USE OF PROTEIN TYROSINE PHOSPHATASE ZETA AS A BIOMOLECULAR TARGET IN THE TREATMENT AND VISUALIZATION OF BRAIN TUMORS

- AGY THERAPEUTICS

The present invention relates to the use of proteins which are differentially expressed in primary brain tumor tissues, as compared to normal brain tissues, as biomolecular targets for brain tumor treatment therapies. Specifically, the present invention relates to the use of immunotherapeutic and immunoimaging agents which specifically bind to human protein tyrosine phosphatase-zeta (PTP&xgr;) for the treatment and visualization of brain tumors in patients. The present invention also provides compounds and pharmaceutically acceptable compositions for administration in the methods of the invention.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
FIELD OF USE

[0001] The present invention relates to the use of proteins which are differentially expressed in primary brain tumor tissues, as compared to normal brain tissues, as biomolecular targets for brain tumor treatment therapies. Specifically, the present invention relates to the use of immunotherapeutic and immunoimaging agents which specifically bind to human protein tyrosine phosphatase-zeta (PTP&xgr;) for the treatment and visualization of brain tumors in patients. The present invention also provides compounds and pharmaceutically acceptable compositions for administration in the methods of the invention.

BACKGROUND OF THE INVENTION

[0002] Brain Tumor Biology and Etiology

[0003] Brain tumors are considered to have one of the least favorable prognoses for long term survival: the average life expectancy of an individual diagnosed with a central nervous system (CNS) tumor is just eight to twelve months. Several unique characteristics of both the brain and its particular types of neoplastic cells create daunting challenges for the complete treatment and management of brain tumors. Among these are 1) the physical characteristics of the intracranial space, 2) the relative biological isolation of the brain from the rest of the body, 3) the relatively essential and irreplaceable nature of the organ mass, and 4) the unique nature of brain tumor cells.

[0004] First and foremost, the intracranial space and physical layout of the brain create significant obstacles to treatment and recovery. The brain is made of, primarily, astrocytes (which make up the majority of the brain mass, and serve as a scaffold and support for the neurons), neurons (which carry the actual electrical impulses of the nervous system,) and a minor contingent of other cells such as insulating oligodendrocytes (which produce myelin). These cell types give rise to primary brain tumors (e.g., astrocytomas, neuroblastomas, glioblastomas, oligodendrogliomas, etc.) Although the World Health Organization has recently established standard guidelines, the nomenclature for brain tumors is somewhat imprecise, and the terms astrocytoma and glioblastoma are often used broadly. The brain is encased in the relatively rigid shell of the skull, and is cushioned by the cerebrospinal fluid, much like a fetus in the womb. Because of the relatively small volume of the skull cavity, minor changes in the volume of tissue in the brain can dramatically increase intracranial pressure, causing damage to the entire organ (i.e., “water on the brain”). Thus, even small tumors can have a profound and adverse affect on the brain's function. In contrast, tumors in the relatively distensible abdomen may reach several pounds in size before the patient experiences adverse symptoms. The cramped physical location of the cranium also makes surgery and treatment of the brain a difficult and delicate procedure. However, because of the dangers of increased intracranial pressure from the tumor, surgery is often the first strategy of attack in treating brain tumors.

[0005] In addition to its physical isolation, the brain is chemically and biologically isolated from the rest of the body by the so-called “Blood-Brain-Barrier” (or BBB). This physiological phenomenon arises because of the “tightness” of the epithelial cell junctions in the lining of the blood vessels in the brain. Although nutrients, which are actively transported across the cell lining, may reach the brain, other molecules from the bloodstream are excluded. This prevents toxins, viruses, and other potentially dangerous molecules from entering the brain cavity. However, it also prevents therapeutic molecules, including many chemotherapeutic agents that are useful in other types of tumors, from crossing into the brain. Thus, many therapies directed at the brain must be delivered directly into the brain cavity (e.g., by an Ommaya reservoir), or administered in elevated dosages to ensure the diffusion of an effective amount across the BBB.

[0006] With the difficulties of administering chemotherapies to the brain, radiotherapy approaches have also been attempted. However, the amount of radiation necessary to completely destroy potential tumor-producing cells also produce unacceptable losses of healthy brain tissue. The retention of patient cognitive function while eliminating the tumor mass is another challenge to brain tumor treatment. Neoplastic brain cells are often pervasive, and travel throughout the entire brain mass. Thus, it is impossible to define a true “tumor margin,” unlike, for example, in lung or bladder cancers. Unlike reproductive (ovarian, uterine, testicular, prostate, etc.), breast, kidney, or lung cancers, the entire organ, or even significant portions, cannot be removed to prevent the growth of new tumors. In addition, brain tumors are very heterogeneous, with different cell doubling times, treatment resistances, and other biochemical idiosyncrasies between the various cell populations that make up the tumor. This pervasive and variable nature greatly adds to the difficulty of treating brain tumors while preserving the health and function of normal brain tissue.

[0007] Although current surgical methods offer considerably better post-operative life for patients, the current combination therapy methods (surgery, low-dosage radiation, and chemotherapy) have only improved the life expectancy of patients by one month, as compared to the methods of 30 years ago. Without effective agents to prevent the growth of brain tumor cells that are present outside the main tumor mass, the prognosis for these patients cannot be significantly improved. Although some immuno-affinity agents have been proposed and tested for the treatment of brain tumors, see, e.g., the tenascin-targeting agents described in U.S. Pat. No. 5,624,659, these agents have not proven sufficient for the treatment of brain tumors. Thus, therapeutic agents which are directed towards new molecular targets, and are capable of specifically targeting and killing brain tumor cells, are urgently needed for the treatment of brain tumors.

[0008] Protein Tyrosine Phosphatase Receptors: Generally and PTP-Zeta (&xgr;)

[0009] Vital cellular functions, such as cell proliferation and signal transduction, are regulated in part by the balance between the activities of protein kinases and protein phosphatases. These protein-modifying enzymes add or remove a phosphate group from serine, threonine, or tyrosine residues in specific proteins. Some tyrosine kinases (PTK's) and phosphatases (PTPase's) have been theorized to have a role in some types of oncogenesis, which is thought to result from an imbalance in their activities. There are two classes of PTPase molecules: low molecular weight proteins with a single conserved phosphatase domain such as T-cell protein-tyrosine phosphatase (PTPT; MIM 176887), and high molecular weight receptor-linked PTPases with two tandemly repeated and conserved phosphatase domains separated by 56 to 57 amino acids. Examples of this latter group of receptor proteins include: leukocyte-common antigen (PTPRC; MIM 151460) and leukocyte antigen related tyrosine phosphatase (PTPRF; MIM 179590).

[0010] Kaplan et al. cloned 3 human receptor PTP genes, including PTP-&ggr; (“Cloning of three human tyrosine phosphatases reveals a multigene family of receptor-linked protein-tyrosine-phosphatases expressed in brain.” Proc. Nat. Acad. Sci. 87: 7000-7004 (1990).) It was shown that one PTPG allele was lost in 3 of 5 renal carcinoma cell lines and in 5 of 10 lung carcinoma tumor samples tested. PTP-&ggr; mRNA was expressed in kidney cell lines and lung cell lines but not in several hematopoietic cell lines tested. Thus, the PTP-&ggr; gene appeared to have characteristics suggesting that it may be a tumor suppressor gene in renal and lung carcinoma. Barnea et al. (“Identification of a carbonic anhydrase-like domain in the extracellular region of RPTP-gamma defines a new subfamily of receptor tyrosine phosphatases.” Molec. Cell. Biol. 13: 1497-1506 (1993)) cloned cDNAs for the human and mouse PTP-&ggr; gene (designated PTP-&ggr; by that group) from brain cDNA libraries, and analyzed their predicted polypeptide sequences. The human (1,445-amino acid) and mouse (1,442-amino acid) sequences share 95% identity at the amino acid level and predict a putative extracellular domain, a single transmembrane domain, and a cytoplasmic region with 2 tandem catalytic tyrosine phosphatase domains. The extracellular domain contains a stretch of 266 amino acids that are highly similar to the zinc-containing enzyme carbonic anhydrase (MIM 114800), suggesting that PTP-&ggr; and PTP&xgr; represent a subfamily of receptor tyrosine phosphatases. The gene for PTP-&ggr; has 30 exons and is approximately 780 kb in size. It is much larger than the other receptor PTP genes, with the CD45 gene (MIM 151460) being around 100 kb and the others even smaller.

[0011] Another receptor-type tyrosine phosphatase, protein tyrosine phosphatase zeta (PTP&xgr;) [also known as PTPRZ, HPTP-ZETA, HPTPZ, RPTP-BETA(&bgr;), or RPTPB] was isolated as a cDNA sequence by two groups in the early nineties. The complete cDNA sequence of the protein is provided in SEQ ID NO. 1, and the complete deduced amino acid sequence is provided in SEQ ID NO. 2. Splicing variants and features are indicated in the sequences. Levy et al. (“The cloning of a receptor-type protein tyrosine phosphatase expressed in the central nervous system” J. Biol. Chem. 268: 10573-10581, (1993)) isolated cDNA clones from a human infant brain step mRNA expression library, and deduced the complete amino acid sequence of a large receptor-type protein tyrosine phosphatase containing 2,307 amino acids.

