CA2449288A1 - Formulations of lymphokines and method of use thereof for local or both local and systemic control of proliferative cell disorders - Google Patents

Formulations of lymphokines and method of use thereof for local or both local and systemic control of proliferative cell disorders Download PDF

Info

Publication number
CA2449288A1
CA2449288A1 CA002449288A CA2449288A CA2449288A1 CA 2449288 A1 CA2449288 A1 CA 2449288A1 CA 002449288 A CA002449288 A CA 002449288A CA 2449288 A CA2449288 A CA 2449288A CA 2449288 A1 CA2449288 A1 CA 2449288A1
Authority
CA
Canada
Prior art keywords
formulation
local
peg
tumor
lymphokine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CA002449288A
Other languages
French (fr)
Other versions
CA2449288C (en
Inventor
Gaylen M. Zentner
Miroslav Baudys
Maria Jurek
Wolfram Samlowski
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
BTG International Ltd
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2449288A1 publication Critical patent/CA2449288A1/en
Application granted granted Critical
Publication of CA2449288C publication Critical patent/CA2449288C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

Therapeutic formulations comprising an effective amount of IL-2 or other lymphokine and a biodegradable polymeric carrier having reverse gelation properties and the methods of use thereof for local or both local and systemic control of proliferative cell disorders are disclosed. The formulation can be administered intratumorally/peritumorally and forms an IL-2 containing depot.
The IL-2-containing depot provides for continuous, prolonged release of IL-2 sufficient to stimulate the production of cytotoxic T lymphocytes which function both locally and systemically, without causing unacceptable side effects.

Description

FORMULATIONS OF LYMPHOKINES AND METHOD OF USE THEREOF
FOR LOCAL OR BOTH LOCAL AND SYSTEMIC CONTROL OF
PROLIFERATIVE CELL DISORDERS
Field of the Invention The present invention is in the field of localized sustained delivery of a lyrnphokine into a warm blooded animal. More particularly, this invention relates to therapeutic formulations and the methods of use thereof for local or both local and systemic control of proliferative cell disorders.
Background of the invention Recent advances in understanding the biology of the immune system have lead to the identification of important modulators of immune responses, generally called cytokines or if produced by lymphocytes, lymphokines. Rosenberg, Steven A.
"The Immunotherapy and Gene Therapy of Cancer." J. Clin. Oncology 10:180-199 (1992).
Cytokines are small proteins secreted primarily, but not exclusively, by cells of the immune system that promote the proliferation andlor differentiative functions of other cells. Examples of cytokines include interleukins, interferons, hematopoietic colony stimulating factors (CSF), and proinflammatory factors such as tumor necrosis factor (TNF). The administration of cytokines and related immunomodulators has resulted in objective tumor responses in patients with various types of neoplasms.
Sarna, Gregory et al., "A Pilot Study of Intralymphatic Interleukin-2. II. Clinical and Biological Effects", .l. Biol. Response Modifies, 9:81-86 (1990). However, current modes of cytokine administration are frequently associated with toxicities that limit the therapeutic value of these agents.
Interleukin-2 (IL-2), a lyrnphokine produced by normal peripheral blood lymphocytes that induces proliferation of antigen or mitogen stimulated T
cells after exposure to plant lectins, antigens, or other stimuli, was first described by Morgan, D.
A., et al., Science (1976), 193:1007-1008. IL-2 acts on the three major types of lymphocytes: T cells, B cells, and NK cells, stimulating them to proliferate and augmenting their differentiative functions. IL-2 potentiates both innate or natural host defenses by stimulating NK cells and antigen-specific acquired immune reactivity by stimulating T cells and B cells. IL-2 was initially made by cultivating human peripheral blood lymphocytes (PBL) or other IL-2-producing cell lines, described in, for example, U.S. Pat. No. 4,401,756. Recombinant DNA technology has provided an
2 alternative to PBLs and other cell lines for producing IL-2. Taniguchi, T. et al., Nature (1983), 302:305-310 and Devos, R., Nucleic Acids ResearcYc (1983), 11:4307-have reported cloning the human IL-2 gene and expressing it in microorganisms.
U.S. Pat. No. 4,518,584 describes and claims mutated proteins of IL-2 in which the cysteine normally occurring at position 125 of the wild-type or native molecule has been replaced with a neutral amino acid, such as serine or alanine. U.S.
Pat. Nos. 4,530,787 and 4,569,790 disclose and claim methods for purifying recombinant native IL-2 and muteins thereof, as well as the purified form of IL-2.
U.S. Pat. No. 4,604,377 discloses an IL-2 composition suitable for reconstitution in a pharmaceutically acceptable aqueous vehicle composed of oxidized microbially produced recombinant IL-2.
In response to tumor antigens, a subset of lymphocytes, termed helper T-cells, secrete small quantities of IL-2. This IL-2 acts locally at the site of tumor antigen stimulation to activate cytotoxic T-cells and natural killer cells that mediate systemic W mor cell destruction. Intravenous, intralymphatic and intralesional administration of IL-2 has resulted in clinically significant responses in some cancer patients.
It has been shown that systemic administration of recombinant IL-2 in high doses causes regression of established metastatic cancers in mice, and stimulates lympholcine-activated filler cells, and tumor-infiltrating lymphocytes in humans.
Rosenberg et al., J. Exp. Med. (1985) 161:1169-1188; Rosenberg, S. et al., New Ehg. J. Med.
(1985)313:1485-1492; Rosenberg et al., Science (1986) 233:1318-1321). However, severe toxicities (hypotension and edema) limit the dose and efficacy of intravenous and intralyrnphatic IL-2 administration. Hoover, H.C. Jr. et al. Cancer Res.
(1984 ),44(4):1671-6. The toxicity of systemically administered lympholcines is not surprising as these agents mediate local cellular interactions and they are normally secreted in only very small quantities. Additionally, other lymphokines, such as interleulcin-4 (IL-4), alpha interferon oc-INF) and gamma interferon (y 1NF) have been used to stimulate immune responses to tumor cells. Lilce IL-2, the current modes of administration have adverse side effects.
To circumvent the toxicity of systemic IL-2 administration several investigators have examined intralesional injection of IL-2. This approach eliminates the toxicity associated with systemic IL-2 administration. However, multiple intralesional injections are required to optimize therapeutic efficacy.
Bubenik, J. et al., InZmichology Letters. 23:287-292 (1989/1990); Borden, Ernest C., and Sondel, Paul M Ca~cer~. 1990 Feb 1;65(3 Suppl):800-14; Rosenberg, Steven A. et al., 108:853-864 (1988). Hence, these injections are impractical for many patients, particularly when tumor sites axe not accessible for injection without potential morbidity.
US Patent No. 6,045,788 discloses that low doses of natural and recombinant IL-2, may be continuously administered to patients for prolonged periods of time, e.g.
longer than three months, in order to activate and/or stimulate their immune system without causing substantial toxicity. However, frequent administration for prolonged periods of time is required, or, in the alternative, an optimum sustained release formulation that is not described in the patent is needed.
In addition to the potential toxicity, another problem of systemic administration of IL-2 is rapid renal elimination. When IL-2 is given intravenously it is eliminated rapidly with an initial elimination half life and terminal elimination half -life of 6 to 12 minutes and 40 to f0 minutes, respectively.
Therefore, there is a need for a formulation of IL-2 that forms IL-2 containing depot after being administered locally to a warm-blooded animal. The IL-2 containing depot provides a continuous, prolonged release of IL-2 sufficient to simulate the production of cytotoxic T lymphocytes (CTTL's) which function locally or both locally and systemically, without causing unacceptable side effects. An optimum material for use as an injectable or implantable polymeric drug Garner in the formulation of the present invention should be biodegradable, compatible with the drugs, and allow fabrication with simple, safe solvents, such as water, and not require additional polymerization or other covalent bond forming reactions following administration.
Summary of the Invention The invention provides a therapeutic formulation comprising a biodegradable block copolymeric drug carrier having thermally reversible gelation properties and an effective amount of lymphokine, said formulation can be administered intratumorally/peritumorally and forms a lymphokine containing depot. The lymphokine -containing depot provides for continuous, prolonged release of lympholcines such as IL-2 sufficient to simulate the production of cytotoxic T
lymphocytes which function locally or both locally and systemically, without causing any significant undesirable systemic side effects. The term "lymphokine"
refers to any agent having IL-2 or other interleulcin activity, including natural, recombinant and mutated IL-2 or other interleulcins, analogs and derivatives thereof.
Preferred lymphokines can be selected from the group consisting of interleukin-2 (IL-2), interleulcin-4, interleukin-12 and their derivatives.
The present invention also provides a method of converting a highly toxic drug, i.e. IL-2 or other lymphokines, into a fully effective drug with minimal unacceptable side effects by incorporating IL-2 or other lymphokines into a biodegradable drug carrier capable of forming a drug containing depot after local administration. Preferably, the biodegradable drug carrier has reverse thermal gelation properties, namely, such drug carriers are in a liquid state below body temperature but transition to a gel or solid state as they are warmed, and exist as gels at or about body temperature. In the remainder of this disclosure, the narrative will generically refer to IL-2, but the present teachings, unless otherwise indicated, extend to the remainder of the agents encompassed herein. Preferably, the IL-2 is a recombinant human IL-2.
The invention further provides a method for therapeutic or prophylactic treatment of the tumor burden in a warm blooded animal host, comprising locally administering to said host a formulation comprising an effective amount of IL-2, or other lymphokines, formulated with a biodegradable drug carrier having reverse thermal gelation properties.
The present invention can be used to treat/cure proliferative cellular disorders such as cancer, or warts. The formulations of the present invention, when administered locally, provide for sustained local release of high levels of IL-2, or other lymphokines(still a low total dose based on systemic exposure), directly to a diseased tissue to be treated, i.e. tumors or warts, stimulating the production of cytotoxic T-lymphocytes that attack the diseased tissue at both the local site and throughout the body as well, without causing unacceptable systemic side effects. Without the presence of the drug carriers used in the present formulation, the IL-2 is largely ineffective due to rapid clearance from the injection site before CTTL
stimulation occurs. Because the drug, namely IL-2 or other lympholcines, stimulates the body's own defenses to attack the diseased tissues, the potential problem of drug resistance is eliminated. Therefore, the present invention provides formulations and methods of use for the treatment of many types of local and metastatic cancer from simple, low-s dose local injections. The formulations of the present invention may be achninistered at intervals ranging from daily to monthly. The formulations of the present invention also provide for local, or both local and systemic anti-cancer therapy from the localized injection of a dose of IL-2 that would be sub-therapeutic if administered systemically.
Brief Descriuti0ns of the d)rawin~s The additional features and advantages of the invention will become apparent from a consideration of the following detailed description presented and the accompanying drawings in which:
FIG. 1A illustrates IL-2 release ifa vitro from the IL-2 formulation, according to Example 1, as analyzed and monitored by ELISA.
FIG. 1B illustrates IL-2 release in vit~°o from the IL-2 formulation, according to Example 1, as analyzed and monitored by a cell proliferation assay.
FIG. 2 illustrates the cytotoxicity assay results of the IL-2 formulation according to Example 1, measured by percentage of specific cell lysis.
FIG. 3 illustrates the effects of the administration of the IL-2 formulation, according to Example l, on fibrosarcoma tumor growth as measured by change in tumor size over time.
FIG. 4 illustrates IL-2 release if2 vitro from the IL-2 formulation, according to Example 4, monitored by ELISA.
FIG. 5 illustrates the dose escalation effect of the administration of the IL-formulation, according to Example 4, on tumor growth as measured by change in tumor size over time.
FIG. 6 illustrates the effect of the repetitive administration of the IL-2 formulation, according to Example 4, on tumor growth as measured by change in tumor size over time.

