US20020072489A1 - Angiogenically effective unit dose of FGF-2 and method of use - Google Patents

Angiogenically effective unit dose of FGF-2 and method of use Download PDF

Info

Publication number
US20020072489A1
US20020072489A1 US09/771,302 US77130201A US2002072489A1 US 20020072489 A1 US20020072489 A1 US 20020072489A1 US 77130201 A US77130201 A US 77130201A US 2002072489 A1 US2002072489 A1 US 2002072489A1
Authority
US
United States
Prior art keywords
unit dose
fgf
mutein
rfgf
active fragment
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.)
Abandoned
Application number
US09/771,302
Other languages
English (en)
Inventor
Martha Whitehouse
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.)
Novartis Vaccines and Diagnostics Inc
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
Priority claimed from US09/385,114 external-priority patent/US6440934B1/en
Application filed by Individual filed Critical Individual
Priority to US09/771,302 priority Critical patent/US20020072489A1/en
Priority to EP02713467A priority patent/EP1361887A2/en
Priority to JP2002559054A priority patent/JP2004533411A/ja
Priority to AU2002245315A priority patent/AU2002245315A1/en
Priority to PCT/US2002/002239 priority patent/WO2002058720A2/en
Assigned to CHIRON CORPORATION reassignment CHIRON CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WHITEHOUSE, MARTHA JO
Publication of US20020072489A1 publication Critical patent/US20020072489A1/en
Priority to US10/862,152 priority patent/US7112560B2/en
Priority to US11/511,028 priority patent/US7541333B2/en
Priority to JP2008235760A priority patent/JP2009024026A/ja
Assigned to NOVARTIS VACCINES AND DIAGNOSTICS, INC. reassignment NOVARTIS VACCINES AND DIAGNOSTICS, INC. MERGER (SEE DOCUMENT FOR DETAILS). Assignors: CHIRON CORPORATION, NOVARTIS BIOTECH PARTNERSHIP, INC.
Priority to JP2009277091A priority patent/JP2010053145A/ja
Priority to JP2009277090A priority patent/JP2010053144A/ja
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/726Glycosaminoglycans, i.e. mucopolysaccharides
    • A61K31/727Heparin; Heparan
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • A61K38/18Growth factors; Growth regulators
    • 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
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • 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
    • A61K38/22Hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers

