US20060275212A1 - Treatment and diagnosis of cancer - Google Patents

Treatment and diagnosis of cancer Download PDF

Info

Publication number
US20060275212A1
US20060275212A1 US11/481,344 US48134406A US2006275212A1 US 20060275212 A1 US20060275212 A1 US 20060275212A1 US 48134406 A US48134406 A US 48134406A US 2006275212 A1 US2006275212 A1 US 2006275212A1
Authority
US
United States
Prior art keywords
cells
biological agent
cancerous
antibody
prostate
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/481,344
Inventor
Neil Bander
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.)
Cornell Research Foundation Inc
Original Assignee
Cornell Research Foundation 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
Priority claimed from US08/838,682 external-priority patent/US6107090A/en
Application filed by Cornell Research Foundation Inc filed Critical Cornell Research Foundation Inc
Priority to US11/481,344 priority Critical patent/US20060275212A1/en
Publication of US20060275212A1 publication Critical patent/US20060275212A1/en
Assigned to CORNELL RESEARCH FOUNDATION, INC. reassignment CORNELL RESEARCH FOUNDATION, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BANDER, NEIL H.
Priority to US11/939,422 priority patent/US8951737B2/en
Priority to US14/617,788 priority patent/US20150316553A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6869Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from a cell of the reproductive system: ovaria, uterus, testes, prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6891Pre-targeting systems involving an antibody for targeting specific cells
    • A61K47/6899Antibody-Directed Enzyme Prodrug Therapy [ADEPT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57434Specifically defined cancers of prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants

