen Steffens Method and device for producing granulates that comprise at least one pharmaceutical active substance
US6837696B2 (en) 2001-09-28 2005-01-04 Mcneil-Ppc, Inc. Apparatus for manufacturing dosage forms
NZ532568A (en) 2001-09-28 2005-07-29 Mcneil Ppc Inc Modified release dosage forms
WO2003031546A1 (en) 2001-10-09 2003-04-17 The Procter & Gamble Company Aqueous compositions for treating a surface
US6592901B2 (en) 2001-10-15 2003-07-15 Hercules Incorporated Highly compressible ethylcellulose for tableting
JP2003125706A (en) * 2001-10-23 2003-05-07 Lion Corp Mouth freshening preparation
PE20030527A1 (en) 2001-10-24 2003-07-26 Gruenenthal Chemie DELAYED-RELEASE PHARMACEUTICAL FORMULATION CONTAINING 3- (3-DIMETHYLAMINO-1-ETHYL-2-METHYL-PROPYL) PHENOL OR A PHARMACEUTICALLY ACCEPTABLE SALT OF THE SAME AND ORAL TABLETS CONTAINING IT
US20030091630A1 (en) 2001-10-25 2003-05-15 Jenny Louie-Helm Formulation of an erodible, gastric retentive oral dosage form using in vitro disintegration test data
TWI312285B (en) * 2001-10-25 2009-07-21 Depomed Inc Methods of treatment using a gastric retained gabapentin dosage
US20030104052A1 (en) 2001-10-25 2003-06-05 Bret Berner Gastric retentive oral dosage form with restricted drug release in the lower gastrointestinal tract
US20030152622A1 (en) 2001-10-25 2003-08-14 Jenny Louie-Helm Formulation of an erodible, gastric retentive oral diuretic
CA2409552A1 (en) 2001-10-25 2003-04-25 Depomed, Inc. Gastric retentive oral dosage form with restricted drug release in the lower gastrointestinal tract
US6723340B2 (en) * 2001-10-25 2004-04-20 Depomed, Inc. Optimal polymer mixtures for gastric retentive tablets
EP1441703B1 (en) 2001-10-29 2018-01-03 Massachusetts Institute of Technology Zero-order release profile dosage form manufactured by three-dimensional printing
US20030125347A1 (en) 2001-11-02 2003-07-03 Elan Corporation Plc Pharmaceutical composition
US20040126428A1 (en) 2001-11-02 2004-07-01 Lyn Hughes Pharmaceutical formulation including a resinate and an aversive agent
CA2484528A1 (en) 2001-12-06 2003-06-19 Michael P. Hite Isoflavone composition for oral delivery
FR2833838B1 (en) 2001-12-21 2005-09-16 Ellipse Pharmaceuticals METHOD FOR MANUFACTURING A TABLET INCLUDING A MORPHINIC ANALGESIC AND TABLET OBTAINED
AUPS044502A0 (en) 2002-02-11 2002-03-07 Commonwealth Scientific And Industrial Research Organisation Novel catalysts and processes for their preparation
US20040033253A1 (en) 2002-02-19 2004-02-19 Ihor Shevchuk Acyl opioid antagonists
US20030190343A1 (en) 2002-03-05 2003-10-09 Pfizer Inc. Palatable pharmaceutical compositions for companion animals
US6572889B1 (en) 2002-03-07 2003-06-03 Noveon Ip Holdings Corp. Controlled release solid dosage carbamazepine formulations
US6753009B2 (en) 2002-03-13 2004-06-22 Mcneil-Ppc, Inc. Soft tablet containing high molecular weight polyethylene oxide
LT2425821T (en) 2002-04-05 2017-07-25 Euro-Celtique S.A. Pharmaceutical preparation containing oxycodone and naloxone
DE10217232B4 (en) 2002-04-18 2004-08-19 Ticona Gmbh Process for the production of filled granules from polyethylene of high or ultra-high molecular weight
WO2003089506A1 (en) 2002-04-22 2003-10-30 Purdue Research Foundation Hydrogels having enhanced elasticity and mechanical strength properties
US20050106249A1 (en) 2002-04-29 2005-05-19 Stephen Hwang Once-a-day, oral, controlled-release, oxycodone dosage forms
WO2003092648A1 (en) 2002-04-29 2003-11-13 Alza Corporation Methods and dosage forms for controlled delivery of oxycodone
AU2003234395B2 (en) 2002-05-13 2008-01-24 Endo Pharmaceuticals Inc. Abuse-resistant opioid solid dosage form
US7776314B2 (en) 2002-06-17 2010-08-17 Grunenthal Gmbh Abuse-proofed dosage system
DE10250083A1 (en) 2002-06-17 2003-12-24 Gruenenthal Gmbh Dosage form protected against abuse
WO2004004693A1 (en) 2002-07-05 2004-01-15 Collgegium Pharmaceutical Abuse-deterrent pharmaceutical compositions of opiods and other drugs
US20040011806A1 (en) * 2002-07-17 2004-01-22 Luciano Packaging Technologies, Inc. Tablet filler device with star wheel
US20070196481A1 (en) * 2002-07-25 2007-08-23 Amidon Gregory E Sustained-release tablet composition
WO2004017947A1 (en) 2002-08-21 2004-03-04 Phoqus Pharmaceuticals Limited Use of an aqueous solution of citric acid and a water-soluble sugar like lactitol as granulation liquid in the manufacture of tablets
US7388068B2 (en) 2002-08-21 2008-06-17 Clariant Produkte (Deutschland) Gmbh Copolymers made of alkylene oxides and glycidyl ethers and use thereof as polymerizable emulsifiers
US20040052844A1 (en) * 2002-09-16 2004-03-18 Fang-Hsiung Hsiao Time-controlled, sustained release, pharmaceutical composition containing water-soluble resins
MXPA05002828A (en) 2002-09-17 2005-05-27 Wyeth Corp Oral formulations.
