CA2155540A1 - Methods for treating amyotrophic lateral sclerosis with cntf - Google Patents

Methods for treating amyotrophic lateral sclerosis with cntf

Info

Publication number
CA2155540A1
CA2155540A1 CA 2155540 CA2155540A CA2155540A1 CA 2155540 A1 CA2155540 A1 CA 2155540A1 CA 2155540 CA2155540 CA 2155540 CA 2155540 A CA2155540 A CA 2155540A CA 2155540 A1 CA2155540 A1 CA 2155540A1
Authority
CA
Canada
Prior art keywords
cntf
day
als
patients
administered
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
CA 2155540
Other languages
French (fr)
Inventor
Frank Collins
Mark Dietz
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.)
Syntex Synergen Neuroscience Joint Venture
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2155540A1 publication Critical patent/CA2155540A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • A61K38/185Nerve growth factor [NGF]; Brain derived neurotrophic factor [BDNF]; Ciliary neurotrophic factor [CNTF]; Glial derived neurotrophic factor [GDNF]; Neurotrophins, e.g. NT-3

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Psychology (AREA)
  • Neurology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Neurosurgery (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A method is provided for the treatment of ALS which is comprised of administering a therapeutically effective amount of ciliary neurotrophic factor (CNTF).

Description

~ W094117818 21 5 ~ ~ 4 0 PCT~S94/0~27 METHOD~ FOR TREATING ~IYO,K~PHIC LATE~AL
3ÇS.~8I~ WITH CNTF

BACKGROUND OF THE lN V~N'l'lON
The present invention includes methods for the treatment of amyotrophic lateral sclerosis (ALS) in human patients.
Amyotrophic lateral sclerosis (ALS) is a progressive degenerative disorder of motor neurons in the spinal cord, brainstem, and motor cortex, manifested clinically by muscular weakness, atrophy, and corticospinal tract signs in varying combinations.
ALS is a common disease, with an annual incidence rate of 0.4 to l.76 per lOO,OOO population (approximately î,OOO-4,300 in the U.S.). Most patients are more than 50 years old at the onset of symptoms and the incidence increases with each decade of life. ALS
occurs in a random pattern throughout the world; it is estimated that in about 5% of cases ALS is familial, being inherited as an autosomal dominant trait.
ALS affects neuromuscular functions in the hand, leg, thoracic, abdominal, or posterior neck muscles.
Typical initial symptoms in the hand include uselessness of hand, awkwardness in tasks requiring fine finger movements, stiffness of the fingers, and slight weakness or wasting of the hand muscles.
Cramping and fasciculations of the muscles of the forearm, upper arm, and shoulder girdle muscles also appears. With time, the other hand and arm may be similarly affected. Eventually a patient exhibits atrophic weakness of the hands and forearms, slight spasticity of the legs, and generalized hyperreflexia.
Muscle strength and bulk diminish, abductors, adductors, and extensors of fingers and thumb tend to become weak before the long flexors, on which the handgrip depends, and the dorsal interosseous spaces become hollowed, giving rise to the "cadaveric" or "skeleton hand". The patient may walk about with WO94/17818 PCT~S94/01227 useless, dangling arms. Later the atrophic weakness spreads to the neck, tongue, pharyngeal and laryngeal muscles.
The principal finding is a loss of nerve cells in the anterior horns of the spinal cord and motor nuclei of the lower brainstem. There is an extensive neuronal loss and gliosis involving the premotor area, particularly the superior frontal gyri, and the inferolateral cortex of the temporal lobes. Many of the surviving nerve cells are small, shrunken, and filled with lipofuscin. Lost cells are replaced by fibrous astrocytes.
ALS has been observed in conjunction with presenile and senile dementia, and with Parkinsons disease. The course of ALS, irrespective of its particular mode of onset and pattern of evolution, is inexorably progressive. Half of the patients die within 3 years and 90% within 6 years (Adams and Victor (1989) in Principles of Neuroloqy 4th ed., McGraw-Hill, Inc., New York.) In ~;~eA~es which cause extensive damage to striated muscle fibers, intracellular muscle fiber enzymes leak out of the cell and enter the bloodstream.
One of the most commonly measured enzymes, and one of the most sensitive measures of muscle damage, is creatine phosphokinase (CPK). Serum CPK levels are used to monitor muscle damage in certain neuromuscular diseases, and as a marker for effective treatment. In disease which cause denervation paralysis with muscular atrophy, such as ALS, serum CPK levels may be elevated up to 2-3 times normal. This phenomenon, occurring in almost half of all patients with ALS, probably represents ongoing muscle damage secondary to progressive denervation.
A stAn~Ardized test has been developed to measure the clinical deficit and progression of ALS. The test battery, developed by the Neuromuscular Research Unit WO94/17818 ~ 5 4 0 PCT~S94/01227 at the New England Medical Center is referred to as the Tufts Quantitative Neuromuscular Exam or TQNE. See, Andres et al. NeuroloqY 38: 409-413 (1988). The TQNE
is a validated test battery specifically designed to measure the motor function and strength of ALS patients as the disease progresses. The battery includes testing of five major functional areas: bulbar, respiratory, arm, leg, and fine motor activities.
The present invention includes the use of the protein neurotrophic factor ciliary neurotrophic factor (CNTF) to treat ALS. Neurotrophic factors are naturally occurring proteins that promote the survival and functional activities of nerve cells. Neurotrophic factors have been found in the target cells to which an innervating nerve cell connects. Such target-derived neurotrophic factors regulate the number of contacts formed between innervating nerve cells and the target cell population, and are necessary for the survival and maintenance of these nerve cells.
Neurotrophic factors are also found in cells that are not innervated. An example of such a neurotrophic factor is CNTF. Human CNTF and the gene encoding human CNTF are described in detail in United States patent numbers 4,997,929, 5,141,856 and co-pending United States patent application serial number 07/857,544 filed March 24, 1992. Each of these documents are specifically incorporated herein by this reference.
Although the biological role of CNTF has not been conclusively established, CNTF appears to be released upon injury to the nervous system and may limit the extent of injury or neuronal damage.
Highly-purified CNTF has been shown to support the survival in cell cultures of chick embryonic parasym-pathetic, sympathetic, sensory, and motor neurons.
There is significant evidence to support the proposition that CNTF is a neurotrophic factor for peripheral primary neurons in vivo and in vitro. See, WO94/17818 PCT~S94/01~7 ~15~0 U.S. patent application serial number 07/735,538 filed July 23, 1991, specifically incorporated herein by this reference.
In the present invention, methods are provided for the treatment of ALS by the administration of ciliary neurotrophic factor (CNTF).