Definitions

  • the present invention relates to the treatment and diagnosis of cancer with biological agents.
  • cancer In spite of improved treatments for certain forms of cancer, it is still a leading cause of death in the United States. Since the chance for complete remission of cancer is, in most cases, greatly enhanced by early diagnosis, it is very desirable that physicians be able to detect cancers before a substantial tumor develops. However, the development of methods that permit rapid and accurate detection of many forms of cancers continues to challenge the medial community.
  • One such illustrative form of cancer is prostate cancer.
  • Prostate cancer is the most common cancer in men with an estimated 317,000 cases in 1996 in the United States. It is the second leading cause of death among men who die from neoplasia with an estimated 40,000 deaths per year. Prompt detection and treatment is needed to limit mortality caused by prostate cancer.
  • prostatic dancer When it metastasizes, prostatic dancer has a distinct predilection for bone and lymph nodes. Saitoh et al., “Metastatic Patterns of Prostatic Cancer. Correlation Between Sites And Number Of Organs Involved,” Cancer, 54:3078-3084 (1984). At the time of clinical diagnosis, as many as 25% of patients have bone metastasis demonstrable by radionuclide scans. Murphy, G. P., et al., “The National Survey Of Prostate Cancer In The United States By The American College Of Surgeons,” J. Urol., 127:928-939 (1982). Accurate clinical evaluation of nodal involvement has proven to be difficult.
  • Imaging techniques such as computed tomography (“CT”) or magnetic resonance (“MR”) imaging are unable to distinguish metastatic prostate cancer involvement of lymph nodes by criterion other than size (i.e., >1 cm). Therefore, by definition, these imaging modalities are inherently insensitive in the detection of small volume ( ⁇ 1 cm) disease as well as non-specific in the detection of larger volume adenopathy.
  • CT computed tomography
  • MR magnetic resonance
  • serum acid phosphatase is elevated in about 65-90 percent of patients having carcinoma of the prostate with bone metastasis; in about 30 percent of patients without roentgenological evidence of bone metastasis; and in about only 5-10 percent of patients lacking clinically demonstrable metastasis.
  • WO 79/00475 to Chu et. al. describes a method for the detection of prostatic acid phosphatase isoenzyme patterns associated with prostatic cancer which obviates many of the above drawbacks.
  • practical problems are posed by the need for a source of cancerous prostate tissue from which the diagnostically relevant prostatic acid phosphatase isoenzyme patterns associated with prostatic cancer are extracted for the preparation of antibodies thereto.
  • the management of prostate cancer can be achieved by watchful waiting, curative treatment, and palliation.
  • radical prostatectomy offers the best chance for eradication of the disease.
  • the drawback of this procedure is that most cancers had spread beyond the bounds of the operation by the time they were detected.
  • prostate-specific antigen testing has permitted early detection of prostate cancer. As a result, surgery is less extensive with fewer complications. Patients with bulky, high-grade tumors are less likely to be successfully treated by radical prostatectomy.
  • prostate-specific antigen concentrations After surgery, if there are detectable serum prostate-specific antigen concentrations, persistent cancer is indicated. In many cases, prostate-specific antigen concentrations can be reduced by radiation treatment. However, this concentration often increases again within two years.
  • Radiation therapy has also been widely used as an alternative to radical prostatectomy.
  • Patients generally treated by radiation therapy are those who are older and less healthy and those with higher-grade, more clinically advanced tumors.
  • Particularly preferred procedures are external-beam therapy which involves three dimensional, conformal radiation therapy where the field of radiation is designed to conform to the volume of tissue treated; interstitial-radiation therapy where seeds of radioactive compounds are implanted using ultrasound guidance; and a combination of external-beam therapy and interstitial-radiation therapy.
  • Hormonal therapy is the main form of treating men with disseminated prostate cancer. Orchiectomy reduces serum testosterone concentrations, while estrogen treatment is similarly beneficial. Diethylstilbestrol from estrogen is another useful hormonal therapy which has a disadvantage of causing cardiovascular toxicity.
  • gonadotropin-releasing hormone agonists are administered testosterone concentrations are ultimately reduced.
  • Flutamide and other nonsteroidal, anti-androgen agents block binding of testosterone to its intracellular receptors. As a result, it blocks the effect of testosterone, increasing serum testosterone concentrations and allows patients to remain potent—a significant problem after radical prostatectomy and radiation treatments.
  • Cytotoxic chemotherapy is largely ineffective in treating prostate cancer. Its toxicity makes such therapy unsuitable for elderly patients.
  • prostate cancer is relatively resistant to cytotoxic agents.
  • mAbs radiolabeled monoclonal antibodies
  • mAbs offer the potential to enhance both the sensitivity and specificity of detecting prostatic cancer within lymph nodes and elsewhere. While many mAbs have previously been prepared against prostate related antigens, none of these mAbs were specifically generated with an imaging objective in mind. Nevertheless, the clinical need has led to evaluation of some of these mAbs as possible imaging agents.
  • the monoclonal antibodies developed for detection and/or treatment of prostate cancer recognize antigens specific to malignant prostatic tissues. Such antibodies are thus used to distinguish malignant prostatic tissue (for treatment or detection) from benign prostatic tissue. See U.S. Pat. No. 4,970,299 to Bazinet et al. and U.S. Pat. No. 4,902,615 to Freeman et al.
  • Horoszewicz “Monoclonal Antibodies to a New Antigenic Marker in Epithelial Prostatic Cells and Serum of Prostatic Cancer Patients,” Anticancer Research, 7:927-936 (1987) (“Horoszewicz”) and U.S. Pat. No. 5,162,504 to Horoszewicz describe an antibody, designated 7E11, which recognizes prostate specific membrane antigen (“PSMA”).
  • PSMA prostate specific membrane antigen
  • PSMA is an integral membrane protein known to have a short intracellular tail and a long extracellular domain.
  • Biochemical characterization and mapping (Troyer et al., “Biochemical Characterization and Mapping of the 7E11-C5.3 Epitope of the Prostate-specific Membrane Antigen,” Urol. Oncol., 1:29-37 (1995)) have shown that the epitope or antigenic site to which the 7E11 antibody binds is present on the intracellular portion of the molecule. Because antibody molecules do not, under normal circumstances, cross the cell membrane unless they bind to the extracellular portion of a molecule and become translocated intracellularly, the 7E11 antibody does not have access to its antigenic target site in an otherwise healthy, viable cell.
  • imaging using 7E11 is limited to the detection of dead cells within tumor deposits. Additionally, the therapeutic use of the 7E11 antibody is limited, because only cells that are already dead or tissue containing a large proportion of dead cells can be effectively targeted.
  • prostate cancer is merely a representative model.
  • diagnosis and treatment of numerous other cancers have similar problems.
  • the present invention is directed to overcoming the deficiencies of prior art antibodies in diagnosing and treating prostate and other types of cancer.
  • One aspect of the present invention relates to a method of ablating or killing cancerous cells.
  • the process involves providing a biological agent which, when contacted with an extracellular domain of prostate specific membrane antigen, recognizes the extracellular domain of prostate specific membrane antigen.
  • These biological agents are contacted with vascular endothelial cells proximate to the cancerous cells under conditions effective to permit both binding of the biological agent to the vascular endothelial cells proximate to the cancerous cells and killing or ablating of the cancerous cells.
  • the biological agent can be used alone or can be bound to a substance effective to kill or ablate the cancerous cells upon binding of the biological agent to vascular endothelial cells that are proximate to the cancerous cells.
  • the biological agent when contacted with an extracellular domain of prostate specific membrane antigen, binds to and is internalized with the prostate specific membrane antigen of such cells.
  • Preferred biological agents for use in the method of ablating or killing cancerous cells in accordance with the present invention are antibodies or binding portions thereof, probes, or ligands.
  • the methods of the present invention are particularly useful in killing or ablating renal, urothelial, colon, rectal, lung, and breast cancerous cells and cancerous cells of metastatic adenocarcinoma to the liver.
  • Another aspect of the present invention relates to a method of detecting cancerous tissue in a biological sample.
  • This method involves providing a biological agent which, when contacted with an extracellular domain of prostate specific membrane antigen, binds to the extracellular domain of prostate specific membrane antigen.
  • the biological agent is bound to a label effective to permit detection of vascular endothelial cells proximate to or within the cancerous tissue upon binding of the biological agent to the vascular endothelial cells proximate to or within the cancerous tissue.
  • the biological sample is contacted with the biological agent having a label under conditions effective to permit binding of the biological agent to the vascular endothelial cells proximate to or within the cancerous tissue in the biological sample.
  • the presence of cancerous tissue in the biological sample is detected by detection of the label.
  • the biological agent is one that, when contacted with an extracellular domain of prostate specific membrane antigen, binds to and is internalized with the prostate specific membrane antigen.
  • Preferred biological agents for use in the method of detecting cancerous tissue in accordance with the present invention are antibodies or binding portions thereof, probes, or ligands. The method is especially useful in detecting renal, urothelial, colon, rectal, lung, and breast cancerous tissue and cancerous tissue of metastatic adenocarcinoma to the liver.
  • Still another aspect of the present invention relates to a method of ablating or killing normal, benign hyperplastic, and cancerous prostate epithelial cells.
  • the process involves providing a biological agent which recognizes an extracellular domain of prostate specific membrane antigen.
  • the biological agent can be used alone or can be bound to a substance effective to kill the cells upon binding of the biological agent to the cells.
  • These biological agents are then contacted with the cells under conditions effective to permit both binding of the biological agent to the extracellular domain of the prostate specific membrane antigen and killing or ablating of the cells.
  • the biological agent binds to and is internalized with the prostate specific membrane antigen of such cells.
  • Preferred biological agents for use in the method of ablating or killing normal, benign hyperplastic, and cancerous prostate epithelial cells in accordance with the present invention are antibodies or binding portions thereof, probes, or ligands.
  • Another aspect of the present invention relates to a method of detecting normal, benign hyperplastic, and cancerous prostate epithelial cells or portions thereof in a biological sample.
  • This method involves providing a biological agent which binds to an extracellular domain of prostate specific membrane antigen.
  • the biological agent is bound to a label effective to permit detection of the cells or portions thereof upon binding of the biological agent to the cells or portions thereof.
  • the biological sample is contacted with the biological agent having a label under conditions effective to permit binding of the biological agent to the extracellular domain of the prostate specific membrane antigen of any of the cells or portions thereof in the biological sample.
  • the presence of any cells or portions thereof in the biological sample is detected by detection of the label.
  • the biological agent binds to and is internalized with the prostate specific membrane antigen of such cells.
  • Preferred biological agents for use in the method of detecting normal, benign hyperplastic, and cancerous prostate epithelial cells in accordance with the present invention are antibodies or binding portions thereof, probes, or ligands.
  • Another aspect of the present invention pertains to a biological agent that recognizes an extracellular domain of prostate specific membrane antigen.
  • the isolated biological agent binds to and is internalized with the prostate specific membrane antigen.
  • Preferred isolated biological agents which recognize an extracellular domain of prostate specific membrane antigen in accordance with the present invention are isolated antibodies or binding portions thereof, probes, or ligands. Hybridoma cell lines that produce monoclonal antibodies of these types are also disclosed.
  • the biological agents of the present invention recognize the extracellular domain of antigens of normal, benign hyperplastic, and cancerous prostate epithelial cells. Unlike the 7E11 antibody, which recognizes an epitope of prostate-associated antigens which are exposed extracellularly only after cell lysis, the biological agents of the present invention bind to antigenic epitopes which are extracellularly exposed in living prostate cells. Using the biological agents of the present invention, living, unfixed normal, benign hyperplastic, and cancerous prostate epithelial cells can be targeted, which makes treatment and diagnosis more effective. In a preferred embodiment for treating prostate cancer, the biological agents of the present invention also bind to and are internalized with the prostate specific membrane antigen, which permits the therapeutic use of intracellularly acting cytotoxic agents.
  • FIG. 1 is an immuno-electron micrograph of gold-labeled monoclonal antibody J591 on the surface of LNCaP cells after incubation at 4° C.
  • FIG. 2 is an immuno-electron micrograph of LNCaP cells treated with gold-labeled monoclonal antibody J591 after 5 minutes incubation at 37° C.
  • FIG. 3 is an immuno-electron micrograph of LNCaP cells treated with gold-labeled monoclonal antibody J591 after 10 minutes incubation at 37° C.
  • FIG. 4 is an immuno-electron micrograph of LNCaP cells treated with gold-labeled monoclonal antibody J591 after 15 minutes incubation at 37° C.
  • FIG. 5 is an immuno-electron micrograph of LNCaP cells treated with gold-labeled monoclonal antibody J591 after 15 minutes at 37° C. showing J591 within endosomes.
  • FIG. 6 summarizes the sequencing strategy of the heavy chain of monoclonal antibody J591.
  • FIG. 7 shows the nucleotide sequence of the heavy chain of monoclonal antibody J591 (designated SEQ. ID. No. 1), the nucleotide sequence of the corresponding reverse, non-coding strand (designated SEQ. ID. No. 2), and the corresponding deduced amino acid sequences (designated SEQ. ID. Nos. 3, 4, and 5).
  • FIG. 8 is a comparison of the heavy chain of monoclonal antibody J591 with the consensus sequence for Mouse Heavy Chains Subgroup IIA.
  • FIG. 9 summarizes the sequencing strategy of the kappa light chain of monoclonal antibody J591.
  • FIG. 10 shows the nucleotide sequences of the kappa light chain of monoclonal antibody J591 (designated SEQ. ID. No. 9), the nucleotide sequence of the corresponding reverse, non-coding strand (designated SEQ. ID. No. 10), and the corresponding deduced amino acid sequence (designated SEQ. ID. Nos. 11, 12, and 13).
  • FIG. 11 is a comparison of the kappa light chain of monoclonal antibody J591 with the consensus sequence for Mouse Kappa Chains Subgroup V.
  • FIGS. 12A-12F are micrographs (250 ⁇ magnification) showing the immunohistochemical reactivity of mAb J591 to neovasculature of various carcinomas.
  • One aspect of the present invention relates to a method of ablating or killing normal, benign hyperplastic, and cancerous prostate epithelial cells.
  • the process involves providing a biological agent, such as an antibody or binding portion thereof, probe, or ligand, which binds to an extracellular domain of prostate specific membrane antigen of (i.e., a portion of prostate specific membrane antigen which is external to) such cells.
  • the biological agent can be used alone or can be bound to a substance effective to kill the cells upon binding of the biological agent to the cells.
  • These biological agents are then contacted with the cells under conditions effective to permit both binding of the biological agent to the extracellular domain of the prostate specific membrane antigen and killing or ablating of the cells.
  • such contacting is carried out in a living mammal by administering the biological agent to the mammal under conditions effective to permit both binding of the biological agent to the extracellular domain of the prostate specific membrane antigen and killing or ablating of the cells.
  • Such administration can be carried out orally or parenterally.
  • the biological agent binds to and is internalized with the prostate specific membrane antigen of such cells.
  • the biological agent can be used alone.
  • the biological agent can be bound to a substance effective to kill the cells upon binding of the biological agent to prostate specific membrane antigen and upon internalization of the biological agent with the prostate specific membrane antigen.
  • the mechanism by which the biological agent is internalized with the prostate specific membrane antigen is not critical to the practice of the present invention.
  • the biological agent can induce internalization of the prostate specific membrane antigen.
  • internalization of the biological agent can be the result of routine internalization of prostate specific membrane antigen.
  • biological agents i.e., biological agents, such as an antibody or binding portion thereof, probe, or ligand which, when contacted with an extracellular domain of prostate specific membrane antigen, recognizes the extracellular domain of prostate specific membrane antigen and, preferably, is internalized therewith
  • the biological agent can be used alone or can be bound to a substance effective to kill the cancerous cells upon binding of the biological agent to vascular endothelial cells proximate thereto. These biological agents are contacted with vascular endothelial cells proximate to the cancerous cells.
  • the contacting is carried out under conditions that are effective to permit binding of the biological agent to the vascular endothelial cells proximate to the cancerous cells and, in addition, that are effective to kill or ablate the cancerous cells.
  • the mechanism by which the cancerous cells are killed or ablated is not critical to the practice of the present invention.
  • the cancerous cells can be killed or ablated directly by the biological agent as a consequence of their proximity to the vascular endothelial cells to which the biological agent binds.
  • the biological agent can kill, ablate, or otherwise change the properties of the vascular endothelial cells to which it binds so that blood flow to the cancerous cells proximate thereto is stopped or otherwise reduced, thereby causing the cancerous cells to be killed or ablated.
  • the method of the present invention is particularly useful for killing or ablating vascular endothelial cells in cancerous tissue as well as the cancerous cells contained in cancerous tissue.
  • the biological agent employed is one that, when contacted with an extracellular domain of prostate specific membrane antigen, binds to and is internalized with the extracellular domain of prostate specific membrane antigen.
  • the methods of the present invention are particularly useful to kill or ablate cancerous prostate epithelial cells as well as cancerous cells other than cancerous prostate epithelial cells. Examples of cancerous cells which are not cancerous prostate epithelial cells are renal, urothelial, colon, rectal, lung, and breast cancerous cells and cancerous cells of metastatic adenocarcinoma to the liver.
  • the method of the present invention can be used to kill or ablate any cell which expresses an extracellular domain of prostate specific membrane antigen or a portion thereof or whose subsistence is dependent upon cells which express an extracellular domain of prostate specific membrane antigen or a portion thereof
  • the method of the present invention is particularly useful to kill or ablate cancerous cells, because the vascular endothelial cells supplying blood to cancerous tissues (e.g., tumors, collections of cancerous cells, or other cancerous masses) express an extracellular domain of prostate specific membrane antigen, irrespective of the type of cancer involved.
  • cancerous tissues e.g., tumors, collections of cancerous cells, or other cancerous masses
  • vascular endothelial cells supplying blood to normal tissues do not express an extracellular domain of prostate specific membrane antigen.
  • Another aspect of the present invention relates to a method of detecting normal, benign hyperplastic, and cancerous epithelial cells or portions thereof in a biological sample.
  • This method involves providing a biological agent, such as an antibody or binding portion thereof, probe, or ligand, which binds to an extracellular domain of prostate specific membrane antigen of such cells.
  • the biological agent is bound to a label effective to permit detection of the cells or portions (e.g., prostate specific membrane antigen or fragments thereof liberated from such normal, benign hyperplastic, and cancerous cells) thereof upon binding of the biological agent to the cells or portions thereof.
  • the biological sample is contacted with the biological agent having a label under conditions effective to permit binding of the biological agent to the extracellular domain of the prostate specific membrane antigen of any of the cells or portions thereof in the biological sample.
  • the presence of any cells or portions thereof in the biological sample is detected by detection of the label.
  • such contacting is carried out in a living mammal and involves administering the biological agent to the mammal under conditions effective to permit binding of the biological agent to the prostate specific membrane antigen of any of the cells or portions thereof in the biological sample. Again, such administration can be carried out orally or parenterally.
  • the method of the present invention can be used to screen patients for diseases associated with the presence of normal, benign hyperplastic, and cancerous epithelial cells or portions thereof. Alternatively, it can be used to identify the recurrence of such diseases, particularly when the disease is localized in a particular biological material of the patient. For example, recurrence of prostatic disease in the prostatic fossa may be encountered following radical prostatectomy. Using the method of the present invention, this recurrence can be detected by administering a short range radiolabeled antibody to the mammal and then detecting the label rectally, such as with a transrectal detector probe.
  • the contacting step can be carried out in a sample of serum or urine or other body fluids, such as to detect the presence of PSMA in the body fluid.
  • the biological agent recognize substantially no antigens circulating in the blood other than PSMA. Since intact prostate cells do not excrete or secrete PSMA into the extracellular environment, detecting PSMA in serum, urine, or other body fluids generally indicates that prostate cells are being lysed.
  • the biological agents and methods of the present invention can be used to determine the effectiveness of a prostate cancer treatment protocol by monitoring the level of PSMA in serum, urine or other body fluids.
  • the biological agent such as the antibody or binding portion thereof, probe, or ligand, binds to and is internalized with the prostate specific membrane antigen of such cells.
  • the biological agent is bound to a label effective to permit detection of the cells or portions thereof upon binding of the biological agent to and internalization of the biological agent with the prostate specific membrane antigen.
  • Another aspect of the present invention relates to a method of detecting cancerous tissue in a biological sample.
  • This method involves providing the above-described biological agent (i.e., a biological agent, such as an antibody or binding portion thereof, probe, or ligand which, when contacted with an extracellular domain of prostate specific membrane antigen, recognizes the extracellular domain of prostate specific membrane antigen).
  • the biological agent is bound to a label that is effective to permit detection of vascular endothelial cells proximate to or within the cancerous tissue upon binding of the biological agent to vascular endothelial cells proximate to or within the cancerous tissue.
  • the biological sample is then contacted with the biological agent having a label. Contacting is carried out under conditions effective to permit binding of the biological agent to the vascular endothelial cells proximate to or within the cancerous tissue in the biological sample.
  • the presence of cancerous cells or portions thereof in the biological sample is detected by detection of the label.
  • a portion of the biological sample can be used.
  • a tissue biopsy sample can be contacted with the biological agent to determine the presence of cancerous tissue in the tissue biopsy sample as well as in the larger biological sample from which it is taken.
  • the biological agent can be contacted with a serum or urine sample to acertain whether any vascular endothelial cells expressing an extracellular domain of prostate specific membrane antigen are present therein.
  • vascular endothelial cells expressing an extracellular domain of prostate specific membrane antigen are found in the vasculature of cancerous tissues but not in the vasculature of normal tissues, detection of the label in a serum or urine sample indicates the presence of cancerous tissue in the larger biological sample from which it is taken (e.g., a patient).
  • the biological agent employed is one that, when contacted with an extracellular domain of prostate specific membrane antigen, binds to and is internalized with the prostate specific membrane antigen.
  • the methods of the present invention can be used to detect cancerous prostate epithelial cells as well as cancerous tissues containing cancerous cells other than cancerous prostate epithelial cells.
  • cancerous tissues containing cancerous cells other than cancerous prostate epithelial cells which can be detected with the methods of the present invention include renal, urothelial, colon, rectal, lung, and breast cancerous tissue and cancerous tissue of metastatic adenocarcinoma to the liver.
  • biological agents suitable for either killing, ablating, or detecting cancerous cells and normal, benign hyperplastic, and cancerous prostate epithelial cells include antibodies, such as monoclonal or polyclonal antibodies.
  • antibody fragments, half-antibodies, hybrid derivatives, probes, and other molecular constructs may be utilized.
  • These biological agents such as antibodies, binding portions thereof, probes, or ligands, bind to extracellular domains of prostate specific membrane antigens or portions thereof in normal, benign hyperplastic, and cancerous prostate epithelial cells.
  • the biological agents when practicing the methods of the present invention to kill, ablate, or detect normal, benign hyperplastic, and cancerous prostate epithelial cells, the biological agents bind to all such cells, not only to cells which are fixed or cells whose intracellular antigenic domains are otherwise exposed to the extracellular environment. Consequently, binding of the biological agents is concentrated in areas where there are prostate epithelial cells, irrespective of whether these cells are fixed or unfixed, viable or necrotic. Additionally or alternatively, these biological agents, such as antibodies, binding portions thereof, probes, or ligands, bind to and are internalized with prostate specific membrane antigens or portions thereof in normal, benign hyperplastic, and cancerous prostate epithelial cells.
  • Monoclonal antibody production may be effected by techniques which are well-known in the art. Basically, the process involves first obtaining immune cells (lymphocytes) from the spleen of a mammal (e.g., mouse) which has been previously immunized with the antigen of interest either in vivo or in vitro. The antibody-secreting lymphocytes are then fused with (mouse) myeloma cells or transformed cells, which are capable of replicating indefinitely in cell culture, thereby producing an immortal, immunoglobulin-secreting cell line. The resulting fused cells, or hybridomas, are cultured, and the resulting colonies screened for the production of the desired monoclonal antibodies.
  • lymphocytes immune cells
  • mammal e.g., mouse
  • myeloma cells or transformed cells which are capable of replicating indefinitely in cell culture, thereby producing an immortal, immunoglobulin-secreting cell line.
  • the resulting fused cells, or hybridomas are cultured, and
  • Colonies producing such antibodies are cloned, and grown either in vivo or in vitro to produce large quantities of antibody.
  • a description of the theoretical basis and practical methodology of fusing such cells is set forth in Kohler and Milstein, Nature 256:495 (1975), which is hereby incorporated by reference.
  • Mammalian lymphocytes are immunized by in vivo immunization of the animal (e.g., a mouse) with the protein or polypeptide of the present invention. Such immunizations are repeated as necessary at intervals of up to several weeks to obtain a sufficient titer of antibodies. Following the last antigen boost, the animals are sacrificed and spleen cells removed.
  • Fusion with mammalian myeloma cells or other fusion partners capable of replicating indefinitely in cell culture is effected by standard and well-known techniques, for example, by using polyethylene glycol (“PEG”) or other fusing agents (See Milstein and Kohler, Eur. J. Immunol. 6:511 (1976), which is hereby incorporated by reference).
  • PEG polyethylene glycol
  • This immortal cell line which is preferably murine, but may also be derived from cells of other mammalian species, including but not limited to rats and humans, is selected to be deficient in enzymes necessary for the utilization of certain nutrients, to be capable of rapid growth, and to have good fusion capability. Many such cell lines are known to those skilled in the art, and others are regularly described.
  • Procedures for raising polyclonal antibodies are also well known. Typically, such antibodies can be raised by administering the protein or polypeptide of the present invention subcutaneously to New Zealand white rabbit's which have first been bled to obtain pre-immune serum.
  • the antigens can be injected at a total volume of 100 ⁇ l per site at six different sites. Each injected material will contain synthetic surfactant adjuvant pluronic polyols, or pulverized acrylamide gel containing the protein or polypeptide after. SDS-polyacrylamide gel electrophoresis.
  • the rabbits are then bled two weeks after the first injection and periodically boosted with the same antigen three times every six weeks. A sample of serum is then collected 10 days after each boost.
  • Polyclonal antibodies are then recovered from the serum by affinity chromatography using the corresponding antigen to capture the antibody. Ultimately, the rabbits are euthenized with pentobarbital 150 mg/Kg IV. This and other procedures for raising polyclonal antibodies are disclosed in E. Harlow, et. al., editors, Antibodies: A Laboratory Manual (1988), which is hereby incorporated by reference.
  • binding portions of such antibodies include Fab fragments, F(ab′) 2 fragments, and Fv fragments.
  • Fab fragments fragments
  • F(ab′) 2 fragments fragments
  • Fv fragments fragments
  • These antibody fragments can be made by conventional procedures, such as proteolytic fragmentation procedures, as described in J. Goding, Monoclonal Antibodies: Principles and Practice , pp. 98-118 (N.Y. Academic Press 1983), which is hereby incorporated by reference.
  • the processes of the present invention can utilize probes or ligands found either in nature or prepared synthetically by recombinant DNA procedures or other biological or molecular procedures.
