WO2021089715A1 - Use of colchicine in the treatment and prevention of lung cancer - Google Patents

Use of colchicine in the treatment and prevention of lung cancer Download PDF

Info

Publication number
WO2021089715A1
WO2021089715A1 PCT/EP2020/081164 EP2020081164W WO2021089715A1 WO 2021089715 A1 WO2021089715 A1 WO 2021089715A1 EP 2020081164 W EP2020081164 W EP 2020081164W WO 2021089715 A1 WO2021089715 A1 WO 2021089715A1
Authority
WO
WIPO (PCT)
Prior art keywords
colchicine
cancer
release
tumor
therapy
Prior art date
Application number
PCT/EP2020/081164
Other languages
French (fr)
Inventor
Michael Riel
Original Assignee
Murray And Poole Enterprises, Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Murray And Poole Enterprises, Ltd. filed Critical Murray And Poole Enterprises, Ltd.
Publication of WO2021089715A1 publication Critical patent/WO2021089715A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Colchicine chemical name (-)-N-[(7S, 12aS)-l, 2,3, 10-tetramethoxy-9-oxo-5, 6,7,9- tetrahydrobenzo[a]heptalen-7-yl]-acetamide, is an alkaloid found in extracts of Colchicum autumnale, Gloriosa superba, and other plants. It is a microtubule-disrupting agent used in the treatment of conditions that may be treated, relieved or prevented with anti inflammatory treatment.
  • Colchicine is well recognized as a valid therapy in acute flares of gouty arthritis, familial Mediterranean fever (FMF), and Behcet's disease. It has also been used to treat many inflammatory disorders prone to fibrosis. In the recent past, colchicine has been proposed to be effective in therapy in cardiovascular diseases. Thus, colchicine has a proven track record in treatment of various diseases and conditions associated with inflammation.
  • FMF familial Mediterranean fever
  • Behcet's disease has also been used to treat many inflammatory disorders prone to fibrosis.
  • colchicine has been proposed to be effective in therapy in cardiovascular diseases.
  • colchicine has a proven track record in treatment of various diseases and conditions associated with inflammation.
  • colchicine has been proposed as a first treatment option for recurrent pericarditis (class I indication) and optional for acute pericarditis (class Ila indication) in the 2004 European guidelines on the management of pericardial diseases (Maisch et al. , Guidelines on the Diagnosis and Management of Pericardial Diseases, Eur Heart J., 2004, 25, 916-928).
  • colchicine is effective for secondary prevention of recurrent pericarditis Imazio et al., Ann. Intern. Med., 2011, 155 (7), 409-14). Colchicine has also been proposed to reduce postpericardiotomy reactions revealed as pericarditis (Imazio et al., Am. Heart J., 2011, 162 (3), 527-532; Meurin and Tabet, Arch. Cardiovasc. Dis., 2011, 104 (8-9), 425-427).
  • Colchicine for the treatment of post-pericardiotomy syndrome was tested for the first time in a preliminary prospective, open-label, randomized trial of colchicine (1.5 mg/day) compared with placebo beginning on the third post-operative day in 163 patients who underwent cardiac surgery (Finkelstein et al., Herz, 200227, 791-194).
  • colchicine For the treatment of gout, the recommended dose of colchicine (COLCRYS®) is 1.8 mg/day in one or multiple doses in one hour. For adults with gout, treatment is initiated with a dose of 1.2 mg at the first sign of symptoms followed by 0.6 mg one hour later. (Physician's Desk Reference, 68th ed., (2014)).
  • colchicine is associated with many adverse side effects.
  • COLCRYS® for instance, is an immediate release formulation of colchicine.
  • Adverse effects associated with the administration of COLCRYS® include, but are not limited to, nausea, vomiting, abdominal pain, diarrhea, hair loss, weakness, nerve irritation, severe anemia, low white blood counts, and low platelets (Physician's Desk Reference, 68th ed., (2014)).
  • IL-Ib interleukin- 1b
  • colchicine is a microtubule inhibitor that has anti-inflammatory activity in part through interference with the NLRP3 inflammasome structure that leads to a similar reduction in iterleukukin- ⁇ b levels.
  • the instant invention addresses these and other needs by providing a modified formulation of low dose colchicine characterized by a sustained release of an active ingredient that is well tolerated with few, if any, of the commonly experienced adverse side effects associated with immediate release colchicine and colchicine at higher dosages.
  • a modified formulation of low dose colchicine characterized by a sustained release of an active ingredient that is well tolerated with few, if any, of the commonly experienced adverse side effects associated with immediate release colchicine and colchicine at higher dosages.
  • This invention addresses the need for safe and effective cancer treatments, such as lung cancer, that serve either as a single agent that treats, prevents, inhibits, reduces, suppresses, prevents, slows or delays the progression of, shrinks, or directly attacks cancer cells or that can act in combination with other immune modulating therapies to enhance their therapeutic benefit.
  • Colchicine is inexpensive, tolerated at low doses, has a proven safety track record, and fits the needs for this group of people.
  • Colchicine has a long history as a safe anti-inflammatory agent used in several clinical settings including arthritis, gout and pericarditis, diseases known to be driven in part by NLRP3 mediated inflammation.
  • colchicine As an anti-mitotic microtubule inhibitor, colchicine has been shown to disrupt inflammasome construction resulting in lower systemic levels of both IL-Ib and CRP (Martinon, Nature, 2006; Martinez, Atherosclerosis, 2018). Small clinical studies have suggested benefits of colchicine in the setting of gastric or hypopharyngeal tumors, and gout patients treated with colchicine have been reported to have a reduced cancer burden (Zhang, BioSci. Rep., 2019; Kim, Anticancer Res., 2017; Kuo, Medicine, 2015).
  • a method for inhibiting, delaying, or reducing the occurrence of lung cancer in a subject with cancer comprising administering to the subject an effective amount of colchicine.
  • the colchicine may induce anti-tumor responses, inhibit IL-Ib mediated signaling, inhibit inflammation activation, inhibit release of TNF-a and IL-10.
  • the colchicine is included in an amount between about 0.25mg and about l.Omg. The colchicine could be included in an amount between about 0.5mg.
  • a method for treating lung cancer in a subject comprising administering to the subject an effective amount of colchicine.
  • the colchicine may induce anti-tumor responses, inhibit IL-Ib mediated signaling, inhibit inflammation activation, inhibit release of TNF-a and IL-10.
  • the colchicine is included in an amount between about 0.25mg and about l.Omg.
  • the colchicine could be included in an amount between about 0.5mg.
  • a method for treating lung cancer in a subject comprising administering to the subject an effective amount of colchicine and an effective amount of at least one other immune modulating therapy.
  • the immune modulating therapy is selected from the group consisting of a cancer vaccine, an immuno stimulatory agent, adoptive T cell or antibody therapy, immune checkpoint blockade and a combination thereof.
  • the immune modulating agent is selected from the group consisting of interleukins, cytokines, chemokines, antagonists of immune checkpoint blockades and a combination thereof.
  • the immune modulating therapy may be a cancer therapy.
  • the cancer therapy is selected from the group consisting of surgery or surgical procedures, radiation therapy, chemotherapy or a combination thereof.
  • colchicine and the immune modulating agent or immune modulating therapy are administered separately or concurrently.
  • the colchicine is included in an amount between about 0.25mg and about l.Omg.
  • the colchicine could be included in an amount between about 0.5mg.
  • FIG. 1 shows the dissolution profiles for colchicine sustained-release formulations containing 10%, 15% and 20%, respectively, of an exemplary retarding agent.
  • FIG. 2 shows the dissolution profiles for colchicine sustained-release formulation containing 30% of an exemplary retarding agent and tablet hardnesses of 50N and 130N, respectively.
  • FIG. 3 shows the dissolution profiles for colchicine sustained-release formulations according to FIGS. 1 and 2.
  • FIG. 4 shows the dissolution profile for a colchicine sustained-release formulation containing 0% of an exemplary retarding agent.
  • FIG. 5 shows the dissolution profile for a colchicine sustained-release formulation containing 25% of an exemplary retarding agent.
  • FIG. 6 shows the dissolution profile for colchicine formulation containing 0%, 23.3%, 26.6% and 30% of an exemplary retarding agent.
  • FIGS. 7 A, 7B, 7C and 7D show plasma colchicine levels (ng/mL) as a function of time (hrs) for colchicine formulations according to FIG. 6.
  • FIG. 8 shows plasma colchicine levels (ng/mL) as a function of time (hrs) for colchicine formulations according to FIG. 6.
  • FIG. 9 shows the levels of colchicine in neutrophils on Day 1 and Day 10 for colchicine formulations according to FIG. 6.
  • colchicine includes colchicine or any pharmaceutically acceptable salts thereof.
  • “Pharmaceutically acceptable” means that which is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary use as well as human pharmaceutical use.
  • “Pharmaceutically acceptable salts” includes derivatives of colchicine, wherein the colchicine is modified by making acid or base addition salts thereof, and further refers to pharmaceutically acceptable solvates, including hydrates, and co-crystals of such compounds and such salts.
  • Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid addition salts of basic residues such as amines; alkali or organic addition salts of acidic residues; and the like, and combinations comprising one or more of the foregoing salts.
  • the pharmaceutically acceptable salts include non-toxic salts and the quaternary ammonium salts of the colchicine.
  • non-toxic acid salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; other acceptable inorganic salts include metal salts such as sodium salt, potassium salt, cesium salt, and the like; and alkaline earth metal salts, such as calcium salt, magnesium salt, and the like, and combinations comprising one or more of the foregoing salts.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like
  • other acceptable inorganic salts include metal salts such as sodium salt, potassium salt, cesium salt, and the like
  • alkaline earth metal salts such as calcium salt, magnesium salt, and the like, and combinations comprising one or more of the foregoing salts.
  • Organic salts includes salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, mesylic, esylic, besylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, HOOC — (CH2)n — COOH where n is 0-4, and the like; organic amine salts such as triethylamine salt, pyridine salt, picoline salt, ethanolamine salt, triethanolamine salt, dicyclohexylamine salt, N,N'- dibenzylethylenediamine salt, and the like; and amino acid salts such as arginate, as
  • colchicine salts include colchicine hydrochloride, colchicine dihydrochloride, and co-crystals, hydrates or solvates thereof.
  • “Pharmacokinetic parameters” describe the in vivo characteristics of an active agent (or a metabolite or a surrogate marker for the active agent) over time, such as plasma concentration (C), Cmax, Cn, C24, Tmax, and AUC.
  • Cmax is the measured plasma concentration of the active agent at the point of maximum, or peak, concentration.
  • Cmin is the measured plasma concentration of the active agent at the point of minimum concentration.
  • Cn is the measured plasma concentration of the active agent at about n hours after administration.
  • “C24” is the measured plasma concentration of the active agent at about 24 hours after administration.
  • Tmax refers to the time at which the measured plasma concentration of the active agent is the highest after administration of the active agent.
  • AUC is the area under the curve of a graph of the measured plasma concentration of an active agent vs. time, measured from one time point to another time point.
  • AUCO-t is the area under the curve of plasma concentration versus time from time 0 to time t, where t can be the last time point with measurable plasma concentration for an individual formulation.
  • the AUC0- ⁇ or AUCO-INF is the calculated area under the curve of plasma concentration versus time from time 0 to time infinity.
  • AUCO-t is the area under the curve of plasma concentration over the dosing interval (i.e., from time 0 to time t (tau), where tau is the length of the dosing interval.
  • Ke or Kel the terminal elimination rate constant calculated from a semi-log plot of the plasma concentration versus time curve
  • tl/2 the terminal elimination half-life, calculated as 0.693/Kel
  • CL/F denotes the apparent total body clearance after administration, calculated as Total Dose/Total AUC ⁇
  • Varea/F denotes the apparent total volume of distribution after administration, calculated as Total Dose/(Total AUC ⁇ xKel).
  • Effectiveness means the ability of an active agent administered to a patient to produce a therapeutic effect in the patient.
  • Bioavailability means the extent or rate at which an active agent is absorbed into a living system or is made available at the site of physiological activity.
  • bioavailability data for a given formulation may provide an estimate of the relative fraction of the administered dose that is absorbed into the systemic circulation.
  • Bioavailability can be characterized by one or more pharmacokinetic parameters.
  • a “dosage form” means a unit of administration of an active agent.
  • dosage forms include tablets, capsules, injections, suspensions, liquids, emulsions, creams, ointments, suppositories, inhalable forms, transdermal forms, and the like.
  • An “immediate release formulation” refers to a formulation that releases greater than or equal to about 80% of the pharmaceutical agent in less than or equal to about 30 min.
  • an enhancing agent is defined as any non- pharmaceutically active ingredient that improves the therapeutic potential of a formulation.
  • sustained release is defined herein as release of a pharmaceutical agent in a continuous manner over a prolonged period of time.
  • Prolonged period of time it is meant a continuous period of time of greater than about 1 hour, greater than about 4 hours, greater than about 8 hours, greater than about 12 hours, greater than about 16 hours, or up to more than about 24 hours.
  • rate of release or “release rate” or “dissolution rate” of a drug refers to the quantity of drug released from a dosage form per unit time, e.g., milligrams of drug released per hour (mg/hr) or a percentage of a total drug dose released per hour.
  • Drug release rates for dosage forms are typically measured as an in vitro rate of drug release, i.e., a quantity of drug released from the dosage form per unit time measured under appropriate conditions and in a suitable fluid.
  • the release rates referred to herein are determined by placing a dosage form to be tested in a medium in an appropriate dissolution bath. Aliquots of the medium, collected at pre-set intervals, are then injected into a chromatographic system fitted with an appropriate detector to quantify the amounts of drug released during the testing intervals.
  • side effect refers to a secondary and usually adverse effect of a drug.
  • cancer refers to or describe the physiological condition in mammals in which a population of cells are characterized by unregulated cell growth.
  • examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia.
  • cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, gastric, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, brain cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, esophageal cancer, salivary gland carcinoma, sarcoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancers.
  • Tumor and “neoplasm” as used herein refer to any mass of tissue that result from excessive cell growth or proliferation, either benign (noncancerous) or malignant (cancerous) including pre-cancerous lesions.
  • the terms “metastasis,” “metastases,” “metastatic,” and other grammatical equivalents as used herein refer to cancer cells which spread or transfer from the site of origin (e.g., a primary tumor) to other regions of the body with the development of a similar cancerous lesion at the new location.
  • a “metastatic” or “metastasizing” cell is one that loses adhesive contacts with neighboring cells and migrates via the bloodstream or lymph from the primary site of disease to invade neighboring body structures.
  • metastatic cancers that are amenable to treatment via the methods provided herein include, but are not limited to metastatic sarcomas, breast carcinomas, ovarian cancer, head and neck cancer, and pancreatic cancer.
  • the term "therapeutically effective amount” refers to an amount of an antibody, polypeptide, polynucleotide, small organic molecule, or other drug effective to "treat” a disease or disorder in a subject or mammal.
  • the therapeutically effective amount of the drug can reduce the number of cancer cells; retard or stop cancer cell division, reduce or retard an increase in tumor size; inhibit, e.g., suppress, retard, prevent, stop, delay, or reverse cancer cell infiltration into peripheral organs including, for example, the spread of cancer into soft tissue and bone; inhibit, e.g., suppress, retard, prevent, shrink, stop, delay, or reverse tumor metastasis; inhibit, e.g., suppress, retard, prevent, stop, delay, or reverse tumor growth; relieve to some extent one or more of the symptoms associated with the cancer, reduce morbidity and mortality; improve quality of life; or a combination of such effects.
  • the drug prevents growth and/or kills existing cancer cells, it can be referred to as cytostatic
  • Terms such as “treating” or “treatment” or “to treat” or “alleviating” or “to alleviate” refer to both 1) therapeutic measures that cure, slow down, lessen symptoms of, reverse, and/or halt progression of a diagnosed pathologic condition or disorder and 2) prophylactic or preventative measures that prevent and/or slow the development of a targeted pathologic condition or disorder.
  • those in need of treatment include those already with the disorder; those prone to have the disorder; and those in whom the disorder is to be prevented.
  • a subject is successfully "treated” according to the methods of the present invention if the patient shows one or more of the following: a reduction in the number of or complete absence of cancer cells; a reduction in the tumor size; or retardation or reversal of tumor growth, inhibition, e.g., suppression, prevention, retardation, shrinkage, delay, or reversal of metastases, e.g., of cancer cell infiltration into peripheral organs including, for example, the spread of cancer into soft tissue and bone; inhibition of, e.g., suppression of, retardation of, prevention of, shrinkage of, reversal of, delay of, or an absence of tumor metastases; inhibition of, e.g., suppression of, retardation of, prevention of, shrinkage of, reversal of, delay of, or an absence of tumor growth; relief of one or more symptoms associated with the specific cancer; reduced morbidity and mortality; improvement in quality of life; or some combination of effects.
  • a reduction in the number of or complete absence of cancer cells e.g., suppression
  • Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
  • subject or “individual” or “animal” or “patient” or “mammal,” is meant any subject, particularly a mammalian subject, for whom diagnosis, prognosis, or therapy is desired.
  • Mammalian subjects include humans, domestic animals, farm animals, and zoo, sports, or pet animals such as dogs, cats, guinea pigs, rabbits, rats, mice, horses, cattle, cows, bears, and so on.
  • eukaryote”, “animal”, “mammal”, etc. is well known in the art and can, for example, be deduced from Wehner und Gehring (1995; Thieme Verlag).
  • animals are to be treated which are economically, agronomically or scientifically important.
  • Scientifically important organisms include, but are not limited to, mice, rats, and rabbits.
  • Non-limiting examples of agronomically important animals are sheep, cattle and pigs, while, for example, cats and dogs may be considered as economically important animals.
  • the subject/patient is a mammal; in another embodiment, the subject/patient is a human or a non-human mammal (such as, e.g., a guinea pig, a hamster, a rat, a mouse, a rabbit, a dog, a cat, a horse, a monkey, an ape, a marmoset, a baboon, a gorilla, a chimpanzee, an orang utan, a gibbon, a sheep, cattle, or a pig); most preferably, the subject/patient is a human.
  • a non-human mammal such as, e.g., a guinea pig, a hamster, a rat, a mouse, a rabbit, a dog, a cat, a horse, a monkey, an ape, a marmoset, a baboon, a gorilla, a chimpanzee, an orang
  • colchicine used according to the present invention will be described in detail.
  • the chemical structure of colchicine (ChemID 2012) is as follows:
  • colchicine N[5,6,7,9-tetrahydro-l,2,3,10-tetratmethoxy 9- oxobenzo[a]heptalen-7-yl],(S)-acetamide; molecular fo r m u 1 a : C 22 H 25 N Oe ; CAS number: 64-86-8.
  • Colchicine is an anti-inflammatory drug with a long history in human medicine, used for the symptomatic treatment of inflammatory diseases, most prominently gout. It is a natural product which can be extracted from two plants of the lily family, Colchicum autumnale and Gloriosa superba. Colchicine is a tricyclic alkaloid and has a molecular mass of 399.437.
  • the active ingredient colchicine as well as its tablet formulation is listed in various national and international pharmacopeias such as the United States Pharmacopeia (USP).
  • colchicine The pharmacotherapeutic mechanism of action of colchicine in diverse disorders is not fully understood, though it is known that the drug accumulates preferentially in leucocytes, particularly neutrophils which is important for its therapeutic effect.
  • Availability of tubulin is essential for several cellular functions such as mitosis. Therefore colchicine effectively functions as a "mitotic poison" or spindle poison.
  • colchicine By inhibiting microtubule self-assembly, colchicine interferes with many cellular functions involved in the immune response such as modulation of the production of chemokines chemokines and prostanoids and inhibition of neutrophil and endothelial cell adhesion molecules. Eventually it decreases neutrophil degranulation, chemotaxis and phagocytosis, thus reducing the initiation and amplification of inflammation. Colchicine also inhibits uric acid crystal deposition (a process important to the genesis of gout), which is enhanced by a low pH in the tissues, probably by inhibiting oxidation of glucose and subsequent lactic acid reduction in leukocytes (Imazio, Brucato el al.
  • colchicine excerpts its therapeutic effect by suppressing the acute pericardial inflammation.
  • the exact cellular and molecular mechanisms of how colchicine relieves pain and inflammation in acute pericarditis and prevents recurrences are not fully understood.
  • Inflammation has particular pathophysiologic relevance for lung cancer where chronic bronchitis, triggered by asbestos, silica, smoking, and other external inhaled toxins, results in a persistent inflammatory response (O’Callaghan, J. Thoracic. Oncol., 5:2024-36, 2010; Lee, Crit. Rev. Oncol. Hematol., 66:208-17, 2008).
  • Inflammatory activation in the lung is mediated in part through activation of the Nod-like receptor protein 3 (NLRP3) inflammasome with consequent local generation of active IL-Ib, a process that can lead to both chronic fibrosis and cancer (Dostert, Science, 320:674-7, 2008; Gasse, /. Clin.
  • Colchicine in the context of the present invention can be used for treating and/or preventing lung cancer, or inhibiting, delaying, or reducing tumor growth or metastases, or tumor growth and metastases in a subject with lung cancer.
  • the present invention additionally provides colchicine formulations to prevent and/or treat lung cancer, and to inhibit, delay, or reduce tumor growth or metastases in a subject in need of such inhibition, delay, or reduction, e.g., a lung cancer patient.
  • the colchicine formulation may be a sustained release formulation, wherein colchicine is released from the formulation at a sustained rate along a pre-determined or desired release profile. Such release is achieved by incorporation into the formulation of an extended release component and an optional immediate release component.
  • the colchicine formulation may be an immediate release formulation, wherein colchicine is released from the formulation quickly upon contacting a fluid such as water and allow fast leaching out of colchicine to the environment over a short period of time, such as several minutes or in an hour.
  • the colchicine formulation of the present invention may be formulated in a dosage form selected from a tablet, a pill, a capsule, a caplet, a troche, a sachet, a cachet, a pouch, sprinkles, or any other form suitable for oral administration.
  • colchicine as described herein i.e., inter alia, in the form of a (pharmaceutical) composition
  • the dissolution rate is such that at least approximately 80% of colchicine is released within the first 60 mins. In general, at least approximately 90% of colchicine will be released within 2 hours.
  • the immediate-release particles may be produced by any known method, such as dry or wet granulation method as described below. In one example, colchicine is mixed with disintegrants and/or binders, and adsorbents and then the mixture is subjected to either fluid bed granulation or spray drying to produce particles with desired immediate-release property.
  • disintegrants include, but are not limited to, cross-linked polyvinyl pyrrolidone or crospovidone, starch derivatives such as carboxy methyl cellulose and cellulose derivatives; calcium alginate; carboxymethylcellulose calcium; carboxymethylcellulose sodium; croscarmellose sodium; docusate sodium; hydroxypropyl cellulose; magnesium aluminum silicate; methylcellulose; polacrilin potassium; sodium alginate; sodium starch glycolate and pregelatinized starch.
  • adsorbents include, but are not limited to, aluminum hydroxide adjuvant; aluminum oxide; aluminum phosphate adjuvant; attapulgite; bentonite; powdered cellulose; colloidal silicon dioxide; hectorite; kaolin; magnesium aluminum silicate; magnesium carbonate; microcrystalline cellulose; pectin; polycarbophil; and saponite.
  • colchicine as described herein is administered in the form of a sustained release preparation.
  • Other expressions like “extended release”, “controlled release”, “modified release” or “delayed release” “preparation” or “formulation” are understood herein to have the same meaning as “sustained release preparation”.
  • Such preparations can in principal be in any form conceivable to the skilled person and include pharmaceutical forms for oral (solid, semi-solid, liquid), dermal (dermal patch), sublingual, parenteral (injection), ophthalmic (eye drops, gel or ointment) or rectal (suppository) administration, as long as a sustained release is ensured.
  • sustained release preparations encompass all pharmaceutical forms that create a steady drug release profile making the drug substance available over an extended period of time following application to the patient.
  • Such an extended period of time may be between 10, 20, 30, 40, 50 or 60 minutes and about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours.
  • Extended release may also be defined functionally as the release of over 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 99 percent (%) of colchicine after about 10, 20, 30, 40, 50 or 60 minutes and about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours.
  • Extended release as used herein may also be defined as making colchicine available to the patient regardless of uptake, as some colchicine may never be absorbed by the patient.
  • Various extended release dosage forms may be designed readily by one of skill in art as disclosed herein to achieve delivery and sustained release of colchicine to the liver and/or both the small and large intestines, to only the small intestine, or to only the large intestine.
  • sustained release preparations may be pH independent. This allows such preparations to dissolve in almost any environment.
  • sustained release preparations may be pH dependent. This allows release to be accomplished at some generally predictable location in the lower intestinal tract more distal to that which would have been accomplished if there had been no delayed release alterations.
  • a method for delay of release is, e.g., a coating. Any coatings should be applied to a sufficient thickness such that the entire coating does not dissolve in the gastrointestinal fluids at pH below about 5, but does dissolve at pH about 5 and above. It is expected that any anionic polymer exhibiting a pH-dependent solubility profile can be used as an enteric coating in the practice of the present invention to achieve delivery to the lower gastrointestinal tract.
  • Polymers and compatible mixtures thereof may be used to provide the coating for the delayed or the extended release of active ingredients, and some of their properties, include, but are not limited to: shellac, also called purified lac, a refined product obtained from the resinous secretion of an insect. This coating dissolves in media of pH >7.
  • sustained release preparations may be influenced by the presence of alcohol in the body.
  • the presence of alcohol is a patient’s body can increase dissolution of the composition and can cause immediate release of the entire dose. This effect is known as “dose dumping” and is dependent on the alcohol solubility of the materials.
  • dose dumping is dependent on the alcohol solubility of the materials.
  • sustained release preparations which contain a higher dose for slow release over 24 hours, for instance, this effect can have safety concerns and can even be life threatening.
  • sustained release preparations may be prepared using time release hydrophilic matrices.
  • time release hydrophilic matrices are known in the field of drug formulations.
  • one such hydrophilic matrix is hydroxypropyl methylcellulose (HPMC) or Hypromellose.
  • Hydrophilic matrices provide an initial release of the drug product in the initial phase mainly triggered by a rapid swelling of the surface of the matrix tablet, combined with an erosion process leading to an immediate release of the drug substance distributed close to the surface of the tablet.
  • about 50%, about 45%, about 40%, about 35%, about 30%, about 25%, about 20%, about 15%, or about 10% of the drug substance may immediately be released depending on the desired release profile.
  • At least about 20% of the drug substance may immediately be released. In another embodiment, at least about 20% of the drug substance may be released within about the first 30 minutes.
  • the term "about” or “approximately” refers to a variation of 10% from the indicated values (e.g., 50%, 45%, 40%, etc.), or in case of a range of values, means a 10% variation from both the lower and upper limits of such ranges. For instance, “about 50%” refers to a range of between 45% and 55%.
  • This gelling prevents the tablet core from dissolving and disintegrating immediately, thereby allowing the main part of the drug substances to dissolve slowly over time within in this gel structure and diffuse into solution following the rules of Fick's law.
  • the diffusion itself may be triggered in this formulation approach by the concentration of the Hypromellose and the viscosity of the formed gel, defined over the molecular weight of the Hypromellose. Therefore, drug release profiles can be modified by varying different viscosity grades of Hypromellose or mixtures thereof. All corresponding formulation and process parameters achieving the predicted release profile are common knowledge and can be adjusted using actual development technologies e.g. formulation screenings, statistical trials designs.
  • the substance responsible for sustained release of the controlled-release formulation can further mix with a binder.
  • the binder is added to increase the mechanical strength of the granules and tablets during formation.
  • Binders can be added to the formulation in different ways: (1) as a dry powder, which is mixed with other ingredients before wet agglomeration, (2) as a solution, which is used as agglomeration liquid during wet agglomeration, and is referred to as a solution binder, and (3) as a dry powder, which is mixed with the other ingredients before compaction.
  • the binder is referred to as a dry binder.
  • Solution binders are a common way of incorporating a binder into granules.
  • the binder used in the formulation is in the form of a dry powder binder.
  • binders useful for the core include hydrogenated vegetable oil, castor oil, paraffin, higher aliphatic alcohols, higher aliphatic acids, long chain fatty acids, fatty acid esters, wax-like materials such as fatty alcohols, fatty acid esters, fatty acid glycerides, hydrogenated fats, hydrocarbons, normal waxes, stearic acid, stearyl alcohol, hydrophobic and hydrophilic polymers having hydrocarbon backbones, and mixtures thereof.
  • water-soluble polymer binders include modified starch, gelatin, polyvinylpyrrolidone, cellulose derivatives (such as for example hydroxypropyl methylcellulose (HPMC) and hydroxypropyl cellulose (HPC)), polyvinyl alcohol and mixtures thereof.
  • the binder is HPMC.
  • the binder is Hypromellose 6mPa*s.
  • the binder can be present in an amount of from about 1% to about 30% by weight of the formulation.
  • the sustained release formulation may include a disintegrant.
  • a disintegrant refers to an agent used in pharmaceutical preparation of tablets, which causes them to disintegrate and release their medicinal substances on contact with moisture.
  • the disintegrant may be water soluble to support the disintegrantation of a tablet in the stomach.
  • Non-limiting examples of disintegrants for use in the formulation include sucrose, lactose, in particular lactose monohydrate, trehalose, maltose, mannitol and sorbitol, croscarmellose sodium, crospovidone, alginic acid, sodium alginate, methacrylic acid DVB, cross-linked PVP, microcrystalline cellulose, polacrilin potassium, sodium starch glycolate, starch, pregelatinized starch and mixtures thereof.
  • the disintegrant is selected from microcrystalline cellulose (e.g. Avicel PH101), cross-linked polyvinylpyrrolidone (e.g. KOLLIDON® CL), cross-linked sodium carboxymethylcellulose (e.g.
  • AC-DI-SOL(TM) starch or starch derivatives such as sodium starch glycolate (e.g. EXPLOTAB®), or combinations with starch (e.g. PRIMOJEL(TM)), swellable ion-exchange resins, such as AMBERLITE(TM) IRP 88, formaldehyde-casein (e.g. ESMA SPRENG(TM)), and mixtures thereof.
  • starch or starch derivatives such as sodium starch glycolate (e.g. EXPLOTAB®), or combinations with starch (e.g. PRIMOJEL(TM)), swellable ion-exchange resins, such as AMBERLITE(TM) IRP 88, formaldehyde-casein (e.g. ESMA SPRENG(TM)), and mixtures thereof.
  • the sustained release formulation may include a filling agent or filler.
  • a filling agent refers to an inert substance used as filler to create desired bulk, flow properties, and compression characteristics in preparation of tablets.
  • filling agents for use in the formulation include sucrose, lactose, in particular lactose monohydrate, trehalose, maltose, mannitol and sorbitol, croscarmellose sodium, crospovidone, alginic acid, sodium alginate, methacrylic acid DVB, cross-linked PVP, microcrystalline cellulose, polacrilin potassium, sodium starch glycolate, starch, pregelatinized starch and mixtures thereof.
  • lactose monohydrate is included as a filling agent in an amount of about 10% to about 80%, preferably about 59%, by weight of the tablet.
  • pregelatinized starch is included as a filling agent in an amount of about 5% to about 50%, preferably about 7.5%, by weight of the tablet.
  • the sustained release formulation of the present invention may include a release retarding agent for maintaining a uniform release rate of the drug.
  • retarding agents include, but are not limited to, cellulose ethers, cellulose esters, acrylic acid copolymers, waxes, gums, glyceryl fatty acid esters and sucrose fatty acid esters.
  • the retarding agent is RETALAC® (Meggle), a spray agglomerated blend of 50 parts lactose monohydrate and 50 parts hypromellose.
  • the viscosity of hypromellose used herein may range from 6 mPa*s - 100,000 mPa*s.
  • the viscosity of hypromellose used is 4000 mPa*s. Adjusting the amount of retarding agent in the composition may alter the release rate of the drug.
  • the retarding agent of the formulation of the present invention releases colchicine in a continuous and uniform manner and is adjusted in such a way that about 80% of the active ingredient is released in vitro in the predetermined period of time.
  • the period of time may be not more than 24 hours, not more than 16 hours, not more than 12 hours, not more than 8 hours, not more than 6 hours, not more than 4 hours, not more than 3.5 hours, or not more than 1.5 hours depending on desired attributes of the final product.
  • the release rate can vary based on whether the experiment is conducted in vitro or in vivo. Therefore, if the desired release rate is between about 1.5 to about 3.5 hours in vitro or between about 1.5 to about 6 hours in vitro, the release rate under in vivo conditions, depending on the experimental conditions, may actually be different.
  • the sustained release formulation of the present invention releases colchicine in a continuous and uniform manner in such a way that about 80% of the active ingredient is released in vitro in between about 1.5 and about 3.5 hours.
  • the formulation of the present invention may include a glidant.
  • a glidant can be used to improve powder flow properties prior to and during tableting and to reduce caking.
  • Suitable glidants include colloidal silicon dioxide, magnesium trisilicate, powdered cellulose, talc, tribasic calcium phosphate and the like.
  • talc is included as a glidant in an amount of about 0.05% to about 5%, preferably about 1%, by weight of the tablet.
  • the formulation of the present invention may include a lubricant. Lubricants can be added to pharmaceutical formulations to decrease any friction that occurs between the solid and the die wall during tablet manufacturing.
  • lubricants are added to certain tablet formulations of the present invention including certain embodiments of the formulation described herein.
  • Non-limiting examples of lubricants useful for the core include glyceryl behenate, stearic acid, hydrogenated vegetable oils (such as hydrogenated cottonseed oil (STEROTEX®), hydrogenated soybean oil (STEROTEX® HM) and hydrogenated soybean oil & castor wax (STEROTEX® K)), stearyl alcohol, leucine, polyethylene glycol (MW 1450, suitably 4000, and higher), magnesium stearate, glyceryl monostearate, polyethylene glycol, ethylene oxide polymers (for example, available under the registered trademark CARBOWAX® from Union Carbide, Inc., Danbury, Conn.), sodium lauryl sulfate, magnesium lauryl sulfate, sodium oleate, sodium stearyl fumarate, DL-leucine, colloidal silica, mixtures thereof and others as known in the art.
  • stearic acid is included as a lubricant in an amount of about 0.05% to about 5%,