FIG. 7 illustrates the effect of the repetitive administration of the IL-2 formulation, according to Example 4, on mouse stuvival rate in a MetA
fibrosarcoma tumor model.
FIG. 8 illustrates the effect of the repetitive dosing of the IL-2 formulation, according to Example 4,on mouse survival rate in a B 16 melanoma tumor model.
Detailed Description of the Invention Before the present therapeutic formulations and methods of use thereof for drug delivery, and in particular for IL-2 delivery, are disclosed and described, it is to be understood that this invention is not limited to the particular configurations, process steps, and materials disclosed herein. Such configurations, process steps, and materials may vary somewhat, which are intended to be within the scope of the invention. It is also to be understood that the terminology employed herein is used for the purpose of describing particular embodiments only and is not intended to be limiting since the scope of the present invention will be limited only by the appended claims and equivalents thereof.
It must be noted that, as used in this specification and the appended claims, the singular forms "a", "an", and "the", includes plural referents tmless the context clearly dictates otherwise. Thus, for example, reference to a composition for delivering "a drug" includes reference to one, two, or more drugs. In describing and claiming the present invention, the following terminology will be used in accordance with the definitions set forth below.
"Parenteral" shall mean intratumoral, peritumoral, intralesional, perilesional, , intrathecal, intraperitoneal, and infra-abdominal.
The term "cell proliferative disorders" includes disorders of cell proliferation and differentiation, commonly considered neoplastic or malignant such as cancers.
Cancer includes, for example, carcinomas, melanomas, myelomas, sarcomas, and the like. The term cancer also includes pre-cancerous tissues and cells that are known to progress into true cancer if left untreated. Other examples of cell proliferative disorder are warts.
The term "therapeutic" treatment refers to administration to patients of a drug, particularly IL-2, after the patient has developed cancer, (i.e., after a tumor burden has been determined), as measured by any means in the art, with resultant decrease or elimination of the existing tumor burden being the goal.
The term "prophylactic" treatment refers to such administration to prevent recurrence of the cancer after therapeutic treatment has been administered.
The term "pharmacologically effective amount" refers to the amount of each active component of the method or composition herein that is sufficient in showing a meaningful patient benefit, i.e., prolongation of life and/or reduction of disease and/or improvement in any clinically significant way. When the effective amounts defined herein are employed, more efficacy is obtained using the combination than using either component alone. As applied to an individual active ingredient administered alone, the term refers to that ingredient alone; when combinations are used, the term refers to combined amounts in the preparation that result in the therapeutic or prophylactic effect.
The term "recombinant" refers to drugs produced by recombinant DNA
techniques wherein in the particular case of IL-2 the gene coding for the IL-2 is cloned by known recombinant DNA technology.
The term "pharmaceutically acceptable" refers to a carrier medium that does not interfere with the effectiveness of the biological activity of the dr~ig and that is not toxic to the host to which it is administered.
"Gelation temperature" means the temperature at which the biodegradable block copolymer undergoes reverse thermal gelation. In other words, the temperaW re below which the block copolymer is apparently soluble or exists as a uniform colloidal system in water and exists as a free-flowing fluid, and above which the block copolymer undergoes phase transition to increase in viscosity or to form a semi-solid gel. The terms "gelation temperature" and "reverse thermal gelation temperature" or the like shall be used interchangeably in referring to the gelation temperature.
"Polymer solution", "aqueous solution", "solution", "homogeneous solution"
and the like, when used in reference to a biodegradable block copolymer contained in such solution, shall mean a water based solution having such block copolymer dissolved or in a uniform colloidal state therein at a functional concentration, and maintaiiled at a temperature below the gelation temperature of the block copolymer.
They shall include water without additives or aqueous solutions containing additives g or excipients such as pH buffers, components for tonicity adjustment, antioxidants, preservatives, drug stabilizers, etc., as commonly used in the preparation of pharmaceutical formulations.
"Reverse thermal gelation" is the phenomenon whereby a solution of a block copolymer spontaneously increases in viscosity, and in many instances transforms into a semisolid gel, as the temperature of the solution is increased above the gelation temperature of the copolymer. For the purposes of the invention, the term "gel"
includes both the semisolid gel and the high viscosity state that exists above the gelation temperature. When cooled to below the gelation temperature, the gel spontaneously reverses over a period of a few minutes to several hours to reform the lower viscosity free-flowing fluid. All interactions to create the gel are physical in nature and do not involve the formation or brealcing of covalent bonds.
"Drug delivery liquid" or "dnzg delivery liquid having reverse thermal gelation properties" shall mean polymer solutions that contain a drug (the drug peg se can either be dissolved, dispersed or colloidal) suitable for administration to a warm-blooded animal. The drug containing polymer solution forms a gelled drug depot when the temperature is raised to or above the gelation temperature of the drug delivery liquid.
"Depot" means a localized site in the body containing concentrated active agents or drugs. Examples of formulations that form depots are gels, implants, microspheres, matrices, particles, etc.
"Gel" means the semi-solid phase that spontaneously occurs as the temperatL~re of the "polymer solution" or "drug delivery liquid" is raised to or above the gelation temperature of the block copolymer.
"Gel mixture" or "mixture of triblock copolymers" refers to a reverse thermal gelation system comprising two or more ABA or BAB tribloclc copolymer components. The mixture can be made either by simply mixing two or more individually synthesized triblock copolymer components, or by synthesizing two or more types of copolymer systems in one synthesizing vessel. The mixture prepared by the above two processes may be combined with water to form a polymer solution that may have the same or different gelation properties and gel qualities.

