EP1406600A1 - Compositions and methods of administering tubulin binding agents for the treatment of ocular diseases - Google Patents

Compositions and methods of administering tubulin binding agents for the treatment of ocular diseases

Info

Publication number
EP1406600A1
EP1406600A1 EP02756487A EP02756487A EP1406600A1 EP 1406600 A1 EP1406600 A1 EP 1406600A1 EP 02756487 A EP02756487 A EP 02756487A EP 02756487 A EP02756487 A EP 02756487A EP 1406600 A1 EP1406600 A1 EP 1406600A1
Authority
EP
European Patent Office
Prior art keywords
recited
ocular
subject
combretastatin
approximately
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.)
Withdrawn
Application number
EP02756487A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP1406600A4 (en
Inventor
David Sherris
Mark Wood
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.)
Oxi Gene Inc
Original Assignee
Oxi Gene Inc
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 Oxi Gene Inc filed Critical Oxi Gene Inc
Publication of EP1406600A1 publication Critical patent/EP1406600A1/en
Publication of EP1406600A4 publication Critical patent/EP1406600A4/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/075Ethers or acetals
    • A61K31/085Ethers or acetals having an ether linkage to aromatic ring nuclear carbon
    • A61K31/09Ethers or acetals having an ether linkage to aromatic ring nuclear carbon having two or more such linkages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • A61P9/10Drugs 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to the administration of vascular targeting agents, particularly tubulin binding agents, for the treatment of ocular diseases.
  • the eye is fundamentally one of the most important organs during life. Because of aging, diseases and other factors which can adversely affect vision, the ability to maintain the health of the eye becomes all important. A leading cause of blindness is the inability to introduce drugs or therapeutic agents into the eye and to maintain these drugs or agents at a therapeutically effective concentration therein. Oral ingestion of a drug or injection of a drug at a site other than the eye provides the drug systemically. However, such systemic administration does not provide effective levels of the drug specifically to the eye and thus may necessitate administration of often unacceptably high levels of the agent in order to achieve effective intraocular concentrations.
  • the macula is a region of the retina that contains an elevated concentration of the photo-sensor cells that are responsible for fine-detail vision (a generalized anatomic diagram of the human eye is illustrated in Fig. 1).
  • Macular degeneration is the imprecise historical name given to a poorly understood group of diseases that cause the photo-sensor cells of the macula to lose function.
  • the result of macular degeneration is the loss of vital central vision and detailed vision.
  • a patient stricken with macular degeneration experiences a blank spot in the center of their visual field and often loses the ability to read small print. (Source: Macular Degeneration Foundation, San Jose, CA: www.eyesight.org)
  • the macula contains highly active photoreceptors that consume a great deal of energy. Generating this energy requires a rich supply of oxygen and nutrients.
  • the macula has one of the highest rates of blood-flow through its supply-vessels (a. .a. choroid). Anything that interferes with this rich blood supply can cause the macula to malfunction.
  • T e oxygen-deprived macula responds by producing cytokines that signal endothelial cell growth and neovascularization.
  • macular degeneration dry-form and wet-form.
  • Approximately 85% to 90% of the cases of macular degeneration are the dry type.
  • the deterioration of the retina is associated with the formation of yellow deposits under the macula known as drusen.
  • the deposition of drusen correlates with decrease in the thickness of retinal cells that comprise the macula.
  • the amount of central vision loss is directly related to the location and severity of the drusen-induced retinal thinning.
  • the dry-form of macular degeneration tends to progress more slowly than the wet- form of the disease.
  • There is no effective treatment for dry-form macular degeneration A small percentage individuals suffering from the dry-form of macular degeneration progress to the wet-form of macular degeneration.
  • Fig. 2 illustrates a normal macula and dry-form macular degeneration.
  • the wet-form of macular degeneration is a rapidly progressing disease that almost always results in severe vision loss.
  • Vision-loss associated with Wet macular degeneration is the result of sub-retinal neovascularization.
  • the rapid growth of the sub-retinal blood vessels causes the overlying layer of retinal cells to buckle and become detached from the nutrient- rich choroid.
  • the proliferating vessels penetrate the retina and infiltrate the vitreous humor.
  • Fig. 3 illustrates a normal macula and wet-form macular degeneration.
  • the current standard treatment for macular degeneration is Laser Photocoagulation.
  • An ophthalmologist performing laser photocoagulation locates the aberrant vessels with fluorescent angiography and selectively burns the vessels with the laser ablation technique.
  • a side effect of laser surgery is the destruction of the retinal layer immediately overlying the aberrant vessels.
  • Patients treated with laser photocoagulation have a measurable loss of vision immediately after treatment and this is an unacceptable negative side effect.
  • laser surgery is viewed as a stopgap treatment that is only moderately effective at slowing the disease.
  • Photodynamic therapy is the current state of the art treatment for macular degeneration.
  • the U.S. Food and Drug Administration approved verteporfin for injection (VisudyneTM developed by Ciba Vision & QLT) to treat the wet form of age-related macular degeneration.
  • a patient being treated with photodynamic therapy is injected with the photo- reactive compound (verteporfin) and immediately treated with a non-destructive ophthalmic laser.
  • the ophthalmologist performing the surgery identifies the aberrant vessels and directs the laser beam toward the aberrant vessels.
  • Verteporfin when activated by the laser, generates a transient burst of energy that effectively scorches any cells within the vicinity of the activated molecule.
  • Ionizing radiation is used to kill proliferating vessels (proliferating cells are more sensitive to radiation than quiescent cells). Ionizing radiation is usually administered in a beam large enough to expose most of the eye.
  • a group at the University of Southern Ireland reported that they had tried X-rays on a small number patients with the wet form of macular degeneration. Their positive results have been supported by several similar studies with X-rays done by other research teams in Europe.
  • Another debilitating ocular disease is Retinopathy of Prematurity (ROP).
  • ROP is an eye disease that occurs in a significant percentage of premature babies. The last 12 weeks of a full-term delivery (weeks 28 to 40) are particularly active months in the development of the fetal eye.
  • the pre-natal development of the retinal blood supply (choroid) initiates at the optic nerve on week 16 and progresses in a radial fashion towards the anterior region of the retina until birth (week 40). If birth is premature, the retinal vasculature does not have enough time to fully develop and the anterior edges of the retina become deprived of oxygen. The lack of anterior-retinal oxygenation is the underlying cause of ROP. (Source: The Association of Retinopathy of Prematurity and Related Diseases, Franklin, MI)
  • the ring of scar tissue may extend for 360 degrees around the inside of the eye. When this scar tissue contracts it pulls the retina and produces a retinal detachment. If enough scar tissue forms, the retina can become completely detached.
  • Premature neonates are at risk for developing ROP because they have been taken out of the protective environment of the uterus and are exposed to a variety of angiogenic stimuli, including medications, high levels of oxygen, and variations in light and temperature. Some or all of these factors may have an effect on the development of ROP. Fortunately, most premature infants do not develop ROP, and most infants with ROP improve spontaneously. If ROP does develop, it usually occurs between 34 and 40 weeks after conception, regardless of gestational age at birth.
  • cryotherapy A technique termed cryotherapy has been shown to have a beneficial effect for the treatment of ROP.
  • Cryotherapy involves placing a sub-zero probe on the outer wall of the eye (sclera). The probe causes a zone of ice crystallization on the retinal surface between the sclera and the vitreous.
  • Multiple applications of cryotherapy are performed in order to treat the entire avascular area, which is anterior to the neovascular ridge. Treatment of the ridge itself is avoided, since the ridge tends to bleed and cause vitreous hemorrhage if frozen.