SUMMARY OF THE lNV~NllON
The present invention includes methods for the treatment of ALS by administering a human protein ciliary neurotrophic factor to a patient in need thereof. In particular, the invention provides methods for administering therapeutically effective amounts of CNTF by therapeutically effective routes of administration in order to treat patients suffering from ALS.
In one preferred embodiment of the invention, recombinant human CNTF is administered to human patients suffering from ALS in doses of less than about l0 ~g/kg/day. More specifically, daily doses of about 2-3 ~g/kg/day are utilized.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention as claimed.

DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
Reference will now be made in detail to the presently preferred embodiments of the invention, which, together with the following examples, serve to explain the principles of the invention.
The present invention includes a method for r treating a patient suffering from ALS by administering to that patient the human neurotrophic factor CNTF.
In one embodiment of this invention, preferred CNTFs are naturally occurring proteins. The naturally-occurring proteins are preferred in part because they WO94/17818 ~15 ~ 5 ~0 PCT~S94/01227 pose a possibly lower risk of producing unforeseen and undesirable physiological side effects in patients treated therewith. Human CNTFs are preferred for use in this invention. However, to the extent that non-human CNTFs are substantially equivalent to human CNTFsand possess equivalent biological activity, they are considered to be within the scope of this invention.
For purposes of the specification and claims, a protein is deemed to be "naturally-occurring" if it or a substantially equivalent protein can be found to exist normally in healthy humans. "Naturally-occurring" proteins specifically includes forms of proteins found to exist in healthy humans that are partially truncated at the amino or carboxyl terminus of such proteins or that have amino acids that are deamidated or otherwise chemically modified.
"Naturally-occurring" proteins may be obtained by recombinant DNA methods as well as by isolation from cells which ordinarily produce them. "Naturally-occurring" also encompasses proteins that contain orlack an NH2-terminal methionyl group as a consequence of expression in E. coli.
"Substantially equivalent" as used throughout the specification and claims is defined to mean possessing a very high degree of amino acid residue homology (See generally M. Dayhoff, Atlas of Protein Sequence and Structure, vol. 5, p. 124 (1972), National Biochemical Research Foundation, Washington, D.C., specifically incorporated herein by reference) as well as possessing comparable biological activity.
Substantially equivalent proteins of this invention includes proteins that have a significant degree of homology with the naturally occurring protein, but have been modified to contain a limited number of mutations or deletions within the amino acid sequence. Only such modified proteins retaining the biological activity of the naturally occurring proteins WO94/17818 PCT~S94/01227 ~5~ 3 ~