EP1539098B1 (en) 2002-09-20 2011-08-10 Fmc Corporation Cosmetic composition containing microcrystalline cellulose
EP1555022B1 (en) 2002-09-21 2008-02-20 Shuyi Zhang Sustained release formulation of acetaminophen and tramadol
US8623412B2 (en) 2002-09-23 2014-01-07 Elan Pharma International Limited Abuse-resistant pharmaceutical compositions
DE10250088A1 (en) 2002-10-25 2004-05-06 Grünenthal GmbH Dosage form protected against abuse
US20050186139A1 (en) 2002-10-25 2005-08-25 Gruenenthal Gmbh Abuse-proofed dosage form
US20050191244A1 (en) 2002-10-25 2005-09-01 Gruenenthal Gmbh Abuse-resistant pharmaceutical dosage form
DE10250087A1 (en) 2002-10-25 2004-05-06 Grünenthal GmbH Dosage form protected against abuse
WO2004037222A2 (en) 2002-10-25 2004-05-06 Labopharm Inc. Sustained-release tramadol formulations with 24-hour efficacy
DE10250084A1 (en) 2002-10-25 2004-05-06 Grünenthal GmbH Dosage form protected against abuse
DE10252667A1 (en) * 2002-11-11 2004-05-27 Grünenthal GmbH New spiro-((cyclohexane)-tetrahydropyrano-(3,4-b)-indole) derivatives, are ORL1 receptor ligands useful e.g. for treating anxiety, depression, epilepsy, senile dementia, withdrawal symptoms or especially pain
US20040091528A1 (en) 2002-11-12 2004-05-13 Yamanouchi Pharma Technologies, Inc. Soluble drug extended release system
US20040185097A1 (en) 2003-01-31 2004-09-23 Glenmark Pharmaceuticals Ltd. Controlled release modifying complex and pharmaceutical compositions thereof
US7442387B2 (en) 2003-03-06 2008-10-28 Astellas Pharma Inc. Pharmaceutical composition for controlled release of active substances and manufacturing method thereof
ATE454169T1 (en) 2003-03-13 2010-01-15 Controlled Chemicals Inc OXYCODONE CONJUGATES WITH LOWER ABUSE POTENTIAL AND EXTENDED DURATION
DE602004031096D1 (en) 2003-03-26 2011-03-03 Egalet As MORPHINE SYSTEM WITH CONTROLLED RELEASE
EP2186510B1 (en) 2003-03-26 2013-07-10 Egalet Ltd. Matrix compositions for controlled delivery of drug substances
TWI347201B (en) 2003-04-21 2011-08-21 Euro Celtique Sa Pharmaceutical products,uses thereof and methods for preparing the same
WO2004093819A2 (en) 2003-04-21 2004-11-04 Euro-Celtique, S.A. Tamper resistant dosage form comprising co-extruded, adverse agent particles and process of making same
US8778382B2 (en) 2003-04-30 2014-07-15 Purdue Pharma L.P. Tamper resistant transdermal dosage form
US8906413B2 (en) 2003-05-12 2014-12-09 Supernus Pharmaceuticals, Inc. Drug formulations having reduced abuse potential
CN1473562A (en) 2003-06-27 2004-02-11 辉 刘 Mouth cavity quick dissolving quick disintegrating freeze-dried tablet and its preparing method
US20050015730A1 (en) * 2003-07-14 2005-01-20 Srimanth Gunturi Systems, methods and computer program products for identifying tab order sequence of graphically represented elements
RU2339365C2 (en) 2003-08-06 2008-11-27 Грюненталь Гмбх Drug dosage form, protected from unintended application
DE102004020220A1 (en) 2004-04-22 2005-11-10 Grünenthal GmbH Process for the preparation of a secured against misuse, solid dosage form
US20070048228A1 (en) 2003-08-06 2007-03-01 Elisabeth Arkenau-Maric Abuse-proofed dosage form
DE102005005446A1 (en) * 2005-02-04 2006-08-10 Grünenthal GmbH Break-resistant dosage forms with sustained release
DE10336400A1 (en) 2003-08-06 2005-03-24 Grünenthal GmbH Anti-abuse dosage form
DE10361596A1 (en) 2003-12-24 2005-09-29 Grünenthal GmbH Process for producing an anti-abuse dosage form
CL2004002016A1 (en) 2003-08-06 2005-05-20 Gruenenthal Chemie THERMOFORMED DOSAGE FORM FOR PROOF OF ABUSE CONTAINING (A) ONE OR MORE SUSCEPTIBLE ACTIVE PRINCIPLES OF ABUSE, (B) OPTIONALLY AUXILIARY SUBSTANCES, (C) AT LEAST A DEFINED SYNTHETIC OR NATURAL POLYMER AND (D) OPTIONALLY AT LEAST
DE102004032051A1 (en) 2004-07-01 2006-01-19 Grünenthal GmbH Process for the preparation of a secured against misuse, solid dosage form
PT1658054E (en) 2003-08-06 2007-09-18 Gruenenthal Gmbh Dosage form that is safeguarded from abuse
US8075872B2 (en) * 2003-08-06 2011-12-13 Gruenenthal Gmbh Abuse-proofed dosage form