  • Sweeteners that can also be used in the taste-masking coating of certain embodiments of the matrix dosage forms include glucose (corn syrup), dextrose, invert sugar, fructose, and mixtures thereof (when not used as a carrier); saccharin and its various salts, such as sodium salt; dipeptide sweeteners such as aspartame; dihydrochalcone compounds, glycyrrhizin; Steva Rebaudiana (Stevioside); chloro derivatives or sucrose such as sucralose; and sugar alcohols such as sorbitol, mannitol, xylitol, and the like.
  • hydrogenated starch hydrolysates and the synthetic sweeteners such as 3,6-dihydro-6- methyl-1-1-1, 2, 3-oxathiazin-4-l-2, 2-dioxide, particularly the potassium salt (acesulfame- K), and sodium and calcium salts thereof.
  • the sweeteners can be used alone or in any combination thereof.
  • the formulation of the present invention can further contain one or more pharmaceutically acceptable excipients such as granulating aids or agents, colorants, flavorants, pH adjusters, anti-adherents, glidants and like excipients conventionally used in pharmaceutical compositions.
  • a coloring excipient can be advantageously added as giving rise to visual change preventing abuse. It can color simultaneously the liquid or the particles or one independently of the other.
  • coloring excipients include indigotine, cochineal carminic acid, yellow orange S, allura red AC, iron oxides, cucurmin, riboflavin, tartrazine, quinoline yellow, azorubine, amaranth, carmines, erythosine, red 2G, patented blue V, glittering blue FCF, chlorophylls, copper complexes of chlorophylls, green S, caramel, glittering black BN, carbo medicinalis vegetabilis, brown FK and HT, carotenoids, Annatto extracts, paprika extracts, lycopene, lutein, canthaxanthin, beetroot red, anthocyanes, calcium carbonate, titanium dioxide, aluminium, silver, gold or litholmbin BK or any other coloring excipient suitable for an oral administration.
  • a formulation may be coated.
  • Coatings may provide a variety of functions.
  • coatings may be used, for example, to achieve delayed release, resistance to acid, targeted release in the lower GI tract, avoidance of bad taste in mouth.
  • coatings may be used to protect the API/tablct from light and provide for better mechanical resistance.
  • a coating may serve other functions as well and a person skilled in the art knows the purpose of tablet coating.
  • the pharmaceutical composition and/or the solid carrier particles can be coated with one or more enteric coatings, seal coatings, film coatings, barrier coatings, compress coatings, fast disintegrating coatings, or enzyme degradable coatings. Multiple coatings may be applied for desired performance. Further, one or more of the actives may be provided for immediate release, pulsatile release, controlled release, extended release, delayed release, targeted release, synchronized release, or targeted delayed release.
  • the formulation may include combinations of typical pharmaceutical actives (e.g., pseudephedrin) and vitamins (e.g., Vitamin C), minerals (Ca, Mg, Zn, K) or other supplements (e.g., St. John's Wort, echinacae, amino acids).
  • solid carriers can be made of various component types and levels or thicknesses of coats, with or without an active ingredient. Such diverse solid carriers can be blended in a dosage form to achieve a desired performance.
  • the liquid formulations may be delivered to, and adapted for, oral, nasal, buccal, ocular, urethral, transmucosal, vaginal, topical or rectal delivery, although oral delivery is used mostly.
  • the drug release profile can easily be adapted by adding a coating, e.g., a hard or soft gelatin coating, a starch coating, a resin or polymer coating and/or a cellulosic coating.
  • such dosage forms may be further coated with, for example, a seal coating, an enteric coating, an extended release coating, or a targeted delayed release coating.
  • a seal coating as used herein relates to a mixture of pharmaceutically acceptable excipients that is applied to, combined with, mixed with or otherwise added to the carrier or composition.
  • the coating may be applied to an active that is compressed, molded or extruded and may also include: gelatin, and/or pellets, beads, granules or particles of the carrier or composition.
  • the coating may be applied through an aqueous dispersion or after dissolving in appropriate solvent.
  • the carrier may or may not be fully or partially biodegradable.
  • polymethacrylate acrylic polymers can be employed as coating polymers.
  • the coating is an acrylic resin lacquer used in the form of an aqueous dispersion, such as that which is commercially available from Rohm Pharma under the trade name EUDRAGIT® or from BASF under the trade name KOLLICOAT®.
  • EUDRAGIT® E100 is used as the coating polymer, which is a cationic copolymer based on dimethylaminoethyl methacrylate and neutral methacrylic esters having a average molecular weight is approximately 150,000.
  • Coating methods can consist in spraying a solution of the polymer on the tablets, either in a pan coater or a fluid bed coating apparatus.
  • the solvent may be organic or aqueous, depending on the nature of the polymer used. In a preferable embodiment, the solvent is alcohol. Coating methods are well known in the art.
  • compositions of the present invention can also be formulated as enteric coated delayed release oral dosage forms, i.e., as an oral dosage form of a pharmaceutical composition as described herein that uses an enteric coating to effect release in the lower gastrointestinal tract.
  • enteric coated dosage form will generally include microparticles, microgranules, micropellets or microbeads of the active ingredient and/or other composition components, which are themselves coated or uncoated.
  • the enteric coated oral dosage form may also be a capsule (coated or uncoated) containing pellets, beads or granules of the solid carrier or the composition, which are themselves coated or uncoated.
  • Carriers for use with the present invention include permeable and semipermeable matrices or polymers that control the release characteristics of the formulation.
  • Such polymers include, for example, cellulose acylates, acetates, and other semi-permeable polymers such as those described in U.S. Pat. No. 4,285,987 (hereby incorporated by reference), as well as the selectively permeable polymers formed by the coprecipitation of a polycation and a polyanioni as disclosed in U.S. Pat. Nos. 3,173,876; 3,276,586; 3,541,005; 3,541,006 and 3,546,142 (relevant portions incorporated herein by reference).
  • Other carriers for use with the present invention include, e.g., starch, modified starch, and starch derivatives, gums, including but not limited to xanthan gum, alginic acid, other alginates, benitoniite, veegum, agar, guar, locust bean gum, gum arabic, quince psyllium, flax seed, okra gum, arabinoglactin, pectin, tragacanth, scleroglucan, dextran, amylose, amylopectin, dextrin, etc., cross-linked polyvinylpyrrolidone, ion-exchange resins, such as potassium polymethacrylate, carrageenan (and derivatives), gum karaya, biosynthetic gum, etc.
  • gums including but not limited to xanthan gum, alginic acid, other alginates, benitoniite, veegum, agar, guar,
  • polystyrene- acrylic and its copolymers include: polycarbonates (linear polyesters of carbonic acid); microporous materials (bisphenol, a microporous poly(vinylchloride), micro-porous polyamides, microporous modacrylic copolymers, microporous styrene- acrylic and its copolymers); porous polysulfones, halogenated poly(vinylidene), polychloroethers, acetal polymers, polyesters prepared by esterification of a dicarboxylic acid or anhydride with an alkylene polyol, poly(alkylenesulfides), phenolics, polyesters, asymmetric porous polymers, cross-linked olefin polymers, hydrophilic microporous homopolymers, copolymers or interpolymers having a reduced bulk density, and other similar materials, poly (urethane), cross-linked chain-extended poly (urethane), poly(imides), poly(benzimi
  • Additional additives and their levels, and selection of a primary coating material or materials will depend on the following properties: pH levels at target site, desirability to make tablet pH dependent or pH independent, solubility in alcohol, resistance to dissolution and disintegration in the stomach; impermeability to gastric fluids and drug/carrier/enzyme while in the stomach; ability to dissolve or disintegrate rapidly at the target intestine site; physical and chemical stability during storage; non-toxicity; easy application as a coating (substrate friendly); and economical practicality.
  • a controlled-release tablet or capsule or the like comprises colchicine as a core coated with an immediate release layer.
  • a controlled-release double layer tablet or capsule or the like comprises a layer of sustained release and a layer of immediate release.
  • a controlled-release tablet with more than two layers comprises (i) one or two more layers of substance controlling the sustained release and (ii) one or two more layers of immediate release.
  • the composition comprising colchicine is further coated with at least one release- slowing intermediate layer of slightly soluble intermediate layer to control release of colchicine.
  • colchicine immediate release dosage forms have been used in the treatment of gout or FMF.
  • all approved pharmaceuticals containing colchicine are approved only for gout and/or FMF and are immediate release tablets.
  • Colchicine can be used in the prevention of certain other inflammatory diseases such as pericarditis, PPS and, most recently, patients with stable coronary heart diseases.
  • the difference between treatment and prevention with regard to colchicine is that in treatment, an overt disease and/or ongoing inflammation has to be treated.
  • high levels of colchicine are required, which usually goes hand in hand with unwanted side effects, most prominently gastrointestinal insults, as well as increased risk of colchicine related toxicity.
  • colchicine formulated as a sustained release preparation, as described above.
  • An advantage of colchicine administered as sustained release is, e.g., a flattening of the serum level curve (lower but broader peak levels) reduces the incidence of serious adverse events related to colchicine toxicity, also in case of potential drug interactions, thereby increasing compliance.
  • Much of colchicine related toxicity comes from the fact that one or both of the excretion pathways (liver and kidney) is reduced in its activity, either by other drugs or by a disease (e.g. kidney insufficiency).
  • a disease e.g. kidney insufficiency
  • the body In the case of a slower and extended drug absorption (extended release), the body has also more time to excrete the colchicine from the system. In this case, it is less likely that colchicine levels reach toxic levels in case of defective excretion (due to drug interaction or disease).
  • colchicine administered as a sustained release formulation is that plasma levels remain more evenly distributed (i.e., the variability of plasma levels, such as the differences in Cmax, Tmax, AUC or other pharmacokinetic parameters, among patients is reduced), resulting in fewer “non-responders” to the treatment.
  • administration of colchicine as sustained release is resistant to dose dumping, therefore the dissolution of the composition is not significantly influenced by alcohol.
  • colchicine does not have to go deep into the tissue (like for gout), it may be active in the blood system directly (in the vessels) where it acts on the plaques and especially on inflammatory blood cells (neutrophils). This means, less total colchicine and lower serum levels can be therapeutic. Fast and high colchicine levels, e.g., as for treating an acute gout flare can be avoided. Thus, lower levels of colchicine, e.g., about 0.1 to about 0.75mg sustained release formulations as described above (or even less frequent doses), may be sufficient to achieve the desired clinical outcome.
  • colchicine In the normal situation, most colchicine is absorbed from the small intestine and most passes the liver (some is also excreted in the urine via kidney). There it is metabolized but quite a large proportion of colchicine goes through the liver un-metabolized. This means, it goes through the liver into the bile and from there it is excreted into the big intestine (colon). There it can be resorbed into the body again which leads to the characteristic second peak (accounts for about 50% of totally absorbed colchicine and is thought to be responsible for gastrointestinal problems, such as diarrhea). If colchicine is formulated as a sustained release preparation as described above, a slower release of colchicine results in a slower resorption.
  • Colchicine administered as sustained release in accordance with the present invention may also be beneficial for other known side/adverse effects associated with colchicine treatment/administration (the skilled person is well aware of the adverse effects that may occur upon colchicine administration or colchicine treatment).
  • administration of colchicine as sustained release, as described above results in a safety increase and safety benefit.
  • the current invention additionally encompasses a method of preparing formulations of colchicine, comprising a sustained release component, and an optional immediate release component, wherein colchicine is released from the formulation at the sustained rate along the pre-determined or desired release profile.
  • the colchicine compositions described in the present invention is in the form of a tablet.
  • tablette means a compressed pharmaceutical dosage form of any shape or size.
  • the tablets described herein may be obtained from the compositions comprising colchicine and a pharmaceutically acceptable excipient. Any of the colchicine compositions can be in the form of any other dosage form known in the art, specifically, any oral dosage form, for example a capsule.
  • a controlled release formulation for use in oral dosage forms.
  • the formulation includes a mixture containing hypromellose as a hydrophilic matrix, which is effective to provide controlled release of a pharmaceutically active ingredient.
  • Matrix systems are well known in the art.
  • the drug is homogenously dispersed in a polymer in association with conventional excipients. This admixture is typically compressed under pressure to produce a tablet.
  • the API is released from the tablet by diffusion and erosion.
  • Matrix systems are described in detail by (i) Handbook of Pharmaceutical Controlled Release Technology, Ed. D. L. Wise, Marcel Dekker, Inc. New York, N.Y. (2000), and (ii) Treatise on Controlled Drug Delivery, Fundamentals, Optimization, Applications, Ed. A. Kydonieus, Marcel Dekker, Inc. New York, N.Y. (1992), the contents of both of which are hereby incorporated by reference.
  • the tablet surface wets and the polymer begins to partially hydrate forming an outer gel layer.
  • This outer gel layer becomes fully hydrated and begins to erode into the aqueous fluids. Water continues to permeate toward the core of the tablet permitting another gel layer to form beneath the dissolving outer gel layer.
  • These successive concentric gel layers sustain uniform release of the API by diffusion from the gel layer and exposure through tablet erosion.
  • the hypromellose when included in a compressed tablet matrix, the hypromellose provides a hydrophilic swellable structure capable of functioning as the gel layer. In this way, the drug release is controlled.
  • the colchicine formulation of the present invention can be manufactured by either wet or dry granulation of a colchicine composition, blending the resulting granulate with excipients, and then compressing the composition into tablets.
  • wet granulation is used to prepare wet granules comprising colchicine.
  • a granulating liquid is used in wet granulation process. Both aqueous and non-aqueous liquids may be used as the granulating liquid.
  • the granulating liquid is an aqueous liquid, or more specifically, purified or de-ionized water. The amount of the granulating liquid used may depend on many factors, for example, the type of the granulating liquid, the amount of the granulating liquid used, the type of excipient used, the nature of the active agent, and the active agent loading.
  • the colchicine particles and suitable excipients are mixed with the granulating liquid for a sufficiently long period to facilitate good distribution of all starting materials and good content uniformity.
  • Wet granulation is generally performed at temperatures between about 20° C. to about 35° C., or more specifically, at room temperature (about 25° C.).
  • the granulate is dried at increased temperatures to yield a dry granulate.
  • the step of drying may be performed for a sufficiently long period until the desired residual moisture content is reached. In an embodiment, this may be about 45°C for about 12-48 hours. It should be appreciated that the overall time to perform the granulation process may depend on a variety of factors, including but not limited to, the solvents used, batch size, instruments used, etc.
  • any equipment may be used to contact the granulating liquid with the colchicine and the excipients as long as uniform distribution of the granulating liquid is achieved.
  • small-scale production can be achieved by mixing and wetting the colchicine and the excipients in mortars or stainless steel bowls, while for larger quantities, V-blenders with intensifier bars, planetary mixers, rotary granulators, high shear granulators, and fluid- bed granulation equipment may be used.
  • the granulator is a high shear granulator.
  • a method of making a colchicine composition comprises wet granulating colchicine with pharmaceutically acceptable excipients and a granulating liquid to obtain wet granules, and mixing the granules in a next step with a second excipient to obtain a colchicine composition.
  • the pharmaceutically acceptable excipient comprises a binder and a filler.
  • the binder may be Hypromellose.
  • the filler may be lactose monohydrate and pregelatinized starch.
  • purified water is used as the granulating liquid.
  • the second excipient mixed with the granules may be a filler.
  • the filler may be lactose monohydrate.
  • the colchicine compositions can contain about 0.1 wt % to about 10 wt %, or more specifically, about 0.25 wt % to about 0.75 wt %, of colchicine, based on the total weight of the colchicine composition.
  • the method of making a composition comprises wet granulating colchicine with a pharmaceutically acceptable excipient to obtain wet granules, and mixing the granules with a filler to obtain a colchicine composition.
  • the method further includes drying the mixture.
  • the wet granules are dried to obtain dried granules, and then the dried granules are mixed with a binder, a filler, or both to obtain the composition.
  • the dried granules can be milled to obtain milled granules before mixing the milled dried granules with the binder, a filler, or both.
  • the method can further include mixing the colchicine composition with a glidant, a lubricant, or both to obtain a blend or compressing the blend to obtain a tablet.
  • the glidant may be Talc.
  • the lubricant may be Stearic acid.
  • the method can further include coating the tablet.
  • a method of making a colchicine tablet comprises wet granulating colchicine with a pharmaceutically acceptable excipient to obtain wet granules; drying the wet granules to obtain dried granules; milling the dried granules to obtain milled granules; mixing the milled granules with a filler to obtain the composition; mixing the composition with a glidant, a lubricant, or both to obtain a blend; and compressing the blend to obtain a colchicine tablet of the present invention.
  • the wet granules are dried to obtain dried granules before mixing with a second excipient, for example a filler.
  • Wet granules can be dried by any suitable means to remove the granulating liquid and to form dried granules containing colchicine and the pharmaceutically acceptable excipient.
  • the conditions and duration of drying depend on factors such as the liquid used and the weight of the granulating particles. Examples of suitable drying methods include, but are not limited to, tray drying, forced air drying, microwave drying, vacuum drying and fluid bed drying.
  • dried granules may be mixed directly with an excipient, for example, a filler, a binder, or a lubricant, for further processing.
  • dried granules may optionally be subjected to additional processing steps prior to mixing with the excipient.
  • dried granules may be sized to reduce particle size prior to mixing with an excipient. Exemplary sizing operations include milling or sieving. Any suitable equipment for reducing the particle size may be used in the present invention.
  • Suitable excipients may be added extragranularly and mixed with the granules to form colchicine compositions.
  • extragranular or “extragranularly” means that the referenced material, for example, a suitable excipient, is added or has been added as a dry component after wet granulation.
  • a filler, a binder, a glidant and a lubricant are added extragranularly to the granules and mixed to form a blend.
  • the blend may be encapsulated directly into capsule shells, for example, hard gelatin shells, to form capsule formulations. Alternatively, the blend may be compressed into tablets.
  • the granules are dried granules or milled, dried granules.
  • Mixing can be carried out for a sufficient time to produce homogeneous mixtures or blends. Mixing may be accomplished by blending, stiffing, shaking, tumbling, rolling, or by any other method to achieve a homogeneous blend. In some embodiments, the components to be mixed are combined under low shear conditions in a suitable apparatus, such as a V-blender, tote blender, double cone blender or any other apparatus capable of functioning under low shear conditions.
  • a suitable apparatus such as a V-blender, tote blender, double cone blender or any other apparatus capable of functioning under low shear conditions.
  • the homogenous mixtures or blends are then compressed using any method suitable in the industry.
  • the mechanical force will define the physical properties of the tablets, especially the crushing strength of the resulting tablet.
  • the mechanical strength interacts with the initial swelling of the tablet and dilution speed of the tablet core. This effect is well known in the art and can be adjusted and controlled during the lifecycle of the product.
  • the compression strengths used may range from about 30N to about 130N. In one embodiment, the compression strength may be about 100N. In another embodiment, the compression strength may be about 100N +/- 15N.
  • the colchicine tablets prepared from the above described methods exhibit acceptable physical characteristics including good friability and hardness. As per EP and USP guidelines, the colchicine tablets disclosed herein have friability in the range of about 0% to less than about 1%.
  • the colchicine tablet can be coated. Coating the tablet may be performed by any known process.
  • a coating for the colchicine tablet disclosed herein can be any suitable coating, such as, for example, a functional or a non-functional coating, or multiple functional or non-functional coatings.
  • functional coating is meant to include a coating that modifies the release properties of the total formulation, for example, a sustained- release coating.
  • non-functional coating is meant to include a coating that is not a functional coating, for example, a cosmetic coating.
  • a non-functional coating can have some impact on the release of the active agent due to the initial dissolution, hydration, perforation of the coating, etc., but would not be considered to be a significant deviation from the non-co ated composition.
  • a colchicine composition comprises colchicine, a binder, a filler, a retarding agent, a glidant, and a lubricant.
  • a colchicine composition comprises about 0.25 to about 0.75 mg colchicine; about 10 to about 80 mg lactose monohydrate; about 5 to about 50 mg pregelatinized starch; about 1 to about 30 mg Hypromellose 6mPa*s; about 5 to about 40 mg Retalac (compound of lactose monohydrate and Hypromellose 4000mPa*s 50/50 w/w %); about 0.5 to about 5 mg Talc; and about 0.5 to about 5 mg Stearic acid 50.
  • the colchicine composition comprises about 0.5 mg colchicine, about 59 mg lactose monohydrate; about 7.5 mg pregelatinized starch; about 1 mg Hypromellose 6mPa*s; about 30 mg Retalac (compound of lactose monohydrate and Hypromellose 4000mPa*s 50/50 w/w %); about 1 mg Talc; and about 1 mg Stearic acid 50.
  • the colchicine dosage form has a total weight of about 100 mg.
  • the colchicine composition can be in the form of a tablet.
  • the present invention also presents a method to treat and/or prevent, or inhibit, delay, or reduce tumor growth or metastases in a subject in need of such inhibition, delay, or reduction, e.g., a lung cancer patient, comprising administering to the subject a therapeutically effective amount of a colchicine formulation of the present invention.
  • the method of the current invention possesses the flexibility to selectively adjust the pharmacokinetics of the administered formulations depending on the nature of the condition and needs of the patients due to the novel design of the colchicine formulation that comprises an extended release component and an optional immediate release component, and the release profiles of both components can be selectively modified during the preparation process as described above to comply with the predetermined release profile.
  • treatment includes the application or administration of a colchicine formulation as described herein to a patient, where the patient has, or has the risk of developing lung cancer.
  • treatment is also intended to include the application or administration of a pharmaceutical composition comprising the colchicine formulation, to a patient, where the patient has, or has the risk of developing lung cancer.
  • Colchicine is an anti-inflammatory agent, known as the drug of choice for management of gout. It has been shown that its anti-inflammatory effect is mediated by inhibition of the pro-inflammatory cytokines IL-1 and IL-Ib release. Moreover, by restraint of the NF-KB pathway and blocking cell mitosis, colchicine may exert an inhibitory effect on tumorigenesis. Addition of colchicine to human LPS stimulated PBMC exerted a disruptive effect on cellular microtubules with a consequent increased IL-Ib and a decreased TNF-a release.
  • tumor types that on average harbor a greater concentration of infiltrating leukocytes include: melanoma, colorectal cancer, non-small cell lung cancer and head and neck squamous cell carcinoma.
  • the lymphocytic infiltrate is thought to reflect how foreign or "visible" a tumor is to the immune system which in turn is connected to the extent of “disregulation” or “aberration” within the tumor.
  • they despite effective recruitment of immune cells to the tumor, they generally do not succumb to immune- mediated regression. This is believed to be the effect of suppressive or resistance mechanisms adopted by the tumor.
  • M2 macrophage myeloid derived suppressor cells
  • regulatory T cells regulatory T cells
  • dendritic cells dendritic cells on defective antigen presentation
  • suppressive cytokines impaired costimulation for effector T cells and antigen loss.
  • checkpoint inhibitors In tumors with high numbers of infiltrating leukocytes, a new class of therapeutics called checkpoint inhibitors has been shown to release or remove suppressive immune elements including regulatory T cells.
  • the leading class of these molecules target the proteins PDCD1, PDCDL1 or CTLA4 although there are others approved and still many others in various stages of clinical approval across the range of suppressive mechanisms.
  • “Disregulation” in tumors with high lymphocytic content is highly correlated with the total number of genetic mutations that tumor harbors which in turn translates to the production of defective and novel (foreign) proteins which serve as antigens that prime the immune response. In a very simplistic sense:
  • colchicine Given its anti-inflammatory effect, colchicine is anticipated to be most effective in the scenario described above including reduction of IL-10 as has been published.
  • some of the anti-tumor activity may come from direct effects of colchicine on tumor cells. It is known that colchicine is internalized especially by fast growing cells. Colchicine may inhibit cell division of tumor cells by inhibition of micro tubule function.
  • Colchicine in the context of the present invention can be used to treat and/or prevent, or inhibit, delay, or reduce tumor growth or metastases in a subject in need of such inhibition, delay, or reduction, e.g., a lung cancer patient.
  • cancers that can be treated here include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, leukemia, squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, gastric cancer, pancreatic cancer, neuroendocrine cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, brain cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, esophageal cancer, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, head and neck cancer, and a combination thereof.
  • colchicine can be used to inhibit, delay, or reduce tumor growth or metastases in a subject having lung cancer.
  • the colchicine formulation herein can be used to promote a positive therapeutic response with respect to tumor growth and/or metastases.
  • positive therapeutic response with respect to cancer treatment is intended an improvement in the disease in association with the anti-tumor activity of these binding molecules, e.g., antibodies or fragments thereof, and/or an improvement in the symptoms associated with the disease.
  • the methods provided herein are directed to inhibiting, preventing, reducing, alleviating, delaying, or lessening growth of a tumor and/or the development of metastases of primary tumors in a patient. That is the prevention of distal tumor outgrowths, can be observed.
  • an improvement in the disease may be characterized as a complete response.
  • complete response is intended an absence of clinically detectable metastases with normalization of any previously abnormal radiographic studies, e.g. at the site of the primary tumor or the presence of tumor metastases in bone marrow.
  • an improvement in the disease may be categorized as being a partial response.
  • partial response is intended at least about a 50% decrease in all measurable metastases (i.e., the number of tumor cells present in the subject at a remote site from the primary tumor).
  • an improvement in the disease may be categorized as being relapse free survival or “progression free survival”.
  • relapse free survival is intended the time to recurrence of a tumor at any site.
  • progression free survival is the time before further growth of tumor at a site being monitored can be detected.
  • Inhibition, delay, or reduction of metastases can be assessed using screening techniques such as imaging, for example, fluorescent antibody imaging, bone scan imaging, and tumor biopsy sampling including bone marrow aspiration (BMA), or immunohistochemistry.
  • imaging for example, fluorescent antibody imaging, bone scan imaging, and tumor biopsy sampling including bone marrow aspiration (BMA), or immunohistochemistry.
  • BMA bone marrow aspiration
  • the subject undergoing therapy with a colchicine molecule of the present invention can experience the beneficial effect of an improvement in the symptoms associated with the disease.
  • Clinical response can be assessed using screening techniques such as magnetic resonance imaging (MRI) scan, x-radiographic imaging, computed tomographic (CT) scan, flow cytometry or fluorescence-activated cell sorter (FACS) analysis, histology, gross pathology, and blood chemistry, including but not limited to changes detectable by ELISA, RIA, chromatography, and the like.
  • MRI magnetic resonance imaging
  • CT computed tomographic
  • FACS fluorescence-activated cell sorter
  • the colchicine formulation as described herein is useful in the prevention of tumor growth and/or metastases.
  • the term “prevention” is well known in the art.
  • a patient/subject suspected of being prone to suffer from a disorder or disease as defined herein may, in particular, benefit from a prevention of the disorder or disease.
  • the subject/patient may have a susceptibility or predisposition for a disorder or disease, including but not limited to hereditary predisposition.
  • Such a predisposition can be determined by standard assays, using, for example, genetic markers or phenotypic indicators.
  • a disorder or disease to be prevented in accordance with the present invention has not been diagnosed or cannot be diagnosed in the patient/subject (for example, the patient/subject does not show any clinical or pathological symptoms).
  • prevention comprises the use of compounds of the present invention before any clinical and/or pathological symptoms are diagnosed or determined or can be diagnosed or determined by the attending physician.
  • Prevention includes, without limitation, to avoid the disease or condition from occurring in patient and/or subject that may be predisposed to the disease but does not yet experience or exhibit symptoms of the disease (prophylactic treatment).
  • Colchicine according to the present invention may also be used in combination with at least one other immune modulating therapy, to inhibit, delay, or reduce tumor growth or metastases in a subject in need of such inhibition, delay, or reduction, e.g., a cancer patient.
  • the immune modulating therapy may include cancer vaccines, immuno stimulatory agents, adoptive T cell or antibody therapy, and inhibitors of immune checkpoint blockade (Lizee et al. 2013. Harnessing the Power of the Immune System to Target Cancer. Annu. Rev. Med. Vol. 64 No. 71-90).
  • Cancer vaccines activate the body’s immune system and natural resistance to an abnormal cell, such as cancer, resulting in eradication or control of the disease.
  • Cancer vaccines generally consist of a tumor antigen in an immunogenic formulation that activates tumor antigen-specific helper cells and/or CTLs and B cells.
  • Vaccines can be in a variety of formulations, including, but not limited to, dendritic cells, especially autologous dendritic cells pulsed with tumor cells or tumor antigens, heterologous tumor cells transfected with an immune stimulating agent such as GM-CSF, recombinant vims, or proteins or peptides that are usually administered together with a potent immune adjuvant such as CpG.
  • Immuno stimulatory agents act to enhance or increase the immune response to tumors, which is suppressed in many cancer patients through various mechanisms.
  • Immune modulating therapies may target lymphocytes, macrophages, dendritic cells, natural killer cells (NK Cell), or subsets of these cells such as cytotoxic T lymphocytes (CTL) or Natural Killer T (NKT) cells. Because of interacting immune cascades, an effect on one set of immune cells will often be amplified by spreading to other cells, e.g. enhanced antigen presenting cell activity promotes response of T and B lymphocytes.
  • immunostimulatory agents include, but are not limited to, HER2, cytokines such as G-CSF, GM-CSF and IL-2, cell membrane fractions from bacteria, glycolipids that associate with CD Id to activate Natural Killer T (NKT) cells, CpG oligonucleotides.
  • Macrophages myelophagocytic cells of the immune system, are a fundamental part of the innate defense mechanisms, which can promote specific immunity by inducing T cell recruitment and activation. Despite this, their presence within the tumor microenvironment has been associated with enhanced tumor progression and shown to promote cancer cell growth and spread, angiogenesis and immunosuppression. Key players in the setting of their phenotype are the microenvironmental signals to which macrophages are exposed, which selectively tune their functions within a functional spectrum encompassing the Ml (tumor inhibiting macrophage) and M2 (tumor promoting macrophage) extremes. Sica et al., Seminars in Cancer Biol. ‘:349-355 (2008).
  • TAMs tumor-associated macrophages
  • Adoptive Cell Transfer may employ T cell-based cytotoxic responses to attack cancer cells.
  • Autologous T cells that have a natural or genetically engineered reactivity to a patient's cancer are generated and expanded in vitro and then transferred back into the cancer patient.
  • adoptive transfer of in vitro expanded autologous tumor-infiltrating lymphocytes was an effective treatment for patients with metastatic melanoma. (Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME (April 2008). "Adoptive cell transfer: a clinical path to effective cancer immunotherapy". Nat. Rev. Cancer 8 (4): 299-308). This can be achieved by taking T cells that are found within resected patient tumor.
  • TIL tumor- infiltrating lymphocytes
  • TIL-2 tumor- infiltrating lymphocytes
  • IL-2 anti-CD3 and allo-reactive feeder cells.
  • IL-2 anti-CD3
  • allo-reactive feeder cells These T cells are then transferred back into the patient along with exogenous administration of IL-2 to further boost their anti-cancer activity.
  • autologous T cells have been transduced with a chimeric antigen receptor that renders them reactive to a targeted tumor antigen (Liddy etal., Nature Med. 18: 980-7, (2012); Grupp et ah, New England J. Med. 368. T509- 18, (2013)).
  • Other adoptive cell transfer therapies employ autologous dendritic cells exposed to natural or modified tumor antigens ex vivo that are re-infused into the patient.
  • Provenge is such an PDA approved therapy in which autologous cells are incubated with a fusion protein of prostatic acid phosphatase and GM-CSP to treat patients with prostate tumors.
  • GM-CSP is thought to promote the differentiation and activity of antigen presenting dendritic cells (Small et ah, J. Clin. Oncol. 18: 3894-903(2000); US Patent 7,414,108)).
  • Immune checkpoint blockade therapies enhance T- cell immunity by removing a negative feedback control that limits ongoing immune responses. These types of therapies target inhibitory pathways in the immune system that are crucial for modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage. Tumors can evolve to exploit certain immune-checkpoint pathways as a major mechanism of immune resistance against T cells that are specific for tumor antigens. Since many immune checkpoints are initiated by ligand-receptor interactions, these checkpoints can be blocked by antibodies to either receptor or ligand or may be modulated by soluble recombinant forms of the ligands or receptors.
  • immune checkpoint blockade therapies are those which target Cytotoxic T- lymphocyte-associated antigen 4 (CTLA-4), PD-1, its ligand PD-L1, LAG3 and B7-H3.
  • CTLA-4 Cytotoxic T- lymphocyte-associated antigen 4
  • PD-1 its ligand PD-L1, LAG3 and B7-H3.
  • Cyclophosphamide a commonly used chemotherapeutic agent, can enhance immune responses. Cyclophosphamide differentially suppresses the function of regulatory T cells (Tregs) relative to effector T cells. Tregs are important in regulating anticancer immune responses. Tumor- infiltrating Tregs have previously been associated with poor prognosis. While agents that target Tregs specifically are currently unavailable, cyclophosphamide has emerged as a clinically feasible agent that can preferentially suppress Tregs relative to other T cells and, therefore, allows more effective induction of antitumor immune responses.
  • Tregs regulatory T cells