  • the mechanism of action of cryotherapy is not completely understood. The working hypothesis is that the cryotherapy probably damages the avascular anterior retinal layer.
  • Cryotherapy was found to reduce the risk of retinal detachment from 43% in the untreated eyes to 21% in the treated eyes. Cryotherapy does, however, have potential complications; the procedure is often performed under general anesthesia which can be risky for premature infants.
  • Laser photocoagulation uses similar principles in the treatment of ROP.
  • the laser treatment is applied to the anterior retina that does not yet have a blood supply.
  • the purpose of the treatment is to eliminate the abnormal vessels before they lay down enough scar tissue to produce a retinal detachment.
  • the avascular anterior retina is marginally thinned by the laser reducing the need for oxygen and dampening the angiogenic stimuli, much like cryotherapy.
  • Laser therapy is superior to cryotherapy in that it is directed at the retina and not the entire thickness of the eye wall. Because laser therapy involves less tissue and is not painful, post-treatment inflammation is greatly reduced. When compared to cryotherapy, laser therapy is superior because there is a reduced need for anesthetics.
  • Scleral buckling involves placing a silicone band around the equator of the eye and tightening it to produce a slight indentation on the inside of the eye. This band relieves the traction of the vitreous gel pulling on the fibrous scar tissue and the retina. This allows the retina to flatten onto the wall of the eye and resume normal function. Infants who have had scleral buckling may maintain good vision in the eye, particularly if the macula did not detach. The encircling band usually needs to be removed some months or years later because the eye will continue to grow, producing gradually increasing compression of the globe and induced nearsightedness.
  • Vitrectomy involves making several small incisions into the eye, and using a suction/cutter device to remove the vitreous gel.
  • the vitreous is replaced with a saline solution to keep the eye formed, and the eye is able to maintain its shape and pressure indefinitely without the vitreous gel.
  • the scar tissue on the retina can be peeled or cut away, allowing the retina to relax and lay back down against the eye wall.
  • the retina may take some weeks for the retina to become re-attached after the surgery, and if holes or tears in the retina occur during the procedure, the retina usually will not re- attach.
  • the lens of the eye often has to be removed to allow complete dissection of the scar tissue, but some newer techniques are being tried that can preserve the lens.
  • the success rate for vitrectomy surgery for ROP is, however, somewhat limited.
  • the published anatomic success rate which means getting the retina reattached to the wall of the eye, ranges from 25% to 50% of patients undergoing surgery.
  • the functional success rate which means the ability to see well, is significantly lower.
  • Of eyes that have "successful" vitrectomy surgery (anatomic success) only about 1/4 are able to see well enough to reach out and grab an object or recognize patterns.
  • diabetes Another debilitating ocular disease occurs in patients who suffer from diabetes mellitus. Approximately 14 million Americans have diabetes mellitus. In addition to causing numerous systemic complications (such as kidney failure, hypertension, and cardiovascular disease), diabetes is one of the leading causes of blindness among working-age Americans. In fact, the risk of blindness to persons with diabetes is 25 times greater than that of the general population. Many patients with diabetic eye problems are asymptomatic despite the presence of vision-threatening disease. If diabetic eye disease is left untreated, it can lead to serious visual loss. Decreased vision due to diabetes can be caused by several mechanisms, and treatment needs to be tailored to the individual's needs. (Source: The Center for Disease Control, "The Prevention and Treatment of Diabetes Mellitus - A Guide for Primary Care Practitioners": www.cdc.gov/health/diseases.htm)
  • the first category is termed background diabetic retinopathy or non-proliferative retinopathy. This is essentially the earliest stage of diabetic retinopathy.
  • This stage is characterized by damage to small retinal blood vessels which results in the effusion of fluid (blood) into the retina. Most visual loss during this stage is due to the fluid accumulating in the macula. This accumulation of fluid is called macular edema, and can cause temporary or permanent decreased vision.
  • the second category of diabetic retinopathy is termed proliferative diabetic retinopathy. Proliferative retinopathy is the end result of diabetes-induced damage sustained by the retinal capillary bed (choroid). Damage to the choroid causes oxygen deprivation in the retina.
  • the retinal tissue responds to its anoxic environment by producing angiogenic cytokines that stimulate neovascularization.
  • neovascularization of the retina causes bleeding in the eye, retinal scar tissue, retinal detachments, and any of one of these symptoms can cause decreased vision or blindness.
  • Diabetics often also suffer from neovascular glaucoma, which manifests in rubeosis, blood vessels growing on the iris that causes closure of the angle.
  • Diabetic retinopathy can occur in both Type I diabetics (onset of diabetes prior to age
  • Type II diabetics onset after age 40
  • Type II diabetics onset after age 40
  • diabetic retinopathy may be present in a Type II patient at the time diabetes is discovered.
  • diabetic retinopathy depends upon multiple factors, including the type and degree of retinopathy, associated ocular factors such as cataract or vitreous hemorrhage, and the medical history of the patient. Treatment options include the same options that were discussed for ROP, namely laser photocoagulation, cryotherapy (freezing), and vitrectomy surgery. Blindness due to diabetic retinopathy is preventable in most cases. Intraocular cancerous tumors of any type are mostly uncommon. Ocular tumors are, however, extremely serious in that uveal (eye) cancers generally metastasize to and from other areas of the body.
  • Retinoblastoma is a cancer of one or both eyes which occurs in young children. There are approximately 350 new diagnosed cases per year in the Unites States. Retinoblastoma affects one in every 15,000 to 30,000 live babies that are born in the United States.
  • Retinoblastoma affects children of all races and both boys and girls.
  • the retinoblastoma tumor(s) originate in the retina, the light sensitive layer of the eye which enables the eye to see.
  • the treatment of retinoblastoma is individualized for each patient and depends upon the age of the child, the involvement of one or both eyes, and whether or not the cancer has spread to other parts of the body. If left untreated, the child could die.
  • Treatments for retinoblastoma include enucleation, external beam radiation, radioactive plaques, laser therapy, cryotherapy and chemoreduction.
  • Enucleation is the most common form of treatment for retinoblastoma. During an enucleation, the eye is surgically removed. This is necessary because it is the only way to remove the cancer completely. It is not possible to remove the cancer from within the eye without removing the entire eye. Although partial enucleation is possible for some other eye cancers, it is risky and may even contribute to the spread of the cancer for retinoblastoma patients. When both eyes are involved, sometimes the more involved or "worse" eye is enucleated, while the other eye may be treated with one of the vision-preserving treatments, such as external-beam radiation, plaque therapy, cryotherapy, laser treatment, and chemoreduction which are described below.
  • the vision-preserving treatments such as external-beam radiation, plaque therapy, cryotherapy, laser treatment, and chemoreduction which are described below.
  • Radioactive plaques are disks of radioactive material that were developed in the 1930's to radiate retinoblastoma.
  • Laser therapy is a non-invasive treatment for retinoblastoma.
  • Lasers effectively destroy smaller retinoblastoma tumors. This type of treatment is usually performed by focusing light through the pupil onto and surrounding the cancers in the eye.
  • a diopexy probe has enabled treatment of the cancer by aiming the light through the wall of the eye and not through the pupil.
  • Laser treatment is done under local or general anesthesia, usually does not have any post-operative pain associated with it, and does not require any post-operative medications.
  • Laser can be used alone or in addition to external-beam radiation, plaques, or cryotherapy.
  • Cryotherapy may also be performed on patients suffering from retinoblastoma.