  • Suitable probes or ligands are molecules which bind to the extracellular domains of prostate specific membrane antigens identified by the monoclonal antibodies of the present invention.
  • Other suitable probes or ligands are molecules which bind to and are internalized with prostate specific membrane antigens.
  • Such probes or ligands can be, for example, proteins, peptides, lectins, or nucleic acid probes.
  • the biological agents can be administered orally, parenterally, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intranasal instillation, by intracavitary or intravesical instillation, intraocularly, intraarterially, intralesionally, or by application to mucous membranes, such as, that of the nose, throat, and bronchial tubes.
  • They may be administered alone or with pharmaceutically or physiologically acceptable carriers, excipients, or stabilizers, and can be in solid or liquid form such as, tablets, capsules, powders, solutions, suspensions, or emulsions.
  • the solid unit dosage forms can be of the conventional type.
  • the solid form can be a capsule, such as an ordinary gelatin type containing the biological agent, such as an antibody or binding portion thereof, of the present invention and a carrier, for example, lubricants and inert fillers such as, lactose, sucrose, or cornstarch.
  • these compounds are tableted with conventional tablet bases such as lactose, sucrose, or cornstarch in combination with binders like acacia, cornstarch, or gelatin, disintegrating agents such as, cornstarch, potato starch, or alginic acid, and a lubricant like stearic acid or magnesium stearate.
  • the biological agent of the present invention may also be administered in injectable dosages by solution or suspension of these materials in a physiologically acceptable diluent with a pharmaceutical carrier.
  • a pharmaceutical carrier include sterile liquids such as water and oils, with or without the addition of a surfactant and other pharmaceutically and physiologically acceptable carrier, including adjuvants, excipients or stabilizers.
  • sterile liquids such as water and oils, with or without the addition of a surfactant and other pharmaceutically and physiologically acceptable carrier, including adjuvants, excipients or stabilizers.
  • Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil.
  • water, saline, aqueous dextrose and related sugar solution, and glycols such as, propylene glycol or polyethylene glycol, are preferred liquid carriers, particularly for injectable solutions.
  • the biological agent of the present invention in solution or suspension may be packaged in a pressurized aerosol container together with suitable propellants, for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants.
  • suitable propellants for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants.
  • the materials of the present invention also may be administered in a non-pressurized form such as in a nebulizer or atomizer.
  • the biological agents may be utilized to detect cancerous tissues (particularly the vascular endothelial cells therein) and normal, benign hyperplastic, and cancerous prostate epithelial cells in vivo. This is achieved by labeling the biological agent, administering the labeled biological agent to a mammal, and then imaging the mammal.
  • radiolabels such as 131 I, 111 In, 123 I, 99 mTc, 32 P, 125 I, 3 H, 14 C, and 188 Rh
  • fluorescent labels such as fluorescein and rhodamine
  • nuclear magnetic resonance active labels such as positron emitting isotopes detectable by a positron emission tomography (“PET”) scanner
  • chemiluminescers such as luciferin
  • enzymatic markers such as peroxidase or phosphatase.
  • Short-range radiation emitters such as isotopes detectable by short-range detector probes, such as a transrectal probe, can also be employed.
  • isotopes and transrectal detector probes when used in combination, are especially useful in detecting prostatic fossa recurrences and pelvic nodal disease.
  • the biological agent can be labeled with such reagents using techniques known in the art. For example, see Wensel and Meares, Radioimmunoimaging and Radioimmunotherapy , Elsevier, N.Y. (1983), which is hereby incorporated by reference, for techniques relating to the radiolabeling of antibodies. See also, D. Colcher et al., “Use of Monoclonal Antibodies as Radiopharmaceuticals for the Localization of Human Carcinoma Xenografts in Athymic Mice”, Meth. Enzymol. 121: 802-816 (1986), which is hereby incorporated by reference.
  • a radiolabeled biological agent of this invention can be used for in vitro diagnostic tests.
  • the specific activity of a tagged biological agent depends upon the half-life, the isotopic purity of the radioactive label, and how the label is incorporated into the biological agent.
  • Table 2 lists several commonly-used isotopes, their specific activities and half-lives. In immunoassay tests, the higher the specific activity, in general, the better the sensitivity.
  • Tritium labeling procedures are described in U.S. Pat. No. 4,302,438, which is hereby incorporated by reference.
  • Iodinating, tritium labeling, and 35 S labeling procedures especially adapted for murine monoclonal antibodies are described by Goding, J. W. (supra, pp 124-126) and the references cited therein, which are hereby incorporated by reference.
  • Other procedures for iodinating biological agents, such as antibodies, binding portions thereof, probes, or ligands are described by Hunter and Greenwood, Nature 144:945 (1962), David et al., Biochemistry 13:1014-1021 (1974) and U.S.
  • Radiolabeling elements which are useful in imaging include 123 I, 131 I, 111 In, and 99m Tc, for example.
  • Procedures for iodinating biological agents are described by Greenwood, F. et al., Biochem. J. 89:114-123 (1963); Marchalonis, J., Biochem. J. 113:299-305 (1969); and Morrison, M. et al., Immunochemistry, 289-297 (1971), which are hereby incorporated by reference.
  • Procedures for 99m Tc-labeling are described by Rhodes, B. et al. in Burchiel, S. et al.
  • the biological agent is administered to the patient, is localized to the tumor bearing the antigen with which the biological agent reacts, and is detected or “imaged” in vivo using known techniques such as radionuclear scanning using e.g., a gamma camera or emission tomography. See e.g., A. R. Bradwell et al., “Developments in Antibody Imaging”, Monoclonal Antibodies for Cancer Detection and Therapy , R. W. Baldwin et al., (eds.), pp. 65-85 (Academic Press 1985), which is hereby incorporated by reference.
  • a positron emission transaxial tomography scanner such as designated Pet VI located at Brookhaven National Laboratory, can be used where the radiolabel emits positrons (e.g., 11 C, 18 F, 15 O, and 13 N).
  • Fluorophore and chromophore labeled biological agents can be prepared from standard moieties known in the art. Since antibodies and other proteins absorb light having wavelengths up to about 310 nm, the fluorescent moieties should be selected to have substantial absorption at wavelengths above 310 nm and preferably above 400 nm. A variety of suitable fluorescers and chromophores are described by Stryer, Science, 162:526 (1968) and Brand, L. et al., Annual Review of Biochemistry, 41:843-868 (1972), which are hereby incorporated by reference. The biological agents can be labeled with fluorescent chromophore groups by conventional procedures such as those disclosed in U.S. Pat. Nos. 3,940,475, 4,289,747, and 4,376,110, which are hereby incorporated by reference.
  • fluorescers having a number of the desirable properties described above are the xanthene dyes, which include the fluoresceins derived from 3,6-dihydroxy-9-henylxanthhydrol and resamines and rhodamines derived from 3,6-diamino-9-phenylxanthydrol and lissanime rhodamine B.
  • the rhodamine and fluorescein derivatives of 9-o-carboxyphenylxanthhydrol have a 9-o-carboxyphenyl group.
  • Fluorescein compounds having reactive coupling groups such as amino and isothiocyanate groups such as fluorescein isothiocyanate and fluorescamine are readily available.
  • Another group of fluorescent compounds are the naphthylamines, having an amino group in the ⁇ or ⁇ position.
  • Biological agents can be labeled with fluorchromes or chromophores by the procedures described by Goding, J. (supra, pp 208-249).
  • the biological agents can be labeled with an indicating group containing the NMR-active 19 F atom, or a plurality of such atoms inasmuch as (i) substantially all of naturally abundant fluorine atoms are the 19 F isotope and, thus, substantially all fluorine-containing compounds are NMR-active; (ii) many chemically active polyfluorinated compounds such as trifluoracetic anhydride are commercially available at relatively low cost, and (iii) many fluorinated compounds have been found medically acceptable for use in humans such as the perfluorinated polyethers utilized to carry oxygen as hemoglobin replacements.
  • a whole body NMR determination is carried out using an apparatus such as one of those described by Pykett, Scientific American, 246:78-88 (1982), which is hereby incorporated by reference, to locate and image cancerous tissues (particularly the vascular endothelial cells therein) and prostate epithelial cells.
  • the antibodies of the present invention (or other biological agents of the present invention), labeled as described above, can be coadministered along with an antibody or other biological agent which recognizes only living or only dead prostate epithelial cells labeled with a label which can be distinguished from the label used to label the subject antibody.
  • an antibody or other biological agent which recognizes only living or only dead prostate epithelial cells labeled with a label which can be distinguished from the label used to label the subject antibody.
  • concentration of the two labels at various locations or times, spatial and temporal concentration variations of living and dead normal, benign hyperplastic, and cancerous prostate epithelial cells can be ascertained.
  • this method can be carried out using the labeled antibodies of the present invention, which recognize both living and dead epithelial prostate cells, and labeled 7E11 antibodies, which recognize only dead epithelial prostate cells.
  • the biological agents can also be utilized to kill or ablate cancerous cells and normal, benign hyperplastic, and cancerous prostate epithelial cells in vivo. This involves using the biological agents by themselves or with a cytotoxic drug to which the biological agents of the present invention (i.e., biological agents recognizing normal, benign hyperplastic, and cancerous prostate epithelial cells) are bound. This involves administering the biological agents bonded to a cytotoxic drug to a mammal requiring such treatment. In the case of normal, benign-hyperplastic, and cancerous prostate epithelial cells, since the biological agents recognize prostate epithelial cells, any such cells to which the biological agents bind are destroyed.
  • cancerous tissues since the biological agents recognize vascular endothelial cells that are proximate to cancerous cells, binding of the biological agent/cytotoxic drug complex to these vascular endothelial cells destroys them, thereby cutting off the blood flow to the proximate cancerous cells and, thus, killing or ablating these cancerous cells.
  • the biological agents by virtue of their binding to vascular endothelial cells that are proximate to cancerous cells, are localized proximate to the cancerous cells.
  • suitable biological agents including those containing substances effective to kill cells nondiscriminatingly but only over a short range
  • the biological agents of the present invention may be used to deliver a variety of cytotoxic drugs including therapeutic drugs, a compound emitting radiation, molecules of plants, fungal, or bacterial origin, biological proteins, and mixtures thereof.
  • the cytotoxic drugs can be intracellularly acting cytotoxic drugs, such as short-range radiation emitters, including, for example, short-range, high-energy ⁇ -emitters.
  • Enzymatically active toxins and fragments thereof are exemplified by diphtheria toxin A fragment, nonbinding active fragments of diphtheria toxin, exotoxin A (from Pseudomonas aeruginosa ), ricin A chain, abrin A chain, modeccin A chain, ⁇ -sacrin, certain Aleurites fordii proteins, certain Dianthin proteins, Phytolacca americana proteins (PAP, PAPII and PAP-S), Morodica charantia inhibitor, curcin, crotin, Saponaria officinalis inhibitor, gelonin, mitogillin, restrictocin, phenomycin, and enomycin, for example.
  • cytotoxic moieties are derived from adriamycin, chlorambucil, daunomycin, methotrexate, neocarzinostatin, and platinum, for example.
  • Procedures for conjugating the biological agents with the cytotoxic agents have been previously described. Procedures for conjugating chlorambucil with antibodies are described by Flechner, I, European Journal of Cancer, 9:741-745 (1973); Ghose, T. et al., British Medical Journal, 3:495-499 (1972); and Szekerke, M., et al., Neoplasma, 19:211-215 (1972), which are hereby incorporated by reference. Procedures for conjugating daunomycin and adriamycin to antibodies are described by Hurwitz, E. et al., Cancer Research, 35:1175-1181 (1975) and Arnon, R. et al.
  • a first biological agent is conjugated with a prodrug which is activated only when in close proximity with a prodrug activator.
  • the prodrug activator is conjugated with a second biological agent according to the present invention, preferably one which binds to a non-competing site on the prostate specific membrane antigen molecule.
  • a second biological agent preferably one which binds to a non-competing site on the prostate specific membrane antigen molecule.
  • Monoclonal antibody J415 binds to a binding site which is non-competing with the site to which J591, J533, and E99 bind.
  • the first biological agent can be one of J591, J533, and E99
  • the second biological agent can be J415.
  • the first biological agent can be J415
  • the second biological agent can be one of J591, J533, and E99.
  • Drug-prodrug pairs suitable for use in the practice of the present invention are described in Blakely et al., “ZD2767, an Improved System for Antibody-directed Enzyme Prodrug Therapy That Results in Tumor Regressions in Colorectal Tumor Xenografts,” Cancer Research, 56:3287-3292 (1996), which is hereby incorporated by reference.
  • the biological agent can be coupled to high energy radiation emitters, for example, a radioisotope, such as 131 I, a ⁇ -emitter, which, when localized at the tumor site, results in a killing of several cell diameters.
  • a radioisotope such as 131 I
  • a ⁇ -emitter which, when localized at the tumor site, results in a killing of several cell diameters.
  • radioisotopes include ⁇ -emitters, such as 212 Bi, 213 Bi, and 211 At, and ⁇ -emitters, such as 186 Re and 90 Y. Radiotherapy is expected to be particularly effective, because prostate epithelial cells and vascular endothelial cells within cancers are relatively radiosensitive.
  • such killing or ablation can be effected by initiating endogenous host immune functions, such as complement-mediated or antibody-dependent cellular cytotoxicity.
  • the biological agent of the present invention can be used and sold together with equipment, as a kit, to detect the particular label.
  • Biological agents of the present invention can be used in conjunction with other therapeutic treatment modalities.
  • Such other treatments include surgery, radiation, cryosurgery, thermotherapy, hormone treatment, chemotherapy, vaccines, and other immunotherapies.
  • Also encompassed by the present invention is a method of killing or ablating which involves using the biological agents for prophylaxis.
  • these materials can be used to prevent or delay development or progression of prostate or other cancers.
  • the biological agents according to the present invention only target cancerous cells (such as cells of cancerous tissues containing vascular endothelial cells) and prostate epithelial cells, other tissue is spared. As a result, treatment with such biological agents is safer, particularly for elderly patients.
  • Treatment according to the present invention is expected to be particularly effective, because it directs high levels of biological agents, such as antibodies or binding portions thereof, probes, or ligands, to the bone marrow and lymph nodes where prostate cancer metastases and metastases of many other cancers predominate.
  • the methods of the present invention are particularly well-suited for treating prostate cancer, because tumor sites for prostate cancer tend to be small in size and, therefore, easily destroyed by cytotoxic agents.
  • Treatment in accordance with the present invention can be effectively monitored with clinical parameters, such as, in the case of prostate cancer, serum prostate specific antigen and/or pathological features of a patient's cancer, including stage, Gleason score, extracapsular, seminal, vesicle or perineural invasion, positive margins, involved lymph nodes, etc.
  • these parameters can be used to indicate when such treatment should be employed.
  • the biological agents of the present invention bind to living prostate cells, therapeutic methods for treating prostate cancer using these biological agents are much more effective than those which target lysed prostate cells.
  • diagnostic and imaging methods which determine the location of living normal, benign hyperplastic, or cancerous prostate epithelial cells (as well as vascular endothelial cells within cancers) are much improved by employing the biological agents of the present invention.
  • the ability to differentiate between living and dead prostate cells can be advantageous, especially to monitor the effectiveness of a particular treatment regimen.
  • Hybridomas E99, J415, J533, and J591 have been deposited pursuant to, and in satisfaction of, the requirements of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure with the American Type Culture Collection (“A.T.C.C.”) at 12301 Parklawn Drive, Rockville, Md. 20852.
  • Hybridoma E99 was deposited on May 2, 1996, and received A.T.C.C. Designation Number HB-12101.
  • Hybridoma J415 was deposited on May 30, 1996, and received A.T.C.C. Designation Number HB-12109.
  • Hybridomas J533 and J591 were deposited on Jun. 6, 1996, and received A.T.C.C. Designation Numbers HB-12127 and HB-12126, respectively.
  • LNCaP Horoszewicz, J. S., et al., “LNCaP Model Of Human Prostatic Carcinoma,” Cancer Res., 43:1809-1818 (1983), which is hereby incorporated by reference
  • Hybridomas were initially cloned in RPMI-1640 medium supplemented with 10% FCS, 0.1 mM nonessential amino acids, 2 mM L-glutamine, 100 units/ml of penicillin, 100 ug/ml of streptomycin and HAT medium (GIBCO, Grand Island, N.Y.). Subclones were cultured in the same medium without aminopterin.
  • mice Female BALB/c mice were immunized intraperitoneally with LNCaP (6 ⁇ 10 6 cells) three times at 2 week intervals. A final intraperitoneal booster immunization was administered with fresh prostate epithelial cells which had been grown in vitro. Three days later, spleen cells were fused with SP-2 mouse myeloma cells utilizing standard techniques (Ueda, R., et al., “Cell Surface Antigens Of Human Renal Cancer Defined By Mouse Monoclonal Antibodies: Identification Of Tissue-Specific Kidney Glycoproteins,” Proc. Natl. Acad. Sci. USA, 78:5122-5126 (1981), which is hereby incorporated by reference).
  • mAbs were dialyzed in 0.1 M NaHCO 3 for 2 hours.
  • One ml of mAb at 1 mg/ml was mixed with 0.1 ml of biotinamidocaproate N-hydroxysuccinamide ester (Sigma) in dimethylsulfoxide (1 mg/ml) and stirred for 4 hours at room temperature. Unbound biotin was removed by dialysis against phosphate buffered saline (“PBS”).
  • PBS phosphate buffered saline
  • Cryostat sections of prostate tissues were placed inside rings of Falcon 3034 plate covers (Becton-Dickenson, Lincoln Park, N.J.) previously coated with 0.45% gelatin solution as described in Marusich, M. F., “A Rapid Method For Processing Very Large Numbers Of Tissue Sections For Immunohistochemical Hybridoma Screening,” J. Immunol. Methods, 111:143-145 (1988), which is hereby incorporated by reference. Plates were stored at ⁇ 80° C. Cryostat sections were fixed with 2% paraformaldehyde in PBS for 10 min at room temperature, and, after washing with PBS, endogenous peroxidase activity was blocked by treatment with 0.3% hydrogen peroxide in PBS for 10 min at room temperature.
  • the anti-mouse immunoglobulin mixed hemadsorption assay was performed as described in Ueda, R., et al., “Cell Surface Antigens Of Human Renal Cancer Defined By Mouse Monoclonal Antibodies: Identification Of Tissue-Specific Kidney Glycoproteins,” Proc. Natl. Acad. Sci. USA, 78:5122-5126 (1981), which is hereby incorporated by reference.
  • To prepare the indicator cells anti-mouse Ig (DAKO Corp.) was conjugated to type O human RBC using 0.01% chromium chloride.
  • Serological assays were performed on cells previously plated in Terasaki plates (Nunc, Denmark). Antibodies were incubated with target cells at room temperature for 1 hour. Target cells were then washed, and indicator cells added for 1 hour.
  • LNCaP cells (2 ⁇ 10 7 ) were biotinylated with biotin-NHSS (at final concentration of 5 mM) for 30 minutes on ice. After washing, the biotinylated cells were resuspended in 1 ml lysis buffer (20 mM Tris/HCl pH 8.0, 1 mM EDTA, 1 mM PMSF, 1% triton X-100) for 30 min on ice. The suspension was centrifuged at 1500 g ⁇ 100 min at 4° C., and the supernatant was centrifuged at 12,000 rpm ⁇ 15 min at 4° C.
  • 1 ml lysis buffer (20 mM Tris/HCl pH 8.0, 1 mM EDTA, 1 mM PMSF, 1% triton X-100
  • the resulting lysate was preabsorbed with rabbit or goat anti-mouse IgG-coated pansorbin for 1 hour at 4° C.
  • the pre-absorbed lysate was incubated with the mAb overnight at 4° C.
  • Rabbit or goat anti-mouse Ig-coated agarose beads were added for 2 hours at 4° C. and then washed.
  • the beads were resuspended in Tris-base/NaCl, added to sample buffer with 2-mercaptoethanol, and boiled for 5 min. After centrifuging, the supernatant was run on an SDS-PAGE 12% gel. The gel was transferred to a nitrocellulose membrane which was blocked and stained with straptavidin-peroxidase.
  • the membrane was developed with diaminobenzidine (“DAB”).
  • DAB diaminobenzidine
  • Sequential immunoprecipitation was similar except that the lysate was initially pre-cleared with one mAb overnight at 4° C. A second mAb was then used to immunoprecipitate the pre-cleared lysate.
  • the membranes were incubated with sheep anti-mouse-Ig-peroxidase 1/5000 in 5% dry milk/TBST for 60 min at room temperature. After repeat washing, the membranes were developed using a chemiluminescent tag designated “ECL” (Amersham Life Sciences, International, Arlington Heights, Ill.) according to the manufacturer's directions. The results of the Western Blot experiment are presented in Table 4.
  • LNCaP cells were tested, without fixation, in vitro, by immunofluorescence.
  • LNCaP cells were washed and incubated with mAb for 1 hour at room temperature and then with a rabbit anti-mouse Ig-fluorescein (DAKO Corp., Santa Barbara, Calif.). Wells were read with a fluorescent microscope.
  • Negative control consisted of an isotype-matched irrelevant mAb, while an anti-class I MHC mAb served as a positive control.
  • Plates were coated with LNCaP cell line lysate as a source of prostate specific membrane antigen and washed to remove unbound material. “Cold” (unlabeled) monoclonal antibody was incubated on the plate for 1 hour at room temperature to allow binding to its antigenic site. Subsequently, a second monoclonal antibody, labeled either with biotin or 125 I, was added for an additional hour. Plates were washed to remove unbound material.
  • the amount of the second monoclonal antibody bound to the prostate specific membrane antigen-coated plate was determined either by avidin-alkaline phosphatase in an enzyme-linked immunoassay (in the case of biotin-labeled second monoclonal antibody) or by physically counting the well in a gamma counter (in the case of 125 I-labeled second monoclonal antibody). Controls consisted of using the same monoclonal antibody both cold and labeled to define “100% competition” or using monoclonal antibody to a totally different molecule (e.g., monoclonal antibody I-56, which detects inhibin, a prostate related protein different from prostate specific membrane antigen) to define “0% competition”.
  • monoclonal antibody I-56 which detects inhibin, a prostate related protein different from prostate specific membrane antigen
  • J591, J533, and E99 each interfere, compete, or block binding of one another but do not block binding of J415 and vice versa.
  • Having pairs of monoclonal antibodies which bind to non-competing sites permits development of antibody sandwich assays for detecting soluble antigens, such as solubilized prostate specific membrane antigen or fragment thereof, in, for example, body fluids.
  • the antigen e.g., prostate specific membrane antigen or a fragment thereof
  • the antigen could be “captured” from body fluid with J591 and, in another step, detected by labeled J415.
  • the non-competing sites are each represented once on the prostate specific membrane antigen molecule
  • adding a combination of J591 plus J415 would bind twice as many monoclonal antibody molecules as either monoclonal antibody alone. Binding two non-competing antigenic binding sites also can result in greater antigen cross-linking and, perhaps, increased internalization.
  • monoclonal antibody J591 can be conjugated with an inactive pro-drug and J415 can be conjugated with a pro-drug activator. Since prodrug and activator would be bound in close proximity only at the site of prostate specific membrane antigen-expressing cells (e.g., prostate cancer cells), prodrug activation to the active form would occur only at those sites.
  • prostate specific membrane antigen-expressing cells e.g., prostate cancer cells
  • FIGS. 1-4 are immuno-electron micrographs which follow the interaction of gold-labeled monoclonal antibody J591 with the cell surface as a function of time. In these figures, the location of the monoclonal antibody is indicated by the black dots.
  • FIG. 1 shows the cell prior to 37° C. incubation. J591 can be seen bound to the cell along the external aspect of the cell membrane.
  • M denotes the cell's mitochondria
  • N denotes its nucleus.
  • FIG. 2 shows the cell after incubation at 37° C. for 5 minutes. The arrow indicates formation of a clathrin-coated pit.
  • FIG. 3 which shows the cell after a 10 minute 37° C.
  • FIG. 4 shows that, after incubation at 37° C. for 15 minutes, monoclonal antibody J591 is contained in endocytic vesicles within the cell, as indicated by the arrows. As can be seen in FIG. 5 , after incubation at 37° C. for 15 minutes, monoclonal antibody J591 is also contained within endosomes, as indicated by the arrows.
  • Total RNA was prepared from 10 7 murine hybridoma J591 cells. A sample of the conditioned medium from these cells was tested for binding to the specific antigen for J591 on prostate cells. The conditioned medium was positive by both ELISA and Western Blot for binding to the antigen.
  • VH and VK cDNA were prepared using reverse transcriptase and mouse ⁇ constant region and mouse IgG constant region primers.
  • the first strand cDNAs were amplified by PCR using a variety of mouse signal sequence primers (6 for VH and 7 for VK).
  • the amplified DNAs were gel-purified and cloned into the vector pT7Blue.
  • VH and VK clones obtained were screened for correct inserts by PCR, and the DNA sequence of selected clones was determined by the dideoxy chain termination method.
  • the corresponding deduced amino acid sequences of J591 VH are also shown in FIG. 7 .
  • the coding strand of the J591 heavy chain's variable region (exclusive of signal sequence and constant region components) has the following nucleotide sequence (designated SEQ. ID. No.
  • the reverse, non-coding strand of the J591 heavy chain's variable region has the following nucleotide sequence (designated SEQ. ID. No. 7): TGAGGAGACTGTGAGAGTGGTGCCTTGGCCCCAGTAGTCAAAGTTCCAAC CAGCTGCACAATAATAGACTGCAGAATCCTCAGATGTTAGGCTGCGGAGC TCCATGTAGGCTGTACTGGAGGACTTGTCTACAGTCAATGTGGCCTTGTC CTCGAACTTCTGATTGTAGGTGGTACCACCATTGTTAGGATTGATGTTTC CAATCCACTCAAGGCTCTTTCCATGGCTCTGCTTCACCCAGTGTATGGTA TATTCAGTGAATGTGTATCCAGAAGTCTTGCAGGATATCCTCACTGAAGT CCCAGGCTTCACCAGT CCCAGGCTTCACCAGTTCAGGTCCAGACTGTTGCAGCTGGACCTCAGCTGGACCTCAGCTGGACCTCAGCTGGACCTCAGCTGGACCTCAGCTGGACCTCAGCTGGACCTCACTGCTGG
  • the protein sequence corresponding to the J591 heavy chain's variable region has the following nucleotide sequence (designated SEQ. ID. No. 8): EVQLQQSGPELVKPGTSVRISCKTSGYTFTEYTIHWVKQSHGKSLEWIGN INPNNGGTTYNQKFEDKATLTVDKSSSTAYMELRSLTSEDSAVYYCAAGW NFDYWGQGTTLTVSS
  • the J591 VH is in Mouse Heavy Chains Subgroup IIA (Kabat et al., Sequences of Proteins of Immunological Interest , U.S. Department of Health and Human Services (1991) (“Kabat”), which is hereby incorporated by reference).
  • the sequence of J591 VH is compared to the consensus sequence for this subgroup in FIG. 8 .
  • the nucleic acid sequence of J591 VK corresponding to the ten identical clones is presented in FIG. 10 , along with the nucleic acid sequence of the corresponding reverse, non-coding strand (designated SEQ. ID. No. 10) and the deduced amino acid sequences, which are designated SEQ. ID. No. 11, SEQ. ID. No. 12, and SEQ. ID. No. 13. These sequences include part of the signal sequence and part of the constant region of the antibody.
  • the coding strand of the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to the ten identical clones has the following nucleotide sequence (designated SEQ. ID. No.
  • the reverse, non-coding strand of the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to the ten identical clones has the following nucleotide sequence (designated SEQ. ID. No. 15): .
  • the protein sequence corresponding to the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to the ten identical clones has the following nucleotide sequence (designated SEQ. ID. No. 16): NIVMTQSPKSMSMSVGERVTLTCKASENVVTYVSWYQQKPEQSPKLLIYG ASNRYTGVPDRFTGSGSATDFTLTISSVQAEDLADYHCGQGYSYPYTFGG GTKLEIK
  • the coding strand of the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to clone VK17 has the following nucleotide sequence (designated SEQ. ID. No. 17): GACATTGTGATGACCCAGTCTCACAAATTCATGTCCACATCAGTAGGAGA CAGGGTCAGCATCATCTGTAAGGCCAGTCAAGATGTGGGTACTGCTGTAG ACTGGTATCAACAGAAACCAGGACAATCTCCTAAACTACTGATTTATTGG GCATCCACTCGGCACACTGGAGTCCCTGATCGCTTCACAGGCAGTGGATC TGGGACAGACTTCACTCTCACCATTACTAATGTTCAGTCTGAAGACTTGG CAGATTATTTCTGTCAGCAATATAACAGCTATCCTCTCACGTTCGGTGCT GGGACCATGCTGGACCTGAAA
  • the reverse, non-coding strand of the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to clone VK17 has the following nucleotide sequence (designated SEQ. ID. No. 18): TTTCAGGTCCAGCATGGTCCCAGCACCGAACGTGAGAGGATAGCTGTTAT ATTGCTGACAGAAATAATCTGCCAAGTCTTCAGACTGAACATTAGTAATG GTGAGAGTGAAGTCTGTCCCAGATCCACTGCCTGTGAAGCGATCAGGGAC TCCAGTGTGCCGAGTGGATGCCCAATAAATCAGTAGTTTAGGAGATTGTC CTGGTTTCTGTTGATACCAGTCTACAGCAGTACCCACATCTTGACTGGCC TTACAGATGATGCTGACCCTGTCTCCTACTGATGTGGACATGAATTTGTG AGACTGGGTCATCACAATGTC
  • the protein sequence corresponding to the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to clone VK17 has the following nucleotide sequence (designated SEQ. ID. No. 19): DIVMTQSHKFMSTSVGDRVSIICKASQDVGTAVDWYQQKPGQSPKLLIYW ASTRHTGVPDRFTGSGSGTDFTLTITNVQSEDLADYFCQQYNSYPLTFGA GTMLDLK
  • J591 VK is in the Mouse Kappa Chains Subgroup V (Kabat, which is hereby incorporated by reference).
  • the sequence of J591 VK corresponding to the ten identical clones is compared to the consensus sequence for the subgroup in FIG. 11 .
  • Preferred J591's are those having heavy chain variable region DNA coding strand sequences corresponding to SEQ. ID. No. 6 and non-coding strand (reverse) sequences corresponding to SEQ. ID. No. 7.
  • the heavy chain variable region of J591 preferably has an amino acid sequence corresponding to SEQ. ID. No. 8.
  • the light chain variable region of J591 preferably has a DNA coding strand sequence corresponding to SEQ. ID. No. 17, a DNA non-coding strand (reverse) sequence corresponding to SEQ. ID. No. 18, and a amino acid sequence corresponding to SEQ. ID. No. 19.
  • Cancer tissues from 23 carcinomas were pre-cooled in liquid nitrogen, snap-frozen in OCT compound (Miles, Elkhart, Ind.) on dry ice, and stored at ⁇ 80° C.
  • Cryostat tissue sections (5 ⁇ m) were fixed in cold acetone (4° C.) for 10 minutes.
  • mAbs (5 ⁇ g/ml or hybridoma supernatants) were incubated for 1 hour at room temperature.
  • Antibody binding was detected using rabbit anti-mouse Ig-peroxidase (Dako, Carpinteria, Calif.) as a secondary antibody and DAB (Sigma, St. Louis, Mo.) as chromogen. Isotype-matched irrelevant antibody was used as negative control.
  • FIGS. 2A-2F show the immunohistochemical reactivity of mAb J591 to neovasculature of renal, urothelial, colon, lung, and breast carcinomas, and metastatic adenocarcinoma to the liver.