  • Tregs are important in regulating anticancer immune responses. Tumor- infiltrating Tregs have previously been associated with poor prognosis. While agents that target Tregs specifically are currently unavailable, cyclophosphamide has emerged as a clinically feasible agent that can preferentially suppress Tregs relative to other T cells and, therefore, allows more effective induction of antitumor immune responses.
  • therapy with a colchicine molecule may be combined with either low dose chemotherapy or radiation therapy.
  • standard chemotherapy is often immunosuppressive
  • low doses of chemotherapeutic agents such as cyclophosphamide, doxorubicin, and paclitaxel have been shown to enhance responses to vaccine therapy for cancer (Machiels et ah, Cancer Res. 57:3689-3697 (2001)).
  • chemotherapy may differentially inactivate T regulatory cells (Treg) and myeloid derived suppressor cells (MDSC) that negatively regulate immune responses in the tumor environment.
  • Reg T regulatory cells
  • MDSC myeloid derived suppressor cells
  • Radiation therapy has been generally employed to exploit the direct tumorcidal effect of ionizing radiation. Indeed, high dose radiation can, like chemotherapy, be immunosuppressive.
  • the immune modulating therapy may be an immune modulating agent, including, but not limited to, interleukins such as IL-2, IL-7, IL-12; cytokines such as granulocyte-macrophage colony- stimulating factor (GM-CSF), interferons; various chemokines such as CXCL13, CCL26, CXCL7; antagonists of immune checkpoint blockades such as anti-CTLA-4, anti-PD-1, anti-PD-Ll, anti-LAG3 and anti-B7-H3; synthetic cytosine phosphate-guanosine (CpG), oligodeoxynucleotides, glucans, modulators of regulatory T cells (Tregs) such as cyclophosphamide, or other immune modulating agents.
  • interleukins such as IL-2, IL-7, IL-12
  • cytokines such as granulocyte-macrophage colony- stimulating factor (GM-CSF), interferons
  • the immune modulating agent is an agonist antibody to 4- IBB (CD 137).
  • 4- IBB agonist antibody to 4- IBB
  • the additional immune modulating therapy is administered prior to, during, or subsequent to the colchicine molecule.
  • the combined therapies comprise administration of a colchicine molecule in combination with administration of another immune modulating agent
  • the methods of the invention encompass co-administration, using separate formulations or a single pharmaceutical formulation, with simultaneous or consecutive administration in either order.
  • the immune modulating therapy may be a cancer therapy agent, including, but not limited to, surgery or surgical procedures (e.g. splenectomy, hepatectomy, lymphadenectomy, leukophoresis, bone marrow transplantation, and the like); radiation therapy; chemotherapy, optionally in combination with autologous bone marrow transplant, or other cancer therapy; where the additional cancer therapy is administered prior to, during, or subsequent to the colchicine therapy.
  • surgery or surgical procedures e.g. splenectomy, hepatectomy, lymphadenectomy, leukophoresis, bone marrow transplantation, and the like
  • radiation therapy e.g. splenectomy, hepatectomy, lymphadenectomy, leukophoresis, bone marrow transplantation, and the like
  • chemotherapy optionally in combination with autologous bone marrow transplant, or other cancer therapy
  • the additional cancer therapy is administered prior to, during, or subsequent to the colchicine therapy.
  • the methods of the invention encompass co administration, using separate formulations or a single pharmaceutical
  • treatment includes the application or administration of colchicine as described herein as a single agent or in combination with at least one other immune modulating therapy to a patient, or application or administration of colchicine as a single agent or in combination with at least one other immune modulating therapy to an isolated tissue or cell line from a patient, where the patient has, or has the risk of developing metastases of cancer cells.
  • treatment is also intended to include the application or administration of a pharmaceutical composition comprising colchicine in combination with at least one other immune modulating therapy or application or administration of a pharmaceutical composition comprising colchicine and at least one other immune modulating therapy to an isolated tissue or cell line from a patient, where the patient has, or has the risk of developing metastases of cancer cells.
  • Colchicine as single agents or in combination with at least one other immune modulating therapy are useful for the treatment of various malignant and non-malignant tumors.
  • anti-tumor activity is intended a reduction in the rate of IL-Ib production or accumulation associated directly with the tumor or indirectly with stromal cells of the tumor environment, and hence a decline in growth rate of an existing tumor or of a tumor that arises during therapy, and/or destruction of existing neoplastic (tumor) cells or newly formed neoplastic cells, and hence a decrease in the overall size of a tumor and/or the number of metastatic sites during therapy.
  • therapy with colchicine as a single agent or in combination with at least one other immune modulating therapy causes a physiological response, for example, a reduction in metastases, that is beneficial with respect to treatment of disease states associated with IL-Ib -expressing cells in a human.
  • the invention relates to the use of colchicine according to the present invention as a single agent or in combination with at least one other immune modulating therapy as a medicament, in the treatment or prophylaxis of cancer or for use in a precancerous condition or lesion to inhibit, reduce, prevent, delay, or minimalize the growth or metastases of tumor cells.
  • colchicine as a single agent or in combination with at least one other immune modulating therapy can be used to promote a positive therapeutic response with respect to a malignant human cell.
  • positive therapeutic response with respect to cancer treatment is intended an improvement in the disease in association with the anti-tumor activity of these binding molecules, e.g., antibodies or fragments thereof, and/or an improvement in the symptoms associated with the disease.
  • the methods provided herein are directed to inhibiting, preventing, reducing, alleviating, delaying, or lessening growth of a tumor and/or the development of metastases of primary tumors in a patient. That is the prevention of distal tumor outgrowths, can be observed.
  • an improvement in the disease may be characterized as a complete response.
  • complete response is intended an absence of clinically detectable metastases with normalization of any previously abnormal radiographic studies, e.g. at the site of the primary tumor or the presence of tumor metastases in bone marrow.
  • an improvement in the disease may be categorized as being a partial response.
  • partial response is intended at least about a 50% decrease in all measurable metastases (i.e., the number of tumor cells present in the subject at a remote site from the primary tumor).
  • an improvement in the disease may be categorized as being relapse free survival or “progression free survival”.
  • relapse free survival is intended the time to recurrence of a tumor at any site.
  • progression free survival is the time before further growth of tumor at a site being monitored can be detected.
  • Inhibition, delay, or reduction of metastases can be assessed using screening techniques such as imaging, for example, fluorescent antibody imaging, bone scan imaging, and tumor biopsy sampling including bone marrow aspiration (BMA), or immunohistochemistry.
  • imaging for example, fluorescent antibody imaging, bone scan imaging, and tumor biopsy sampling including bone marrow aspiration (BMA), or immunohistochemistry.
  • BMA bone marrow aspiration
  • the subject undergoing therapy with colchicine as described herein can experience the beneficial effect of an improvement in the symptoms associated with the disease.
  • Clinical response can be assessed using screening techniques such as magnetic resonance imaging (MRI) scan, x-radiographic imaging, computed tomographic (CT) scan, flow cytometry or fluorescence-activated cell sorter (FACS) analysis, histology, gross pathology, and blood chemistry, including but not limited to changes detectable by ELISA, RIA, chromatography, and the like.
  • MRI magnetic resonance imaging
  • CT computed tomographic
  • FACS fluorescence-activated cell sorter
  • colchicine is in fixed (within the same pharmaceutical preparation) or unfixed (different pharmaceutical preparation) combination.
  • Fixed combination can mean, e.g., in a single tablet, solution, cream, capsule, gel, ointment, salve, patch, suppository or transdermal delivery system.
  • Unfixed combination as used herein is to be understood as meaning that the active ingredients/components are in more than one vehicle (e.g. tablets, solutions, creams, capsules, gels, ointments, salves, patches, suppositories or transdermal delivery systems).
  • Each of the vehicles can contain a desired pharmaceutical composition or active component.
  • the route of administration of the colchicine formulation can be, for example, oral, parenteral, by inhalation or topical.
  • parenteral as used herein includes, e.g., intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, rectal, or vaginal administration. While all these forms of administration are clearly contemplated as being within the scope of the invention, an example of a form for administration would be a solution for injection, in particular for intravenous or intraarterial injection or drip.
  • a suitable pharmaceutical composition for injection can comprise a buffer (e.g.
  • colchicine as a single agent or in combination with at least one other immune modulating therapy can be delivered directly to the site of the adverse cellular population thereby increasing the exposure of the diseased tissue to the therapeutic agent.
  • Colchicine when used as a composition in the context of the present invention may include one or more pharmaceutically acceptable carriers and thus may be prepared in the form of a local formulation, in order for it to be administered.
  • the pharmaceutically acceptable carrier may include saline, sterile water, linger liquid, buffer saline, a dextrose solution, a malto dextrin solution, glycerol, ethanol and mixtures of one or more thereof, and also may include an additive such as an antioxidant, a buffer, a bacteriostatic agent or the like, as necessary.
  • a diluent, a dispersant, a surfactant, a binder and a lubricant may be added when the composition according to the present invention is prepared, e.g., in the form of a local formulation such as an ointment, lotion, cream, gel, skin emulsion, skin suspension, patch or spray.
  • Non-limiting examples for administration of the compound and or compositions according to the present invention include coated and uncoated tablets, soft gelatine capsules, hard gelatine capsules, lozenges, troches, solutions, emulsions, suspensions, syrups, elixiers, powders and granules for reconstitution, dispersible powders and granules, medicated gums, chewing tablets and effervescent tablets.
  • the composition according to the present invention can administered in any pharmaceutical form for oral (e.g. solid, semi solid, liquid), dermal (e.g. dermal patch), sublingual, parenteral (e.g. injection), ophthalmic (e.g. eye drops, gel or ointment) or rectal (e.g. suppository) administration.
  • the composition is formulated as a tablet, capsule, suppository, dermal patch or sublingual formulation.
  • compositions used in this invention comprise pharmaceutically acceptable carriers, including, e.g., ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene - polyoxypropylene-block polymers, polyethylene glycol, and wool fat.
  • pharmaceutically acceptable carriers including, e.g., ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as
  • Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions.
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include, e.g., water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • pharmaceutically acceptable carriers include, but are not limited to, 0.01-0.1 M and preferably 0.05 M phosphate buffer or 0.8% saline.
  • Intravenous vehicles include sodium phosphate solutions, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, antioxidants, chelating agents, and inert gases and the like.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the composition must be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and will preferably be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Suitable formulations for use in the therapeutic methods disclosed herein are described in Remington's Pharmaceutical Sciences (Mack Publishing Co.) 16th ed. (1980).
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal and the like.
  • isotonic agents for example, sugars, polyalcohols, such as mannitol, sorbitol, or sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum mono stearate and gelatin.
  • sterile injectable solutions can be prepared by incorporating an active compound (e.g ., colchicine according to the present invention) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated herein, as required, followed by filtered sterilization.
  • an active compound e.g ., colchicine according to the present invention
  • dispersions are prepared by incorporating the active compound into a sterile vehicle, which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying, which yields a powder of an active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the preparations for injections are processed, filled into containers such as ampoules, bags, bottles, syringes or vials, and sealed under aseptic conditions according to methods known in the art. Further, the preparations may be packaged and sold in the form of a kit. Such articles of manufacture can have labels or package inserts indicating that the associated compositions are useful for treating a subject suffering from, or predisposed to a disease or disorder.
  • Parenteral formulations can be a single bolus dose, an infusion or a loading bolus dose followed with a maintenance dose. These compositions can be administered at specific fixed or variable intervals, e.g., once a day, or on an "as needed" basis.
  • compositions used in this invention can be orally administered in an acceptable dosage form including, e.g., capsules, tablets, aqueous suspensions or solutions. Certain pharmaceutical compositions also can be administered by nasal aerosol or inhalation. Such compositions can be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other conventional solubilizing or dispersing agents.
  • the amount of the colchicine formulation to be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • the composition can be administered as a single dose, multiple doses or over an established period of time in an infusion. Dosage regimens also can be adjusted to provide the optimum desired response (e.g ., a therapeutic or prophylactic response).
  • the composition of the present invention can be parenterally administered.
  • Preparations for parenteral administration include sterile aqueous or non- aqueous solutions, suspensions, and emulsions.
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include, e.g., water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • pharmaceutically acceptable carriers include, but are not limited to, 0.01-0.1 M and preferably 0.05 M phosphate buffer or 0.8% saline.
  • Intravenous vehicles include sodium phosphate solutions, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, antioxidants, chelating agents, and inert gases and the like.
  • Parenteral formulations can be a single bolus dose, an infusion or a loading bolus dose followed with a maintenance dose. These compositions can be administered at specific fixed or variable intervals, e.g., once a day, or on an "as needed" basis.
  • the composition according to the present invention may be administered in a dose range varying depending on the patient's body weight, age, gender, health condition, diet, administration time, administration method, excretion rate and disease severity.
  • the compounds of the present invention as compounds per se in their use as pharmacophores or as pharmaceutical compositions can be administered to the patient and/or subject at a suitable dose.
  • the dosage regiment will be determined by the attending physician and clinical factors. As is well known in the medical arts, dosages for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently.
  • the regimen as a regular administration of the pharmaceutical composition comprising the herein defined should be, e.g., in a range as described below. Progress can be monitored by periodic assessment.
  • composition according to the present invention can be administered with a single dose or with 2, 3, 4, 5, 6, 7, 8, 9, or 10 doses, if desired.
  • the composition can be administered 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 times per day.
  • colchicine according to the present invention is administered once per day. More preferably, colchicine according to the present invention is administered once per day as a single dose.
  • the composition according to the present invention can be administered regularly for long periods of time.
  • the composition can be administered regularly for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more years.
  • the composition can be administered regularly for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more months.
  • the composition can be administered regularly for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more weeks.
  • the term “regularly” refers to administration of the composition at regular times or intervals over a period of time. For instance, the composition may be administered to a patient once daily for three (3) years. In other embodiments, the composition may be administered to a patient once every other day for five (5) years.
  • the frequency of administration may vary based on a number of factors, including, but not limited to, the severity of disease, the overall health of the patient, any additional medications the patient is taking, and whether the treatment is prophylactic or not. It should also be appreciated that the frequency of administration may be adjusted at any point.
  • the amount/concentration/dose of the composition according to the present invention can be between O.lmg and 5.0mg, O.lmg and 2.0mg, O.lmg to 1.5mg, O.lmg to l.Omg, O.lmg to 0.75mg, O.lmg to 0.5mg, 0.25mg to 5.0mg, 0.25mg to 2.0mg, 0.25mg to 1.5mg, 0.25mg to l.Omg, 0.25mg to 0.75mg or 0.25mg to 0.5mg.
  • the composition according to the present invention is administered at a daily dose of colchicine of between about O.lmg and about 0.75mg or between about O.lmg and about 0.5mg. In another embodiment, the composition according to the present invention is administered at a daily dose of colchicine of between about 0.25mg to about 0.75mg or between about 0.25mg to about 0.5mg. In an embodiment, the composition according to the present invention is administered at a daily dose of about 0.5mg colchicine.
  • the amount/concentration of colchicine as used herein can be administered at the first day of administration in a higher dose (concentration/amount) compared to the administration of colchicine at the following days(s) of administration (maintenance administration/maintenance dose of administration).
  • a higher dose concentration/amount
  • such decreased dose can be started after 2, 3, 4, 5, 6, 7, 8, 9 or 10 days of initial administration of the higher dose.
  • the higher dose/amount/concentration of colchicine e.g.
  • the composition of the invention is administered with a dose of colchicine of between about 1.0 mg to about 2.0 mg at the first day (preferably as a single dose) of administration and the maintenance dose of colchicine at the following day(s) of administration is between about 0.5 mg to about 1.0 mg.
  • the colchicine formulation of the present invention can be administered to a human or other animal in accordance with the aforementioned methods of treatment in an amount sufficient to produce a therapeutic effect.
  • the colchicine formulation can be administered to such human or other animal in a conventional dosage form prepared by combining the colchicine formulation of the invention with a conventional pharmaceutically acceptable carrier or diluent according to known techniques. It will be recognized by one of skill in the art that the form and character of the pharmaceutically acceptable carrier or diluent is dictated by the amount of active ingredient with which it is to be combined, the route of administration and other well- known variables.
  • terapéuticaally effective dose or amount or “effective amount” is intended an amount of the colchicine formulation that when administered brings about a positive therapeutic response with respect to treatment of a patient with a disease to be treated, e.g., an improvement in the disease can be evidenced by, for example, a delayed onset of clinical symptoms of the disease or condition, a reduction in severity of some or all clinical symptoms of the disease or condition, a slower progression of the disease or condition, an improvement in the overall health or well-being of the subject, or by other parameters well known in the art that are specific to the particular disease.
  • compositions of the present invention for the inhibition, delay, or reduction of metastases, vary depending upon many different factors, including means of administration, target site, physiological state of the patient, whether the patient is human or an animal, other medications administered, and whether treatment is prophylactic or therapeutic.
  • the patient is a human, but non human mammals including transgenic mammals can also be treated.
  • Treatment dosages may be titrated using routine methods known to those of skill in the art to optimize safety and efficacy.
  • the amount of colchicine administered as a single agent or in combination with at least one other immune modulating therapy is readily determined by one of ordinary skill in the art without undue experimentation given the disclosure of the present invention.
  • Factors influencing the mode of administration and the respective amount of colchicine to be administered as a single agent or in combination with at least one other immune modulating therapy include, but are not limited to, the severity of the disease, the history of the disease, the potential for metastases, and the age, height, weight, health, and physical condition of the individual undergoing therapy.
  • the amount of colchicine as a single agent or in combination with at least one other immune modulating therapy to be administered will be dependent upon the mode of administration and whether the subject will undergo a single dose or multiple doses of this agent.
  • the invention also provides for the use of the colchicine formulation in the manufacture of a medicament for treating a subject with a cancer, wherein the medicament is used in a subject that has been pretreated or is concurrently being treated with at least one other therapy.
  • pretreated or “pretreatment” is intended the subject has received one or more other therapies prior to receiving the medicament comprising the colchicine formulation.
  • Pretreated or “pretreatment” includes subjects that have been treated with at least one other therapy within 2 years, within 18 months, within 1 year, within 6 months, within 2 months, within 6 weeks, within 1 month, within 4 weeks, within 3 weeks, within 2 weeks, within 1 week, within 6 days, within 5 days, within 4 days, within 3 days, within 2 days, or even within 1 day prior to initiation of treatment with the medicament comprising the colchicine formulation.
  • concurrent or “concomitant” is intended the subject is receiving one or more other therapies while at the same time receiving the medicament comprising the colchicine formulation. It is not necessary that the subject was a responder to pretreatment with the prior therapy or therapies or a responder to the concurrent therapy or therapies.
  • the subject that receives the medicament comprising the colchicine formulation could have responded, or could have failed to respond, to pretreatment with the prior therapy, or to one or more of the prior therapies where pretreatment comprised multiple therapies.
  • other cancer therapies for which a subject can have received pretreatment prior to receiving the medicament comprising the colchicine as a single agent or in combination with at least one other immune modulating therapy include, but are not limited to, surgery; radiation therapy; chemotherapy, optionally in combination with autologous bone marrow transplant, where suitable chemotherapeutic agents include, but are not limited to, those listed herein above; other anti-cancer monoclonal antibody therapy; small molecule-based cancer therapy, including, but not limited to, the small molecules listed herein above; vaccine/immunotherapy-based cancer therapies; steroid therapy; other cancer therapy; or any combination thereof.
  • Example 1 Colchicine Sustained Release Tablet [00170] This example illustrates a colchicine sustained release tablet. The tablet uses the ingredients and concentrations shown in Table 1 below.
  • concentrations may be altered to change certain properties of the formulations, for instance, the dissolution profile.
  • Table 2 shows the ranges for each ingredient.
  • Example 2 Method of Making a Colchicine Sustained Release Tablet
  • Granulation The granulation was performed in a Kenwood mixer. The colchicine and hypromellose 6Mpa*s were first weighed and separately dissolved into purified water. This solution of hypromellose 6Mpa*s was filled in the mixer containing the lactose within 1.5 min followed by a 3 minute mixing time. Subsequently, the dissolved colchicine was sprayed with a filing agent (e.g., lactose monohydrate) under continuous mixing over a period of 15 minutes. These steps were performed at room temperature. The wet granulate was then passed through a 1.0 mm hand sieve.
  • a filing agent e.g., lactose monohydrate
  • Blending Following the granulation process, the granulate is compounded with a filling agent (e.g., lactose monohydrate), a retarding agent (e.g., Retalac), and other excipients (e.g., flow enhancer, glidants and/or lubricants) to support the tablet compression process.
  • a filling agent e.g., lactose monohydrate
  • a retarding agent e.g., Retalac
  • other excipients e.g., flow enhancer, glidants and/or lubricants
  • these ingredients were placed manually through a 0.8 mm sieve and mixed with the granulate in a cube mixer (Erweka) for 10 minutes.
  • the glidant used may be Talc.
  • the lubricants used may be Stearic acid.
  • the granulate will then be blended using a suitable mixer.
  • Tableting was performed on a Korsch (EK 0) tablet press with a round tabletting tool, biconvex, 6 mm in diameter. Average tablet hardness was approximately 100N +/- 15 N. Tablets measured about lOOmg in mass, friability was not measurable. Breaking strength and hardness were measured with a Erweka Multickeck. Friability was measured with a Erweka Friabilator and a Mettler analytical balance. The dimensions were measured using a Mitutoyo caliper.
  • the dissolution of the sustained release formulation of colchicine was measured at various time points.
  • the compositions were dissolved in 500ml of water at 37°C and stirred continuously over a period of 6 hours. Samples were drawn at several time points to study the kinetics of the dissolution process of the drug substance within the hydrophilic matrix system. Colchicine content in the samples was analyzed using HPFC analysis.
  • the dissolution profiles of the various batches are provided in Tables 4-6 and FIGS. 1-4.
  • the dissolution profile for Batches 1-3 is summarized in Table 4 and FIG. 1.
  • the profile for Batch 1 shows approximately a 92% release within about 30 minutes, followed by a constant release. Complete dissolution occurred after 2 hours.
  • Batch 2 shows approximately a 83% release within about 30 minutes, followed by a constant release.
  • Batch 3 shows approximately a 74% release within about 30 minutes, followed by a constant release. Complete dissolution occurred after 2 hours.
  • the dissolution profile for Batch 4 is summarized in Table 5 and FIG. 2.
  • Batch 4A shows approximately a 30% release within about 30 minutes, followed by a constant release. Complete dissolution occurred after 6 hours.
  • Batch 4B shows approximately a 23% release within about 30 minutes, followed by a constant release. Complete dissolution occurred within 6 hours.
  • a difference in tablet hardness results in a difference in release rates.
  • FIG. 3 shows the dissolution profiles for Batches 1-4.
  • the dissolution profile for Batch 6 is summarized in Table 7 and FIG. 5A.
  • Batch 6 represents a sustained release version of the composition.
  • the profile for Batch 6 in FIG. 5A shows about a 45% release within 30 minutes, about a 65% release in 60 minutes, and about a 80% release in 90 minutes. Complete dissolution occurs within about 2 hours.
  • Batch 4 was further modified by altering the concentration of retarding agent (i.e. Retalac) in the composition (Retalac 0%, 23.3%, 26,6% or 30%).
  • concentration of retarding agent i.e. Retalac
  • FIG. 6 the concentration of retarding agent (i.e. Retalac) in the composition (Retalac 0%, 23.3%, 26,6% or 30%).
  • the dissolution profiles of the four compositions are shown in FIG. 6.
  • the composition with 23.3% Retalac shows about a 65% release within 30 min, about a 80% release in 60 min, and about a 90% release in 90 min. Complete dissolution occurs within about 120 min.
  • the composition with 26.6% Retalac shows about a 55% release within 30 min, about a 75% release in 60 min, and about a 85% release in 90 min.
  • Complete dissolution occurs within about 180 min.
  • FIG. 6 the concentration of retarding agent (i.e. Retalac) in the composition (Retalac
  • the composition with 30% Retalac shows about a 40% release within 30 min, about a 55% release in 60 min, and about a 70% release in 90 min. Complete dissolution occurs in more than about 180 min. As further shown in FIG. 6, the composition with 0% Retalac (i.e., immediate release composition) shows complete dissolution within about 15 min.
  • the release profile of the sustained release composition can be changed to a specific or desired target release by adjusting the amount of retarding agent (i.e., Retalac) as well as tablet hardness.
  • the release depends upon a variety of factors, including erosion of the outer layer of colchicine (i.e., the immediate release portion) as well as diffusion of the inner layer of colchicine (i.e., the sustained release portion). Since the percentage of colchicine is low in the sustained release formulation and the tablets are small, this balance between erosion and diffusion is very sensitive and has to be fine-tuned to reach a very specific dissolution profile.
  • FIG. 7A, FIG. 7B, FIG. 7C, FIG. D, and FIG. 8 show plasma colchicine levels (ng/mL) as a function of time (hrs) for colchicine formulations according to FIG. 6
  • the therapeutic effect of the sustained release formulation containing of colchicine is evaluated in a clinical study that is a multidose, randomized, cross-over study, which will evaluate bioavailability of about 3 different 0.50 mg sustained release formulations (e.g. Test Products 1,2,3 and FIG. 6) of colchicine to a 0.5 mg immediate release formulation (control product), administered to healthy volunteers.
  • Test products 1, 2, and 3 used varying levels of retarding agent (i.e., Retalac), 23.3%, 26.6% or 30%, respectively, thereby having different sustained release profiles.
  • the primary aim of the study is to assess pharmacokinetics (PK) of test and control drug in blood as well as in neutrophils or leucocytes.
  • the primary objective of this study is to compare the pharmacokinetics of the test product vs. control product of colchicine in healthy human volunteers.
  • levels of colchicine in blood herein referred as to blood PK
  • neutrophils or leucocytes herein referred as to neutrophil PK
  • Cmax peak levels
  • AUC area under the curve
  • Colchicine concentrations in neutrophils are measured as it is generally recognized that neutrophils which reconstitute 60-70% of leukocytes, play a central role in inflammatory responses in general and are thought to be major players in several diseases where colchicine is used as treatment. Therefore for the purpose of this experiment, either leucocytes or neutrophils may be analyzed. Hence, the term neutrophils as used herein also refers to, if used, leucocytes. As colchicine is known to preferentially accumulate in neutrophils and inhibit many of their pro-inflammatory functions, they are thought to be a major target of colchicine therapy.
  • Study duration The study drug is administered at a single dose/day for about 8- 14 days consecutive days.
  • the study consists of 24h blood PK (high frequency data collection).
  • Neutrophil PK is analysed in intervals that allow for conclusive determination of colchicine levels in the latter over the duration of the experiment.
  • Treatment regimen Below is an example of what the treatment regime may look like.
  • Treatment schema The drug is given once a day in the morning.
  • PK analysis includes colchicine blood concentrations. Adequate techniques for the isolation of blood and quantification of colchicine are applied.
  • neutrophils will be isolated or purified from raw blood from several time points. Neutrophils will be collected at the beginning the study as well as at least once at the end of the study. PK analysis includes colchicine concentrations in isolated neutrophils.
  • Results The results are shown in FIGS. 7-9.
  • FIGS. 7A-7D show plasma colchicine levels (ng/mL) as a function of time (hrs) in the immediate release formulation (FIG. 7A) as compared to the sustained release formulations (FIGS. 7B, 7C and 7D).
  • the sustained release formulation of FIG. 7A has a Cmax of 1.77 +/- 0.8 ng/mL, an AUC (o-t) of 8.08 +/- 2.9 ng/mL and a Tmax of 0.83 +/- 0.2 hr.
  • the sustained release formulation of FIG. 7B has a Cmax of 1.56 +/- 0.4 ng/mL, an AUC (o-t) of 7.94 +/- 0.9 ng/mL and a Tmax of 1.47 +/- 0.2 hr.
  • the sustained release formulation of FIG. 7C has a Cmax of 1.56 +/- 0.4 ng/mL, an AUC (o-t) of 7.94 +/- 0.9 ng/mL and a Tmax of 1.47 +/- 0.2 hr.
  • the sustained release formulation of FIG. 7D has a Cmax of 0.91 +/- 0.3 ng/mL, an AUC (o-t) of 5.04 +/- 2.3 ng/mL and a Tmax of 1.89 +/- 0.4 hr.
  • FIG. 8 shows the overlapping profiles of the immediate release formulation and the three sustained release profiles. As can be seen, there is less inter-subject variation in plasma levels, lower Cmax and similar AUC for the sustained release formulation of FIG. 7B when compared to the immediate release formulation. Since the colchicine concentration on plasma increases and decreases more steadily and in a more controlled way, and the variability of colchicine levels in plasma from patient to patient is less, the safety and efficacy profile is more predicable.
  • FIG. 9 shows the levels of colchicine in neutrophils on Day 1 and Day 10 in the immediate release formulation and the three sustained release formulations. As can be seen, the levels of colchicine increase from Day 1 to Day 10, suggesting that colchicine accumulates in neutrophils over time. The results further show that the levels in neutrophils are similar after 10 days regardless of the formulation, suggesting that the therapeutic effect will be equal for the tested sustained release and immediate release formulations (as the site of action of colchicine is neutrophils).
  • Example 5 Therapeutic effects of colchicine in patients with cancer [00202] To assess the therapeutic effects of a sustained release formulation in patients with or in patients with an increased risk of cancer, a prospective randomized observer blinded end-point trial will be conducted to determine whether adding 0.5 mg/day of colchicine to standard anti-cancer therapies inhibits, delays, or reduces tumor growth or metastases in a cancer patient.
  • a sufficient amount of patients with cancer or in risk of cancer are to be randomized to long-term low dose sustained release colchicine or to standard therapy alone. Cancer incidence, morbidity and mortality will be analyzed in regular intervals. Patients in the colchicine arm are expected to have a significantly lower cancer incidence and mortality than patients not treated with colchicine.
  • Example 6 Anti-tumor effects of colchicine in animals [00204] A sufficient amount of specimen of a suitable animal species will be included in this study utilizing commonly used tumor models. A therapeutically effective amount of colchicine is administered to the test animals. Placebo will be administered to a control group. Tumor growth will be induced in all animals. Animals treated with colchicine are expected to have significantly less tumor growth, or tumor growth is prevented as compared to the control group.
  • Example 7 Anti- tumor effects of colchicine in vitro
  • a suitable in vitro model for studying the anti-tumor effects of drugs is utilized for this test.
  • Such a test may include commonly used tumor cell lines with or without combination with cultivated PBMCs.
  • Colchicine is expected to inhibit or reduce growth of tumor cell lines upon incubation with therapeutically effective amount of colchicine and/or cultivation with PBMCs pretreated with a therapeutically effective amount of colchicine.
  • Example 8 Effects of colchicine on lung cancer
  • the investigators propose to conduct a randomized, double-blind, placebo- controlled, multi-center trial of oral daily colchicine ⁇ dose discussion pending, may need 2 doses probably 0.5 and 1.0 mg daily >> to formally test whether chronic interleukin- 1b inhibition will slow the invasion, progression, and metastases associated with early stage lung cancers and thus significantly improve cancer survival.
  • the trial will be conducted using multiple internet based and social media techniques previously employed to great success without sacrificing quality of adherence, compliance, endpoint adjudication, or rates of long-term follow-up.
  • Patients will be eligible for the proposed trial if they have been recently diagnosed with non-metastatic stage IA2, IA3, and IB lung cancer and have just completed initial resection. Such patients are typically followed by the thoracic surgery community in a setting where no post-resection therapy has proven effective and where routine care includes close observation and surveillance for recurrent disease through serial CT scans over a period of 3 to 5 years. As such, the trial has been designed to minimally alter usual care practice and should thus be easy to conduct in the many dedicated thoracic surgical centers who have made a fundamental commitment to this effort.
  • the proposed trial primary endpoint will be recurrence of cancer or cancer-related death at 5 years (conservatively estimated to be 35-40 percent) while the proposed trial secondary endpoint will consist of cancer-related death alone (conservatively estimated to be 30 to 35 percent).
  • the 300mg dose of canakinumab reduced incident lung cancer by 67 % and fatal lung cancer by 77% (at a median time period of only 3 years).
  • the proposed trial assumes a treatment effect of only half that size (35% relative risk reduction); as such, power exceeds 93% for the primary endpoint and 86% for the secondary endpoint with total sample sizes ranging from 1000 to 1200 participants.
  • randomization will be stratified on age and smoking status, with pre-specified subgroup analyses conducted on the basis of tumor genetic profiles and other typical clinical parameters.