[0012] Levy found that the protein, which they designated PTP-&bgr; (PTP&xgr;), is a transmembrane protein with 2 cytoplasmic PTPase domains and a 1,616-amino acid extracellular domain. As in PTP-&ggr; (MIM 176886), the 266 N-terminal residues of the extracellular domain are have a high degree of similarity to carbonic anhydrases (see MIM 114880). The human gene encoding PTP&xgr; has been mapped to chromosome 7q31.3-q32 by chromosomal in situ hybridization (Ariyama et al., “Assignment of the human protein tyrosine phosphatase, receptor-type, zeta (PTPRZ) gene to chromosome band 7q31.3” Cytogenet. Cell Genet. 70: 52-54 (1995)). Northern blot analysis has shown that showed that PTP-zeta is expressed only in the human central nervous system. By in situ hybridization, Levy et al. (1993) localized the expression to different regions of the adult human brain, including the Purkinje cell layer of the cerebellum, the dentate gyrus, and the subependymal layer of the anterior horn of the lateral ventricle. Levy stated that this was the first mammalian tyrosine phosphatase whose expression is restricted to the nervous system. In addition, high levels of expression in the murine embryonic brain were said to suggest an important role in CNS development.

[0013] Gebbink et al. isolated a mouse cDNA of 5.7 kb, encoding a ‘new’ member of the family of receptor-like protein-tyrosine phosphatases, termed RPTP-&mgr; (“Cloning, expression and chromosomal localization of a new putative receptor-like protein tyrosine phosphatase.” FEBS Lett. 290: 123-130 (1991)). The cDNA predicted a protein of 1,432 amino acids (not including the signal peptide) with a calculated molecular mass of 161,636. In addition, they cloned the human homolog, which showed 98.7% amino acid identity to the mouse protein. The predicted mouse protein consisted of a 722-amino acid extracellular region, containing 13 potential N-glycosylation sites, a single transmembrane domain, and a 688-amino acid intracellular part containing two tandem repeats homologous to the catalytic domains of other tyrosine phosphatases. RNA blot analysis showed a single transcript that was most abundant in lung but present in much lower amounts in brain and heart as well. The human PTP-&mgr; gene was assigned to 18 pter-q11 by Southern analysis of human/rodent somatic cell hybrid clones.

[0014] PTP-&egr; cDNA was isolated by Krueger et al. (Structural diversity and evolution of human receptor-like protein tyrosine phosphatases. EMBO J. 9:3241-3252, 1990.1990). The 700-amino acid protein has a short extracellular domain and two tandemly repeated intracellular PTPase domains. High levels of PTP-&egr; transcription were noted in the mouse brain and testes. Both isoforms of PTP-&egr;-a transmembrane, receptor-type isoform and a shorter, cytoplasmic one—appear to arise from a single gene through the use of alternative promoters and 5-prime exons.

[0015] Thus, the PTP receptor family of proteins has been characterized as a fairly diverse family of membrane-bound receptors, and non-membrane bound isoforms, which share a common PTPase cytosol domain architecture. Although their expression in fetal and embryonic tissues has suggested a developmental biology role for the proteins, their full function in normal and disease state biology is still not fully understood.

SUMMARY OF THE INVENTION

[0016] The present invention provides novel methods and reagents for specifically targeting brain tumor neoplastic cells for both therapeutic and imaging purposes. Thus, in a first aspect, the present invention provides PTP&xgr; affinity-based compounds and compositions useful in treating a brain tumor in a patient. The compositions and compounds of this aspect of the invention generally fall into two groups: PTP&xgr;-binding conjugate compounds, which comprise a cytotoxic moiety (C), which inhibits the growth of tumor cells; and PTP&xgr;-binding compound compositions in which the PTP&xgr; binding moiety alters the normal function of PTP&xgr; in the tumor cell, thus inhibiting cell growth.

[0017] In a first group of embodiments of this aspect of the invention, PTP&xgr;-binding therapeutic conjugate compounds are provided. These compounds have the general formula &agr;(Pz)C, wherein &agr;(Pz) is one or more moieties which specifically binds to a human protein tyrosine phosphatase-zeta, and C is one or more cytotoxic moieties. In preferred embodiments &agr;(Pz) is an antibody or an antibody fragment. In particularly preferred embodiments, &agr;(Pz) is an antibody or an antibody fragment which elicits a reduced immune response when administered to a human patient. Preferred cytotoxic moieties for use in these embodiments of the invention include radioactive moieties, chemotoxic moieties, and toxin proteins. The invention also provides compositions comprising these PTP&xgr;-binding therapeutic conjugate compounds in a pharmaceutically acceptable carrier.

[0018] In a second group of embodiments of this first aspect of the invention, PTP&xgr;-binding therapeutic compounds are provided which alter the normal function of PTP&xgr; in brain tumor cells and inhibit brain tumor cell growth. These PTP&xgr;-binding therapeutic compounds have the general formula &agr;(Pz), wherein &agr;(Pz) is one or more moieties which specifically binds to a human protein tyrosine phosphatase-zeta, and wherein the binding of &agr;(Pz) alters the function of protein tyrosine phosphatase-zeta. In preferred embodiments &agr;(Pz) is an antibody or an antibody fragment. In particularly preferred embodiments, &agr;(Pz) is an antibody or an antibody fragment which elicits a reduced immune response when administered to a human patient. It is preferred that the therapeutic compounds of this second group of embodiments of the first aspect of the invention be formulated into therapeutic compositions comprising the PTP&xgr;-binding compound in a pharmaceutically acceptable carrier.

[0019] In a second aspect, the present invention provides methods for using these compounds and compositions to treat a brain tumor in a patient. The methods comprise administering an effective amount of a composition, comprising a PTP&xgr;-binding compound from the first or second group of embodiments of the first aspect and a pharmaceutically acceptable carrier, to a patient in need thereof. Brain tumors treated in this fashion may be glioblastomas, astrocytomas, neuroblastomas, or any type of brain tumor. Administration of the therapeutic composition may be by any acceptable means. One preferred method for administration is by intrathecal administration, although intravascular administration is also preferred.

[0020] In a third aspect, the present invention provides PTP&xgr; affinity-based compounds and compositions for the visualization of brain tumors in patients. These compounds have the general formula &agr;(Pz)I, wherein &agr;(Pz) is one or more moieties which specifically binds to a human protein tyrosine phosphatase-zeta, and I is one or more imaging moieties. In preferred embodiments &agr;(Pz) is an antibody or an antibody fragment. In particularly preferred embodiments, &agr;(Pz) is an antibody or an antibody fragment which elicits a reduced immune response when administered to a human patient. Preferred I moieties include radiographic moieties (useful in, e.g., x-ray, scintillation, or other radiation imaging methods,) positron-emitting moieties, magnetic spin contrast moieties, and optically visible moieties (such as visible particles, fluorescent dyes, and visible-spectrum dyes.) It is preferred that the imaging compounds of these embodiments of the third aspect of the invention be formulated into therapeutic compositions comprising the PTP&xgr;-binding compound in a pharmaceutically acceptable carrier.

[0021] In a fourth aspect, the present invention provides methods of using the compounds and compositions of the third aspect of the invention to visualize a brain tumor in a patient. These methods generally comprise administering an effective amount of an imaging compound of the general formula &agr;(Pz)I in a pharmaceutically acceptable carrier to the patient, and then visualizing the imaging moieties of the compound. Administration of the imaging composition may be by any acceptable means. Intravascular administration of the imaging composition is preferred in these methods, although intrathecal administration is also preferred. Preferred methods of visualizing the imaging moieties of the compounds include radiographic imaging techniques (e.g., x-ray imaging and scintillation imaging techniques), positron-emission tomography, magnetic resonance imaging techniques, and direct or indirect (e.g., endoscopic) visual inspection.

BRIEF DESCRIPTION OF THE DRAWINGS

[0022] FIG. 1: A diagram of the three known splicing variant isoforms of PTP&xgr;. The approximate position of the domains of the isoforms is indicated at the left. Isoform PTP&xgr;-&agr; is the full length isoform, which contains the primary amino acid sequence aa 25-2314 of SEQ ID NO. 2 (aa 1-24 are a signal polypeptide). In Isoform PTP&xgr;-&bgr;, aa 755-1614 are missing. Isoform PTP&xgr;-S (phosphacan), is a secreted isoform which comprise the extracellular domains of PTP&xgr;-&agr;, in which the transmembrane and cytosol domains are missing.

DETAILED DESCRIPTION OF THE INVENTION

[0023] Applicants have identified protein tyrosine phosphatase zeta (PTP&xgr;) as a gene which is differentially regulated between brain cancer tissue (glioblastoma) and normal brain tissue. Applicants have performed differential cloning between cancerous and normal brains and have identified the PTP&xgr; gene by DNA sequence analysis. Based on the observation in other diseases, particularly other cancers, in which overexpressed genes can contribute to the pathology of the disease, these overexpressed genes and their protein products are expected to mediate the initiation and progression of brain tumors. Thus, the overexpressed PTP&xgr; protein, which is presented on the cell surface, provides an excellent target for immunotherapeutic agents which either deliver cytotoxic agents to directly promote tumor cell death, or which alter the function of PTP&xgr; to inhibit the normal physiology of the tumor cell. In addition, immunoimaging agents targeted to PTP&xgr; may be utilized to visualize the tumor mass either in diagnostic methods (e.g., magnetic resonance imaging (MRI) or radiography), or in surgery (e.g., by the use of optically visual dye moieties in the immunoimaging agent).