"Solution", "solubilized", "dissolved" and all other terms that refer to a solution or dissolved state includes a homogeneous solution, micellar solution, or any apparently uniform colloidal state such as an emulsion or a suspension.
"Biodegradable" means that the block copolymer can chemically break down or degrade within the body to form nontoxic components. The rate of degradation can be the same or different from the rate of drug release.
"Drug" shall mean any organic or inorganic compound or substance having biological or pharmacological activity that can be adapted or used for a therapeutic purpose.
"Peptide," "polypeptide," "oligopeptide" and "protein" shall be used interchangeably when referring to peptide or protein drugs and shall not be limited as to any particular molecular weight, peptide sequence or length, field of bioactivity or therapeutic use unless specifically stated.
"Interleukin" shall mean any protein/polypeptide agent or its derivative, analog or mimetic, having interleulcin activity, particularly IL-2 and IL-12 activities, including natural and recombinant IL-2 and IL-12, pharmaceutically-acceptable fusion proteins of natural and recombinant interleukins, derivatives and mixW res thereof.
"hIL-2" refers to a protein exhibiting the spectrum of activities characterizing human interleukin-2. Specifically, the protein must be capable of stimulating the proliferation of hIL-2 dependent cytolytic and helper T cell lines, as set forth in the standard assays of S. Gillis et al, J. Immuhol. (1978) 120:2027-2032 and of J.
Watson, J. Exp. Med. (1979) 150:1510-1519. Modified IL-2 and IL-12 are also included in this definition so long as biological activity is not destroyed thereby.
"PLGA" shall mean a copolymer or copolymer radicals derived from the condensation copolyrnerization of lactic acid and glycolic acid, or, by the ring opening copolymerization of lactide and glycolide. The term lactic acid and lactate are used interchangeably; glycolic acid and glycolate are also used interchangeably.
"PLA" shall mean a polymer derived from the condensation of lactic acid or by the ring opening polymerization of lactide.
"PGA" shall mean a polymer derived from the condensation of glycolic acid or by the ring opening polymerization of glycolide.