Abstract

Use of colchicine to inhibit, delay, or reduce the occurrence of lung cancer in a subject with cancer, comprising administering to the subject an effective amount of colchicine is provided.

Description

USE OF COLCHICINE IN THE TREATMENT AND PREVENTION OF LUNG
CANCER
BACKGROUND OF THE INVENTION
[0001] Colchicine, chemical name (-)-N-[(7S, 12aS)-l, 2,3, 10-tetramethoxy-9-oxo-5, 6,7,9- tetrahydrobenzo[a]heptalen-7-yl]-acetamide, is an alkaloid found in extracts of Colchicum autumnale, Gloriosa superba, and other plants. It is a microtubule-disrupting agent used in the treatment of conditions that may be treated, relieved or prevented with anti inflammatory treatment.
[0002] Colchicine is well recognized as a valid therapy in acute flares of gouty arthritis, familial Mediterranean fever (FMF), and Behcet's disease. It has also been used to treat many inflammatory disorders prone to fibrosis. In the recent past, colchicine has been proposed to be effective in therapy in cardiovascular diseases. Thus, colchicine has a proven track record in treatment of various diseases and conditions associated with inflammation.
[0003] In particular, colchicine has been proposed as a first treatment option for recurrent pericarditis (class I indication) and optional for acute pericarditis (class Ila indication) in the 2004 European guidelines on the management of pericardial diseases (Maisch et al. , Guidelines on the Diagnosis and Management of Pericardial Diseases, Eur Heart J., 2004, 25, 916-928).
[0004] Imazio et al. ( Circulation , 2005, 112 (13), 2012-2016) showed that colchicine was effective for the treatment and the prevention of recurrent pericarditis in a prospective, randomized, open-label designed study of 120 patients with a first episode of acute pericarditis (idiopathic, viral, postpericardiotomy syndromes, and connective tissue diseases), who were randomly assigned to conventional treatment with aspirin or conventional treatment plus colchicine (1.0 to 2.0 mg for the first day and then 0.5 to 1.0 mg/day for 3 months). The primary end point was recurrence rate, which was significantly reduced from 32.3% down to 10.7% at 18 months in the colchicine group (p=0.004).
[0005] Further, the same group showed that colchicine could be efficient after conventional treatment failure to manage acute pericarditis (Imazio at al., Arch InternMed, 2005, 165 (17), 1987-91). In a prospective, randomized, open-label design, 84 consecutive patients with a first episode of recurrent pericarditis were randomly assigned to receive conventional treatment with aspirin alone or conventional treatment plus colchicine (1.0- 2.0 mg the first day and then 0.5- 1.0 mg/d for 6 months). The primary end point was the recurrence rate, which was significantly decreased in the colchicine group (actuarial rates at 18 months were 24.0% vs 50.6% with conventional treatment).
[0006] It has also been shown that colchicine is effective for secondary prevention of recurrent pericarditis Imazio et al., Ann. Intern. Med., 2011, 155 (7), 409-14). Colchicine has also been proposed to reduce postpericardiotomy reactions revealed as pericarditis (Imazio et al., Am. Heart J., 2011, 162 (3), 527-532; Meurin and Tabet, Arch. Cardiovasc. Dis., 2011, 104 (8-9), 425-427).
[0007] Colchicine for the treatment of post-pericardiotomy syndrome (PPS) was tested for the first time in a preliminary prospective, open-label, randomized trial of colchicine (1.5 mg/day) compared with placebo beginning on the third post-operative day in 163 patients who underwent cardiac surgery (Finkelstein et al., Herz, 200227, 791-194).
[0008] The effectiveness of colchicine for the prevention of PPS has also been shown in a multicentre, double-blind, randomized trial, in which 360 patients (mean age 65.7+12.3 years, 66% males), 180 in each treatment arm, were randomized to receive placebo or colchicine (1.0 mg twice daily for the first day followed by a maintenance dose of 0.5 mg twice daily for 1 month in patients >70 kg, and halved doses for patients ,70 kg or intolerant to the highest dose) on the third post-operative day (Imazio et al., European Heart Journal, 2010, 31, 2749-2754).
[0009] For the treatment of gout, the recommended dose of colchicine (COLCRYS®) is 1.8 mg/day in one or multiple doses in one hour. For adults with gout, treatment is initiated with a dose of 1.2 mg at the first sign of symptoms followed by 0.6 mg one hour later. (Physician's Desk Reference, 68th ed., (2014)).
[0010] In fact, colchicine is associated with many adverse side effects. COLCRYS®, for instance, is an immediate release formulation of colchicine. Adverse effects associated with the administration of COLCRYS® include, but are not limited to, nausea, vomiting, abdominal pain, diarrhea, hair loss, weakness, nerve irritation, severe anemia, low white blood counts, and low platelets (Physician's Desk Reference, 68th ed., (2014)).
[0011] Evidence, including the outcomes of the CANTOS clinical trial, indicates that inflammation in the tumor microenvironment is mediated in part through interleukin- 1b (IL-Ib), which is believed to play a role in growth, invasion, biologic transformation, and metastasis of early stage lung cancer. In the CANTOS trial, a randomized, double-blind, placebo-controlled trial of atherosclerosis patients who were treated with the interleukin- 1 inhibitor canakinumab given subcutaneously once every 3 months, a dose-dependent reduction in new lung cancers was observed, culminating in a statistically significant reduction in incident lung cancer (HR 0.33, 95% Cl 0.18 - 0.59, P < 0.0001) and a statistically significant reduction in lung cancer mortality (HR 0.23, 95% Cl 0.10 - 0.54, P = 0.0002) over an average follow-up period of 3.7 years. Within CANTOS, the magnitude of inflammation inhibition achieved as assessed by on-treatment levels of the inflammatory biomarkers hsCRP and interleukin-6 have consistently proven to correlate with clinical benefit (see, Ridker et al., Lancet , 391:319-328, 2018, and Ridker et al., Eur. Heart /., 39:3499-3507, 2018), data internally consistent with the dose-response effects seen for lung cancer.
[0012] However, since canakinumab is an expensive monoclonal antibody, there continues to be a need for proof-of-principal with an inexpensive and orally available pharmaceutical agent capable of address lung cancer and lung tumorigenesis. Thus, the need exists for an inexpensive oral agent with similar anti-inflammatory properties. As explained herein, colchicine is a microtubule inhibitor that has anti-inflammatory activity in part through interference with the NLRP3 inflammasome structure that leads to a similar reduction in iterleukukin-ΐb levels.
[0013] Further, while significant therapeutics are available for subjects experiencing or diagnosed with lung cancer, or suffering from advanced forms of such cancer, subjects diagnosed with cancer at an early stage have few, if any, options for treatment. Due to the increasing use of CT scanning in diagnostic radiology, the detection of early stage lung cancer is a common clinical problem that poses substantial burden for patients and society. For patients in which detected tumors are greater than 4 cm or where either regional lymph node metastases or distant metastases are present, treatment protocols exist that have improved clinical outcomes, most notably with the use of surgery and adjunctive PD-1 inhibition. Examples of ongoing work in this arena includes the ALCHEMIST trials within National Cancer Trials Network which are limited to tumors >4cm, most with known metastases. By contrast, for patients in which detected tumors are non-metastatic and less than or equal to 4 cm (1A2, IA3, and IB), no proven therapeutic options beyond primary resection exist. Yet, in current registries, 5 year cancer recurrence rates in such patients exceed 35 percent. A safe and effective medical therapy in this setting would thus represent a major advance for this increasingly common clinical problem. Given the number of patients diagnosed annually in the United States with early stage lung cancer where no proven medical therapy exists and mortality rates at five years approach 30 to 40 percent, this invention addresses a major unmet clinical need using a novel biologic approach for early lung cancer treatment for instance, for patients with newly diagnosed non-metastatic lung cancer.
[0014] The instant invention addresses these and other needs by providing a modified formulation of low dose colchicine characterized by a sustained release of an active ingredient that is well tolerated with few, if any, of the commonly experienced adverse side effects associated with immediate release colchicine and colchicine at higher dosages. Thus, provided is an effective, once-daily dosage form of colchicine or salts thereof, that may improve patient.
[0015] This invention addresses the need for safe and effective cancer treatments, such as lung cancer, that serve either as a single agent that treats, prevents, inhibits, reduces, suppresses, prevents, slows or delays the progression of, shrinks, or directly attacks cancer cells or that can act in combination with other immune modulating therapies to enhance their therapeutic benefit. Colchicine is inexpensive, tolerated at low doses, has a proven safety track record, and fits the needs for this group of people. Colchicine has a long history as a safe anti-inflammatory agent used in several clinical settings including arthritis, gout and pericarditis, diseases known to be driven in part by NLRP3 mediated inflammation. As an anti-mitotic microtubule inhibitor, colchicine has been shown to disrupt inflammasome construction resulting in lower systemic levels of both IL-Ib and CRP (Martinon, Nature, 2006; Martinez, Atherosclerosis, 2018). Small clinical studies have suggested benefits of colchicine in the setting of gastric or hypopharyngeal tumors, and gout patients treated with colchicine have been reported to have a reduced cancer burden (Zhang, BioSci. Rep., 2019; Kim, Anticancer Res., 2017; Kuo, Medicine, 2015).
BRIEF SUMMARY OF THE INVENTION
[0016] In accordance with an example embodiment of the present disclosure, there is provided a method for inhibiting, delaying, or reducing the occurrence of lung cancer in a subject with cancer, comprising administering to the subject an effective amount of colchicine.
[0017] According to aspects of the present disclosure, the colchicine may induce anti-tumor responses, inhibit IL-Ib mediated signaling, inhibit inflammation activation, inhibit release of TNF-a and IL-10. [0018] According to aspects of the present disclosure, the colchicine is included in an amount between about 0.25mg and about l.Omg. The colchicine could be included in an amount between about 0.5mg.
[0019] In accordance with an example embodiment of the present disclosure, there is provided a method for treating lung cancer in a subject, comprising administering to the subject an effective amount of colchicine.
[0020] According to aspects of the present disclosure, the colchicine may induce anti-tumor responses, inhibit IL-Ib mediated signaling, inhibit inflammation activation, inhibit release of TNF-a and IL-10.
[0021] According to aspects of the present disclosure, the colchicine is included in an amount between about 0.25mg and about l.Omg. The colchicine could be included in an amount between about 0.5mg.
[0022] In accordance with an example embodiment of the present disclosure, there is provided a method for treating lung cancer in a subject, comprising administering to the subject an effective amount of colchicine and an effective amount of at least one other immune modulating therapy.
[0023] According to aspects of the present disclosure, the immune modulating therapy is selected from the group consisting of a cancer vaccine, an immuno stimulatory agent, adoptive T cell or antibody therapy, immune checkpoint blockade and a combination thereof. In some embodiments, the immune modulating agent is selected from the group consisting of interleukins, cytokines, chemokines, antagonists of immune checkpoint blockades and a combination thereof. In some embodiments, the immune modulating therapy may be a cancer therapy. In some embodiments, the cancer therapy is selected from the group consisting of surgery or surgical procedures, radiation therapy, chemotherapy or a combination thereof.
[0024] According to aspects of the present disclosure, colchicine and the immune modulating agent or immune modulating therapy are administered separately or concurrently.
[0025] According to aspects of the present disclosure, the colchicine is included in an amount between about 0.25mg and about l.Omg. The colchicine could be included in an amount between about 0.5mg. BRIEF DESCRIPTION OF THE DRAWINGS/FIGURES
[0026] FIG. 1 shows the dissolution profiles for colchicine sustained-release formulations containing 10%, 15% and 20%, respectively, of an exemplary retarding agent.
[0027] FIG. 2 shows the dissolution profiles for colchicine sustained-release formulation containing 30% of an exemplary retarding agent and tablet hardnesses of 50N and 130N, respectively.
[0028] FIG. 3 shows the dissolution profiles for colchicine sustained-release formulations according to FIGS. 1 and 2.
[0029] FIG. 4 shows the dissolution profile for a colchicine sustained-release formulation containing 0% of an exemplary retarding agent.
[0030] FIG. 5 shows the dissolution profile for a colchicine sustained-release formulation containing 25% of an exemplary retarding agent.
[0031] FIG. 6 shows the dissolution profile for colchicine formulation containing 0%, 23.3%, 26.6% and 30% of an exemplary retarding agent.
[0032] FIGS. 7 A, 7B, 7C and 7D show plasma colchicine levels (ng/mL) as a function of time (hrs) for colchicine formulations according to FIG. 6.
[0033] FIG. 8 shows plasma colchicine levels (ng/mL) as a function of time (hrs) for colchicine formulations according to FIG. 6.
[0034] FIG. 9 shows the levels of colchicine in neutrophils on Day 1 and Day 10 for colchicine formulations according to FIG. 6.
DETAILED DESCRIPTION OF THE INVENTION
I. Definitions
[0035] For the purposes of this invention, the term “colchicine” includes colchicine or any pharmaceutically acceptable salts thereof.
[0036] “Pharmaceutically acceptable” means that which is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary use as well as human pharmaceutical use.
[0037] “Pharmaceutically acceptable salts” includes derivatives of colchicine, wherein the colchicine is modified by making acid or base addition salts thereof, and further refers to pharmaceutically acceptable solvates, including hydrates, and co-crystals of such compounds and such salts. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid addition salts of basic residues such as amines; alkali or organic addition salts of acidic residues; and the like, and combinations comprising one or more of the foregoing salts. The pharmaceutically acceptable salts include non-toxic salts and the quaternary ammonium salts of the colchicine. For example, non-toxic acid salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; other acceptable inorganic salts include metal salts such as sodium salt, potassium salt, cesium salt, and the like; and alkaline earth metal salts, such as calcium salt, magnesium salt, and the like, and combinations comprising one or more of the foregoing salts. Pharmaceutically acceptable organic salts includes salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, mesylic, esylic, besylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, HOOC — (CH2)n — COOH where n is 0-4, and the like; organic amine salts such as triethylamine salt, pyridine salt, picoline salt, ethanolamine salt, triethanolamine salt, dicyclohexylamine salt, N,N'- dibenzylethylenediamine salt, and the like; and amino acid salts such as arginate, asparaginate, glutamate, and the like; and combinations comprising one or more of the foregoing salts; organic amine salts such as triethylamine salt, pyridine salt, picoline salt, ethanolamine salt, triethanolamine salt, dicyclohexylamine salt, N,N' dibenzylethylenediamine salt, and the like; and amino acid salts such as arginate, asparaginate, glutamate, and the like; and combinations comprising one or more of the foregoing salts. All forms of such derivatives of colchicine are contemplated herein, including all crystalline, amorphous, and polymorph forms. Specific colchicine salts include colchicine hydrochloride, colchicine dihydrochloride, and co-crystals, hydrates or solvates thereof.
[0038] “Pharmacokinetic parameters” describe the in vivo characteristics of an active agent (or a metabolite or a surrogate marker for the active agent) over time, such as plasma concentration (C), Cmax, Cn, C24, Tmax, and AUC. “Cmax” is the measured plasma concentration of the active agent at the point of maximum, or peak, concentration. “Cmin” is the measured plasma concentration of the active agent at the point of minimum concentration. “Cn” is the measured plasma concentration of the active agent at about n hours after administration. “C24” is the measured plasma concentration of the active agent at about 24 hours after administration. The term “Tmax” refers to the time at which the measured plasma concentration of the active agent is the highest after administration of the active agent. “AUC” is the area under the curve of a graph of the measured plasma concentration of an active agent vs. time, measured from one time point to another time point. For example AUCO-t is the area under the curve of plasma concentration versus time from time 0 to time t, where t can be the last time point with measurable plasma concentration for an individual formulation. The AUC0-¥ or AUCO-INF is the calculated area under the curve of plasma concentration versus time from time 0 to time infinity. In steady-state studies, AUCO-t is the area under the curve of plasma concentration over the dosing interval (i.e., from time 0 to time t (tau), where tau is the length of the dosing interval. Other pharmacokinetic parameters are the parameter Ke or Kel, the terminal elimination rate constant calculated from a semi-log plot of the plasma concentration versus time curve; tl/2 the terminal elimination half-life, calculated as 0.693/Kel; CL/F denotes the apparent total body clearance after administration, calculated as Total Dose/Total AUC¥; and Varea/F denotes the apparent total volume of distribution after administration, calculated as Total Dose/(Total AUC¥xKel).
[0039] “Efficacy” means the ability of an active agent administered to a patient to produce a therapeutic effect in the patient.
[0040] “Bioavailability” means the extent or rate at which an active agent is absorbed into a living system or is made available at the site of physiological activity. For active agents that are intended to be absorbed into the bloodstream, bioavailability data for a given formulation may provide an estimate of the relative fraction of the administered dose that is absorbed into the systemic circulation. “Bioavailability” can be characterized by one or more pharmacokinetic parameters.
[0041] A “dosage form” means a unit of administration of an active agent. Examples of dosage forms include tablets, capsules, injections, suspensions, liquids, emulsions, creams, ointments, suppositories, inhalable forms, transdermal forms, and the like.
[0042] An “immediate release formulation” refers to a formulation that releases greater than or equal to about 80% of the pharmaceutical agent in less than or equal to about 30 min. [0043] For the purposes of this application, an enhancing agent (“enhancer”) is defined as any non- pharmaceutically active ingredient that improves the therapeutic potential of a formulation.
[0044] “Sustained release” is defined herein as release of a pharmaceutical agent in a continuous manner over a prolonged period of time.
[0045] By “prolonged period of time” it is meant a continuous period of time of greater than about 1 hour, greater than about 4 hours, greater than about 8 hours, greater than about 12 hours, greater than about 16 hours, or up to more than about 24 hours.
[0046] As used herein, unless otherwise noted, “rate of release” or “release rate” or “dissolution rate” of a drug refers to the quantity of drug released from a dosage form per unit time, e.g., milligrams of drug released per hour (mg/hr) or a percentage of a total drug dose released per hour. Drug release rates for dosage forms are typically measured as an in vitro rate of drug release, i.e., a quantity of drug released from the dosage form per unit time measured under appropriate conditions and in a suitable fluid. The release rates referred to herein are determined by placing a dosage form to be tested in a medium in an appropriate dissolution bath. Aliquots of the medium, collected at pre-set intervals, are then injected into a chromatographic system fitted with an appropriate detector to quantify the amounts of drug released during the testing intervals.
[0047] As used herein, “side effect” refers to a secondary and usually adverse effect of a drug.
[0048] As used herein, the terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals in which a population of cells are characterized by unregulated cell growth. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia. More particular examples of such cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, gastric, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, brain cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, esophageal cancer, salivary gland carcinoma, sarcoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancers.
[0049] “Tumor” and “neoplasm” as used herein refer to any mass of tissue that result from excessive cell growth or proliferation, either benign (noncancerous) or malignant (cancerous) including pre-cancerous lesions. [0050] The terms “metastasis,” "metastases," "metastatic," and other grammatical equivalents as used herein refer to cancer cells which spread or transfer from the site of origin (e.g., a primary tumor) to other regions of the body with the development of a similar cancerous lesion at the new location. A “metastatic” or “metastasizing” cell is one that loses adhesive contacts with neighboring cells and migrates via the bloodstream or lymph from the primary site of disease to invade neighboring body structures. The terms also refer to the process of metastasis, which includes, but is not limited to detachment of cancer cells from a primary tumor, intravasation of the tumor cells to circulation, their survival and migration to a distant site, attachment and extravasation into a new site from the circulation, and microcolonization at the distant site, and tumor growth and development at the distant site. In certain embodiments, metastatic cancers that are amenable to treatment via the methods provided herein include, but are not limited to metastatic sarcomas, breast carcinomas, ovarian cancer, head and neck cancer, and pancreatic cancer.
[0051] The term "therapeutically effective amount" refers to an amount of an antibody, polypeptide, polynucleotide, small organic molecule, or other drug effective to "treat" a disease or disorder in a subject or mammal. In the case of cancer, the therapeutically effective amount of the drug can reduce the number of cancer cells; retard or stop cancer cell division, reduce or retard an increase in tumor size; inhibit, e.g., suppress, retard, prevent, stop, delay, or reverse cancer cell infiltration into peripheral organs including, for example, the spread of cancer into soft tissue and bone; inhibit, e.g., suppress, retard, prevent, shrink, stop, delay, or reverse tumor metastasis; inhibit, e.g., suppress, retard, prevent, stop, delay, or reverse tumor growth; relieve to some extent one or more of the symptoms associated with the cancer, reduce morbidity and mortality; improve quality of life; or a combination of such effects. To the extent the drug prevents growth and/or kills existing cancer cells, it can be referred to as cytostatic and/or cytotoxic.
[0052] Terms such as "treating" or "treatment" or “to treat” or "alleviating" or “to alleviate” refer to both 1) therapeutic measures that cure, slow down, lessen symptoms of, reverse, and/or halt progression of a diagnosed pathologic condition or disorder and 2) prophylactic or preventative measures that prevent and/or slow the development of a targeted pathologic condition or disorder. Thus those in need of treatment include those already with the disorder; those prone to have the disorder; and those in whom the disorder is to be prevented. A subject is successfully "treated" according to the methods of the present invention if the patient shows one or more of the following: a reduction in the number of or complete absence of cancer cells; a reduction in the tumor size; or retardation or reversal of tumor growth, inhibition, e.g., suppression, prevention, retardation, shrinkage, delay, or reversal of metastases, e.g., of cancer cell infiltration into peripheral organs including, for example, the spread of cancer into soft tissue and bone; inhibition of, e.g., suppression of, retardation of, prevention of, shrinkage of, reversal of, delay of, or an absence of tumor metastases; inhibition of, e.g., suppression of, retardation of, prevention of, shrinkage of, reversal of, delay of, or an absence of tumor growth; relief of one or more symptoms associated with the specific cancer; reduced morbidity and mortality; improvement in quality of life; or some combination of effects. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
[0053] By "subject" or "individual" or "animal" or "patient" or "mammal," is meant any subject, particularly a mammalian subject, for whom diagnosis, prognosis, or therapy is desired. Mammalian subjects include humans, domestic animals, farm animals, and zoo, sports, or pet animals such as dogs, cats, guinea pigs, rabbits, rats, mice, horses, cattle, cows, bears, and so on. The meaning of the terms “eukaryote”, “animal”, “mammal”, etc. is well known in the art and can, for example, be deduced from Wehner und Gehring (1995; Thieme Verlag). In the context of this invention, it is also envisaged that animals are to be treated which are economically, agronomically or scientifically important. Scientifically important organisms include, but are not limited to, mice, rats, and rabbits. Non-limiting examples of agronomically important animals are sheep, cattle and pigs, while, for example, cats and dogs may be considered as economically important animals. In one embodiment, the subject/patient is a mammal; in another embodiment, the subject/patient is a human or a non-human mammal (such as, e.g., a guinea pig, a hamster, a rat, a mouse, a rabbit, a dog, a cat, a horse, a monkey, an ape, a marmoset, a baboon, a gorilla, a chimpanzee, an orang utan, a gibbon, a sheep, cattle, or a pig); most preferably, the subject/patient is a human. II. Colchicine
[0054] In the following, colchicine used according to the present invention will be described in detail. The chemical structure of colchicine (ChemID 2012) is as follows:
Figure imgf000013_0001
[0055] The chemical name of colchicine is: N[5,6,7,9-tetrahydro-l,2,3,10-tetratmethoxy 9- oxobenzo[a]heptalen-7-yl],(S)-acetamide; molecular fo r m u 1 a : C 22 H 25 N Oe ; CAS number: 64-86-8.
[0056] Colchicine is an anti-inflammatory drug with a long history in human medicine, used for the symptomatic treatment of inflammatory diseases, most prominently gout. It is a natural product which can be extracted from two plants of the lily family, Colchicum autumnale and Gloriosa superba. Colchicine is a tricyclic alkaloid and has a molecular mass of 399.437. The active ingredient colchicine as well as its tablet formulation is listed in various national and international pharmacopeias such as the United States Pharmacopeia (USP).
[0057] The positive effect of its plant source in the treatment of rheumatism and swelling was described first already around 1500 B.C. in Egypt. Its use in gout was first described around 1500 years ago (Graham and Roberts, 1953, Ann Rheum Dis 12(1): 16-9). Today, the therapeutic value of colchicine is well established in a number of inflammatory diseases and approved by FDA for the prophylaxis and treatment of acute gout flares and familial Mediterranean fever (FMF). Other important established, though off-label uses are amongst others, Behcet's disease and recurrent pericarditis. In all known indications, it is generally administered orally as solid tablets in strengths of 0.5-0.6mg/tablet (e.g. Europe and United States, respectively). The pharmacotherapeutic mechanism of action of colchicine in diverse disorders is not fully understood, though it is known that the drug accumulates preferentially in leucocytes, particularly neutrophils which is important for its therapeutic effect. Three major interactions of colchicine with specific proteins modulate its pharmacokinetics: tubulin, cytochrome P450 3A4 (CYP3A4), and P-glycoprotein. It is assumed that most therapeutic effects of the drug are related to its capacity to bind to b- tubulin, thus inhibiting self-assembly and polymerization of microtubules. Availability of tubulin is essential for several cellular functions such as mitosis. Therefore colchicine effectively functions as a "mitotic poison" or spindle poison. By inhibiting microtubule self-assembly, colchicine interferes with many cellular functions involved in the immune response such as modulation of the production of chemokines chemokines and prostanoids and inhibition of neutrophil and endothelial cell adhesion molecules. Eventually it decreases neutrophil degranulation, chemotaxis and phagocytosis, thus reducing the initiation and amplification of inflammation. Colchicine also inhibits uric acid crystal deposition (a process important to the genesis of gout), which is enhanced by a low pH in the tissues, probably by inhibiting oxidation of glucose and subsequent lactic acid reduction in leukocytes (Imazio, Brucato el al. 2009, Eur Heart J, 30(5): 532-9; Cocco, Chu el al. 2010, Eur J Intern Med, 21(6): 503-8; Stanton, Gemert et al. 2011, Med Res Rev , 31(3): 443-81). In the management of pericarditis, colchicine excerpts its therapeutic effect by suppressing the acute pericardial inflammation. However, the exact cellular and molecular mechanisms of how colchicine relieves pain and inflammation in acute pericarditis and prevents recurrences are not fully understood.
[0058] The cancer findings from CANTOS relating to IL-Ib inhibition are fully consistent with long-standing evidence that many malignancies arise in areas of chronic inflammation (Coussens, 420:860-7, Nature, 2002; Grivenikov, Cell, 140:883-899, 2010) and that inadequate resolution of inflammation has a major role in tumor invasion, progression, and metastasis (Apte, Cancer Metastasis Rev., 25:387-408, 2006; Porta, Immunobiology, 214:761-77, 2009; Balkwill, Semin. Cancer Biol., 22:33-40, 2012). Inflammation has particular pathophysiologic relevance for lung cancer where chronic bronchitis, triggered by asbestos, silica, smoking, and other external inhaled toxins, results in a persistent inflammatory response (O’Callaghan, J. Thoracic. Oncol., 5:2024-36, 2010; Lee, Crit. Rev. Oncol. Hematol., 66:208-17, 2008). Inflammatory activation in the lung is mediated in part through activation of the Nod-like receptor protein 3 (NLRP3) inflammasome with consequent local generation of active IL-Ib, a process that can lead to both chronic fibrosis and cancer (Dostert, Science, 320:674-7, 2008; Gasse, /. Clin. Inv., 117:3786-99, 2007). In mice, inflammasome activation and pro-IL-Ib processing accelerates tumor invasiveness, growth, and metastatic spread (Apte, Cancer Metastasis Rev., 2006). For example, in IL- 1b -/- mice, neither local tumors nor lung metastases develop following localized or intravenous inoculation with melanoma cell lines, data suggesting that IL-Ib is required for tumor invasiveness (Voronov, Proc. Nat’l. Acad. Sci. USA, 100:2645-50, 2003). These data build on pioneering studies dating from the 1990’ s reporting that IL-1 induces augmentation of experimental metastases (Giavazzi, Cancer Res., 50:4771-4775, 1990), while IL-1 antagonism reduces the number and size of metastases in several tumor models (Chirivi, Cancer Res., 53:5051-5054, 1993; Vidal- Vanaclocha, Cancer Res., 54:2667- 2672, 1994).
[0059] Colchicine in the context of the present invention can be used for treating and/or preventing lung cancer, or inhibiting, delaying, or reducing tumor growth or metastases, or tumor growth and metastases in a subject with lung cancer.
III. Colchicine Formulations
[0060] The present invention additionally provides colchicine formulations to prevent and/or treat lung cancer, and to inhibit, delay, or reduce tumor growth or metastases in a subject in need of such inhibition, delay, or reduction, e.g., a lung cancer patient. In one embodiment, the colchicine formulation may be a sustained release formulation, wherein colchicine is released from the formulation at a sustained rate along a pre-determined or desired release profile. Such release is achieved by incorporation into the formulation of an extended release component and an optional immediate release component. In another embodiment, the colchicine formulation may be an immediate release formulation, wherein colchicine is released from the formulation quickly upon contacting a fluid such as water and allow fast leaching out of colchicine to the environment over a short period of time, such as several minutes or in an hour. The colchicine formulation of the present invention may be formulated in a dosage form selected from a tablet, a pill, a capsule, a caplet, a troche, a sachet, a cachet, a pouch, sprinkles, or any other form suitable for oral administration.
[0061] According to the present invention, colchicine as described herein (i.e., inter alia, in the form of a (pharmaceutical) composition) is administered in the form of immediate release preparation. As an immediate formulation, the dissolution rate is such that at least approximately 80% of colchicine is released within the first 60 mins. In general, at least approximately 90% of colchicine will be released within 2 hours. The immediate-release particles may be produced by any known method, such as dry or wet granulation method as described below. In one example, colchicine is mixed with disintegrants and/or binders, and adsorbents and then the mixture is subjected to either fluid bed granulation or spray drying to produce particles with desired immediate-release property. Examples of disintegrants include, but are not limited to, cross-linked polyvinyl pyrrolidone or crospovidone, starch derivatives such as carboxy methyl cellulose and cellulose derivatives; calcium alginate; carboxymethylcellulose calcium; carboxymethylcellulose sodium; croscarmellose sodium; docusate sodium; hydroxypropyl cellulose; magnesium aluminum silicate; methylcellulose; polacrilin potassium; sodium alginate; sodium starch glycolate and pregelatinized starch. Examples of adsorbents include, but are not limited to, aluminum hydroxide adjuvant; aluminum oxide; aluminum phosphate adjuvant; attapulgite; bentonite; powdered cellulose; colloidal silicon dioxide; hectorite; kaolin; magnesium aluminum silicate; magnesium carbonate; microcrystalline cellulose; pectin; polycarbophil; and saponite.