[0024] Applicants have identified PTP&xgr; by a direct examination of the expression level of genes in actual tumor cells. These samples provide a more accurate and realistic picture of tumor cell biology, especially on the detailed transcriptome level, than animal models or established cell tissue culture cell lines. Several groups have found that cell lines established from astrocytomas and other cell lines do not exhibit expression patterns which reflect the actual expression of the original tumor. For instance, Schreiber, et. al., “Primary brain tumors differ in their expression of octamer deoxyribonucleic acid-binding transcription factors from long-term cultured glioma cell lines.” Neurosurgery 34: 129-35 (1994), showed that nervous system-specific transcription factors known as N-Oct proteins are differentially expressed in human neuroblastoma and glioblastoma cell lines in vitro. However, when these results were compared to freshly isolated human primary and metastatic brain tumors, of the five astrocytomas and three glioblastomas analyzed, all but two tumors displayed the complete N-Oct protein profile, irrespective of histopathological tumor grade. Similarly, Eberle, et al., “The expression of angiogenin in tissue samples of different brain tumors and cultured glioma cells.” Anticancer Res 20: 1679-84 (2000), could show that angiogenin is detectable in different kinds of intracranial tumor tissue samples. Although angiogenin could be detected in primary cultivated glioma cells, it was not detected in the permanent cell lines. Finally, Hartmann, et al., “The rate of homozygous CDKN2A/p16 deletions in glioma cell lines and in primary tumors.” Int J Oncol 15: 975-82 (1999), showed that the rate of homozygous deletions of CDKN2A/p16 is variable between different tumor entities, but the rate of deletions is higher in established cell lines in comparison with primary tumors. Hartmann hypothesized that such incongruencies may reflect statistical sampling errors, true differences depending on tissue derivatization and CDKN2A/p16 loss under selective pressure in tissue culture. After comparing established cell lines derived from human glioblastomas and their corresponding primary tumors by multiplex PCR methodology, they found that in 2 of 11 cases (18%) the primary tumor had no p16 alteration whereas the corresponding cell lines had a homozygous p16 deletion, and that CDKN2A/p16 was lost already in the earliest passages of the cell lines. Thus, Hartmann concluded that the deletion was the result of selective cell-culture pressures in many cases.

[0025] These inconsistent results arise because the tumor tissue samples are obtained from their native milieu, without allowing them the opportunity to alter their gene expression levels in response to artificial environmental stimuli. As recently reported by the Brain Tumor Progress Review group of the National Cancer Institute in November, 2000, conventionally used glioblastoma cell lines contain genetic and gene expression alterations that are well defined and do not necessarily reflect the primary tumors from which they were derived. In addition, these cell lines are highly homogenous, unlike a primary brain tumor. Therefore, data derived soley from a cell line cannot reliably reflect the biology, heterogeneity, or therapeutic response of a primary brain tumor.

[0026] Applicants obtained tumor tissue, snap frozen in the operation hall from unknown patients, which was confirmed as glioblastoma grade IV by neuropathology. These tissues served as the experimental sample. Human whole brain tissue (Clontech Laboratories, Palo Alto, USA) served as control sample. Poly-A+RNA prepared from the cells was converted into double-stranded cDNA (dscDNA).

[0027] Briefly, the ds-cDNA's from control and disease states were subjected to kinetic re-annealing hybridization during which normalization of transcript abundances and enrichment for differentially expressed transcripts (i.e., subtraction) occurs. Normalized-subtracted ds-cDNAs were cloned into a plasmid vector, a large number of recombinant bacterial clones were picked, and their recombinant inserts were isolated by PCR. High-density cDNA arrays of those PCR products were screened with cDNA probes derived from the original control and disease states. Thus, only clones displaying a significant transcriptional induction and/or repression were sequenced and carried forward for massive expression profiling using a variety of temporal, spatial and disease-related probe sets.

[0028] The selected PCR products (fragments of 200-2000 bp in size) from clones showing a significant transcriptional induction and/or repression were sequenced and functionally annotated in AGY's proprietary database structure (See WO01/13105). Because large sequence fragments were utilized in the sequencing step, the data generated has a much higher fidelity and specificity than other approaches, such as SAGE. The resulting sequence information was compared to public databases using the BLAST (blastn) and tblastx algorithm. PTP&xgr; was identified as being expressed in glioblastoma cells at a level approximately 2.0 to 4.0 times the expression in normal brain cells. In the selected group from the subtractive library, 20 clones out of 20,000 were found to align with the PTP&xgr; coding sequence.

[0029] Characteristics and Use of PTP&xgr;

[0030] Thus, PTP&xgr; was selected as a prime target for selective immuno-therapeutic agents in treating or imaging brain tumors. The complete cDNA sequence encoding PTP&xgr; is provided in SEQ ID NO. 1, and the complete amino acid sequence encoding PTP&xgr; is provided in SEQ ID NO. 2. Three different splice variants have been described, which include two membrane bound variants (full length: PTP&xgr;-&agr;, and shorter version PTP&xgr;-&bgr;) and one secreted form (Phosphacan). See FIG. 1. Isoform PTP&xgr;-&agr; is the full length isoform, which contains the primary amino acid sequence aa 25-2314 of SEQ ID NO. 2 (aa 1-24 are a signal polypeptide). In Isoform PTP&xgr;-&bgr;, aa 755-1614 are missing. Isoform PTP&xgr;-S (phosphacan), is a secreated isoform, which is comprises the extracellular domains of PTP&xgr;-&agr;. Northern Blot analysis have shown that the PTP zeta is exclusively expressed in the human central nervous system. In mouse embryos, the PTP&xgr; transcript was mainly detected in the ventricular and subventricular zone of the brain and the spinal cord. The same pattern was detected in adult mice. Detailed studies have shown that during rat embryogenesis the two transmembrane splice variants of PTP&xgr; are mainly expressed in glial precursor cells and that the secretory version (Phosphacan) is more abundant in mature astrocytes which have already migrated in the ventricle zone.

[0031] As used herein, a compound which specifically binds to human protein tyrosine phosphatase-zeta (PTP&xgr;) is any compound (such as an antibody) which has a binding affinity for any naturally occurring isoform, spice variant, or polymorphism of PTP&xgr;, explicitly including the three splice variants describe herein. As one of ordinary skill in the art will appreciate, such “specific” binding compounds (e.g., antibodies) may also bind to other closely related proteins which exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater than 99% identity) with the amino acid sequence of PTP&xgr;. Such proteins include truncated forms or domains of PTP&xgr;, and recombinantly engineered alterations of PTP&xgr;. For example, an portion of SEQ ID NO. 1 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below.

[0032] In general, it is preferred that the antibodies utilized in the compositions and methods of the invention bind to the membrane-bound isoforms of the protein, as this will more specifically target the cytotoxic therapeutic agent, or the imaging agent, to the brain tumor cell. However, embodiments which utilize antibodies which bind to the secreted isoform of the protein are also useful in the invention, as one of ordinary skill would expect that the concentration of the secreted isoform would also be increased adjacent to brain tumor cells which over-express the protein.

[0033] The amino acid sequence of full length PTP&xgr; consists of 2307 amino acids, as the sequence was deduced by Levy (in which aa 1722-1728 of SEQ ID NO. 2 were missing) (See also U.S. Pat. Nos. 5,604,094, and 6,160,090, fully incorporated herein by reference), or 2314 amino acids as the sequence was deduced by Krueger, et al., (“A human transmembrane protein-tyrosine phosphatase, PTP zeta, is expressed in brain and has an N-terminal receptor domain homologous to carbonic anhydrases” Proc. nat. Acad. Sci. U.S.A. 89:7417-7421(1992)). Amino acids 1-24 of SEQ ID NO. 2 are a signal sequence which directs the proper placement of the transmembrane protein. The extracellular domain of the mature PTP&xgr; protein spans amino acids 25-1635 of SEQ ID NO. 2 in the long and secreted forms (this forms the entire secreted form), and amino acids 25-754,1615-1635 in the short isoform. The transmembrane region of the protein spans amino acids 1636-1661 of SEQ ID NO. 2, and the balance of the protein forms the cytoplasmic domain, amino acids 1662-2314.

[0034] When raising antibodies to PTP&xgr;, the entire protein (any of the three isoforms) or a portion thereof may be utilized. For instance, the extracellular domain of the long or short form, the entire secreted form, or a portion of extracellular domain may be utilized. For instance, amino acids 25-754, which are common to both &agr; and &bgr; isoforms, may be used. Such larger PTP&xgr; proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo alto, Calif.) or another suitable genetic source. When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.). In these cases, the PTP&xgr; protein (or a portion thereof) can serve as the PTP&xgr; antigen. When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the PTP&xgr; antigen. Commonly utilized conjugate proteins which are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full PTP&xgr; sequence may be utilized. Preferably, one or more 8-30 aa peptide portions of an extracellular domain of PTP&xgr; are utilized, with peptides in the range of 10-20 being a more economical choice. Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA. Alternatively, peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system. In order to ensure proper protein glycosylation and processing, an animal cell system (e.g., Sf9 or other insect cells, CHO or other mammalian cells) is preferred. Other information useful in designing an antigen for the production of antibodies to PTP&xgr;, including glycosylation sites, is provided in SEQ ID NO. 2

[0035] The extracellular domain of human PTP&zgr; is known to bind to tenascin-C, tenascin-R, pleiotrophin (NM—002825), midkine (NM—002391), FGF-2 (XM—00366), Nr-CAM (NM—005010), L1/Ng-CAM , contactin (NM—001843), N-CAM (XM—006332), and axonin-1NM—005076.) The first 5 of these molecules are either components of the extracellular matrix in gliomas or are soluble factors known to be present in gliomas, and the latter 4 are neuronal surface molecules. The binding of PTP&xgr; to these molecules may play a significant role in the oncogenesis and growth of neoplastic cells in the brain. Thus, in alternative embodiments of the compositions and methods of the invention, antibody moieties are utilized which bind to PTP&xgr; at a site on the protein which alters the binding of an extracellular ligand molecule to PTP&xgr;. Such PTP&xgr; activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).