are included within the definition. Those skilled in the art are competent to prepare mutated proteins based on the known sequence of the naturally occurring proteins according to well known procedures without undue experimentation. The biological activity of such mutations may also be determined (without undue experimentation) according to the procedures described herein by those skilled in the art.
"Biological activity" as used throughout the specification and claims refers to the natural neurotrophic activity of the CNTF proteins of this invention. One measure of the biological activity of CNTF accepted by those skilled in the art is the ability to support the survival in cell cultures of chick embryonic ciliary ganglia as described in U.S.
patent 5,011,914. A protein is considered to have the same or comparable biological activity as the naturally occurring proteins of this invention if the protein has a specific activity within two orders of magnitude as the naturally-occurring proteins of this invention.
Particularly preferred CNTFs of the present invention are the naturally-occurring proteins that have previously been described in a currently pending United States patent application. This application is U.S. Patent Application Serial No. 07/857,544 filed March 24, 1992 of Collins et al., which is entitled "Purified Ciliary Neurotrophic Factor." (See also, U.S. patents 5,011,914; 5,141,856; and 4,997,929).
Other preferred forms of CNTF are described in U.S.
Patent Application Serial No. 07/753,176 filed August 30, 1992 of Collins et al., which is entitled "Purification of Recombinant Ciliary Neurotrophic Factor and C-Terminal Truncated Ciliary Neurotrophic Factor". Each of these patents and patent applications are specifically incorporated herein by reference.
The nucleic acid sequences of the genes encoding human and animal CNTFs and the amino acid sequences of WO94/17818 ~1 ~ 5 ~ 4 0 PCT~S94/0~27 such protein5 are given in U.S. patent numbers 4,997,929 and 5,14l,856. The present invention encompasses non-glycosylated forms of CNTF as well as truncated forms of the naturally-occurring and recombinant CNTF proteins as described in the above patents. In a further embodiment, CNTF is modified by attachment of one or more polyethylene glycol (PEG) or other repeating polymeric moieties. In the preferred embodiment of this invention the CNTF used is naturally-occurring recombinant human CNTF (rhCNTF).
Methods for producing the CNTFs of this invention are also disclosed in the above-mentioned patents. One disclosed method consists of isolating CNTF from various sources, such as peripheral nerve tissues. A
second disclosed method involves isolating the genes responsible for coding CNTF, cloning the gene in suitable vectors and cell types, and expressing the gene in order to produce the CNTF. The latter method, which is exemplary of recombinant DNA methods in general, is a preferred method of the present inventiori. Recombinant DNA methods are preferred in part because they are capable of achieving comparatively higher amounts at greater purity.
Preferably, the above described CNTFs are produced by the aforementioned method in "substantially pure"
form. By "substantially pure" it is meant that CNTF, in an unmodified form, has comparable biological activity to the purified CNTF described in United States patent 4,997,929. It is to be recognized, however, that derivatives of CNTF may have different specific activities. In a preferred embodiment of the present invention, a therapeutic composition comprising CNTF is administered in an effective amount to patients in order to effectively treat the symptoms of ALS.
Because it is possible that the function of the preferred CNTFs is imparted by one or more discrete and separable portions of the CNTF protein, it is also W094/17818 PCT~S94/0~27 ~ 4 ~ -8-envisioned that the method of the present invention could be practiced by a~;n;~tering a therapeutic composition whose active ingredient consists of that portion (or those portions) of CNTF which controls (or control) CNTF function.
The therapeutic composition of the present invention is preferably administered parenterally by injection. In the most preferred embodiment the parenteral administration is subcutaneous. Also, other effective administration forms, such as parenteral slow-release formulations, intrathecally by continuous infusion from an implanted pump, inhalant mists, orally active formulations, or suppositories, are also envisioned. The CNTF of this invention is preferably formulated with a pharmaceutically acceptable carrier.
A pharmaceutically acceptable carrier is a carrier that is not harmful to the patient and does not degrade, deactivate, or in any other way h; n~er the effects of the CNTF. One preferred carrier is physiological saline solution, but it is contemplated that other pharmaceutically acceptable carriers may also be used.