[0062] According to the present invention, colchicine as described herein (i.e., inter alia, in the form of a (pharmaceutical) composition) is administered in the form of a sustained release preparation. Other expressions like “extended release”, “controlled release”, “modified release” or “delayed release” “preparation” or “formulation” are understood herein to have the same meaning as “sustained release preparation”. Such preparations can in principal be in any form conceivable to the skilled person and include pharmaceutical forms for oral (solid, semi-solid, liquid), dermal (dermal patch), sublingual, parenteral (injection), ophthalmic (eye drops, gel or ointment) or rectal (suppository) administration, as long as a sustained release is ensured.
[0063] In accordance with the invention, sustained release preparations encompass all pharmaceutical forms that create a steady drug release profile making the drug substance available over an extended period of time following application to the patient. Such an extended period of time may be between 10, 20, 30, 40, 50 or 60 minutes and about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours. Extended release may also be defined functionally as the release of over 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 99 percent (%) of colchicine after about 10, 20, 30, 40, 50 or 60 minutes and about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or 24 hours. Extended release as used herein may also be defined as making colchicine available to the patient regardless of uptake, as some colchicine may never be absorbed by the patient. Various extended release dosage forms may be designed readily by one of skill in art as disclosed herein to achieve delivery and sustained release of colchicine to the liver and/or both the small and large intestines, to only the small intestine, or to only the large intestine.
[0064] In some embodiments, sustained release preparations may be pH independent. This allows such preparations to dissolve in almost any environment. In other embodiments, sustained release preparations may be pH dependent. This allows release to be accomplished at some generally predictable location in the lower intestinal tract more distal to that which would have been accomplished if there had been no delayed release alterations. A method for delay of release is, e.g., a coating. Any coatings should be applied to a sufficient thickness such that the entire coating does not dissolve in the gastrointestinal fluids at pH below about 5, but does dissolve at pH about 5 and above. It is expected that any anionic polymer exhibiting a pH-dependent solubility profile can be used as an enteric coating in the practice of the present invention to achieve delivery to the lower gastrointestinal tract. Polymers and compatible mixtures thereof may be used to provide the coating for the delayed or the extended release of active ingredients, and some of their properties, include, but are not limited to: shellac, also called purified lac, a refined product obtained from the resinous secretion of an insect. This coating dissolves in media of pH >7.
[0065] In some embodiments, sustained release preparations may be influenced by the presence of alcohol in the body. The presence of alcohol is a patient’s body can increase dissolution of the composition and can cause immediate release of the entire dose. This effect is known as “dose dumping” and is dependent on the alcohol solubility of the materials. For sustained release preparations which contain a higher dose for slow release over 24 hours, for instance, this effect can have safety concerns and can even be life threatening.
[0066] To achieve a uniform or continuous rate of release, sustained release preparations may be prepared using time release hydrophilic matrices. These time release hydrophilic matrices are known in the field of drug formulations. For example, one such hydrophilic matrix is hydroxypropyl methylcellulose (HPMC) or Hypromellose. Hydrophilic matrices provide an initial release of the drug product in the initial phase mainly triggered by a rapid swelling of the surface of the matrix tablet, combined with an erosion process leading to an immediate release of the drug substance distributed close to the surface of the tablet. In an embodiment, about 50%, about 45%, about 40%, about 35%, about 30%, about 25%, about 20%, about 15%, or about 10% of the drug substance may immediately be released depending on the desired release profile. In an embodiment, at least about 20% of the drug substance may immediately be released. In another embodiment, at least about 20% of the drug substance may be released within about the first 30 minutes. As used herein, the term "about" or “approximately” refers to a variation of 10% from the indicated values (e.g., 50%, 45%, 40%, etc.), or in case of a range of values, means a 10% variation from both the lower and upper limits of such ranges. For instance, “about 50%” refers to a range of between 45% and 55%. Within the initial swelling of the tablet surface a gel formation of the hydrophilic matrix starts. This gelling prevents the tablet core from dissolving and disintegrating immediately, thereby allowing the main part of the drug substances to dissolve slowly over time within in this gel structure and diffuse into solution following the rules of Fick's law. The diffusion itself may be triggered in this formulation approach by the concentration of the Hypromellose and the viscosity of the formed gel, defined over the molecular weight of the Hypromellose. Therefore, drug release profiles can be modified by varying different viscosity grades of Hypromellose or mixtures thereof. All corresponding formulation and process parameters achieving the predicted release profile are common knowledge and can be adjusted using actual development technologies e.g. formulation screenings, statistical trials designs.
[0067] In an embodiment, the substance responsible for sustained release of the controlled-release formulation can further mix with a binder. The binder is added to increase the mechanical strength of the granules and tablets during formation. Binders can be added to the formulation in different ways: (1) as a dry powder, which is mixed with other ingredients before wet agglomeration, (2) as a solution, which is used as agglomeration liquid during wet agglomeration, and is referred to as a solution binder, and (3) as a dry powder, which is mixed with the other ingredients before compaction. In this form the binder is referred to as a dry binder. Solution binders are a common way of incorporating a binder into granules. In certain embodiments, the binder used in the formulation is in the form of a dry powder binder. Non-limiting examples of binders useful for the core include hydrogenated vegetable oil, castor oil, paraffin, higher aliphatic alcohols, higher aliphatic acids, long chain fatty acids, fatty acid esters, wax-like materials such as fatty alcohols, fatty acid esters, fatty acid glycerides, hydrogenated fats, hydrocarbons, normal waxes, stearic acid, stearyl alcohol, hydrophobic and hydrophilic polymers having hydrocarbon backbones, and mixtures thereof. Specific examples of water-soluble polymer binders include modified starch, gelatin, polyvinylpyrrolidone, cellulose derivatives (such as for example hydroxypropyl methylcellulose (HPMC) and hydroxypropyl cellulose (HPC)), polyvinyl alcohol and mixtures thereof. In an embodiment, the binder is HPMC. In another embodiment, the binder is Hypromellose 6mPa*s. In an embodiment, the binder can be present in an amount of from about 1% to about 30% by weight of the formulation.
[0068] In another embodiment of the invention, the sustained release formulation may include a disintegrant. A disintegrant refers to an agent used in pharmaceutical preparation of tablets, which causes them to disintegrate and release their medicinal substances on contact with moisture. In an embodiment, the disintegrant may be water soluble to support the disintegrantation of a tablet in the stomach. Non-limiting examples of disintegrants for use in the formulation include sucrose, lactose, in particular lactose monohydrate, trehalose, maltose, mannitol and sorbitol, croscarmellose sodium, crospovidone, alginic acid, sodium alginate, methacrylic acid DVB, cross-linked PVP, microcrystalline cellulose, polacrilin potassium, sodium starch glycolate, starch, pregelatinized starch and mixtures thereof. In at least one embodiment the disintegrant is selected from microcrystalline cellulose (e.g. Avicel PH101), cross-linked polyvinylpyrrolidone (e.g. KOLLIDON® CL), cross-linked sodium carboxymethylcellulose (e.g. AC-DI-SOL(TM)), starch or starch derivatives such as sodium starch glycolate (e.g. EXPLOTAB®), or combinations with starch (e.g. PRIMOJEL(TM)), swellable ion-exchange resins, such as AMBERLITE(TM) IRP 88, formaldehyde-casein (e.g. ESMA SPRENG(TM)), and mixtures thereof.
[0069] In another embodiment of the invention, the sustained release formulation may include a filling agent or filler. A filling agent refers to an inert substance used as filler to create desired bulk, flow properties, and compression characteristics in preparation of tablets. Non-limiting examples of filling agents for use in the formulation include sucrose, lactose, in particular lactose monohydrate, trehalose, maltose, mannitol and sorbitol, croscarmellose sodium, crospovidone, alginic acid, sodium alginate, methacrylic acid DVB, cross-linked PVP, microcrystalline cellulose, polacrilin potassium, sodium starch glycolate, starch, pregelatinized starch and mixtures thereof. In an embodiment, lactose monohydrate is included as a filling agent in an amount of about 10% to about 80%, preferably about 59%, by weight of the tablet. In an embodiment, pregelatinized starch is included as a filling agent in an amount of about 5% to about 50%, preferably about 7.5%, by weight of the tablet.
[0070] In another embodiment, the sustained release formulation of the present invention may include a release retarding agent for maintaining a uniform release rate of the drug. Examples of retarding agents include, but are not limited to, cellulose ethers, cellulose esters, acrylic acid copolymers, waxes, gums, glyceryl fatty acid esters and sucrose fatty acid esters. In one embodiment, the retarding agent is RETALAC® (Meggle), a spray agglomerated blend of 50 parts lactose monohydrate and 50 parts hypromellose. The viscosity of hypromellose used herein may range from 6 mPa*s - 100,000 mPa*s. In an embodiment, the viscosity of hypromellose used is 4000 mPa*s. Adjusting the amount of retarding agent in the composition may alter the release rate of the drug. In one embodiment, the retarding agent of the formulation of the present invention releases colchicine in a continuous and uniform manner and is adjusted in such a way that about 80% of the active ingredient is released in vitro in the predetermined period of time. By way of example, and by no means limiting the scope of the invention, the period of time may be not more than 24 hours, not more than 16 hours, not more than 12 hours, not more than 8 hours, not more than 6 hours, not more than 4 hours, not more than 3.5 hours, or not more than 1.5 hours depending on desired attributes of the final product. It is understood that the release rate can vary based on whether the experiment is conducted in vitro or in vivo. Therefore, if the desired release rate is between about 1.5 to about 3.5 hours in vitro or between about 1.5 to about 6 hours in vitro, the release rate under in vivo conditions, depending on the experimental conditions, may actually be different. In an embodiment, the sustained release formulation of the present invention releases colchicine in a continuous and uniform manner in such a way that about 80% of the active ingredient is released in vitro in between about 1.5 and about 3.5 hours.
[0071] In another embodiment, the formulation of the present invention may include a glidant. A glidant can be used to improve powder flow properties prior to and during tableting and to reduce caking. Suitable glidants include colloidal silicon dioxide, magnesium trisilicate, powdered cellulose, talc, tribasic calcium phosphate and the like. In one embodiment, talc is included as a glidant in an amount of about 0.05% to about 5%, preferably about 1%, by weight of the tablet. [0072] In another embodiment, the formulation of the present invention may include a lubricant. Lubricants can be added to pharmaceutical formulations to decrease any friction that occurs between the solid and the die wall during tablet manufacturing. High friction during tableting can cause a series of problems, including inadequate tablet quality (capping or even fragmentation of tablets during ejection, and vertical scratches on tablet edges) and may even stop production. Accordingly, lubricants are added to certain tablet formulations of the present invention including certain embodiments of the formulation described herein. Non-limiting examples of lubricants useful for the core include glyceryl behenate, stearic acid, hydrogenated vegetable oils (such as hydrogenated cottonseed oil (STEROTEX®), hydrogenated soybean oil (STEROTEX® HM) and hydrogenated soybean oil & castor wax (STEROTEX® K)), stearyl alcohol, leucine, polyethylene glycol (MW 1450, suitably 4000, and higher), magnesium stearate, glyceryl monostearate, polyethylene glycol, ethylene oxide polymers (for example, available under the registered trademark CARBOWAX® from Union Carbide, Inc., Danbury, Conn.), sodium lauryl sulfate, magnesium lauryl sulfate, sodium oleate, sodium stearyl fumarate, DL-leucine, colloidal silica, mixtures thereof and others as known in the art. In one embodiment, stearic acid is included as a lubricant in an amount of about 0.05% to about 5%, preferably about 1%, by weight of the tablet.
[0073] Sweeteners that can also be used in the taste-masking coating of certain embodiments of the matrix dosage forms include glucose (corn syrup), dextrose, invert sugar, fructose, and mixtures thereof (when not used as a carrier); saccharin and its various salts, such as sodium salt; dipeptide sweeteners such as aspartame; dihydrochalcone compounds, glycyrrhizin; Steva Rebaudiana (Stevioside); chloro derivatives or sucrose such as sucralose; and sugar alcohols such as sorbitol, mannitol, xylitol, and the like. Also contemplated are hydrogenated starch hydrolysates and the synthetic sweeteners such as 3,6-dihydro-6- methyl-1-1-1, 2, 3-oxathiazin-4-l-2, 2-dioxide, particularly the potassium salt (acesulfame- K), and sodium and calcium salts thereof. The sweeteners can be used alone or in any combination thereof.
[0074] The formulation of the present invention can further contain one or more pharmaceutically acceptable excipients such as granulating aids or agents, colorants, flavorants, pH adjusters, anti-adherents, glidants and like excipients conventionally used in pharmaceutical compositions. In an embodiment, a coloring excipient can be advantageously added as giving rise to visual change preventing abuse. It can color simultaneously the liquid or the particles or one independently of the other. Among suitable coloring excipients the following may be cited: indigotine, cochineal carminic acid, yellow orange S, allura red AC, iron oxides, cucurmin, riboflavin, tartrazine, quinoline yellow, azorubine, amaranth, carmines, erythosine, red 2G, patented blue V, glittering blue FCF, chlorophylls, copper complexes of chlorophylls, green S, caramel, glittering black BN, carbo medicinalis vegetabilis, brown FK and HT, carotenoids, Annatto extracts, paprika extracts, lycopene, lutein, canthaxanthin, beetroot red, anthocyanes, calcium carbonate, titanium dioxide, aluminium, silver, gold or litholmbin BK or any other coloring excipient suitable for an oral administration.
[0075] In an embodiment, a formulation may be coated. Coatings may provide a variety of functions. In some embodiments, coatings may be used, for example, to achieve delayed release, resistance to acid, targeted release in the lower GI tract, avoidance of bad taste in mouth. In some embodiments, coatings may be used to protect the API/tablct from light and provide for better mechanical resistance. Of course it should be appreciated that a coating may serve other functions as well and a person skilled in the art knows the purpose of tablet coating.
[0076] The pharmaceutical composition and/or the solid carrier particles can be coated with one or more enteric coatings, seal coatings, film coatings, barrier coatings, compress coatings, fast disintegrating coatings, or enzyme degradable coatings. Multiple coatings may be applied for desired performance. Further, one or more of the actives may be provided for immediate release, pulsatile release, controlled release, extended release, delayed release, targeted release, synchronized release, or targeted delayed release. In fact, the formulation may include combinations of typical pharmaceutical actives (e.g., pseudephedrin) and vitamins (e.g., Vitamin C), minerals (Ca, Mg, Zn, K) or other supplements (e.g., St. John's Wort, echinacae, amino acids). For release/absorption control, solid carriers can be made of various component types and levels or thicknesses of coats, with or without an active ingredient. Such diverse solid carriers can be blended in a dosage form to achieve a desired performance. The liquid formulations may be delivered to, and adapted for, oral, nasal, buccal, ocular, urethral, transmucosal, vaginal, topical or rectal delivery, although oral delivery is used mostly. [0077] When formulated with microparticles or nanoparticles, the drug release profile can easily be adapted by adding a coating, e.g., a hard or soft gelatin coating, a starch coating, a resin or polymer coating and/or a cellulosic coating. Although not limited to microparticles or nanoparticles (as in, e.g., microcapsules or nanocapsules), such dosage forms may be further coated with, for example, a seal coating, an enteric coating, an extended release coating, or a targeted delayed release coating. The term "enteric coating" as used herein relates to a mixture of pharmaceutically acceptable excipients that is applied to, combined with, mixed with or otherwise added to the carrier or composition. The coating may be applied to an active that is compressed, molded or extruded and may also include: gelatin, and/or pellets, beads, granules or particles of the carrier or composition. The coating may be applied through an aqueous dispersion or after dissolving in appropriate solvent. The carrier may or may not be fully or partially biodegradable.
[0078] In an embodiment, polymethacrylate acrylic polymers can be employed as coating polymers. In at least one embodiment, the coating is an acrylic resin lacquer used in the form of an aqueous dispersion, such as that which is commercially available from Rohm Pharma under the trade name EUDRAGIT® or from BASF under the trade name KOLLICOAT®. In a more preferable embodiments, EUDRAGIT® E100 is used as the coating polymer, which is a cationic copolymer based on dimethylaminoethyl methacrylate and neutral methacrylic esters having a average molecular weight is approximately 150,000. Different coating polymers of the certain embodiments can be mixed together in any desired ratio in order to ultimately obtain a coating having a desirable drug dissolution profile. Coating methods can consist in spraying a solution of the polymer on the tablets, either in a pan coater or a fluid bed coating apparatus. The solvent may be organic or aqueous, depending on the nature of the polymer used. In a preferable embodiment, the solvent is alcohol. Coating methods are well known in the art.
[0079] The compositions of the present invention can also be formulated as enteric coated delayed release oral dosage forms, i.e., as an oral dosage form of a pharmaceutical composition as described herein that uses an enteric coating to effect release in the lower gastrointestinal tract. The enteric coated dosage form will generally include microparticles, microgranules, micropellets or microbeads of the active ingredient and/or other composition components, which are themselves coated or uncoated. The enteric coated oral dosage form may also be a capsule (coated or uncoated) containing pellets, beads or granules of the solid carrier or the composition, which are themselves coated or uncoated.
[0080] Carriers for use with the present invention include permeable and semipermeable matrices or polymers that control the release characteristics of the formulation. Such polymers include, for example, cellulose acylates, acetates, and other semi-permeable polymers such as those described in U.S. Pat. No. 4,285,987 (hereby incorporated by reference), as well as the selectively permeable polymers formed by the coprecipitation of a polycation and a polyanioni as disclosed in U.S. Pat. Nos. 3,173,876; 3,276,586; 3,541,005; 3,541,006 and 3,546,142 (relevant portions incorporated herein by reference).
[0081] Other carriers for use with the present invention include, e.g., starch, modified starch, and starch derivatives, gums, including but not limited to xanthan gum, alginic acid, other alginates, benitoniite, veegum, agar, guar, locust bean gum, gum arabic, quince psyllium, flax seed, okra gum, arabinoglactin, pectin, tragacanth, scleroglucan, dextran, amylose, amylopectin, dextrin, etc., cross-linked polyvinylpyrrolidone, ion-exchange resins, such as potassium polymethacrylate, carrageenan (and derivatives), gum karaya, biosynthetic gum, etc. Other useful polymers include: polycarbonates (linear polyesters of carbonic acid); microporous materials (bisphenol, a microporous poly(vinylchloride), micro-porous polyamides, microporous modacrylic copolymers, microporous styrene- acrylic and its copolymers); porous polysulfones, halogenated poly(vinylidene), polychloroethers, acetal polymers, polyesters prepared by esterification of a dicarboxylic acid or anhydride with an alkylene polyol, poly(alkylenesulfides), phenolics, polyesters, asymmetric porous polymers, cross-linked olefin polymers, hydrophilic microporous homopolymers, copolymers or interpolymers having a reduced bulk density, and other similar materials, poly (urethane), cross-linked chain-extended poly (urethane), poly(imides), poly(benzimidazoles), collodion, regenerated proteins, semi-solid cross-linked poly(vinylpyrrolidone) .
[0082] Additional additives and their levels, and selection of a primary coating material or materials will depend on the following properties: pH levels at target site, desirability to make tablet pH dependent or pH independent, solubility in alcohol, resistance to dissolution and disintegration in the stomach; impermeability to gastric fluids and drug/carrier/enzyme while in the stomach; ability to dissolve or disintegrate rapidly at the target intestine site; physical and chemical stability during storage; non-toxicity; easy application as a coating (substrate friendly); and economical practicality.
[0083] Further to the above, various formulations, not limiting the scope of the present invention, illustrating the invention are described hereafter. A controlled-release tablet or capsule or the like comprises colchicine as a core coated with an immediate release layer. A controlled-release double layer tablet or capsule or the like comprises a layer of sustained release and a layer of immediate release. A controlled-release tablet with more than two layers comprises (i) one or two more layers of substance controlling the sustained release and (ii) one or two more layers of immediate release.
[0084] According to one embodiment of the invention, the composition comprising colchicine is further coated with at least one release- slowing intermediate layer of slightly soluble intermediate layer to control release of colchicine.
[0085] Traditionally, colchicine immediate release dosage forms (mostly tablets, also injections or oral solutions) have been used in the treatment of gout or FMF. Worldwide, all approved pharmaceuticals containing colchicine are approved only for gout and/or FMF and are immediate release tablets. Colchicine can be used in the prevention of certain other inflammatory diseases such as pericarditis, PPS and, most recently, patients with stable coronary heart diseases. The difference between treatment and prevention with regard to colchicine is that in treatment, an overt disease and/or ongoing inflammation has to be treated. Thus high levels of colchicine are required, which usually goes hand in hand with unwanted side effects, most prominently gastrointestinal insults, as well as increased risk of colchicine related toxicity. In prevention, one does not have to suppress ongoing inflammation but rather suppress an outbreak of inflammation. Thus, supposedly lower and steadier levels of colchicine are required and are beneficial. As described in the present invention, this is achieved by administering colchicine formulated as a sustained release preparation, as described above.
[0086] In the case of inhibiting, delaying, or reducing tumor growth or metastases in a subject in need of such inhibition, delay, or reduction, e.g., a cancer patient, lower or higher levels of colchicine may be necessary to inhibit IL-Ib activity. The sustained release system facilitates more steady levels of colchicine and reduces the incidence of adverse events.
[0087] An advantage of colchicine administered as sustained release is, e.g., a flattening of the serum level curve (lower but broader peak levels) reduces the incidence of serious adverse events related to colchicine toxicity, also in case of potential drug interactions, thereby increasing compliance. Much of colchicine related toxicity comes from the fact that one or both of the excretion pathways (liver and kidney) is reduced in its activity, either by other drugs or by a disease (e.g. kidney insufficiency). In the case of a slower and extended drug absorption (extended release), the body has also more time to excrete the colchicine from the system. In this case, it is less likely that colchicine levels reach toxic levels in case of defective excretion (due to drug interaction or disease). Another potential advantage of colchicine administered as a sustained release formulation is that plasma levels remain more evenly distributed (i.e., the variability of plasma levels, such as the differences in Cmax, Tmax, AUC or other pharmacokinetic parameters, among patients is reduced), resulting in fewer “non-responders” to the treatment. In addition, administration of colchicine as sustained release is resistant to dose dumping, therefore the dissolution of the composition is not significantly influenced by alcohol.
[0088] Further, sustaining the release expands the time where colchicine is present in the blood in therapeutic levels. This results in a more efficient inhibition of disease progression and, thus, improving the clinical outcome.
[0089] Furthermore, for the prophylactic uses described herein, colchicine does not have to go deep into the tissue (like for gout), it may be active in the blood system directly (in the vessels) where it acts on the plaques and especially on inflammatory blood cells (neutrophils). This means, less total colchicine and lower serum levels can be therapeutic. Fast and high colchicine levels, e.g., as for treating an acute gout flare can be avoided. Thus, lower levels of colchicine, e.g., about 0.1 to about 0.75mg sustained release formulations as described above (or even less frequent doses), may be sufficient to achieve the desired clinical outcome.
[0090] In the normal situation, most colchicine is absorbed from the small intestine and most passes the liver (some is also excreted in the urine via kidney). There it is metabolized but quite a large proportion of colchicine goes through the liver un-metabolized. This means, it goes through the liver into the bile and from there it is excreted into the big intestine (colon). There it can be resorbed into the body again which leads to the characteristic second peak (accounts for about 50% of totally absorbed colchicine and is thought to be responsible for gastrointestinal problems, such as diarrhea). If colchicine is formulated as a sustained release preparation as described above, a slower release of colchicine results in a slower resorption. This results in more complete metabolism of colchicine in the liver (because it is less busy with colchicine at a time) and, thus, less recirculation of un metabolized colchicine. This consequently reduces the incidence of gastrointestinal problems and increases compliance. Colchicine administered as sustained release in accordance with the present invention may also be beneficial for other known side/adverse effects associated with colchicine treatment/administration (the skilled person is well aware of the adverse effects that may occur upon colchicine administration or colchicine treatment). Thus, administration of colchicine as sustained release, as described above, results in a safety increase and safety benefit.
IV. Methods of Preparing a Sustained Release Formulation
[0091] The current invention additionally encompasses a method of preparing formulations of colchicine, comprising a sustained release component, and an optional immediate release component, wherein colchicine is released from the formulation at the sustained rate along the pre-determined or desired release profile.
[0092] In one embodiment, the colchicine compositions described in the present invention is in the form of a tablet. As used herein, the term “tablet” means a compressed pharmaceutical dosage form of any shape or size. The tablets described herein may be obtained from the compositions comprising colchicine and a pharmaceutically acceptable excipient. Any of the colchicine compositions can be in the form of any other dosage form known in the art, specifically, any oral dosage form, for example a capsule.
[0093] In a first aspect of the invention, there is provided a controlled release formulation for use in oral dosage forms. The formulation includes a mixture containing hypromellose as a hydrophilic matrix, which is effective to provide controlled release of a pharmaceutically active ingredient.
[0094] Matrix systems are well known in the art. In a typical matrix system, the drug is homogenously dispersed in a polymer in association with conventional excipients. This admixture is typically compressed under pressure to produce a tablet. The API is released from the tablet by diffusion and erosion. Matrix systems are described in detail by (i) Handbook of Pharmaceutical Controlled Release Technology, Ed. D. L. Wise, Marcel Dekker, Inc. New York, N.Y. (2000), and (ii) Treatise on Controlled Drug Delivery, Fundamentals, Optimization, Applications, Ed. A. Kydonieus, Marcel Dekker, Inc. New York, N.Y. (1992), the contents of both of which are hereby incorporated by reference. [0095] When the tablet is exposed to aqueous media, such as in the gastrointestinal tract, the tablet surface wets and the polymer begins to partially hydrate forming an outer gel layer. This outer gel layer becomes fully hydrated and begins to erode into the aqueous fluids. Water continues to permeate toward the core of the tablet permitting another gel layer to form beneath the dissolving outer gel layer. These successive concentric gel layers sustain uniform release of the API by diffusion from the gel layer and exposure through tablet erosion. In the case of the mixtures of the present invention, when included in a compressed tablet matrix, the hypromellose provides a hydrophilic swellable structure capable of functioning as the gel layer. In this way, the drug release is controlled.
[0096] In accordance with one embodiment, the colchicine formulation of the present invention can be manufactured by either wet or dry granulation of a colchicine composition, blending the resulting granulate with excipients, and then compressing the composition into tablets.
[0097] In one embodiment, wet granulation is used to prepare wet granules comprising colchicine. A granulating liquid is used in wet granulation process. Both aqueous and non-aqueous liquids may be used as the granulating liquid. In one embodiment, the granulating liquid is an aqueous liquid, or more specifically, purified or de-ionized water. The amount of the granulating liquid used may depend on many factors, for example, the type of the granulating liquid, the amount of the granulating liquid used, the type of excipient used, the nature of the active agent, and the active agent loading.
[0098] In one embodiment, the colchicine particles and suitable excipients are mixed with the granulating liquid for a sufficiently long period to facilitate good distribution of all starting materials and good content uniformity. Wet granulation is generally performed at temperatures between about 20° C. to about 35° C., or more specifically, at room temperature (about 25° C.). Following wet granulation, the granulate is dried at increased temperatures to yield a dry granulate. In an embodiment, the step of drying may be performed for a sufficiently long period until the desired residual moisture content is reached. In an embodiment, this may be about 45°C for about 12-48 hours. It should be appreciated that the overall time to perform the granulation process may depend on a variety of factors, including but not limited to, the solvents used, batch size, instruments used, etc.
[0099] Any equipment may be used to contact the granulating liquid with the colchicine and the excipients as long as uniform distribution of the granulating liquid is achieved. For example, small-scale production can be achieved by mixing and wetting the colchicine and the excipients in mortars or stainless steel bowls, while for larger quantities, V-blenders with intensifier bars, planetary mixers, rotary granulators, high shear granulators, and fluid- bed granulation equipment may be used. In one embodiment, the granulator is a high shear granulator.
[00100] In one embodiment, a method of making a colchicine composition comprises wet granulating colchicine with pharmaceutically acceptable excipients and a granulating liquid to obtain wet granules, and mixing the granules in a next step with a second excipient to obtain a colchicine composition. In one embodiment, the pharmaceutically acceptable excipient comprises a binder and a filler. In an embodiment, the binder may be Hypromellose. In an embodiment, the filler may be lactose monohydrate and pregelatinized starch. In another embodiment, purified water is used as the granulating liquid. In an embodiment, the second excipient mixed with the granules may be a filler. In an embodiment, the filler may be lactose monohydrate. The colchicine compositions can contain about 0.1 wt % to about 10 wt %, or more specifically, about 0.25 wt % to about 0.75 wt %, of colchicine, based on the total weight of the colchicine composition.
[00101] In an embodiment, the method of making a composition comprises wet granulating colchicine with a pharmaceutically acceptable excipient to obtain wet granules, and mixing the granules with a filler to obtain a colchicine composition. In some embodiments, the method further includes drying the mixture. In another embodiment, the wet granules are dried to obtain dried granules, and then the dried granules are mixed with a binder, a filler, or both to obtain the composition. In another embodiment, the dried granules can be milled to obtain milled granules before mixing the milled dried granules with the binder, a filler, or both. The method can further include mixing the colchicine composition with a glidant, a lubricant, or both to obtain a blend or compressing the blend to obtain a tablet. In one embodiment, the glidant may be Talc. In another embodiment, the lubricant may be Stearic acid. The method can further include coating the tablet.