  • Cryotherapy is performed under local or general anesthesia and freezes smaller retinoblastoma tumors.
  • a pen-like probe is placed on the sclera adjacent to the tumor and the tumor is frozen.
  • Cryotherapy usually has to be repeated many times to successfully destroy all of the cancer cells.
  • An adverse side effect of cryotherapy is that it causes the lids and eye to swell for 1 to 5 days; sometimes the swelling is so much that the children are unable to open their lids for a few days. Eye drops or ointment is often given to reduce the swelling.
  • Chemoreduction is the treatment of retinoblastoma with chemotherapy.
  • Chemotherapy is generally administered intravenously to the child, passes through the blood stream, and causes the tumors to shrink within a few weeks if successful.
  • Chemotherapy, with one or more drugs can be given once, twice, or more. Depending on the drug(s) and on the institution, the child may or may not be hospitalized during this process. After chemotherapy, the child is re-examined and the remaining tumor(s) are treated with cryotherapy, laser, or radioactive plaque. Children may require as many as twenty treatments with re-examinations of the eye under anesthesia every 3 weeks.
  • retinoblastoma can spread (metastasize) outside of the eye to the brain, the central nervous system (brain and spinal cord), and the bones.
  • chemotherapy is prescribed by a pediatric oncologist and is administered through the peripheral blood vessels or into the brain for months to years after initial diagnosis of metastatic disease.
  • Choroidal metastasis is the most frequently occurring intraocular malignancy and can be the initial manifestation of systemic malignancy. Choroidal metastases resemble nonpigmented melanomas. They have a similar appearance to melanoma on fluorescein angio-gram and show subtle echographic differences on ultrasonograms. Choroidal metastases, however, grow more rapidly and are more likely to cause large exudative retinal detachments.
  • Primary ocular lymphoma is one of the most interesting intraocular tumors. Its relationship with primary central nervous system lymphoma and the propensity of the tumor to proliferate in the subretinal pigment epithelial space, where no lymphoid tissue exists, are just two compelling aspects of this highly aggressive lymphoma.
  • the clinical manifestations of primary ocular lymphoma are notorious for mimicking benign uveitic entities and thus delaying the correct diagnosis for months.
  • the neoplastic cells in ocular lymphoma can remain confined to the space between the retinal pigment epithelium and Bruch's membrane. Because the vitritis associated with these aggregates of lymphoma often consists of reactive lymphocytes, vitreous biopsy can be nondiagnostic.
  • ocular lymphoma consists of large, cytologically atypical cells that stain positive for leukocyte common antigen. Aspirates are usually associated with large amounts of necrotic debris. Immunophenotypic analysis has been problematic in the past. Some early studies failed to find any surface markers and concluded that ocular lymphoma was a null- cell tumor. Pretreatment of cells with hyaluronidase has increased the yield of irnmuno- pathologic studies.
  • Choroidal melanoma is a primary cancer of the eye. It arises from the pigmented cells of the choroid of the eye and is not a tumor that started somewhere else and spread to the eye. Although some choroidal melanomas are more life-threatening than others, almost all should be treated as if they were malignant. Some choroidal melanomas appear to remain dormant and do not grow. Most enlarge slowly over time and lead to loss of vision. These tumors can spread to other parts of the body and lead eventually to death. Numerous cases have been reported of ocular melanoma metastasizing to the liver.
  • High-energy particles (helium ion or proton beam radiation) from a cyclotron can also be used to irradiate tumors.
  • Surgery is performed first to sew small metal clips to the sclera so that the particle beam can be aimed accurately.
  • Treatment is given over several successive days. The equipment needed for these treatments is available only in a few medical centers in the world. Good results have been reported in some patients, but many patients treated in this way have been followed for only a few years. Therefore, the long-term results of these forms of radiation therapy compared with the more commonly used plaque are unknown. Over the years, other treatments have been used for a small number of patients.
  • Another ocular cancer is intraocular melanoma, a rare cancer in which cancer cells are found in the part of the eye called the uvea.
  • the uvea contains cells called melanocytes, which contain pigment. When these cells become cancerous, the cancer is referred to as a melanoma.
  • the uvea includes the iris (the colored part of the eye), the ciliary body (a muscle in the eye), and the choroid (a layer of tissue in the back of the eye).
  • the iris opens and closes to change the amount of light entering the eye.
  • the ciliary body changes the shape of the lens inside the eye so it can focus.
  • the choroid layer is next to the retina, the part of the eye that makes a picture.
  • melanoma that starts in the iris, it may look like a dark spot on the iris. If melanoma is in the ciliary body or the choroid, a person may have blurry vision or may have no symptoms, and the cancer may grow before it is noticed.
  • the chance of recovery (prognosis) from intraocular melanoma depends on the size and cell type of the cancer, where the cancer is in the eye, and whether the cancer has spread.
  • There are treatments for all patients with intraocular melanoma Three types of treatment are commonly administered, namely surgery (removal of the cancer), radiation therapy (using high-dose x-rays or other high-energy rays to "kill" the cancer cells), and photocoagulation (destroying blood vessels that feed the tumor).
  • Surgery is the most common treatment of intraocular melanoma.
  • a doctor may remove the cancer using one of the following operations: - Iridectomy- removal of only parts of the iris;
  • Iridotrabeculectomy removal of parts of the iris and the supporting tissues around the cornea, the clear layer covering the front of the eye;
  • Radiation therapy can also be used to apply x-rays or other high-energy rays to the area where the cancer cells exist so as kill cancer cells and shrink the tumors. Radiation can be used alone or in combination with surgery. Photocoagulation treatment may also be used wherein a tiny beam of light, usually from a laser, is applied to the eye to destroy blood vessels and kill the tumor.
  • the subject invention provides such a therapy, providing for efficacious non-systemic administration of a tubulin binding agent for the treatment of ocular disease, with minimal side effects.
  • VTA vascular targeting agent
  • Neovascularization of ocular tissue is a pathogenic condition characterized by vascular proliferation and occurs in a variety of ocular diseases with varying degrees of vision failure.
  • the administration of a VTA for the pharmacological control of the neovascularization associated with non-malignant vascular proliferative disorders such as wet macular degeneration, proliferative diabetic retinopathy or retinopathy of prematurity would potentially benefit patients for which few therapeutic options are available.
  • the invention provides the administration of a VTA for the pharmacological control of neovascularization associated with malignant vascular proliferative disorders such as ocular tumors.
  • the blood-retinal barrier is composed of specialized nonfenestrated tightly- joined endothelial cells that form a transport barrier for certain substances between the retinal capillaries and the retinal tissue.
  • the nascent vessels of the cornea and retina associated with the retinopathies are aberrant, much like the vessels associated with solid tumors.
  • Tubulin binding agents, inhibitors of tubulin polymerization and vascular targeting agents may be able to attack the aberrant vessels because these vessels do not share architectural similarities with the blood retinal barrier.
  • Tubulin binding agents may halt the progression of the disease much like they do with a tumor- vasculature.
  • Systemic administration may be accomplished by administration of the tubulin binding agents into the bloodstream at a site which is separated by a measurable distance from the diseased or affected organ or tissue, in this case they eye.
  • Preferred modes of systemic administration include parenteral or oral administration.
  • Fig. 1 is a simplified front and side anatomic illustration of a mammalian eye
  • Fig. 2A illustrates normal macula
  • Fig. 2B illustrates dry-form macular degeneration
  • Fig. 2C illustrates wet-form macular degeneration
  • FIG. 3A and 3B are magnified photographs of a portion of the cornea showing the inhibition of vessel growth on Day 28 following in administration of CA4P administration in comparison with a vehicle control eye;