US20050063214A1 (en) 2003-09-22 2005-03-24 Daisaburo Takashima Semiconductor integrated circuit device
PT1663229E (en) 2003-09-25 2010-07-13 Euro Celtique Sa Pharmaceutical combinations of hydrocodone and naltrexone
WO2005032524A2 (en) 2003-09-30 2005-04-14 Alza Corporation Osmotically driven active agent delivery device providing an ascending release profile
US20060172006A1 (en) 2003-10-10 2006-08-03 Vincent Lenaerts Sustained-release tramadol formulations with 24-hour clinical efficacy
US20060009478A1 (en) 2003-10-15 2006-01-12 Nadav Friedmann Methods for the treatment of back pain
WO2005041968A2 (en) 2003-10-29 2005-05-12 Alza Corporation Once-a-day, oral, controlled-release, oxycodone dosage forms
US7201920B2 (en) 2003-11-26 2007-04-10 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of opioid containing dosage forms
JP2007513147A (en) 2003-12-04 2007-05-24 ファイザー・プロダクツ・インク Spray congealing process for producing a multiparticulate crystalline pharmaceutical composition, preferably containing poloxamer and glyceride, using an extruder
WO2005055981A2 (en) * 2003-12-09 2005-06-23 Euro-Celtique S.A. Tamper resistant co-extruded dosage form containing an active agent and an adverse agent and process of making same
WO2005060942A1 (en) 2003-12-19 2005-07-07 Aurobindo Pharma Ltd Extended release pharmaceutical composition of metformin
DE10360792A1 (en) 2003-12-23 2005-07-28 Grünenthal GmbH Spirocyclic cyclohexane derivatives
CA2551815A1 (en) 2003-12-29 2005-07-21 Alza Corporation Novel drug compositions and dosage forms
WO2005079752A2 (en) 2004-02-11 2005-09-01 Rubicon Research Private Limited Controlled release pharmaceutical compositions with improved bioavailability
GB0403098D0 (en) 2004-02-12 2004-03-17 Euro Celtique Sa Extrusion
GB0403100D0 (en) 2004-02-12 2004-03-17 Euro Celtique Sa Particulates
TWI350762B (en) 2004-02-12 2011-10-21 Euro Celtique Sa Particulates
CN1921814B (en) 2004-02-23 2012-02-29 欧洲凯尔特公司 Abuse resistance opioid transdermal delivery device and its pharmaceutical use
TWI365880B (en) * 2004-03-30 2012-06-11 Euro Celtique Sa Process for preparing oxycodone hydrochloride having less than 25 ppm 14-hydroxycodeinone and oxycodone hydrochloride composition,pharmaceutical dosage form,sustained release oeal dosage form and pharmaceutically acceptable package having less than 25 pp
US20050220877A1 (en) 2004-03-31 2005-10-06 Patel Ashish A Bilayer tablet comprising an antihistamine and a decongestant
DE102004019916A1 (en) 2004-04-21 2005-11-17 Grünenthal GmbH Anti-abuse drug-containing patch
PL1740156T3 (en) 2004-04-22 2011-12-30 Gruenenthal Gmbh Method for the production of an abuse-proof, solid form of administration
WO2005105036A1 (en) 2004-04-28 2005-11-10 Natco Pharma Limited Controlled release mucoadhesive matrix formulation containing tolterodine and a process for its preparation
TWI356036B (en) * 2004-06-09 2012-01-11 Smithkline Beecham Corp Apparatus and method for pharmaceutical production
PL1612203T3 (en) 2004-06-28 2007-12-31 Gruenenthal Gmbh Crystalline forms of (-)-(1R,2R)-3-(3-dimethylamino-1-ethyl-2-methylpropyl)-phenol hydrochloride
ITMI20041317A1 (en) 2004-06-30 2004-09-30 Ibsa Inst Biochimique Sa PHARMACEUTICAL FORMULATIONS FOR THE SAFE ADMINISTRATION OF DRUGS USED IN THE TREATMENT OF DRUG ADDICTION AND PROCEDURE FOR THEIR OBTAINING
DE102004032103A1 (en) 2004-07-01 2006-01-19 Grünenthal GmbH Anti-abuse, oral dosage form
KR101204657B1 (en) 2004-07-01 2012-11-27 그뤼넨탈 게엠베하 Oral dosage form safeguarded against abuse containing 1r,2r-3-3-dimethylamino-1-ethyl-2-methyl-propyl-phenol
DE102004032049A1 (en) 2004-07-01 2006-01-19 Grünenthal GmbH Anti-abuse, oral dosage form
EP1765298B1 (en) 2004-07-01 2012-10-24 Gruenenthal Gmbh Method for producing a solid dosage form, which is safeguarded against abuse, while using a planetary gear extruder
EP1765303B2 (en) 2004-07-01 2022-11-23 Grünenthal GmbH Oral tablet safeguarded against abuse
ES2306167T3 (en) 2004-07-27 2008-11-01 Unilever N.V. COMPOSITIONS FOR HAIR CARE.