[0036] Selection of antibodies which alter (enhance or inhibit) the binding of a ligand to PTP&xgr; may be accomplished by a straightforward binding inhibition/enhancement assay. According to standard techniques, the binding of a labeled (e.g., fluorescently or enzyme-labeled) antibody to PTP&xgr;, which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand. The change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand. Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as PTP&xgr; ligand-binding inhibitors or enhancers.

[0037] In addition, antibodies which are useful for altering the function of PTP&xgr; may be assayed in functional formats, such as glioblastoma cell culture or mouse/rat CNS tumor model studies. In glioblastoma cell models of activity, expression of PTP&xgr; is first verified in the particular cell strain to be used. If necessary, the cell line may be stably transfected with a PTP&xgr; coding sequence under the control of an appropriate constituent promoter, in order to express PTP&xgr; at a level comparable to that found in primary tumors. The ability of the glioblastoma cells to survive in the presence of the candidate function-altering anti-PTP&xgr; antibody is then determined. Similarly, In vivo models for human brain tumors, particularly nude mice/SCID mice model or rat models, have been described [Antunes, L., Angioi-Duprez, K. S., Bracard, S. R., Klein-Monhoven, N. A., Le Faou, A. E., Duprez, A. M., and Plenat, F. M. (2000). Analysis of tissue chimerism in nude mouse brain and abdominal xenograft models of human glioblastoma multiforme: what does it tell us about the models and about glioblastoma biology and therapy? J Histochem Cytochem 48, 847-58; Price, A., Shi, Q., Morris, D., Wilcox, M. E., Brasher, P. M., Rewcastle, N. B., Shalinsky, D., Zou, H., Appelt, K., Johnston, R. N., Yong, V. W., Edwards, D., and Forsyth, P. (1999). Marked inhibition of tumor growth in a malignant glioma tumor model by a novel synthetic matrix metalloproteinase inhibitor AG3340. Clin Cancer Res 5, 845-54; and Senner, V., Sturm, A., Hoess, N., Wassmann, H., and Paulus, W. (2000). In vivo glioma model enabling regulated gene expression. Acta Neuropathol (Berl) 99, 603-8.] Once correct expression of PTP&xgr; in the tumor model is verified, the effect of the candidate anti-PTP&xgr; antibodies on the tumor masses in these models can evaluated, wherein the ability of the anti-PTP&xgr; antibody candidates to alter PTP&xgr; activity is indicated by a decrease in tumor growth or a reduction in the tumor mass. Thus, antibodies which exhibit the appropriate anti-tumor effect may be selected without direct knowledge of a binding ligand.

[0038] Antibodies for Use in the Antibody-Therapeutics Methods of the Invention

[0039] Generally, as the term is utilized in the specification, “antibody” or “antibody moiety” is intended to include any polypeptide chain-containing molecular structure that has a specific shape which fits to and recognizes an epitope, where one or more non-covalent binding interactions stabilize the complex between the molecular structure and the epitope. Antibodies which bind specifically to a human protein PTP&xgr; are referred to as anti-PTP&xgr; antibodies, or &agr;(Pz). The specific or selective fit of a given structure and its specific epitope is sometimes referred to as a “lock and key” fit. The archetypal antibody molecule is the immunoglobulin, and all types of immunoglobulins (IgG, IgM, IgA, IgE, IgD, etc.), from all sources (e.g., human, rodent, rabbit, cow, sheep, pig, dog, other mammal, chicken, turkey, emu, other avians, etc.) are considered to be “antibodies.” Antibodies utilized in the present invention may be polyclonal antibodies, although monoclonal antibodies are preferred because they may be reproduced by cell culture or recombinantly, and may be modified to reduce their antigenicity.

[0040] Polyclonal antibodies may be raised by a standard protocol by injecting a production animal with an antigenic composition, formulated as described above. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988. In one such technique, an PTP&xgr; antigen comprising an antigenic portion of the PTP&xgr; polypeptide is initially injected into any of a wide variety of mammals (e.g., mice, rats, rabbits, sheep or goats). Alternatively, in order to generate antibodies to relatively short peptide portions of PTP&xgr; (see discussion above), a superior immune response may be elicited if the polypeptide is joined to a carrier protein, such as ovalbumin, BSA or KLH. The peptide-conjugate is injected into the animal host, preferably according to a predetermined schedule incorporating one or more booster immunizations, and the animals are bled periodically. Polyclonal antibodies specific for the polypeptide may then be purified from such antisera by, for example, affinity chromatography using the polypeptide coupled to a suitable solid support.

[0041] Alternatively, for monoclonal antibodies, hybridomas may be formed by isolating the stimulated immune cells, such as those from the spleen of the inoculated animal. These cells are then fused to immortalized cells, such as myeloma cells or transformed cells, which are capable of replicating indefinitely in cell culture, thereby producing an immortal, immunoglobulin-secreting cell line. The immortal cell line utilized is preferably selected to be deficient in enzymes necessary for the utilization of certain nutrients. Many such cell lines (such as myelomas) are known to those skilled in the art, and include, for example: thymidine kinase (TK) or hypoxanthine-guanine phosphoriboxyl transferase (HGPRT). These deficiencies allow selection for fused cells according to their ability to grow on, for example, hypoxanthine aminopterinthymidine medium (HAT).

[0042] Preferably, the immortal fusion partners utilized are derived from a line that does not secrete immunoglobulin. The resulting fused cells, or hybridomas, are cultured under conditions that allow for the survival of fused, but not unfused, cells and the resulting colonies screened for the production of the desired monoclonal antibodies. Colonies producing such antibodies are cloned, expanded, and grown so as to produce large quantities of antibody, see Kohler and Milstein, 1975 Nature 256:495 (the disclosures of which are hereby incorporated by reference).

[0043] Large quantities of monoclonal antibodies from the secreting hybridomas may then be produced by injecting the clones into the peritoneal cavity of mice and harvesting the ascites fluid therefrom. The mice, preferably primed with pristine, or some other tumor-promoter, and immunosuppressed chemically or by irradiation, may be any of various suitable strains known to those in the art. The ascites fluid is harvested from the mice and the monoclonal antibody purified therefrom, for example, by CM Sepharose column or other chromatographic means. Alternatively, the hybridomas may be cultured in vitro or as suspension cultures. Batch, continuous culture, or other suitable culture processes may be utilized. Monoclonal antibodies are then recovered from the culture medium or supernatant.

[0044] Several monoclonal antibodies against PTP&xgr; are currently available from commercial sources. For instance, BD Transduction Labs supplies a mouse anti-human MAB (WB, IH, IF), denominated “R20720”, which recognizes the two transmembrane isoforms (PTP&xgr;-&agr; and PTP&xgr;-&bgr;). Chemicon supplied a mouse anti-human MAB (WB, IH, IP), denominated “MAB5210”, which recognizes both of the transmembrane isoforms, and also recognizes the soluble isoform (phosphacan, PTP&xgr;-S). These antibodies are suitable for use in the compositions of the present invention, especially in Fab fragment form (which eliminates significant portions of the antigenic mouse constant heavy and light chain regions.) However, it is preferred that such antibodies by humanized or chimerized according to one of the procedures outlined below.

[0045] In addition, the antibodies or antigen binding fragments may be produced by genetic engineering. In this technique, as with the standard hybridoma procedure, antibody-producing cells are sensitized to the desired antigen or immunogen. The messenger RNA isolated from the immune spleen cells or hybridomas is used as a template to make cDNA using PCR amplification. A library of vectors, each containing one heavy chain gene and one light chain gene retaining the initial antigen specificity, is produced by insertion of appropriate sections of the amplified immunoglobulin cDNA into the expression vectors. A combinatorial library is constructed by combining the heavy chain gene library with the light chain gene library. This results in a library of clones which co-express a heavy and light chain (resembling the Fab fragment or antigen binding fragment of an antibody molecule). The vectors that carry these genes are co-transfected into a host (e.g. bacteria, insect cells, mammalian cells, or other suitable protein production host cell.). When antibody gene synthesis is induced in the transfected host, the heavy and light chain proteins self-assemble to produce active antibodies that can be detected by screening with the antigen or immunogen.

[0046] Preferably, recombinant antibodies are produced in a recombinant protein production system which correctly glycosylates and processes the immunoglobulin chains, such as insect or mammalian cells. An advantage to using insect cells which utilize recombinant baculoviruses for the production of antibodies for use in the present invention is that the baculovirus system allows production of mutant antibodies much more rapidly than stably transfected mammalian cell lines. In addition, insect cells have been shown to correctly process and glycosylate eukaryotic proteins, which prokaryotic cells do not. Finally, the baculovirus expression of foreign protein has been shown to constitute as much as 50-75% of the total cellular protein late in viral infection, making this system an excellent means of producing milligram quantities of the recombinant antibodies.

[0047] The use of the baculovirus Autographia californica nuclear polyhedrosis virus (AcNPV) and recombinant viral stocks in Spodoptera frugiperda (Sf9) cells to prepare large quantities of protein has been described by Smith et al. (1985), Summers and Smith (1987). A preferred method of preparing recombinant antibodies is through the expression of DNA encoding recombinant antibody (produced by screening,. as above, or by protein engineering to include more human-like domains, as discussed below) via the baculoviral expression system in Sf9 insect cells. Production of recombinant proteins in Sf9 cells is well known in the art, and one of ordinary skill would be able to select from a number of acceptable protocols (e.g., that described in U.S. Pat. No. 6,603,905).