  • Fig. 4A and 4B illustrate microscopic histology of changes to the cornea (inhibition of vessel growth) on Day 28 following systemic administration of CA4P in comparison with a vehicle control eye.
  • Fig. 5A illustrates the effect of a single dose of CA4P the vascularization of an ocular tumor in an animal model of retinoblastoma.
  • Fig. 5B illustrates the degree of tumor regression in an animal model of retinoblastoma following repetitive dosing of CA4P.
  • the human eye possess several structurally unique properties: it is exposed to the environment, it is highly enervated, it has a high rate of blood flow in the choroid yet the anterior chamber and vitreous humor are completely avascular and isolated from the circulatory system.
  • the exceptional architecture of the eye provides ample opportunity for delivery of tubulin binding agents by one or more non-systemic methods of administration for the treatment of ocular conditions, diseases, tumors and disorders.
  • a simplified anatomic illustration of the eye is shown in Fig. 1.
  • neovascularization of ocular tissue is a pathogenic condition that occurs in a variety of ocular diseases and is associated with varying degrees of vision failure. Pharmacological control of neovascularization would potentially benefit patients suffering from diseases such as wet macular degeneration, proliferative diabetic retinopathy and retinopathy of prematurity.
  • Tubulin binding agents inhibit tubulin assembly by binding to mbulin-binding cofactors or cofactor-tubulin complexes in a cell during mitosis and prevent the division and thus proliferation of the cell.
  • Tubulin binding agents comprise a broad class of compounds which inhibit tubulin polymerization, and which generally function as tumor selective vascular targeting agents useful for cancer chemotherapy, as well as for other non-cancer applications such as ocular disease.
  • systemic administration does not generally provide effective levels of the drug specifically to the eye. Since drugs administered systemically may be metabohzed in the body before even reaching the eye, higher levels of the drug may need to be administered in order to achieve effective intraocular concentrations.
  • Non-systemic or local administration of drugs directly to the eye(s) of a patient suffering from an ocular disease allows the effective concentration of drug to be administered and benefits the patient immeasurably.
  • Ocular indications treatable by the non-systemic administration of the tubulin binding agents in accordance with the present invention include non-malignant vascular proliferative diseases characterized by corneal, retinal, or choroidal neovascularization, as well as malignant vascular proliferative diseases such as ocular tumors and cancers.
  • Corneal neovascularization occurs in the following: trachoma (Chlamydia trachomatis), viral interstitial keratitis, microbial keratoconjunctivitis, corneal transplantation and burns.
  • Retinal and/or choroidal neovascularization occurs in macular degeneration, diabetic retinopathy, and retinopathy of prematurity.
  • Anterior chamber neovascularization occurs in glaucoma.
  • non-systemic methods of administering tubulin binding agents contemplated by the present invention are: intravitreal administration (injection), sub- conjunctival administration, peri-ocular administration, sub-Tenon's injection, iontophoretic delivery, topical administration with ophthalmic drops, gels, or ointments, and via ocular insert or implant.
  • Tubulin binding agents may be administered intravitreally via an injection directly into the vitreous humor of the eye.
  • Tubulin binding agents may also be administered beneath the conjunctiva by sub-conjunctival injection, and around the eye via peri-ocular injection.
  • Tubulin binding agents may also be administered by injection into the sub-Tenon's space (under Tenon's capsule) with a blunt tip Connor Cannula.
  • the medical professional administering the dosage of tubulin binding agent can avoid puncturing the globe and damaging the optic nerve.
  • the injection site is cauterized and the space serves as a depot for the drug.
  • Administration into the sub-Tenon's space is less invasive than intravitreal injection.
  • a tubulin binding agent may be formulated as a biocompatible, biodegradable, and/or bioerodible ocular implant or insert containing the tubulin binding agent so as to provide slow release of the drug and maintenance of a therapeutically effective drug concentration for an extended period of time.
  • Drug-containing bioerodible ocular implants for implantation or insertion into a mammalian eye are described, for example, in U.S. Pat. No. 5,904,144 and U.S. Pat. No. 5,766,242, which are incorporated by reference herein in its entirety.
  • Ocular implants generally comprise a capsule that is placed in a desired location in the eye.
  • the capsule may include one or more medicaments or may include cells that produce a biologically active molecule for continuous, controlled delivery to the eye.
  • the amount of drug that may be employed in this embodiment will vary depending on the effective dosage of the drug and the rate of release from the insert or implant on or within the eye.
  • Iontophoresis uses an electrical current to drive the flux of ionic compounds across a cell membrane. This technique is currently utilized for transdermal delivery of ionic drugs.
  • the two principal mechanisms by which iontophoresis drives the transport of drugs are: (a) iontophoresis, in which a charged ion is repelled from an electrode of the same charge, and (b) electroosmosis, the convective movement of solvent that occurs through a charged "pore" in response to the preferential passage of counter-ions when the electric field is applied.
  • the tubulin binding agents may also be formulated for topical administration to the eye in the form of sterile, ophthalmic drops.
  • the preferred tubulin binding agent is combretastatin A4 ("CA4"), a potent vascular targeting agent.
  • CA4 is essentially insoluble in water. This characteristic interferes with the formulation of pharmaceutical preparations of this compound.
  • CA4P the more preferable prodrag form of combretastatin A4
  • the invention is not hmited in this respect, however, and formulations of CA4 may work as well or better than CA4P.
  • Combretastatins are derived from tropical and subtropical shrubs and trees of the Combretaceae family, which represent a practically unexplored reservoir of new substances with potentially useful biological properties.
  • Illustrative is the genus Combretum with 25 species (10% of the total) known in the primitive medical practices of Africa and India for uses as diverse as treating leprosy (See: Watt, J. M. et al, "The Medicinal and Poisonous Plants of Southern and Eastern Africa", E. & S. Livingstone, Ltd., London, 1962, p. 194) (Combretum sp. root) and cancer (Combretum latifolium).
  • Combretastatins have been found to be antineoplastic substances. Numerous combretastatins have been isolated, structurally elucidated and synthesized. U.S. Pat. Nos. 5,409,953 and 5,59,786 describe the isolation and synthesis of Combretastatins designated as A-l, A-2, A-3, B-l, B-2, B-3 and B-4. The disclosures of these patents are incorporated by reference herein in their entirety.
  • a related Combretastatin, designated Combretastatin A4 was described in U.S. Pat. No. 4,996,237 to Pettit, and which is incorporated by reference herein in its entirety.
  • CA4P is a derivative of the natural combretastatin A4 subtype described in U.S. Patent No. 5,561,122, the entire disclosure of which is incorporated by reference herein.
  • the preferred CA4P compound substitutes a disodium phosphate derivative for the -OH group in the CA4 structure and which allows metabolic conversion of CA4P back into the water insoluble CA4 in vivo.
  • the invention is not, however, limited to the phosphate derivative, and other prodrug moieties may be substituted for the -OH group in the CA4 compound.
  • phosphate prodrug salts other than the disodium salt of CA4P are expected to perform in substantially the same way for the purposes of this invention. Examples of other phosphate prodrug salts are described in PCT patent applications WO 02/22626 and WO 99/35150, the disclosures of which are incorporated herein.
  • CA4P is the first in a new class of drugs—anti-tumor vascular targeting agents—that shrink solid tumors by selectively targeting and destroying the tumor-specific blood vessels formed by angiogenesis.
  • Anti-tumor vascular targeting and angiogenesis inhibition are related cancer therapies that radically depart from conventional approaches to treating cancer. In contrast to traditional methods involving a direct attack on cancer cells, these new drugs target a tumor's life support system, the network of newly emerging blood vessels that form as a result of angiogenesis, the sprouting of new blood vessels from previously existing ones. Preclinical studies have shown that the use of these therapies can cause a tumor to shrink and ultimately disappear.