US20060068009A1 (en) 2004-09-30 2006-03-30 Scolr Pharma, Inc. Modified release ibuprofen dosage form
US20070077297A1 (en) 2004-09-30 2007-04-05 Scolr Pharma, Inc. Modified release ibuprofen dosage form
US20070231268A1 (en) 2004-11-24 2007-10-04 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of orally administered pharmaceutical products
US20060177380A1 (en) 2004-11-24 2006-08-10 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of orally administered pharmaceutical products
US20080152595A1 (en) 2004-11-24 2008-06-26 Acura Pharmaceuticals, Inc. Methods and compositions for deterring abuse of orally administered pharmaceutical products
CA2594713A1 (en) 2005-01-26 2006-08-03 Taiho Pharmaceutical Co., Ltd. Anticancer drug containing .alpha.,.alpha.,.alpha.-trifluorothymidine and thymidine phosphorylase inhibitor
EP2319499A1 (en) 2005-01-28 2011-05-11 Euro-Celtique S.A. Alcohol resistant dosage forms
DE102005005449A1 (en) 2005-02-04 2006-08-10 Grünenthal GmbH Process for producing an anti-abuse dosage form
US20060194759A1 (en) 2005-02-25 2006-08-31 Eidelson Stewart G Topical compositions and methods for treating pain and inflammation
EP1695700A1 (en) 2005-02-28 2006-08-30 Euro-Celtique S.A. Dosage form containing oxycodone and naloxone
SI1861405T1 (en) 2005-03-04 2009-10-31 Euro Celtique Sa Method of reducing alpha, beta- unsaturated ketones in opioid compositions
US7732427B2 (en) 2005-03-31 2010-06-08 University Of Delaware Multifunctional and biologically active matrices from multicomponent polymeric solutions
JP5256425B2 (en) 2005-04-08 2013-08-07 リングアル コンセグナ ピーティーワイ エルティーディー Oral delivery system
RU2405539C2 (en) 2005-05-10 2010-12-10 Новартис Аг Method of obtaining compositions by extrusion of resistant to pressing pharmaceutical substances
CN101188999B (en) 2005-06-03 2012-07-18 尹格莱特股份有限公司 A pharmaceutical delivery system for delivering active component dispersed in dispersion medium
WO2007005716A2 (en) * 2005-06-30 2007-01-11 Cinergen, Llc Methods of treatment and compositions for use thereof
WO2007008752A2 (en) * 2005-07-07 2007-01-18 Farnam Companies, Inc. Sustained release pharmaceutical compositions for highly water soluble drugs
US8858993B2 (en) 2005-07-25 2014-10-14 Metrics, Inc. Coated tablet with zero-order or near zero-order release kinetics
CN101232871A (en) 2005-08-03 2008-07-30 伊士曼化工公司 Tocopheryl polyethylene glycol succinate powder and process for preparing same
ZA200803414B (en) 2005-10-14 2009-09-30 Kitasato Inst Novel dihydropseudoerythromycin derivatives
US20070092573A1 (en) * 2005-10-24 2007-04-26 Laxminarayan Joshi Stabilized extended release pharmaceutical compositions comprising a beta-adrenoreceptor antagonist
PL116330U1 (en) 2005-10-31 2007-04-02 Alza Corp Method for the reduction of alcohol provoked rapid increase in the released dose of the orally administered opioide with prolonged liberation
US8329744B2 (en) 2005-11-02 2012-12-11 Relmada Therapeutics, Inc. Methods of preventing the serotonin syndrome and compositions for use thereof
US9125833B2 (en) 2005-11-02 2015-09-08 Relmada Therapeutics, Inc. Multimodal abuse resistant and extended release opioid formulations
US8652529B2 (en) 2005-11-10 2014-02-18 Flamel Technologies Anti-misuse microparticulate oral pharmaceutical form
FR2892937B1 (en) 2005-11-10 2013-04-05 Flamel Tech Sa MICROPARTICULAR ORAL PHARMACEUTICAL FORM ANTI-MEASURING
US20100172989A1 (en) 2006-01-21 2010-07-08 Abbott Laboratories Abuse resistant melt extruded formulation having reduced alcohol interaction
US20070190142A1 (en) * 2006-01-21 2007-08-16 Abbott Gmbh & Co. Kg Dosage forms for the delivery of drugs of abuse and related methods
US20090022798A1 (en) 2007-07-20 2009-01-22 Abbott Gmbh & Co. Kg Formulations of nonopioid and confined opioid analgesics
US20090317355A1 (en) 2006-01-21 2009-12-24 Abbott Gmbh & Co. Kg, Abuse resistant melt extruded formulation having reduced alcohol interaction
EP1813276A1 (en) 2006-01-27 2007-08-01 Euro-Celtique S.A. Tamper resistant dosage forms
FR2898056B1 (en) * 2006-03-01 2012-01-20 Ethypharm Sa SQUEEZE-RESISTANT TABLETS TO PREVENT UNLAWFUL MISUSE
AU2007224221B2 (en) 2006-03-02 2013-02-14 SpecGx LLC Processes for preparing morphinan-6-one products with low levels of alpha, beta-unsaturated ketone compounds
US20100226855A1 (en) 2006-03-02 2010-09-09 Spherics, Inc. Rate-Controlled Oral Dosage Formulations
CA2932389A1 (en) 2006-03-24 2007-10-04 Auxilium International Holdings, Inc. Stabilized compositions containing alkaline labile drugs
MX2008012265A (en) 2006-03-24 2009-02-20 Auxilium Int Holdings Inc Process for the preparation of a hot-melt extruded laminate.