Definitions

  • the present invention is directed to a unit dose composition for inducing cardiac angiogenesis in a human comprising a therapeutically effective amount FGF-2 or an angiogenically active fragment or mutein thereof.
  • the present invention is also directed to a method for administering a single unit dose composition to a human to induce cardiac angiogenesis while minimizing systemic risk to the patient.
  • the present invention is useful because the disclosed unit dose composition, and method for its administration, provide an alternative to angioplasty or surgical intervention for the treatment of coronary artery disease (CAD) and further provide an adjunct for reducing post myocardial infarct (MI) injury in humans.
  • CAD coronary artery disease
  • MI post myocardial infarct
  • the fibroblast growth factors are a family of at least eighteen structurally related polypeptides (named FGF-1 to FGF-23) that are characterized by a high degree of affinity for proteoglycans, such as heparin.
  • the various FGF molecules range in size from 15 to at least 32 kD, and exhibit a broad range of biological activities in normal and malignant conditions including nerve cell adhesion and differentiation [Schubert et al., J. Cell Biol. 104:635-643 (1987)]; wound healing [U.S. Pat. No.
  • the FGF family is a family of pluripotent growth factors that stimulate to varying extents fibroblasts, smooth muscle cells, epithelial cells and neuronal cells.
  • FGF When FGF is released by normal tissues, such as in fetal development or wound healing, it is subject to temporal and spatial controls. However, many of the members of the FGF family are also oncogenes. Thus, in the absence of temporal and spatial controls, they have the potential to stimulate tumor growth while providing angiogenesis.
  • Coronary artery disease is a progressive condition in humans wherein one or more coronary arteries gradually become occluded through the buildup of plaque (atherosclerosis).
  • the coronary arteries of patients having this disease are often treated by balloon angioplasty or the insertion of stents to prop open the partially occluded arteries.
  • many of these patients are required to undergo coronary artery bypass surgery at great expense and risk. It would be desirable to provide such patients with a medicament that would enhance coronary blood flow so as to reduce the need to undergo bypass surgery.
  • Battler et al. who administered bovine bFGF (i.e., FGF-2) to pigs in a myocardial infarct model, considered the varying results that are obtained from one animal species to another, and expressly discloses that the divergent results “thus emphasiz[e] the caution that must be exercised in extrapolating results from different animal models.” Battler et al., at page 2005, col.1. Further, Battler points out that “the dosage and mode of administration of bFGF [i.e., bovine FGF-2] may have profound implications for the biologic effect achieved.” Battler, et al., at page 2005, col.1.
  • Applicants have discovered that administering a single unit dose of about 0.2 ⁇ g/kg to about 48 ⁇ g/kg of rFGF-2 or an angiogenically active fragment or mutein thereof into one or more coronary vessels (IC) or a peripheral vein (IV) of a human patient in need of coronary angiogenesis, unexpectedly provided the human patient with a rapid and therapeutic coronary angiogenesis that resulted in an unexpectedly large increase (i.e., 96 and 100 seconds of increase in the mean change from baseline for all groups at 2 and 6 months) in the treated patient's exercise tolerance time (ETT) that persisted for an unexpectedly long duration (i.e., 6 months as of this writing).
  • ETT exercise tolerance time
  • coronary angiogenesis is meant the formation of new blood vessels, ranging in size from capillaries to arterioles which act as collaterals in coronary circulation.
  • angioplasty is considered a therapeutic success if it provides an increase in a patient's ETT of greater than 30 seconds compared to the placebo.
  • the invention is directed to a unit dose of rFGF-2 comprising a safe and therapeutically effective amount of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • the safe and therapeutically effective amount comprises about 0.2 ⁇ g/kg to about 48 ⁇ g/kg of rFGF-2 or an angiogenically active fragment or mutein thereof, based upon ideal body weight.
  • the safe and therapeutically effective amount of the unit dose comprises 0.2 ⁇ g/kg to 2.0 ⁇ g/kg, greater than 2.0 ⁇ g/kg to less than 24 ⁇ g/kg, or 24 ⁇ g/kg to 48 ⁇ g/kg IC of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • the safe and therapeutically effective amount of the unit dose comprises 18 ⁇ g/kg to 36 ⁇ g/kg IV of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • the unit dose of the present invention comprises 0.008 mg to 7.2 mg, more typically 0.3 mg to 3.5 mg, of FGF-2 or an angiogenically active fragment or mutein thereof.
  • a suitable FGF-2 is the rFGF-2 of SEQ ID NO: 2 or an angiogenically active fragment or mutein thereof.
  • the present invention is directed to a method of treating a human patient for CAD or to induce coronary angiogenesis therein.
  • the method comprises administering into one or more coronary vessels or a peripheral vein of a human patient in need of treatment for coronary artery disease (or in need of angiogenesis) a safe and therapeutically effective amount of a recombinant FGF-2 (rFGF-2) or an angiogenically active fragment or mutein thereof.
  • rFGF-2 recombinant FGF-2
  • angiogenically active fragment or mutein thereof typically, a portion of the safe and therapeutically effective amount is administered to each of 2 coronary vessels.
  • the safe and therapeutically effective amount comprises about 0.2 ⁇ g/kg to about 48 ⁇ g/kg, of rFGF-2 or an angiogenically active fragment or mutein thereof in a pharmaceutically acceptable carrier. In other embodiments, the safe and therapeutically effective amount comprises 0.2 ⁇ g/kg to 2 ⁇ g/kg, >2 ⁇ g/kg to ⁇ 24 ⁇ g/kg, or 24 ⁇ g/kg to 48 ⁇ g/kg of rFGF-2 an angiogenically active fragment or mutein thereof in a pharmaceutically acceptable carrier.
  • the amount of rFGF-2 or angiogenically active fragment or mutein thereof that is used in the above method comprises 0.008 mg to 7.2 mg, more typically 0.3 mg to 3.5 mg, of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • FGF-2 is a glycosoaminoglycan (e.g., heparin) binding protein and the presence of a glycosoaminoglycan optimizes activity and AUC (see FIGS. 3 and 4)
  • the IC dosages of RFGF-2 of the present invention typically are administered from 0-30 minutes prior to the administration of a glycosoaminoglycan, such as a heparin.
  • the heparin is administered IC or IV, typically IV.
  • the heparin is combined with the unit dose composition.
  • rFGF-2 releases nitric oxide, a potent vasodilator, aggressive fluid management prior to (proactively) and during the infusion is critical to patient's safety.
  • Administration of IV fluids e.g., 500-1000 mL of normal saline
  • administration of boluses of IV fluids e.g., 200 mL normal saline
  • decreases of systolic blood pressure e.g., ⁇ 90 mm Hg
  • EDTA is a potent chelator of calcium which is required for normal myocardial contraction and cardiac conduction, minimizing the concentration of EDTA is critical to patient's safety.
  • a concentration of EDTA less than 100 ⁇ g/ml in the unit dose composition optimized the safety of administration of rFGF-2 by IC or IV infusion to human patients.
  • the mean score for exertional capacity increased by 10.9 to 20.2 at 2 months; and by 16.5 to 24.1 at 6 months.
  • the mean score increased by 32.1 to 46.2 at 2 months; and by 16.7 to 23.2 at 6 months.
  • the mean score increased by 20.0 to 32.9 at 2 months; and by 11.4 to 36.7 at 6 months.
  • the mean score increased by 8.5 to 19.8 at 2 months; and by 6.3 to 19.8 at 6 months.
  • the mean score increased by 20.2 to 27.8 at 2 months; and by 23.8 to 34.0 at 6 months.
  • a change of 8 points on any scale is considered clinically significant.
  • MRI was also performed on 33 human patients diagnosed with CAD to assess the effect of administering a single unit dose of rFGF-2 on their cardiac ejection fraction, regional myocardial function and perfusion (delayed arrival zone). Specifically, the patients were administered a single unit dose of 0.33 ⁇ g/kg to 48 ⁇ g/kg IC or 18 ⁇ g/kg to 36 ⁇ g/kg IV of rFGF-2 of SEQ ID NO: 2.
  • the 33 human CAD patients were assessed by resting cardiac magnetic resonance imaging (MRI) at baseline (i.e., prior to treatment), and 1, 2 and 6 months after treatment with a single unit dose of rFGF-2 of the invention by IC or IV routes, the patients exhibited a highly statistically significant response to the method of treatment as objectively measured by increased target wall thickening, target wall motion, and target area collateral extent, and by decreased target area delayed arrival extent.
  • MRI cardiac magnetic resonance imaging
  • FIG. 1A is a plot of the mean rFGF-2 plasma concentration versus time profiles for eight different doses of rFGF-2 (SEQ ID NO: 2) administered by IC infusion in humans over a 20 minute period.
  • the eight doses of rFGF-2 presented in FIG. 1A are 0.33, 0.65, 2, 6, 12, 24, 36, and 48 ⁇ g/kg of lean body mass (LBM).
  • FIG. 1B is a plot of the mean FGF-2 plasma concentration versus time profiles for 2 different doses of rFGF-2 (SEQ ID NO: 2) administered by IV infusion in humans over a 20 minute period.
  • the 2 IV doses of rFGF-2 in FIG. 1B are 18 and 36 ⁇ g/kg.
  • the mean concentration-time profile following IC administration of 36 ⁇ g/kg rFGF-2 is included for comparison.
  • FIG. 2 is a plot of mean FGF-2 area under the curve (AUC) in pg*min/ml corresponding to FIGS. 1A and 1B. This plot shows the dose linearity of systemic rFGF-2 exposure following IC or IV infusion. The systemic exposure for the IC route is similar to that observed following IV administration.
  • FIG. 3 is a plot of individual human patient FGF-2 plasma clearance (CL) values as a function of the time of heparin administration in “minutes prior to rFGF-2 infusion” and shows the influence of timing of heparin administration on rFGF-2 plasma clearance (CL).
  • FIG. 4 is a plot individual human patient FGF-2 dose normalized area under curves (AUCs) as a function of the time of heparin administration in “minutes prior to rFGF-2 infusion” and shows the influence of timing of heparin administration on FGF-2 AUC.
  • AUCs area under curves
  • FIG. 5 summarizes the analysis plan for the Phase II Clinical Trial.
  • FIG. 6 summarizes patient characteristics for the patient population in the Phase II Clinical Trial.
  • FIG. 7 shows patient disposition over the course of the Phase II Clinical Trail.
  • FIG. 8 shows the safety variables for the patient population for the Phase II Clinical Trial.
  • FIG. 9 depicts change in exercise time for the placebo, and 3 FGF-2 treatment groups.
  • FIG. 10 shows the change in angina frequency score for patients in placebo and the three treatment groups.
  • FIG. 11 shows the change in other domains of the Seattle Angina Questionnaire for patients in placebo and the three treatment groups.
  • FIG. 12 shows change in Short Form-36 physical component summary score.
  • FIG. 13 shows change in ETT and angina frequency score stratified by baseline CCS Class 3 or 4.
  • FIG. 14 shows the change in ETT and angina frequency score stratified by baseline AFS less than or equal to 40.
  • rFGF-2 or an angiogenically active fragment or mutein thereof when administered in a safe and therapeutically effective amount into one or more coronary vessels or into a peripheral vein of a human patient diagnosed with CAD provides the patient with a safe and therapeutically efficacious treatment for the patient's coronary artery disease that lasts at least 4 to 6 months, more typically at least 2 months, before a further treatment is needed.
  • This duration of the effect and the magnitude of the improvements in ETT, SAQ and MRI were unexpected for a single unit dose of medicament.
  • rFGF-2 an amount of rFGF-2 or an angiogenically active fragment or mutein thereof that when administered in accordance with this invention, is free from major complications that cannot be medically managed, and that provides for objective cardiac improvement in patients having symptoms of CAD despite optimum medical management.
  • acute hypotension that can be managed by administration of fluids, is considered “safe” for the purpose of this invention.
  • the safe and therapeutically effective amount of rFGF-2 comprises about 0.2 ⁇ g/kg to about 48 ⁇ g/kg of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • a suitable FGF-2 for use in the present invention is the rFGF-2 of SEQ ID NO: 2 or an angiogenically active fragment or mutein thereof.
  • the present invention has multiple aspects.
  • the present invention is directed to a unit dose composition for inducing angiogenesis in a human patient, the unit dose comprising a therapeutically effective (i.e., an angiogenically effective) amount of rFGF-2 or an angiogenically active fragment or mutein thereof, the amount comprising about 0.2 ⁇ g/kg to about 48 ⁇ g/kg of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • a safe and therapeutically effective amount comprises about 0.2 ⁇ g/kg to about 2 ⁇ g/kg, about 2 ⁇ g/kg to about 24 ⁇ g/kg, or about 24 ⁇ g/kg to about 48 ⁇ g/kg of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • the unit dose composition of the present invention comprises from 0.008 mg to 7.2 mg of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • the unit dose composition contains a sufficient amount of FGF-2 to accommodate dosing any one of the majority of human CAD patients, ranging from the smallest patient (e.g., 40 kg) at the lowest dosage (about 0.2 ⁇ g/kg) through the larger patients (e.g., 150 kg) at the highest dosage (about 48 ⁇ g/kg).
  • the unit dose comprises 0.3 mg to 3.5 mg of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • the unit dose composition is typically provided in solution or lyophilized form containing the above referenced amount of rFGF-2 and an effective amount of one or more pharmaceutically acceptable buffers, stabilizers and/or other excipients as later described herein.
  • the active agent in the above described unit dose composition is a recombinant FGF-2 or an angiogenically active fragment or mutein thereof.
  • Methods for making recombinant FGF-2 are well-known in the art.
  • the recombinant FGF-2 of SEQ ID NO: 2 is made as described in U.S. Pat. No. 5,155,214, entitled “Basic Fibroblast Growth Factor,” which issued on Oct. 13, 1992, and which is expressly incorporated herein by reference in its entirety.