Abstract

The present invention is directed to the use of antibodies or binding portions thereof, probes, ligands, or other biological agents which either recognize an extracellular domain of prostate specific membrane antigen or bind to and are internalized with prostate specific membrane antigen. These biological agents can be labeled and used for detection of cancerous tissues, particularly cancerous tissues proximate to or containing vascular endothelial cells, which express an extracellular domain of prostate specific membrane antigen. The labeled biological agents can also be used to detect normal, benign hyperplastic, and cancerous prostate epithelial cells or portions thereof. They also can be used alone or bound to a substance effective to ablate or kill such cells as a therapy for prostate or other cancers. Also disclosed are four hybridoma cell lines, each of which produces a monoclonal antibody recognizing extracellular domains of prostate specific membrane antigens of normal, benign hyperplastic, and cancerous prostate epithelial cells or portions thereof.

Description

  • The present application claims the benefit of U.S. Provisional Patent Application 60/022,125, filed Jul. 18, 1996, and is a continuation-in-part of U.S. patent application Ser. No. 08/838,682, filed Apr. 9, 1997, which claims the benefit of U.S. Provisional Patent Application Ser. No. 60/016,976, filed May 6, 1996.
  • FIELD OF THE INVENTION
  • The present invention relates to the treatment and diagnosis of cancer with biological agents.
  • BACKGROUND OF THE INVENTION
  • In spite of improved treatments for certain forms of cancer, it is still a leading cause of death in the United States. Since the chance for complete remission of cancer is, in most cases, greatly enhanced by early diagnosis, it is very desirable that physicians be able to detect cancers before a substantial tumor develops. However, the development of methods that permit rapid and accurate detection of many forms of cancers continues to challenge the medial community. One such illustrative form of cancer is prostate cancer.
  • Prostate cancer is the most common cancer in men with an estimated 317,000 cases in 1996 in the United States. It is the second leading cause of death among men who die from neoplasia with an estimated 40,000 deaths per year. Prompt detection and treatment is needed to limit mortality caused by prostate cancer.
  • Detection of Prostate Cancer
  • When it metastasizes, prostatic dancer has a distinct predilection for bone and lymph nodes. Saitoh et al., “Metastatic Patterns of Prostatic Cancer. Correlation Between Sites And Number Of Organs Involved,” Cancer, 54:3078-3084 (1984). At the time of clinical diagnosis, as many as 25% of patients have bone metastasis demonstrable by radionuclide scans. Murphy, G. P., et al., “The National Survey Of Prostate Cancer In The United States By The American College Of Surgeons,” J. Urol., 127:928-939 (1982). Accurate clinical evaluation of nodal involvement has proven to be difficult. Imaging techniques such as computed tomography (“CT”) or magnetic resonance (“MR”) imaging are unable to distinguish metastatic prostate cancer involvement of lymph nodes by criterion other than size (i.e., >1 cm). Therefore, by definition, these imaging modalities are inherently insensitive in the detection of small volume (<1 cm) disease as well as non-specific in the detection of larger volume adenopathy. A recent study assessed the accuracy of MR in patients with clinically localized prostate cancer. Rifkin et al., “Comparison Of Magnetic Resonance Imaging And Ultrasonography In Staging Early Prostate Cancer,” N. Engel. J. Med., 323:621-626 (1990). In this study, 194 patients underwent an MR and 185 of these patients had a lymph node dissection. 23 (13%) patients had pathologically involved lymph nodes. MR was suspicious in only 1 of these 23 cases resulting in a sensitivity of 4%. Similar results have also been noted with CT scans. Gasser et al., “MRI And Ultrasonography In Staging Prostate Cancer,” N. Engl. J. Med. (Correspondence), 324(7):49-495 (1991).
  • The elevation of serum acid phosphatase activity in patients having metastasized prostate carcinoma was first reported by Gutman et al., J. Clin. Invest 17:473 (1938). In cancer of the prostate, prostatic acid phosphatase is released from the cancer tissue into the blood stream with the result that the total serum acid phosphatase level can be greatly increased above normal values. Numerous studies of this enzyme and its relation to prostatic cancer have been made since that time, e.g. Yam, Amer. J. Med. 56:604 (1974). However, the measurement of serum acid phosphatase is elevated in about 65-90 percent of patients having carcinoma of the prostate with bone metastasis; in about 30 percent of patients without roentgenological evidence of bone metastasis; and in about only 5-10 percent of patients lacking clinically demonstrable metastasis.
  • Prior art attempts to develop a specific test for prostatic acid phosphatase have met with only limited success, because techniques which rely on enzyme activity on a so-called “specific” substrate cannot take into account other biochemical and immunochemical differences among the many acid phosphatases which are unrelated to enzyme activity of prostate origin. In the case of isoenzymes, i.e. genetically defined enzymes having the same characteristic enzyme activity and a similar molecular structure but differing in amino acid sequences and/or content and, therefore, immunochemically distinguishable, it would appear inherently impossible to distinguish different isoenzyme forms merely by the choice of a particular substrate. It is, therefore, not surprising that none of these prior art methods is highly specific for the direct determination of prostatic acid phosphatase activity; e.g. see Cancer 5:236 (1952); J. Lab. Clin. Med. 82:486 (1973); Clin. Chem. Acta. 44:21 (1973); and J. Physiol. Chem. 356:1775 (1975).
  • In addition to the aforementioned problems of non-specificity which appear to be inherent in many of the prior art reagents employed for the detection of prostate acid phosphatase, there have been reports of elevated serum acid phosphatase associated with other diseases, which further complicates the problem of obtaining an accurate clinical diagnosis of prostatic cancer. For example, Tuchman et al., Am. J. Med. 27:959 (1959) noted that serum acid phosphatase levels appear to be elevated in patients with Gaucher's disease.
  • Due to the inherent difficulties in developing a “specific” substrate for prostate acid phosphatase, several researchers have developed immunochemical methods for the detection of prostate acid phosphatase. However, the previously reported immunochemical methods have drawbacks of their own which have precluded their widespread acceptance. For example, Shulman et al., Immunology 93:474 (1964) described an immuno-diffusion test for the detection of human prostate acid phosphatase. Using antisera prepared from a prostatic fluid antigen obtained by rectal massage from patients with prostatic disease, no cross-reactivity precipitin line was observed in the double diffusion technique against extracts of normal kidney, testicle, liver, and lung. However, this method has the disadvantages of limited sensitivity, even with the large amounts of antigen employed, and of employing antisera which may cross-react with other, antigenically unrelated serum protein components present in prostatic fluid.
  • WO 79/00475 to Chu et. al. describes a method for the detection of prostatic acid phosphatase isoenzyme patterns associated with prostatic cancer which obviates many of the above drawbacks. However, practical problems are posed by the need for a source of cancerous prostate tissue from which the diagnostically relevant prostatic acid phosphatase isoenzyme patterns associated with prostatic cancer are extracted for the preparation of antibodies thereto.
  • In recent years, considerable effort has been spent to identify enzyme or antigen markers for various types of malignancies with the view towards developing specific diagnostic reagents. The ideal tumor marker would exhibit, among other characteristics, tissue or cell-type specificity. Previous investigators have demonstrated the occurrence of human prostate tissue-specific antigens.
  • Treatment of Prostate Cancer
  • As described in W. J. Catalona, “Management of Cancer of the Prostate,” New Engl. J. Med., 331(15):996-1004 (1994), the management of prostate cancer can be achieved by watchful waiting, curative treatment, and palliation.
  • For men with a life expectancy of less than 10 years, watchful waiting is appropriate where low-grade, low-stage prostate cancer is discovered at the time of a partial prostatectomy for benign hyperplasia. Such cancers rarely progress during the first five years after detection. On the other hand, for younger men, curative treatment is often more appropriate.
  • Where prostate cancer is localized and the patient's life expectancy is 10 years or more, radical prostatectomy offers the best chance for eradication of the disease. Historically, the drawback of this procedure is that most cancers had spread beyond the bounds of the operation by the time they were detected. However, the use of prostate-specific antigen testing has permitted early detection of prostate cancer. As a result, surgery is less extensive with fewer complications. Patients with bulky, high-grade tumors are less likely to be successfully treated by radical prostatectomy.
  • After surgery, if there are detectable serum prostate-specific antigen concentrations, persistent cancer is indicated. In many cases, prostate-specific antigen concentrations can be reduced by radiation treatment. However, this concentration often increases again within two years.
  • Radiation therapy has also been widely used as an alternative to radical prostatectomy. Patients generally treated by radiation therapy are those who are older and less healthy and those with higher-grade, more clinically advanced tumors. Particularly preferred procedures are external-beam therapy which involves three dimensional, conformal radiation therapy where the field of radiation is designed to conform to the volume of tissue treated; interstitial-radiation therapy where seeds of radioactive compounds are implanted using ultrasound guidance; and a combination of external-beam therapy and interstitial-radiation therapy.
  • For treatment of patients with locally advanced disease, hormonal therapy before or following radical prostatectomy or radiation therapy has been utilized. Hormonal therapy is the main form of treating men with disseminated prostate cancer. Orchiectomy reduces serum testosterone concentrations, while estrogen treatment is similarly beneficial. Diethylstilbestrol from estrogen is another useful hormonal therapy which has a disadvantage of causing cardiovascular toxicity. When gonadotropin-releasing hormone agonists are administered testosterone concentrations are ultimately reduced. Flutamide and other nonsteroidal, anti-androgen agents block binding of testosterone to its intracellular receptors. As a result, it blocks the effect of testosterone, increasing serum testosterone concentrations and allows patients to remain potent—a significant problem after radical prostatectomy and radiation treatments.
  • Cytotoxic chemotherapy is largely ineffective in treating prostate cancer. Its toxicity makes such therapy unsuitable for elderly patients. In addition, prostate cancer is relatively resistant to cytotoxic agents.
  • Use of Monoclonal Antibodies in Prostate Cancer Detection and Treatment
  • Theoretically, radiolabeled monoclonal antibodies (“mAbs”) offer the potential to enhance both the sensitivity and specificity of detecting prostatic cancer within lymph nodes and elsewhere. While many mAbs have previously been prepared against prostate related antigens, none of these mAbs were specifically generated with an imaging objective in mind. Nevertheless, the clinical need has led to evaluation of some of these mAbs as possible imaging agents. Vihko et al., “Radioimaging of Prostatic Carcinoma With Prostatic Acid Phosphatase-Specific Antibodies,” Biotechnology in Diagnostics, 131-134 (1985); Babaian et al., “Radioimmunological Imaging of Metastatic Prostatic Cancer With 111-Indium-Labeled Monoclonal Antibody PAY 276,” J. Urol., 137:439-443 (1987); Leroy et al., “Radioimmunodetection Of Lymph Node Invasion In Prostatic Cancer. The Use Of Iodine 123 (123-I)-Labeled Monoclonal Anti-Prostatic Acid Phosphatase (PAP) 227 A F (ab′) 2 Antibody Fragments In Vivo,” Cancer, 64:1-5 (1989); Meyers et al., “Development Of Monoclonal Antibody Imaging Of Metastatic Prostatic Carcinoma,” The Prostate, 14:209-220 (1989).
  • In some cases, the monoclonal antibodies developed for detection and/or treatment of prostate cancer recognize antigens specific to malignant prostatic tissues. Such antibodies are thus used to distinguish malignant prostatic tissue (for treatment or detection) from benign prostatic tissue. See U.S. Pat. No. 4,970,299 to Bazinet et al. and U.S. Pat. No. 4,902,615 to Freeman et al.
  • Other monoclonal antibodies react with surface antigens on all prostate epithelial cells whether cancerous or benign. See U.S. Pat. No. 4,446,122 and Re 33,405 to Chu et al., U.S. Pat. No. 4,863,851 to McEwan et al., and U.S. Pat. No. 5,055,404 to Ueda et al. However, the antigens detected by these monoclonal antibodies are present in the blood and, therefore, compete with antigens at tumor sites for the monoclonal antibodies. This causes background noise which makes the use of such antibodies inadequate for in vivo imaging. In therapy, such antibodies, if bound to a cytotoxic agent, could be harmful to other organs.
  • Horoszewicz et al., “Monoclonal Antibodies to a New Antigenic Marker in Epithelial Prostatic Cells and Serum of Prostatic Cancer Patients,” Anticancer Research, 7:927-936 (1987) (“Horoszewicz”) and U.S. Pat. No. 5,162,504 to Horoszewicz describe an antibody, designated 7E11, which recognizes prostate specific membrane antigen (“PSMA”). Israeli et al., “Molecular Cloning of a Complementary DNA Encoding a Prostate-specific Membrane Antigen,” Cancer Research, 53:227-230 (1993) (“Israeli”) describes the cloning and sequencing of PSMA and reports that PSMA is prostate-specific and shows increased expression levels in metastatic sites and in hormone-refractory states. Other studies have indicated that PSMA is more strongly expressed in prostate cancer cells relative to cells from the normal prostate or from a prostate with benign hyperplasia. Furthermore, PSMA is not found in serum (Troyer et al., “Detection and Characterization of the Prostate-Specific Membrane Antigen (PSMA) in Tissue Extracts and Body Fluids,” Int. J. Cancer, 62:552-558 (1995)).
  • These characteristics make PSMA an attractive target for antibody mediated targeting for imaging and therapy of prostate cancer. Imaging studies using indium-labeled 7E11 have indicated that the antibody localizes quite well to both the prostate and to sites of metastasis. In addition, 7E11 appears to have clearly improved sensitivity for detecting lesions compared to other currently available imaging techniques, such as CT and MR imaging or bone scan. Bander, “Current Status of Monoclonal Antibodies for Imaging and Therapy of Prostate Cancer,” Sem. In Oncology, 21:607-612 (1994).
  • However, the use of 7E11 and other known antibodies to PSMA to mediate imaging and therapy has several disadvantages. First, PSMA is an integral membrane protein known to have a short intracellular tail and a long extracellular domain. Biochemical characterization and mapping (Troyer et al., “Biochemical Characterization and Mapping of the 7E11-C5.3 Epitope of the Prostate-specific Membrane Antigen,” Urol. Oncol., 1:29-37 (1995)) have shown that the epitope or antigenic site to which the 7E11 antibody binds is present on the intracellular portion of the molecule. Because antibody molecules do not, under normal circumstances, cross the cell membrane unless they bind to the extracellular portion of a molecule and become translocated intracellularly, the 7E11 antibody does not have access to its antigenic target site in an otherwise healthy, viable cell.
  • Consequently, imaging using 7E11 is limited to the detection of dead cells within tumor deposits. Additionally, the therapeutic use of the 7E11 antibody is limited, because only cells that are already dead or tissue containing a large proportion of dead cells can be effectively targeted.
  • Although the inadequacies and problems in the diagnosis and treatment of one particular type of cancer are the focus of the preceding discussion, prostate cancer is merely a representative model. The diagnosis and treatment of numerous other cancers have similar problems.
  • The present invention is directed to overcoming the deficiencies of prior art antibodies in diagnosing and treating prostate and other types of cancer.
  • SUMMARY OF THE INVENTION
  • One aspect of the present invention relates to a method of ablating or killing cancerous cells. The process involves providing a biological agent which, when contacted with an extracellular domain of prostate specific membrane antigen, recognizes the extracellular domain of prostate specific membrane antigen. These biological agents are contacted with vascular endothelial cells proximate to the cancerous cells under conditions effective to permit both binding of the biological agent to the vascular endothelial cells proximate to the cancerous cells and killing or ablating of the cancerous cells. The biological agent can be used alone or can be bound to a substance effective to kill or ablate the cancerous cells upon binding of the biological agent to vascular endothelial cells that are proximate to the cancerous cells.
  • In a particularly preferred embodiment of the method of ablating or killing cancerous cells in accordance with the present invention, the biological agent, when contacted with an extracellular domain of prostate specific membrane antigen, binds to and is internalized with the prostate specific membrane antigen of such cells. Preferred biological agents for use in the method of ablating or killing cancerous cells in accordance with the present invention are antibodies or binding portions thereof, probes, or ligands. The methods of the present invention are particularly useful in killing or ablating renal, urothelial, colon, rectal, lung, and breast cancerous cells and cancerous cells of metastatic adenocarcinoma to the liver.
  • Another aspect of the present invention relates to a method of detecting cancerous tissue in a biological sample. This method involves providing a biological agent which, when contacted with an extracellular domain of prostate specific membrane antigen, binds to the extracellular domain of prostate specific membrane antigen. The biological agent is bound to a label effective to permit detection of vascular endothelial cells proximate to or within the cancerous tissue upon binding of the biological agent to the vascular endothelial cells proximate to or within the cancerous tissue. The biological sample is contacted with the biological agent having a label under conditions effective to permit binding of the biological agent to the vascular endothelial cells proximate to or within the cancerous tissue in the biological sample. The presence of cancerous tissue in the biological sample is detected by detection of the label.
  • In a particularly preferred embodiment of the method of detecting cancerous tissue in accordance with the present invention, the biological agent is one that, when contacted with an extracellular domain of prostate specific membrane antigen, binds to and is internalized with the prostate specific membrane antigen. Preferred biological agents for use in the method of detecting cancerous tissue in accordance with the present invention are antibodies or binding portions thereof, probes, or ligands. The method is especially useful in detecting renal, urothelial, colon, rectal, lung, and breast cancerous tissue and cancerous tissue of metastatic adenocarcinoma to the liver.
  • Still another aspect of the present invention relates to a method of ablating or killing normal, benign hyperplastic, and cancerous prostate epithelial cells. The process involves providing a biological agent which recognizes an extracellular domain of prostate specific membrane antigen. The biological agent can be used alone or can be bound to a substance effective to kill the cells upon binding of the biological agent to the cells. These biological agents are then contacted with the cells under conditions effective to permit both binding of the biological agent to the extracellular domain of the prostate specific membrane antigen and killing or ablating of the cells.
  • In a particularly preferred embodiment of the method of ablating or killing normal, benign hyperplastic, and cancerous prostate epithelial cells in accordance with the present invention, the biological agent binds to and is internalized with the prostate specific membrane antigen of such cells. Preferred biological agents for use in the method of ablating or killing normal, benign hyperplastic, and cancerous prostate epithelial cells in accordance with the present invention are antibodies or binding portions thereof, probes, or ligands.
  • Another aspect of the present invention relates to a method of detecting normal, benign hyperplastic, and cancerous prostate epithelial cells or portions thereof in a biological sample. This method involves providing a biological agent which binds to an extracellular domain of prostate specific membrane antigen. The biological agent is bound to a label effective to permit detection of the cells or portions thereof upon binding of the biological agent to the cells or portions thereof. The biological sample is contacted with the biological agent having a label under conditions effective to permit binding of the biological agent to the extracellular domain of the prostate specific membrane antigen of any of the cells or portions thereof in the biological sample. The presence of any cells or portions thereof in the biological sample is detected by detection of the label.
  • In a particularly preferred embodiment of the method of detecting normal, benign hyperplastic, and cancerous prostate epithelial cells in accordance with the present invention, the biological agent binds to and is internalized with the prostate specific membrane antigen of such cells. Preferred biological agents for use in the method of detecting normal, benign hyperplastic, and cancerous prostate epithelial cells in accordance with the present invention are antibodies or binding portions thereof, probes, or ligands.
  • Another aspect of the present invention pertains to a biological agent that recognizes an extracellular domain of prostate specific membrane antigen. In a preferred embodiment, the isolated biological agent binds to and is internalized with the prostate specific membrane antigen. Preferred isolated biological agents which recognize an extracellular domain of prostate specific membrane antigen in accordance with the present invention are isolated antibodies or binding portions thereof, probes, or ligands. Hybridoma cell lines that produce monoclonal antibodies of these types are also disclosed.
  • The biological agents of the present invention recognize the extracellular domain of antigens of normal, benign hyperplastic, and cancerous prostate epithelial cells. Unlike the 7E11 antibody, which recognizes an epitope of prostate-associated antigens which are exposed extracellularly only after cell lysis, the biological agents of the present invention bind to antigenic epitopes which are extracellularly exposed in living prostate cells. Using the biological agents of the present invention, living, unfixed normal, benign hyperplastic, and cancerous prostate epithelial cells can be targeted, which makes treatment and diagnosis more effective. In a preferred embodiment for treating prostate cancer, the biological agents of the present invention also bind to and are internalized with the prostate specific membrane antigen, which permits the therapeutic use of intracellularly acting cytotoxic agents.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is an immuno-electron micrograph of gold-labeled monoclonal antibody J591 on the surface of LNCaP cells after incubation at 4° C.
  • FIG. 2 is an immuno-electron micrograph of LNCaP cells treated with gold-labeled monoclonal antibody J591 after 5 minutes incubation at 37° C.
  • FIG. 3 is an immuno-electron micrograph of LNCaP cells treated with gold-labeled monoclonal antibody J591 after 10 minutes incubation at 37° C.
  • FIG. 4 is an immuno-electron micrograph of LNCaP cells treated with gold-labeled monoclonal antibody J591 after 15 minutes incubation at 37° C.
  • FIG. 5 is an immuno-electron micrograph of LNCaP cells treated with gold-labeled monoclonal antibody J591 after 15 minutes at 37° C. showing J591 within endosomes.
  • FIG. 6 summarizes the sequencing strategy of the heavy chain of monoclonal antibody J591.
  • FIG. 7 shows the nucleotide sequence of the heavy chain of monoclonal antibody J591 (designated SEQ. ID. No. 1), the nucleotide sequence of the corresponding reverse, non-coding strand (designated SEQ. ID. No. 2), and the corresponding deduced amino acid sequences (designated SEQ. ID. Nos. 3, 4, and 5).
  • FIG. 8 is a comparison of the heavy chain of monoclonal antibody J591 with the consensus sequence for Mouse Heavy Chains Subgroup IIA.
  • FIG. 9 summarizes the sequencing strategy of the kappa light chain of monoclonal antibody J591.
  • FIG. 10 shows the nucleotide sequences of the kappa light chain of monoclonal antibody J591 (designated SEQ. ID. No. 9), the nucleotide sequence of the corresponding reverse, non-coding strand (designated SEQ. ID. No. 10), and the corresponding deduced amino acid sequence (designated SEQ. ID. Nos. 11, 12, and 13).
  • FIG. 11 is a comparison of the kappa light chain of monoclonal antibody J591 with the consensus sequence for Mouse Kappa Chains Subgroup V.
  • FIGS. 12A-12F are micrographs (250× magnification) showing the immunohistochemical reactivity of mAb J591 to neovasculature of various carcinomas.
  • DETAILED DESCRIPTION OF THE INVENTION
  • One aspect of the present invention relates to a method of ablating or killing normal, benign hyperplastic, and cancerous prostate epithelial cells. The process involves providing a biological agent, such as an antibody or binding portion thereof, probe, or ligand, which binds to an extracellular domain of prostate specific membrane antigen of (i.e., a portion of prostate specific membrane antigen which is external to) such cells. The biological agent can be used alone or can be bound to a substance effective to kill the cells upon binding of the biological agent to the cells. These biological agents are then contacted with the cells under conditions effective to permit both binding of the biological agent to the extracellular domain of the prostate specific membrane antigen and killing or ablating of the cells. In its preferred form, such contacting is carried out in a living mammal by administering the biological agent to the mammal under conditions effective to permit both binding of the biological agent to the extracellular domain of the prostate specific membrane antigen and killing or ablating of the cells. Such administration can be carried out orally or parenterally.
  • In a particularly preferred embodiment of the method of ablating or killing normal, benign hyperplastic, and cancerous prostate epithelial cells in accordance with the present invention, the biological agent binds to and is internalized with the prostate specific membrane antigen of such cells. Again, the biological agent can be used alone. Alternatively, the biological agent can be bound to a substance effective to kill the cells upon binding of the biological agent to prostate specific membrane antigen and upon internalization of the biological agent with the prostate specific membrane antigen.
  • The mechanism by which the biological agent is internalized with the prostate specific membrane antigen is not critical to the practice of the present invention. For example, the biological agent can induce internalization of the prostate specific membrane antigen. Alternatively, internalization of the biological agent can be the result of routine internalization of prostate specific membrane antigen.
  • The above-described biological agents. (i.e., biological agents, such as an antibody or binding portion thereof, probe, or ligand which, when contacted with an extracellular domain of prostate specific membrane antigen, recognizes the extracellular domain of prostate specific membrane antigen and, preferably, is internalized therewith) can be used to ablate or kill cancerous cells. In this aspect of the present invention, the biological agent can be used alone or can be bound to a substance effective to kill the cancerous cells upon binding of the biological agent to vascular endothelial cells proximate thereto. These biological agents are contacted with vascular endothelial cells proximate to the cancerous cells. The contacting is carried out under conditions that are effective to permit binding of the biological agent to the vascular endothelial cells proximate to the cancerous cells and, in addition, that are effective to kill or ablate the cancerous cells. The mechanism by which the cancerous cells are killed or ablated is not critical to the practice of the present invention. For example, the cancerous cells can be killed or ablated directly by the biological agent as a consequence of their proximity to the vascular endothelial cells to which the biological agent binds. Alternatively, the biological agent can kill, ablate, or otherwise change the properties of the vascular endothelial cells to which it binds so that blood flow to the cancerous cells proximate thereto is stopped or otherwise reduced, thereby causing the cancerous cells to be killed or ablated. Thus, the method of the present invention is particularly useful for killing or ablating vascular endothelial cells in cancerous tissue as well as the cancerous cells contained in cancerous tissue.
  • In a particularly preferred embodiment of the method of ablating or killing cancerous cells in accordance with the present invention, the biological agent employed is one that, when contacted with an extracellular domain of prostate specific membrane antigen, binds to and is internalized with the extracellular domain of prostate specific membrane antigen. The methods of the present invention are particularly useful to kill or ablate cancerous prostate epithelial cells as well as cancerous cells other than cancerous prostate epithelial cells. Examples of cancerous cells which are not cancerous prostate epithelial cells are renal, urothelial, colon, rectal, lung, and breast cancerous cells and cancerous cells of metastatic adenocarcinoma to the liver. Although the method of the present invention can be used to kill or ablate any cell which expresses an extracellular domain of prostate specific membrane antigen or a portion thereof or whose subsistence is dependent upon cells which express an extracellular domain of prostate specific membrane antigen or a portion thereof, the method of the present invention is particularly useful to kill or ablate cancerous cells, because the vascular endothelial cells supplying blood to cancerous tissues (e.g., tumors, collections of cancerous cells, or other cancerous masses) express an extracellular domain of prostate specific membrane antigen, irrespective of the type of cancer involved. In contrast, vascular endothelial cells supplying blood to normal tissues do not express an extracellular domain of prostate specific membrane antigen.
  • Another aspect of the present invention relates to a method of detecting normal, benign hyperplastic, and cancerous epithelial cells or portions thereof in a biological sample. This method involves providing a biological agent, such as an antibody or binding portion thereof, probe, or ligand, which binds to an extracellular domain of prostate specific membrane antigen of such cells. The biological agent is bound to a label effective to permit detection of the cells or portions (e.g., prostate specific membrane antigen or fragments thereof liberated from such normal, benign hyperplastic, and cancerous cells) thereof upon binding of the biological agent to the cells or portions thereof. The biological sample is contacted with the biological agent having a label under conditions effective to permit binding of the biological agent to the extracellular domain of the prostate specific membrane antigen of any of the cells or portions thereof in the biological sample. The presence of any cells or portions thereof in the biological sample is detected by detection of the label. In its preferred form, such contacting is carried out in a living mammal and involves administering the biological agent to the mammal under conditions effective to permit binding of the biological agent to the prostate specific membrane antigen of any of the cells or portions thereof in the biological sample. Again, such administration can be carried out orally or parenterally.
  • The method of the present invention can be used to screen patients for diseases associated with the presence of normal, benign hyperplastic, and cancerous epithelial cells or portions thereof. Alternatively, it can be used to identify the recurrence of such diseases, particularly when the disease is localized in a particular biological material of the patient. For example, recurrence of prostatic disease in the prostatic fossa may be encountered following radical prostatectomy. Using the method of the present invention, this recurrence can be detected by administering a short range radiolabeled antibody to the mammal and then detecting the label rectally, such as with a transrectal detector probe.
  • Alternatively, the contacting step can be carried out in a sample of serum or urine or other body fluids, such as to detect the presence of PSMA in the body fluid. When the contacting is carried out in a serum or urine sample, it is preferred that the biological agent recognize substantially no antigens circulating in the blood other than PSMA. Since intact prostate cells do not excrete or secrete PSMA into the extracellular environment, detecting PSMA in serum, urine, or other body fluids generally indicates that prostate cells are being lysed. Thus, the biological agents and methods of the present invention can be used to determine the effectiveness of a prostate cancer treatment protocol by monitoring the level of PSMA in serum, urine or other body fluids.
  • In a particularly preferred embodiment of the method of detecting normal, benign hyperplastic, and cancerous prostate epithelial cells in accordance with the present invention, the biological agent, such as the antibody or binding portion thereof, probe, or ligand, binds to and is internalized with the prostate specific membrane antigen of such cells. Again, the biological agent is bound to a label effective to permit detection of the cells or portions thereof upon binding of the biological agent to and internalization of the biological agent with the prostate specific membrane antigen.
  • Another aspect of the present invention relates to a method of detecting cancerous tissue in a biological sample. This method involves providing the above-described biological agent (i.e., a biological agent, such as an antibody or binding portion thereof, probe, or ligand which, when contacted with an extracellular domain of prostate specific membrane antigen, recognizes the extracellular domain of prostate specific membrane antigen). The biological agent is bound to a label that is effective to permit detection of vascular endothelial cells proximate to or within the cancerous tissue upon binding of the biological agent to vascular endothelial cells proximate to or within the cancerous tissue. The biological sample is then contacted with the biological agent having a label. Contacting is carried out under conditions effective to permit binding of the biological agent to the vascular endothelial cells proximate to or within the cancerous tissue in the biological sample. The presence of cancerous cells or portions thereof in the biological sample is detected by detection of the label.
  • Rather than contacting the entire biological sample with the biological agent, it is contemplated that a portion of the biological sample can be used. For example, a tissue biopsy sample can be contacted with the biological agent to determine the presence of cancerous tissue in the tissue biopsy sample as well as in the larger biological sample from which it is taken. Alternatively, the biological agent can be contacted with a serum or urine sample to acertain whether any vascular endothelial cells expressing an extracellular domain of prostate specific membrane antigen are present therein. Since vascular endothelial cells expressing an extracellular domain of prostate specific membrane antigen are found in the vasculature of cancerous tissues but not in the vasculature of normal tissues, detection of the label in a serum or urine sample indicates the presence of cancerous tissue in the larger biological sample from which it is taken (e.g., a patient).
  • In a particularly preferred embodiment of the method of detecting cancerous tissues in accordance with the present invention, the biological agent employed is one that, when contacted with an extracellular domain of prostate specific membrane antigen, binds to and is internalized with the prostate specific membrane antigen. The methods of the present invention can be used to detect cancerous prostate epithelial cells as well as cancerous tissues containing cancerous cells other than cancerous prostate epithelial cells. Examples of cancerous tissues containing cancerous cells other than cancerous prostate epithelial cells which can be detected with the methods of the present invention include renal, urothelial, colon, rectal, lung, and breast cancerous tissue and cancerous tissue of metastatic adenocarcinoma to the liver.
  • As indicated above, biological agents suitable for either killing, ablating, or detecting cancerous cells and normal, benign hyperplastic, and cancerous prostate epithelial cells include antibodies, such as monoclonal or polyclonal antibodies. In addition, antibody fragments, half-antibodies, hybrid derivatives, probes, and other molecular constructs may be utilized. These biological agents, such as antibodies, binding portions thereof, probes, or ligands, bind to extracellular domains of prostate specific membrane antigens or portions thereof in normal, benign hyperplastic, and cancerous prostate epithelial cells. As a result, when practicing the methods of the present invention to kill, ablate, or detect normal, benign hyperplastic, and cancerous prostate epithelial cells, the biological agents bind to all such cells, not only to cells which are fixed or cells whose intracellular antigenic domains are otherwise exposed to the extracellular environment. Consequently, binding of the biological agents is concentrated in areas where there are prostate epithelial cells, irrespective of whether these cells are fixed or unfixed, viable or necrotic. Additionally or alternatively, these biological agents, such as antibodies, binding portions thereof, probes, or ligands, bind to and are internalized with prostate specific membrane antigens or portions thereof in normal, benign hyperplastic, and cancerous prostate epithelial cells.
  • Monoclonal antibody production may be effected by techniques which are well-known in the art. Basically, the process involves first obtaining immune cells (lymphocytes) from the spleen of a mammal (e.g., mouse) which has been previously immunized with the antigen of interest either in vivo or in vitro. The antibody-secreting lymphocytes are then fused with (mouse) myeloma cells or transformed cells, which are capable of replicating indefinitely in cell culture, thereby producing an immortal, immunoglobulin-secreting cell line. The resulting fused cells, or hybridomas, are cultured, and the resulting colonies screened for the production of the desired monoclonal antibodies. Colonies producing such antibodies are cloned, and grown either in vivo or in vitro to produce large quantities of antibody. A description of the theoretical basis and practical methodology of fusing such cells is set forth in Kohler and Milstein, Nature 256:495 (1975), which is hereby incorporated by reference.
  • Mammalian lymphocytes are immunized by in vivo immunization of the animal (e.g., a mouse) with the protein or polypeptide of the present invention. Such immunizations are repeated as necessary at intervals of up to several weeks to obtain a sufficient titer of antibodies. Following the last antigen boost, the animals are sacrificed and spleen cells removed.
  • Fusion with mammalian myeloma cells or other fusion partners capable of replicating indefinitely in cell culture is effected by standard and well-known techniques, for example, by using polyethylene glycol (“PEG”) or other fusing agents (See Milstein and Kohler, Eur. J. Immunol. 6:511 (1976), which is hereby incorporated by reference). This immortal cell line, which is preferably murine, but may also be derived from cells of other mammalian species, including but not limited to rats and humans, is selected to be deficient in enzymes necessary for the utilization of certain nutrients, to be capable of rapid growth, and to have good fusion capability. Many such cell lines are known to those skilled in the art, and others are regularly described.
  • Procedures for raising polyclonal antibodies are also well known. Typically, such antibodies can be raised by administering the protein or polypeptide of the present invention subcutaneously to New Zealand white rabbit's which have first been bled to obtain pre-immune serum. The antigens can be injected at a total volume of 100 μl per site at six different sites. Each injected material will contain synthetic surfactant adjuvant pluronic polyols, or pulverized acrylamide gel containing the protein or polypeptide after. SDS-polyacrylamide gel electrophoresis. The rabbits are then bled two weeks after the first injection and periodically boosted with the same antigen three times every six weeks. A sample of serum is then collected 10 days after each boost. Polyclonal antibodies are then recovered from the serum by affinity chromatography using the corresponding antigen to capture the antibody. Ultimately, the rabbits are euthenized with pentobarbital 150 mg/Kg IV. This and other procedures for raising polyclonal antibodies are disclosed in E. Harlow, et. al., editors, Antibodies: A Laboratory Manual (1988), which is hereby incorporated by reference.
  • In addition to utilizing whole antibodies, the processes of the present invention encompass use of binding portions of such antibodies. Such binding portions include Fab fragments, F(ab′)2 fragments, and Fv fragments. These antibody fragments can be made by conventional procedures, such as proteolytic fragmentation procedures, as described in J. Goding, Monoclonal Antibodies: Principles and Practice, pp. 98-118 (N.Y. Academic Press 1983), which is hereby incorporated by reference.
  • Alternatively, the processes of the present invention can utilize probes or ligands found either in nature or prepared synthetically by recombinant DNA procedures or other biological or molecular procedures. Suitable probes or ligands are molecules which bind to the extracellular domains of prostate specific membrane antigens identified by the monoclonal antibodies of the present invention. Other suitable probes or ligands are molecules which bind to and are internalized with prostate specific membrane antigens. Such probes or ligands can be, for example, proteins, peptides, lectins, or nucleic acid probes.
  • It is particularly preferred to use the monoclonal antibodies identified below in Table 1.
    TABLE 1
    ATCC Designation for
    Monoclonal Antibody Name Hybridoma Cell Line
    E99 HB-12101
    J415 HB-12109
    J533 HB-12127
    J591 HB-12126