US20070224637A1 (en) 2006-03-24 2007-09-27 Mcauliffe Joseph C Oxidative protection of lipid layer biosensors
US10960077B2 (en) 2006-05-12 2021-03-30 Intellipharmaceutics Corp. Abuse and alcohol resistant drug composition
US9023400B2 (en) 2006-05-24 2015-05-05 Flamel Technologies Prolonged-release multimicroparticulate oral pharmaceutical form
US20070292508A1 (en) 2006-06-05 2007-12-20 Balchem Corporation Orally disintegrating dosage forms
US20080069891A1 (en) 2006-09-15 2008-03-20 Cima Labs, Inc. Abuse resistant drug formulation
CN101091721A (en) 2006-06-22 2007-12-26 孙明 Method for preparing new type asshide
JP4029109B1 (en) 2006-07-18 2008-01-09 タマ生化学株式会社 Complex powder of vitamin E and proline and method for producing the same
EP2049087A2 (en) 2006-07-21 2009-04-22 LAB International SRL Hydrophilic abuse deterrent delivery system
SA07280459B1 (en) 2006-08-25 2011-07-20 بيورديو فارما إل. بي. Tamper Resistant Oral Pharmaceutical Dosage Forms Comprising an Opioid Analgesic
US8445018B2 (en) 2006-09-15 2013-05-21 Cima Labs Inc. Abuse resistant drug formulation
US8187636B2 (en) 2006-09-25 2012-05-29 Atlantic Pharmaceuticals, Inc. Dosage forms for tamper prone therapeutic agents
KR101400824B1 (en) * 2006-09-25 2014-05-29 후지필름 가부시키가이샤 Resist composition, resin for use in the resist composition, compound for use in the synthesis of the resin, and pattern-forming method usign the resist composition
US20080085304A1 (en) 2006-10-10 2008-04-10 Penwest Pharmaceuticals Co. Robust sustained release formulations
GB0624880D0 (en) 2006-12-14 2007-01-24 Johnson Matthey Plc Improved method for making analgesics
WO2008086804A2 (en) 2007-01-16 2008-07-24 Egalet A/S Use of i) a polyglycol and n) an active drug substance for the preparation of a pharmaceutical composition for i) mitigating the risk of alcohol induced dose dumping and/or ii) reducing the risk of drug abuse
US20080181932A1 (en) 2007-01-30 2008-07-31 Drugtech Corporation Compositions for oral delivery of pharmaceuticals
CN100579525C (en) 2007-02-02 2010-01-13 东南大学 Sustained release preparation of licardipine hydrochloride and its preparing process
CN101057849A (en) 2007-02-27 2007-10-24 齐齐哈尔医学院 Slow-releasing preparation containing metformin hydrochloride and glipizide and its preparation method
JP5452236B2 (en) 2007-03-02 2014-03-26 ファーナム・カンパニーズ・インコーポレーテッド Sustained release composition using wax-like substance
DE102007011485A1 (en) 2007-03-07 2008-09-11 Grünenthal GmbH Dosage form with more difficult abuse
EP1980245A1 (en) 2007-04-11 2008-10-15 Cephalon France Bilayer lyophilized pharmaceutical compositions and methods of making and using same
WO2008132707A1 (en) 2007-04-26 2008-11-06 Sigmoid Pharma Limited Manufacture of multiple minicapsules
WO2008142627A2 (en) 2007-05-17 2008-11-27 Ranbaxy Laboratories Limited Multilayered modified release formulation comprising amoxicillin and clavulanate
US8202542B1 (en) 2007-05-31 2012-06-19 Tris Pharma Abuse resistant opioid drug-ion exchange resin complexes having hybrid coatings
WO2008148798A2 (en) 2007-06-04 2008-12-11 Egalet A/S Controlled release pharmaceutical compositions for prolonged effect
US20100035886A1 (en) 2007-06-21 2010-02-11 Veroscience, Llc Parenteral formulations of dopamine agonists
JP2010532358A (en) 2007-07-01 2010-10-07 ピーター ハバウシ,ジョセフ Formulation with chewable outer layer
JP2010534204A (en) 2007-07-20 2010-11-04 アボット ゲーエムベーハー ウント カンパニー カーゲー Formulation of non-opioid analgesics and entrapped opioid analgesics
WO2009034541A2 (en) 2007-09-11 2009-03-19 Ranbaxy Laboratories Limited Controlled release pharmaceutical dosage forms of trimetazidine
PL2200593T3 (en) 2007-09-13 2017-02-28 Cima Labs Inc. Abuse resistant drug formulation
WO2009051819A1 (en) 2007-10-17 2009-04-23 Axxia Pharmaceuticals, Llc Polymeric drug delivery systems and thermoplastic extrusion processes for producing such systems
ES2619329T3 (en) 2007-11-23 2017-06-26 Grünenthal GmbH Tapentadol compositions
EP2067471B1 (en) 2007-12-06 2018-02-14 Durect Corporation Oral pharmaceutical dosage forms
BRPI0820770A2 (en) 2007-12-12 2015-06-16 Basf Se Water-soluble polymeric drug salt, solid administration form, and method for producing water-soluble polymeric drug salt.
BRPI0821732A2 (en) 2007-12-17 2015-06-16 Labopharm Inc Controlled release formulations, solid dosage form, and use of controlled release formulation
CA2713128C (en) 2008-01-25 2016-04-05 Gruenenthal Gmbh Pharmaceutical dosage form
KR100970665B1 (en) 2008-02-04 2010-07-15 삼일제약주식회사 Sustained release tablet containing alfuzosin or its salt
JP2011513391A (en) 2008-03-05 2011-04-28 パナセア バイオテック リミテッド Sustained release pharmaceutical composition containing mycophenolate and method thereof
EP2100598A1 (en) 2008-03-13 2009-09-16 Laboratorios Almirall, S.A. Inhalation composition containing aclidinium for treatment of asthma and chronic obstructive pulmonary disease
JP2011523402A (en) 2008-05-06 2011-08-11 インスティトゥト デ インベスティガシオン エン オルトドンシア、エセ.エレ. Substances that promote tooth movement
WO2009135680A1 (en) 2008-05-09 2009-11-12 Grünenthal GmbH Process for the preparation of an intermediate powder formulation and a final solid dosage form under usage of a spray congealing step
MX2010014566A (en) 2008-07-03 2011-02-15 Novartis Ag Melt granulation process.