In one preferred embodiment it is envisioned that the carrier and the CNTF constitute a physiologically-compatible, slow-release formulation. The primary solvent in such a carrier may be either aqueous or non-aqueous in nature. In addition, the carrier may contain other pharmacologically-acceptable excipients for modifying or maint~;ning the pH, osmolarity, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor of the formulation. Similarly, the carrier may contain still other pharmacologically-acceptable excipients for modifying or maint~; n; ng the r stability, rate of dissolution, release, or absorption of the CNTF. Such excipients are those substances usually and customarily employed to formulate dosages for parenteral administration in either unit dose or multi-dose form or for intrathecal delivery by ~ WO94/17818 ~15 5 ~ 4 0 PCT~S94/01227 continuous or periodic infusion from an implanted pump or intrathecally by periodic injection.
Once the therapeutic composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or dehydrated or lyophilized powder. Such formulations may be stored either in a ready to use form or requiring reconstitution immediately prior to administration. The preferred storage of such formulations is at temperatures at least as low as 4C
and preferably at -70C. It is also preferred that such formulations cont~i~;ng CNTF are stored and administered at or near physiological pH. It is presently believed that storage and administration in a formulation at a pH below approximately p~ 5.5 and above approximately pH 8.0 is undesirable.
Preferably, the manner of parenterally administering the formulations cont~;ning CNTF is via a subcutaneous or intramuscular route. The most preferred a~ini~tration is parenterally via a subcutaneous route. To achieve the desired dose of CNTF, single or repeated subcutaneous or intramuscular injections may be administered. It is believed that the administration of CNTF in doses below approximately 0.005 ~g/kg/day may not be effective, while the administration of doses of greater than 10mg/kg/day may have undesirable side effects. In a preferred embodiment of the invention the dose of CNTF is between 0.5-50 ~g/kg/day. To treat or prevent the progression of ALS, it may be desirable to administer the CNTF
periodically. The periodic administrations may constitute monthly, bi-weekly, weekly, daily or hourly regimes. The required frequency of administration will be apparent to those treating the patient based on standard observational techniques.
In Example 2 below, patients suffering from ~LS
were administered daily injections of rhCNTF in dosages WO94117818 ~ t ~ Q PCT~S94/01227 including 2 ~g/kg/day, 5 ~g/kg/day, lO ~g/kg/day and 20 ~gkg/day. The progression of the ALS symptoms in the patients were monitored by the TQNE evaluation.
Surprisingly, as seen in the results shown in Tables III and IV, the most positive trends were seen in the patient group which had been administered 2 ~g/kg/day.
Thus, in a preferred embodiment of the invention, patients suffering from ALS are administered rhCNTF in doses of less than about lO ~g/kg and preferable about 2-3 ~g/kg. Further, in the preferred embodiment the rhCNTF is administered once daily.
It is also contemplated that certain formulations cont~;n;ng CNTF are to be administered orally.
Preferably, CNTF which is administered in this fashion is encapsulated. The encapsulated CNTF may be formulated with or without those carriers customarily used in the compounding of solid dosage forms.
Preferably, the capsule is designed so that the active portion of the formulation is released at that point in the gastro-intestinal tract when bioavailability is maximized and pre-systemic degradation is minimized.
Additional excipients may be included to facilitate absorption of CNTF. Diluents, flavorings, low melting point waxes, vegetable oils, lubricants, suspending agents, tablet disintegrating agents, and binders may also be employed.
Regardless of the manner of administration, the specific dose is calculated according to the approximate body weight or surface area of the patient.
Further refinement of the calculations necessary to determine the appropriate dosage for treatment involving each of the above mentioned formulations is routinely made by those of ordinary skill in the art and is within the ambit of tasks routinely performed by them without undue experimentation, especially in light of the dosage information and assays disclosed herein.
These dosages may be ascertained through use of the ~ W094/17818 2 ~ ~ 5 5 4 0 PCT~S94/01227 established assays for determining dosages utilized in conjunction with appropriate dose-response data.
According to the present invention, a patient in need of a treatment for ALS is administered a therapeutically effective amount of CNTF. As described above, the dosage sufficient to deliver a "therapeutically effective amount" of CNTF can be determined by those of ordinary skill in the art without undue experimentation. A "therapeutically effective amount" may be defined as the amount of CNTF
sufficient to giVe rise to subjective or objective improvements in the condition of the patient suffering from ALS.
It should be noted that the CNTF formulations described herein may be used for veterinary as well as human applications and that the term "patient" should not be construed in a limiting manner. In the case of veterinary applications, the dosage ranges should be the same as specified above.
It is understood that the application of teachings of the present invention to a specific problem or environment will be within the capabilities of one having ordinary skill in the art in light of the teachings contained herein. Examples of representative uses of the present invention appear in the following examples.