"Biodegradable polyester or poly(ortho esters" refers to any biodegradable polyester or poly(ortho ester), wherein the polyesters are preferably synthesized from monomers selected from the group consisting of D,L-lactide, D-lactide, L-lactide, D,L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid, s-caprolactone, E-5 hydroxy hexanoic acid, y butyrolactone, 'y hydroxy butyric acid, 8-valerolactone, 8-hydroxy valeric acid, hydroxybutyric acid, malic acid, and copolymers thereof.
"ReGel~" is a tradename of MacroMed Incorporated for a class of low molecular weight, biodegradable block copolymers having reverse thermal gelation properties as described in US Patent Nos. 6,004,573, 6,117949, 6,201, 072, and 10 6,287,588, hereby incorporated by reference. It also includes compositions disclosed in pending U.S. patent applications Serial Nos. 09/906,041 and 09/559,799 hereby incorporated by reference. The biodegradable drug carrier comprises ABA-type or BAB-type triblock copolymers or mixtures thereof, wherein the A-bloclcs are relatively hydrophobic and comprise biodegradable polyesters or poly(ortho esters, and the B-blocks are relatively hydrophilic and comprise polyethylene glycol (PEG), said copolymer having a hydrophobic content of between 50.1 to 83% by weight and a hydrophilic content of between 17 to 49.9% by weight, and an overall block copolymer molecular weight of between 2000 and 8000. The drug carriers exhibit water solubility at temperatures below normal mammalian body temperatures and undergo reversible thermal gelation to then exist as a gel at temperatures equal to physiological mammalian body temperatures. The biodegradable, hydrophobic A
polymer block comprises a polyester or poly (ortho ester), wherein the polyester is synthesized from monomers selected from the group consisting of D,L-lactide, D-lactide, L-lactide, D,L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid, ~-caprolactone, s-hydroxyhexanoic acid, y butyrolactone, y hydroxybutyric acid, 8-valerolactone, 8-hydroxyvaleric acid, hydroxybutyric acids, malic acid, and copolymers thereof and having an average molecular weight of between about 600 and 3000. The hydrophilic B-block segment is preferably polyethylene glycol (PEG) having an average molecular weight of between about S00 and 2200.
The concentration at which the bloclc copolymers are soluble at temperatures below the gelation temperature may be considered as the functional concentration.
Generally speaking, block copolymer concentrations of as low as 3% and up to about 50% by weight can be used and still be functional. However, concentrations in the range of about 5 to 40% by weight are preferred and concentrations in the range of about 10 to 30% by weight are most preferred. At the lower functional concentration ranges, phase transition may result in the formation of a weak gel or viscous liquid, however these systems are still functional for purposes of the invention.
The formulation of the present invention may also comprise a reconstitution enhancing and enabling agent comprising a liquid polyethylene glycol (PEG), a PEG
derivative, or a mixture of PEG and a PEG derivative, said PEG or PEG
derivative having a molecular weight of ISO to 1100 Daltons. The PEG derivative is comprised of PEG that has been derivatized with a member selected from the group consisting of D,L-lactide, D-lactide, L-lactide, D,L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid and copolymers thereof. The PEG derivative can also be a member represented by Rl-CO-O-(PEG)-CO-RZ or Rl-O-(PEG)-RZ wherein Rl and RZ
are independently members selected from the group consisting of H and C1 to Clo I S alkyl.
The mixture of the biodegradable copolymer and the drug, such as IL-2, may be prepared as.an aqueous solution or uniform colloid of the copolymer below the gelation temperature to form an IL-2 delivery liquid wherein IL-2 may be either partially or completely dissolved. This IL-2 delivery liquid is then administered via local routes~of delivery, such as intratumoral or peritumoral, to a patient whereupon it undergoes reversible thermal gelation since body temperature will be above the gelation temperature. This substantially reduces or in many cases eliminates the systemic toxicity of IL-2 and maximizes the immune system response to cancer cells.
Other potential applications of the IL-2 formulations of the present invention include administration locally at the site of surgical intervention/tumor excision to kill cancer cells remaining after surgery. Other possible local routes of delivery to treat malignant tumors include intraperitoneal administration for tumors/metastases localized in the peritoneum, and intracranial administration for tumors localized in the sl~.ill.
The present invention also provides a general method for converting highly toxic cytolcines and related dn~gs, such as lymphol~ines, including IL-2, IL-4, IL-12 and their derivatives and mimetics, into effective anticancer agents, while minimizing negative side effects. The formulations of the present invention provide for sustained delivery of the drug from a local depot formed intratumorally/peritiunorally at a single or several localized tumor site(s), that induce both local or both local and systemic immune responses.
By forming a local sustained delivery drug depot intratumorally/peritumorally, a relatively high local cytolcine steady state concentration at the twnor site is achieved.
The high local concentration of IL-2 stimulates the activation and production of tumor specific CTTL's that attaclc tumor tissue both locally as well as throughout the body, while the overall systemic IL-2 level is low enough to avoid any significant unacceptable side effects.
The formulation and method of the present invention are useful for treatment of any tumors that present as a manifestation of a cancer disease that can be formed by any malignant or pre-cancerous tissue, including carcinomas, sarcomas, melanomas, myelomas and the like. The formulations and methods of the present invention provide a way of converting a highly toxic drug, particularly a cytokine such as IL-2, IL-4, IL-12 and their derivatives and mimetics, into an effective therapeutic formulation without significant undesirable side effects. Even though the overall lymphokine dose administered is lowered by an order of magnitude as compared to the dose of IL-2 or other lyrnpholcine required for systemic treatment, the formulations of the present invention still provide for better or comparable pharmacological effect.
In one embodiment of the present invention, IL-2 is incorporated in a biodegradable block copolymeric drug carrier, having reverse thermal gelation properties (ReGel°), in a soluble form or as a suspension or other colloidal form and kept below its gelation temperature. After being administered intratumorally or peritumorally to a patient or a diseased animal (warm blooded mammalian host) whose bodies' temperature is above the ReGel° gelation temperature, the formulation forms local depots which can provide for continuous, sustained release of IL-2. The IL-2/ ReGel° formulations can also contain various additives, such as polyols including sugars, surfactants, amino acids, other proteins and buffer salts.
These additives can serve as functional and/or physical stabilizers of a particular lympholcine, including IL-2, IL-4, IL-12 and their derivatives and mirnetics, before injection in the liquid state or at the local IL-2 depot site in the gel after injection.