[0048] It should be noted that antibodies which have a reduced propensity to induce a violent or detrimental immune response in humans (such as anaphylactic shock), and which also exhibit a reduced propensity for priming an immune response which would prevent repeated dosage with the antibody therapeutic or imaging agent (e.g., the human-anti-murine-antibody “HAMA” response), are preferred for use in the invention. These antibodies are preferred for all administrative routes, including intrathecal administration. Even through the brain is relatively isolated in the cranial cavity, behind the blood brain barrier, an immune response still can occur in the form of increased leukocyte infiltration, and inflammation. Although some increased immune response against the tumor is desirable, the concurrent binding and inactivation of the therapeutic or imaging agent generally outweighs this benefit. Thus, humanized, chimeric, or xenogenic human antibodies, which produce less of an immune response when administered to humans, are preferred for use in the present invention.

[0049] Chimeric antibodies may be made by recombinant means by combining the murine variable light and heavy chain regions (VK and VH), obtained from a murine (or other animal-derived) hybridoma clone, with the human constant light and heavy chain regions, in order to produce an antibody with predominantly human domains. The production of such chimeric antibodies is well known in the art, and may be achieved by standard means (as described, e.g., in U.S. Pat. No. 5,624,659, incorporated fully herein by reference.) Humanized antibodies are engineered to contain even more human-like immunoglobulin domains, and incorporate only the complementarity-determining regions of the animal-derived antibody. This is accomplished by carefully examining the sequence of the hyper-variable loops of the variable regions of the monoclonal antibody, and fitting them to the structure of the human antibody chains. Although facially complex, the process is straightforward in practice. See, e.g., U.S. Pat. No. 6,187,287, incorporated fully herein by reference.

[0050] Alternatively, polyclonal or monoclonal antibodies may be produced from animals which have been genetically altered to produce human immunoglobulins, such as the Abgenix XenoMouse or the Medarex HuMAb® technology. The transgenic animal may be produced by initially producing a “knock-out” animal which does not produce the animal's natural antibodies, and stably transforming the animal with a human antibody locus (e.g., by the use of a human artificial chromosome.) Only human antibodies are then made by the animal. Techniques for generating such animals, and deriving antibodies therefrom, are described in U.S. Pat. Nos. 6,162,963 and 6,150,584, incorporated fully herein by reference. Such fully human xenogenic antibodies are a preferred antibody for use in the methods and compositions of the present invention.

[0051] Alternatively, single chain antibodies (Fv, as described below) can be produced from phage libraries containing human variable regions. See U.S. Pat. No. 6,174,708, incorporated fully herein by reference. Also see Kuan, C. T., Reist, C. J., Foulon, C. F., Lorimer, I. A., Archer, G., Pegram, C. N., Pastan, I., Zalutsky, M. R., and Bigner, D. D. (1999). 125I-labeled anti-epidermal growth factor receptor-vIII single-chain Fv exhibits specific and high-level targeting of glioma xenografts. Clin Cancer Res 5, 1539-49;Lorimer, I. A., Keppler-Haflkemeyer, A., Beers, R. A., Pegram, C. N., Bigner, D. D., and Pastan, I. (1996). Recombinant immunotoxins specific for a mutant epidermal growth factor receptor: targeting with a single chain antibody variable domain isolated by phage display. Proc Natl Acad Sci USA 93, 14815-20; Pastan, I. H., Archer, G. E., McLendon, R. E., Friedman, H. S., Fuchs, H. E., Wang, Q. C., Pai, L. H., Herndon, J., and Bigner, D. D. (1995). Intrathecal administration of single-chain immunotoxin, LMB-7 [B3(Fv)-PE38], produces cures of carcinomatous meningitis in a rat model. Proc Natl Acad Sci USA 92, 2765-9, all of which are incorporated by reference fully herein.

[0052] In addition to entire immunoglobulins (or their recombinant counterparts), immunoglobulin fragments comprising the epitope binding site (e.g., Fab′, F(ab′)2, or other fragments) are useful as antibody moieties in the present invention. Such antibody fragments may be generated from whole immunoglobulins by ficin, pepsin, papain, or other protease cleavage. “Fragment,” or minimal immunoglobulins may be designed utilizing recombinant immunoglobulin techniques. For instance “Fv” immunoglobulins for use in the present invention may be produced by linking a variable light chain region to a variable heavy chain region via a peptide linker (e.g., poly-glycine or another sequence which does not form an alpha helix or beta sheet motif).

[0053] Fv fragments are heterodimers of the variable heavy chain domain (VH) and the variable light chain domain (VL). The heterodimers of heavy and light chain domains that occur in whole IgG, for example, are connected by a disulfide bond. Recombinant Fvs in which VH and VL are connected by a peptide linker are typically stable, see, for example, Huston et al., Proc. Natl. Acad, Sci. USA 85:5879-5883 (1988) and Bird et al., Science 242:423-426 (1988), both fully incorporated herein, by reference. These are single chain Fvs which have been found to retain specificity and affinity and have been shown to be useful for imaging tumors and to make recombinant immunotoxins for tumor therapy. However, researchers have bound that some of the single chain Fvs have a reduced affinity for antigen and the peptide linker can interfere with binding. Improved Fv's have been also been made which comprise stabilizing disulfide bonds between the VH and VL regions, as described in U.S. Pat. No. 6,147,203, incorporated fully herein by reference. Any of these minimal antibodies may be utilized in the present invention, and those which are humanized to avoid HAMA reactions are preferred for use in embodiments of the invention.

[0054] In addition, derivatized immunoglobulins with added chemical linkers, detectable moieties [fluorescent dyes, enzymes, substrates, chemiluminescent moieties], or specific binding moieties [such as streptavidin, avidin, or biotin] may be utilized in the methods and compositions of the present invention. For convenience, the term “antibody” or “antibody moiety” will be used throughout to generally refer to molecules which specifically bind to an epitope of PTP&xgr;, although the term will encompass all immunoglobulins, derivatives, fragments, recombinant or engineered immunoglobulins, and modified immunoglobulins, as described above.

[0055] Candidate anti-PTP&xgr; antibodies can be tested for anti-PTP&xgr; activity by any suitable standard means. As a first screen, the antibodies may be tested for binding against the PTP&xgr; antigen utilized to produce them, or against the entire PTP&xgr; extracellular domain or protein. As a second screen, anti-PTP&xgr; candidates may be tested for binding to an appropriate glioblastoma cell line (i.e., one which approximates primary tumor PTP&xgr; expression), or to primary tumor tissue samples. For these screens, the anti-PTP&xgr; candidate antibody may be labeled for detection (e.g., with fluorescein or another fluorescent moiety, or with an enzyme such as horseradish peroxidase). After selective binding to PTP&xgr; is established, the candidate antibody, or an antibody conjugate produced as described below, may be tested for appropriate activity (i.e., the ability to decrease tumor cell growth and/or to aid in visualizing tumor cells) in an in vivo model, such as an appropriate glioblastoma cell line, or in a mouse or rat human brain tumor model, as described above.

[0056] Therapeutic and Imaging Moieties, and Methods for Conjugating them with anti-PTP&xgr; Antibodies to Use in the Compositions and Methods of the Invention

[0057] As described above, the anti-PTP&xgr; antibodies for use in the present invention may have utility on their own without conjugation, if they alter the native activity of PTP&xgr; in the tumor cell. Such antibodies, which may be selected as described above, may be utilized without further modification to include a cytotoxic or imaging moiety. These types of compositions have the advantage of reduced toxicity (in that only the toxicity f the antibody moieties themselves must be taken into account when dosing), and are simpler to manufacture: thus, non-conjugated activity altering anti-PTP&xgr; antibody therapeutics are a preferred embodiment of the invention. However, the conjugation of cytotoxic or imaging agents is yet another preferred embodiment when utilizing these antibodies, as the added moieties also add functionality to the therapeutic.

[0058] Thus, in many preferred embodiments of the invention, the anti-PTP&xgr; antibodies may be coupled or conjugated to one or more therapeutic cytotoxic or imaging moieties. As used herein, “cytotoxic moiety” (C) simply means a moiety which inhibits cell growth or promotes cell death when proximate to or absorbed by the cell. Suitable cytotoxic moieties in this regard include radioactive isotopes (radionuclides), chemotoxic agents such as differentiation inducers and small chemotoxic drugs, toxin proteins, and derivatives thereof. As utilized herein, “imaging moiety” (I) means a moiety which can be utilized to increase contrast between a tumor and the surrounding healthy tissue in a visualization technique (e.g., radiography, positron-emission tomography, magnetic resonance imaging, direct or indirect visual inspection.) Thus, suitable imaging moieties include radiography moieties (e.g. heavy metals and radiation emitting moieties), positron emitting moieties, magnetic resonance contrast moieties, and optically visible moieties (e.g., fluorescent or visible-spectrum dyes, visible particles, etc.). It will be appreciated by one of ordinary skill that some overlap exists between what is a therapeutic moiety and what is an imaging moiety. For instance 212Pb and 212Bi are both useful radioisotopes for therapeutic compositions, but are also electron-dense, and thus provide contrast for X-ray radiographic imaging techniques, and can also be utilized in scintillation imaging techniques.

[0059] In general, therapeutic or imaging agents may be conjugated to the anti-PTP&xgr; moiety by any suitable technique, with appropriate consideration of the need for pharmokinetic stability and reduced overall toxicity to the patient. A therapeutic agent may be coupled to a suitable antibody moiety either directly or indirectly (e.g. via a linker group). A direct reaction between an agent and an antibody is possible when each possesses a functional group capable of reacting with the other. For example, a nucleophilic group, such as an amino or sulfhydryl group, may be capable of reacting with a carbonyl-containing group, such as an anhydride or an acid halide, or with an alkyl group containing a good leaving group (e.g., a halide). Alternatively, a suitable chemical linker group may be used. A linker group can function as a spacer to distance an antibody from an agent in order to avoid interference with binding capabilities. A linker group can also serve to increase the chemical reactivity of a substituent on a moiety or an antibody, and thus increase the coupling efficiency. An increase in chemical reactivity may also facilitate the use of moieties, or functional groups on moieties, which otherwise would not be possible.