  • angiogenesis inhibitors and anti-tumor vascular targeting agents both target a tumor's blood vessels, they differ in their approach and in the end result.
  • angiogenesis inhibition the aim is to prevent tumor growth by inhibiting the formation of tumor-specific blood vessels that feed and sustain the tumor.
  • anti-tumor vascular targeting the goal is to obliterate tumors by selectively attacking and destroying their existing blood vessels, creating a rapid and irreversible shutdown of these blood vessels. Such an effect is not observed with anti-angiogenesis drugs. Only antivascular targeting activity can destroy existing blood vessels supporting tumor growth.
  • Combretastatin also has the ability to inhibit the proliferation of endothelial cells which produce and line new tumor vasculature (anti-angiogenic activity). Hence, it is thought that Combretastatin can behave both as a anti-tumor vascular targeting agent and as an anti- angiogenic drug. In preclinical studies, both therapies have been shown to leave blood vessels associated with normal tissue unaffected.
  • the present invention contemplates the administration of CA4P both alone, and/or in combination with current state of the art medicaments for the treatment of ocular diseases.
  • Vasculature formed by angiogenesis has also been observed in diseases other than cancer including diseases of the eye, e.g. macular degeneration, proliferative diabetic retinopathy and retinopathy of prematurity.
  • diseases of the eye e.g. macular degeneration, proliferative diabetic retinopathy and retinopathy of prematurity.
  • Preliminary work toward reducing such vasculature in an experimental eye model was carried out from the laboratory of Donald Armstrong, Ph.D., D.Sc, University of Florida, College of Veterinary Medicine, Division of Ophthalmology, who demonstrated that CA4P accelerated the regression rate of preformed vessels in the eye of experimental animal models.
  • Figs. 3A, 3B, 4A and 4B illustrate the regression of preformed vessels in the eyes of rabbits studied in this experiment.
  • CA4 and CA4P are currently undergoing clinical testing for treatment of a variety of diseases and indications including use as an anti-tumor vascular targeting agent, and as inhibitor of angiogenesis. Furthermore, CA4P has demonstrated the ability to treat ocular diseases, such as subretinal neovascularization.
  • the present invention also contemplates the use of synthetic analogs of the Combretastatins as described in Bioorg. Med. Chem. Lett. 11(2001) 871-874, 3073-3076, J. Med. Chem. (2002), 45: 1697-1711, WO 02/50007, WO 01/12579, WO 00/35865, WO
  • tubulin binding agents which may be administered as VTAs include the following agents or their prodrugs: 2,3-disubstituted Benzo[b]thiophenes (US Pat. Nos. 5,886, 025; 6,162,930, and 6,350,777), 2,3-disubstituted benzo[b]furans (WO 98/39323), 2- 3-disubstituted indoles (WO01/19794), disubstituted dihydronaphthalenes (WO01/68654), or Colchicine analogs (WO 99/02166).
  • additional non-cytotoxic prodrugs of vascular targeting agents which are converted to a substantially cytotoxic drug by action of an endothelial enzyme selectively induced at enhanced levels at sites of vascular proliferation are disclosed in WO00/48606.
  • tubulin binding agents which may be administered in accordance with the present invention include: taxanes, vinblastine (vinca alkaloids), colchicines (colchicinoids), dolastatins, podophyllotoxins, steganacins, amphtethiniles, flavanoids, rhizoxins, curacins A, ephothilones A and B, welwistatins, phenstatins, 2-strylquinazolin- 4(3H)-ones, stilbenes, 2-aryl-l, 8-naphthyridin-4(lH)-ones, and 5,6-dihydroindolo(2,l- a)isoquinolines.
  • the tubulin binding agents to the eye of a subject in need thereof, it is important to consider that the human eye possesses several structurally unique properties: it is exposed to the environment, it is highly enervated, it has a high rate of blood flow in the choroid yet the anterior chamber and vitreous humor are completely avascular and isolated from the circulatory system.
  • the exceptional architecture of the eye provides ample opportunity for alternative drug delivery methods.
  • four non-systemic modes of administration are contemplated by the present invention, namely intravitreal administration (injection), sub-Tenon's injection, iontophoretic delivery, implants/inserts and ophthalmic drop delivery.
  • neovascular retinopathies as well as ocular tumors, are thus a viable target for CA4P therapy and other tubulin binding agents for a variety of reasons, namely:
  • Tubulin binding agents may be able to attack the aberrant nascent vessels associated with the retinopathy because these vessels do not share architectural similarities with the BRB. Tubulin binding agents may halt the progression of the disease much like it does with a solid tumor vasculature. In addition, tubulin binding agents may able to cause the regression of nascent vessels as has been observed in various pre-clinical studies.
  • tubulin binding agents may be effective drugs when used in combination with current state of the art treatments.
  • CA4P When delivered systemically or nonsystemically, CA4P shows promise as a vascular targeting agent in animal models of corneal, retinal, or choroidal angiogenesis and in animal models with ocular tumors.
  • CA4P as well as other vascular targeting and tubulin binding agents show promise when delivered systemically in models of corneal, retinal, or choroidal angiogenesis, as well as other ocular diseases and tumors.
  • Preferred modes of systemic administration include parenteral and oral administration.
  • Parenteral administration is the route of administration of drugs by injection under or through one or more layers of the skin or mucous membranes.
  • Parenteral routes of administration include any route other than the oral-gastrointestinal (enteral) tract.
  • Parenteral administration includes the intravenous, intramuscular and subcutaneous routes.
  • compositions of the invention are formulated to be compatible with its intended route of administration.
  • Pharmaceutical compositions for ophthalmic topical administration may include ophthalmic solutions, ophthalmic gels, sprays, ointments, perfusion and inserts.
  • a topically delivered formulation of tubulin binding agent should remain stable for a period of time long enough to attain the desired therapeutic effects. In addition the agent must penetrate the surface structures of the eye and accumulate in significant quantities at the site of the disease. Additionally, a topically delivered agent should not cause an excessive amount of local toxicity.
  • Ophthalmic solutions in the form of eye drops generally consist of aqueous media.
  • buffers organic carriers, inorganic carriers, emulsifiers, wetting agents, etc.
  • Pharmaceutically acceptable buffers for ophthalmic topical formulations include phosphate, borate, acetate and glucoronate buffers, amongst others.
  • Drug carriers may include water, water mixture of lower alkanols, vegetable oils, polyalkylene glycols, petroleum based jelly, ethylcellulose, ethyl oleate, carboxymethylcellulose, polyvinylpyrrolidone, and isoproplyl myristrate.
  • Ophthalmic sprays generally produce the same results as eye drops and can be formulated in a similar manner. Some ophthalmic drugs have poor penetrability across ocular barriers and are not administrable as drops or spray. Ointments may thus be used to prolong contact time and increase the amount of drag absorbed. Continuous and constant perfusion of the eye with drug solutions can be achieved by placing polyethylene tubing in the conjunctival sac. The flow rate of the perfusate is adjustable via a minipump system to produce continuous irrigation of the eye. Inserts are similar to soft contact lens positioned on the cornea, except that inserts are generally placed in the upper cul-de-sac or, less frequently, in the lower conjunctival sac rather than attached to the open cornea. Inserts are generally made of biologically soluble materials which dissolve in lacrimal fluid or disintegrate while releasing the drug.
  • the active compounds are coated upon implants or inserts which are implanted into the eye.
  • an implant contemplated by the present invention is an implant from Oculex Pharmaceuticals, Inc., Sunnyvale, CA.
  • the Oculex implant is a biodegradable BDDTM drug delivery device comprised of a biodegradable micro- size polymer system that enables microencapsulated drug therapies to be implanted within the eye. This implant permits the desired drug to be directly released into the area of the eye requiring medication for a predetermined period of time from days, to months to as long as many years.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals. Additional known information with regard to the methods for making the formulations in accordance with the present invention can be found in standard references in the field, such as for example, "Remington's Pharmaceutical Sciences", Mack Publishing Co., Easter, PA, 15 th Ed. (1975).