~MPLE l:
A number of human subjects with amyotrophic lateral sclerosis (ALS) were given subcutaneous doses of recombinant human CNTF (rhCNTF) as part of an open-t label, asc~n~;ng study assessing the safety, tolerability and pharmacokinetics of CNTF. During a portion of this study, patients were given a single subcutaneous administration of rhCNTF each day for 28 days. (The rhCNTF was prepared in an E. coli expression system as described in U.S. patent numbers WO94/17818 PCT~S94/01227 4,997,929 and 5,14l,856 and U.S. patent application serial no. 07/753,176).

Inclusion criteria: Patients included in the test met the following criteria:
1. Unequivocal diagnosis of uncomplicated ALS
2. The patient was male or female between the ages of 21-85 years.
3. The patient, if female, was postmenopausal for at least two years, surgically sterile, or, if the patient was of childbearing potential, she had been practicing a method of birth control considered effective and medically acceptable by the investigator for a minimum of 2 months prior to the study and at least 2 months after the study ended.
4. The patient was an outpatient at the time of enrollment.
5. The patient was willing to be housed for 7 days in a medical unit, and was able to comply with the study visit schedule.
6. The patient's laboratory values for white blood cells (WBCs) and differential, hemoglobin, hematocrit, platelets, serum electrolytes, SGOT, SGPT, alkaline phosphatase, BUN an creatinine, total bilirubin, and urinalysis were within clinically acceptable limits.
7. The general physical condition of the patient was such that the investigator considered the patient to be acceptable for this study and would survive at least six months.
8. The patient was given an informed consent or assent that has been approved by an institutional review board.

Exclusion criteria: Patients were excluded from the WO94/17818 2 1 5 S~ ~0 PCT~S94/01227 test if they met any of the following criteria:
1. The patient has uncontrolled (over the last 30 days), clinically significant cardiovascular, pulmonary, endocrine, or gastrointestinal disease. The patient has clinically significant hematologic, metabolic, hepatic, or renal disease.
2. The patient's FVC and/or FEV1 is < 40% of predicted.
3. The patient has evidence for GMl antibodies, paraproteinemia, familial ALS, or "pure"
motor syndromes (Spinal muscular atrophy, etc.) 4. The patient is lactating or pregnant.
5. The patient has received another investigational drug within the past 30 days.
6. The patient has any other major neurologic disease in addition to ALS.
7. The patient has had a major surgical operation or severe infection within one month prior to the day of dosing.
8. The patient has a history of recent ethanol or drug abuse, or noncompliance with treatment on other experimental protocols.
9. The patient has limited mental capacity such that he/she cannot provide written informed consent, information regarding adverse experiences or comply with evaluation procedures.
Clinical Supplies The rhCNTF was supplied in a sterile solution ready for injection. This solution was comprised of a Tris (lOmM) and sodium phosphate buffered solution (lOmM) at pH 7.2, containing 205 mM sodium chloride, 10% v/v glycerol, 0.2% w/w polysorbate 80, and 0.1%
human serum albumin. The concentration of rhCNTF was WO94/17818 PCT~S94/01227 2 ~ -14-either l.0 mg~mL or 4 mg/mL. The rhCNTF and placebo were packaged in 3 cc glass vials with Teflon-faced butyl rubber stoppers. Each vial contained l.7 mL of formulated material. This study also used a placebo which was the vehicle for dilution of rhCNTF. The rhCNTF and placebo were stored frozen at -20 or -70 degrees Celsius. Frozen, formulated rhCNTF and placebo were thawed at room temperature. Prior to use, the liquid formulations were gently inverted several times to afford a homogenous solution.
Dosing of rhCNTF was performed with a l or 3 cc syringe fitted with a 27 gauge needle. In order to administer very low doses, for which the volume of l mg/mL solution is too small to measure accurately, a solution of O.l mg/mL was produced by adding 0.43 mL of l mg/mL solution to one vial of placebo (l.7 mL).