[00102] In another embodiment, a method of making a colchicine tablet comprises wet granulating colchicine with a pharmaceutically acceptable excipient to obtain wet granules; drying the wet granules to obtain dried granules; milling the dried granules to obtain milled granules; mixing the milled granules with a filler to obtain the composition; mixing the composition with a glidant, a lubricant, or both to obtain a blend; and compressing the blend to obtain a colchicine tablet of the present invention. [00103] In some embodiments, the wet granules are dried to obtain dried granules before mixing with a second excipient, for example a filler. Wet granules can be dried by any suitable means to remove the granulating liquid and to form dried granules containing colchicine and the pharmaceutically acceptable excipient. The conditions and duration of drying depend on factors such as the liquid used and the weight of the granulating particles. Examples of suitable drying methods include, but are not limited to, tray drying, forced air drying, microwave drying, vacuum drying and fluid bed drying.
[00104] After drying, dried granules may be mixed directly with an excipient, for example, a filler, a binder, or a lubricant, for further processing. Alternatively, dried granules may optionally be subjected to additional processing steps prior to mixing with the excipient. For example, dried granules may be sized to reduce particle size prior to mixing with an excipient. Exemplary sizing operations include milling or sieving. Any suitable equipment for reducing the particle size may be used in the present invention.
[00105] Suitable excipients may be added extragranularly and mixed with the granules to form colchicine compositions. As used herein, the term “extragranular” or “extragranularly” means that the referenced material, for example, a suitable excipient, is added or has been added as a dry component after wet granulation. In one embodiment, a filler, a binder, a glidant and a lubricant are added extragranularly to the granules and mixed to form a blend. The blend may be encapsulated directly into capsule shells, for example, hard gelatin shells, to form capsule formulations. Alternatively, the blend may be compressed into tablets. In some embodiments, the granules are dried granules or milled, dried granules.
[00106] Mixing can be carried out for a sufficient time to produce homogeneous mixtures or blends. Mixing may be accomplished by blending, stiffing, shaking, tumbling, rolling, or by any other method to achieve a homogeneous blend. In some embodiments, the components to be mixed are combined under low shear conditions in a suitable apparatus, such as a V-blender, tote blender, double cone blender or any other apparatus capable of functioning under low shear conditions.
[00107] The homogenous mixtures or blends are then compressed using any method suitable in the industry. The mechanical force will define the physical properties of the tablets, especially the crushing strength of the resulting tablet. The mechanical strength interacts with the initial swelling of the tablet and dilution speed of the tablet core. This effect is well known in the art and can be adjusted and controlled during the lifecycle of the product. For the colchicine sustained-release formulation of the present invention, the compression strengths used may range from about 30N to about 130N. In one embodiment, the compression strength may be about 100N. In another embodiment, the compression strength may be about 100N +/- 15N.
[00108] The colchicine tablets prepared from the above described methods exhibit acceptable physical characteristics including good friability and hardness. As per EP and USP guidelines, the colchicine tablets disclosed herein have friability in the range of about 0% to less than about 1%.
[00109] The colchicine tablet can be coated. Coating the tablet may be performed by any known process. A coating for the colchicine tablet disclosed herein can be any suitable coating, such as, for example, a functional or a non-functional coating, or multiple functional or non-functional coatings. By “functional coating” is meant to include a coating that modifies the release properties of the total formulation, for example, a sustained- release coating. By “non-functional coating” is meant to include a coating that is not a functional coating, for example, a cosmetic coating. A non-functional coating can have some impact on the release of the active agent due to the initial dissolution, hydration, perforation of the coating, etc., but would not be considered to be a significant deviation from the non-co ated composition.
[00110] In one embodiment, a colchicine composition comprises colchicine, a binder, a filler, a retarding agent, a glidant, and a lubricant. In an embodiment, a colchicine composition comprises about 0.25 to about 0.75 mg colchicine; about 10 to about 80 mg lactose monohydrate; about 5 to about 50 mg pregelatinized starch; about 1 to about 30 mg Hypromellose 6mPa*s; about 5 to about 40 mg Retalac (compound of lactose monohydrate and Hypromellose 4000mPa*s 50/50 w/w %); about 0.5 to about 5 mg Talc; and about 0.5 to about 5 mg Stearic acid 50. In an embodiment, the colchicine composition comprises about 0.5 mg colchicine, about 59 mg lactose monohydrate; about 7.5 mg pregelatinized starch; about 1 mg Hypromellose 6mPa*s; about 30 mg Retalac (compound of lactose monohydrate and Hypromellose 4000mPa*s 50/50 w/w %); about 1 mg Talc; and about 1 mg Stearic acid 50. The colchicine dosage form has a total weight of about 100 mg. The colchicine composition can be in the form of a tablet. V. Treatment Methods Using Colchicine As a Single Agent or In Combination With At Least One Immune Modulating Therapy to Treat and/or Prevent Lung Cancer
[00111] The present invention also presents a method to treat and/or prevent, or inhibit, delay, or reduce tumor growth or metastases in a subject in need of such inhibition, delay, or reduction, e.g., a lung cancer patient, comprising administering to the subject a therapeutically effective amount of a colchicine formulation of the present invention. The method of the current invention possesses the flexibility to selectively adjust the pharmacokinetics of the administered formulations depending on the nature of the condition and needs of the patients due to the novel design of the colchicine formulation that comprises an extended release component and an optional immediate release component, and the release profiles of both components can be selectively modified during the preparation process as described above to comply with the predetermined release profile.
[00112] In one embodiment, treatment includes the application or administration of a colchicine formulation as described herein to a patient, where the patient has, or has the risk of developing lung cancer. In another embodiment, treatment is also intended to include the application or administration of a pharmaceutical composition comprising the colchicine formulation, to a patient, where the patient has, or has the risk of developing lung cancer.
[00113] Colchicine is an anti-inflammatory agent, known as the drug of choice for management of gout. It has been shown that its anti-inflammatory effect is mediated by inhibition of the pro-inflammatory cytokines IL-1 and IL-Ib release. Moreover, by restraint of the NF-KB pathway and blocking cell mitosis, colchicine may exert an inhibitory effect on tumorigenesis. Addition of colchicine to human LPS stimulated PBMC exerted a disruptive effect on cellular microtubules with a consequent increased IL-Ib and a decreased TNF-a release. In a recent study it has been shown that colchicine added to co cultures of PBMC with either FIT-29 or RKO human colon cancer cells promoted cancer cells- stimulated PBMC to produce IL-Ib and to inhibit the release of TNF-a and IL-10.
[00114] On an indication and patient-by-patient basis, not all tumors contain immune cells. Tumor types that on average harbor a greater concentration of infiltrating leukocytes include: melanoma, colorectal cancer, non-small cell lung cancer and head and neck squamous cell carcinoma. In these tumors, the lymphocytic infiltrate is thought to reflect how foreign or "visible" a tumor is to the immune system which in turn is connected to the extent of “disregulation” or “aberration” within the tumor. Yet despite effective recruitment of immune cells to the tumor, they generally do not succumb to immune- mediated regression. This is believed to be the effect of suppressive or resistance mechanisms adopted by the tumor. These mechanisms generally include the activity of myeloid derived suppressor cells (MDSC), M2 macrophage, regulatory T cells, and dendritic cells on defective antigen presentation, the production of suppressive cytokines, impaired costimulation for effector T cells and antigen loss.
[00115] In tumors with high numbers of infiltrating leukocytes, a new class of therapeutics called checkpoint inhibitors has been shown to release or remove suppressive immune elements including regulatory T cells. The leading class of these molecules target the proteins PDCD1, PDCDL1 or CTLA4 although there are others approved and still many others in various stages of clinical approval across the range of suppressive mechanisms.
[00116] “Disregulation” in tumors with high lymphocytic content is highly correlated with the total number of genetic mutations that tumor harbors which in turn translates to the production of defective and novel (foreign) proteins which serve as antigens that prime the immune response. In a very simplistic sense:
[00117] High tumor burden — > tumor formation — > increased antigenicity — > increased lymphocytic infiltrate — > induction of resistance marked by the production of suppressive cytokines such as IL-10.
[00118] Given its anti-inflammatory effect, colchicine is anticipated to be most effective in the scenario described above including reduction of IL-10 as has been published.
[00119] Alternatively, some of the anti-tumor activity may come from direct effects of colchicine on tumor cells. It is known that colchicine is internalized especially by fast growing cells. Colchicine may inhibit cell division of tumor cells by inhibition of micro tubule function.
[00120] Colchicine in the context of the present invention can be used to treat and/or prevent, or inhibit, delay, or reduce tumor growth or metastases in a subject in need of such inhibition, delay, or reduction, e.g., a lung cancer patient. Other potential cancers that can be treated here include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, leukemia, squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, gastric cancer, pancreatic cancer, neuroendocrine cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, brain cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, esophageal cancer, salivary gland carcinoma, kidney cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, head and neck cancer, and a combination thereof. Especially, in the context of the present invention, colchicine can be used to inhibit, delay, or reduce tumor growth or metastases in a subject having lung cancer.
[00121] In accordance with the present invention, the colchicine formulation herein can be used to promote a positive therapeutic response with respect to tumor growth and/or metastases. By "positive therapeutic response" with respect to cancer treatment is intended an improvement in the disease in association with the anti-tumor activity of these binding molecules, e.g., antibodies or fragments thereof, and/or an improvement in the symptoms associated with the disease. In particular, the methods provided herein are directed to inhibiting, preventing, reducing, alleviating, delaying, or lessening growth of a tumor and/or the development of metastases of primary tumors in a patient. That is the prevention of distal tumor outgrowths, can be observed. Thus, for example, an improvement in the disease may be characterized as a complete response. By "complete response" is intended an absence of clinically detectable metastases with normalization of any previously abnormal radiographic studies, e.g. at the site of the primary tumor or the presence of tumor metastases in bone marrow. Alternatively, an improvement in the disease may be categorized as being a partial response. By "partial response" is intended at least about a 50% decrease in all measurable metastases (i.e., the number of tumor cells present in the subject at a remote site from the primary tumor). Alternatively, an improvement in the disease may be categorized as being relapse free survival or “progression free survival”. By “relapse free survival” is intended the time to recurrence of a tumor at any site. “Progression free survival” is the time before further growth of tumor at a site being monitored can be detected.
[00122] Inhibition, delay, or reduction of metastases can be assessed using screening techniques such as imaging, for example, fluorescent antibody imaging, bone scan imaging, and tumor biopsy sampling including bone marrow aspiration (BMA), or immunohistochemistry. In addition to these positive therapeutic responses, the subject undergoing therapy with a colchicine molecule of the present invention, can experience the beneficial effect of an improvement in the symptoms associated with the disease. [00123] Clinical response can be assessed using screening techniques such as magnetic resonance imaging (MRI) scan, x-radiographic imaging, computed tomographic (CT) scan, flow cytometry or fluorescence-activated cell sorter (FACS) analysis, histology, gross pathology, and blood chemistry, including but not limited to changes detectable by ELISA, RIA, chromatography, and the like.
[00124] In another embodiment, the colchicine formulation as described herein is useful in the prevention of tumor growth and/or metastases. In the context of the present invention, the term “prevention” is well known in the art. For example, a patient/subject suspected of being prone to suffer from a disorder or disease as defined herein may, in particular, benefit from a prevention of the disorder or disease. The subject/patient may have a susceptibility or predisposition for a disorder or disease, including but not limited to hereditary predisposition. Such a predisposition can be determined by standard assays, using, for example, genetic markers or phenotypic indicators. It is to be understood that a disorder or disease to be prevented in accordance with the present invention has not been diagnosed or cannot be diagnosed in the patient/subject (for example, the patient/subject does not show any clinical or pathological symptoms). Thus, the term “prevention” comprises the use of compounds of the present invention before any clinical and/or pathological symptoms are diagnosed or determined or can be diagnosed or determined by the attending physician. Prevention includes, without limitation, to avoid the disease or condition from occurring in patient and/or subject that may be predisposed to the disease but does not yet experience or exhibit symptoms of the disease (prophylactic treatment).
[00125] Colchicine according to the present invention may also be used in combination with at least one other immune modulating therapy, to inhibit, delay, or reduce tumor growth or metastases in a subject in need of such inhibition, delay, or reduction, e.g., a cancer patient.
[00126] In one embodiment, the immune modulating therapy may include cancer vaccines, immuno stimulatory agents, adoptive T cell or antibody therapy, and inhibitors of immune checkpoint blockade (Lizee et al. 2013. Harnessing the Power of the Immune System to Target Cancer. Annu. Rev. Med. Vol. 64 No. 71-90).
[00127] Cancer Vaccines. Cancer vaccines activate the body’s immune system and natural resistance to an abnormal cell, such as cancer, resulting in eradication or control of the disease. Cancer vaccines generally consist of a tumor antigen in an immunogenic formulation that activates tumor antigen- specific helper cells and/or CTLs and B cells. Vaccines can be in a variety of formulations, including, but not limited to, dendritic cells, especially autologous dendritic cells pulsed with tumor cells or tumor antigens, heterologous tumor cells transfected with an immune stimulating agent such as GM-CSF, recombinant vims, or proteins or peptides that are usually administered together with a potent immune adjuvant such as CpG.
[00128] Immunostimulatory Agents. Immuno stimulatory agents act to enhance or increase the immune response to tumors, which is suppressed in many cancer patients through various mechanisms. Immune modulating therapies may target lymphocytes, macrophages, dendritic cells, natural killer cells (NK Cell), or subsets of these cells such as cytotoxic T lymphocytes (CTL) or Natural Killer T (NKT) cells. Because of interacting immune cascades, an effect on one set of immune cells will often be amplified by spreading to other cells, e.g. enhanced antigen presenting cell activity promotes response of T and B lymphocytes. Examples of immunostimulatory agents include, but are not limited to, HER2, cytokines such as G-CSF, GM-CSF and IL-2, cell membrane fractions from bacteria, glycolipids that associate with CD Id to activate Natural Killer T (NKT) cells, CpG oligonucleotides.
[00129] Macrophages, myelophagocytic cells of the immune system, are a fundamental part of the innate defense mechanisms, which can promote specific immunity by inducing T cell recruitment and activation. Despite this, their presence within the tumor microenvironment has been associated with enhanced tumor progression and shown to promote cancer cell growth and spread, angiogenesis and immunosuppression. Key players in the setting of their phenotype are the microenvironmental signals to which macrophages are exposed, which selectively tune their functions within a functional spectrum encompassing the Ml (tumor inhibiting macrophage) and M2 (tumor promoting macrophage) extremes. Sica et al., Seminars in Cancer Biol. ‘:349-355 (2008). Increased macrophage numbers during cancer generally correlates with poor prognosis (Qualls and Murray, Curr. Topics in Develop. Biol. 94: 309-328 (2011)). Of the multiple unique stromal cell types common to solid tumors, tumor-associated macrophages (TAMs) are significant for fostering tumor progression. Targeting molecular pathways regulating TAM polarization holds great promise for anticancer therapy. Ruffell et al, Trends in Immunol. 33:119-126 (2012).
[00130] Adoptive Cell Transfer. Adoptive cell transfer may employ T cell-based cytotoxic responses to attack cancer cells. Autologous T cells that have a natural or genetically engineered reactivity to a patient's cancer are generated and expanded in vitro and then transferred back into the cancer patient. One study demonstrated that adoptive transfer of in vitro expanded autologous tumor-infiltrating lymphocytes was an effective treatment for patients with metastatic melanoma. (Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME (April 2008). "Adoptive cell transfer: a clinical path to effective cancer immunotherapy". Nat. Rev. Cancer 8 (4): 299-308). This can be achieved by taking T cells that are found within resected patient tumor. These T cells are referred to as tumor- infiltrating lymphocytes (TIL) and are presumed to have trafficked to the tumor because of their specificity for tumor antigens. Such T cells can be induced to multiply in vitro using high concentrations of IL-2, anti-CD3 and allo-reactive feeder cells. These T cells are then transferred back into the patient along with exogenous administration of IL-2 to further boost their anti-cancer activity. In other studies, autologous T cells have been transduced with a chimeric antigen receptor that renders them reactive to a targeted tumor antigen (Liddy etal., Nature Med. 18: 980-7, (2012); Grupp et ah, New England J. Med. 368. T509- 18, (2013)).
[00131] Other adoptive cell transfer therapies employ autologous dendritic cells exposed to natural or modified tumor antigens ex vivo that are re-infused into the patient. Provenge is such an PDA approved therapy in which autologous cells are incubated with a fusion protein of prostatic acid phosphatase and GM-CSP to treat patients with prostate tumors. GM-CSP is thought to promote the differentiation and activity of antigen presenting dendritic cells (Small et ah, J. Clin. Oncol. 18: 3894-903(2000); US Patent 7,414,108)).
[00132] Immune Checkpoint Blockade. Immune checkpoint blockade therapies enhance T- cell immunity by removing a negative feedback control that limits ongoing immune responses. These types of therapies target inhibitory pathways in the immune system that are crucial for modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage. Tumors can evolve to exploit certain immune-checkpoint pathways as a major mechanism of immune resistance against T cells that are specific for tumor antigens. Since many immune checkpoints are initiated by ligand-receptor interactions, these checkpoints can be blocked by antibodies to either receptor or ligand or may be modulated by soluble recombinant forms of the ligands or receptors. Neutralization of immune checkpoints allows tumor- specific T cells to continue to function in the otherwise immunosuppressive tumor microenvironment. Examples of immune checkpoint blockade therapies are those which target Cytotoxic T- lymphocyte-associated antigen 4 (CTLA-4), PD-1, its ligand PD-L1, LAG3 and B7-H3.
[00133] Cyclophosphamide. Cyclophosphamide, a commonly used chemotherapeutic agent, can enhance immune responses. Cyclophosphamide differentially suppresses the function of regulatory T cells (Tregs) relative to effector T cells. Tregs are important in regulating anticancer immune responses. Tumor- infiltrating Tregs have previously been associated with poor prognosis. While agents that target Tregs specifically are currently unavailable, cyclophosphamide has emerged as a clinically feasible agent that can preferentially suppress Tregs relative to other T cells and, therefore, allows more effective induction of antitumor immune responses.
[00134] In another embodiment, therapy with a colchicine molecule may be combined with either low dose chemotherapy or radiation therapy. Although standard chemotherapy is often immunosuppressive, low doses of chemotherapeutic agents such as cyclophosphamide, doxorubicin, and paclitaxel have been shown to enhance responses to vaccine therapy for cancer (Machiels et ah, Cancer Res. 57:3689-3697 (2001)). In some cases, chemotherapy may differentially inactivate T regulatory cells (Treg) and myeloid derived suppressor cells (MDSC) that negatively regulate immune responses in the tumor environment. Radiation therapy has been generally employed to exploit the direct tumorcidal effect of ionizing radiation. Indeed, high dose radiation can, like chemotherapy, be immunosuppressive. Numerous observations, however, suggest that under appropriate conditions of dose fractionation and sequencing, radiation therapy can enhance tumor- specific immune responses and the effects of immune modulating agents. One of several mechanisms that contribute to this effect is cross-presentation by dendritic cells and other antigen presenting cells of tumor antigens released by radiation-induced tumor-cell death (Higgins et ah, Cancer Biol. Ther. 8. T 440- 1449 (2009)). In effect, radiation therapy may induce in situ vaccination against a tumor (Ma etal., Seminar Immunol. 22. T 13-124 (2010)) and this could be amplified by combination with therapy with colchicine.
[00135] In one embodiment, the immune modulating therapy may be an immune modulating agent, including, but not limited to, interleukins such as IL-2, IL-7, IL-12; cytokines such as granulocyte-macrophage colony- stimulating factor (GM-CSF), interferons; various chemokines such as CXCL13, CCL26, CXCL7; antagonists of immune checkpoint blockades such as anti-CTLA-4, anti-PD-1, anti-PD-Ll, anti-LAG3 and anti-B7-H3; synthetic cytosine phosphate-guanosine (CpG), oligodeoxynucleotides, glucans, modulators of regulatory T cells (Tregs) such as cyclophosphamide, or other immune modulating agents. In one embodiment, the immune modulating agent is an agonist antibody to 4- IBB (CD 137). As recently reported, such agonist antibody to 4- IBB can give rise to a novel class of KLRG1+ T cells that are highly cytotoxic for tumors (Curran et ah, J. Exp. Med. 270:743-755 (2013)). In all cases, the additional immune modulating therapy is administered prior to, during, or subsequent to the colchicine molecule. Where the combined therapies comprise administration of a colchicine molecule in combination with administration of another immune modulating agent, the methods of the invention encompass co-administration, using separate formulations or a single pharmaceutical formulation, with simultaneous or consecutive administration in either order.
[00136] In one embodiment, the immune modulating therapy may be a cancer therapy agent, including, but not limited to, surgery or surgical procedures (e.g. splenectomy, hepatectomy, lymphadenectomy, leukophoresis, bone marrow transplantation, and the like); radiation therapy; chemotherapy, optionally in combination with autologous bone marrow transplant, or other cancer therapy; where the additional cancer therapy is administered prior to, during, or subsequent to the colchicine therapy. Where the combined therapies comprise administration of colchicine as described herein in combination with administration of another therapeutic agent, the methods of the invention encompass co administration, using separate formulations or a single pharmaceutical formulation, with simultaneous or consecutive administration in either order.
[00137] In one embodiment, treatment includes the application or administration of colchicine as described herein as a single agent or in combination with at least one other immune modulating therapy to a patient, or application or administration of colchicine as a single agent or in combination with at least one other immune modulating therapy to an isolated tissue or cell line from a patient, where the patient has, or has the risk of developing metastases of cancer cells. In another embodiment, treatment is also intended to include the application or administration of a pharmaceutical composition comprising colchicine in combination with at least one other immune modulating therapy or application or administration of a pharmaceutical composition comprising colchicine and at least one other immune modulating therapy to an isolated tissue or cell line from a patient, where the patient has, or has the risk of developing metastases of cancer cells. [00138] Colchicine as single agents or in combination with at least one other immune modulating therapy are useful for the treatment of various malignant and non-malignant tumors. By "anti-tumor activity" is intended a reduction in the rate of IL-Ib production or accumulation associated directly with the tumor or indirectly with stromal cells of the tumor environment, and hence a decline in growth rate of an existing tumor or of a tumor that arises during therapy, and/or destruction of existing neoplastic (tumor) cells or newly formed neoplastic cells, and hence a decrease in the overall size of a tumor and/or the number of metastatic sites during therapy. For example, therapy with colchicine as a single agent or in combination with at least one other immune modulating therapy causes a physiological response, for example, a reduction in metastases, that is beneficial with respect to treatment of disease states associated with IL-Ib -expressing cells in a human.
[00139] In one embodiment, the invention relates to the use of colchicine according to the present invention as a single agent or in combination with at least one other immune modulating therapy as a medicament, in the treatment or prophylaxis of cancer or for use in a precancerous condition or lesion to inhibit, reduce, prevent, delay, or minimalize the growth or metastases of tumor cells.
[00140] In accordance with the methods of the present invention, colchicine as a single agent or in combination with at least one other immune modulating therapy can be used to promote a positive therapeutic response with respect to a malignant human cell. By "positive therapeutic response" with respect to cancer treatment is intended an improvement in the disease in association with the anti-tumor activity of these binding molecules, e.g., antibodies or fragments thereof, and/or an improvement in the symptoms associated with the disease. In particular, the methods provided herein are directed to inhibiting, preventing, reducing, alleviating, delaying, or lessening growth of a tumor and/or the development of metastases of primary tumors in a patient. That is the prevention of distal tumor outgrowths, can be observed. Thus, for example, an improvement in the disease may be characterized as a complete response. By "complete response" is intended an absence of clinically detectable metastases with normalization of any previously abnormal radiographic studies, e.g. at the site of the primary tumor or the presence of tumor metastases in bone marrow. Alternatively, an improvement in the disease may be categorized as being a partial response. By "partial response" is intended at least about a 50% decrease in all measurable metastases (i.e., the number of tumor cells present in the subject at a remote site from the primary tumor). Alternatively, an improvement in the disease may be categorized as being relapse free survival or “progression free survival”. By “relapse free survival” is intended the time to recurrence of a tumor at any site. “Progression free survival” is the time before further growth of tumor at a site being monitored can be detected.
[00141] Inhibition, delay, or reduction of metastases can be assessed using screening techniques such as imaging, for example, fluorescent antibody imaging, bone scan imaging, and tumor biopsy sampling including bone marrow aspiration (BMA), or immunohistochemistry. In addition to these positive therapeutic responses, the subject undergoing therapy with colchicine as described herein can experience the beneficial effect of an improvement in the symptoms associated with the disease.
[00142] Clinical response can be assessed using screening techniques such as magnetic resonance imaging (MRI) scan, x-radiographic imaging, computed tomographic (CT) scan, flow cytometry or fluorescence-activated cell sorter (FACS) analysis, histology, gross pathology, and blood chemistry, including but not limited to changes detectable by ELISA, RIA, chromatography, and the like.
[00143] In an embodiment, the use of colchicine is in fixed (within the same pharmaceutical preparation) or unfixed (different pharmaceutical preparation) combination. “Fixed combination” is to be understood as meaning a combination whose active ingredients are combined at fixed doses in the same vehicle (single formula) that delivers them together to the point of application. Fixed combination can mean, e.g., in a single tablet, solution, cream, capsule, gel, ointment, salve, patch, suppository or transdermal delivery system. “Unfixed combination” as used herein is to be understood as meaning that the active ingredients/components are in more than one vehicle (e.g. tablets, solutions, creams, capsules, gels, ointments, salves, patches, suppositories or transdermal delivery systems). Each of the vehicles can contain a desired pharmaceutical composition or active component.
VI. Pharmaceutical Compositions and Administration Methods
[00144] Methods of preparing and administering the colchicine formulation of the present invention to a subject in need thereof are well known to or are readily determined by those skilled in the art. The route of administration of the colchicine formulation can be, for example, oral, parenteral, by inhalation or topical. The term parenteral as used herein includes, e.g., intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, rectal, or vaginal administration. While all these forms of administration are clearly contemplated as being within the scope of the invention, an example of a form for administration would be a solution for injection, in particular for intravenous or intraarterial injection or drip. A suitable pharmaceutical composition for injection can comprise a buffer (e.g. acetate, phosphate or citrate buffer), a surfactant (e.g. polysorbate), optionally a stabilizer agent (e.g. human albumin), etc. However, in other methods compatible with the teachings herein, colchicine as a single agent or in combination with at least one other immune modulating therapy can be delivered directly to the site of the adverse cellular population thereby increasing the exposure of the diseased tissue to the therapeutic agent.
[00145] Colchicine when used as a composition in the context of the present invention may include one or more pharmaceutically acceptable carriers and thus may be prepared in the form of a local formulation, in order for it to be administered. The pharmaceutically acceptable carrier may include saline, sterile water, linger liquid, buffer saline, a dextrose solution, a malto dextrin solution, glycerol, ethanol and mixtures of one or more thereof, and also may include an additive such as an antioxidant, a buffer, a bacteriostatic agent or the like, as necessary. Furthermore, a diluent, a dispersant, a surfactant, a binder and a lubricant may be added when the composition according to the present invention is prepared, e.g., in the form of a local formulation such as an ointment, lotion, cream, gel, skin emulsion, skin suspension, patch or spray.
[00146] Non-limiting examples for administration of the compound and or compositions according to the present invention include coated and uncoated tablets, soft gelatine capsules, hard gelatine capsules, lozenges, troches, solutions, emulsions, suspensions, syrups, elixiers, powders and granules for reconstitution, dispersible powders and granules, medicated gums, chewing tablets and effervescent tablets. The composition according to the present invention can administered in any pharmaceutical form for oral (e.g. solid, semi solid, liquid), dermal (e.g. dermal patch), sublingual, parenteral (e.g. injection), ophthalmic (e.g. eye drops, gel or ointment) or rectal (e.g. suppository) administration. In an embodiment, the composition is formulated as a tablet, capsule, suppository, dermal patch or sublingual formulation.
[00147] The pharmaceutical compositions used in this invention comprise pharmaceutically acceptable carriers, including, e.g., ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene - polyoxypropylene-block polymers, polyethylene glycol, and wool fat.
[00148] Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include, e.g., water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. In the subject invention, pharmaceutically acceptable carriers include, but are not limited to, 0.01-0.1 M and preferably 0.05 M phosphate buffer or 0.8% saline. Other common parenteral vehicles include sodium phosphate solutions, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, antioxidants, chelating agents, and inert gases and the like.
[00149] More particularly, pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In such cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and will preferably be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Suitable formulations for use in the therapeutic methods disclosed herein are described in Remington's Pharmaceutical Sciences (Mack Publishing Co.) 16th ed. (1980). [00150] Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols, such as mannitol, sorbitol, or sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum mono stearate and gelatin.