    These antibodies can be used alone or as a component in a mixture with other antibodies or other biological agents to treat cancers or image cancerous tissues (particularly the vascular endothelial cells therein) or prostate epithelial cells with varying surface antigen characteristics.
  • Regardless of whether the biological agents are used for treatment or diagnosis, they can be administered orally, parenterally, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intranasal instillation, by intracavitary or intravesical instillation, intraocularly, intraarterially, intralesionally, or by application to mucous membranes, such as, that of the nose, throat, and bronchial tubes. They may be administered alone or with pharmaceutically or physiologically acceptable carriers, excipients, or stabilizers, and can be in solid or liquid form such as, tablets, capsules, powders, solutions, suspensions, or emulsions.
  • The solid unit dosage forms can be of the conventional type. The solid form can be a capsule, such as an ordinary gelatin type containing the biological agent, such as an antibody or binding portion thereof, of the present invention and a carrier, for example, lubricants and inert fillers such as, lactose, sucrose, or cornstarch. In another embodiment, these compounds are tableted with conventional tablet bases such as lactose, sucrose, or cornstarch in combination with binders like acacia, cornstarch, or gelatin, disintegrating agents such as, cornstarch, potato starch, or alginic acid, and a lubricant like stearic acid or magnesium stearate.
  • The biological agent of the present invention may also be administered in injectable dosages by solution or suspension of these materials in a physiologically acceptable diluent with a pharmaceutical carrier. Such carriers include sterile liquids such as water and oils, with or without the addition of a surfactant and other pharmaceutically and physiologically acceptable carrier, including adjuvants, excipients or stabilizers. Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil. In general, water, saline, aqueous dextrose and related sugar solution, and glycols such as, propylene glycol or polyethylene glycol, are preferred liquid carriers, particularly for injectable solutions.
  • For use as aerosols, the biological agent of the present invention in solution or suspension may be packaged in a pressurized aerosol container together with suitable propellants, for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants. The materials of the present invention also may be administered in a non-pressurized form such as in a nebulizer or atomizer.
  • The biological agents may be utilized to detect cancerous tissues (particularly the vascular endothelial cells therein) and normal, benign hyperplastic, and cancerous prostate epithelial cells in vivo. This is achieved by labeling the biological agent, administering the labeled biological agent to a mammal, and then imaging the mammal.
  • Examples of labels useful for diagnostic imaging in accordance with the present invention are radiolabels such as 131I, 111In, 123I, 99mTc, 32P, 125I, 3H, 14C, and 188Rh, fluorescent labels such as fluorescein and rhodamine, nuclear magnetic resonance active labels, positron emitting isotopes detectable by a positron emission tomography (“PET”) scanner, chemiluminescers such as luciferin, and enzymatic markers such as peroxidase or phosphatase. Short-range radiation emitters, such as isotopes detectable by short-range detector probes, such as a transrectal probe, can also be employed. These isotopes and transrectal detector probes, when used in combination, are especially useful in detecting prostatic fossa recurrences and pelvic nodal disease. The biological agent can be labeled with such reagents using techniques known in the art. For example, see Wensel and Meares, Radioimmunoimaging and Radioimmunotherapy, Elsevier, N.Y. (1983), which is hereby incorporated by reference, for techniques relating to the radiolabeling of antibodies. See also, D. Colcher et al., “Use of Monoclonal Antibodies as Radiopharmaceuticals for the Localization of Human Carcinoma Xenografts in Athymic Mice”, Meth. Enzymol. 121: 802-816 (1986), which is hereby incorporated by reference.
  • A radiolabeled biological agent of this invention can be used for in vitro diagnostic tests. The specific activity of a tagged biological agent, such as a tagged antibody, binding portion thereof, probe, or ligand, depends upon the half-life, the isotopic purity of the radioactive label, and how the label is incorporated into the biological agent. Table 2 lists several commonly-used isotopes, their specific activities and half-lives. In immunoassay tests, the higher the specific activity, in general, the better the sensitivity.
    TABLE 2
    Specific Activity of Pure
    Isotope Isotope (Curies/mole) Half-Life
    14C 6.25 × 101 5720 years
    3H 2.01 × 104 12.5 years
    35S 1.50 × 106 87 days
    125I 2.18 × 106 60 days
    32P 3.16 × 106 14.3 days
    131I 1.62 × 107 8.1 days
  • Procedures for labeling biological agents with the radioactive isotopes listed in Table 2 are generally known in the art. Tritium labeling procedures are described in U.S. Pat. No. 4,302,438, which is hereby incorporated by reference. Iodinating, tritium labeling, and 35S labeling procedures especially adapted for murine monoclonal antibodies are described by Goding, J. W. (supra, pp 124-126) and the references cited therein, which are hereby incorporated by reference. Other procedures for iodinating biological agents, such as antibodies, binding portions thereof, probes, or ligands, are described by Hunter and Greenwood, Nature 144:945 (1962), David et al., Biochemistry 13:1014-1021 (1974) and U.S. Pat. Nos. 3,867,517 and 4,376,110, which are hereby incorporated by reference. Radiolabeling elements which are useful in imaging include 123I, 131I, 111In, and 99mTc, for example. Procedures for iodinating biological agents are described by Greenwood, F. et al., Biochem. J. 89:114-123 (1963); Marchalonis, J., Biochem. J. 113:299-305 (1969); and Morrison, M. et al., Immunochemistry, 289-297 (1971), which are hereby incorporated by reference. Procedures for 99mTc-labeling are described by Rhodes, B. et al. in Burchiel, S. et al. (eds.), Tumor Imaging: The Radioimmunochemical Detection of Cancer, New York: Masson 111-123 (1982) and the references cited therein, which are hereby incorporated by reference. Procedures suitable for 111In-labeling biological agents are described by Hnatowich, D. J. et al., J. Immul. Methods, 65:147-157 (1983), Hnatowich, D. et al., J. Applied Radiation, 35:554-557 (1984), and Buckley, R. G. et al., F.E.B.S. 166:202-204 (1984), which are hereby incorporated by reference.
  • In the case of a radiolabeled biological agent, the biological agent is administered to the patient, is localized to the tumor bearing the antigen with which the biological agent reacts, and is detected or “imaged” in vivo using known techniques such as radionuclear scanning using e.g., a gamma camera or emission tomography. See e.g., A. R. Bradwell et al., “Developments in Antibody Imaging”, Monoclonal Antibodies for Cancer Detection and Therapy, R. W. Baldwin et al., (eds.), pp. 65-85 (Academic Press 1985), which is hereby incorporated by reference. Alternatively, a positron emission transaxial tomography scanner, such as designated Pet VI located at Brookhaven National Laboratory, can be used where the radiolabel emits positrons (e.g., 11C, 18F, 15O, and 13N).
  • Fluorophore and chromophore labeled biological agents can be prepared from standard moieties known in the art. Since antibodies and other proteins absorb light having wavelengths up to about 310 nm, the fluorescent moieties should be selected to have substantial absorption at wavelengths above 310 nm and preferably above 400 nm. A variety of suitable fluorescers and chromophores are described by Stryer, Science, 162:526 (1968) and Brand, L. et al., Annual Review of Biochemistry, 41:843-868 (1972), which are hereby incorporated by reference. The biological agents can be labeled with fluorescent chromophore groups by conventional procedures such as those disclosed in U.S. Pat. Nos. 3,940,475, 4,289,747, and 4,376,110, which are hereby incorporated by reference.
  • One group of fluorescers having a number of the desirable properties described above are the xanthene dyes, which include the fluoresceins derived from 3,6-dihydroxy-9-henylxanthhydrol and resamines and rhodamines derived from 3,6-diamino-9-phenylxanthydrol and lissanime rhodamine B. The rhodamine and fluorescein derivatives of 9-o-carboxyphenylxanthhydrol have a 9-o-carboxyphenyl group. Fluorescein compounds having reactive coupling groups such as amino and isothiocyanate groups such as fluorescein isothiocyanate and fluorescamine are readily available. Another group of fluorescent compounds are the naphthylamines, having an amino group in the α or β position.
  • Biological agents can be labeled with fluorchromes or chromophores by the procedures described by Goding, J. (supra, pp 208-249). The biological agents can be labeled with an indicating group containing the NMR-active 19F atom, or a plurality of such atoms inasmuch as (i) substantially all of naturally abundant fluorine atoms are the 19F isotope and, thus, substantially all fluorine-containing compounds are NMR-active; (ii) many chemically active polyfluorinated compounds such as trifluoracetic anhydride are commercially available at relatively low cost, and (iii) many fluorinated compounds have been found medically acceptable for use in humans such as the perfluorinated polyethers utilized to carry oxygen as hemoglobin replacements. After permitting such time for incubation, a whole body NMR determination is carried out using an apparatus such as one of those described by Pykett, Scientific American, 246:78-88 (1982), which is hereby incorporated by reference, to locate and image cancerous tissues (particularly the vascular endothelial cells therein) and prostate epithelial cells.
  • In cases where it is important to distinguish between regions containing live and dead prostate epithelial cells or to distinguish between live and dead prostate epithelial cells, the antibodies of the present invention (or other biological agents of the present invention), labeled as described above, can be coadministered along with an antibody or other biological agent which recognizes only living or only dead prostate epithelial cells labeled with a label which can be distinguished from the label used to label the subject antibody. By monitoring the concentration of the two labels at various locations or times, spatial and temporal concentration variations of living and dead normal, benign hyperplastic, and cancerous prostate epithelial cells can be ascertained. In particular, this method can be carried out using the labeled antibodies of the present invention, which recognize both living and dead epithelial prostate cells, and labeled 7E11 antibodies, which recognize only dead epithelial prostate cells.
  • The biological agents can also be utilized to kill or ablate cancerous cells and normal, benign hyperplastic, and cancerous prostate epithelial cells in vivo. This involves using the biological agents by themselves or with a cytotoxic drug to which the biological agents of the present invention (i.e., biological agents recognizing normal, benign hyperplastic, and cancerous prostate epithelial cells) are bound. This involves administering the biological agents bonded to a cytotoxic drug to a mammal requiring such treatment. In the case of normal, benign-hyperplastic, and cancerous prostate epithelial cells, since the biological agents recognize prostate epithelial cells, any such cells to which the biological agents bind are destroyed. Although such administration may destroy normal prostate epithelial cells, this is not problematic, because the prostate is not required for life or survival. Although the prostate may indirectly contribute to fertility, this is not likely to be a practical consideration in patients receiving the treatment of the present invention. In the case of cancerous tissues, since the biological agents recognize vascular endothelial cells that are proximate to cancerous cells, binding of the biological agent/cytotoxic drug complex to these vascular endothelial cells destroys them, thereby cutting off the blood flow to the proximate cancerous cells and, thus, killing or ablating these cancerous cells. Alternatively, the biological agents, by virtue of their binding to vascular endothelial cells that are proximate to cancerous cells, are localized proximate to the cancerous cells. Thus, by use of suitable biological agents (including those containing substances effective to kill cells nondiscriminatingly but only over a short range), cells in cancerous tissue (including cancerous cells) can be selectively killed or ablated.
  • The biological agents of the present invention may be used to deliver a variety of cytotoxic drugs including therapeutic drugs, a compound emitting radiation, molecules of plants, fungal, or bacterial origin, biological proteins, and mixtures thereof. The cytotoxic drugs can be intracellularly acting cytotoxic drugs, such as short-range radiation emitters, including, for example, short-range, high-energy α-emitters.
  • Enzymatically active toxins and fragments thereof are exemplified by diphtheria toxin A fragment, nonbinding active fragments of diphtheria toxin, exotoxin A (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, α-sacrin, certain Aleurites fordii proteins, certain Dianthin proteins, Phytolacca americana proteins (PAP, PAPII and PAP-S), Morodica charantia inhibitor, curcin, crotin, Saponaria officinalis inhibitor, gelonin, mitogillin, restrictocin, phenomycin, and enomycin, for example. Procedures for preparing enzymatically active polypeptides of the immunotoxins are described in WO84/03508 and WO85/03508, which are hereby incorporated by reference. Certain cytotoxic moieties are derived from adriamycin, chlorambucil, daunomycin, methotrexate, neocarzinostatin, and platinum, for example.
  • Procedures for conjugating the biological agents with the cytotoxic agents have been previously described. Procedures for conjugating chlorambucil with antibodies are described by Flechner, I, European Journal of Cancer, 9:741-745 (1973); Ghose, T. et al., British Medical Journal, 3:495-499 (1972); and Szekerke, M., et al., Neoplasma, 19:211-215 (1972), which are hereby incorporated by reference. Procedures for conjugating daunomycin and adriamycin to antibodies are described by Hurwitz, E. et al., Cancer Research, 35:1175-1181 (1975) and Arnon, R. et al. Cancer Surveys, 1:429-449 (1982), which are hereby incorporated by reference. Procedures for preparing antibody-ricin conjugates are described in U.S. Pat. No. 4,414,148 and by Osawa, T., et al. Cancer Surveys, 1:373-388 (1982) and the references cited therein, which are hereby incorporated by reference. Coupling procedures as also described in EP 86309516.2, which is hereby incorporated by reference.
  • In a particularly preferred embodiment of the present invention, especially well-suited for killing or ablating normal, benign hyperplastic, and cancerous prostate epithelial cells, a first biological agent is conjugated with a prodrug which is activated only when in close proximity with a prodrug activator. The prodrug activator is conjugated with a second biological agent according to the present invention, preferably one which binds to a non-competing site on the prostate specific membrane antigen molecule. Whether two biological agents bind to competing or non-competing binding sites can be determined by conventional competitive binding assays. For example, monoclonal antibodies J591, J533, and E99 bind to competing binding sites on the prostate specific membrane antigen molecule. Monoclonal antibody J415, on the other hand, binds to a binding site which is non-competing with the site to which J591, J533, and E99 bind. Thus, for example, the first biological agent can be one of J591, J533, and E99, and the second biological agent can be J415. Alternatively, the first biological agent can be J415, and the second biological agent can be one of J591, J533, and E99. Drug-prodrug pairs suitable for use in the practice of the present invention are described in Blakely et al., “ZD2767, an Improved System for Antibody-directed Enzyme Prodrug Therapy That Results in Tumor Regressions in Colorectal Tumor Xenografts,” Cancer Research, 56:3287-3292 (1996), which is hereby incorporated by reference.
  • Alternatively, the biological agent can be coupled to high energy radiation emitters, for example, a radioisotope, such as 131I, a γ-emitter, which, when localized at the tumor site, results in a killing of several cell diameters. See, e.g., S. E. Order, “Analysis, Results, and Future Prospective of the Therapeutic Use of Radiolabeled Antibody in Cancer Therapy”, Monoclonal Antibodies for Cancer Detection and Therapy, R. W. Baldwin et al. (eds.), pp 303-316 (Academic Press 1985), which is hereby incorporated by reference. Other suitable radioisotopes include α-emitters, such as 212Bi, 213Bi, and 211At, and β-emitters, such as 186Re and 90Y. Radiotherapy is expected to be particularly effective, because prostate epithelial cells and vascular endothelial cells within cancers are relatively radiosensitive.
  • Where the biological agents are used alone to kill or ablate cancerous cells or prostate epithelial cells, such killing or ablation can be effected by initiating endogenous host immune functions, such as complement-mediated or antibody-dependent cellular cytotoxicity.
  • The biological agent of the present invention can be used and sold together with equipment, as a kit, to detect the particular label.
  • Biological agents of the present invention can be used in conjunction with other therapeutic treatment modalities. Such other treatments include surgery, radiation, cryosurgery, thermotherapy, hormone treatment, chemotherapy, vaccines, and other immunotherapies.
  • Also encompassed by the present invention is a method of killing or ablating which involves using the biological agents for prophylaxis. For example, these materials can be used to prevent or delay development or progression of prostate or other cancers.
  • Use of the therapeutic methods of the present invention to treat prostate and other cancers has a number of benefits. Since the biological agents according to the present invention only target cancerous cells (such as cells of cancerous tissues containing vascular endothelial cells) and prostate epithelial cells, other tissue is spared. As a result, treatment with such biological agents is safer, particularly for elderly patients. Treatment according to the present invention is expected to be particularly effective, because it directs high levels of biological agents, such as antibodies or binding portions thereof, probes, or ligands, to the bone marrow and lymph nodes where prostate cancer metastases and metastases of many other cancers predominate. Moreover, the methods of the present invention are particularly well-suited for treating prostate cancer, because tumor sites for prostate cancer tend to be small in size and, therefore, easily destroyed by cytotoxic agents. Treatment in accordance with the present invention can be effectively monitored with clinical parameters, such as, in the case of prostate cancer, serum prostate specific antigen and/or pathological features of a patient's cancer, including stage, Gleason score, extracapsular, seminal, vesicle or perineural invasion, positive margins, involved lymph nodes, etc. Alternatively, these parameters can be used to indicate when such treatment should be employed.
  • Because the biological agents of the present invention bind to living prostate cells, therapeutic methods for treating prostate cancer using these biological agents are much more effective than those which target lysed prostate cells. For the same reasons, diagnostic and imaging methods which determine the location of living normal, benign hyperplastic, or cancerous prostate epithelial cells (as well as vascular endothelial cells within cancers) are much improved by employing the biological agents of the present invention. In addition, the ability to differentiate between living and dead prostate cells can be advantageous, especially to monitor the effectiveness of a particular treatment regimen.
  • Hybridomas E99, J415, J533, and J591 have been deposited pursuant to, and in satisfaction of, the requirements of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure with the American Type Culture Collection (“A.T.C.C.”) at 12301 Parklawn Drive, Rockville, Md. 20852. Hybridoma E99 was deposited on May 2, 1996, and received A.T.C.C. Designation Number HB-12101. Hybridoma J415 was deposited on May 30, 1996, and received A.T.C.C. Designation Number HB-12109. Hybridomas J533 and J591 were deposited on Jun. 6, 1996, and received A.T.C.C. Designation Numbers HB-12127 and HB-12126, respectively.
  • The present invention is further illustrated by the following examples.
  • EXAMPLES Example 1 Human Tissues
  • Fresh specimens of benign and malignant tissues were obtained from the Department of Pathology of New York Hospital Cornell University Medical Center (“NYH-CUMC”),
  • Example 2 Tissue Culture
  • Cultured cell lines of human cancers were obtained from the Laboratory of Urological Oncology of NYH-CUMC. The prostate cancer cell lines PC-3 (Mickey, D. D., et al., “Characterization Of A Human Prostate Adenocarcinoma Cell Line (DU145) As A Monolayer Culture And As A Solid Tumor In Athymic Mice,” Prog. Clin. Biol. Res., 37:67-84 (1980), which is hereby incorporated by reference), DU-145 (Mickey, D. D., et al., “Characterization Of A Human Prostate Adenocarcinoma Cell Line (DU145) As A Monolayer Culture And As A Solid Tumor In Athymic Mice,” Prog. Clin. Biol. Res., 37:67-84 (1980), which is hereby incorporated by reference), and LNCaP (Horoszewicz, J. S., et al., “LNCaP Model Of Human Prostatic Carcinoma,” Cancer Res., 43:1809-1818 (1983), which is hereby incorporated by reference) were obtained from the American Type Culture Collection (Rockville, Md.). Hybridomas were initially cloned in RPMI-1640 medium supplemented with 10% FCS, 0.1 mM nonessential amino acids, 2 mM L-glutamine, 100 units/ml of penicillin, 100 ug/ml of streptomycin and HAT medium (GIBCO, Grand Island, N.Y.). Subclones were cultured in the same medium without aminopterin.
  • Example 3 Preparation of Mouse Monoclonal Antibodies
  • Female BALB/c mice were immunized intraperitoneally with LNCaP (6×106 cells) three times at 2 week intervals. A final intraperitoneal booster immunization was administered with fresh prostate epithelial cells which had been grown in vitro. Three days later, spleen cells were fused with SP-2 mouse myeloma cells utilizing standard techniques (Ueda, R., et al., “Cell Surface Antigens Of Human Renal Cancer Defined By Mouse Monoclonal Antibodies: Identification Of Tissue-Specific Kidney Glycoproteins,” Proc. Natl. Acad. Sci. USA, 78:5122-5126 (1981), which is hereby incorporated by reference). Supernatants of the resulting clones were screened by rosette and complement cytotoxicity assays against viable LNCaP. Clones which were positive by these assays were screened by immunochemistry vs normal kidney, colon, and prostate. Clones which were LNCap+/NmlKid/colon/prostate+ were selected and subcloned 3 times by limiting dilution. The immunoglobulin class of cultured supernatant from each clone was determined by immunodiffusion using specified rabbit antisera (Calbiochem, San Diego, Calif.). mAbs were purified using the MAPS-II kit (Bio-Rad, Richmond, Calif.).
  • Example 4 Biotinylation of mAbs
  • Purified mAbs were dialyzed in 0.1 M NaHCO3 for 2 hours. One ml of mAb at 1 mg/ml was mixed with 0.1 ml of biotinamidocaproate N-hydroxysuccinamide ester (Sigma) in dimethylsulfoxide (1 mg/ml) and stirred for 4 hours at room temperature. Unbound biotin was removed by dialysis against phosphate buffered saline (“PBS”).
  • Example 5 Immunohistochemical Staining of Prostate Tissues
  • Cryostat sections of prostate tissues were placed inside rings of Falcon 3034 plate covers (Becton-Dickenson, Lincoln Park, N.J.) previously coated with 0.45% gelatin solution as described in Marusich, M. F., “A Rapid Method For Processing Very Large Numbers Of Tissue Sections For Immunohistochemical Hybridoma Screening,” J. Immunol. Methods, 111:143-145 (1988), which is hereby incorporated by reference. Plates were stored at −80° C. Cryostat sections were fixed with 2% paraformaldehyde in PBS for 10 min at room temperature, and, after washing with PBS, endogenous peroxidase activity was blocked by treatment with 0.3% hydrogen peroxide in PBS for 10 min at room temperature. After sections were incubated with 2% BSA in PBS for 20 min, mAbs were added for 60 min at room temperature. Slides were extensively washed with PBS and incubated with peroxidase-conjugated rabbit anti-mouse Ig (DAKO Corp., Santa Barbara, Calif.) diluted 1:100 in. 10% normal human serum in PBS for 60 min at room temperature. After a diaminobenzidine reaction, sections were counterstained with hematoxylin.
  • Example 6 Serological Analysis
  • The anti-mouse immunoglobulin mixed hemadsorption assay was performed as described in Ueda, R., et al., “Cell Surface Antigens Of Human Renal Cancer Defined By Mouse Monoclonal Antibodies: Identification Of Tissue-Specific Kidney Glycoproteins,” Proc. Natl. Acad. Sci. USA, 78:5122-5126 (1981), which is hereby incorporated by reference. To prepare the indicator cells, anti-mouse Ig (DAKO Corp.) was conjugated to type O human RBC using 0.01% chromium chloride. Serological assays were performed on cells previously plated in Terasaki plates (Nunc, Denmark). Antibodies were incubated with target cells at room temperature for 1 hour. Target cells were then washed, and indicator cells added for 1 hour.
  • Example 7 Immunoprecipitation
  • LNCaP cells (2×107) were biotinylated with biotin-NHSS (at final concentration of 5 mM) for 30 minutes on ice. After washing, the biotinylated cells were resuspended in 1 ml lysis buffer (20 mM Tris/HCl pH 8.0, 1 mM EDTA, 1 mM PMSF, 1% triton X-100) for 30 min on ice. The suspension was centrifuged at 1500 g×100 min at 4° C., and the supernatant was centrifuged at 12,000 rpm×15 min at 4° C. The resulting lysate was preabsorbed with rabbit or goat anti-mouse IgG-coated pansorbin for 1 hour at 4° C. The pre-absorbed lysate was incubated with the mAb overnight at 4° C. Rabbit or goat anti-mouse Ig-coated agarose beads were added for 2 hours at 4° C. and then washed. The beads were resuspended in Tris-base/NaCl, added to sample buffer with 2-mercaptoethanol, and boiled for 5 min. After centrifuging, the supernatant was run on an SDS-PAGE 12% gel. The gel was transferred to a nitrocellulose membrane which was blocked and stained with straptavidin-peroxidase. The membrane was developed with diaminobenzidine (“DAB”).
  • Sequential immunoprecipitation was similar except that the lysate was initially pre-cleared with one mAb overnight at 4° C. A second mAb was then used to immunoprecipitate the pre-cleared lysate.
  • Approximately 2000 clones were screened, of which four clones were selected as described in Example 3, above. After subcloning, supernatants from the 4 hybridomas, E99, J415, J533, and J591, were assayed by immunofluorescence against viable (i.e. unfixed) LNCaP, immunoprecipitation, and sequential immunoprecipitation to confirm reactivity to PSMA.
  • The immunofluorescence study using the LNCaP target cell (described originally in Horoszewicz, which is hereby incorporated by reference, to make the 7E11 antibody and the prototype cell line for expression for PSMA) shows that E99 antibody binds to and renders viable LNCaP cells immunofluorescent. This is in contrast to the 7E11 antibody, which, as noted originally in Horoszewicz, which is hereby incorporated by reference, gives only poor or no binding to viable LNCaP cells but exhibits strong binding once the cells are fixed (killed).
  • The reactivities of the four mAbs with normal human tissues were examined immunohistochemically; these results are presented in Table 3.
    TABLE 3
    Reactivity of mAbs with human normal tissues
    by indirect immunoperosidase staining
    E99 J415 J533 J591
    Tissues 3) 1) 1) 1)
    Prostate*
    Kidney
    Glomerulus
    Prox. Tubule
    Ureter
    Bladder
    Testis
    Uterus
    Esophagus
    Small Intestine
    Stomach
    Colon
    Spleen
    Thyroid
    Lung
    Pancreas
    Liver
    BPH 0-3+ 0-3+ 0-4+ 0-4+
    Prostate Cancer 0-3+ 0-3+ 0-4+ 0-4+
    LNCaP (scid) 3+ 3+ 4+ 4+
    LuCaP (scid) 0-2+ 0-2+ 0-3+ 0-3+