The formulations of the present invention can be administered once but are preferably administered repeatedly on a daily to monthly basis in order to provide for improved therapeutic results such as regression in tumor size thus managing cancer disease chronically or causing complete disappearance or non-reappearance of the tumor. Repeated administration at the same injection site is made possible by a fast removal of the formulation from the injection site over a period of several days to four weeks depending on drug carrier type due to spontaneous, in vivo, chemical degradation of the biodegradable block copolymers.
Generally, the formulation of the present invention can be administered peritumorally or intratumorally at a tumor site that has been identified by a well-established diagnostic imaging technique. One specific route of administration of IL-2 formulation of the present invention for malignant tumor treatment requiring precise tumor localization is intracranial administration for tumors within the skull.
Another specific local route of administration of IL-2 formulation of the present invention for malignant tumor treatment that does not require specific tumor localization is intraperitoneal IL-2 admiustration for primary tumors and tumor metastases localized in the peritoneum. Another specific administration route that does not require any sophisticated imaging technique is subcutaneous injection of an IL-2 formulation of the present invention next to tumors localized in the skin, for example primary and metastatic melanoma tumors. Another specific administration route is administration of an IL-2 formulation locally at the site of surgical intervention/tumor excision to bill cancer cells remaining after surgery.
The optimum dose administered will depend on the type of cancer, type of host, tumor localization, route, schedule and sequence of administration, already existing tumor burden (disease state), the type of cytokine, particularly IL-2, IL-4, IL-12 and their derivatives and mimetics, or other lympholcine used. However, the general range is within the range of 1000 LU. to 1x108 LU. for the IL-2-formulations of the present invention.
The method of this invention involves administering to a warm-blooded mammalian host, including a mouse, rat, rabbit, primate, pig or human host, preferably a human patient, a pharmacologically effective amount of a drug, particularly IL-2. This system will not cause unacceptable toxicity or mechanical damage to the surrounding tissue due to the biocompatibility of the materials and pliability of the gel, and will be completely biodegraded to lactic acid, glycolic acid, or other corresponding monomers, and polyethylene glycol within a specific time interval. The IL-2 release rate, gel strength, gelation temperature and degradation rate can be controlled by proper design and preparation of the various dmg carriers. For example, the weight percent of A-blocks and B-blocks, the mole percentages of monomers comprising the A-blocks, and the molecular weight and polydispersity of the ABA or BAB triblock copolymers can be modified. IL-2 release is also controllable through adjustment of the concentration of polymer in the drug delivery liquid.
A dosage form comprised of a polymer solution that contains IL-2, i.e. drug delivery liquid, is administered to the body. This formulation then spontaneously gels, due to the reverse thermal gelation properties of the block copolymer, to form a dntg depot as the temperature of the formulation rises to body temperature. In most instances, the IL-2 will make up between about 1000 LU. to 1x108 LU./ml of the drug delivery liquid. In some instances, the functionality or physical stability of the interleukins can be increased by addition of various additives to the aqueous solutions or suspensions. Additives, such as polyols (including sugars), amino acids;
surfactants, preservatives, antioxidants, stabilizing agents, tonicity adjusting agents, other proteins and certain salts may be used. These additives can be readily incorporated into the drug delivery liquid.
The dosage amount that appears to be most effective is one that results in regression in size of the tumor, complete disappearance or non-reappearance of the tumor, and is not toxic or has an acceptable toxicity to the host. This optimum dose level will depend on many factors, for example, on the type of host and type of cancer, route, schedule and sequence of administration, existing tumor burden, the type of IL-2 and the definition of toxicity.
In one embodiment of the present invention, IL-2 (20x106 LU.) is combined with ReGel° to form a liquid formulation, which is then injected peritLtmorally. The IL-2/ReGel° forms a gel at the injection site when it is warmed to body temperature and provides for the prolonged release from the depot wherein the IL-2 is released slowly and continuously for several days. The IL-2 that is released from the gel is active and stimulates CTTL's both locally and systemically. In contrast, a single injection of IL-2 in a conventional formulation (i.e., without the ReGel° carrier) has been proven ineffective in controlling tumor growth. Therefore, the IL-2/ReGel~
formulation of the present invention provides Local or both local and systemic 5 anticancer therapy from the local injection of a dose of IL-2 that would be sub-therapeutic if dosed systemically.
The following Examples are presented to illustrate the process of preparing the composition and method of using the composition of the present invention.

10 This example illustrates the ih vitro release of IL-2 from the IL-2 formulation of the present invention.
The biodegradable block copolymer carrier in this Example is a 23 wt solution of a block copolymer having a PLG/PEG-1000 weight ratio of 2.4, a L/G
mole ratio of 75/25, a molecular weight of 4,000 Daltons and gelation temperature of IS 14°C.
A fixed volume (1 mL) of sterile IL-2 formulation containing 50,000 LU. (3 ~.g) of IL-2 (Proleukin", commercially available from Chiron) was placed into the bottom of 50 mL tissue culture flasks in duplicate. The flasks were incubated at 37°C
in the presence of 25 mL of tissue culture medium (RPMI 1640, BioWhittalcer, Inc.).
At pre-selected time points, aliquots (0.5 mL) of release medium were withdrawn and assayed for IL-2 content after appropriate dilution. The IL-2 content was assayed by a specific enzyme linked immunoassay (OptEIA Human IL-2 Set, Pharmingen) and by a quantitative cell proliferative assay using CTTL-20 indicator cells following thymidine incorporation into the cells (24 hrs incubation at 37°C). For the later, a dose response curve was prepared with serial dilutions of standard IL-2. The results are plotted as cumulative IL-2 release as function of time (Fig.lA for IL-2 measured by ELISA, Fig IB for IL-2 measured by cell proliferative assay). Both methods showed that IL-2 release was quantitative, lasted 3 to 4 days, and the released IL-2 was fiilly bioactive.

This Example illustrates the ability of IL-2 released from formulation of the present invention to induce cytotoxic lymphocytes by using the same IL-2 formulation described in Example 1.
A fixed volume (0.5 mL) of sterile the IL-2 formulation as described in Example 1, containing escalating doses of IL-2 (Proleulcin°; 12,500 LU.; 25,000 LU.;
50,000 LU.) was placed into the bottom of 25 mL tissue culture flasks, in duplicate.
The flaslcs were incubated at 37°C for 3 days in the presence of 25 mL
of tissue culture medium (RPMI 1640, BioWhittaker, Inc.) containing marine splenocytes.
Activated lymphocytes were harvested and analyzed for their capacity to bill fibrosarcoma tumor cells in a 5' Cr release assay by determining the percentage (%) of tumor cells undergoing lysis. As shown in Fig.2, the IL-2 released from ReGel° is fully bioactive as compared to free IL-2 added to the release medium at the beginning of the release period. The E:T cell ratio in Fig. 2 represents the ratio of effector cells (activated lymphocytes) to target cells (RD-995 tumor cells), the number of which per sample (104) was lcept constant. In conclusion, the IL-2 released from the formulation of the present invention was fully bioactive and the formulation can be used as an effective delivery system fox sustained local peritumoral IL-2 delivery i~c vivo.
EXAMI°LE 3 The example illustrates tumor regression by a single peritumoral injection of the IL-2 formulation of the present invention in mice.
Mice (C3H/HEN) were implanted subcutaneously with RD-995 fibrosarcoma tumor cells. When the solid tumors were 4-5 mm in size, the mice (divided into groups of b) were injected with 0.2 mL IL-2 formulations as described in Example 1, 2 x 0.1 mL on the opposite sides of tumor perimeter. The IL-2 formulation contains escalating doses of IL-2 100,000 LU.; 250,000 LU. or 500,000 LU. In the control group, the drug carrier alone was injected. Tumor size measurements were obtained every other day for 3 weeks (Fig. 3). Tumor growth was arrested in each group as compared to the control group.
EXA19~PLE 4 This example illustrates the in vitro release of IL-2 from a dnig carrier that has a gelation temperature above 20°C.