MX2011001864A (en) 2008-08-20 2011-06-20 Univ Texas Hot-melt extrusion of modified release multi-particulates.
WO2010044842A1 (en) 2008-10-16 2010-04-22 University Of Tennessee Research Foundation Tamper resistant oral dosage forms containing an embolizing agent
US20100104638A1 (en) 2008-10-27 2010-04-29 Wei-Guo Dai Extended release oral acetaminophen/tramadol dosage form
EP2376065A2 (en) 2008-11-14 2011-10-19 Portola Pharmaceuticals, Inc. Solid composition for controlled release of ionizable active agents with poor aqueous solubility at low ph and methods of use thereof
PL2379111T3 (en) 2008-12-12 2013-08-30 Paladin Labs Inc Narcotic drug formulations with decreased abuse potential
CN102316857A (en) 2008-12-16 2012-01-11 莱博法姆公司 Prevent the controlled release formulation misapplied
EP2700400A1 (en) 2009-01-26 2014-02-26 Egalet Ltd. Controlled release formulation with continuous efficacy
US8603526B2 (en) 2009-02-06 2013-12-10 Egalet Ltd. Pharmaceutical compositions resistant to abuse
SI2408436T1 (en) 2009-03-18 2017-06-30 Evonik Roehm Gmbh Controlled release pharmaceutical composition with resistance against the influence of ethanol employing a coating comprising neutral vinyl polymers and excipients
EP2246063A1 (en) 2009-04-29 2010-11-03 Ipsen Pharma S.A.S. Sustained release formulations comprising GnRH analogues
GB0909680D0 (en) 2009-06-05 2009-07-22 Euro Celtique Sa Dosage form
EP2445487A2 (en) 2009-06-24 2012-05-02 Egalet Ltd. Controlled release formulations
WO2011008298A2 (en) 2009-07-16 2011-01-20 Nectid, Inc. Novel axomadol dosage forms
CN102573806B (en) 2009-07-22 2015-02-25 格吕伦塔尔有限公司 Tamper-resistant dosage form for oxidation-sensitive opioids
CN102573805A (en) 2009-07-22 2012-07-11 格吕伦塔尔有限公司 Hot-melt extruded controlled release dosage form
US9579285B2 (en) 2010-02-03 2017-02-28 Gruenenthal Gmbh Preparation of a powdery pharmaceutical composition by means of an extruder
CA2795158C (en) 2010-04-02 2019-10-22 Alltranz Inc. Abuse-deterrent transdermal formulations of opiate agonists and agonist-antagonists
GB201006200D0 (en) 2010-04-14 2010-06-02 Ayanda As Composition
US10463633B2 (en) 2010-04-23 2019-11-05 Kempharm, Inc. Therapeutic formulation for reduced drug side effects
FR2960775A1 (en) 2010-06-07 2011-12-09 Ethypharm Sa MICROGRANULES RESISTANT TO MISMATCH
WO2012028319A1 (en) 2010-09-02 2012-03-08 Grünenthal GmbH Tamper resistant dosage form comprising inorganic salt
EP2611425B1 (en) 2010-09-02 2014-07-02 Grünenthal GmbH Tamper resistant dosage form comprising an anionic polymer
WO2012028317A1 (en) 2010-09-02 2012-03-08 Grünenthal GmbH Tamper resistant dosage form comprising an anionic polymer
EP2635258A1 (en) 2010-11-04 2013-09-11 AbbVie Inc. Drug formulations
US20120231083A1 (en) 2010-11-18 2012-09-13 The Board Of Trustees Of The University Of Illinois Sustained release cannabinoid medicaments
GB201020895D0 (en) 2010-12-09 2011-01-26 Euro Celtique Sa Dosage form
AU2011346758C1 (en) 2010-12-23 2015-09-03 Purdue Pharma L.P. Tamper resistant solid oral dosage forms
AU2012219322A1 (en) 2011-02-17 2013-05-09 QRxPharma Ltd. Technology for preventing abuse of solid dosage forms
EP3685827A1 (en) 2011-03-04 2020-07-29 Grünenthal GmbH Aqueous pharmaceutical formulation of tapentadol for oral administration
US8858963B1 (en) 2011-05-17 2014-10-14 Mallinckrodt Llc Tamper resistant composition comprising hydrocodone and acetaminophen for rapid onset and extended duration of analgesia
ES2628303T3 (en) 2011-06-01 2017-08-02 Fmc Corporation Solid dose controlled release forms
EP2726065A4 (en) 2011-06-30 2014-11-26 Neos Therapeutics Lp Abuse resistant drug forms
WO2013017242A1 (en) 2011-07-29 2013-02-07 Grünenthal GmbH Tamper-resistant tablet providing immediate drug release
CN103841964A (en) 2011-07-29 2014-06-04 格吕伦塔尔有限公司 Tamper-resistant tablet providing immediate drug release
FR2979242A1 (en) 2011-08-29 2013-03-01 Sanofi Sa COMPRESSES AGAINST ABUSIVE USE, BASED ON PARACETAMOL AND OXYCODONE
BR112014008120A2 (en) 2011-10-06 2017-04-11 Gruenenthal Gmbh inviolable oral pharmaceutical dosage form comprising opioid agonist and opioid antagonist
MX349725B (en) 2011-11-17 2017-08-10 Gruenenthal Gmbh Tamper-resistant oral pharmaceutical dosage form comprising a pharmacologically active ingredient, an opioid antagonist and/or aversive agent, polyalkylene oxide and anionic polymer.