[0060] Suitable linkage chemistries include maleimidyl linkers and alkyl halide linkers (which react with a sulfhydryl on the antibody moiety) and succinimidyl linkers (which react with a primary amine on the antibody moiety). Several primary amine and sulflhydryl groups are present on immunoglobulins, and additional groups may be designed into recombinant immunoglobulin molecules. It will be evident to those skilled in the art that a variety of bifunctional or polyfunctional reagents, both homo- and hetero-functional (such as those described in the catalog of the Pierce Chemical Co., Rockford, Ill.), may be employed as a linker group. Coupling may be effected, for example, through amino groups, carboxyl groups, sulfhydryl groups or oxidized carbohydrate residues. There are numerous references describing such methodology, e.g., U.S. Pat. No. 4,671,958. As an alternative coupling method, cytotoxic or imaging moieties may be coupled to the anti-PTP&xgr; antibody moiety through a an oxidized carbohydrate group at a glycosylation site, as described in U.S. Pat. Nos. 5,057,313 and 5,156,840. Yet another alternative method of coupling the antibody moiety to the cytotoxic or imaging moiety is by the use of a non-covalent binding pair, such as streptavidin/biotin, or avidin/biotin. In these embodiments, one member of the pair is covalently coupled to the antibody moiety and the other member of the binding pair is covalently coupled to the cytotoxic or imaging moiety.

[0061] Where a cytotoxic moiety is more potent when free from the antibody portion of the immunoconjugates of the present invention, it may be desirable to use a linker group which is cleavable during or upon internalization into a cell, or which is gradually cleavable over time in the extracellular environment. A number of different cleavable linker groups have been described. The mechanisms for the intracellular release of a cytotoxic moiety agent from these linker groups include cleavage by reduction of a disulfide bond (e.g., U.S. Pat. No. 4,489,710), by irradiation of a photolabile bond (e.g., U.S. Pat. No. 4,625,014), by hydrolysis of derivatized amino acid side chains (e.g., U.S. Pat. No. 4,638,045), by serum complement-mediated hydrolysis (e.g., U.S. Pat. No. 4,671,958), and acid-catalyzed hydrolysis (e.g., U.S. Pat. No. 4,569,789).

[0062] It may be desirable to couple more than one cytotoxic and/or imaging moiety to an antibody. By poly-derivatizing the anti-PTP&xgr; antibody, several cytotoxic strategies may be simultaneously implemented, an antibody may be made useful as a contrasting agent for several visualization techniques, or a therapeutic antibody may be labeled for tracking by a visualization technique. In one embodiment, multiple molecules of an imaging or cytotoxic moiety are coupled to one antibody molecule. In another embodiment, more than one type of moiety may be coupled to one antibody. Regardless of the particular embodiment, immunoconjugates with more than one moiety may be prepared in a variety of ways. For example, more than one moiety may be coupled directly to an antibody molecule, or linkers which provide multiple sites for attachment (e.g., dendrimers) can be used. Alternatively, a carrier with the capacity to hold more than one cytotoxic or imaging moiety can be used.

[0063] A carrier may bear the agents in a variety of ways, including covalent bonding either directly or via a linker group, and non-covalent associations. Suitable covalent-bond carriers include proteins such as albumins (e.g., U.S. Pat. No. 4,507,234), peptides, and polysaccharides such as aminodextran (e.g., U.S. Pat. No. 4,699,784), each of which have multiple sites for the attachment of moieties. A carrier may also bear an agent by non-covalent associations, such as non-covalent bonding or by encapsulation, such as within a liposome vesicle (e.g., U.S. Pat. Nos. 4,429,008 and 4,873,088). Encapsulation carriers are especially useful for imaging moiety conjugation to anti-PTP&xgr; antibody moieties for use in the invention, as a sufficient amount of the imaging moiety (dye, magnetic resonance contrast reagent, etc.) for detection may be more easily associated with the antibody moiety. In addition, encapsulation carriers are also useful in chemotoxic therapeutic embodiments, as they can allow the therapeutic compositions to gradually release a chemotoxic moiety over time while concentrating it in the vicinity of the tumor cells.

[0064] Carriers and linkers specific for radionuclide agents (both for use as cytotoxic moieties or positron-emission imaging moieties) include radiohalogenated small molecules and chelating compounds. For example, U.S. Pat. No. 4,735,792 discloses representative radiohalogenated small molecules and their synthesis. A radionuclide chelate may be formed from chelating compounds that include those containing nitrogen and sulfur atoms as the donor atoms for binding the metal, or metal oxide, radionuclide. For example, U.S. Pat. No. 4,673,562, to Davison et al. discloses representative chelating compounds and their synthesis. Such chelation carriers are also useful for magnetic spin contrast ions for use in magnetic resonance imaging tumor visualization methods, and for the chelation of heavy metal ions for use in radiographic visualization methods.

[0065] Preferred radionuclides for use as cytotoxic moieties are radionulcides which are suitable for pharmacological administration. Such radionuclides include 123I, 125I, 131I, 90Y, 211At, 67Cu, 186Re, 188Re, 212Pb, and 212Bi. Iodine and astatine isotopes are more preferred radionuclides for use in the therapeutic compositions of the present invention, as a large body of literature has been accumulated regarding their use. 131I is particularly preferred, as are other &bgr;-radiation emitting nuclides, which have an effective range of several millimeters. 123I, 125I, 131I, or 211At may be conjugated to antibody moieties for use in the compositions and methods utilizing any of several known conjugation reagents, including Iodogen, N-succinimidyl 3-[211At]astatobenzoate, N-succinimidyl 3-[131I]iodobenzoate (SIB), and, N-succinimidyl 5-[131I]iodob-3-pyridinecarboxylate (SIPC). Any iodine isotope may be utilized in the recited iodo-reagents. For example, a suitable antibody for use in the present invention may be easily made by coupling an Fab fragment of the BD Transduction Labs R20720 anti-PTP&xgr; MAb with 131I Iodogen according to the manufacturer's instructions. Other radionuclides may be conjugated to anti-PTP&xgr; antibody moieties by suitable chelation agents known to those of skill in the nuclear medicine arts.

[0066] Preferred chemotoxic agents include small-molecule drugs such as methotrexate, and pyrimidine and purine analogs. Preferred chemotoxin differentiation inducers include phorbol esters and butyric acid. Chemotoxic moieties may be directly conjugated to the anti-PTP&xgr; antibody moiety via a chemical linker, or may encapsulated in a carrier, which is in turn coupled to the anti-PTP&xgr; antibody moiety.

[0067] Preferred toxin proteins for use as cytotoxic moieties include ricin, abrin, diphtheria toxin, cholera toxin, gelonin, Pseudomonas exotoxin, Shigella toxin, pokeweed antiviral protein, and other toxin proteins known in the medicinal biochemistry arts. As these toxin agents may elicit undesirable immune responses in the patient, especially if injected intravascularly, it is preferred that they be encapsulated in a carrier for coupling to the anti-PTP&xgr; antibody moiety.

[0068] Preferred radiographic moieties for use as imaging moieties in the present invention include compounds and chelates with relatively large atoms, such as gold, iridium, technetium, barium, thallium, iodine, and their isotopes. It is preferred that less toxic radiographic imaging moieties, such as iodine or iodine isotopes, be utilized in the compositions and methods of the invention. Examples of such compositions which may be utilized for x-ray radiography are described in U.S. Pat. No. 5,709,846, incorporated fully herein by reference. Such moieties may be conjugated to the anti-PTP&xgr; antibody moiety through an acceptable chemical linker or chelation carrier. Positron emitting moieties for use in the present invention include 18F, which can be easily conjugated by a fluorination reaction with the anti-PTP&xgr; antibody moiety according to the method described in U.S. Pat. No. 6,187,284.

[0069] Preferred magnetic resonance contrast moieties include chelates of chromium(III), manganese(II), iron(II), nickel(II), copper(II), praseodymium(III), neodymium(III), samarium(III) and ytterbium(III) ion. Because of their very strong magnetic moment, the gadolinium(III), terbium(III), dysprosium(III), holmium(III), erbium(III), and iron(III) ions are especially preferred. Examples of such chelates, suitable for magnetic resonance spin imaging, are described in U.S. Pat. No. 5,733,522, incorporated fully herein by reference. Nuclear spin contrast chelates may be conjugated to the anti-PTP&xgr; antibody moieties through a suitable chemical linker.

[0070] Optically visible moieties for use as imaging moieties include fluorescent dyes, or visible-spectrum dyes, visible particles, and other visible labeling moieties. Fluorescent dyes such as fluorescein, coumarin, rhodamine, bodipy Texas red, and cyanine dyes, are useful when sufficient excitation energy can be provided to the site to be inspected visually. Endoscopic visualization procedures may be more compatible with the use of such labels. For many procedures where imaging agents are useful, such as during an operation to resect a brain tumor, visible spectrum dyes are preferred. Acceptable dyes include FDA-approved food dyes and colors, which are non-toxic, although pharmacuetially acceptable dyes which have been approved for internal administration are preferred. In preferred embodiments, such dyes are encapsulated in carrier moieties, which are in turn conjugated to the anti-PTP&xgr; antibody. Alternatively, visible particles, such as colloidal gold particles or latex particles, may be coupled to the anti-PTP&xgr; antibody moiety via a suitable chemical linker.