Anecdotal Results Groups of six patients each were daily injected with 0.002, 0.005, O.Ol or 0.02 mg/kg of rhCNTF. 7 placebo patients at various dosage amounts were included in the study. Anecdotal results of the patients completing the 28 day regime are given below in Table I.
In summary, 2 of the 7 placebo patients experienced subjective improvement. Subjective improvement or a decrease in hyperreflexia, cramps or fasciculations on exam was reported by 15 of the 21 ALS
patients receiving rhCNTF who completed the trial.
CPK Results Serum levels of creatine phosphokinase (CPK) were measured in the test group of patients at various times for most patients, levels of serum CPK were determined prior to the initiation of a~r;ni~tration, on the day the treatment began and weekly thereafter. The results of these measurements are shown in Table II below. In ~ WO94/17818 21~ 5 ~ 4 0 PCT~S94/01227 Table II, under the column Drug/Placebo, D represents patients who received CNTF and P represents patients who received placebo, and the number represents the dosage of CNTF in mg/kg/day.
In summary, in patients who received daily injections of rhCNTF, serum CPK levels decreased in 5 of 6 patients, receiving 0.005 mg/kg/day of rhCNTF, 6 of 6 patients receiving 0.0~ mg/kg/day of rhCNTF and 3 of 3 patients receiving 0.02 mg/kg/day. Patients receiving placebo showed no obvious correlations to a decrease in serum CPK levels.
It is reasonable to conclude from this data that the reduction in CPK correlates to a reduction in muscle damage or breakdown. This result may be a function of a primary protective effect on muscle cells or from a secondary protective effect from improvement in the function of innervating motor neurons. Either explanation is possible in that CNTF receptors are found on both neurons and on muscle cells.
Example 2 ALS patients meeting the inclusion and exclusion criteria set forth in Example 1 were given a daily single subcutaneous administration of rhCNTF. The condition of the patients in the study was monitored by TQNE.
The Neuromuscular Research Unit at the New England Medical Center has developed a st~n~rzied test battery to measure the clinical deficit in ALS, the Tufts Quantitative Neuromuscular Exam (TQNE). See, Andres, PL, et al. Neurology 1986; 36: 937-941 (Quantitative motor assessment in amyotrophic lateral sclerosis); and Andres, PL, et al. Neurology 1988; 38: 409-413 (Use of composite scores (Megascores) to measure deficit in ALS). The major component of the TQNE is the measurement of maximum voluntary isometric contraction (MVIC) of 10 muscle groups in the arms and lO muscle wo 94,l78l8 ~ ~ 5 5 ~ ~ ~ PCT~S94/01227 groups in the legs using a strain guage tensiometer.
The TQNE is most commonly used to measure disease progression and efficacy of investigational agents.

The TQNE measurements obtained at a given visit are transformed to megascores as described in Andres 1988 reference sura, by averaging the Z-transformed items (the raw score minus the mean, divided by the st~n~rd deviation of the TUFTS ALS population for that score) that compose that category.

Table 3 shows TQNE changes over 28 days of treatment in patients receiving daily subcutaneous injections of rhCNT~. The left column lists muscle groups and their respective megascores. Each column group to the right of this is a dose level (placebo, 2 5, lO, and 20 ~g/kg/day). Individual columns group in each are patients who had evaluable TQNEs at baseline and at Day 28. A "+" sign demonstrate a 28-day score better than the baseline score. A "-" sign demonstrates a 28-day score worse than the baseline score. "@" signifies no changes. Table 4 shows a summary table of megascore improvement per dose group.

Although changes in this small number of patients are not statistically significant, there is a positive trend in the 2 ~g/kg/day dose group.

TABLE I

DO8E L~ L PATIBNT ~ RY
0.002 mg\kg\day Pt. 2-0002; walking improved (more upright, longer distances) by 2nd week of study, lnsted until 3 weeks after study.
Pt. 2-0004; d~_..aso~ frequency of falling; before study, OOD; during study, 4 times over 28 days. One week after CNTF stop,oed, returned to 000.
Pt. 2-0006; improved movement of toes on left foot, still present one month after stopping CNTF.
Placebo; ; .:_ ~ in speech and climbing stairs, as ~ell as ~ _rea3 fasciculations.
Pt. 3-0001; hyperreflexin diminished.
Pt. 3-0002; "perceived i o:.
0.005 Placebo; improved strength and longer ability to use walker.
Pt. 4-0101; fewer cramps Pt. 4-0102; fewer fasciculations Pt. 3-0104; strength improved, walked without cane.
Pt. 3-0105; cramps stop,oed after day 2, walking and speed improved 0.01 Pt. 2-0203; by secor,d week, reported improved ability to use a straw (pull liquid into mouth), which persisted 2 weeks after end of study.
Pt. 2-0201; fewer cramps and fasciculations, more hand motility Pt. 2-0203; fewer fasciculations Pt. 2-0204; fewer fnsciculations 0.02 Pt. 5-0301, increased strength in arms and hands, started within several days nfter starting CNTF. Also, not dropping things as much. Neuro exam ' ,~.
Pt. 3-0304; improved left foot/leg strength, hyperreflexia diminished, less de~ on cane.