  • all other references cited herein, whether occurring before or after this sentence, are expressly incorporated herein by reference in their entirety.
  • a DNA of SEQ ID NO: 1, which encodes a bFGF (hereinafter “FGF-2”) of SEQ ID NO: 2 is inserted into a cloning vector, such as pBR322, pMB9, Col E1, pCRI, RP4 or ⁇ -phage, and the cloning vector is used to transform either a eukaryotic or prokaryotic cell, wherein the transformed cell expresses the FGF-2.
  • the host cell is a yeast cell, such as Saccharomyces cerevisiae.
  • the resulting full-length FGF-2 that is expressed has 146 amino acids in accordance with SEQ ID NO: 2.
  • FGF-2 of SEQ ID NO: 2 has four cysteines, i.e., at residue positions 25, 69, 87 and 92, there are no internal disulfide linkages. ['214 at col. 6, lines 59-61.] However, in the event that cross-linking occurred under oxidative conditions, it would likely occur between the residues at positions 25 and 69.
  • the FGF-2 of SEQ ID NO: 2 which has 146 amino acid residues, differs from naturally occurring human FGF-2 by only 2 amino acid residue.
  • the amino acids at residue positions 112 and 128 of the FGF-2 of SEQ ID NO: 2 are Ser and Pro, respectively, whereas in human FGF-2, they are Thr and Ser, respectively.
  • bovine FGF-2 like the corresponding human FGF-2 is initially synthesized in vivo as a polypeptide having 155 amino acid residues. Abraham et al. “ Human Basic Fibroblast Growth Factor: Nucleotide Sequence and Genomic Organization,” EMBO J., 5(10):2523-2528 (1986).
  • the FGF-2 of SEQ ID NO: 2 When the FGF-2 of SEQ ID NO: 2 is compared to the full length 155 residue bovine FGF-2 of Abraham, the FGF-2 of SEQ ID NO: 2 lacks the first nine amino acid residues, Met Ala Ala Gly Ser Ile Thr Thr Leu (SEQ ID NO: 3), at the N-terminus of the corresponding full length molecule.
  • the recombinant FGF-2 employed in the present compositions and method was purified to pharmaceutical quality (98% or greater purity) using the techniques described in detail in U.S. Pat. No. 4,956,455, entitled “Bovine Fibroblast Growth Factor” which issued on Sep. 11, 1990 and which is incorporated herein by reference in its entirety.
  • the first 2 steps employed in the purification of the recombinant FGF-2 of Applicants' unit dose composition are “conventional ion-exchange and reverse phase HPLC purification steps as described previously.”
  • the third step, which the '455 patent refers to as the “key purification step” ['455 at col. 7, lines 5-6] is heparin-SEPHAROSE® affinity chromatography, wherein the strong heparin binding affinity of the FGF-2 is utilized to achieve several thousand-fold purification when eluting at approximately 1.4M and 1.95M NaCl ['455 at col. 9, lines 20-25].
  • Polypeptide homogeneity was confirmed by reverse-phase high pressure liquid chromatography (RP-HPLC). Buffer exchange was achieved by SEPHADEX® G-25(M) gel filtration chromatography.
  • the active agent in the unit dose of the present invention also comprises an “angiogenically active fragment” of FGF-2.
  • angiogenically active fragment of FGF-2 is meant a fragment of FGF-2 that has about 80% of the 146 residues of SEQ ID NO: 2 and that retains at least 50%, preferably at least 80%, of the angiogenic activity of the FGF-2 of SEQ ID NO: 2.
  • the FGF-2 fragment should have two cell binding sites and at least one of the two heparin binding sites.
  • the two putative cell binding sites of the analogous human FGF-2 occur at residue positions 36-39 and 77-81 thereof. See Yoshida, et al., “ Genomic Sequence of hst, a Transforming Gene Encoding a Protein Homologous to Fibroblast Growth Factors and the int -2- Encoded Protein ,” PNAS USA, 84:7305-7309 (October 1987) at FIG. 3.
  • the two putative heparin binding sites of hFGF-2 occur at residue positions 18-22 and 107-111 thereof See Yoshida (1987) at FIG. 3.
  • the angiogenically active fragments of FGF-2 typically encompass those terminally truncated fragments of FGF-2 that have at least residues that correspond to residues 30-110 of FGF-2 of SEQ ID NO: 2; more typically, at least residues that correspond to residues 18-146 of FGF-2 of SEQ ID NO: 2.
  • the unit dose of the present invention also comprises an “angiogenically active . . . mutein” of the rFGF-2 of SEQ ID NO: 2.
  • angiogenically active . . . mutein as used herein, is meant an isolated and purified recombinant protein or polypeptide that has 65% sequence identity (homology) to any naturally occurring FGF-2, as determined by the Smith-Waterman homology search algorithm ( Meth. Mol. Biol.
  • the angiogenically active mutein has at least 75%, more preferably at least 85%, and most preferably, at least 90% sequence identity to the naturally occurring FGF-2.
  • GAP Genetics Computing Group
  • BESTFIT Altschul et al.
  • BLAST Altschul et al.
  • FASTA Altschul et al.
  • TFASTA et al.
  • GAP Genetics Computing Group
  • CLUSTAL CLUSTAL in the PC/Gene program by Intellegenetics, Mountain View Calif.
  • the percentage of sequence identity is determined by using the default parameters determined by the program.
  • sequence identity is intended to refer to the percentage of the same amino acids that are found similarly positioned within the mutein sequence when a specified, contiguous segment of the amino acid sequence of the mutein is aligned and compared to the amino acid sequence of the naturally occurring FGF-2.
  • an “angiogenically active mutein” of an angiogenic agent of the present invention uses standard techniques for site directed mutagenesis, as known in the art and/or as taught in Gilman, et al., Gene, 8:81 (1979) or Roberts, et al., Nature, 328:731 (1987).
  • site directed mutagenesis techniques one or more point mutations are introduced into the cDNA sequence of SEQ ID NO: 1 to introduce one or more amino acid substitutions or an internal deletion.
  • Conservative amino acid substitutions are those that preserve the general charge, hydrophobicity/hydrophilicity, and/or steric bulk of the amino acid being substituted.
  • substitutions between the following groups are conservative: Gly/Ala, Val/Ile/Leu, Lys/Arg, Asn/Gln, Glu/Asp, Ser/Cys/Thr, and Phe/Trp/Tyr.
  • Significant (up to 35%) variation from the sequence of the naturally occurring angiogenic FGF-2 is permitted as long as the resulting protein or polypeptide retains angiogenic activity within the limits specified above.
  • Cysteine-depleted muteins are muteins within the scope of the present invention. These muteins are constructed using site directed mutagenesis as described above, or according to the method described in U.S. Pat. No. 4,959,314 (“the '314 patent”), entitled “Cysteine-Depleted Muteins of Biologically Active Proteins.”
  • the '314 patent discloses how to determine biological activity and the effect of the substitution. Cysteine substitution is particularly useful in proteins having 2 or more cysteines that are not involved in disulfide formation. Suitable substitutions include the substitution of serine for one or both of the cysteines at residue positions 87 and 92, which are not involved in disulfide formation. Preferably, substitutions are introduced at the FGF-2 N-terminus, which is not associated with angiogenic activity. However, as discussed above, conservative substitutions are suitable for introduction throughout the molecule.
  • the unit dose composition of the present invention comprises a safe and an angiogenically effective dose of rFGF-2 or an angiogenically active fragment or mutein thereof, and a pharmaceutically acceptable carrier.
  • the safe and angiogenically effective dose of the pharmaceutical composition of the present invention is in a form and a size suitable for administration to a human patient and comprises (i) 0.2 ⁇ g/kg to 48 ⁇ g/kg of rFGF-2 or an angiogenically active fragment or mutein thereof, (ii) and a pharmaceutically acceptable carrier.
  • the safe and angiogenically effective dose comprises 0.2 ⁇ g/kg to 2 ⁇ g/kg, >2 ⁇ g/kg to ⁇ 24 ⁇ g/kg or 24 ⁇ g/kg to 48 ⁇ g/kg of FGF-2 or an angiogenically active fragment or mutein thereof, and a pharmaceutically acceptable carrier.
  • the unit dose of the present invention comprises 0.008 mg to 7.2 mg, more typically 0.3 mg to 3.5 mg, of the FGF-2 or an angiogenically active fragment or mutein thereof.
  • the second recited component of the unit dose composition of the present invention is a “pharmaceutically acceptable carrier.”
  • pharmaceutically acceptable carrier any of the carriers or diluents that are well-known in the art for the stabilization and/or administration of a proteinaceous medicament that does not itself induce the production of antibodies harmful to the patient receiving the composition, and which may be administered without undue toxicity.
  • the choice of the pharmaceutically acceptable carrier and its subsequent processing enables the unit dose composition of the present invention to be provided in either liquid or solid form.
  • the pharmaceutically acceptable carrier comprises a stable carrier or diluent suitable for intravenous (“IV”) or intracoronary (“IC”) injection or infusion.
  • IV intravenous
  • IC intracoronary
  • Suitable carriers or diluents for injectable or infusible solutions are nontoxic to a human recipient at the dosages and concentrations employed, and include sterile water, sugar solutions, saline solutions, protein solutions or combinations thereof.
  • the pharmaceutically acceptable carrier includes a buffer and one or more stabilizers, reducing agents, anti-oxidants and/or anti-oxidant chelating agents.
  • buffers, stabilizers, reducing agents, anti-oxidants and chelating agents in the preparation of protein based compositions, particularly pharmaceutical compositions, is well-known in the art. See, Wang et al., “ Review of Excipients and pHs for Parenteral Products Used in the United States,” J. Parent. Drug Assn., 34(6):452462 (1980); Wang et al., “ Parenteral Formulations of Proteins and Peptides: Stability and Stabilizers,” J. Parent. Sci.
  • Suitable buffers include acetate, adipate, benzoate, citrate, lactate, maleate, phosphate, tartarate and the salts of various amino acids. See Wang (1980) at page 455.
  • Suitable stabilizers include carbohydrates such as threlose or glycerol.
  • Suitable reducing agents which maintain the reduction of reduced cysteines, include dithiothreitol (DTT also known as Cleland's reagent) or dithioerythritol at 0.01% to 0.1% wt/wt; acetylcysteine or cysteine at 0.1% to 0.5% (pH 2-3); and thioglycerol at 0.1% to 0.5% (pH 3.5 to 7.0) and glutathione.
  • DTT dithiothreitol
  • acetylcysteine or cysteine at 0.1% to 0.5%
  • thioglycerol at 0.1% to 0.5% (pH 3.5 to 7.0) and glutathi
  • Suitable antioxidants include sodium bisulfite, sodium sulfite, sodium metabisulfite, sodium thiosulfate, sodium formaldehyde sulfoxylate, and ascorbic acid. See Akers (1988) at pages 225.
  • Suitable chelating agents which chelate trace metals to prevent the trace metal catalyzed oxidation of reduced cysteines, include citrate, tartarate, ethylenediaminetetraacetic acid (EDTA) in its disodium, tetrasodium, and calcium disodium salts, and diethylenetriamine pentaacetic acid (DTPA).
  • Suitable sugars include glycerol, trehalose, glucose, galactose and mannitol, sorbitol.
  • a suitable protein is human serum albumin.
  • a typical unit dose composition of the present invention comprises from about 0.001 mg to 8 mg, more typically 0.03 to 5 mg rFGF-2 or an angiogenically active fragment or mutein thereof, dissolved a pharmaceutically acceptable carrier.
  • a suitable pharmaceutically acceptable carrier comprises 10 mM thioglycerol, 135 mM NaCl, 10 mM sodium citrate, and 1 mM EDTA, pH 5.
  • a suitable diluent or flushing agent for the above-described unit dose composition is any of the above-described carriers. Typically, the diluent is the carrier solution.
  • rFGF-2 or an angiogenically active fragment or mutein thereof is unstable for long periods of time in liquid form.
  • the unit dose composition should be stored frozen at ⁇ 60° C. When thawed, the solution is stable for 6 months at refrigerated conditions.
  • a typical unit dose would comprise about 1-40 ml, more typically 10-40 ml, of the above-described composition having 0.008-7.2 mg of rFGF-2 or an angiogenically active fragment or mutein dissolved therein.
  • a suitable rFGF-2 for use in the unit dose is the rFGF-2 of SEQ ID NO: 2 or an angiogenically active fragment or mutein thereof.
  • the unit dose composition is provided in lyophilized (freeze-dried) form.
  • the unit dose of rFGF-2 is capable of being stored at refrigerated temperatures for substantially longer than 6 months without loss of therapeutic effectiveness. Lyophilization is accomplished by the rapid freeze drying (i.e., removing water) under reduced pressure of a plurality of vials, each containing the above described liquid form of the unit dose of the rFGF-2 of the present invention therein. Lyophilizers, which perform the above described lyophilization, are commercially available and readily operable by those skilled in the art.
  • the resulting lyophilized unit dose composition in lyophilized cake form, is formulated to contain within the resulting lyophilized cake one or more of the buffers, stabilizers, anti-oxidants, reducing agents, salts and/or sugars described above for the corresponding liquid formulation.
  • a lyophilized unit dose composition containing all such other components need only be reconstituted to a known volume or concentration with sterile aqueous diluent such as sterile water, a sterile sugar solution, or a sterile saline solution. Alternatively, it could be reconstituted with a sterile buffer solution as described above, but lacking a chelating agent, such as EDTA.
  • the unit dose composition is stable from 6 months to 2 years at refrigerated temperatures. Thus, storage of the unit dose composition in lyophilized form is readily accommodated using conventional refrigeration equipment.
  • the unit dose composition of the present invention is administered via a cardiac catheter or other injection device, which has dead space
  • the vial is optionally formulated to contain up to 50% extra (e.g., a total of about 10.8 mg) of rFGF-2 or angiogenically active fragment or mutein thereof.
  • the extra solution is suitable for filling the dead space in the delivery equipment.
  • the pharmaceutical composition is loaded in the cardiac catheter in front of a pharmaceutically acceptable buffer, diluent or carrier, which is then used to deliver the appropriate amount of the one or more dosages to the one or more sites in the myocardium that are in need of angiogenesis.