[0071] Delivery of Therapeutic and Imaging Agents to the Patient:

[0072] The Blood Brain Barrier (BBB) and Administration Strategies:

[0073] At one time, the BBB was not considered to present a problem in the diagnosis and treatment of brain tumors, because early scans of human brain tumors suggested that the BTB (blood tumor barrier) was “leaky.” This leakiness is relative, however: as the size of the molecule increases, the rate of movement across the barrier decreases. The BBB has been demonstrated to be heterogeneous in experimental human tumor xenograft animal models and in human patients. This lack of uniformity is because of the reduced integrity of tight junctions in the capillary endothelial cells of the tumor neovasculature, intratumoral variation in permeability, and altered intratumoral blood flow (Fuchs et al, 1990, Cancer research 50, 1954-59, Groothuis et al., 1984, Prog.Exp.Tumor Res.) Thus, although the BBB may not pose a delivery problem for some tumors in some patients, this cannot be said for all brain tumors across the board. In addition, a preferred mode of administration of the therapeutics of the invention is after removal of the main tumor mass (resection of the tumor), which destroys much of the “leaky” neovasculature. Moreover, as brain carcinomas are usually pervasive throughout the organ, therapies which are directed towards eradicating all tumor-producing cells cannot rely exclusively on the localized tumor neovasculature.

[0074] A first strategy for drug delivery through the BBB entails disruption of the BBB, either by osmotic means such as mannitol or leukotrienes, or biochemically by the use of vasoactive substances such as bradykinin. The potential for using BBB opening to target specific agents to brain tumors is also an option. In preferred embodiments, a BBB disrupting agent is co-administered with the therapeutic or imaging compositions of the invention when the compositions are administered by intravascular injection. Other strategies to go through the BBB may entail the use of endogenous transport systems, including carrier-mediated transporters such as glucose and amino acid carriers, receptor-mediated transcytosis for insulin or transferrin, and active efflux transporters such as p-glycoprotein. Active transport moieties may also be conjugated to the therapeutic or imaging compounds for use in the invention to facilitate transport across the epithelial wall of the blood vessel. However, the best current strategy for drug delivery behind the BBB is by intrathecal delivery of therapeutics or imaging agents directly to the cranium, as through an Ommaya reservoir.

[0075] Delivery/Administration of Therapeutic Antibodies:

[0076] For administration, the antibody-therapeutic or antibody-imaging agent will generally be mixed, prior to administration, with a non-toxic, pharmaceutically acceptable carrier substance. Usually, this will be an aqueous solution, such as normal saline or phosphate-buffered saline (PBS), Ringer's solution, lactate-Ringer's solution, or any isotonic physiologically acceptable solution for administration by the chosen means. Preferably, the solution is sterile and pyrogen-free, and is manufactured and packaged under current Good Manufacturing Processes (GMP's), as approved by the FDA. The clinician of ordinary skill is familiar with appropriate ranges for pH, tonicity, and additives or preservatives when formulating pharmaceutical compositions for administration by intravascular injection, intrathecal injection, injection into the cerebro-spinal fluid, direct injection into the tumor, or by other routes. In addition to additives for adjusting pH or tonicity, the antibody-therapeutics and antibody-imaging agents may be stabilized against aggregation and polymerization with amino acids and non-ionic detergents, polysorbate, and polyethylene glycol. Optionally, additional stabilizers may include various physiologically-acceptable carbohydrates and salts. Also, polyvinylpyrrolidone may be added in addition to the amino acid. Suitable therapeutic immunoglobulin solutions which are stabilized for storage and administration to humans are described in U.S. Pat. No. 5,945,098, incorporated fully herein by reference. Other agents, such as human serum albumin (HSA), may be added to the therapeutic or imaging composition to stabilize the antibody conjugates.

[0077] The compositions of the invention may be administered using any medically appropriate procedure, e.g., intravascular (intravenous, intraarterial, intracapillary) administration, injection into the cerebrospinal fluid, intracavity or direct injection in the tumor. Intrathecal administration maybe carried out through the use of an Ommaya reservoir, in accordance with known techniques. (F.Balis et al., Am J. Pediatr. Hematol. Oncol. 11, 74, 76 (1989). For the imaging compositions of the invention, administration via intravascular injection is preferred for pre-operative visualization of the tumor. Post-operative visualization or visualization concurrent with an operation may be through intrathecal or intracavity administration, as through an Ommaya reservoir, or also by intravascular administration.

[0078] Intravascular injection may be by intravenous or intraarterial injection: carotid artery injection is thought to assist in administration to the brain, and is thus preferred. Antibody-agents injected into the blood stream have been shown to cross the blood-brain barrier and to infiltrate the cranial cavity to some extent, usually in the range of 10−4 to 10−3% injected dose per gram. This rate of uptake may be sufficient for imaging reagents, and also may be useful for tumor-cell specific cytotoxic agents (e.g, those specifically directed to the inhibition of the function of tumor-cell overexpressed proteins). However, in order to achieve therapeutic concentrations of the antibody-therapeutic agents without unacceptable toxicity to the patient, it is preferred that the therapeutics compositions be administered by intrathecal injection, direct injection, or injection into the cerebro-spinal fluid.

[0079] Thus, a preferred method for administration of the therapeutic compositions of the invention is by depositing it into the inner cavity of a cystic tumor by any suitable technique, such as by direct injection (aided by stereotaxic positioning of an injection syringe, if necessary) or by placing the tip of an Ommaya reservoir into a cavity, or cyst, for administration. Where the tumor is a solid tumor, the antibody may be administered by first creating a resection cavity in the location of the tumor. This procedure differs from an ordinary craniotomy and tumor resection only in a few minor respects. As tumor resection is a common treatment procedure, and is often indicated to relieve pressure, administration of the therapeutic compositions of the invention following tumor resection is a preferred embodiment of the treatment methods of the invention. Following gross total resection in a standard neurosurgical fashion, the cavity is preferable rinsed with saline until all bleeding is stopped by cauterization. Next the pia-arachnoid membrane, surrounding the tumor cavity at the surface, is cauterized to enhance the formation of fibroblastic reaction and scarring in the pia-arachnoid area. The result is the formation of an enclosed, fluid-filled cavity within the brain tissue at the location from where the tumor was removed. After the cyst has been formed, either the tip of an Ommaya reservoir or a micro catheter, which is connected to a pump device and allows the continues infusion of an antibody solution into the cavity, can be placed into the cavity. See, e.g., U.S. Pat. No. 5,558,852, incorporated fully herein by reference.

[0080] Alternatively, a convention-enhanced delivery catheter may be implanted directly into the tumor mass, into a natural or surgically created cyst, or into the normal brain mass. Such convention-enhanced pharmaceutical composition delivery devices greatly improve the diffusion of the composition throughout the brain mass. The implanted catheters of these delivery devices utilize high-flow microinfusion (with flow rates in the range of about 0.5 to 15.0 &mgr;l/minute), rather than diffusive flow, to deliver the therapeutic or imaging composition to the brain and/or tumor mass. Such devices are described in U.S. Pat. No. 5,720,720, incorporated fully herein by reference.

[0081] The effective amount of the therapeutic antibody-conjugate composition or of the imaging antibody-conjugate compositions to be given to a particular patient will depend on a variety of factors, several of which will be different from patient to patient. A competent clinician will be able to determine an effective amount of a therapeutic antibody-conjugate composition to administer to a patient to retard the growth and promote the death of tumor cells, or an effective amount of an imaging composition to administer to a patient to facilitate the visualization of a tumor. Dosage of the antibody-conjugate will depend on the treatment of the tumor, route of administration, the nature of the therapeutics, sensitivity of the tumor to the therapeutics, etc. Utilizing LD50 animal data, and other information available for the conjugated cytotoxic or imaging moiety, a clinician can determine the maximum safe dose for an individual, depending on the route of administration. For instance, an intravenously administered dose may be more than an intrathecally administered dose, given the greater body of fluid into which the therapeutic composition is being administered. Similarly, compositions which are rapidly cleared from the body may be administered at higher doses, or in repeated doses, in order to maintain a therapeutic concentration. As imaging moieties are typically less toxic than cytotoxic moieties, they may be administered in higher doses in some embodiments. Utilizing ordinary skill, the competent clinician will be able to optimize the dosage of a particular therapeutic or imaging composition in the course of routine clinical trials.

[0082] Typically the dosage will be 0.001 to 100 milligrams of conjugate per Kilogram subject body weight. Doses in the range of 0.01 to 1 mg per kilogram of patient body weight may be utilized for a radionuclide therapeutic composition which is administered intrathecally. Relatively large doses, in the range of 0.1 to 10 mg per kilogram of patient body weight, may used for imaging conjugates with a relatively non-toxic imaging moiety. The amount utilized will depend on the sensitivity of the imaging method, and the relative toxicity of the imaging moiety. In a therapeutic example, where the therapeutic composition comprises a 131I cytotoxic moiety, the dosage to the patient will typically start at a lower range of 10 mCi, and go up to 100, 300 or even 500 mCi. Stated otherwise, where the therapeutic agent is 131I, the dosage to the patient will typically be from 5,000 Rads to 100,000 Rads (preferably at least 13,000 Rads, or even at least 50,000 Rads). Doses for other radionuclides are typically selected so that the tumoricidal dose will be equivalent to the foregoing range for 131I. Similarly, chemotoxic or toxin protein doses may be scaled accordingly.