N ~ N O ~ X ~ X X ~ ~ N
N ~ N Ul , NUl o -- CO ~ ~ oO N3~ O

N N N _ ~ ~ -- r~ o O U~
H

-- O ~ _ 0 ~ O r-~

~Nj~ C$ ~~ N~ N~ ~ ~ N ~ N N N ` ~ 00 N ~ N _ N ~ ' ' r Q _ N N N N O N N NIt~ O O trt In Ut U~ O
Il 1111111~ 111111 111111 11 1111 11 11 11 11 11 Q Q ~ Q C Q Q Q Q ~L ~ Q Q Q Q Q Q

OOOOOOOOOOOO------O

WO 94/17818 215 ~5 ~ ~ . PCT/US94/01227 o ;~; N ~ G C~

NlC , N ~ ~ E

~ _ ~ . ~ ;~ ~ u~ , 8 o ~ ~ 8 lC ;~ - $ ~ X Si;
~ N N NN _ N

et:
O æ - ~ N ~8 O` ' `
X

O O O O O o N N o U ~1111 U11 11 11 11 Cl OI--~O ~ G 0 1--No No No oN No No No oN o ~C~ i 5 ~ -20-Z N ~ J
W ~,,, ...... ,, 3 ~
~ N . ~ . . . . ~ . .
~, - ~ .

H U~ a H O ~ ~ ' ~ ' I I ' ' ' ' t 0 ~3 ~ W , ' ' ' , . , ~ t W '~ a ~ , ~ .

. .
O' _ N ~ . ~ I , , , ~ , , , ~ I
~; N ~
U~
~n , t ' ' ' ~ . .

N ~ ~ ~ t I ~ . ~ ~ +

X ~ g ~ ~

WO 94/17818 21 S 5 5 4 ~ PCT/US94101227 o -o O ~

o o o o o o l ~ ~ ~
o o o z o o 3 ~ o o o N I ~ I ' ' O O O U~

1 ~

.~ ' ~ ~n C I I ~ E;

O ~ S
)'~ 0 0 ~ S~
O
a ~ ~ ~, c g J ~ _ K ~ _iY ~ 1~ C~
+ I ~

~1355 4~3 o~ o o o ~ o U ~ X
~;
o o o o o ~n _ W
Z ,c a ~ ~ 0 u~

~ 0 ~` 1 0 ~ o ~C
A ~

:Z

Pl ~ N

0 0 0 ~
m

Claims (11)

CLAIMS:
1. A method for the treatment of amyotrophic lateral sclerosis (ALS) in patients suffering therefrom comprised of the administration of a therapeutically effective amount of ciliary neurotrophic factor (CNTF) in a pharmaceutically acceptable carrier.
2. The method of claim 1 wherein said CNTF is human CNTF.
3. The method of claim 1 wherein said CNTF is naturally occurring CNTF.
4. The method of claim 1 wherein said CNTF is administered parenterally.
5. The method of claim 4 wherein said CNTF is administered subcutaneously.
6. The method of claim 4 wherein said CNTF is administered intramuscularly.
7. The method of claim 1 wherein said CNTF is administered in a dose of between 0.005 µg/kg/day and 10 mg/kg/day.
8. The method of claim 7 wherein said CNTF is administered in a dose of between 0.5 and 50 µg/kg/day.
9. A method for the treatment of amyotrophic lateral sclerosis (ALS) in patients suffering therefrom comprised of the administration of a dose of less than about 10 µg/kg/day of ciliary neurotrophic factor (CNTF) in a pharmaceutically acceptable carrier.
10. The method of claim 9 wherein said CNTF is administered in a dose of about 2-3 µg/kg/day.
11. A method for the treatment of amyotrophic lateral sclerosis (ALS) in patients suffering therefrom comprised of the subcutaneous administration of a dose of about 2-3 µg/kg/day of recombinant human ciliary neurotrophic factor (rhCNTF) in a pharmaceutically acceptable carrier.
CA 2155540 1993-02-08 1994-02-03 Methods for treating amyotrophic lateral sclerosis with cntf Abandoned CA2155540A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US1521893A 1993-02-08 1993-02-08
US08/015,218 1993-02-08
US11644093A 1993-09-03 1993-09-03
US08/116,440 1993-09-03