  • the pharmaceutically acceptable carrier for the above described unit dose composition comprises a buffer and one or more stabilizers, reducing agents, anti-oxidants and/or anti-oxidant chelating agents. It is also within the scope of the present invention that the unit dose composition contain an amount of a glycosoaminoglycan (also known as a “proteoglycan” or a “mucopolysaccharide”), such as heparin, that is effective to bind to the FGF-2 and to the endothelial cell receptors so as to enhance the angiogenic effectiveness of the FGF-2 or angiogenically active fragment or mutein thereof.
  • a glycosoaminoglycan also known as a “proteoglycan” or a “mucopolysaccharide”
  • heparin such as heparin
  • the amount of heparin that is administered is about 10-80 U per kg of patient weight (U/kg), typically about 40 U/kg. Expressed in absolute terms, the total amount of heparin administered to any one patient does not exceed 5,000 U.
  • the unit dose composition of the present invention would not only contain an angiogenically effective amount of rFGF-2 or an angiogenically active fragment or mutein thereof, it would also contain from about 10-80 U/kg of heparin, typically about 40 U/kg.
  • the typical volume of diluent is from about 1 to 40 ml. While larger volumes of diluent could be used, such larger volumes would typically result in longer administration times.
  • a single dose comprising from 0.2 ⁇ g/kg to 48 ⁇ g/kg of the rFGF-2 or an angiogenically active fragment or mutein thereof is withdrawn from the vial as reconstituted product for administration to the patient.
  • an average 70 kg man that is being dosed at 24 ⁇ g/kg would have a sufficient volume of the reconstituted product withdrawn from the vial to receive an IC infusion of (70 kg ⁇ 30 ⁇ g/kg) 2100 ⁇ g (i.e., 2.1 mg).
  • the present invention is directed to a method for treating a human patient for CAD or MI, using the above described unit dose composition.
  • the present invention is directed to a method for treating a human patient for coronary artery disease, comprising administering a safe and therapeutically effective amount of a recombinant FGF-2 or an angiogenically active fragment or mutein thereof to one or more, typically 2, patent coronary vessels or a peripheral vein of a human patient in need of treatment for coronary artery disease.
  • the human patient in need of treatment for coronary artery disease is typically a human patient with coronary artery disease who remains symptomatic with angina despite optional medical management.
  • a preferred coronary vessel is a coronary artery, although grafted saphenous veins and grafted internal mammary arteries, as provided by coronary angioplasty, are also suitable.
  • Suitable peripheral veins for administering the unit dose composition include those peripheral veins found throughout the human body that are routinely used by treating physicians and nurses for administration of fluids and medicaments. Examples of such veins include the cephalic, the median cubital, and the basilic of the arm.
  • the unit dose of rFGF-2 or angiogenic fragment or mutein thereof is typically administered within an hour, more typically over a period of about 20 minutes into one or more (typically, 2) patent coronary vessels.
  • the unit dose composition is typically administered at a rate of 0.5 to 2.0 ml/minute, more typically at about 1 ml/minute.
  • the coronary vessels can be native vessels or grafts, so long as they are not occluded.
  • the volume of the unit dose of rFGF-2 or angiogenic fragment or mutein thereof is typically 1040 ml; more typically 20 ml.
  • the length of time for infusion of the unit dose is not critical and can be shortened or lengthened depending on the rate and volume of infusion.
  • the unit dose of rFGF-2 or angiogenic fragment or mutein thereof is administered typically within an hour, more typically over a 20 minute period, into a peripheral vein using a conventional IV setup.
  • the unit dose composition is typically administered at a rate of 1 ml/minute.
  • the method of the present invention provides treatment of the underlying condition in CAD or MI and not merely transitory relief from the symptoms, such as provided by nitrates.
  • the safe and therapeutically effective amount of the method of the present invention comprises 0.2 ⁇ g/kg to 48 ⁇ g/kg of rFGF-2 or an angiogenically active fragment or mutein thereof in a pharmaceutically acceptable carrier.
  • the safe and therapeutically effective amount comprises 0.2 ⁇ g/kg to 2 ⁇ g/kg, >2 ⁇ g/kg to ⁇ 24 ⁇ g/kg, or 24 ⁇ g/kg to 48 ⁇ g/kg of rFGF-2 or an angiogenically active fragment or mutein thereof in a pharmaceutically acceptable carrier.
  • the safe and therapeutically effective amount is about .008 mg to about 7.2 mg of rFGF-2 or an angiogenically active fragment or mutein thereof; more typically, 0.3 mg to 3.5 mg of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • a suitable rFGF-2 is the rFGF-2 of SEQ ID NO: 2 or an angiogenically active fragment or mutein thereof.
  • the present invention is also directed to a method for inducing angiogenesis in a heart of a human patient comprising, administering a single unit dose composition of a recombinant FGF-2 or an angiogenically active fragment or mutein thereof to one or more coronary vessels or to a peripheral vein in a human patient in need of coronary angiogenesis, said unit dose composition comprising from about 0.008 mg to 7.2 mg of recombinant rFGF-2 or an angiogenically active fragment or mutein thereof in a pharmaceutically acceptable carrier. More typically, the unit dose composition comprises about 0.3-3.5 mg rFGF-2 or an angiogenically active fragment or mutein thereof in a pharmaceutically acceptable carrier.
  • a single unit dose composition containing a therapeutically effective amount of an rFGF-2 or an angiogenically fragment or mutein thereof is administered to at least one coronary vessel of a human patient in need of angiogenesis, using standard cardiac catheterization techniques already known and used in the art for the intracoronary administration of medicaments, e.g., thrombolytics, streptokinase, or radio-opaque dyes or magnetic particles used to visualize the coronary arteries.
  • medicaments e.g., thrombolytics, streptokinase, or radio-opaque dyes or magnetic particles used to visualize the coronary arteries.
  • a coronary catheter is inserted into an artery (e.g., femoral or subclavian) of the patient in need of treatment and the catheter is pushed forward, with visualization, until it is positioned in the appropriate coronary vessel of the patient to be treated.
  • the pharmaceutical composition in solution form is administered by infusing the unit dose substantially continuously over a period of 10 to 30 minutes.
  • the pharmaceutical composition of the invention could be administered over a longer period of time, the Applicants perceive no benefit and a potentially increased risk of thrombosis in doing so.
  • a portion (e.g., one half) of the unit dose is administered in a first coronary vessel.
  • the catheter is repositioned into a second secondary coronary vessel and the remainder of the unit dose is administered with flushing of the catheter.
  • portions of the unit dose may be administered to a plurality of coronary vessels until the entire unit dose has been administered.
  • the present invention is directed to a method for inducing angiogenesis in a heart of a human patient, comprising administering into one or more coronary vessels (IC) or into a peripheral vein (IV) of a human patient in need of coronary angiogenesis, a single unit dose composition comprising an angiogenically effective amount of rFGF-2 or an angiogenically active fragment or mutein thereof in a pharmaceutically acceptable carrier.
  • IC coronary vessels
  • IV peripheral vein
  • the angiogenically effective amount comprises about 0.2 ⁇ g/kg to about 48 ⁇ g/kg (or in absolute terms about 0.008 mg to about 7.2 mg) of a recombinant FGF-2 or an angiogenically active fragment or mutein thereof. More typically, the angiogenically effective amount comprises about 0.3 mg to 3.5 mg of a recombinant FGF-2 or an angiogenically active fragment or mutein thereof.
  • a suitable rFGF-2 for use in the above-identified method is the rFGF-2 of SEQ ID NO: 2 or an angiogenically active fragment thereof.
  • the unit dose composition is administered IC to patent coronary vessels or IV to a peripheral vein. In another embodiment, the unit dose composition is administered with heparin as described herein.
  • the present invention is also directed to a method for treating a human patient for an MI comprising, administering into one or more coronary vessels or into a peripheral vein of said human patient, a single unit dose composition comprising a therapeutically effective amount of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • the unit dose composition typically comprises about 0.2 ⁇ g/kg to about 48 ⁇ g/kg (or in absolute terms about 0.008 mg to about 7.2 mg) of a recombinant FGF-2 or an angiogenically active fragment or mutein thereof in a pharmaceutically acceptable carrier.
  • a suitable rFGF-2 for use in the above-identified method is the rFGF-2 of SEQ ID NO: 2 or an angiogenically active fragment thereof.
  • the present invention is directed to an improved method for treating a patient for unstable angina or acute myocardial infarction, requiring angioplasty, the method comprising providing angioplasty to the patient in need of treatment; the improvement comprising administering into one or more coronary vessels or into a peripheral vein of said human patient, a single unit dose composition comprising a therapeutically effective amount of rFGF-2 or an angiogenically active fragment or mutein thereof.
  • the unit dose composition comprises about 0.2 ⁇ g/kg to about 48 ⁇ g/kg (or in absolute terms about 0.008 mg to about 7.2 mg) of a recombinant FGF-2 or an angiogenically active fragment or mutein thereof in a pharmaceutically acceptable carrier.
  • a suitable rFGF-2 for use in the above-identified method is the rFGF-2 of SEQ ID NO: 2 or an angiogenically active fragment thereof.
  • the rFGF-2 or the angiogenically active fragment or mutein thereof is associated with release of nitric oxide, a recognized smooth muscle dilator, which upon administration to the patient causes a sudden drop in the patient's blood pressure. Accordingly, in the methods of the present invention, it is preferable to hydrate the patient with IV fluids prior to administering the unit dose of the present invention. Moreover, for safety and tolerability of the unit dose, aggressive fluid management during and after rFGF-2 administration is also preferred.
  • glycosoaminoglycan also known as a “proteoglycan” or a “mucopolysaccharide”
  • heparin a glycosoaminoglycan
  • the effective amount of glycosaminoglycan (such as heparin) that is administered is about 10-80 U/kg, more typically, about 40 U/kg.
  • the total amount of heparin administered to any one patient immediately prior to dosing generally does not exceed 5,000 U.
  • EDTA is a potent chelator of calcium, which is required for normal myocardial contraction and cardiac conduction, minimizing the concentration of EDTA is critical to patient's safety.
  • a concentration of EDTA less than 100 ⁇ g/ml optimized the safety of administration of rFGF-2 by IC or IV infusion to human patients.
  • a Phase I clinical trial directed to treating human patients for CAD by administering a single unit dose composition of the present invention was conducted and is described in Examples 1-3 herein.
  • 66 human patients diagnosed with CAD who satisfied the criteria of Example 2 herein, received a single unit dose of rFGF-2 in accordance with the method of the present invention.
  • 52 human patients were administered a unit dose of 0.33 ⁇ g/kg to 48 ⁇ g/kg of rFGF-2 by IC infusion over about a 20 minute period.
  • Fourteen human patients were administered a unit dose of either 18 ⁇ g/kg or 36 ⁇ g/kg of rFGF-2 by IV infusion over about a 20 minute period.
  • the 66 treated patients were then assessed relative to baseline (i.e., prior to treatment with the single unit dose), and again at 1 month, 2 months and 6 months after treatment with the single unit dose, using three sets of art-recognized assessment criteria: 1) changes in their exercise tolerance time (ETT); 2) the Seattle Angina Questionnaire, which provides an assessment based upon a mixed combination of objective and subjective criteria; and 3) the measurement of physical changes in the heart as assessed by MRI.
  • ETT exercise tolerance time
  • Seattle Angina Questionnaire which provides an assessment based upon a mixed combination of objective and subjective criteria
  • ETT of the 66 patients of the Phase I clinical trial of Examples 1-3 was measured at baseline, and at 1 month, 2 months and 6 months after dosing (with a single unit dose composition of the invention) using a Bruce treadmill protocol. Subjects were excluded from the analysis if the treadmill protocol was not the same as used at baseline. Therefore, the number of subjects varied over time. In addition, any patients who had emergency revascularization were excluded from the analysis. A dose was considered effective if the mean change in ETT from baseline increased by greater than 60 seconds. The results of the ETT assessment are provided in Table 1.
  • the mean change from baseline at one month was less than 60 seconds for all dose groups. However, the percentage of patients stopping their treadmill test because of angina decreased in all groups over time. At 2 months and 6 months after dosing, the mean changes from baseline were greater in the high dose IC and IV groups of patients than in the low and mid dose IC groups. The persistence of increased ETT at 6 months (133.1 sec and 87.5 sec) in the high dose IC (24-48 :g/kg) and IV (18 & 38 :g/kg) groups, respectively, was unexpected. The greatest mean increases in ETT of 107.9 and 133.1 seconds at 2 and 6 months, respectively, occurred in the high dose (2448 :g/kg) IC group.
  • the IV group exhibited significant mean increases in ETT of 93.4 seconds and 87.5 seconds, at 2 months and 6 months respectively, which was not predicted by the rat and pig animal models used herein. Overall, the persistence of the effect (increase in ETT) at 2 months and its magnitude for both the IC and IV groups was wholly unexpected.
  • the 66 human patients of the Phase I clinical trial described in Examples 1-3 herein were also evaluated using the Seattle Angina Questionnaire (SAQ).
  • the possible range of scores for each of the five scales is 0 to 100 with the higher scores indicating a better quality of life.
  • a mean change of 8 points or more between the mean baseline scores (i.e., before treatment) and the posttreatment scores is recognized as being “clinically significant.”