  • systemic routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transmucosal, and rectal administration.
  • Solutions or suspensions used for parenteral or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders forthe extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound (e.g., a vascular targeting agent) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • the active compound e.g., a vascular targeting agent
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • suppositories e.g., with conventional suppository bases such as cocoa butter and other glycerides
  • retention enemas for rectal delivery.
  • compositions and formulations comprising a tubulin binding agent in association with a pharmaceutically acceptable carrier, diluent, or excipient, such as for example, but not limited to, water, glucose, lactose, hydroxypropyl methylcellulose, as well as other pharmaceutically acceptable carriers, diluents or excipients generally known in the art.
  • a pharmaceutically acceptable carrier such as for example, but not limited to, water, glucose, lactose, hydroxypropyl methylcellulose, as well as other pharmaceutically acceptable carriers, diluents or excipients generally known in the art.
  • pharmacologically effective amount means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by a researcher or clinician.
  • the dosage of CA4P for administration to the eye of a subject is in the range of from approximately 0.01 mg/ml to lOOmg/ml.
  • the concentration of CA4P achieved in the eye should be therapeutically relevant and is in the range of approximately 1 nanomolar to 100 millimolar.
  • the more preferred concentration of CA4P in the eye is in the range of from approximately 1 micromolar to 100 micromolar.
  • an amount of combretastatin A4 prodrug in the range of from approximately 0.1 mg/m 2 to approximately 120 mg/m 2 is advantageously administered parenterally.
  • tubulin binding agents in accordance with the present invention will be formulated for administration to mammals, particularly humans.
  • the invention is not limited in this respect and formulations may be prepared according to veterinary guidelines for administration to animals as well.
  • the invention is further defined by reference to the following examples. It will be apparent to those skilled in the art that many modifications, both to the materials and methods, may be practiced without departing from the purpose and interest of the invention.
  • Example 1 Ocular Irritation Studies and Determination of Mean Tolerable Dosage (MTD) of CA4P when Administered Locally in the Eye Using Three Routes of Administration
  • the test article, CA4P was evaluated for the potential to cause intraocular irritation following intravitreal injection in rabbits. Following general anesthesia, a 0.2 ml dose of CA4P was administered to the right eyes of eight rabbits. A 0.2 ml dose of 0.9% sodium chloride USP solution was administered to the left eyes of the rabbits to serve as a negative control. Four different concentrations of CA4P were tested. Each of the four concentrations (0.1 mg/ml, 1.0 mg/ml, 10 mg/ml and 100 mg/ml) were dosed to the right eyes of two rabbits. Approximately 48 hours after the treatment, the eyes were examined with a biomicroscopic slit-lamp and an indirect ophthalmoscope.
  • the test article, CA4P was evaluated for primary ocular irritation.
  • a single 0.2 ml dose of CA4P dilution (0.1, 1.0, 10 and 100 mg/ml) was placed in the lower conjunctival sac of the left eye to serve as a comparative control.
  • the contralateral eye received buffered saline solution. Ocular reactions were evaluated at 24, 48 and 72 hours after the sample instillation.
  • test article was not considered an irritant as compared to the buffered saline solution control article.
  • a non-systemic method of drug administration must penetrate the relevant structures of the eye and deliver the drug in therapeutically significant quantities at the disease site.
  • radiolabelled drug biodistribution experiments were performed.
  • 14 C-CA4P Three different concentrations of 14 C-CA4P were tested (1, 10, 100 mg/ml) corresponding to doses of 1, 5, and 5 uCi of applied radioactivity respectively.
  • a blank control group was also included. Rabbits were anaesthetized at 1, 6, 24, and 48 hours for blood sampling.
  • Ocular tissue samples were dissected from the cornea, aqueous humor, vitreous humor, choroid, or retina, placed in 20-ml glass scintillation vials, vortexed, and incubated for 24hrs with 500ul digesting fluid. Plasma was separated from whole blood by centrifugation (l,800g for 10 minutes). Both ocular tissue samples and plasma were incubated at room temperature with 16ml of Hionic FluorTM scintillation fluid for a period of 24 hours prior to radioactivity counting.
  • Each sample was counted for 5 minutes in a Betamatic V counter (Bio-Tek Kontron Instruments, St Quentin en Yvelines, France). The conversion of counts per minute ("cpm") into disintegrations per minute (“dpm”) was performed automatically by the beta-counter, using calibration curves obtained from 14 C- standards and quenching curves from the respective blank matrices spiked with 14 C- standards.
  • the concentration of drug was determined according to nanogram equivalents of CA4P (ng-Eq/g of tissue) which was calculated from the measured dpm value, the weight of the tissue specimen, and the specific activity of the drug (0.37 mCi/mg), followed by subtraction of the corresponding background value from control eye tissue.
  • a tissue concentration of luM CA4P is equal to 440nEq/g tissue.
  • Table 1 recites the biodistribution results following intravitreal injection.
  • the degree of ocular penetration was dependent on the concentration of CA4P placed on the surface of the eye.
  • the highest concentrations of drug in the eye (“C max ”) were achieved within the first hour following administration.
  • Therapeutically relevant concentrations of drug (>luM) were delivered to the retina at all concentrations tested. High concentrations of drug were also found in the vitreous and sclera. Relatively little drug was found in the aqueous humor of the eye or the blood plasma.
  • Biodistribution Table 2 recites the biodistribution results following Sub-Tenon's injection. In all tissues examined, the degree of ocular penetration was dependent on the concentration of CA4P placed on the surface of the eye. The highest concentrations of drug in the eye were within the first hour following administration. Therapeutically relevant concentrations of drug (>luM) were delivered to the retina and choroid at the 100 and lOmg/ml administered dose. A high concentration of drug was also observed in the sclera. Relatively little drug was found in the vitreous, aqueous humor, or the blood plasma.
  • Subconjunctival injections were administered at doses of 0.1, 1, 10, and 100 mg/ml.
  • 14 C-CA4P solutions were formulated with 0.24% ION KOH and 0.01% Benzalkonium chloride and injected in a volume of lOOul.
  • Animals were treated at an applied dose of 5uCi with a single subconjunctival injection in right eyes. After delivery, the eye was gently held closed for 2-5 seconds.
  • Table 3 The results of the experiment are recited in Table 3 below.
  • Table 4 recites the biodistribution results following Periocular injection.
  • the degree of ocular penetration was dependent on the concentration of CA4P placed on the surface of the eye.
  • the highest concentrations of drug in the eye were within the first hour following administration.
  • Therapeutically relevant concentrations of drug >luM were delivered to the retina and choroid at all administered doses.
  • a high concentration of drug was also observed in the sclera. Relatively little drug was found in the vitreous or the blood plasma.
  • Topical gels and solutions were developed for use as topical formulation suitable for the topical delivery of CA4P to the surface of the eye.
  • Topical solutions (1, 3, and 10%) were directly prepared in 0.9% NaCl (Aguettant, Lyon, France) and steriUzed with 0.2um filter (pH 6.4 to 8.5, osmolarity 290 to 459 mosmol/kg H20.
  • Low viscosity topical gels (l,3,and 10%) were prepared in 0.5% carboxymethylcellulose (Sigma Aldrich Chirnie, St. Quentin Fallavier Cedex, France) with 0.9% NaCl.
  • the physicochemical specifications of each gel are listed in Table 5
  • Topical formulations were applied to the surface of right eyes at an applied dose of 5uCi in a volume of 50ul. Cornea was sampled instead of sclera. Samples were taken at 0.5, 1, 6, and 24 hours. Table 6 recites the biodistribution results following administration of each topical