Publications (1)

Publication Number Publication Date
CA2155540A1 true CA2155540A1 (en) 1994-08-18

Family

ID=26687105

Family Applications (1)

Application Number Title Priority Date Filing Date
CA 2155540 Abandoned CA2155540A1 (en) 1993-02-08 1994-02-03 Methods for treating amyotrophic lateral sclerosis with cntf

Country Status (4)

Country Link
EP (1) EP0682525A1 (en)
AU (1) AU6169894A (en)
CA (1) CA2155540A1 (en)
WO (1) WO1994017818A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0831883A1 (en) * 1995-05-12 1998-04-01 The Rockefeller University Treatment of alzheimer disease by modulation of synapsins
WO1997012635A1 (en) * 1995-10-02 1997-04-10 Cytotherapeutics, Inc. Method for treating amyotrophic lateral sclerosis
AU8021098A (en) * 1997-06-13 1998-12-30 Roche Diagnostics Gmbh Improvement of the regeneration of myelin sheaths
WO2002006341A1 (en) * 2000-07-14 2002-01-24 Children's Medical Center Corporation A trophic factor capable of producing a neurosalutary effect in a subject
AU2011239512B2 (en) 2010-04-16 2016-01-21 Biogen Ma Inc. Anti-VLA-4 antibodies

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4923696A (en) * 1987-05-04 1990-05-08 Baylor College Of Medicine Method to prepare a neurotrophic composition
US5011914A (en) * 1989-01-05 1991-04-30 Collins Franklin D Purified ciliary neurotrophic factor
US4997929A (en) * 1989-01-05 1991-03-05 Synergen, Inc. Purified ciliary neurotrophic factor
US5093317A (en) * 1989-06-05 1992-03-03 Cephalon, Inc. Treating disorders by application of insulin-like growth factor
IE903130A1 (en) * 1989-09-15 1991-03-27 Regeneron Pharma Ciliary neurotrophic factor

Also Published As

Publication number Publication date
WO1994017818A1 (en) 1994-08-18
AU6169894A (en) 1994-08-29
EP0682525A1 (en) 1995-11-22

Similar Documents

Publication Publication Date Title
US8207115B2 (en) Treatment of cartilage disorders with FGF-18
EP1993589B1 (en) Treatments for neurological disorders
KR20070085838A (en) Method for treatment of movement disorders
JPH11147829A (en) Treatment for treating behavioral abnormality and paraesthesia while reducing pain
EP0639079A1 (en) Methods for treating interleukin-1 and tumor necrosis factor mediated diseases
US20070225368A1 (en) Therapy for Multiple Sclerosis
ES2392883T3 (en) Treatment of cartilage disorders with FGF-18
US9289493B2 (en) Method for treating inflammatory conditions with mucosally administered interleukin-2
CN115397443A (en) Stem cell-derived exosomes comprising pain modulators and uses thereof
KR20130016127A (en) Treatment of traumatic brain injury
JP2019506439A (en) Targeted therapeutic lysosomal enzyme fusion proteins, related formulations and uses thereof
PT792160E (en) NEUROTROPHIC FACTOR DERIVED FROM GLIAL CELLS USED AS A NEUROPROTECTOR AGENT
JP2001512447A (en) How to administer amifostine
JP2003520237A (en) Combinations containing mirtrazapine and gepirone for the treatment of depression and related diseases
CA2155540A1 (en) Methods for treating amyotrophic lateral sclerosis with cntf
Tomlinson et al. Proximal myopathy after perhexiline maleate treatment.
Tsao et al. The childhood muscular dystrophies: making order out of chaos
US6143714A (en) Methods of using hepatocyte growth factor to promote survival, growth and differentiation of motor neurons
JP2009504636A (en) Pharmaceutical composition for treatment of nerve injury comprising plasma or serum
US11969522B2 (en) Use of immune modulators to improve nerve regeneration
US20220362219A1 (en) Retina regeneration through epigenetics manipulation
JP7350019B2 (en) Compositions for treating amyotrophic lateral sclerosis (ALS) and methods of using the same for treatment
US5691313A (en) Methods of treating impotency with ciliary neurotrophic factor
EP1161441A1 (en) Compositions for promoting nerve regeneration
KR20230142502A (en) Use of LINK-TSG6 for treating osteoarthritis pain

Legal Events

Date Code Title Description
FZDE Dead