[00151] In any case, sterile injectable solutions can be prepared by incorporating an active compound ( e.g ., colchicine according to the present invention) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated herein, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle, which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying, which yields a powder of an active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The preparations for injections are processed, filled into containers such as ampoules, bags, bottles, syringes or vials, and sealed under aseptic conditions according to methods known in the art. Further, the preparations may be packaged and sold in the form of a kit. Such articles of manufacture can have labels or package inserts indicating that the associated compositions are useful for treating a subject suffering from, or predisposed to a disease or disorder.
[00152] Parenteral formulations can be a single bolus dose, an infusion or a loading bolus dose followed with a maintenance dose. These compositions can be administered at specific fixed or variable intervals, e.g., once a day, or on an "as needed" basis.
[00153] Certain pharmaceutical compositions used in this invention can be orally administered in an acceptable dosage form including, e.g., capsules, tablets, aqueous suspensions or solutions. Certain pharmaceutical compositions also can be administered by nasal aerosol or inhalation. Such compositions can be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other conventional solubilizing or dispersing agents. [00154] The amount of the colchicine formulation to be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. The composition can be administered as a single dose, multiple doses or over an established period of time in an infusion. Dosage regimens also can be adjusted to provide the optimum desired response ( e.g ., a therapeutic or prophylactic response).
[00155] In some situations, the composition of the present invention can be parenterally administered. Preparations for parenteral administration include sterile aqueous or non- aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include, e.g., water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. In the subject invention, pharmaceutically acceptable carriers include, but are not limited to, 0.01-0.1 M and preferably 0.05 M phosphate buffer or 0.8% saline. Other common parenteral vehicles include sodium phosphate solutions, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, antioxidants, chelating agents, and inert gases and the like.
[00156] Parenteral formulations can be a single bolus dose, an infusion or a loading bolus dose followed with a maintenance dose. These compositions can be administered at specific fixed or variable intervals, e.g., once a day, or on an "as needed" basis.
[00157] The composition according to the present invention may be administered in a dose range varying depending on the patient's body weight, age, gender, health condition, diet, administration time, administration method, excretion rate and disease severity. The compounds of the present invention as compounds per se in their use as pharmacophores or as pharmaceutical compositions can be administered to the patient and/or subject at a suitable dose. The dosage regiment will be determined by the attending physician and clinical factors. As is well known in the medical arts, dosages for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. Generally, the regimen as a regular administration of the pharmaceutical composition comprising the herein defined should be, e.g., in a range as described below. Progress can be monitored by periodic assessment.
[00158] The composition according to the present invention can be administered with a single dose or with 2, 3, 4, 5, 6, 7, 8, 9, or 10 doses, if desired. The composition can be administered 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 times per day. Preferably, colchicine according to the present invention is administered once per day. More preferably, colchicine according to the present invention is administered once per day as a single dose.
[00159] The composition according to the present invention can be administered regularly for long periods of time. In an embodiment, the composition can be administered regularly for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more years. In another embodiment, the composition can be administered regularly for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more months. In other embodiments, the composition can be administered regularly for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more weeks. As used herein, the term “regularly” refers to administration of the composition at regular times or intervals over a period of time. For instance, the composition may be administered to a patient once daily for three (3) years. In other embodiments, the composition may be administered to a patient once every other day for five (5) years. It should be appreciated that the frequency of administration may vary based on a number of factors, including, but not limited to, the severity of disease, the overall health of the patient, any additional medications the patient is taking, and whether the treatment is prophylactic or not. It should also be appreciated that the frequency of administration may be adjusted at any point.
[00160] The amount/concentration/dose of the composition according to the present invention can be between O.lmg and 5.0mg, O.lmg and 2.0mg, O.lmg to 1.5mg, O.lmg to l.Omg, O.lmg to 0.75mg, O.lmg to 0.5mg, 0.25mg to 5.0mg, 0.25mg to 2.0mg, 0.25mg to 1.5mg, 0.25mg to l.Omg, 0.25mg to 0.75mg or 0.25mg to 0.5mg. In an embodiment, the composition according to the present invention is administered at a daily dose of colchicine of between about O.lmg and about 0.75mg or between about O.lmg and about 0.5mg. In another embodiment, the composition according to the present invention is administered at a daily dose of colchicine of between about 0.25mg to about 0.75mg or between about 0.25mg to about 0.5mg. In an embodiment, the composition according to the present invention is administered at a daily dose of about 0.5mg colchicine. [00161] In a preferred embodiment, the amount/concentration of colchicine as used herein can be administered at the first day of administration in a higher dose (concentration/amount) compared to the administration of colchicine at the following days(s) of administration (maintenance administration/maintenance dose of administration). Alternatively such decreased dose (maintenance dose) can be started after 2, 3, 4, 5, 6, 7, 8, 9 or 10 days of initial administration of the higher dose. In case the course of treatment is any such as described above, the higher dose/amount/concentration of colchicine (e.g. at the first day of administration) can be any as described above, provided that the maintenance dose (the dose/amount/concentration of colchicine at the days following the higher dose/amount/concentration) is lower than the initial dose/amount/concentration of colchicine (e.g. at the first day of administration). Preferably, the composition of the invention is administered with a dose of colchicine of between about 1.0 mg to about 2.0 mg at the first day (preferably as a single dose) of administration and the maintenance dose of colchicine at the following day(s) of administration is between about 0.5 mg to about 1.0 mg.
[00162] In keeping with the scope of the present disclosure, the colchicine formulation of the present invention can be administered to a human or other animal in accordance with the aforementioned methods of treatment in an amount sufficient to produce a therapeutic effect. The colchicine formulation can be administered to such human or other animal in a conventional dosage form prepared by combining the colchicine formulation of the invention with a conventional pharmaceutically acceptable carrier or diluent according to known techniques. It will be recognized by one of skill in the art that the form and character of the pharmaceutically acceptable carrier or diluent is dictated by the amount of active ingredient with which it is to be combined, the route of administration and other well- known variables.
[00163] By "therapeutically effective dose or amount" or "effective amount" is intended an amount of the colchicine formulation that when administered brings about a positive therapeutic response with respect to treatment of a patient with a disease to be treated, e.g., an improvement in the disease can be evidenced by, for example, a delayed onset of clinical symptoms of the disease or condition, a reduction in severity of some or all clinical symptoms of the disease or condition, a slower progression of the disease or condition, an improvement in the overall health or well-being of the subject, or by other parameters well known in the art that are specific to the particular disease.
[00164] Therapeutically effective doses of the compositions of the present invention, for the inhibition, delay, or reduction of metastases, vary depending upon many different factors, including means of administration, target site, physiological state of the patient, whether the patient is human or an animal, other medications administered, and whether treatment is prophylactic or therapeutic. In certain embodiments the patient is a human, but non human mammals including transgenic mammals can also be treated. Treatment dosages may be titrated using routine methods known to those of skill in the art to optimize safety and efficacy.
[00165] The amount of colchicine administered as a single agent or in combination with at least one other immune modulating therapy is readily determined by one of ordinary skill in the art without undue experimentation given the disclosure of the present invention. Factors influencing the mode of administration and the respective amount of colchicine to be administered as a single agent or in combination with at least one other immune modulating therapy include, but are not limited to, the severity of the disease, the history of the disease, the potential for metastases, and the age, height, weight, health, and physical condition of the individual undergoing therapy. Similarly, the amount of colchicine as a single agent or in combination with at least one other immune modulating therapy to be administered will be dependent upon the mode of administration and whether the subject will undergo a single dose or multiple doses of this agent.
[00166] The invention also provides for the use of the colchicine formulation in the manufacture of a medicament for treating a subject with a cancer, wherein the medicament is used in a subject that has been pretreated or is concurrently being treated with at least one other therapy. By "pretreated" or "pretreatment" is intended the subject has received one or more other therapies prior to receiving the medicament comprising the colchicine formulation. "Pretreated" or "pretreatment" includes subjects that have been treated with at least one other therapy within 2 years, within 18 months, within 1 year, within 6 months, within 2 months, within 6 weeks, within 1 month, within 4 weeks, within 3 weeks, within 2 weeks, within 1 week, within 6 days, within 5 days, within 4 days, within 3 days, within 2 days, or even within 1 day prior to initiation of treatment with the medicament comprising the colchicine formulation. By "concurrent" or "concomitant" is intended the subject is receiving one or more other therapies while at the same time receiving the medicament comprising the colchicine formulation. It is not necessary that the subject was a responder to pretreatment with the prior therapy or therapies or a responder to the concurrent therapy or therapies. Thus, the subject that receives the medicament comprising the colchicine formulation could have responded, or could have failed to respond, to pretreatment with the prior therapy, or to one or more of the prior therapies where pretreatment comprised multiple therapies. Examples of other cancer therapies for which a subject can have received pretreatment prior to receiving the medicament comprising the colchicine as a single agent or in combination with at least one other immune modulating therapy include, but are not limited to, surgery; radiation therapy; chemotherapy, optionally in combination with autologous bone marrow transplant, where suitable chemotherapeutic agents include, but are not limited to, those listed herein above; other anti-cancer monoclonal antibody therapy; small molecule-based cancer therapy, including, but not limited to, the small molecules listed herein above; vaccine/immunotherapy-based cancer therapies; steroid therapy; other cancer therapy; or any combination thereof.
[00167] All of the references cited above, as well as all references cited herein, are incorporated herein by reference in their entireties.
[00168] While the invention has been illustrated and described in detail in above, such illustration and description are to be considered illustrative or exemplary and not restrictive. It will be understood that changes and modifications may be made by those of ordinary skill within the scope and spirit of the following claims. In particular, the present invention covers further embodiments with any combination of features from different embodiments described above and below.
[00169] The present invention is additionally described by way of the following illustrative non-limiting examples that provide a better understanding of the present invention and of its many advantages. The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques used in the present invention to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
EXAMPLES
Example 1: Colchicine Sustained Release Tablet [00170] This example illustrates a colchicine sustained release tablet. The tablet uses the ingredients and concentrations shown in Table 1 below.
Figure imgf000050_0001
[00171] The concentrations may be altered to change certain properties of the formulations, for instance, the dissolution profile. Table 2 shows the ranges for each ingredient.
Figure imgf000050_0002
Figure imgf000051_0001
Example 2: Method of Making a Colchicine Sustained Release Tablet
[00172] The above ingredients are utilized to make a tablet to the following working directions:
[00173] Granulation: The granulation was performed in a Kenwood mixer. The colchicine and hypromellose 6Mpa*s were first weighed and separately dissolved into purified water. This solution of hypromellose 6Mpa*s was filled in the mixer containing the lactose within 1.5 min followed by a 3 minute mixing time. Subsequently, the dissolved colchicine was sprayed with a filing agent (e.g., lactose monohydrate) under continuous mixing over a period of 15 minutes. These steps were performed at room temperature. The wet granulate was then passed through a 1.0 mm hand sieve. It was then dried in an oven (Haeraeus ) at 45 0 C for 26 h to a residual moisture content of 0.53 %, and passed through a 0.8 mm sieve shaker (Erweka AR 400). Density parameters were tested (Engelmann). Bulk density 0.53 g / ml, compacted bulk density 0.67 g / ml, Hausner ratio 1.26. Rheology : Flow time 4sec ; Slope angle 23.8 °.
[00174] Blending: Following the granulation process, the granulate is compounded with a filling agent (e.g., lactose monohydrate), a retarding agent (e.g., Retalac), and other excipients (e.g., flow enhancer, glidants and/or lubricants) to support the tablet compression process. To this end, these ingredients were placed manually through a 0.8 mm sieve and mixed with the granulate in a cube mixer (Erweka) for 10 minutes. In one embodiment, the glidant used may be Talc. In another embodiment, the lubricants used may be Stearic acid. The granulate will then be blended using a suitable mixer.
[00175] Compression of Tablets: To form the tablets, a compression force is needed. The mechanical force will define the physical properties of the tablets, especially the crushing strength of the resulting tablet. The mechanical strength interacts with the initial swelling of the tablet and dilution speed of the tablet core. This effect is well known in the art and can be adjusted and controlled during the lifecycle of the product.
[00176] Tableting was performed on a Korsch (EK 0) tablet press with a round tabletting tool, biconvex, 6 mm in diameter. Average tablet hardness was approximately 100N +/- 15 N. Tablets measured about lOOmg in mass, friability was not measurable. Breaking strength and hardness were measured with a Erweka Multickeck. Friability was measured with a Erweka Friabilator and a Mettler analytical balance. The dimensions were measured using a Mitutoyo caliper.
Example 3: Measurements of Dissolution Profiles of Sustained-Release Colchicine Formulations
[00177] The dissolution of the sustained release formulation of colchicine was measured at various time points. The compositions were dissolved in 500ml of water at 37°C and stirred continuously over a period of 6 hours. Samples were drawn at several time points to study the kinetics of the dissolution process of the drug substance within the hydrophilic matrix system. Colchicine content in the samples was analyzed using HPFC analysis.
[00178] Several batches were tested to determine the optimal dissolution profile for the sustained release formulation. The release can be modified by both the concentration of hypromellose or by using different viscosity grades of hypromellose (e.g. lOOOmPa or lOOOOmPa). In the batches tested below, the viscosity grade remained constant, however, the concentration of hypromellose 4000mPa in the tablet was modified.
[00179] Table 3 below summarizes the various compositions that were tested.
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
[00180] The dissolution profiles of the various batches are provided in Tables 4-6 and FIGS. 1-4. In particular, the dissolution profile for Batches 1-3 is summarized in Table 4 and FIG. 1. The profile for Batch 1 shows approximately a 92% release within about 30 minutes, followed by a constant release. Complete dissolution occurred after 2 hours. Batch 2 shows approximately a 83% release within about 30 minutes, followed by a constant release. Complete dissolution occurred after 2 hours. Batch 3 shows approximately a 74% release within about 30 minutes, followed by a constant release. Complete dissolution occurred after 2 hours.
Figure imgf000055_0001
[00181] The dissolution profile for Batch 4 is summarized in Table 5 and FIG. 2. Batch 4A shows approximately a 30% release within about 30 minutes, followed by a constant release. Complete dissolution occurred after 6 hours. Batch 4B shows approximately a 23% release within about 30 minutes, followed by a constant release. Complete dissolution occurred within 6 hours. In this example, a difference in tablet hardness results in a difference in release rates. FIG. 3 shows the dissolution profiles for Batches 1-4.
Figure imgf000056_0001
[00182] The dissolution profile for Batch 5 is summarized in Table 6 and FIG. 4. Batch 5 represents the immediate release version of the composition. The profile for Batch 5 shows a 90% release within 5 minutes. Complete dissolution occurred within 2 hours.
Figure imgf000056_0002
[00183] Table 7 below summarizes another batch, Batch 6, that was tested.
Figure imgf000056_0003
Figure imgf000057_0001
[00184] The dissolution profile for Batch 6 is summarized in Table 7 and FIG. 5A. Batch 6 represents a sustained release version of the composition. The profile for Batch 6 in FIG. 5A shows about a 45% release within 30 minutes, about a 65% release in 60 minutes, and about a 80% release in 90 minutes. Complete dissolution occurs within about 2 hours.
[00185] Batch 4 was further modified by altering the concentration of retarding agent (i.e. Retalac) in the composition (Retalac 0%, 23.3%, 26,6% or 30%). The dissolution profiles of the four compositions are shown in FIG. 6. As shown in FIG. 6, the composition with 23.3% Retalac shows about a 65% release within 30 min, about a 80% release in 60 min, and about a 90% release in 90 min. Complete dissolution occurs within about 120 min. As further shown in FIG. 6, the composition with 26.6% Retalac shows about a 55% release within 30 min, about a 75% release in 60 min, and about a 85% release in 90 min. Complete dissolution occurs within about 180 min. As further shown in FIG. 6, the composition with 30% Retalac shows about a 40% release within 30 min, about a 55% release in 60 min, and about a 70% release in 90 min. Complete dissolution occurs in more than about 180 min. As further shown in FIG. 6, the composition with 0% Retalac (i.e., immediate release composition) shows complete dissolution within about 15 min.
[00186] As mentioned previously, the release profile of the sustained release composition can be changed to a specific or desired target release by adjusting the amount of retarding agent (i.e., Retalac) as well as tablet hardness. The release depends upon a variety of factors, including erosion of the outer layer of colchicine (i.e., the immediate release portion) as well as diffusion of the inner layer of colchicine (i.e., the sustained release portion). Since the percentage of colchicine is low in the sustained release formulation and the tablets are small, this balance between erosion and diffusion is very sensitive and has to be fine-tuned to reach a very specific dissolution profile.
Example 4: Pharmacokinetic Properties of Sustained-Release Colchicine Formulations [00187] FIG. 7A, FIG. 7B, FIG. 7C, FIG. D, and FIG. 8 show plasma colchicine levels (ng/mL) as a function of time (hrs) for colchicine formulations according to FIG. 6 [00188] The therapeutic effect of the sustained release formulation containing of colchicine is evaluated in a clinical study that is a multidose, randomized, cross-over study, which will evaluate bioavailability of about 3 different 0.50 mg sustained release formulations (e.g. Test Products 1,2,3 and FIG. 6) of colchicine to a 0.5 mg immediate release formulation (control product), administered to healthy volunteers. Test products 1, 2, and 3 used varying levels of retarding agent (i.e., Retalac), 23.3%, 26.6% or 30%, respectively, thereby having different sustained release profiles. The primary aim of the study is to assess pharmacokinetics (PK) of test and control drug in blood as well as in neutrophils or leucocytes.
[00189] Research objective: The primary objective of this study is to compare the pharmacokinetics of the test product vs. control product of colchicine in healthy human volunteers. In particular, levels of colchicine in blood (herein referred as to blood PK), neutrophils or leucocytes (herein referred as to neutrophil PK) will be assessed upon treatment with control and test product. [00190] The hypothesis tested is that administration of an equal amount of colchicine in form of a sustained release tablet leads to lower peak levels (Cmax), while maintaining equal absolute bioavailability (area under the curve, AUC). Colchicine concentrations in neutrophils are measured as it is generally recognized that neutrophils which reconstitute 60-70% of leukocytes, play a central role in inflammatory responses in general and are thought to be major players in several diseases where colchicine is used as treatment. Therefore for the purpose of this experiment, either leucocytes or neutrophils may be analyzed. Hence, the term neutrophils as used herein also refers to, if used, leucocytes. As colchicine is known to preferentially accumulate in neutrophils and inhibit many of their pro-inflammatory functions, they are thought to be a major target of colchicine therapy. Therefore it is of special interest to know whether similar concentrations of colchicine are reached in isolated neutrophils or in leukocytes which would give information on potential bioequivalence. Therefore, blood will be drawn in various time points over the course of the study to check colchicine concentrations in blood and neutrophils.
[00191] General Study Design: This study is designed as a randomized, cross-over study. There will be 3 groups of patients (n= 3x at least 8). Group 1 will receive control or test drug for about 8-14 days. After a wash out period, they will receive test drug 1 for about 8- 14 days days. Group 2 will receive the control drug for the same time. After a wash out period, they will receive test drug 2 for the same time. Group 3 will receive the control drug for the same time. After a wash out period, they will receive test drug 3 for the same time. The study participants are treated on an in-patient basis for the first 24h and on an out patient basis for the remaining time. A sufficiently long wash-out period lies between the two trials.
[00192] Subject participation: There will be 3 groups of patients (n= 3x at least 8). For each test drug, healthy volunteers will be randomized to one drug for the first round. After a sufficient wash out period, the same subjects will receive the control drug.
[00193] Study duration: The study drug is administered at a single dose/day for about 8- 14 days consecutive days. The study consists of 24h blood PK (high frequency data collection). Neutrophil PK is analysed in intervals that allow for conclusive determination of colchicine levels in the latter over the duration of the experiment.
[00194] Treatment regimen: Below is an example of what the treatment regime may look like.
Figure imgf000060_0001
[00195] Research techniques and data analysis: Analytical chemistry techniques (HPLC) or immunological techniques (radioimmunoassay) may be utilized for the assessment of colchicine and optionally, its metabolites. Collected data is analyzed by adequate data management and statistics software.
[00196] Subject Population: The study population consists of male healthy volunteers (n=5- 10/group). For inclusion in the study, the patient must be 25 - 40 years of age; not less than 60kg and not more than 120 kg body weight; healthy; no major competing comorbidities or contraindication to colchicine therapy; willing to provide consent and be randomized into the study. Patients who meet the following criteria will be excluded: ongoing therapy with other anti-inflammatory/immunosuppressive drugs; treatment with drugs with known drug interactions with colchicine; renal/hepatic impairment; known hypersensitivity to colchicine.
[00197] Treatment schema: The drug is given once a day in the morning.
[00198] Data collection schema: For blood PK, blood will be collected over a period of
24h at the beginning the study in short intervals (sufficient time points to establish pharmacokinetic data, e.g.: -1, +0.5h, lh, 1.5h, 2h, 3h, 4h, 5h, 7h, lOh, 12h, 15h and 24 h (before next application). Additional time points may be 24h, and 10 days after drug withdrawal at the end of the study to examine the wash out phase of the drug. PK analysis includes colchicine blood concentrations. Adequate techniques for the isolation of blood and quantification of colchicine are applied.
[00199] For neutrophil PK, neutrophils will be isolated or purified from raw blood from several time points. Neutrophils will be collected at the beginning the study as well as at least once at the end of the study. PK analysis includes colchicine concentrations in isolated neutrophils. [00200] Results: The results are shown in FIGS. 7-9. FIGS. 7A-7D show plasma colchicine levels (ng/mL) as a function of time (hrs) in the immediate release formulation (FIG. 7A) as compared to the sustained release formulations (FIGS. 7B, 7C and 7D). The immediate release formulation of FIG. 7A has a Cmax of 1.77 +/- 0.8 ng/mL, an AUC (o-t) of 8.08 +/- 2.9 ng/mL and a Tmax of 0.83 +/- 0.2 hr. The sustained release formulation of FIG. 7B has a Cmax of 1.56 +/- 0.4 ng/mL, an AUC (o-t) of 7.94 +/- 0.9 ng/mL and a Tmax of 1.47 +/- 0.2 hr. The sustained release formulation of FIG. 7C has a Cmax of 1.56 +/- 0.4 ng/mL, an AUC (o-t) of 7.94 +/- 0.9 ng/mL and a Tmax of 1.47 +/- 0.2 hr. The sustained release formulation of FIG. 7D has a Cmax of 0.91 +/- 0.3 ng/mL, an AUC (o-t) of 5.04 +/- 2.3 ng/mL and a Tmax of 1.89 +/- 0.4 hr. FIG. 8 shows the overlapping profiles of the immediate release formulation and the three sustained release profiles. As can be seen, there is less inter-subject variation in plasma levels, lower Cmax and similar AUC for the sustained release formulation of FIG. 7B when compared to the immediate release formulation. Since the colchicine concentration on plasma increases and decreases more steadily and in a more controlled way, and the variability of colchicine levels in plasma from patient to patient is less, the safety and efficacy profile is more predicable. As a result, there will likely be fewer adverse side effects (e.g., gastrointestinal side effects) and/or reduced risk of toxicity (e.g., due to drug-drug interactions or co-morbidities such as renal and/or hepatic impairment).
[00201] FIG. 9 shows the levels of colchicine in neutrophils on Day 1 and Day 10 in the immediate release formulation and the three sustained release formulations. As can be seen, the levels of colchicine increase from Day 1 to Day 10, suggesting that colchicine accumulates in neutrophils over time. The results further show that the levels in neutrophils are similar after 10 days regardless of the formulation, suggesting that the therapeutic effect will be equal for the tested sustained release and immediate release formulations (as the site of action of colchicine is neutrophils).
Example 5: Therapeutic effects of colchicine in patients with cancer [00202] To assess the therapeutic effects of a sustained release formulation in patients with or in patients with an increased risk of cancer, a prospective randomized observer blinded end-point trial will be conducted to determine whether adding 0.5 mg/day of colchicine to standard anti-cancer therapies inhibits, delays, or reduces tumor growth or metastases in a cancer patient.
[00203] A sufficient amount of patients with cancer or in risk of cancer are to be randomized to long-term low dose sustained release colchicine or to standard therapy alone. Cancer incidence, morbidity and mortality will be analyzed in regular intervals. Patients in the colchicine arm are expected to have a significantly lower cancer incidence and mortality than patients not treated with colchicine.
Example 6: Anti-tumor effects of colchicine in animals [00204] A sufficient amount of specimen of a suitable animal species will be included in this study utilizing commonly used tumor models. A therapeutically effective amount of colchicine is administered to the test animals. Placebo will be administered to a control group. Tumor growth will be induced in all animals. Animals treated with colchicine are expected to have significantly less tumor growth, or tumor growth is prevented as compared to the control group.
Example 7: Anti- tumor effects of colchicine in vitro [00205] A suitable in vitro model for studying the anti-tumor effects of drugs is utilized for this test. Such a test may include commonly used tumor cell lines with or without combination with cultivated PBMCs. Colchicine is expected to inhibit or reduce growth of tumor cell lines upon incubation with therapeutically effective amount of colchicine and/or cultivation with PBMCs pretreated with a therapeutically effective amount of colchicine.
Example 8: Effects of colchicine on lung cancer [00206] The investigators propose to conduct a randomized, double-blind, placebo- controlled, multi-center trial of oral daily colchicine <dose discussion pending, may need 2 doses probably 0.5 and 1.0 mg daily >> to formally test whether chronic interleukin- 1b inhibition will slow the invasion, progression, and metastases associated with early stage lung cancers and thus significantly improve cancer survival. To dramatically reduce costs, the trial will be conducted using multiple internet based and social media techniques previously employed to great success without sacrificing quality of adherence, compliance, endpoint adjudication, or rates of long-term follow-up. [00207] Patients will be eligible for the proposed trial if they have been recently diagnosed with non-metastatic stage IA2, IA3, and IB lung cancer and have just completed initial resection. Such patients are typically followed by the thoracic surgery community in a setting where no post-resection therapy has proven effective and where routine care includes close observation and surveillance for recurrent disease through serial CT scans over a period of 3 to 5 years. As such, the trial has been designed to minimally alter usual care practice and should thus be easy to conduct in the many dedicated thoracic surgical centers who have made a fundamental commitment to this effort.
[00208] All eligible patients in the proposed trial will receive either colchicine or placebo at randomization and on a daily basis thereafter for the trial duration << need to discuss if we want an open label run in for compliance first>>. Randomization and study drug initiation will occur 2 to 3 weeks post-operatively at the time of the first routine clinical follow-up visit.
[00209] For the first two years of the trial, following usual care patterns in North American, all participants will return to their respective thoracic surgical or oncology center every three months at which time CT scans will be obtained, routine safety blood work will be done, a simple internet based questionnaire will be completed for adverse effects and clinical events, and further study drug provided. In years 3 to 5, identical procedures will be followed with continued 3 month clinic visits to ascertain safety, evaluate endpoints, and provide study drug; however, and again in accordance with usual care, CT scans will be performed in years 3 through 5 every 6 months rather than every 3 months. Thus, all aspects of the trial conduct and follow-up are designed to seamlessly interface with routine clinical care, further reducing trial costs.
[00210] The proposed trial primary endpoint will be recurrence of cancer or cancer-related death at 5 years (conservatively estimated to be 35-40 percent) while the proposed trial secondary endpoint will consist of cancer-related death alone (conservatively estimated to be 30 to 35 percent). In the CANTOS atherosclerosis trial, the 300mg dose of canakinumab reduced incident lung cancer by 67 % and fatal lung cancer by 77% (at a median time period of only 3 years). To remain conservative, the proposed trial assumes a treatment effect of only half that size (35% relative risk reduction); as such, power exceeds 93% for the primary endpoint and 86% for the secondary endpoint with total sample sizes ranging from 1000 to 1200 participants. For efficiency, randomization will be stratified on age and smoking status, with pre-specified subgroup analyses conducted on the basis of tumor genetic profiles and other typical clinical parameters.
[00211] Many modifications and other embodiments of the inventions set forth herein will come to mind to one skilled in the art to which these inventions pertain having the benefit of the teachings presented in the foregoing descriptions and the associated drawings. Therefore, it is to be understood that the inventions are not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims and list of embodiments disclosed herein. Although specific terms are employed herein, they are used in a generic and descriptive sense only and not for purposes of limitation.