The biodegradable polymer carrier W this Example is a 13 wt% solution of a block copolymer having a PEG(1000)/PEG(1450) weight ratio of 20/80, a PLG/PEG
weight ratio of 2.06, a L/G mole ratio of 85/15, a molecular weight of 4,800 Daltons and gelation temperature of 26°C.
A fixed volume (1 mL) of sterile IL-2 formulation containing 50,000 LU. (3 ~,g) of IL-2 (Proleukin°) was placed into the bottom of 50 mL tissue culture flaslcs, in duplicate. The flasks were incubated at 37°C in the presence of 25 mL
of tissue culture medium (RPMI 1640, BioWhittalcer, Inc.). At pre-selected time points, aliquots of release medimn (0.5 ml) were withdrawn and after appropriate dilution assayed for IL-2 content by a specific enzyme linlced immunoassay (OptEIA
Human IL-2 Set, Pharmingen). The results were plotted as IL-2 cumulative release vs.
time (Fig. 4). IL-2 loaded in ReGel~ was released in vity-o from the gel depot quantitatively in a bioactive form over a period of 4 days.
EXAMPLE ~
This example illustrates tumor regression by a single peritumoral injection of an IL-2 formulation in mice. The IL-2 formulation was the same as described in Example 4.
Mice (C3H/HEN) were implanted subcutaneously with RD-995 fibrosarcoma tumor cells. When the solid tumors were 4-5 mm in size, the mice (divided into groups of 10) were injected with 0.2 mL of IL-2 formulation (2 x 0.1 mL on the opposite sides of tumor perimeter) containing escalating doses of IL-2 (500,000 LU.;
2,000,000 LU. or 4,000,000 LU.). In the control groups, ReGel° only formulation (drug free) or conventional IL-2 formulation (500,000 LU.) were injected peritumorally. Tumor size measurements were obtained every other day for 26 days (Fig. 5). Tumor growth was arrested for about 3 weeks in each ReGel~/IL-2 group as compared to the control groups. Blood pressure was measured on day 3 and 5 after administration in each group of animals. All blood pressure values stayed in the physiological range, and no adverse decreases in blood pressure were detected, indicating that the IL-2 formulation of the present invention causes no adverse side effects.

This example illustrates tumor regression by weeldy peritumoral injection of IL-2 formulation in mice. The IL-2 formulation was the same as described in Example 4.
Mice (C3H/HEN) were implanted subcutaneously with RD-995 fibrosarcoma tumor cells. When the solid tumors were 4-5 mm in size, the mice (divided into groups of 10) were injected with 0.2 mL of IL-2 formulation (2 x 0.1 mL on the opposite sides of tumor perimeter) containing 2,000,000 LU. of IL-2. In the control groups, a drug carrier only formulation (drug free) or a conventional IL-2 formulation (500,000 LU.) was injected periturnorally. Another control group was treated using a conventional IL-2 formulation (180,000 LU.) administered systemically (S.C.) twice a day (B.LD.) for 5 consecutive days (maximmn tolerated dose level for systemic in mice). The administration was repeated at days 7, 14, and 21 except for the conventional systemic B.LD. group. Tumor size measurements were obtained every other day (Fig. 6). Tumor growth was arrested for about 41/2 weeks in the IL-2 formulation group as compared to the control groups. Blood pressure was measured on day 8 after the first administration in each group of animals. All blood pressure values stayed in the physiological range and no adverse decreases in blood pressure were detected.

This example illustrates weekly intraperitoneal injection of an IL-2 formulation in MethA intraperitoneal tumor mouse model. The IL-2 formulation was the same as described in Example 4.
Mice (Balb C) were injected intraperitoneally with 106 MethA fibrosarcoma tumor cells. After 3 days (day 0, Fig. 7), the mice (divided into groups of 10) were inj ected intraperitoneally with 0.2 mL of an IL-2 formulation containing escalating doses of IL-2 (100,000 LU.; 500,000 LU. or 2,000,000 LU.). The control groups consisted of tumor bearing untreated mice, drug free formulation injected mice, mice treated with conventional systemic IL-2 (180,00 LU. S.C. B.LD. x 5 days), and mice injected intraperitoneally with 500,000 LU. of conventional IL-2. The injections were repeated weekly at days 7, 14, 21, 28 and 35 except for the conventional systemic B.LD. group. Mice were followed for survival (Fig. 7). It was demonstrated that the IL-2 formulation of the present invention gave superior survival data as compared to the positive controls using conventional IL-2 formulations.

This example illustrates survival rate by weekly peritumoral injection of the IL-2 formulation of the present invention in mice bearing B16 melanoma solid tumor.
The IL-2 formulation was the same as described in Example 4.
Mice (C57/Bl6) were implanted subcutaneously with B 1 & melanoma tumor cells. When the solid tlunors were 4-5 mm in size, the mice (divided into groups of 8) were injected with 0.2 mL of the IL-2 formulation of the present invention (2 x 0.1 mL on the opposite sides of tumor perimeter) containing 2,000,000 LU. of IL-2.
In the control groups, drug carrier only formulation (drug flee) or a conventional IL-2 formulation (2,000,000 LU.) were injected peritumorally. The administration was repeated at day 7, 14, and 21, 28 and 35. Mice were followed for tumor growth and survival rate. Blood pressure was measured on day 1, 2, 3, 4 and 5 (after first administration), and repeatedly on day 14, 15, 16, 17 and 18 (after third administration) in each anmal group. All blood pressure values stayed in the physiological range and no adverse decreases in blood pressure were detected.
While the W mor growth was significantly slowed with the IL-2 formulation of the present invention as compared to the control groups, the IL-2 formulation of the present invention provided superior survival data as compared to the positive control using conventional IL-2 formulation (Fig. 8).

This example illustrates activation of systemic immunity by weekly peritumoral injection of IL-2 formulation in mice, the drug carrier is the same as described in Example 1.
Mice (C3H/HEN) are implanted subcutaneously with two tumors, one tumor on each flank using RI)-995 fibrosarcoma tumor cells. When the solid tumors are 4-5 mm in size, the mice (divided into groups of 10) are injected with 0.2 mL
formulation as described in Example l, 2 x 0.1 mL on the opposite sides of tumor perimeter containing 2,000,000 LU. of IL-2 at only right sided tmnor (5 mice) or left sided tumor (5 mice). Control groups consist of two tumors bearing entreated mice, drug carrier only injected mice (5 mice at left sided tumor and 5 mice at right sided tumor), and mice injected perittunorally (S mice at left sided tumor and S mice at right sided ttunor) with conventional IL-2 formulation (2,000,000 LU.). In each group, the administration is repeated weekly at the identical pre-selected tumor site for consecutive weeks. Growth of both treated as well as untreated tumor is arrested (or S. tumors regressed completely) in the IL-2/formulation group while in the control groups, both tumors grow progressively. Accordingly, survival data for the IL-formulation group are superior to those obtained for control groups, including peritumoral conventional IL-2 group, demonstrating that systemic immune response is only achieved with the IL-2 formulation of the present invention.
10 E~'AMPLE 10 This example illustrates activation of systemic immunity by weekly peritumoral injection of IL-2 fornulation in mice. The drug carrier is the same as described in Example 4.
Mice (C3H/HEI~ are implanted subcutaneously with two tumors, one tumor 1 S on each flank using RD-995 fibrosarcoma tumor cells. When the solid tumors are 4-S
mm in size, the mice (divided into groups of 10) are injected with 0.2 mL IL-2 formulation as described in Example 4, 2 x 0.1, mL on the opposite sides of tumor perimeter containing 2,000,000 LU. of IL-2 at only right sided tumor (5 mice) or left sided tumor (S mice). Control groups consist of two tumors bearing untreated mice, 20 dntg can-ier only injected mice (5 mice at left sided,tttmor and S mice at right sided tumor), and mice injected peritttmorally (S mice at left sided tumor and S
mice at night sided tumor) with conventional IL-2 formulation (2,000,000 LU.). In each group, the administration is repeated weekly at the identical pre-selected tumor site for consecutive weeks. Growth of both treated as well as untreated tumor is arrested (or tumors regressed completely) in the IL-2 formulation group while in the control groups, both tumors grow progressively. Accordingly, survival data for the IL-fornulation group are superior to those obtained for control groups, including peritumoral conventional IL-2 group, demonstrating that systemic immune response is only achieved with the IL-2 formulation of the present invention.
The above Examples are presented for illustrative purposes only, and are not intended, and should not be construed to limit the invention in any manner.
Various modifications of the compounds and methods of the invention may be made without departing from the spirit or scope thereof. It will be immediately apparent to one skilled in the art which various modifications may be made without departing from the scope of the invention that is limited only by the following claims and their functional equivalents.