  • a dose was considered “effective” if the mean change in score from baseline increased by greater than 14 points. The reason that 14 was chosen (instead of 8) was to allow for the improvement that was seen in the placebo group at 2 months in a clinical trial of another growth factor—VEGF.
  • the patients were categorized according to the same dosage groups that were evaluated for ETT, i.e., 0.33-2.0 ⁇ g/kg IC (low) 6.0-12.0 ⁇ g/kg IC (mid); 24-48 ⁇ g/kg IC (high); and 18 and 36 ⁇ g/kg IV.
  • the questionnaire was administered to subjects in each dosage group at baseline (prior to dosing), and at 2 months and 6 months after being administered a single unit dose composition of rFGF-2 in accordance with the method of the present invention.
  • the first SAQ 'scale is “exertional capacity.”
  • the data on exertional capacity is summarized in Table 2 herein.
  • the second SAQ scale to be evaluated was “angina stability.”
  • the data summarizing the angina stability is presented in Table 3 herein.
  • the third SAQ scale to be evaluated was “angina frequency.”
  • the data summarizing the angina frequency is presented in Table 4 herein.
  • the mean patient scores (27.9, 32.9, 28.9 and 20.0) for angina frequency increased at 2 months (relative to baseline) by an effective amount (>14) for all dosage groups and for all modes of administration (IC or IV).
  • the mean patient scores continued to increase at 6 months only for the mid dose (6.0-12.0 :g/kg) group, suggesting a peak effect at 2 months post-dosing.
  • the mid dose (6.0-12.0 :g/kg) and high dose (24.0-48.0 :g/kg) groups the changes at 2 months and 6 months were similar, suggesting a persistent effect at 6 months on angina frequency.
  • the fifth SAQ scale to be evaluated was “disease perception.”
  • the data summarizing the disease perception is presented in Table 6 herein.
  • Table 6 the scores for disease perception increased from baseline to scores of 20.2-29.2 at 2 months and 23.8-34.0 at 6 months. These scores showed that administering a single unit dose composition in accordance with the method of the present invention was considered to be as effective (or more effective) at 6 months as at 2 months. These scores suggest a persistence of the effectiveness of the method of the present invention on disease perception out to 6 months following administration of a single unit dose composition.
  • target area collateral extent (%).
  • the greatest collateral extent increases were observed for the low and mid IC doses, i.e., at one month (10.4% and 18.3%, respectively), 2 months (14.7% and 18.0%, respectively) and 2 months (16.0% and no value for mid dose, respectively), which was wholly unexpected.
  • the corresponding % increases in target area collateral extent that were observed for the IC high dose group were 6.3%, 8.0% and 9.0%, respectively.
  • target area delayed arrival extent (%).
  • the greatest target area delayed extent decreases were observed for the low dose IC group, which was also highly unexpected.
  • FGF-2 contains a positively charged tail that is known to bind to proteoglycan chains (heparin and heparin-like structures) on cell surfaces and on the endothelial wall of the vasculature. See Moscatelli, et al., “ Interaction of Basic Fibroblast Growth Factor with Extracellular Matrix and Receptors ,” Ann. NY Acad. Sci., 638:177-181(1981).
  • the kidneys and liver are the major organs for the elimination of rFGF-2.
  • the kidneys have a protein cutoff of about 60 kD and thus retain serum albumin (MW 60 kD).
  • FGF-2 146 residues
  • FGF-2 has a molecular weight of about 16.5 kD. Accordingly, renal excretion is to be expected.
  • bFGF-2 bovine FGF-2
  • both the liver and the kidney were shown to contain high counts of the radiolabelled bFGF-2 at 1 hour after IV or IC injection.
  • the liver was identified as the major organ of elimination.
  • FIG. 1A provides a plasma concentration versus time curve showing these phases in humans after IC administration of rFGF-2 of SEQ ID NO: 2 as a function of each of the following eight doses: 0.33 ⁇ g/kg, 0.65 ⁇ g/kg, 2 ⁇ g/kg, 6 ⁇ g/kg, 12 ⁇ g/kg, 24 ⁇ g/kg, 36 ⁇ g/kg, and 48 ⁇ g/kg of lean body mass (LBM).
  • LBM lean body mass
  • FIG. 1A shows the plasma dose linearity for the eight doses of rFGF-2 that were administered by IC infusion over a twenty minute period.
  • FIG. 1A also shows a biphasic plasma level decline, i.e., a fast distribution phase during the first hour, followed by an elimination phase with an estimated T 1 ⁇ 2 of 5-7 hours.
  • the plasma concentrations of FGF-2 of SEQ ID NO: 2 were determined by a commercially available ELISA (R&D Systems, Minneapolis Minn.) that was marketed for analysis of human FGF-2.
  • the ELISA assay showed 100% cross-reactivity with the rFGF-2 of SEQ ID NO: 2.
  • Other members of the FGF family, as well as many other cytokines, were not detected by this assay. Further, heparin does not interfere with the assay.
  • FIG. 1B is a plot of the mean FGF-2 plasma concentration as a function of time for 18 ⁇ g/kg and 36 ⁇ g/kg rFGF-2 administered IV, as compared to 36 ⁇ g/kg rFGF-2 administered IC.
  • the plasma concentration versus time profiles in FIG. 1B for the 36 ⁇ g/kg doses by the IV and IC routes are superimposible. However, a first-pass effect with the IC route is not eliminated.
  • FIG. 2 is a plot of mean FGF-2 area under the curve (AUC) in pg*min/ml corresponding to FIGS. 1A and 1B.
  • FIG. 2 shows the dose linearity of systemic rFGF-2 exposure (AUC) following IC or IV infusion.
  • AUC systemic rFGF-2 exposure
  • FIG. 2 shows that the systemic exposure to rFGF-2 following administration by the IC and IV routes is substantially similar.
  • FIG. 3 is a plot of individual human patient plasma clearance (CL) values (ml/min/kg) as a function of the time of heparin administration in “minutes prior to rFGF-2 infusion.”
  • FIG. 3 shows the influence of timing of heparin administration on FGF-2 plasma clearance (CL).
  • FIG. 3 shows that administering heparin up to 100 minutes prior to rFGF-2 decreases FGF-2 clearance, the preferred time for administering heparin is from 0-30 minutes prior the rFGF-2 administration, wherein the effect of the heparin on decreasing FGF-2 clearance is greatest.
  • FIG. 4 is a plot individual human patient rFGF-2 dose normalized area under curves (AUCs) as a function of the time of heparin administration in “minutes prior to rFGF-2 infusion” and shows the influence of timing of heparin administration on rFGF-2 AUC.
  • FIG. 4 shows that the greatest AUC/dose was achieved when an effective amount of a glycosoaminoglycan, such as heparin, was preadministered within 30 minutes or less of IC rFGF-2 infusion, more preferably within 20 minutes or less of IC or IV rFGF-2 infusion.
  • an effective amount of a glycosoaminoglycan is 10-80 U/kg heparin.
  • the mean pharmacokinetic parameters for rFGF-2 in humans as a function of dosage and mode of administration are summarized in Table 8 herein.
  • the T1 ⁇ 2 for FGF-2 in humans was determined to range from 2.2 ⁇ 3.7 hours at low dose (0.33-2.0 ⁇ g/kg) IC to 7.0+3.5 hours at a dose of 18-36 ⁇ g/kg IV; given the limitations of the assay, the terminal half-life is estimated at 5-7 hours for all groups.
  • the clearances of FGF-2 ranged from 13.2 to 18.2 L/hour/70 kg man.
  • the steady state volume (V ss ) was determined to range from 11.3 ⁇ 10.4 L/70 kg man to 16.8 ⁇ 10.7 L/70 kg man.
  • exercise capacity was significantly improved in patients with congestive heart failure.
  • the rFGF-2 of SEQ ID NO: 2 was formulated as a unit dose and pharmaceutical composition and administered to rats, pigs and ultimately to humans in the Phase I clinical trial referenced herein. The various formulations are described below.
  • the rFGF-2 Unit Dose was provided as a liquid in 3 cc type I glass vials with a laminated gray butyl rubber stopper and red flip-off overseal.
  • the rFGF-2 unit dose contained 1.2 ml of 0.3 mg/ml rFGF-2 of SEQ ID NO: 2 in 10 mM sodium citrate, 10 mM monothioglycerol, 1 mM disodium dihydrate EDTA (molecular weight 372.2), 135 mM sodium chloride, pH 5.0.
  • each vial (and unit dose) contained 0.36 mg rFGF-2.
  • the vials containing the unit dose in liquid form were stored at 20 to 8° C.
  • the rFGF diluent was supplied in 5 cc type I glass vials with a laminated gray butyl rubber stopper and red flip-off overseal.
  • the rFGF-2 diluent contains 10 mM sodium citrate, 10 mM monothioglycerol, 135 mM sodium chloride, pH 5.0.
  • Each vial contained 5.2 ml of rFGF-2 diluent solution that was stored at 2° to 8° C.
  • the rFGF-2 Pharmaceutical Composition that was infused was prepared by diluting the rFGF-2 unit dose with the rFGF diluent such that the infusion volume is 1040 ml. In order to keep the EDTA concentration below a preset limit of 100 ⁇ g/ml, the total infusion volume was increased to a maximum of 40 ml when proportionately higher absolute amounts of FGF-2 were administered to patients with higher body weights.
  • Suboptimal candidates for approved revascularization procedures e.g., angioplasty, stents, coronary artery bypass graft (CABG) (or refuses those interventions)
  • Malignancy any history of malignancy within past ten years, with the exception of curatively treated basal cell carcinoma.
  • Ocular conditions proliferative retinopathy, severe non-proliferative retinopathy, retinal vein occlusion, Eales' disease, or macular edema or funduscopy by ophthalmologist: history of intraocular surgery within 2 months
  • Renal function creatinine clearance below normal range adjusted for age; protein >250 mg or microalbumin >30 mg/24 h urine
  • Hemodynamically relevant arrhythmias e.g., ventricular fibrillation, sustained ventricular tachycardia
  • Any pathological fibrosis e.g., pulmonary fibrosis, scleroderma
  • vascular malformation e.g., AV malformation, hemangiomas
  • the Phase I CAD trial of this example is an open label, dose escalation study of recombinant fibroblast growth factor-2 (rFGF-2) for safety, tolerability and pharmacokinetics.
  • rFGF-2 recombinant fibroblast growth factor-2
  • the study was conducted at 2 sites: Beth Israel Deaconess Hospital (Harvard) in Boston, Mass. and Emory University Hospital in Atlanta, Ga. Enrollment is complete. Subjects were treated with a single infusion of rFGF-2 on Day 1 and followed for 360 days; follow-up is not yet complete in some subjects.
  • the subject population consists of patients with advanced CAD who are exercise limited by coronary ischemia and are considered suboptimal candidates for (or do not want to undergo) one of the established revascularization procedures (e.g., CABG, angioplasty—with or without stent).
  • the major exclusion criteria were history or suspicion of malignancy, uncompensated heart failure or left ventricular ejection fraction ⁇ 20%, renal insufficiency or proteinuria, and various ocular conditions (e.g., proliferative diabetic retinopathy, severe non-proliferative retinopathy).
  • rFGF-2 of SEQ ID NO: 2 Sixty-six subjects have received rFGF-2 of SEQ ID NO: 2 in this trial: 52 received the rFGF-2 as an IC infusion and fourteen received it as an IV infusion. Each subject was observed in the hospital for at least twenty-four hours, and then followed as an outpatient for 360 days with follow-up visits (Days 15, 29, 57, 180 and 360). At least four subjects were studied at each dose; if no subject experienced dose-limiting toxicity as defined by the protocol within 2 days, the dose was escalated. The drug was administered as a single 20 minute infusion divided between 2 major sources of coronary blood supply (IC), using standard techniques for positioning a catheter into the patient's coronary artery (such as already employed in angioplasty) or in a peripheral vein (IV).
  • IC major sources of coronary blood supply
  • IV peripheral vein
  • Angina frequency and quality of life was assessed by the Seattle Angina Questionnaire (SAQ) at a baseline (before rFGF-2 administration) and at 2 months and 6 months after rFGF-2 administration.
  • Exercise tolerance time was assessed by the treadmill test at 1, 2, and 6 months.
  • Rest/exercise nuclear perfusion and gated sestamibi-determined rest ejection fraction (EF), and resting magnetic resonance imaging (MRI) were assessed at baseline, and at 1 month, 2 months and 6 months post rFGF-2 administration.
  • MRI measurements which were thought to objectively measure changes in % in cardiac function and perfusion included: (1) ejection fraction; (2) myocardial infarct extent (%); (3) normal wall thickening (4) normal motion (%); (5) target wall thickening (%); (6) target wall motion (%); (7) target wall area collateral extent (%); and (8) target area delayed arrival extent (%).
  • MTD maximally tolerated dose
  • the starting dose of 0.33 ⁇ g/kg IC was escalated over eight sequential groups to 48 ⁇ g/kg IC, at which dose 2 of ten subjects experienced dose-limiting toxicity (hypotension) as defined by the protocol. Hypotension was manageable with fluids alone in all subjects (no vasopressors or assistive devices). At 36 ⁇ g/kg IC, only 1 of 10 subjects had dose-limiting toxicity which defined this dose as the maximally tolerated dose (MTD). Two additional groups were studied by IV infusion; four subjects of half the MTD (18 ⁇ g/kg) and ten subjects at the MTD (36 ⁇ g/kg).
  • SAEs serious adverse events
  • AEs treatment-emergent adverse events
  • bradycardia was not dose-dependent.
  • Other adverse events (AEs) which were deemed at least possibly related and appeared dose-related occurred within the first several days or week post infusion and included chest pain, shortness of breath, insomnia, anxiety, and nausea. These events were mild to moderate in severity, and most did not require specific intervention.
  • the drug When administered IC, the drug was administered over approximately 20 minutes as a single infusion divided between 2 major sources of coronary blood supply (IC), using standard techniques for positioning a catheter into the patient's coronary artery (such as already employed in angioplasty).
  • IC coronary blood supply
  • IV When administered IV, the drug was administered as an infusion over 20 minutes into a peripheral vein.
  • MI myocardial infarction
  • 4 6.0 ⁇ g/kg
  • 7 36.0 ⁇ g/kg
  • Multiple MIs occurred in 2 patients, i.e., one from group 1 (0.33 ⁇ g/kg) and one from group 3 (2.0 ⁇ g/kg).
  • Emergency revascularization procedures CABG or angioplasty with or without stent were performed during follow-up in 4 patients: one each from groups 1 (0.33 ⁇ g/kg), 3 (2.0 ⁇ g/kg), 4 (6.0 ⁇ g/kg), and 7 (36.0 ⁇ g/kg).
  • Acute hypotension seen at higher doses during or just subsequent to infusion, was managed by administration of IV fluids without need for a vasopressor.