    ● - positive; ▪ - weak, heterogeneous; ◯ - negative

    The above sequential immunoprecipitaion study showed that 7E11, E99, J415, J533, and J591 bind to the same molecule, i.e. PSMA.
  • Example 8 Western Blot Analysis
  • To confirm that antibodies E99, J415, J533, and J591 precipitate an identical band to the 7E11 antibody (i.e., PSMA), Western Blot analyses were performed. Seminal plasma (400 μg/lane) or LNCaP lysate were loaded into lanes of 12% SDS-PAGE gels. After electrophoresis, the gels are transferred to nitrocellulose membranes. The membranes were blocked with 5% dry milk/Tris-buffered saline-tween 20 (“TBST”) for 60 min at room temperature. After washing, the membranes were incubated with primary mAb for 60 min at room temperature. After repeat washing, the membranes were incubated with sheep anti-mouse-Ig-peroxidase 1/5000 in 5% dry milk/TBST for 60 min at room temperature. After repeat washing, the membranes were developed using a chemiluminescent tag designated “ECL” (Amersham Life Sciences, International, Arlington Heights, Ill.) according to the manufacturer's directions. The results of the Western Blot experiment are presented in Table 4.
    TABLE 4
    Western blot data
    Sample
    7E11 E99 J415 J533 J591
    Prostatic
    100 KD 100 KD 100 KD 100 KD 100 KD
    (seminal) band band band band band
    fluid
    LNCaP
    100 KD & 100 KD & 100 KD & 100 KD & 100 KD &
    cell lysate 200 KD 200 KD 200 KD 200 KD 200 KD
    bands bands bands bands bands
  • Example 9 mAb Reactivity to External Domain of PSMA
  • To confirm cell surface (external) expression of the detected PSMA, fresh, viable LNCaP cells were tested, without fixation, in vitro, by immunofluorescence. LNCaP cells were washed and incubated with mAb for 1 hour at room temperature and then with a rabbit anti-mouse Ig-fluorescein (DAKO Corp., Santa Barbara, Calif.). Wells were read with a fluorescent microscope. Negative control consisted of an isotype-matched irrelevant mAb, while an anti-class I MHC mAb served as a positive control.
  • Immunofluorescence and rosette assay results are presented in Table 5.
    TABLE 5
    Comparison of 7E11 with new mAbs
    LNCaP
    viable cells 7E11 E99 J415 J533 J591
    Immunoflu- neg 3+ 3+ 4+ 4+
    orescence
    Rosette neg + + + +
    assay
    LNCaP-fixed +++ ++++ +++ ++ +++
  • Example 10 Competition Studies
  • A competition study was carried out to determine whether J591, J533, E99, and J415 detected the same or different antigenic sites (epitopes) of the prostate specific membrane antigen molecule using the following procedure.
  • Plates were coated with LNCaP cell line lysate as a source of prostate specific membrane antigen and washed to remove unbound material. “Cold” (unlabeled) monoclonal antibody was incubated on the plate for 1 hour at room temperature to allow binding to its antigenic site. Subsequently, a second monoclonal antibody, labeled either with biotin or 125I, was added for an additional hour. Plates were washed to remove unbound material. The amount of the second monoclonal antibody bound to the prostate specific membrane antigen-coated plate was determined either by avidin-alkaline phosphatase in an enzyme-linked immunoassay (in the case of biotin-labeled second monoclonal antibody) or by physically counting the well in a gamma counter (in the case of 125I-labeled second monoclonal antibody). Controls consisted of using the same monoclonal antibody both cold and labeled to define “100% competition” or using monoclonal antibody to a totally different molecule (e.g., monoclonal antibody I-56, which detects inhibin, a prostate related protein different from prostate specific membrane antigen) to define “0% competition”.
  • The results indicated that J591, J533, and E99 each interfere, compete, or block binding of one another but do not block binding of J415 and vice versa. 7E11/CYT356, known to bind PSMA at a different (intracellular) site, did not block any of J591, J533, E99, or J415.
  • Having pairs of monoclonal antibodies which bind to non-competing sites permits development of antibody sandwich assays for detecting soluble antigens, such as solubilized prostate specific membrane antigen or fragment thereof, in, for example, body fluids. For example, the antigen (e.g., prostate specific membrane antigen or a fragment thereof) could be “captured” from body fluid with J591 and, in another step, detected by labeled J415.
  • In another setting, e.g. treatment, one could increase antibody binding by using a combination of non-competing monoclonal antibodies. For example, assuming the non-competing sites are each represented once on the prostate specific membrane antigen molecule, adding a combination of J591 plus J415 would bind twice as many monoclonal antibody molecules as either monoclonal antibody alone. Binding two non-competing antigenic binding sites also can result in greater antigen cross-linking and, perhaps, increased internalization. Furthermore, since the two detected sites are physically located on the same prostate specific membrane antigen molecule, the binding of two monoclonal antibody molecules to that single prostate specific membrane antigen molecule puts the two monoclonal antibody molecules in close proximity to each other, a setting which provides optimal drug-prodrug interaction. For example, monoclonal antibody J591 can be conjugated with an inactive pro-drug and J415 can be conjugated with a pro-drug activator. Since prodrug and activator would be bound in close proximity only at the site of prostate specific membrane antigen-expressing cells (e.g., prostate cancer cells), prodrug activation to the active form would occur only at those sites.
  • Example 11 Microscopy
  • Confocal microscopy and immuno-electron microscopy demonstrated that E99, J591, J533, and J415 are bound to the cell membrane at clathrin-coated pits and then rapidly internalize into endosomes (cytoplasmic vesicles). FIGS. 1-4 are immuno-electron micrographs which follow the interaction of gold-labeled monoclonal antibody J591 with the cell surface as a function of time. In these figures, the location of the monoclonal antibody is indicated by the black dots.
  • Viable LNCaP cells were incubated with J591 for one hour at 4° C. The cells were washed and then held at 37° C. for 0, 5, 10, or 15 minutes, after which time they were fixed and processed for immuno-electron microscopy. FIG. 1 shows the cell prior to 37° C. incubation. J591 can be seen bound to the cell along the external aspect of the cell membrane. In this Figure, “M” denotes the cell's mitochondria, and “N” denotes its nucleus. FIG. 2 shows the cell after incubation at 37° C. for 5 minutes. The arrow indicates formation of a clathrin-coated pit. In FIG. 3, which shows the cell after a 10 minute 37° C. incubation, pinching off or endocytosis of the clathrin-coated pit can be seen, as indicated by the arrow. FIG. 4 shows that, after incubation at 37° C. for 15 minutes, monoclonal antibody J591 is contained in endocytic vesicles within the cell, as indicated by the arrows. As can be seen in FIG. 5, after incubation at 37° C. for 15 minutes, monoclonal antibody J591 is also contained within endosomes, as indicated by the arrows.
  • Example 12 Sequencing of the Variable Region of Monoclonal Antibody J591
  • Total RNA was prepared from 107 murine hybridoma J591 cells. A sample of the conditioned medium from these cells was tested for binding to the specific antigen for J591 on prostate cells. The conditioned medium was positive by both ELISA and Western Blot for binding to the antigen.
  • VH and VK cDNA were prepared using reverse transcriptase and mouse κ constant region and mouse IgG constant region primers. The first strand cDNAs were amplified by PCR using a variety of mouse signal sequence primers (6 for VH and 7 for VK). The amplified DNAs were gel-purified and cloned into the vector pT7Blue.
  • The VH and VK clones obtained were screened for correct inserts by PCR, and the DNA sequence of selected clones was determined by the dideoxy chain termination method.
  • Excluding the primer region (as the sequence of this depended on the sequence of the primer that was used), all the VH clones obtained gave identical sequence. This sequence was obtained from clones produced with three different 5′ primers. One clone had one base pair change within the signal sequence, and one clone contained an aberrant PCR product. Using the sequencing strategy shown in FIG. 6, the nucleotide sequence for the heavy chain was obtained. It is designated SEQ. ID. No. 1 and is presented in FIG. 7, along with the nucleotide sequence of the corresponding reverse, non-coding strand (designated SEQ. ID. No. 2). These sequences include part of the signal sequence and part of the constant region of the antibody. The corresponding deduced amino acid sequences of J591 VH, designated SEQ. ID. No. 3, SEQ. ID. No. 4, and SEQ. ID. No. 5, are also shown in FIG. 7. The coding strand of the J591 heavy chain's variable region (exclusive of signal sequence and constant region components) has the following nucleotide sequence (designated SEQ. ID. No. 6):
    GAGGTCCAGCTGCAACAGTCTGGACCTGAACTGGTGAAGCCTGGGACTTC
    AGTGAGGATATCCTGCAAGACTTCTGGATACACATTCACTGAATATACCA
    TACACTGGGTGAAGCAGAGCCATGGAAAGAGCCTTGAGTGGATTGGAAAC
    ATCAATCCTAACAATGGTGGTACCACCTACAATCAGAAGTTCGAGGACAA
    GGCCACATTGACTGTAGACAAGTCCTCCAGTACAGCCTACATGGAGCTCC
    GCAGCCTAACATCTGAGGATTCTGCAGTCTATTATTGTGCAGCTGGTTGG
    AACTTTGACTACTGGGGCCAAGGCACCACTCTCACAGTCTCCTCA
  • The reverse, non-coding strand of the J591 heavy chain's variable region (exclusive of signal sequence and constant region components) has the following nucleotide sequence (designated SEQ. ID. No. 7):
    TGAGGAGACTGTGAGAGTGGTGCCTTGGCCCCAGTAGTCAAAGTTCCAAC
    CAGCTGCACAATAATAGACTGCAGAATCCTCAGATGTTAGGCTGCGGAGC
    TCCATGTAGGCTGTACTGGAGGACTTGTCTACAGTCAATGTGGCCTTGTC
    CTCGAACTTCTGATTGTAGGTGGTACCACCATTGTTAGGATTGATGTTTC
    CAATCCACTCAAGGCTCTTTCCATGGCTCTGCTTCACCCAGTGTATGGTA
    TATTCAGTGAATGTGTATCCAGAAGTCTTGCAGGATATCCTCACTGAAGT
    CCCAGGCTTCACCAGTTCAGGTCCAGACTGTTGCAGCTGGACCTC
  • The protein sequence corresponding to the J591 heavy chain's variable region (exclusive of signal sequence and constant region components) has the following nucleotide sequence (designated SEQ. ID. No. 8):
    EVQLQQSGPELVKPGTSVRISCKTSGYTFTEYTIHWVKQSHGKSLEWIGN
    INPNNGGTTYNQKFEDKATLTVDKSSSTAYMELRSLTSEDSAVYYCAAGW
    NFDYWGQGTTLTVSS
  • The J591 VH is in Mouse Heavy Chains Subgroup IIA (Kabat et al., Sequences of Proteins of Immunological Interest, U.S. Department of Health and Human Services (1991) (“Kabat”), which is hereby incorporated by reference). The sequence of J591 VH is compared to the consensus sequence for this subgroup in FIG. 8.
  • In contrast to the VH, more than one VK sequence was obtained. Out of the 15 VK clones examined, four gave the sequence of an aberrant mouse Igκ from the fusion partner (Carol et al., Molecular Immunology, 25:991-995 (1988), which is hereby incorporated by reference). These clones originated from two specific 5′ primers. No further work was done with these clones of the remaining clones, ten gave identical nucleotide sequences, and one clone, VK17, gave an alternative VK sequence. The ten identical clones originated from three 5′ primers (different from the two that gave the aberrant sequence), one of which also produced VK17. The sequencing strategy that was employed is shown in FIG. 9.
  • The nucleic acid sequence of J591 VK corresponding to the ten identical clones (designated SEQ. ID. No. 9) is presented in FIG. 10, along with the nucleic acid sequence of the corresponding reverse, non-coding strand (designated SEQ. ID. No. 10) and the deduced amino acid sequences, which are designated SEQ. ID. No. 11, SEQ. ID. No. 12, and SEQ. ID. No. 13. These sequences include part of the signal sequence and part of the constant region of the antibody. The coding strand of the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to the ten identical clones has the following nucleotide sequence (designated SEQ. ID. No. 14):
    AACATTGTAATGACCCAATCTCCCAAATCCATGTCCATGTCAGTAGGAGA
    GAGGGTCACCTTGACCTGCAAGGCCAGTGAGAATGTGGTTACTTATGTTT
    CCTGGTATCAACAGAAACCAGAGCAGTCTCCTAAACTGCTGATATACGGG
    GCATCCAACCGGTACACTGGGGTCCCCGATCGCTTCACAGGCAGTGGATC
    TGCAACAGATTTCACTCTGACCATCAGCAGTGTGCAGGCTGAAGACCTTG
    CAGATTATCACTGTGGACAGGGTTACAGCTATCCGTACACGTTCGGAGGG
    GGGACCAAGCTGGAAATAAAA
  • The reverse, non-coding strand of the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to the ten identical clones has the following nucleotide sequence (designated SEQ. ID. No. 15): .
    TTTTATTTCCAGCTTGGTCCCCCCTCCGAACGTGTACGGATAGCTGTAAC
    CCTGTCCACAGTGATAATCTGCAAGGTCTTCAGCCTGCACACTGCTGATG
    GTCAGAGTGAAATCTGTTGCAGATCCACTGCCTGTGAAGCGATCGGGGAC
    CCCAGTGTACCGGTTGGATGCCCCGTATATCAGCAGTTTAGGAGACTGCT
    CTGGTTTCTGTTGATACCAGGAAACATAAGTAACCACATTCTCACTGGCC
    TTGCAGGTCAAGGTGACCCTCTCTCCTACTGACATGGACATGGATTTGGG
    AGATTGGGTCATTACAATGTT
  • The protein sequence corresponding to the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to the ten identical clones has the following nucleotide sequence (designated SEQ. ID. No. 16):
    NIVMTQSPKSMSMSVGERVTLTCKASENVVTYVSWYQQKPEQSPKLLIYG
    ASNRYTGVPDRFTGSGSATDFTLTISSVQAEDLADYHCGQGYSYPYTFGG
    GTKLEIK
  • The coding strand of the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to clone VK17 has the following nucleotide sequence (designated SEQ. ID. No. 17):
    GACATTGTGATGACCCAGTCTCACAAATTCATGTCCACATCAGTAGGAGA
    CAGGGTCAGCATCATCTGTAAGGCCAGTCAAGATGTGGGTACTGCTGTAG
    ACTGGTATCAACAGAAACCAGGACAATCTCCTAAACTACTGATTTATTGG
    GCATCCACTCGGCACACTGGAGTCCCTGATCGCTTCACAGGCAGTGGATC
    TGGGACAGACTTCACTCTCACCATTACTAATGTTCAGTCTGAAGACTTGG
    CAGATTATTTCTGTCAGCAATATAACAGCTATCCTCTCACGTTCGGTGCT
    GGGACCATGCTGGACCTGAAA
  • The reverse, non-coding strand of the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to clone VK17 has the following nucleotide sequence (designated SEQ. ID. No. 18):
    TTTCAGGTCCAGCATGGTCCCAGCACCGAACGTGAGAGGATAGCTGTTAT
    ATTGCTGACAGAAATAATCTGCCAAGTCTTCAGACTGAACATTAGTAATG
    GTGAGAGTGAAGTCTGTCCCAGATCCACTGCCTGTGAAGCGATCAGGGAC
    TCCAGTGTGCCGAGTGGATGCCCAATAAATCAGTAGTTTAGGAGATTGTC
    CTGGTTTCTGTTGATACCAGTCTACAGCAGTACCCACATCTTGACTGGCC
    TTACAGATGATGCTGACCCTGTCTCCTACTGATGTGGACATGAATTTGTG
    AGACTGGGTCATCACAATGTC
  • The protein sequence corresponding to the J591 light (kappa) chain's variable region (exclusive of signal sequence and constant region components) corresponding to clone VK17 has the following nucleotide sequence (designated SEQ. ID. No. 19):
    DIVMTQSHKFMSTSVGDRVSIICKASQDVGTAVDWYQQKPGQSPKLLIYW
    ASTRHTGVPDRFTGSGSGTDFTLTITNVQSEDLADYFCQQYNSYPLTFGA
    GTMLDLK
  • J591 VK is in the Mouse Kappa Chains Subgroup V (Kabat, which is hereby incorporated by reference). The sequence of J591 VK corresponding to the ten identical clones is compared to the consensus sequence for the subgroup in FIG. 11.
  • Preferred J591's are those having heavy chain variable region DNA coding strand sequences corresponding to SEQ. ID. No. 6 and non-coding strand (reverse) sequences corresponding to SEQ. ID. No. 7. The heavy chain variable region of J591 preferably has an amino acid sequence corresponding to SEQ. ID. No. 8. The light chain variable region of J591 preferably has a DNA coding strand sequence corresponding to SEQ. ID. No. 17, a DNA non-coding strand (reverse) sequence corresponding to SEQ. ID. No. 18, and a amino acid sequence corresponding to SEQ. ID. No. 19.
  • Example 13 Immunohistochemical Staining of Normal and Cancer Tissues
  • Cancer tissues from 23 carcinomas were pre-cooled in liquid nitrogen, snap-frozen in OCT compound (Miles, Elkhart, Ind.) on dry ice, and stored at −80° C. Cryostat tissue sections (5 μm) were fixed in cold acetone (4° C.) for 10 minutes. mAbs (5 μg/ml or hybridoma supernatants) were incubated for 1 hour at room temperature. Antibody binding was detected using rabbit anti-mouse Ig-peroxidase (Dako, Carpinteria, Calif.) as a secondary antibody and DAB (Sigma, St. Louis, Mo.) as chromogen. Isotype-matched irrelevant antibody was used as negative control.
  • mAbs J591, J533, J415, and E99 reacted strongly with vascular endothelia in all 23 carcinomas studied, including 9/9 renal, 5/5 urothelial, 6/6 colon, 1/1 lung, and 1/1 breast carcinomas, and 1/1 metastatic adenocarcinoma to the liver. FIGS. 2A-2F, respectively, show the immunohistochemical reactivity of mAb J591 to neovasculature of renal, urothelial, colon, lung, and breast carcinomas, and metastatic adenocarcinoma to the liver.
  • Although the invention has been described in detail for the purpose of illustration, it is understood that such detail is solely for that purpose and variations can be made by those skilled in the art without departing from the spirit and scope of the invention which is defined by the following claims.