  • CA4P gel formulation In all tissues examined, the degree of ocular penetration was dependent on the concentration of CA4P in each gel formulation. The highest concentrations of drug in the eye were within the first hour following administration. Therapeutically relevant concentrations of drug (>luM) were delivered to the cornea, retina, and choroid with all three gel formulations. Relatively little drug was found in in the blood plasma. Table 6: Biodistribution of CA4P following Topical Administration of a Gel
  • Table 7 recites the biodistribution results following administration of each topical CA4P solution formulation.
  • the degree of ocular penetration was dependent on the concentration of CA4P in each solution formulation.
  • the highest concentrations of drag in the eye were within the first hour following administration.
  • Therapeutically relevant concentrations of drug >luM
  • lOmg/ml dose resulted in delivery of a significant amount of drag to the retina and choroid.
  • CA4P is ionizable at physiological pH and therefore is amenable to iontophoretic delivery.
  • the effectiveness of transcleral iontophoretic delivery of CA4P was evaluated using an ocular rabbit ophthalmic applicator (IOMED Inc., Salt Lake City, UT) composed of an 180ul silicone receptacle shell backed with silver chloride-coated silver foil current distribution component, a connector lead wire, and a single layer of hydrogel-impregnated polyvinyl acetal matrix to which CA4P (lOmg/ml) was administered.
  • the contact surface area of the applicator was 0.54cm 2 .
  • Direct current anodal iontophoresis was performed with each applicator at 2,3,and 4 mA for 20 rnin using an Phoresor II TM PM 700 (IOMED Inc., Salt Lake City, UT) power supply.
  • Passive iontophoresis (0mA for 20min) was used as a control.
  • the animals were euthanized, and eyes were enucleated 30 minutes post-treatment, rinsed with tap water, and frozen at -70 C. Retina and choirodal tissue was dissected from these sample.
  • CA4P, CA, and the internal standard Diethylstilbestrol were quantified from approximately lOOmg of tissue using chromatography tandem mass spectrometry ("LC/MS/MS") method.
  • LC/MS/MS chromatography tandem mass spectrometry
  • An aliquot of methonal extraction was injected onto a SCTEX APIO 3000 LC/MS/MS apparatus equipped with an HPLC colum.
  • Peak area of the m z 315-> 285 product ion of CA43 and m/z 395-> 79 product ion of CA4P were measured against the peak area of the m/z 267 -> 237 product ion of the internal standard.
  • Quantitation was performed using weighted (1/x) linear least squares regression analyses generated from fortified calibration standards prepared immediately prior to each run.
  • ocular neovascularization was induced by administration of lipid hydroperoxide (LHP) by intra-corneal injection at a dosage of 30 ⁇ g to rabbit eyes. Seven to 14 days later, ocular vessels formed in the injected eyes due to LHP insult.
  • the subjects were divided into two groups; those of one group were given combretastatin A4 disodium phosphate by intravenous administration at a dosage of 40mg/kg once a day for five days, while a vehicle without combretastatin A4 disodium phosphate was administered to the other group by i.v. admimstration as a dosage of water for the same time period.
  • the eyes of both groups were examined seven days later. A reduction of vessels of 40% or more was observed in the group treated with combretastatin A4 disodium phosphate, but not in the other group.
  • Example 5 Treatment of Corneal Neovascularization via Systemic Administration of CA4P
  • a rabbit corneal model was used in which neovascularization was induced by tinoleic acid hydroperoxide ("LHP") injection (Ueda et al., Angiogenesis, 1997, 1: 174-184).
  • LHP tinoleic acid hydroperoxide
  • Injection of LHP in the corneal stroma stimulates the localized production of angiogenic cytokines within the cornea.
  • Blood vessels in the circumlimbal plexus respond to the angiogenic stimulation by migrating towards the site of LHP injection.
  • Therapeutic efficacy of systemically delivered CA4P was assessed by measuring the length of these proliferating vessels.
  • Table 10 and 11 summarize the effects of CA4P on vessel length as a function of intervention-time and number of treatments.
  • CA4P treatment was used to intervene within 3 days of the initial angiogenic stimulation (Table 10, Group 2), the drag caused a complete inhibition of neovascular growth. In contrast, vessels in the vehicle control group continued to grow. This effect can be qualified as angiogenesis inhibition or an anti- angiogenic effect.
  • CA4P treatment was used to intervene 10 days after the angiogenic stimulation (Table 11, Group 2), the effect was the same.
  • Figure 3B is a surface photograph of a CA4P-treated eye on Day 28. This photograph further illustrates the inhibition of vessel growth on Day 28 following CA4P administration in comparison with the vehicle control eye depicted in Figure 3A.
  • FIGS. 4A and 4B The micrographs presented in Figures 4A and 4B (magnification 400X) are examples of the stained histological specimens obtained from the same animals on day 28.
  • Figure 4A vessels appeared round and numerous.
  • CA4P treated animals Figure 4B
  • vessels appeared narrow and less numerous.
  • evidence of vessel regression was observed at during later stages of intervention with CA4P (data not shown). It appeared that CA4P was able to reduce the width of the established vessels and significantly inhibit the sprouting of branches from thee vessels, which is indicative of an additional vascular targeting effect.
  • Example 6 Treatment of Choroidal Neovascularization in an animal model of Macular Degeneration via Systemic Administration of CA4P
  • Choroidal neovascularization is a major cause of severe vision loss in patients with age-related or wet macular degeneration.
  • a murine model of Choroidal Neovascularization was tested.
  • the investigator used a krypton laser to create a wound on the Bruch's membrane of a C57BL/6J mouse. Each eye received several burns. The burn elicited a classic wound-healing response that included neovascularization within the choroid.
  • This krypton laser photocoagulation method has been described in Tobe et al., Am. J. of Pathology, 1998, 153(5): 1641-6.
  • Example 7 Treatment of Retinal Neovascularization in a Mouse Model of Retinopathy of Prematurity via Systemic Administration of CA4P
  • the inner retina of the mammalian eye receives oxygen from the superficial retinal capillary bed.
  • This capillary bed is located beneath the inner limiting membrane which serves as the interface between the inner retina and the outer avascular vitreous.
  • the pathology of retinal neovascularization or retinopathy arises from ischemia -induced growth of neovasculature beyond the retinal inner limiting membrane and into the vitreous, causing severe loss of vision and frequently leading to retinal detachment.
  • a well-characterized murine model of oxygen-induced retinal neovascularization closely simulates retinopathy of prematurity ("ROP") exhibited by prematurely born human infants, and exhibits characteristics common to a variety of other ischemia-induced retinopathies, including diabetic retinopathy (Smith et al., Invest. Ophthalmol. Vis. Sci., 1994, 35: 101-11).
  • ROP retinopathy of prematurity
  • neonatal mice are exposed to sustained hyperoxic conditions (75% oxygen for 7 days) that inhibit the development of the superficial retinal capillary bed.
  • sustained hyperoxic conditions (75% oxygen for 7 days
  • the retina responds to the lack of oxygen by producing angiogenic cytokines that cause serious pathological consequences.
  • the localized production of angiogenic cytokines can cause the underdeveloped superficial retinal capillary bed to sprout new vessels that breach the inner limiting membrane.
  • the growth of the aberrant blood vessels in the vitreous causes the formation of severe scar tissues and traction-induced retinal detachment.
  • Retinal neovascularization can be quantified by counting the number chemically stained nuclei of penetrating endothelial cells in retinal tissue section of treated and untreated eyes according to existing methods (Majka et al., Invest. Ophthalmol. Vis. Sci. 2001, 42: 210-15). It is expected that the number of nuclei penetrating the inner limiting membrane would be significantly reduced in CA4P eyes.
  • Example 8 Treatment of Ocular Tumors in a Mouse Model of Retinoblastoma via Subconjuctival Administration of CA4P
  • BSS balanced salt solution
EP02756487A 2001-07-13 2002-07-15 COMPOSITIONS AND METHOD FOR THE ADMINISTRATION OF TUBULIN BINDING AGENTS FOR THE TREATMENT OF EYE DISEASES Withdrawn EP1406600A4 (en)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US38622701P 2001-07-13 2001-07-13
US386227P 2001-07-13
US37784702P 2002-05-03 2002-05-03
US37784502P 2002-05-03 2002-05-03
US37755602P 2002-05-03 2002-05-03
US377556P 2002-05-03
US377847P 2002-05-03
US377845P 2002-05-03
PCT/US2002/022449 WO2003006002A1 (en) 2001-07-13 2002-07-15 Compositions and methods of administering tubulin binding agents for the treatment of ocular diseases