  • the maximally tolerated dose (MTD) in humans was defined as 36 ⁇ g/kg IC.
  • MTD maximally tolerated dose
  • the MTD was 6.5 ⁇ g/kg IC.
  • Doses of rFGF-2 up to 48 ⁇ g/kg IC were administered in human patients with aggressive fluid management, but were defined by the protocol as “not tolerated” due to acute and/or orthostatic hypotension in 2 out of ten patients.
  • the terminal half-life of the infused rFGF-2 was estimated at 5 to 7 hours.
  • a treatment is considered very successful if the angiogenic effects last at least 2 months.
  • the unexpectedly superior angiogenic effects were observed to last up to 6 months in some patients in all dosage groups. Based upon the results already obtained, it is expected that the angiogenic effects may last twelve months or more but do last at least 2 months in the patients, at which time the procedure could be repeated, if necessary.
  • the rFGF-2 of SEQ ID NO: 2 was formulated as a unit dose pharmaceutical composition for administration to humans in the Phase II clinical trial referenced herein. The various formulations are described below.
  • the rFGF-2 Unit Dose was prepared as a liquid in 5 cc type I glass vials with a laminated gray butyl rubber stopper and red flip-off overseal.
  • the rFGF-2 formulation contains 0.3 mg/ml rFGF-2 of SEQ ID NO: 2 in 10 mM sodium citrate, 10 mM monothioglycerol, 0.3 mM disodium dihydrate EDTA (molecular weight 372.2), 135 mM sodium chloride, pH 5.0.
  • Each vial contained 3.7 ml of rFGF-2 drug product solution (1.11 mg rFGF-2 per vial).
  • the resulting unit dose in liquid form is stored at less than ⁇ 60° C.
  • the above described unit dose is diluted with the “rFGF-2 placebo.”
  • the contents of several of the vials may be combined to produce a unit dose of 36 ⁇ g/kg for the Phase II study.
  • the rFGF-2 placebo is supplied as a clear colorless liquid in 5 cc type I glass vials with a laminated gray butyl rubber stopper and red flip-off overseal.
  • the rFGF-2 placebo is indistinguishable in appearance from the drug product and has the following formulation: 10 mM sodium citrate, 10 mM monothioglycerol, 0.3 mM disodium dihydrate EDTA (molecular weight 372.2), 135 mM sodium chloride, pH 5.0.
  • Each vial contains 5.2 ml of rFGF-2 placebo solution.
  • the rFGF-2 placebo is stored at 2° to 8° C.
  • the rFGF-2 Pharmaceutical Composition that is infused is prepared by diluting the rFGF-2 unit dose with the rFGF diluent such that the infusion volume is 20 ml for Phase II.
  • This clinical trial was designed as a phase 2, multicenter, double-blind, placebo-controlled study of rFGF-2 in 300 subjects with CAD. Subjects who met all eligibility criteria were randomly assigned to receive placebo or one of three doses of rFGF-2 (approximately 75 subjects per arm). Doses were 0 (placebo), 0.3, 3.0, and 30.0 ⁇ g/kg based on actual body weight. Study drug was to be administered as a single IC infusion over 20 minutes during cardiac catheterization. Subjects were monitored in the hospital for at least 6 hours postdose at the site, and then followed at specified intervals over 180 days. Long-term safety was assessed in a separate extension protocol using telephone contacts and questionnaires for an additional 6 months.
  • Ejection fraction ⁇ 30% by an accepted imaging technology (ventriculogram, MRI, first pass on single photon emission computed tomography [SPECT]) scan, echocardiogram [ECHO] or multigated [MUGA] nuclear assessment).
  • Inducible and reversible ischemic defect of moderate or greater size involving at least half the area of one major myocardial territory (anterior, inferior, lateral, or septal) on a rest/stressed thallium/sestamibi scan, or multiple inducible and reversible ischemic defects in multiple myocardial territories, the sum total of which involves the equivalent of at least half the area of one major myocardial territory on a rest/stressed thallium/sestamibi scan within 30 days prior to randomization.
  • CBC Complete blood count
  • platelets serum chemistry
  • prothrombin time prothrombin time
  • urinalysis within clinically acceptable ranges for cardiac catheterization within 30 days prior to randomization.
  • Prostate specific antigen within 30 days prior to randomization (males) conforming to the following age-specific ranges: Age PSA (ng/mL) ⁇ 40 years ⁇ 2.0 40-49 years ⁇ 2.7 50-59 years ⁇ 3.7 60-69 years ⁇ 5.1 70-79 years ⁇ 7.0 ⁇ 80 years ⁇ 7.2
  • Malignancy History or suspicion of malignancy within the past 10 years, with the exception of curatively treated basal cell carcinoma, squamous cell carcinoma of the skin in sun-exposed areas, or carcinoma of the cervix.
  • Renal insufficiency as defined by serum creatinine>2.0 mg/dL.
  • Proteinuria as defined by 1+ or greater protein on dipstick.
  • CABG CABG, angioplasty, transient ischemic attack, or stroke within 4 months.
  • Any pathological fibrosis eg, pulmonary fibrosis, scleroderma.
  • vascular malformation eg, arteriovenous malformation, hemangiomas>3 mm.
  • a subject could withdraw consent to participate in the study at any time without prejudice. Additionally, the investigator could withdraw a subject if, according to clinical judgment, it was in the best interest of the subject or if the subject could not comply with the protocol.
  • Subjects were randomly assigned to receive placebo or 0.3, 3.0, or 30.0 ⁇ g/kg rFGF-2.
  • the study drug was to be administered as an infusion of 20 mL over 20 minutes divided between two patent coronary vessels or grafts, using a calibrated, precision, infusion pump.
  • the rFGF-2 used in this study was a 146 amino acid, non-glycosylated, monomeric, 16.5 kDa protein that was expressed in genetically engineered yeast.
  • the rFGF-2 drug product was supplied as a clear, colorless liquid in 5-mL, type I, glass vials with a laminated, gray, butyl stopper and red, flip-off overseal.
  • the rFGF-2 formulation contained 0.3 mg/mL rFGF-2 in 10 mM sodium citrate, 10 mM monothioglycerol, 0.3 mM disodium dihydrate EDTA (molecular weight 372.2),135 mM sodium chloride, pH 5.0.
  • Each vial contained 3.7 mL of rFGF-2 drug-product solution (1.11 mg rFGF-2 per vial).
  • the rFGF-2 drug-product vials were stored at ⁇ 60° C. or less.
  • the rFGF-2 was diluted with placebo according to the subjects' actual body weight.
  • the placebo was supplied as a clear, colorless liquid in 5-mL, type I, glass vials with a laminated, gray, butyl stopper and red, flip-off overseal.
  • the placebo was indistinguishable in appearance from the drug product; it contained 10 mM sodium citrate, 10 mM monothioglycerol, 0.3 mM disodium dihydrate EDTA (molecular weight 372.2), 135 mM sodium chloride, pH 5.0.
  • Each vial contained 5.2 mL of placebo solution.
  • the placebo vials were stored at 2 to 8° C.
  • Subjects typically received a single IV bolus of heparin between 10 and 20 minutes prior to infusion of study drug in order to minimize the risk of thrombosis related to the duration of time the catheter was in place.
  • the primary efficacy variable was the change in exercise capacity as measured by ETT time at Day 90.
  • Secondary efficacy variables included the change in ETT time at Day 180; the change in QoL as measured by the angina frequency score (AFS), treatment satisfaction score (TSS), exertional capacity score (ECS), and disease perception score (DPS) of the SAQ, and the physical and mental components of the SF-36 at Days 90 and 180; the change in ischemic area at rest and with pharmacologic stress by thallium/sestamibi scans at Days 90 and 180; changes in EF, targeted wall thickness and motion, and perfusion by MRI at Days 90 and 180 in a subset of subjects.
  • AFS angina frequency score
  • TSS treatment satisfaction score
  • ECS exertional capacity score
  • DPS disease perception score
  • the Canadian Cardiovascular Classification is based on a classification scheme ranging from Class 0 to Class IV. To be classified in: Class 0, a patient does not experience angina or anginal equivalent symptoms; Class I, with ordinary physical activity symptoms occur only with strenuous, rapid, or prolonged exertion at work or recreation; Class II, a patient experiences slight limitation of ordinary activity due to angina; Class III, a patient experiences marked limitation of activity due to angina; and Class IV, a patient develops angina at rest or with any physical activity.
  • the SAQ is a validated, disease-specific, self-administered questionnaire with 5 scales: angina stability scale (ASS), angina frequency scale (AFS), exercise capacity scale (ECS), treatment satisfaction scale (TSS), and disease perception scale (DPS) (Spertus JA, et al.).
  • the ASS was not included in the analysis of this trial as it refers to a 4 week interval which was not relevant to the evaluation periods.
  • Each component comprises a scale from 1 to 100; lower scores are associated with worse symptoms; >8 point changes are considered clinically relevant.
  • the SF-36 is a validated, general, quality-of-life (QoL) instrument with 10 scales and two summary component scales: a physical component summary scale (PCSS) and a mental component summary scale (MCSS).
  • QoL quality-of-life
  • a segment was determined to have a reversible defect if the assigned abnormal regional grade at stress decreased or normalized on the rest images (reversibility score: stress score ⁇ rest score>1), and to have a fixed defect if the assigned regional grade at stress was abnormal and remained the same on rest imaging.
  • the fixed defects were subgrouped on the basis of the severity of the graded scores, i.e., mild-to-moderate (scores of 1, 2, and 3) and severe (score of 4).
  • the global extent of perfusion abnormality and ischemia was assessed by summing the individual scores from the 20 segments, and expressed as the average stress and average reversibility scores, respectively.
  • a Data and Safety Monitoring Board (DSMB) reviewed SAEs and results of laboratory tests approximately every 6 weeks.
  • Table 7 summarizes demographic features of the subjects enrolled in the trial. These features were similar among the four treatment groups (Table 7). TABLE 7 Summary of Demography Treatment Group Placebo 0.3 ⁇ g/kg 3.0 ⁇ g/kg 30 ⁇ g/kg Age (mean years) 63.9 63.0 62.9 61.8 Percent male sex 86% 84% 80% 86% Percent Caucasian race 97% 90% 94% 93% Weight (mean kg) 88.24 87.25 87.23 87.50 Height (mean cm) 170.94 172.02 172.45 171.99 Percent tobacco use 8% 11% 10% 10%
  • FIG. 9 shows the ETT time at baseline, at Day 90, and the primary efficacy variable, change in ETT time from baseline to Day 90.
  • subjects who had undergone a revascularization and subjects with no assessment were excluded from the analysis unless otherwise specified.
  • the mean increase in exercise time was 44.1 seconds for the placebo group, 48.5 seconds in the low-dose group, 65.0 seconds in the middle-dose group, and 49.1 seconds in the high-dose group.
  • FIG. 9 also shows ETT time at Day 180 and the change in ETT time from baseline to Day 180.
  • the mean increase in exercise time was 54.8 seconds in the placebo group, 75.3 seconds in the low-dose group, 76.3 seconds in the middle-dose group, and 51.3 seconds in the high-dose group.
  • Trends in improvement in ETT were observed at Day 180 in subjects with a baseline CCS of 3 or 4 and/or SAQ angina frequency of less than or equal to 40.
  • FIG. 10 shows the analyses of the SAQ.
  • a change of ⁇ 8 points is considered clinically meaningful and higher SAQ scores are associated with better clinical status.
  • the P values were 0.035 based upon the overall test and 0.007 based upon the test of placebo versus all FGF.
  • FIG. 12 shows the analysis for the SF-36 Physical Component Summary Scale (PCSS) and Mental Component Summary Scale (MCSS).
  • PCSS Physical Component Summary Scale
  • MCSS Mental Component Summary Scale
  • FIG. 13 shows ETT time for subjects with baseline CCS of 3 or 4.
  • the mean change in exercise time from baseline to Day 180 was 33.1 seconds for the placebo group, 70.7 seconds for the low-dose group, 75.7 seconds for the middle-dose group, and 41.5 seconds for the high-dose group (pairwise P values 0.15, 0.086, 0.74, respectively).
  • FIG. 13 show SAQ scores for subjects with baseline CCS of 3 or 4.
  • FIG. 14 shows ETT time for subjects with baseline Angina Frequency Score (AFS) of ⁇ 40, as measured using the Seattle Angina Questionnaire (SAQ).
  • AFS Angina Frequency Score
  • SAQ Seattle Angina Questionnaire
  • FIG. 14 shows AFS for subjects with baseline AFS of ⁇ 40.
  • the magnitude of the difference in AFS between the low and middle dose groups and placebo in subjects with lower AFS at baseline was considered clinically relevant.
  • LVEDD Left Ventricular End Diastolic Diameter
  • Normal wall motion is in the expected range (30-40%) and was unchanged. Normal wall thickening is in the expected range (45-50%) and is unchanged. Target wall, i.e., the wall of the myocardium that is ischemic, motion at rest was the same as normal.
  • DAZ Delayed arrival zone
  • MRI data provided evidence of improved LV function in FGF groups as evidenced by reduced LVEDD and LVESD. Perfusion analysis showed a trend towards improvement in FGF groups.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cardiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Vascular Medicine (AREA)
  • Dermatology (AREA)
  • Molecular Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Urology & Nephrology (AREA)
  • Endocrinology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US09/771,302 1998-10-13 2001-01-26 Angiogenically effective unit dose of FGF-2 and method of use Abandoned US20020072489A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
US09/771,302 US20020072489A1 (en) 1998-10-13 2001-01-26 Angiogenically effective unit dose of FGF-2 and method of use
PCT/US2002/002239 WO2002058720A2 (en) 2001-01-26 2002-01-25 Angiogenically effective unit dose of fgf-2 and method of use
JP2002559054A JP2004533411A (ja) 2001-01-26 2002-01-25 血管形成的に有効なfgf−2の単位用量および使用方法
EP02713467A EP1361887A2 (en) 2001-01-26 2002-01-25 Angiogenically effective unit dose of fgf-2 and method of use
AU2002245315A AU2002245315A1 (en) 2001-01-26 2002-01-25 Angiogenically effective unit dose of fgf-2 and method of use
US10/862,152 US7112560B2 (en) 1998-10-13 2004-06-04 Angiogenically effective unit dose of FGF-2 and method of use
US11/511,028 US7541333B2 (en) 1998-10-13 2006-08-28 Angiogenically effective unit dose of FGF-2 and method of use
JP2008235760A JP2009024026A (ja) 2001-01-26 2008-09-12 血管形成的に有効なfgf−2の単位用量および使用方法
JP2009277091A JP2010053145A (ja) 2001-01-26 2009-12-04 血管形成的に有効なfgf−2の単位用量および使用方法
JP2009277090A JP2010053144A (ja) 2001-01-26 2009-12-04 血管形成的に有効なfgf−2の単位用量および使用方法