CA2858868C (en) 2011-12-09 2017-08-22 Purdue Pharma L.P. Pharmaceutical dosage forms comprising poly (epsilon-caprolactone) and polyethylene oxide
WO2013127830A1 (en) 2012-02-28 2013-09-06 Grünenthal GmbH Tamper-resistant pharmaceutical dosage form comprising nonionic surfactant
US20130225697A1 (en) 2012-02-28 2013-08-29 Grunenthal Gmbh Tamper-resistant dosage form comprising pharmacologically active compound and anionic polymer
NZ629468A (en) 2012-03-02 2017-08-25 Rhodes Pharmaceuticals Lp Tamper resistant immediate release formulations
TR201815502T4 (en) 2012-04-18 2018-11-21 Gruenenthal Gmbh Tamper or pharmaceutical dosage form that is resistant and resistant to dose discharge.
US10064945B2 (en) 2012-05-11 2018-09-04 Gruenenthal Gmbh Thermoformed, tamper-resistant pharmaceutical dosage form containing zinc
CA2870012A1 (en) 2012-05-11 2013-11-14 Grunenthal Gmbh Thermoformed, tamper-resistant pharmaceutical dosage form containing zinc
WO2014059512A1 (en) 2012-10-15 2014-04-24 Isa Odidi Oral drug delivery formulations
WO2014191396A1 (en) 2013-05-29 2014-12-04 Grünenthal GmbH Tamper resistant dosage form with bimodal release profile
MX371432B (en) 2013-05-29 2020-01-30 Gruenenthal Gmbh Tamper-resistant dosage form containing one or more particles.
CA2817728A1 (en) 2013-05-31 2014-11-30 Pharmascience Inc. Abuse deterrent immediate release formulation
KR20160031526A (en) 2013-07-12 2016-03-22 그뤼넨탈 게엠베하 Tamper-resistant dosage form containing ethylene-vinyl acetate polymer
US9770514B2 (en) 2013-09-03 2017-09-26 ExxPharma Therapeutics LLC Tamper-resistant pharmaceutical dosage forms
WO2015065547A1 (en) 2013-10-31 2015-05-07 Cima Labs Inc. Immediate release abuse-deterrent granulated dosage forms
US10744131B2 (en) 2013-12-31 2020-08-18 Kashiv Biosciences, Llc Abuse-resistant drug formulations

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10369109B2 (en) 2002-06-17 2019-08-06 Grünenthal GmbH Abuse-proofed dosage form
US9675610B2 (en) 2002-06-17 2017-06-13 Grünenthal GmbH Abuse-proofed dosage form
US8075872B2 (en) 2003-08-06 2011-12-13 Gruenenthal Gmbh Abuse-proofed dosage form
US8114383B2 (en) 2003-08-06 2012-02-14 Gruenenthal Gmbh Abuse-proofed dosage form
US8192722B2 (en) 2003-08-06 2012-06-05 Grunenthal Gmbh Abuse-proof dosage form
US8309060B2 (en) 2003-08-06 2012-11-13 Grunenthal Gmbh Abuse-proofed dosage form
US10130591B2 (en) 2003-08-06 2018-11-20 Grünenthal GmbH Abuse-proofed dosage form
US10058548B2 (en) 2003-08-06 2018-08-28 Grünenthal GmbH Abuse-proofed dosage form
US8420056B2 (en) 2003-08-06 2013-04-16 Grunenthal Gmbh Abuse-proofed dosage form
US9629807B2 (en) 2003-08-06 2017-04-25 Grünenthal GmbH Abuse-proofed dosage form
US11224576B2 (en) 2003-12-24 2022-01-18 Grünenthal GmbH Process for the production of an abuse-proofed dosage form
US8323889B2 (en) 2004-07-01 2012-12-04 Gruenenthal Gmbh Process for the production of an abuse-proofed solid dosage form
US11844865B2 (en) 2004-07-01 2023-12-19 Grünenthal GmbH Abuse-proofed oral dosage form
US8114384B2 (en) 2004-07-01 2012-02-14 Gruenenthal Gmbh Process for the production of an abuse-proofed solid dosage form
US10729658B2 (en) 2005-02-04 2020-08-04 Grünenthal GmbH Process for the production of an abuse-proofed dosage form
US10675278B2 (en) 2005-02-04 2020-06-09 Grünenthal GmbH Crush resistant delayed-release dosage forms
US8722086B2 (en) 2007-03-07 2014-05-13 Gruenenthal Gmbh Dosage form with impeded abuse
US9750701B2 (en) 2008-01-25 2017-09-05 Grünenthal GmbH Pharmaceutical dosage form
US8383152B2 (en) 2008-01-25 2013-02-26 Gruenenthal Gmbh Pharmaceutical dosage form
US9161917B2 (en) 2008-05-09 2015-10-20 Grünenthal GmbH Process for the preparation of a solid dosage form, in particular a tablet, for pharmaceutical use and process for the preparation of a precursor for a solid dosage form, in particular a tablet
US10493033B2 (en) 2009-07-22 2019-12-03 Grünenthal GmbH Oxidation-stabilized tamper-resistant dosage form
US9925146B2 (en) 2009-07-22 2018-03-27 Grünenthal GmbH Oxidation-stabilized tamper-resistant dosage form