Publications (2)

Publication Number Publication Date
EP1406600A1 true EP1406600A1 (en) 2004-04-14
EP1406600A4 EP1406600A4 (en) 2007-06-06

Family

ID=32034390

Family Applications (1)

Application Number Title Priority Date Filing Date
EP02756487A Withdrawn EP1406600A4 (en) 2001-07-13 2002-07-15 COMPOSITIONS AND METHOD FOR THE ADMINISTRATION OF TUBULIN BINDING AGENTS FOR THE TREATMENT OF EYE DISEASES

Country Status (8)

Country Link
EP (1) EP1406600A4 (it)
JP (2) JP2004536847A (it)
KR (1) KR20040030042A (it)
CN (1) CN1527704B (it)
CA (1) CA2453442C (it)
NZ (1) NZ575466A (it)
WO (1) WO2003006002A1 (it)
ZA (1) ZA200400210B (it)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040229960A1 (en) * 2001-07-13 2004-11-18 David Sherris Compositions and methods of administering tubulin binding agents for the treatment of ocular diseases
JP4005048B2 (ja) * 2003-04-09 2007-11-07 日信工業株式会社 炭素繊維複合材料およびその製造方法
JP2012532113A (ja) * 2009-07-02 2012-12-13 オキシジーン, インコーポレイテッド 後嚢混濁を予防するためのコンブレタスタチン
CN102596929A (zh) * 2009-08-27 2012-07-18 生物学特性有限公司 黄斑变性的治疗
NZ598489A (en) 2009-08-27 2014-03-28 Bionomics Ltd Combination therapy for treating proliferative diseases
US9371432B2 (en) * 2012-08-02 2016-06-21 Amril Ag Natural rubber containing nanocarbon
CN113577020B (zh) * 2021-08-16 2022-09-23 海南鑫开源医药科技有限公司 一种玻璃体腔内注射剂、其制备方法及应用
CN113520995B (zh) * 2021-08-16 2023-03-10 海南鑫开源医药科技有限公司 一种离子敏感型眼用原位凝胶、其制备方法及应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000048606A1 (en) * 1999-02-18 2000-08-24 Oxigene, Inc. Compositions and methods for use in targeting vascular destruction
WO2000048591A1 (en) * 1999-02-16 2000-08-24 Angiogene Pharmaceuticals Ltd. Combinations for the treatment of diseases involving angiogenesis
WO2002022626A1 (en) * 2000-09-14 2002-03-21 Bristol-Myers Squibb Company Combretastatin a-4 phosphate prodrug salts with nitrogen-containing compounds

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1338645C (en) * 1987-01-06 1996-10-15 George R. Pettit Isolation, structural elucidation and synthesis of novel antineoplastic substances denominated "combretastatins"
US5504074A (en) * 1993-08-06 1996-04-02 Children's Medical Center Corporation Estrogenic compounds as anti-angiogenic agents
US7018987B1 (en) * 1998-01-09 2006-03-28 Arizona Broad of Regents acting for and on behalf of Arizona State University Synthesis of combretastatin A-4 prodrugs and trans-isomers thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000048591A1 (en) * 1999-02-16 2000-08-24 Angiogene Pharmaceuticals Ltd. Combinations for the treatment of diseases involving angiogenesis
WO2000048606A1 (en) * 1999-02-18 2000-08-24 Oxigene, Inc. Compositions and methods for use in targeting vascular destruction
WO2002022626A1 (en) * 2000-09-14 2002-03-21 Bristol-Myers Squibb Company Combretastatin a-4 phosphate prodrug salts with nitrogen-containing compounds

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GRIGGS J ET AL: "Targeting tumour vasculature: the development of combretastatin A4." THE LANCET ONCOLOGY FEB 2001, vol. 2, no. 2, February 2001 (2001-02), pages 82-87, XP005063056 ISSN: 1470-2045 *
See also references of WO03006002A1 *

Also Published As

Publication number Publication date
KR20040030042A (ko) 2004-04-08
CN1527704A (zh) 2004-09-08
WO2003006002A8 (en) 2004-05-27
EP1406600A4 (en) 2007-06-06
CA2453442C (en) 2011-02-01
CA2453442A1 (en) 2003-01-23
NZ575466A (en) 2011-01-28
WO2003006002A1 (en) 2003-01-23
ZA200400210B (en) 2005-06-29
JP2004536847A (ja) 2004-12-09
WO2003006002A9 (en) 2004-07-22
CN1527704B (zh) 2011-05-18
JP2009132738A (ja) 2009-06-18

Similar Documents

Publication Publication Date Title
US20030181531A1 (en) Compositions and methods of administering tubulin binding agents for the treatment of ocular diseases
Nuyts et al. Histopathologic effects of mitomycin C after trabeculectomy in human glaucomatous eyes with persistent hypotony
JP2009132738A (ja) 眼の疾患の処置のためのチューブリン結合薬剤を投与するための組成物および方法
US20040229960A1 (en) Compositions and methods of administering tubulin binding agents for the treatment of ocular diseases
US20020161013A1 (en) Method of local anesthesia and analgesia
EP1476147A1 (en) Methods for treating eye disorders
US20100249134A1 (en) Use of Beta-Adrenoceptor Antagonists for the Manufacture of a Medicament for the Treatment of Disorders of the Outer Retina
ZA200510344B (en) Coumarin derivatives for the treatment of ophthalmic disorders
US20100190734A1 (en) Method of treating dry eye disease with azithromycin
US20120196937A1 (en) Treatment of ophthalmic disorders using urea
AU2008202468B2 (en) Compositions and Methods of Administering Tubulin Binding Agents for the Treatment of Ocular Diseases
US8679557B2 (en) Synergistic herbal ophthalmic formulation for lowering the intra ocular pressure in case of glaucoma
RU2354398C2 (ru) Композиции и способы введения средств, связывающих тубулин, для лечения глазных заболеваний
AU2002322494A1 (en) Compositions and methods of administering tubulin binding agents for the treatment of ocular diseases
RU2339369C2 (ru) Лечение офтальмологических нарушений с использованием мочевины и ее производных
JP2004331502A (ja) 視神経細胞保護剤
WO2001056606A1 (fr) Remedes pour troubles ophtalmiques
JP4393863B2 (ja) 視神経細胞保護剤
US20100331311A1 (en) Galantamine as a neuroprotective drug for retinal ganglion cells
RU2458657C1 (ru) Способ торможения грубого рубцевания конъюнктивы

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20040112

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LI LU MC NL PT SE SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1064941

Country of ref document: HK

A4 Supplementary search report drawn up and despatched

Effective date: 20070509

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 27/02 20060101ALI20070503BHEP

Ipc: A61K 31/09 20060101AFI20070503BHEP

17Q First examination report despatched

Effective date: 20090806

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20120731

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1064941

Country of ref document: HK