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US10410398P 1998-10-13 1998-10-13
US10410298P 1998-10-13 1998-10-13
US09/385,114 US6440934B1 (en) 1998-10-13 1999-08-27 Angiogenically effective unit dose of FGF-2 and method of use
US09/771,302 US20020072489A1 (en) 1998-10-13 2001-01-26 Angiogenically effective unit dose of FGF-2 and method of use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/385,114 Continuation-In-Part US6440934B1 (en) 1998-10-13 1999-08-27 Angiogenically effective unit dose of FGF-2 and method of use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/862,152 Continuation US7112560B2 (en) 1998-10-13 2004-06-04 Angiogenically effective unit dose of FGF-2 and method of use

Publications (1)

Publication Number Publication Date
US20020072489A1 true US20020072489A1 (en) 2002-06-13

Family

ID=25091381

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/771,302 Abandoned US20020072489A1 (en) 1998-10-13 2001-01-26 Angiogenically effective unit dose of FGF-2 and method of use
US10/862,152 Expired - Lifetime US7112560B2 (en) 1998-10-13 2004-06-04 Angiogenically effective unit dose of FGF-2 and method of use
US11/511,028 Expired - Fee Related US7541333B2 (en) 1998-10-13 2006-08-28 Angiogenically effective unit dose of FGF-2 and method of use