Claims (37)

1. A method of ablating or killing cancerous cells comprising:
providing a biological agent which, when contacted with an extracellular domain of prostate specific membrane antigen, binds to the extracellular domain of prostate specific membrane antigen and
contacting vascular endothelial cells proximate to the cancerous cells with the biological agent under conditions effective to permit both binding of the biological agent to the vascular endothelial cells proximate to the cancerous cells and ablating or killing of the cancerous cells.
2. A method according to claim 1, wherein the biological agent kills or ablates the vascular endothelial cells proximate to the cancerous cells, thereby killing or ablating the cancerous cells by reducing blood flow thereto.
3. A method according to claim 1, wherein the cancerous cells are renal cancerous cells, urothelial cancerous cells, colon cancerous cells, rectal cancerous cells, lung cancerous cells, breast cancerous cells, or cancerous cells of metastatic adenocarcinoma to the liver.
4. A method according to claim 1, wherein the biological agent is an antibody or binding portion thereof, probe, or ligand.
5. A method according to claim 1, wherein the biological agent, when contacted with an extracellular domain of prostate specific membrane antigen, is internalized with the prostate specific membrane antigen.
6. A method according to claim 1, wherein said contacting is carried out in a living mammal and comprises:
administering the biological agent to the mammal under conditions effective to permit both binding of the biological agent to vascular endothelial cells proximate to the cancerous cells and killing of the cancerous cells.
7. A method according to claim 6, wherein said administering is carried out orally, parenterally, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intranasal instillation, by intracavitary or intravesical instillation, intraocularly, intraarterially, intralesionally, or by application to mucous membranes.
8. A method according to claim 4, wherein an antibody is used in carrying out said method, the antibody being selected from the group consisting of a monoclonal antibody and a polyclonal antibody.
9. A method according to claim 8, wherein the antibody is selected from the group consisting of an E99, a J415, a J533, and a J591 monoclonal antibody.
10. A method according to claim 8, wherein the antibody is a monoclonal antibody produced by a hybridoma cell line having an ATCC Accession Number selected from the group consisting of HB-12101, HB-12109, HB-12127, and HB-12126.
11. A method according to claim 4, wherein a binding portion of an antibody is used in carrying out said method, the binding portion being selected from the group consisting of an Fab fragment, an F(ab′)2 fragment, and an Fv fragment.
12. A method according to claim 4, wherein the probe or ligand is used in carrying out said method.
13. A method according to claim 1, wherein the biological agent is bound to a substance effective to kill or ablate the cancerous cells upon binding of the biological agent to vascular endothelial cells proximate to the cancerous cells.
14. A method according to claim 13, wherein the substance effective to kill or ablate the cancerous cells is a cytotoxic drug.
15. A method according to claim 14, wherein the cytotoxic drug is selected from the group consisting of therapeutic drug, a compound emitting radiation, molecules of plant, fungal, or bacterial origin, biological proteins, and mixtures thereof.
16. A method according to claim 4, wherein the antibody is effective to initiate an endogenous host immune function.
17. A method according to claim 16, wherein the endogenous host immune function is complement-mediated cellular cytoxicity.
18. A method according to claim 16, wherein the endogenous host immune function is antibody-dependent cellular cytoxicity.
19. A method according to claim 1, wherein the biological agent is in a composition further comprising a physiologically acceptable carrier, excipient, or stabilizer.
20. A method according to claim 1, wherein the biological agent is in a composition further comprising a pharmaceutically acceptable carrier, excipient, or stabilizer.
21. A method of detecting cancerous tissue in a biological sample comprising:
providing an biological agent which, when contacted with an extracellular domain of prostate specific membrane antigen, binds to the extracellular domain of prostate specific membrane antigen, wherein the biological agent is bound to a label effective to permit detection of vascular endothelial cells proximate to or within the cancerous tissue upon binding of the biological agent to the vascular endothelial cells proximate to or within the cancerous tissue;
contacting the biological sample with the biological agent having a label under conditions effective to permit binding of the biological agent to the vascular endothelial cells proximate to or within the cancerous tissue in the biological sample; and
detecting a presence of any cancerous tissue in the biological sample by detecting the label.
22. A method according to claim 21, wherein the cancerous tissue is renal cancerous tissue, urothelial cancerous tissue, colon cancerous tissue, rectal cancerous tissue, lung cancerous tissue, breast cancerous tissue, or cancerous tissue of metastatic adenocarcinoma to the liver.
23. A method according to claim 21, wherein the biological agent is an antibody or binding portion thereof, probe, or ligand.
24. A method according to claim 21, wherein the biological agent, when contacted with an extracellular domain of prostate specific membrane antigen, is internalized with the prostate specific membrane antigen.
25. A method according to claim 21, wherein said contacting is carried out in a living mammal and comprises:
administering the biological agent to the mammal under conditions effective to permit binding of the biological agent to the vascular endothelial cells proximate to or within the cancerous tissue in the biological sample.
26. A method according to claim 25, wherein the label is a short-range radiation emitter.
27. A method according to claim 25, wherein said administering is carried out orally, parenterally, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intraversal instillation, by intracavitary or intravesical instillation, intraocularly, intraarterially, intralesionally, or by application to mucous membranes.
28. A method according to claim 23, wherein an antibody is used in carrying out said method, said antibody being selected from the group consisting of a monoclonal antibody and a polyclonal antibody.
29. A method according to claim 28, wherein the antibody is selected from the group consisting of an E99, a J415, a J533, and a J591 monoclonal antibody.
30. A method according to claim 28, wherein the antibody is a monoclonal antibody produced by a hybridoma cell line having an ATCC Accession Number selected from the group consisting of HB-12101, HB-12109, HB-12127, and HB-12126.
31. A method according to claim 23, wherein a binding portion of an antibody is used in carrying out said method, the binding portion being selected from the group consisting of an Fab fragment, an F(ab′)2 fragment, and an Fv fragment.
32. A method according to claim 23, wherein a probe or ligand is used in carrying out said method.
33. A method according to claim 21, wherein the label is selected from the group consisting of a fluorescent label, a radioactive label, a nuclear magnetic resonance active label, a luminescent label, and a chromophore label.
34. A method according to claim 21, wherein the biological agent is in a composition further comprising a physiologically acceptable carrier, excipient, or stabilizer.
35. A method according to claim 21, wherein the biological agent is in a composition further comprising a pharmaceutically acceptable carrier, excipient, or stabilizer.
36. A method according to claim 21, wherein said contacting is carried out in a sample of serum or urine.
37. A method according to claim 21, wherein said contacting is carried out in a tissue biopsy sample.
US11/481,344 1996-05-06 2006-07-05 Treatment and diagnosis of cancer Abandoned US20060275212A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US11/481,344 US20060275212A1 (en) 1996-05-06 2006-07-05 Treatment and diagnosis of cancer
US11/939,422 US8951737B2 (en) 1996-05-06 2007-11-13 Treatment and diagnosis of cancer
US14/617,788 US20150316553A1 (en) 1996-05-06 2015-02-09 Treatment and diagnosis of cancer

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US1697696P 1996-05-06 1996-05-06
US2212596P 1996-07-18 1996-07-18
US08/838,682 US6107090A (en) 1996-05-06 1997-04-09 Treatment and diagnosis of prostate cancer with antibodies to extracellur PSMA domains
US08/895,914 US6136311A (en) 1996-05-06 1997-07-17 Treatment and diagnosis of cancer
US09/357,708 US6770450B1 (en) 1996-05-06 1999-07-20 Treatment and diagnosis of cancer
US09/929,546 US7163680B2 (en) 1996-05-06 2001-08-13 Treatment and diagnosis of cancer
US11/481,344 US20060275212A1 (en) 1996-05-06 2006-07-05 Treatment and diagnosis of cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/929,546 Continuation US7163680B2 (en) 1996-05-06 2001-08-13 Treatment and diagnosis of cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/939,422 Continuation US8951737B2 (en) 1996-05-06 2007-11-13 Treatment and diagnosis of cancer

Publications (1)

Publication Number Publication Date
US20060275212A1 true US20060275212A1 (en) 2006-12-07

Family

ID=29424685

Family Applications (7)

Application Number Title Priority Date Filing Date
US08/895,914 Expired - Lifetime US6136311A (en) 1996-05-06 1997-07-17 Treatment and diagnosis of cancer
US09/357,707 Expired - Lifetime US6649163B1 (en) 1996-05-06 1999-07-20 Treatment and diagnosis of cancer
US09/357,708 Expired - Lifetime US6770450B1 (en) 1996-05-06 1999-07-20 Treatment and diagnosis of cancer
US09/929,546 Expired - Fee Related US7163680B2 (en) 1996-05-06 2001-08-13 Treatment and diagnosis of cancer
US11/481,344 Abandoned US20060275212A1 (en) 1996-05-06 2006-07-05 Treatment and diagnosis of cancer
US11/939,422 Expired - Fee Related US8951737B2 (en) 1996-05-06 2007-11-13 Treatment and diagnosis of cancer
US14/617,788 Abandoned US20150316553A1 (en) 1996-05-06 2015-02-09 Treatment and diagnosis of cancer

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US08/895,914 Expired - Lifetime US6136311A (en) 1996-05-06 1997-07-17 Treatment and diagnosis of cancer
US09/357,707 Expired - Lifetime US6649163B1 (en) 1996-05-06 1999-07-20 Treatment and diagnosis of cancer
US09/357,708 Expired - Lifetime US6770450B1 (en) 1996-05-06 1999-07-20 Treatment and diagnosis of cancer
US09/929,546 Expired - Fee Related US7163680B2 (en) 1996-05-06 2001-08-13 Treatment and diagnosis of cancer

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/939,422 Expired - Fee Related US8951737B2 (en) 1996-05-06 2007-11-13 Treatment and diagnosis of cancer
US14/617,788 Abandoned US20150316553A1 (en) 1996-05-06 2015-02-09 Treatment and diagnosis of cancer

Country Status (1)

Country Link
US (7) US6136311A (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040033229A1 (en) * 2001-10-23 2004-02-19 Maddon Paul J. PSMA antibodies and protein multimers
US20090028872A1 (en) * 2005-09-26 2009-01-29 Jonathan Alexander Terret Human monoclonal antibodies to cd70
US20090060908A1 (en) * 2005-02-18 2009-03-05 Medarex, Inc. Monoclonal Antibodies Against Prostate Specific Membrane Antigen (PSMA) Lacking in Fucosyl Residues
US20100150950A1 (en) * 2006-12-14 2010-06-17 Medarex, Inc. Human antibodies that bind cd70 and uses thereof
US8470330B2 (en) 2001-10-23 2013-06-25 Psma Development Company, Llc PSMA antibodies and uses thereof
EP2740490A1 (en) * 2007-10-03 2014-06-11 Cornell University Treatment of proliferative disorders using antibodies to PSMA
US9242012B2 (en) 2008-09-08 2016-01-26 Psma Development Company, Llc Methods for killing PSMA-expressing, taxane-resistant cancer cells

Families Citing this family (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6749853B1 (en) 1992-03-05 2004-06-15 Board Of Regents, The University Of Texas System Combined methods and compositions for coagulation and tumor treatment
US5965132A (en) 1992-03-05 1999-10-12 Board Of Regents, The University Of Texas System Methods and compositions for targeting the vasculature of solid tumors
US7070782B1 (en) * 1992-11-05 2006-07-04 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen
US7105159B1 (en) 1992-11-05 2006-09-12 Sloan-Kettering Institute For Cancer Research Antibodies to prostate-specific membrane antigen
US6953668B1 (en) 1992-11-05 2005-10-11 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen
US6569432B1 (en) * 1995-02-24 2003-05-27 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen and uses thereof
CA2212846A1 (en) 1995-02-24 1996-08-29 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen and uses thereof
US20040253246A1 (en) * 1996-02-23 2004-12-16 Israeli Ron S. Prostate-specific membrane antigen and uses thereof
US6962981B1 (en) * 1996-03-25 2005-11-08 Medarex, Inc. Monoclonal antibodies specific for the extracellular domain of prostate-specific membrane antigen
US7381407B1 (en) * 1996-03-25 2008-06-03 Medarex, Inc. Monoclonal antibodies specific for the extracellular domain of prostate-specific membrane antigen
US6136311A (en) * 1996-05-06 2000-10-24 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
US6107090A (en) * 1996-05-06 2000-08-22 Cornell Research Foundation, Inc. Treatment and diagnosis of prostate cancer with antibodies to extracellur PSMA domains
US6759515B1 (en) * 1997-02-25 2004-07-06 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
US20060024301A1 (en) * 1997-02-25 2006-02-02 Corixa Corporation Prostate-specific polypeptides and fusion polypeptides thereof
US6903196B1 (en) 1999-06-17 2005-06-07 Utah Ventures Ii, L.P. Methods for identifying and isolating tissue-specific lumen-exposed molecules
US20040146516A1 (en) * 1999-06-17 2004-07-29 Utah Ventures Ii L.P. Lumen-exposed molecules and methods for targeted delivery
US6379550B1 (en) * 2000-07-24 2002-04-30 Health Research, Inc. Method for detecting PSA and its molecular forms using thiophilic gel
US7048931B1 (en) 2000-11-09 2006-05-23 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
EP1390078A4 (en) * 2001-04-26 2005-10-26 Analogic Corp Ct scanner for imaging a preselected portion of a patient
EP1390069A1 (en) * 2001-05-30 2004-02-25 Cornell Research Foundation, Inc. Endopeptidase/anti-psma antibody fusion proteins for treatment of cancer
US7666414B2 (en) * 2001-06-01 2010-02-23 Cornell Research Foundation, Inc. Methods for treating prostate cancer using modified antibodies to prostate-specific membrane antigen
JP4619651B2 (en) * 2001-06-01 2011-01-26 コーネル・リサーチ・ファンデーション・インコーポレイテッド Modified antibodies against prostate-specific membrane antigen and uses thereof
US7514078B2 (en) * 2001-06-01 2009-04-07 Cornell Research Foundation, Inc. Methods of treating prostate cancer with anti-prostate specific membrane antigen antibodies
JP2005501011A (en) * 2001-06-08 2005-01-13 ユタ ベンチャー ザ セカンド リミテッド パートナーシップ Tissue-specific inner membrane protein
WO2003024388A2 (en) * 2001-09-20 2003-03-27 Cornell Research Foundation, Inc. Methods and compositions for treating and preventing skin disorders using binding agents specific for psma
US20030129193A1 (en) * 2001-09-27 2003-07-10 Board Of Regents, The University Of Texas System And Peregrine Pharmaceuticals, Inc. Combined methods for tumor vasculature coaguligand treatment
EP1537146B9 (en) 2002-07-15 2011-07-06 Board Of Regents, The University Of Texas System Antibodies binding to anionic phospholipids and aminophospholipids and their use in treating viral infections
ATE408712T1 (en) * 2003-01-10 2008-10-15 Millennium Pharm Inc METHOD FOR DETERMINING RECURRENCE OF PROSTATE CANCER
US7902338B2 (en) 2003-07-31 2011-03-08 Immunomedics, Inc. Anti-CD19 antibodies
WO2005070456A2 (en) * 2004-01-09 2005-08-04 Millennium Pharmaceuticals, Inc. Diagnosing and treating female reproductive tract or chilhood cancer with pmsa antibodies
WO2005094882A1 (en) * 2004-03-03 2005-10-13 Millennium Pharmaceuticals, Inc. Modified antibodies to prostate-specific membrane antigen and uses thereof
US7713693B1 (en) 2004-09-01 2010-05-11 University Of Louisiana At Monroe Human cancer cell specific gene transcript
US9492400B2 (en) * 2004-11-04 2016-11-15 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
WO2006072625A2 (en) 2005-01-06 2006-07-13 Novo Nordisk A/S Anti-kir combination treatments and methods
WO2007070682A2 (en) * 2005-12-15 2007-06-21 Massachusetts Institute Of Technology System for screening particles
ES2363891T3 (en) 2006-03-20 2011-08-18 The Regents Of The University Of California ANTIBODIES AGAINST THE ANTIGEN OF TRONCAL CELLS OF THE PROSTATE (PSCA) GENETICALLY MODIFIED FOR ADDRESSING TO CANCER.
ES2776100T3 (en) 2006-03-31 2020-07-29 Massachusetts Inst Technology System for targeted delivery of therapeutic agents
JP5630998B2 (en) 2006-05-15 2014-11-26 マサチューセッツ インスティテュート オブ テクノロジー Polymers for functional particles
WO2007137117A2 (en) * 2006-05-17 2007-11-29 Massachusetts Institute Of Technology Aptamer-directed drug delivery
WO2007150030A2 (en) 2006-06-23 2007-12-27 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
WO2008147456A2 (en) * 2006-11-20 2008-12-04 Massachusetts Institute Of Technology Drug delivery systems using fc fragments
EP2134830A2 (en) * 2007-02-09 2009-12-23 Massachusetts Institute of Technology Oscillating cell culture bioreactor
WO2008124634A1 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Polymer-encapsulated reverse micelles
JP2010523595A (en) 2007-04-04 2010-07-15 マサチューセッツ インスティテュート オブ テクノロジー Poly (amino acid) targeting part
CA2698343C (en) 2007-09-04 2018-06-12 The Regents Of The University Of California High affinity anti-prostate stem cell antigen (psca) antibodies for cancer targeting and detection
WO2009051837A2 (en) 2007-10-12 2009-04-23 Massachusetts Institute Of Technology Vaccine nanotechnology
EP2281004A4 (en) * 2008-04-14 2012-02-15 Proscan Rx Pharma Inc Prostate specific membrane antigen antibodies and antigen binding fragments
US8343498B2 (en) * 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US8591905B2 (en) 2008-10-12 2013-11-26 The Brigham And Women's Hospital, Inc. Nicotine immunonanotherapeutics
US8343497B2 (en) 2008-10-12 2013-01-01 The Brigham And Women's Hospital, Inc. Targeting of antigen presenting cells with immunonanotherapeutics
US8277812B2 (en) 2008-10-12 2012-10-02 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IgG humoral response without T-cell antigen
DK3903829T3 (en) 2009-02-13 2023-06-26 Immunomedics Inc IMMUNE CONJUGATES WITH AN INTRACELLULAR CLEAVABLE BOND
ES2712732T3 (en) 2009-02-17 2019-05-14 Cornell Res Foundation Inc Methods and kits for the diagnosis of cancer and the prediction of therapeutic value
IN2012DN01663A (en) 2009-09-16 2015-06-05 Immunomedics Inc
CA2782194C (en) 2009-12-02 2018-01-16 Immunomedics, Inc. Combination of radiolabelled antibodies (rait) and antibody-drug conjugates (adc) for treatment of pancreatic cancer
EP3135302A1 (en) 2009-12-02 2017-03-01 Imaginab, Inc. J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use
CA2831572C (en) 2011-05-02 2019-11-26 Immunomedics, Inc. Ultrafiltration concentration of allotype selected antibodies for small-volume administration
CA2874864C (en) 2012-08-14 2023-02-21 Ibc Pharmaceuticals, Inc. T-cell redirecting bispecific antibodies for treatment of disease
WO2017004144A1 (en) 2015-07-01 2017-01-05 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
HUE057977T2 (en) 2012-12-13 2022-06-28 Immunomedics Inc Dosages of immunoconjugates of antibodies and sn-38 for improved efficacy and decreased toxicity
WO2014165818A2 (en) 2013-04-05 2014-10-09 T Cell Therapeutics, Inc. Compositions and methods for preventing and treating prostate cancer
JP6908964B2 (en) 2013-10-18 2021-07-28 ピーエスエムエー ディベロップメント カンパニー,エルエルシー Combination therapy with PSMA ligand conjugate
US20150110814A1 (en) 2013-10-18 2015-04-23 Psma Development Company, Llc Combination therapies with psma ligand conjugates
WO2015073896A2 (en) 2013-11-15 2015-05-21 Psma Development Company, Llc Biomarkers for psma targeted therapy for prostate cancer
MX2016010683A (en) 2014-02-21 2017-05-11 Ibc Pharmaceuticals Inc Disease therapy by inducing immune response to trop-2 expressing cells.
US9139649B2 (en) 2014-02-25 2015-09-22 Immunomedics, Inc. Humanized anti-CD22 antibody
WO2015200260A1 (en) 2014-06-24 2015-12-30 Immunomedics, Inc. Anti-histone therapy for vascular necrosis in severe glomerulonephritis
PL3204018T3 (en) 2014-10-07 2022-01-03 Immunomedics, Inc. Neoadjuvant use of antibody-drug conjugates
CA2981543A1 (en) 2015-04-22 2016-10-27 Immunomedics, Inc. Isolation, detection, diagnosis and/or characterization of circulating trop-2-positive cancer cells
CN107849148B (en) 2015-05-21 2023-09-19 哈普恩治疗公司 Trispecific binding proteins and methods of use
US10869940B2 (en) 2015-06-12 2020-12-22 The Board Of Trustees Of The Leland Stanford Junior University Targeted photoacoustic compounds, formulations, and uses thereof
ES2953441T3 (en) 2015-06-25 2023-11-13 Immunomedics Inc Combination of anti-hla-dr or anti-Trop-2 antibodies with microtubule inhibitors, parp inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
JOP20160154B1 (en) 2015-07-31 2021-08-17 Regeneron Pharma Anti-psma antibodies, bispecific antigen-binding molecules that bind psma and cd3, and uses thereof
SG10201913625XA (en) 2015-08-07 2020-03-30 Imaginab Inc Antigen binding constructs to target molecules
EA201892693A1 (en) 2016-05-20 2019-04-30 Харпун Терапьютикс, Инк. PROTEINS CONTAINING A SINGLE-BANDY VARIABLE FRAGMENT, CONNECTING CD3
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
US10100106B2 (en) 2016-05-20 2018-10-16 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
AU2017363300A1 (en) 2016-11-23 2019-06-20 Harpoon Therapeutics, Inc. Prostate specific membrane antigen binding protein
JP7215997B2 (en) 2016-11-23 2023-01-31 ハープーン セラピューティクス,インク. Trispecific proteins targeting prostate specific membrane antigen (PSMA) and methods of use
US11266745B2 (en) 2017-02-08 2022-03-08 Imaginab, Inc. Extension sequences for diabodies
EP3589662A4 (en) 2017-02-28 2020-12-30 Harpoon Therapeutics, Inc. Inducible monovalent antigen binding protein
US10730954B2 (en) 2017-05-12 2020-08-04 Harpoon Therapeutics, Inc. MSLN targeting trispecific proteins and methods of use
CA3063359A1 (en) 2017-05-12 2018-11-15 Harpoon Therapeutics, Inc. Mesothelin binding proteins
CN111465612A (en) 2017-10-13 2020-07-28 哈普恩治疗公司 B cell maturation antigen binding proteins
AU2018347582A1 (en) 2017-10-13 2020-05-07 Harpoon Therapeutics, Inc. Trispecific proteins and methods of use
AU2019346466A1 (en) 2018-09-25 2021-05-20 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
EP4106806A1 (en) 2020-02-21 2022-12-28 Harpoon Therapeutics, Inc. Flt3 binding proteins and methods of use
WO2022074152A1 (en) 2020-10-08 2022-04-14 Targimmune Therapeutics Ag Immunotherapy for the treatment of cancer
JPWO2022239720A1 (en) 2021-05-10 2022-11-17
WO2023079142A2 (en) 2021-11-05 2023-05-11 Targimmune Therapeutics Ag Targeted linear conjugates comprising polyethyleneimine and polyethylene glycol and polyplexes comprising the same