Claims (10)

22
1. A formulation for locally administering a lymphokine to a warm blooded animal to provide local or both local and systemic therapeutic effect, comprising:
a) an effective amount of a lymphokine;
b) a biodegradable ABA- or BAB-type tri-block copolymer comprising:
i) 51 to 83 % by weight of a biodegradable, hydrophobic A block comprising a biodegradable polyester or poly(ortho ester), and ii) 17 to 49 % by weight of a hydrophilic B block comprising a polyethylene glycol (PEG), said tri-block copolymer having a weight average molecular weight of between about 2000 to 4990 and possessing reverse thermal gelation properties;
and c) a reconstitution enhancing and enabling agent comprising a polyethylene glycol (PEG), a PEG derivative or a mixture of PEG and a PEG derivative, said PEG
or PEG derivative having a weight averaged molecular weight of 150 to 1100 Daltons;
and wherein said formulation forms a lymphokine containing depot after being administered and provide continuous, sustained release of lymphokine.
2. The formulation according to Claim 1, wherein the lymphokine is a member selected from the group consisting of interleukin-2 (IL-2), interleukin-4, interleukin-12, derivatives and mimetics thereof.
3. The formulation according to Claim 1 where the formulation is an injectable liquid prior administration.
4. The formulation according to Claim 1 further comprising a biocompatible additive selected from the group consisting of polyols including sugars, surfactants, amino acids, proteins, preservatives, antioxidants, stabilizing agents, tonicity adjusting agents, buffer salts and equivalents thereof.
5. The formulation according to Claim 1 wherein the PEG derivative is comprised of PEG that has been derivatized with a member selected from the group consisting of D,L-lactide, D-lactide, L-lactide, D,L-lactic acid, D-lactic acid, L-lactic acid, glycolide, glycolic acid and copolymers thereof.
6. The formulation according to Claim 1 wherein the PEG derivative is represented by R1-CO-O-(PEG)-CO-R2 or R1-O-(PEG)-R2 wherein R1 and R2 are independently members selected from the group consisting of H and C1 to C10 alkyl.
7. A method for the local or both local and systemic control of proliferative cell disorders in a warm-blooded animal, comprising:
a) preparing a lymphokine formulation of one of the Claims 1 to 6, b) administering said formulation adjacent or into the area of said warm-blooded animal where the proliferate cell disorder occurs;
c) allowing said formulation to form a lymphokine containing depot which provides continuous, sustained release of lymphokine such that local or both local and systemic therapeutic effects are achieved without causing unacceptable side effects.
8. The method according to Claim 7, wherein the proliferative cell disorder is cancer or warts.
9. The method according to Claim 7, wherein the administration is via a parenteral means selected from the group consisting of intratumoral, peritumoral, perilesional, intralesional, intrathecal, intraperitoneal, and intra-abdominal.
10. The method according to Claim 7, wherein the formulation is administered daily to monthly.
CA2449288A 2001-06-14 2002-06-14 Formulations of lymphokines and method of use thereof for local or both local and systemic control of proliferative cell disorders Expired - Fee Related CA2449288C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US29859401P 2001-06-14 2001-06-14
US60/298,594 2001-06-14
US10/172,805 US20030003074A1 (en) 2001-06-14 2002-06-13 Formulations of lymphokines and method of use thereof for local or both local and systemic control of proliferative cell disorders
US10/172,805 2002-06-13
PCT/US2002/018807 WO2002102309A2 (en) 2001-06-14 2002-06-14 Formulation of lymphokines and method of use thereof for local or bath local and systemic control of proliferative cell disorders

Publications (2)

Publication Number Publication Date
CA2449288A1 true CA2449288A1 (en) 2002-12-27
CA2449288C CA2449288C (en) 2018-01-02

Family

ID=26868470

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2449288A Expired - Fee Related CA2449288C (en) 2001-06-14 2002-06-14 Formulations of lymphokines and method of use thereof for local or both local and systemic control of proliferative cell disorders

Country Status (8)

Country Link
US (1) US20030003074A1 (en)
EP (1) EP1446423A4 (en)
JP (1) JP2004538271A (en)
KR (1) KR20040052510A (en)
CN (1) CN1610694A (en)
AU (1) AU2002312499B8 (en)
CA (1) CA2449288C (en)
WO (1) WO2002102309A2 (en)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6824822B2 (en) * 2001-08-31 2004-11-30 Alkermes Controlled Therapeutics Inc. Ii Residual solvent extraction method and microparticles produced thereby
US20040185101A1 (en) * 2001-03-27 2004-09-23 Macromed, Incorporated. Biodegradable triblock copolymers as solubilizing agents for drugs and method of use thereof
US7649023B2 (en) * 2002-06-11 2010-01-19 Novartis Ag Biodegradable block copolymeric compositions for drug delivery
US7456254B2 (en) * 2004-04-15 2008-11-25 Alkermes, Inc. Polymer-based sustained release device
KR101040415B1 (en) * 2004-04-15 2011-06-09 알케르메스,인코포레이티드 Polymer-based sustained release device
EP2131815A4 (en) * 2007-03-22 2012-11-07 Alkermes Inc Coacervation process
EP2596802A1 (en) 2011-11-23 2013-05-29 PLS-Design GmbH Pharmaceutical composition for treatment of allergic reactions
EP2674168A1 (en) 2012-06-14 2013-12-18 PLS-Design GmbH Modulation of effector T cell responses by local depletion of complement component C3
EP2674167A1 (en) 2012-06-14 2013-12-18 PLS-Design GmbH Controlled activation of complement components for use as endogenous adjuvant
EP2746396A1 (en) 2012-12-20 2014-06-25 PLS-Design GmbH Selective local inhibition of TNFR1-mediated functions at the site of antigen/allergen presentation
JP6143286B2 (en) * 2013-05-13 2017-06-07 学校法人 関西大学 Anti-adhesion material and method for producing the same
US10668017B2 (en) * 2016-08-15 2020-06-02 Wisconsin Alumni Research Foundation Perivascular drug delivery system
WO2018041981A1 (en) * 2016-08-31 2018-03-08 Institut National De La Sante Et De La Recherche Medicale (Inserm) Immunomodulation after locoregional anti-tumoral treament
JP6403297B2 (en) * 2017-05-02 2018-10-10 川澄化学工業株式会社 Method for producing anti-adhesion material