[0083] The antibody conjugate can be administered to the subject in a series of more than one administration. For therapeutic compositions, regular periodic administration (e.g., every 2-3 days) will sometimes be required, or may be desirable to reduce toxicity. For therapeutic compositions which will be utilized in repeated-dose regimens, antibody moieties which do not provoke HAMA or other immune responses are preferred. The imaging antibody conjugate compositions may be administered at an appropriate time before the visualization technique. For example, administration within an hour before direct visual inspection may be appropriate, or administration within twelve hours before an MRI scan may be appropriate. Care should be taken, however, to not allow too much time to pass between administration and visualization, as the imaging compound may eventually be cleared from the patient's system.

[0084] The foregoing is intended to be illustrative of the embodiments of the present invention, and are not intended to limit the invention in any way. Although the invention has been described with respect to specific modifications, the details thereof are not to be construed as limitations, for it will be apparent that various equivalents, changes and modifications may be resorted to without departing from the spirit and scope thereof and it is understood that such equivalent embodiments are to be included herein. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.

Claims

1. A method to treat a brain tumor comprising administering a therapeutic amount of a composition comprising:

a compound of the general formula &agr;(Pz)C, wherein &agr;(Pz) is one or more moieties which specifically binds to a human protein tyrosine phosphatase-zeta, and C is one or more cytotoxic moieties;
and a pharmaceutically acceptable carrier.

2. The method of claim 1 wherein the therapeutic composition is administered by intrathecal administration.

3. The method of claim 1 wherein the therapeutic composition is administered by intravascular administration.

4. The method of claim 1 wherein the brain tumor is a glioblastoma.

5. The method of claim 1 wherein &agr;(Pz) is selected from the group consisting of an antibody and an antibody fragment.

6. The method of claim 5 wherein the antibody is selected from the group consisting of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

7. The method of claim 5 wherein the antibody fragment is selected from the group consisting of fragments of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

8. The method of claim 1 wherein C is a radioactive moiety.

9. The method of claim 8 wherein the radioactive moiety comprises a pharmaceutically acceptable radioactive isotope selected from the group consisting of 123I, 125I, 131I, 90 Y, 211At, 67Cu, 186Re, 188Re, 212Pb, and 212Bi.

10. The method of claim 8 wherein the radioactive moiety comprises a pharmaceutically acceptable radioactive isotope selected from the group consisting of 123I, 125I, 131I, and 211At.

11. The method of claim 1 wherein C is a chemotoxic moiety.

12. The method of claim 11 wherein the chemotoxic moiety is selected from the group consisting of methotrexate, a pyrimidine analog, a purine analog, a phorbol ester, and butyric acid.

13. The method of claim 1 wherein C is a toxin protein moiety.

14. The method of claim 13 wherein the toxin protein moiety is selected from the group consisting of ricin, abrin, diphtheria toxin, cholera toxin, gelonin, Pseudomonas exotoxin, Shigella toxin, and pokeweed antiviral protein.

15. A compound for the treatment of a brain tumor of the general formula &agr;(Pz)C, wherein &agr;(Pz) is one or more moieties which specifically binds to a human protein tyrosine phosphatase-zeta, and C is one or more cytotoxic moieties.

16. The compound of claim 15 wherein &agr;(Pz) is selected from the group consisting of an antibody and an antibody fragment.

17. The compound of claim 16 wherein the antibody is selected from the group consisting of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

18. The compound of claim 16 wherein the antibody fragment is selected from the group consisting of fragments of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

19. The compound of claim 15 wherein C is a radioactive moiety.

20. The compound of claim 15 wherein the radioactive moiety comprises a pharmaceutically acceptable radioactive isotope selected from the group consisting of 123I, 125I, 131I, 90Y, 211At, 67Cu, 186Re, 188Re, 22Pb, and 212Bi.

21. The compound of claim 15 wherein the radioactive moiety comprises a pharmaceutically acceptable radioactive isotope selected from the group consisting of 123I, 125I and 131I.

22. The compound of claim 15 wherein C is a chemotoxic moiety.

23. The compound of claim 22 wherein the chemotoxic moiety is selected from the group consisting of methotrexate, a pyrimidine analog, a purine analog, a phorbol ester, and butyric acid.

24. The compound of claim 15 wherein C is a toxin protein moiety.

25. The compound of claim 24 wherein the toxin protein moiety is selected from the group consisting of ricin, abrin, diphtheria toxin, cholera toxin, gelonin, Pseudomonas exotoxin, Shigella toxin, and pokeweed antiviral protein.

26. A method to treating a brain tumor comprising administering a therapeutic amount of a composition comprising:

a compound of the general formula &agr;(Pz), wherein &agr;(Pz) is one or more moieties which specifically binds to a human protein tyrosine phosphatase-zeta, wherein the binding of &agr;(Pz) alters the function of protein tyrosine phosphatase-zeta,
and a pharmaceutically acceptable carrier.

27. The method of claim 26 wherein the therapeutic composition is administered by intrathecal administration.

28. The method of claim 26 wherein the therapeutic composition is administered by intravascular administration.

29. The method of claim 26 wherein the brain tumor is a glioblastoma.

30. The method of claim 26 wherein &agr;(Pz) is selected from the group consisting of an antibody and an antibody fragment.

31. The method of claim 30 wherein the antibody is selected from the group consisting of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

32. The method of claim 30 wherein the antibody fragment is selected from the group consisting of fragments of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

33. A composition for the treatment of a brain tumor comprising:

a compound of the general formula &agr;(Pz), wherein &agr;(Pz) is one or more moieties which specifically binds to a human protein tyrosine phosphatase-zeta, wherein the binding of &agr;(Pz) alters the function of protein tyrosine phosphatase-zeta,
and a pharmaceutically acceptable carrier.

34. The composition of claim 33 wherein &agr;(Pz) is selected from the group consisting of an antibody and an antibody fragment.

35. The compound of claim 34 wherein the antibody is selected from the group consisting of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

36. The compound of claim 34 wherein the antibody fragment is selected from the group consisting of fragments of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

37. A method for visualizing a brain tumor in a patient, the method comprising:

a) administering to a patient an effective amount of a composition comprising: a compound of the general formula &agr;(Pz)I, wherein &agr;(Pz) is one or more moieties which specifically binds to a human protein tyrosine phosphatase-zeta, and I is one or more imaging moieties;
 and a pharmaceutically acceptable carrier; and
b) visualizing the imaging moieties of the compound.

38. The method of claim 37 wherein the imaging composition is administered by intrathecal administration.

39. The method of claim 37 wherein the imaging composition is administered by intravascular administration.

40. The method of claim 37 wherein the brain tumor is a glioblastoma.

41. The method of claim 37 wherein &agr;(Pz) is selected from the group consisting of an antibody and an antibody fragment.

42. The method of claim 41 wherein the antibody is selected from the group consisting of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

43. The method of claim 41 wherein the antibody fragment is selected from the group consisting of fragments of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

44. The method of claim 37 wherein I is a radiographic moiety.

45. The method of claim 44 wherein the radiographic moiety comprises iodine or an iodine isotope.

46. The method of claim 44 wherein the visualization in step (b) is by x-ray imaging.

47. The method of claim 44 wherein the visualization in step (b) is by scintillation imaging.

48. The method of claim 37 wherein I is a positron-emitting moiety.

49. The method of claim 48 wherein the positron-emitting moiety comprises 18F.

50. The method of claim 48 wherein the visualization in step (b) is by positron emission tomography.

51. The method of claim 37 wherein I is a magnetic spin contrast moiety.

52. The method of claim 51 wherein the magnetic spin contrast moiety comprises an ion selected from the group consisting of chromium(III), manganese(II), iron(II), nickel(II), copper(II), praseodymium(III), neodymium(III), samarium(III) and ytterbium(III).

53. The method of claim 51 wherein the visualization in step (b) is by magnetic resonance imaging.

54. The method of claim 37 wherein I is selected from the group consisting of an optically visible dye and an optically visible particle.

55. The method of claim 54 wherein the visualization method in step (b) is by direct visual inspection.

56. The method of claim 54 wherein the visualization method in step (b) is by visual inspection through an endoscopic instrument.

57. A composition for the visualization of a brain tumor comprising:

a compound of the general formula &agr;(Pz)I, wherein &agr;(Pz) is one or more moieties which specifically binds to a human protein tyrosine phosphatase-zeta, and I is one or more imaging moieties; and
a pharmaceutically acceptable carrier.

58. The composition of claim 57 wherein &agr;(Pz) is selected from the group consisting of an antibody and an antibody fragment.

59. The composition of claim 58 wherein the antibody is selected from the group consisting of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

60. The composition of claim 58 wherein the antibody fragment is selected from the group consisting of fragments of: monoclonal antibodies, polyclonal antibodies, humanized antibodies, recombinant antibodies, chemically modified antibodies, and synthetic antibody analogs.

61. The composition of claim 57 wherein I is a radiographic moiety.

62. The composition of claim 61 wherein the radiographic moiety comprises iodine or an iodine isotope.

63. The composition of claim 57 wherein I is a magnetic-spin contrast moiety.

64. The composition of claim 63 wherein the magnetic spin contrast moiety comprises an ion selected from the group consisting of chromium(III), manganese(II), iron(II), nickel(II), copper(II), praseodymium(III), neodymium(III), samarium(III) and ytterbium(III).

65. The composition of claim 57 wherein I is a positron-emitting moiety.

66. The composition of claim 65 wherein the positron-emitting moiety comprises 18F.

67. The composition of claim 57 wherein I is selected from the group consisting of an optically visible dye and an optically visible particle.

Patent History
Publication number: 20020146370
Type: Application
Filed: Mar 23, 2001
Publication Date: Oct 10, 2002
Applicant: AGY THERAPEUTICS
Inventors: Sabine Mueller (San Francisco, CA), Thorsten Melcher (San Francisco, CA), Daniel J. Chin (Foster City, CA)
Application Number: 09816703