Claims

WHAT IS CLAIMED IS:
1. A method for inhibiting, delaying, or reducing the occurrence of lung cancer in a subject with cancer, comprising administering to the subject an effective amount of colchicine.
2. The method of claim 1, wherein colchicine induces anti-tumor responses.
3. The method of claim 1, wherein colchicine inhibits IL-Ib mediated signaling.
4. The method of claim 1, wherein colchicine inhibits inflammation activation.
5. The method of claim 1, wherein colchicine inhibits release of TNF-a and IL-10.
6. The method of claim 1, wherein colchicine is included in an amount between about 0.25mg and about l.Omg.
7. The method of claim 1, wherein colchicine is included in an amount between about
0.5mg.
8. A method for treating lung cancer in a subject, comprising administering to the subject an effective amount of colchicine.
9. The method of claim 8, wherein colchicine induces anti-tumor responses.
10. The method of claim 8, wherein colchicine inhibits IL-Ib mediated signaling.
11. The method of claim 8, wherein colchicine inhibits release of TNF-a and IF- 10.
12. The method of claim 8, wherein colchicine inhibits inflammation activation.
13. The method of claim 8, wherein colchicine is included in an amount between about 0.25mg and aboutl.Omg.
14. The method of claim 8, wherein colchicine is included in an amount between about
0.5mg.
15. A method for treating lung cancer in a subject, comprising administering to the subject an effective amount of colchicine and an effective amount of at least one other immune modulating therapy.
16. The method of claim 15, wherein the immune modulating agent is selected from the group consisting of interleukins, cytokines, chemokines, antagonists of immune checkpoint blockades and a combination thereof.
17. The method of claim 15, wherein the immune modulating therapy may be a cancer therapy.
18. The method of claim 17, wherein the cancer therapy is selected from the group consisting of surgery or surgical procedures, radiation therapy, chemotherapy or a combination thereof.
19. The method of any one of claims 15 to 18, wherein colchicine and the immune modulating agent or immune modulating therapy are administered separately or concurrently.
20. The method of any one of claims 15 to 18, wherein colchicine is included in an amount of between about 0.4mg to about 0.75mg.
21. The method of claim 20, wherein colchicine is included in an amount between about
0.5mg.
PCT/EP2020/081164 2019-11-06 2020-11-05 Use of colchicine in the treatment and prevention of lung cancer WO2021089715A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962931684P 2019-11-06 2019-11-06
US62/931,684 2019-11-06

Publications (1)

Publication Number Publication Date
WO2021089715A1 true WO2021089715A1 (en) 2021-05-14

Family

ID=73138841

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2020/081164 WO2021089715A1 (en) 2019-11-06 2020-11-05 Use of colchicine in the treatment and prevention of lung cancer

Country Status (1)

Country Link
WO (1) WO2021089715A1 (en)

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3173876A (en) 1960-05-27 1965-03-16 John C Zobrist Cleaning methods and compositions
US3276586A (en) 1963-08-30 1966-10-04 Rosaen Filter Co Indicating means for fluid filters
US3541005A (en) 1969-02-05 1970-11-17 Amicon Corp Continuous ultrafiltration of macromolecular solutions
US3541006A (en) 1968-07-03 1970-11-17 Amicon Corp Ultrafiltration process
US3546142A (en) 1967-01-19 1970-12-08 Amicon Corp Polyelectrolyte structures
US4285987A (en) 1978-10-23 1981-08-25 Alza Corporation Process for manufacturing device with dispersion zone
US7414108B2 (en) 1997-04-11 2008-08-19 Dendreon Corporation Composition and method for producing an immune response against tumor-related antigens
WO2011022805A1 (en) * 2009-08-26 2011-03-03 Alberta Health Services Novel colchicine derivatives, methods and uses thereof
WO2019055509A1 (en) * 2017-09-12 2019-03-21 Ardea Biosciences, Inc. Cxcr-2 inhibitors for treating disorders
WO2019149884A1 (en) * 2018-02-02 2019-08-08 Murray & Poole Enterprises, Ltd Use of colchicine to inhibit tumor growth and metastases

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3173876A (en) 1960-05-27 1965-03-16 John C Zobrist Cleaning methods and compositions
US3276586A (en) 1963-08-30 1966-10-04 Rosaen Filter Co Indicating means for fluid filters
US3546142A (en) 1967-01-19 1970-12-08 Amicon Corp Polyelectrolyte structures
US3541006A (en) 1968-07-03 1970-11-17 Amicon Corp Ultrafiltration process
US3541005A (en) 1969-02-05 1970-11-17 Amicon Corp Continuous ultrafiltration of macromolecular solutions
US4285987A (en) 1978-10-23 1981-08-25 Alza Corporation Process for manufacturing device with dispersion zone
US7414108B2 (en) 1997-04-11 2008-08-19 Dendreon Corporation Composition and method for producing an immune response against tumor-related antigens
WO2011022805A1 (en) * 2009-08-26 2011-03-03 Alberta Health Services Novel colchicine derivatives, methods and uses thereof
WO2019055509A1 (en) * 2017-09-12 2019-03-21 Ardea Biosciences, Inc. Cxcr-2 inhibitors for treating disorders
WO2019149884A1 (en) * 2018-02-02 2019-08-08 Murray & Poole Enterprises, Ltd Use of colchicine to inhibit tumor growth and metastases

Non-Patent Citations (52)

* Cited by examiner, † Cited by third party
Title
"Physician's Desk Reference", 2014
"Remington's Pharmaceutical Sciences", 1980, MACK PUBLISHING CO
"Treatise on Controlled Drug Delivery, Fundamentals, Optimization, Applications", 1992, MARCEL DEKKER, INC.
APTE, CANCER METASTASIS REV., vol. 25, 2006, pages 387 - 408
BALKWILL, SEMIN. CANCER BIOL., vol. 22, 2012, pages 33 - 40
BHATTACHARYA SURELA ET AL: "Colchicine induces autophagy and senescence in lung cancer cells at clinically admissible concentration: potential use of colchicine in combination with autophagy inhibitor in cancer therapy", TUMOR BIOLOGY, KARGER, BASEL, CH, vol. 37, no. 8, 11 February 2016 (2016-02-11), pages 10653 - 10664, XP036219264, ISSN: 1010-4283, [retrieved on 20160211], DOI: 10.1007/S13277-016-4972-7 *
CAS, no. 64-86-8
CHIRIVI, CANCER RES., vol. 53, 1993, pages 5051 - 5054
COCCOCHU ET AL., EUR J INTERN MED, vol. 21, no. 6, 2010, pages 503 - 8
COUSSENS, NATURE, vol. 420, 2002, pages 860 - 7
CURRAN ET AL., J. EXP. MED., vol. 210, 2013, pages 743 - 755
DOSTERT, SCIENCE, vol. 320, 2008, pages 674 - 7
FINKELSTEIN ET AL., HERZ, vol. 27, 2002, pages 791 - 194
GASSE, J. CLIN. INV., vol. 117, 2007, pages 3786 - 99
GIAVAZZI, CANCER RES., vol. 50, 1990, pages 4771 - 4775
GRAHAMROBERTS, ANN RHEUM DIS, vol. 12, no. 1, 1953, pages 16 - 9
GRIVENIKOV, CELL, vol. 140, 2010, pages 883 - 899
GRUPP ET AL., NEW ENGLAND J. MED., vol. 368, 2013, pages 1509 - 18
HIGGINS ET AL., CANCER BIOL. THER., vol. 8, 2009, pages 1440 - 1449
IMAZIO ET AL., AM. HEART J., vol. 162, no. 3, 2011, pages 527 - 532
IMAZIO ET AL., ANN. INTERN. MED., vol. 155, no. 7, 2011, pages 409 - 14
IMAZIO ET AL., CIRCULATION, vol. 112, no. 13, 2005, pages 2012 - 2016
IMAZIO ET AL., EUROPEAN HEART JOURNAL, vol. 31, 2010, pages 2749 - 2754
IMAZIO, ARCH INTERNMED, vol. 165, no. 17, 2005, pages 1987 - 91
IMAZIOBRUCATO ET AL., EUR HEART J, vol. 30, no. 5, 2009, pages 532 - 9
KIM, ANTICANCER RES., 2017
KUO, MEDICINE, 2015
LEE, CRIT. REV. ONCOL. HEMATOL., vol. 66, 2008, pages 208 - 17
LIDDY ET AL., NATURE MED, vol. 18, 2012, pages 980 - 7
LIN ZU-YAU ET AL: "Anticancer effects of clinically acceptable colchicine concentrations on human gastric cancer cell lines", KAOHSIUNG JOURNAL OF MEDICAL SCIENCES, KAOHSIUNG MEDICAL COLLEGE, KAOHSIUNG, TW, vol. 32, no. 2, 2 February 2016 (2016-02-02), pages 68 - 73, XP029450790, ISSN: 1607-551X, DOI: 10.1016/J.KJMS.2015.12.006 *
LIZEE ET AL.: "Harnessing the Power of the Immune System to Target Cancer", ANNU. REV. MED., vol. 64, no. 71-90, 2013
MA ET AL., SEMINAR IMMUNOL, vol. 22, 2010, pages 113 - 124
MACHIELS ET AL., CANCER RES., vol. 61, 2001, pages 3689 - 3697
MAISCH ET AL.: "Guidelines on the Diagnosis and Management of Pericardial Diseases", EUR HEART J., vol. 25, 2004, pages 916 - 928
MARTINEZ, ATHEROSCLEROSIS, 2018
MARTINON, NATURE, 2006
MARZO-MAS ANA ET AL: "Interactions of long-chain homologues of colchicine with tubulin", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 126, 23 November 2016 (2016-11-23), pages 526 - 535, XP029885654, ISSN: 0223-5234, DOI: 10.1016/J.EJMECH.2016.11.049 *
MEURINTABET, ARCH. CARDIOVASC. DIS., vol. 104, no. 8-9, 2011, pages 425 - 427
MUVAFFAK ASLI ET AL: "Prolonged cytotoxic effect of colchicine released from biodegradable microspheres", JOURNAL OF BIOMEDICAL MATERIALS RESEARCH PART B: APPLIED BIOMATERIALS, JOHN WILEY & SONS, HOBOKEN, NJ, US, vol. 71, no. 2, 15 November 2004 (2004-11-15), pages 295 - 304, XP002791225, ISSN: 1552-4973, DOI: 10.1002/JBM.B.30092 *
O'CALLAGHAN, J. THORACIC. ONCOL., vol. 5, 2010, pages 2024 - 36
PORTA, IMMUNOBIOLOGY, vol. 214, 2009, pages 761 - 77
QUALLSMURRAY, CURR. TOPICS IN DEVELOP. BIOL., vol. 94, 2011, pages 309 - 328
RIDKER ET AL., EUR. HEART J., vol. 39, 2018, pages 3499 - 3507
RIDKER ET AL., LANCET, vol. 391, 2018, pages 319 - 328
ROSENBERG SARESTIFO NPYANG JCMORGAN RADUDLEY ME: "Adoptive cell transfer: a clinical path to effective cancer immunotherapy", NAT. REV. CANCER, vol. 8, no. 4, April 2008 (2008-04-01), pages 299 - 308, XP002571413, DOI: 10.1038/nrc2355
RUFFELL ET AL., TRENDS IN IMMUNOL, vol. 33, 2012, pages 119 - 126
SICA ET AL., SEMINARS IN CANCER BIOL., vol. 18, 2008, pages 349 - 355
SMALL ET AL., J. CLIN. ONCOL., vol. 18, 2000, pages 3894 - 903
STANTONGERNERT ET AL., MED RES REV, vol. 31, no. 3, 2011, pages 443 - 81
VIDAL-VANACLOCHA, CANCER RES., vol. 54, 1994, pages 2667 - 2672
VORONOV, PROC. NAT'L. ACAD. SCI. USA, vol. 100, 2003, pages 2645 - 50
ZHANG, BIOSCI. REP., 2019

Similar Documents

Publication Publication Date Title
US11648206B2 (en) Sustained-release formulations of colchicine and methods of using same
US11877991B2 (en) Use of colchicine to inhibit tumor growth and metastases
JP7269227B2 (en) Niraparib formulation
KR20120046216A (en) 3-cyanoquinoline tablet formulations and uses thereof
JP2019529476A (en) Urothelial cancer and other urogenital organs using N- (4- (6,7-dimethoxyquinolin-4-yloxy) phenyl) -N ′-(4-fluorophenyl) cyclopropane-1,1-dicarboxamide Treatment for malignant tumor
WO2021089715A1 (en) Use of colchicine in the treatment and prevention of lung cancer
JP2022160654A (en) Drug combinations for reducing cell viability and/or cell proliferation
TW202211921A (en) Treatments of prostate cancer with combinations of abiraterone acetate and niraparib
EA045002B1 (en) USE OF COLCHICINE FOR INHIBITION OF TUMOR GROWTH AND METASTASES
JP6697539B2 (en) Colchicine sustained-release preparation and method of using the same
CN113453685A (en) Methods of treating liver cancer using orally administered dioxolane nucleotides in combination with anti-PD 1 or anti-PDL 1 monoclonal antibodies
EP3998066A1 (en) Colchicine salicylate and uses thereof

Legal Events

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

Ref document number: 20803143

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20803143

Country of ref document: EP

Kind code of ref document: A1