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0091539B2 (en) * 1982-03-31 1996-11-27 Ajinomoto Co., Inc. Gene coding for interleukin-2 polypeptide, recombinant DNA carrying said gene, cell lines possessing the recombinant DNA,and method for producing interleukin-2 using said cells
US5102872A (en) * 1985-09-20 1992-04-07 Cetus Corporation Controlled-release formulations of interleukin-2
US4863726A (en) * 1987-05-29 1989-09-05 Cetus Corporation Combination therapy using immunotoxins with interleukin-2
DE68927679T2 (en) * 1988-10-27 1997-06-26 Univ Minnesota Immunosuppressants from liposomes containing lymphokine IL -2
ATE174514T1 (en) * 1989-01-23 1999-01-15 Chiron Corp RECOMBINANT CELLS FOR THERAPY OF INFECTIONS AND HYPERPRODELIVERY DISORDERS AND THEIR PRODUCTION
US5830452A (en) * 1990-11-20 1998-11-03 Chiron Corporation Method for enhancing the anti-tumor therapeutic index of interleukin-2
US5681562A (en) * 1991-06-25 1997-10-28 Sidney Kimmel Cancer Center Lymphokine gene therapy of cancer
NZ244306A (en) * 1991-09-30 1995-07-26 Boehringer Ingelheim Int Composition for introducing nucleic acid complexes into eucaryotic cells, complex containing nucleic acid and endosomolytic agent, peptide with endosomolytic domain and nucleic acid binding domain and preparation
US5419900A (en) * 1993-05-19 1995-05-30 The United States Of America As Represented By The Department Of Of Health And Human Services Immunologic enhancement with intermittent interleukin-2 therapy
US5631236A (en) * 1993-08-26 1997-05-20 Baylor College Of Medicine Gene therapy for solid tumors, using a DNA sequence encoding HSV-Tk or VZV-Tk
US6156305A (en) * 1994-07-08 2000-12-05 Baxter International Inc. Implanted tumor cells for the prevention and treatment of cancer
US5626862A (en) * 1994-08-02 1997-05-06 Massachusetts Institute Of Technology Controlled local delivery of chemotherapeutic agents for treating solid tumors
GB9506466D0 (en) * 1994-08-26 1995-05-17 Prolifix Ltd Cell cycle regulated repressor and dna element
US5641665A (en) * 1994-11-28 1997-06-24 Vical Incorporated Plasmids suitable for IL-2 expression
US5837233A (en) * 1995-03-17 1998-11-17 University Of California Method for treating tumors
US6045788A (en) * 1996-02-28 2000-04-04 Cornell Research Foundation, Inc. Method of stimulation of immune response with low doses of IL-2
US6034072A (en) * 1997-02-10 2000-03-07 Genemedicine, Inc. IL-2 gene expression and delivery systems and uses
PT1034207E (en) * 1997-10-03 2005-07-29 Macromed Inc COPOLYMERS OF POLY (LACTIDO-CO-GLYCOLIDES) AND POLYETHYLENE GLYCOLIDES, OF LOW MOLECULAR WEIGHT, BIODEGRADABLE, WITH PROPERTIES OF REVERSE THERMAL GELIFICATION
US6004573A (en) * 1997-10-03 1999-12-21 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
KR20010053259A (en) * 1998-06-30 2001-06-25 스티븐 엠. 오드레 Thermosensitive biodegradable hydrogels for sustained delivery of biologically active agents
US6998137B2 (en) * 2000-04-07 2006-02-14 Macromed, Inc. Proteins deposited onto sparingly soluble biocompatible particles for controlled protein release into a biological environment from a polymer matrix
US20030228366A1 (en) * 2002-06-11 2003-12-11 Chung Shih Reconstitutable compositions of biodegradable block copolymers

Also Published As

Publication number Publication date
AU2002312499B8 (en) 2007-08-30
KR20040052510A (en) 2004-06-23
CN1610694A (en) 2005-04-27
EP1446423A4 (en) 2010-07-21
WO2002102309A2 (en) 2002-12-27
EP1446423A2 (en) 2004-08-18
WO2002102309A3 (en) 2004-06-17
AU2002312499A1 (en) 2003-01-02
AU2002312499B2 (en) 2007-04-19
CA2449288C (en) 2018-01-02
JP2004538271A (en) 2004-12-24
US20030003074A1 (en) 2003-01-02

Similar Documents

Publication Publication Date Title
CA2449288C (en) Formulations of lymphokines and method of use thereof for local or both local and systemic control of proliferative cell disorders
JP4903256B2 (en) Biodegradable low molecular weight triblock poly (lactide-co-glycolide) -polyethylene glycol copolymer with inverse thermal gelation properties
Packhaeuser et al. In situ forming parenteral drug delivery systems: an overview
US6004573A (en) Biodegradable low molecular weight triblock poly(lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
KR100754550B1 (en) Biodegradable mixtures of triblock polyesterpolyethylene glycol copolymers
US5098702A (en) Combination therapy using interleukin-2 and tumor necrosis factor
CN1161396C (en) Biodegradable low molecular weight triblock polyester polyethylene glocol copolymers having reverse thermal gelation properties
US6117949A (en) Biodegradable low molecular weight triblock poly (lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US4863727A (en) Combination therapy using interleukin-2 and tumor necrosis factor
TWI221777B (en) Polyol/oil suspensions for the sustained release of proteins
Shaker et al. Interleukin-2: evaluation of routes of administration and current delivery systems in cancer therapy
US5425940A (en) Combination therapy using interleukin-2 and tumor necrosis factor
JP2008510004A (en) Biodegradable diblock copolymer having inverse temperature responsive gelling properties and method of use thereof
US20090202642A1 (en) Drug Delivery System Comprising Microparticles and Gelation System
JP3283288B2 (en) Bioactive peptide preparation
Thomas et al. Microparticulate formulations for the controlled release of interleukin-2
US20060159657A1 (en) Formulations of lymphokines and method of use thereof for local or both local and systemic control of proliferative cell disorders
TWI308068B (en)
US11723956B2 (en) Immunomodulation after locoregional anti-tumoral treatment
Beniers et al. In vivo antiproliferative effects of gamma-interferon and tumor necrosis factor alpha in a rat renal cell carcinoma model system
MXPA00003133A (en) BIODEGRADABLE LOW MOLECULAR WEIGHT TRIBLOCK POLY(LACTIDE-co-GLYCOLIDE) POLYETHYLENE GLYCOL COPOLYMERS HAVING REVERSE THERMAL GELATION PROPERTIES

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20200831