Family Applications After (2)

Application Number Title Priority Date Filing Date
US10/862,152 Expired - Lifetime US7112560B2 (en) 1998-10-13 2004-06-04 Angiogenically effective unit dose of FGF-2 and method of use
US11/511,028 Expired - Fee Related US7541333B2 (en) 1998-10-13 2006-08-28 Angiogenically effective unit dose of FGF-2 and method of use

Country Status (5)

Country Link
US (3) US20020072489A1 (ja)
EP (1) EP1361887A2 (ja)
JP (4) JP2004533411A (ja)
AU (1) AU2002245315A1 (ja)
WO (1) WO2002058720A2 (ja)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060025343A1 (en) * 1998-10-13 2006-02-02 Chiron Corporation Angiogenically effective unit dose fo FGF and method of adminstering
US8618052B2 (en) 1998-10-13 2013-12-31 Novartis Vaccines And Diagnostics, Inc. Method of treating coronary artery disease by administering FGF-5
EP3615560A4 (en) * 2017-04-26 2020-12-23 University of Cincinnati FIBROBLAST GROWTH FACTOR LIGAND COMPOSITIONS AND METHODS OF TREATMENT OF PATIENTS UNDERGOING ISCHEMIC REPERFUSION THERAPY

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080311554A1 (en) * 1994-05-06 2008-12-18 Slotman Gus J Methods for monitoring patients with severe sepsis and septic shock and for selecting treatments for these patients
US20030166550A1 (en) 1999-08-27 2003-09-04 Chiron Corporation Angiogenically effective unit dose of FGF-2 and method of use
AU6634400A (en) 1999-08-13 2001-03-13 Chiron Corporation Dose of an angiogenic factor and method of administering to improve myocardial blood flow
CA2405291A1 (en) * 2000-04-06 2001-10-18 Wayne P. Franco Methods of using growth factors for treating heart disease
US20020115603A1 (en) * 2000-06-22 2002-08-22 Chiron Corporation Methods and compositions for the treatment of peripheral artery disease
WO2009058818A2 (en) * 2007-10-29 2009-05-07 The Board Of Regents Of The University Of Texas System Compositions comprising a micro-rna and methods of their use in regulating cardiac remodeling
US8577620B2 (en) * 2008-03-27 2013-11-05 Gus J. Slotman Methods for assessing drug efficacy and response of patient to therapy
KR20220090951A (ko) * 2020-12-23 2022-06-30 건국대학교 산학협력단 섬유아세포 성장인자의 신규 단편 및 이의 용도

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4296100A (en) 1980-06-30 1981-10-20 Franco Wayne P Method of treating the heart for myocardial infarction
US4378347A (en) 1980-06-30 1983-03-29 Franco Wayne P Composition for treating the heart for myocardial infarction
US5155214A (en) 1984-03-05 1992-10-13 The Salk Institute For Biological Studies Basic fibroblast growth factor
US5464774A (en) 1984-03-05 1995-11-07 The Salk Institute For Biological Studies Bovine basic fibroblast growth factor
US4956455A (en) 1984-03-05 1990-09-11 The Salk Institute For Biological Studies Bovine fibroblast growth factor
US5439818A (en) 1985-09-12 1995-08-08 Scios Nova Inc. DNA encoding human recombinant basic fibroblast growth factor
US5604293A (en) 1985-09-12 1997-02-18 Scios Inc. Recombinant human basic fibroblast growth factor
US5238916A (en) 1987-05-29 1993-08-24 The Trustees Of Columbia University In The City Of New York Oncogene encoded polypeptide having growth factor activity and methods of use thereof
US5155217A (en) 1987-05-29 1992-10-13 The Trustees Of Columbia University In The City Of New York Ongogene encoding polypeptide having growth factor activity
US5750659A (en) 1987-06-16 1998-05-12 New York University Mammalian growth factor
KR890701607A (ko) 1987-11-24 1989-12-21 원본미기재 섬유아세포 성장 인자의 유사체
US5108363A (en) 1988-02-19 1992-04-28 Gensia Pharmaceuticals, Inc. Diagnosis, evaluation and treatment of coronary artery disease by exercise simulation using closed loop drug delivery of an exercise simulating agent beta agonist
AU614137B2 (en) 1988-06-06 1991-08-22 Takeda Chemical Industries Ltd. Stabilized fgf composition and production thereof
US5352589A (en) 1988-09-16 1994-10-04 Farmitalia Carlo Erba S.R.L. Deletion mutant of basic fibroblast growth factor and production thereof
US5137734A (en) 1989-03-22 1992-08-11 Dana Farber Cancer Institute Angiogenic monoglycerides
US5194596A (en) 1989-07-27 1993-03-16 California Biotechnology Inc. Production of vascular endothelial cell growth factor
US5350836A (en) 1989-10-12 1994-09-27 Ohio University Growth hormone antagonists
US5612211A (en) * 1990-06-08 1997-03-18 New York University Stimulation, production and culturing of hematopoietic progenitor cells by fibroblast growth factors
DK0486862T3 (da) 1990-11-23 1996-06-24 American Cyanamid Co Kimær fibrolast-vækstfaktor
US5206354A (en) 1990-11-23 1993-04-27 American Cyanamid Company Dna sequence encoding active fragment of fibroblast growth factor, hbf-2
US5213570A (en) 1990-12-14 1993-05-25 Mallinckrodt Medical, Inc. System and method for oxygenation of the pericardium
US5137510A (en) 1990-12-14 1992-08-11 Mallinckrodt Medical, Inc. System and method for oxygenation of the heart using subpericardial fluids
US5269326A (en) 1991-10-24 1993-12-14 Georgetown University Method for transvenously accessing the pericardial space via the right auricle for medical procedures
US5244460A (en) 1991-11-27 1993-09-14 The United States Of America As Represented By The Department Of Health And Human Services Method to foster myocardial blood vessel growth and improve blood flow to the heart
US5491220A (en) 1993-09-24 1996-02-13 Yeda Research And Development Co., Ltd. Surface loop structural analogues of fibroblast growth factors
US5792453A (en) 1995-02-28 1998-08-11 The Regents Of The University Of California Gene transfer-mediated angiogenesis therapy
US5773252A (en) * 1995-06-05 1998-06-30 Human Genome Sciences, Inc. Fibroblast growth factor 15
WO1997016169A1 (en) * 1995-11-01 1997-05-09 Chiron Corporation Treatment of a cardiovascular indication by delivery of therapeutics to the pericardial space
WO1999005190A1 (fr) * 1997-07-22 1999-02-04 Mitsui Chemicals, Inc. COPOLYMERES D'ETHYLENE/α-OLEFINE, COMPOSITIONS, ET PROCEDES DE PREPARATION DE CES COPOLYMERES ET COMPOSITIONS
ATE434440T1 (de) * 1998-09-03 2009-07-15 Novartis Vaccines & Diagnostic Fgf-2 angiogenische wirksame einheitdosis und verfahren zu ihrer verwendung
IL145834A0 (en) * 1999-04-26 2002-07-25 Aventis Pharma Sa Use of a recombinant defective adenovirus comprising a nucleic acid encoding an angiogenic factor for treating pulmonary hypertension
AU6634400A (en) * 1999-08-13 2001-03-13 Chiron Corporation Dose of an angiogenic factor and method of administering to improve myocardial blood flow
US20020115603A1 (en) * 2000-06-22 2002-08-22 Chiron Corporation Methods and compositions for the treatment of peripheral artery disease
CA2436076A1 (en) * 2001-01-26 2002-08-01 Genetix Pharmaceuticals, Inc. Use of compositions containing pdgf-bb for promoting angiogenesis

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060025343A1 (en) * 1998-10-13 2006-02-02 Chiron Corporation Angiogenically effective unit dose fo FGF and method of adminstering
US7511019B2 (en) 1998-10-13 2009-03-31 Novartis Vaccines And Diagnostics, Inc. Method of treating coronary artery disease by administering FGF-4
US20090170776A1 (en) * 1998-10-13 2009-07-02 Novartis Vaccines And Diagnostics, Inc. Angiogenically effective unit dose of fgf and method of administering
US7858584B2 (en) 1998-10-13 2010-12-28 Novartis Vaccines And Diagnostics, Inc. Angiogenically effective unit dose of FGF and method of administering
US8618052B2 (en) 1998-10-13 2013-12-31 Novartis Vaccines And Diagnostics, Inc. Method of treating coronary artery disease by administering FGF-5
US9149507B2 (en) 1998-10-13 2015-10-06 Novartis Vaccines and Dignostics, Inc. Method of treating coronary artery disease by administering FGF-9
EP3615560A4 (en) * 2017-04-26 2020-12-23 University of Cincinnati FIBROBLAST GROWTH FACTOR LIGAND COMPOSITIONS AND METHODS OF TREATMENT OF PATIENTS UNDERGOING ISCHEMIC REPERFUSION THERAPY

Also Published As

Publication number Publication date
AU2002245315A1 (en) 2002-08-06
JP2009024026A (ja) 2009-02-05
JP2004533411A (ja) 2004-11-04
JP2010053145A (ja) 2010-03-11
EP1361887A2 (en) 2003-11-19
WO2002058720A3 (en) 2003-05-15
US20070142283A1 (en) 2007-06-21
US7541333B2 (en) 2009-06-02
US20050143298A1 (en) 2005-06-30
WO2002058720A2 (en) 2002-08-01
US7112560B2 (en) 2006-09-26
JP2010053144A (ja) 2010-03-11

Similar Documents

Publication Publication Date Title
US7541333B2 (en) Angiogenically effective unit dose of FGF-2 and method of use
US7511019B2 (en) Method of treating coronary artery disease by administering FGF-4
JP2020117546A (ja) 脈管形成因子の用量および心筋血流を改善するための投与方法
US6440934B1 (en) Angiogenically effective unit dose of FGF-2 and method of use
US8796211B2 (en) Method of treating coronary artery disease FGF-2
JP2010163457A (ja) 脈管形成に有効な単位用量のfgf−2および使用方法
US20180221446A1 (en) Angiogenically Effective Unit Dose Of FGF And Method Of Administering
EP1525888B1 (en) Angiogenically effective unit dose of FGF-2 and method of use

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHIRON CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WHITEHOUSE, MARTHA JO;REEL/FRAME:012833/0001

Effective date: 20020404

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NOVARTIS VACCINES AND DIAGNOSTICS, INC., CALIFORNI

Free format text: MERGER;ASSIGNORS:CHIRON CORPORATION;NOVARTIS BIOTECH PARTNERSHIP, INC.;REEL/FRAME:021773/0717

Effective date: 20060419