US10080721B2 (en) 2009-07-22 2018-09-25 Gruenenthal Gmbh Hot-melt extruded pharmaceutical dosage form
US9636303B2 (en) 2010-09-02 2017-05-02 Gruenenthal Gmbh Tamper resistant dosage form comprising an anionic polymer
US10300141B2 (en) 2010-09-02 2019-05-28 Grünenthal GmbH Tamper resistant dosage form comprising inorganic salt
US10201502B2 (en) 2011-07-29 2019-02-12 Gruenenthal Gmbh Tamper-resistant tablet providing immediate drug release
US10864164B2 (en) 2011-07-29 2020-12-15 Grünenthal GmbH Tamper-resistant tablet providing immediate drug release
US10695297B2 (en) 2011-07-29 2020-06-30 Grünenthal GmbH Tamper-resistant tablet providing immediate drug release
US9655853B2 (en) 2012-02-28 2017-05-23 Grünenthal GmbH Tamper-resistant dosage form comprising pharmacologically active compound and anionic polymer
US10335373B2 (en) 2012-04-18 2019-07-02 Grunenthal Gmbh Tamper resistant and dose-dumping resistant pharmaceutical dosage form
US10064945B2 (en) 2012-05-11 2018-09-04 Gruenenthal Gmbh Thermoformed, tamper-resistant pharmaceutical dosage form containing zinc
US10154966B2 (en) 2013-05-29 2018-12-18 Grünenthal GmbH Tamper-resistant dosage form containing one or more particles
US9737490B2 (en) 2013-05-29 2017-08-22 Grünenthal GmbH Tamper resistant dosage form with bimodal release profile
US10624862B2 (en) 2013-07-12 2020-04-21 Grünenthal GmbH Tamper-resistant dosage form containing ethylene-vinyl acetate polymer
US10449547B2 (en) 2013-11-26 2019-10-22 Grünenthal GmbH Preparation of a powdery pharmaceutical composition by means of cryo-milling
US9913814B2 (en) 2014-05-12 2018-03-13 Grünenthal GmbH Tamper resistant immediate release capsule formulation comprising tapentadol
US9872835B2 (en) 2014-05-26 2018-01-23 Grünenthal GmbH Multiparticles safeguarded against ethanolic dose-dumping
US9855263B2 (en) 2015-04-24 2018-01-02 Grünenthal GmbH Tamper-resistant dosage form with immediate release and resistance against solvent extraction
US10842750B2 (en) 2015-09-10 2020-11-24 Grünenthal GmbH Protecting oral overdose with abuse deterrent immediate release formulations
AU2020100441B4 (en) * 2020-03-02 2020-06-18 Grünenthal GmbH Dosage form providing prolonged release of tapentadol phosphoric acid salt
AU2020233614B2 (en) * 2020-03-02 2020-11-05 Grünenthal GmbH Dosage form providing prolonged release of tapentadol phosphoric acid salt
AU2020233614A1 (en) * 2020-03-02 2020-10-08 Grünenthal GmbH Dosage form providing prolonged release of tapentadol phosphoric acid salt
AU2020202056B2 (en) * 2020-03-02 2020-10-01 Grünenthal GmbH Dosage form providing prolonged release of tapentadol phosphoric acid salt
AU2020202055B2 (en) * 2020-03-02 2020-10-01 Grünenthal GmbH Dosage form providing prolonged release of tapentadol phosphoric acid salt
AU2020100442B4 (en) * 2020-03-02 2020-06-18 Grünenthal GmbH Dosage form providing prolonged release of tapentadol phosphoric acid salt

Also Published As

Publication number Publication date
CA2713128C (en) 2016-04-05
EP2249811A1 (en) 2010-11-17
BRPI0906467B8 (en) 2021-01-05
US20140322323A1 (en) 2014-10-30
TW200944247A (en) 2009-11-01
JP5774853B2 (en) 2015-09-09
US20160199305A1 (en) 2016-07-14
RU2493830C2 (en) 2013-09-27
KR101616246B1 (en) 2016-05-02
US20090202634A1 (en) 2009-08-13
ZA201006029B (en) 2011-05-25
ECSP10010416A (en) 2010-09-30
BRPI0906467B1 (en) 2019-12-24
US9750701B2 (en) 2017-09-05
KR20100111303A (en) 2010-10-14
IL207128A (en) 2015-01-29
IL207128A0 (en) 2010-12-30
BRPI0906467C1 (en) 2021-05-25
CO6280466A2 (en) 2011-05-20
TWI454288B (en) 2014-10-01
WO2009092601A1 (en) 2009-07-30
CN102014877A (en) 2011-04-13
US8383152B2 (en) 2013-02-26
RU2010135200A (en) 2012-02-27
NZ586792A (en) 2012-09-28
AU2009207796A1 (en) 2009-07-30
MX2010008138A (en) 2010-08-10
JP2011510034A (en) 2011-03-31
AU2009207796B2 (en) 2014-03-27
US20130251759A1 (en) 2013-09-26
BRPI0906467A2 (en) 2016-04-26
CN102014877B (en) 2017-06-06

Similar Documents

Publication Publication Date Title
US9750701B2 (en) Pharmaceutical dosage form
US10675278B2 (en) Crush resistant delayed-release dosage forms

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20140109