Citations (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4454106A (en) * 1982-06-07 1984-06-12 Gansow Otto A Use of metal chelate conjugated monoclonal antibodies
US4472509A (en) * 1982-06-07 1984-09-18 Gansow Otto A Metal chelate conjugated monoclonal antibodies
US4814275A (en) * 1985-05-13 1989-03-21 E.I. Dupont De Nemours And Company Monoclonal hybridoma antibody specific for a human epithelial antigen
US4855353A (en) * 1986-02-14 1989-08-08 Nihon Medi-Physics Co., Ltd. High molecular compounds having amino groups, and their utilization
US4863851A (en) * 1986-10-20 1989-09-05 The Upjohn Company Monoclonal antibody to prostate secretory protein
US4863854A (en) * 1985-08-12 1989-09-05 Sloan-Kettering Institute For Cancer Research Monoclonal antibodies to mucin-like human differentiation antigens
US4885363A (en) * 1987-04-24 1989-12-05 E. R. Squibb & Sons, Inc. 1-substituted-1,4,7-triscarboxymethyl-1,4,7,10-tetraazacyclododecane and analogs
US5013645A (en) * 1987-04-14 1991-05-07 Abbott Laboratories Immunological methods and materials for detection of tumor associated antigens
US5053503A (en) * 1989-02-17 1991-10-01 Centocor Chelating agents
US5057302A (en) * 1987-02-13 1991-10-15 Abbott Laboratories Bifunctional chelating agents
US5118611A (en) * 1988-07-25 1992-06-02 Adeza Biomedical Corporation Adenocarcinoma antigen binding methods and reagents
US5130118A (en) * 1987-11-06 1992-07-14 Abbott Laboratories Methods and materials for the preparation of metal labelled antibody solutions
US5162504A (en) * 1988-06-03 1992-11-10 Cytogen Corporation Monoclonal antibodies to a new antigenic marker in epithelial prostatic cells and serum of prostatic cancer patients
US5198208A (en) * 1987-07-16 1993-03-30 Nycomed Imaging As Aminopolycarboxylic acids and derivatives thereof
US5208324A (en) * 1988-01-26 1993-05-04 Nycomed Imaging As Paramagnetic compounds
US5217704A (en) * 1987-11-06 1993-06-08 Abbott Laboratories Methods and materials for the preparation of metal labelled antibody solutions
US5227471A (en) * 1992-01-30 1993-07-13 Eastern Virginia Medical School Of The Medical College Of Hampton Roads Monoclonal antibody PD41 that binds to a prostate mucin antigen that is expressed in human prostatic carcinoma
US5229289A (en) * 1988-03-11 1993-07-20 The Biomembrane Institute Monoclonal antibodies and vaccine development directed to human cancer-associated antigens by immunization with animal and human and with synthetic carbohydrate-carrier conjugates
US5342924A (en) * 1987-12-31 1994-08-30 Tanox Biosystems, Inc. Extracellular segments of human ε immunoglobulin anchoring peptides and antibodies specific therefor
US5474756A (en) * 1986-01-23 1995-12-12 Bracco International B.V. Method for imaging mammalian tissue using 1-substituted-1,4,7-tricarboxymethyl-1,4,7,10-tetraazacyclododecane and analogs
US5489525A (en) * 1992-10-08 1996-02-06 The United States Of America As Represented By The Department Of Health And Human Services Monoclonal antibodies to prostate cells
US5531978A (en) * 1987-07-16 1996-07-02 Nycomed Imaging As Aminopolycarboxylic acids and derivatives thereof
US5538866A (en) * 1992-11-05 1996-07-23 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen
US5565562A (en) * 1989-02-10 1996-10-15 Celltech Therapeutics Limited Triaza macrocycles
US5599677A (en) * 1993-12-29 1997-02-04 Abbott Laboratories Immunoassays for prostate specific antigen
US5639879A (en) * 1993-04-02 1997-06-17 Associated Universities, Inc. Macrocyclic polyaminocarboxylates for stable radiometal antibody conjugates for therapy, spect and pet imaging
US5660827A (en) * 1992-03-05 1997-08-26 Board Of Regents, The University Of Texas System Antibodies that bind to endoglin
US5697902A (en) * 1985-07-05 1997-12-16 Immunomedics, Inc. Method for imaging and treating organs and tissues
US5804602A (en) * 1996-06-17 1998-09-08 Guilford Pharmaceuticals Inc. Methods of cancer treatment using naaladase inhibitors
US5855866A (en) * 1992-03-05 1999-01-05 Board Of Regenis, The University Of Texas System Methods for treating the vasculature of solid tumors
US5863538A (en) * 1992-03-05 1999-01-26 Board Of Regents, The University Of Texas System Compositions for targeting the vasculature of solid tumors
US5877289A (en) * 1992-03-05 1999-03-02 The Scripps Research Institute Tissue factor compositions and ligands for the specific coagulation of vasculature
US5935818A (en) * 1995-02-24 1999-08-10 Sloan-Kettering Institute For Cancer Research Isolated nucleic acid molecule encoding alternatively spliced prostate-specific membrane antigen and uses thereof
US5958474A (en) * 1996-08-21 1999-09-28 Nestec S.A. Process for preparing food flavor precursors
US6004555A (en) * 1992-03-05 1999-12-21 Board Of Regents, The University Of Texas System Methods for the specific coagulation of vasculature
US6022524A (en) * 1997-04-18 2000-02-08 Dibra S.P.A. Process for the conjugation of chelants with molecules containing amino groups
US6036955A (en) * 1992-03-05 2000-03-14 The Scripps Research Institute Kits and methods for the specific coagulation of vasculature
US6093399A (en) * 1992-03-05 2000-07-25 Board Of Regents, The University Of Texas System Methods and compositions for the specific coagulation of vasculature
US6136311A (en) * 1996-05-06 2000-10-24 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
US6150508A (en) * 1996-03-25 2000-11-21 Northwest Biotherapeutics, Inc. Monoclonal antibodies specific for the extracellular domain of prostate-specific membrane antigen
US6683162B2 (en) * 2000-09-15 2004-01-27 Sloan Kettering Institute Of Cancer Research Targeted alpha particle therapy using actinium-255 conjugates

Family Cites Families (236)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5292668A (en) 1981-12-21 1994-03-08 Boston Biomedical Research Institute, Inc. Bispecific antibody determinants
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4824986A (en) 1985-04-26 1989-04-25 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Metal chelate protein conjugate
US4735210A (en) 1985-07-05 1988-04-05 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US5525338A (en) 1992-08-21 1996-06-11 Immunomedics, Inc. Detection and therapy of lesions with biotin/avidin conjugates
DE3772785D1 (en) 1986-01-23 1991-10-17 Squibb & Sons Inc 1-SUBSTITUTED-4,7,10-TRISCARBOXYMETHYL-1,4,7,10-TETRAAZACYCLODODECAN AND ANALOG.
US5869620A (en) 1986-09-02 1999-02-09 Enzon, Inc. Multivalent antigen-binding proteins
US5260203A (en) 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
FR2604092B1 (en) 1986-09-19 1990-04-13 Immunotech Sa IMMUNOREACTIVES FOR TARGETING ANIMAL CELLS FOR VISUALIZATION OR DESTRUCTION IN VIVO
WO1988002635A1 (en) 1986-10-17 1988-04-21 Cytogen Corporation Method for preparation of protein-chelator-metal ion compositions suitable for injection
US5001225A (en) * 1986-12-08 1991-03-19 Georgetown University Monoclonal antibodies to a pan-malarial antigen
CA1306739C (en) 1987-02-16 1992-08-25 Gohfu Suzukamo Solid base, process for preparing the same and use of the same in preparation of internal olefins
GB9007965D0 (en) 1990-04-09 1990-06-06 Nycomed As Compounds
EP0382736B1 (en) 1987-07-27 1994-11-02 Commonwealth Scientific And Industrial Research Organisation Receptor membranes
US4943525A (en) 1987-11-02 1990-07-24 Bioventures, Inc. Simultaneous immunoassay for the determination of antigens and antibodies
AU619218B2 (en) 1987-11-06 1992-01-23 Abbott Laboratories Methods and materials for the preparation of metal labelled antibody solutions
US5869053A (en) 1988-03-04 1999-02-09 Cancer Research Campaign Technology, Ltd. 5T4 antigen from human trophoblasts
US4892824A (en) 1988-03-15 1990-01-09 Synbiotics Corporation Fast track method for producing monoclonal bi-specific immunoglobulins
US5120525A (en) 1988-03-29 1992-06-09 Immunomedics, Inc. Radiolabeled antibody cytotoxic therapy of cancer
US6010902A (en) 1988-04-04 2000-01-04 Bristol-Meyers Squibb Company Antibody heteroconjugates and bispecific antibodies for use in regulation of lymphocyte activity
US5851527A (en) 1988-04-18 1998-12-22 Immunomedics, Inc. Method for antibody targeting of therapeutic agents
AU634186B2 (en) 1988-11-11 1993-02-18 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
DE3911816A1 (en) 1989-04-11 1990-10-25 Hoechst Ag SUBSTITUTED 1,4,7,10-TETRAAZACYCLOTRIDECANE, METHOD FOR THE PRODUCTION THEREOF AND THE USE THEREOF FOR MARKING SUBSTANCES WITH RADIONUCLIDES
DE3920358A1 (en) 1989-06-22 1991-01-17 Behringwerke Ag BISPECIFIC AND OLIGO-SPECIFIC, MONO- AND OLIGOVALENT ANTI-BODY CONSTRUCTS, THEIR PRODUCTION AND USE
US5980896A (en) 1989-06-30 1999-11-09 Bristol-Myers Squibb Company Antibodies reactive with human carcinomas
US5332567A (en) 1989-08-24 1994-07-26 Immunomedics Detection and treatment of infections with immunoconjugates
US5244816A (en) 1989-10-11 1993-09-14 Akzo N.V. Method for purifying chelator conjugated compounds
WO1991007493A1 (en) 1989-11-13 1991-05-30 Xoma Corporation Chimeric mouse human antibodies with specificity to hiv antigens
GB8928874D0 (en) * 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US5582996A (en) 1990-12-04 1996-12-10 The Wistar Institute Of Anatomy & Biology Bifunctional antibodies and method of preparing same
US6197298B1 (en) 1991-04-19 2001-03-06 Tanox, Inc. Modified binding molecules specific for T lymphocytes and their use as in vivo immune modulators in animals
DE4118120A1 (en) 1991-06-03 1992-12-10 Behringwerke Ag TETRAVALENT BISPECIFIC RECEPTORS, THEIR PRODUCTION AND USE
US5637481A (en) 1993-02-01 1997-06-10 Bristol-Myers Squibb Company Expression vectors encoding bispecific fusion proteins and methods of producing biologically active bispecific fusion proteins in a mammalian cell
GB9115192D0 (en) 1991-07-12 1991-08-28 Antisoma Ltd Cancer treatment
DE4134982A1 (en) 1991-10-23 1993-04-29 Kernforschungsz Karlsruhe USE OF ANTIBODY-CONTAINING PREPARATIONS FOR IMMUNE SUPPRESSION
US5932448A (en) 1991-11-29 1999-08-03 Protein Design Labs., Inc. Bispecific antibody heterodimers
DE4140142A1 (en) 1991-12-05 1993-06-09 Boehringer Mannheim Gmbh, 6800 Mannheim, De MULTIVALENT DEXTRANREAGENT FOR USE IN PRECIPITATION TESTS
AU668374B2 (en) 1991-12-10 1996-05-02 Dana-Farber Cancer Institute Reactive neutralizing human anti-GP120 recombinant antibody, DNA coding the same and use thereof
FR2686899B1 (en) 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
ATE419355T1 (en) 1992-02-06 2009-01-15 Novartis Vaccines & Diagnostic MARKER FOR CANCER AND BIOSYNTHETIC BINDING PROTEIN FOR IT
US6096289A (en) 1992-05-06 2000-08-01 Immunomedics, Inc. Intraoperative, intravascular, and endoscopic tumor and lesion detection, biopsy and therapy
EP1550729B1 (en) 1992-09-25 2009-05-27 Avipep Pty Limited Target binding polypeptide comprising an IG-like VL domain linked to an IG-like VH domain
AU5322494A (en) 1992-10-02 1994-04-26 Trustees Of Dartmouth College Bispecific reagents for redirected targeting of low density lipoprotein
EP0627932B1 (en) 1992-11-04 2002-05-08 City Of Hope Antibody construct
US7105159B1 (en) 1992-11-05 2006-09-12 Sloan-Kettering Institute For Cancer Research Antibodies to prostate-specific membrane antigen
US6953668B1 (en) 1992-11-05 2005-10-11 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen
US7070782B1 (en) 1992-11-05 2006-07-04 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen
AU5670194A (en) 1992-11-20 1994-06-22 Enzon, Inc. Linker for linked fusion polypeptides
US5376249A (en) 1992-11-25 1994-12-27 Perseptive Biosystems, Inc. Analysis utilizing isoelectric focusing
US5837242A (en) 1992-12-04 1998-11-17 Medical Research Council Multivalent and multispecific binding proteins, their manufacture and use
GB9225453D0 (en) 1992-12-04 1993-01-27 Medical Res Council Binding proteins
DE69327229T2 (en) 1992-12-11 2000-03-30 Dow Chemical Co Multivalent single chain antibodies
US5861156A (en) 1993-01-08 1999-01-19 Creative Biomolecules Methods of delivering agents to target cells
US5705614A (en) 1993-04-09 1998-01-06 Chiron Corporation Methods of producing antigen forks
KR100220864B1 (en) 1993-05-17 1999-09-15 오트리브 데이비스 더블유 Improved detection and therapy of lesions with biotin/avidin-metal chelating protein conjugates
US5869049A (en) 1993-09-02 1999-02-09 Trustees Of Dartmouth College Methods of inducing T cell unresponsiveness to bone marrow with gp39 antagonists
ZA946765B (en) 1993-09-02 1996-02-15 Dartmouth College Methods of prolonged suppression of humoral immunity
US6569432B1 (en) 1995-02-24 2003-05-27 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen and uses thereof
GB9324807D0 (en) 1993-12-03 1994-01-19 Cancer Res Campaign Tech Tumour antibody
AU2194695A (en) 1994-03-28 1995-10-17 Regents Of The University Of California, The Method for preparing radionuclide-labeled chelating agent-ligand complexes
US6693190B1 (en) 1994-05-11 2004-02-17 Bracco International B.V. Enhanced relaxivity monomeric and multimeric compounds
DE4421391C1 (en) 1994-06-18 1995-11-30 Gsf Forschungszentrum Umwelt Use of antibodies against T cells for prolonged immunosuppression
US5559099A (en) 1994-09-08 1996-09-24 Genvec, Inc. Penton base protein and methods of using same
US5541087A (en) 1994-09-14 1996-07-30 Fuji Immunopharmaceuticals Corporation Expression and export technology of proteins as immunofusins
US5688690A (en) 1994-09-16 1997-11-18 The Wistar Institute Of Anatomy And Biology Human cytotoxic lymphocyte signal transduction surface protein (P38) and monoclonal antibodies thereto
CA2212846A1 (en) 1995-02-24 1996-08-29 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen and uses thereof
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
GB9504344D0 (en) 1995-03-03 1995-04-19 Unilever Plc Antibody fragment production
US5747035A (en) 1995-04-14 1998-05-05 Genentech, Inc. Polypeptides with increased half-life for use in treating disorders involving the LFA-1 receptor
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
US6096871A (en) 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
AUPO591797A0 (en) 1997-03-27 1997-04-24 Commonwealth Scientific And Industrial Research Organisation High avidity polyvalent and polyspecific reagents
US5773292A (en) 1995-06-05 1998-06-30 Cornell University Antibodies binding portions, and probes recognizing an antigen of prostate epithelial cells but not antigens circulating in the blood
US5830478A (en) 1995-06-07 1998-11-03 Boston Biomedical Research Institute Method for delivering functional domains of diphtheria toxin to a cellular target
DE69638134D1 (en) 1995-06-07 2010-04-08 Immunomedics Inc IMPROVED DELIVERY OF DIAGNOSTIC AND THERAPEUTIC SUBSTANCES AT ONE DESTINATION
BR9606706A (en) 1995-10-16 1999-04-06 Unilever Nv Bispecific or bivalent antibody fragment analog use process to produce the same
EP0856054B1 (en) 1995-10-19 2004-04-28 Bristol-Myers Squibb Company Monoclonal antibody br110 and uses thereof
US20040253246A1 (en) 1996-02-23 2004-12-16 Israeli Ron S. Prostate-specific membrane antigen and uses thereof
IT1283218B1 (en) 1996-03-08 1998-04-16 Bracco Spa POLYKELANTS, THEIR COMPLEXES WITH METALLIC IONS, THEIR PREPARATION AND THEIR USES
ATE318147T1 (en) 1996-03-25 2006-03-15 Medarex Inc SPECIFIC MONOCLONAL ANTIBODIES FOR THE EXTRACELLULAR DOMAIN OF PROTASTA-SPECIFIC MEMBRANE ANTIGEN
US6962981B1 (en) * 1996-03-25 2005-11-08 Medarex, Inc. Monoclonal antibodies specific for the extracellular domain of prostate-specific membrane antigen
US6107090A (en) * 1996-05-06 2000-08-22 Cornell Research Foundation, Inc. Treatment and diagnosis of prostate cancer with antibodies to extracellur PSMA domains
WO1997044664A1 (en) 1996-05-23 1997-11-27 Unilever Plc Improvements in or relating to specific binding assays
DK0922102T3 (en) 1996-07-03 2010-08-16 Genentech Inc Hepatocyte growth factor receptor agonists and applications thereof
GB9712818D0 (en) 1996-07-08 1997-08-20 Cambridge Antibody Tech Labelling and selection of specific binding molecules
US5922845A (en) 1996-07-11 1999-07-13 Medarex, Inc. Therapeutic multispecific compounds comprised of anti-Fcα receptor antibodies
WO1998016254A1 (en) 1996-10-17 1998-04-23 Immunomedics, Inc. Non-antigenic toxin-conjugate and fusion protein of internalizing receptor system
WO1998029736A1 (en) 1996-12-31 1998-07-09 Genometrix Incorporated Multiplexed molecular analysis apparatus and method
GB9704444D0 (en) * 1997-03-04 1997-04-23 Isis Innovation Non-invasive prenatal diagnosis
US6541212B2 (en) 1997-03-10 2003-04-01 The Regents Of The University Of California Methods for detecting prostate stem cell antigen protein
EP0977837A1 (en) 1997-04-03 2000-02-09 Universite Laval Transgenic expression in genital tract and sexual accessory glands
US6368596B1 (en) 1997-07-08 2002-04-09 Board Of Regents, The University Of Texas System Compositions and methods for homoconjugates of antibodies which induce growth arrest or apoptosis of tumor cells
US6977074B2 (en) 1997-07-10 2005-12-20 Mannkind Corporation Method of inducing a CTL response
US6994851B1 (en) 1997-07-10 2006-02-07 Mannkind Corporation Method of inducing a CTL response
JP3992923B2 (en) 1997-09-03 2007-10-17 イムノメディクス, インコーポレイテッド Protein and peptide fluorination process for F-18 positron emission tomography
US6030792A (en) 1997-11-13 2000-02-29 Pfizer Inc Assays for measurement of protein fragments in biological media
US6709844B1 (en) 2000-11-16 2004-03-23 Mannkind Corporation Avoidance of undesirable replication intermediates in plasmid propagation
AU3305699A (en) 1998-02-26 1999-09-15 Beckman Coulter, Inc. Prostate-specific membrane antigens and methods of making and using
DE19911329A1 (en) 1998-03-27 2000-09-21 Benes Ivan Friedrich Radioimmunoconjugate which can be used in human therapy and process for its preparation
HUP9900956A2 (en) 1998-04-09 2002-04-29 Aventis Pharma Deutschland Gmbh. Single-chain multiple antigen-binding molecules, their preparation and use
DK1071700T3 (en) 1998-04-20 2010-06-07 Glycart Biotechnology Ag Glycosylation modification of antibodies to enhance antibody-dependent cellular cytotoxicity
US6200765B1 (en) 1998-05-04 2001-03-13 Pacific Northwest Cancer Foundation Non-invasive methods to detect prostate cancer
US6071490A (en) 1998-05-07 2000-06-06 Immunomedics, Inc. Position emission tomography using gallium-68 chelates
ATE473242T1 (en) 1998-05-20 2010-07-15 Immunomedics Inc THERAPEUTIC AGENTS CONTAINING A BISPECIFIC ANTI-HLA CLASS II INVARIANT CHAIN X ANTI-PATHOGEN ANTIBODIES
US20040141975A1 (en) 1998-06-01 2004-07-22 Raitano Arthur B. Nucleic acid and corresponding protein entitled 98P4B6 useful in treatment and detection of cancer
US20030149531A1 (en) 2000-12-06 2003-08-07 Hubert Rene S. Serpentine transmembrane antigens expressed in human cancers and uses thereof
US6833438B1 (en) 1999-06-01 2004-12-21 Agensys, Inc. Serpentine transmembrane antigens expressed in human cancers and uses thereof
US20060052321A1 (en) 2002-04-05 2006-03-09 Raitano Arthur B Nucleic acid and corresponding protein entitled 98P4B6 useful in treatment and detection of cancer
AU757698C (en) 1998-06-01 2004-04-08 Agensys, Inc. Novel serpentine transmembrane antigens expressed in human cancers and uses thereof
EP1098665B9 (en) 1998-07-13 2003-08-13 The Board Of Regents, The University Of Texas System Cancer treatment methods using therapeutic conjugates that bind to aminophospholipids
WO2000014234A1 (en) 1998-09-08 2000-03-16 Urocor, Inc. Prostate specific promoter and regulation of gene expression
US6361774B1 (en) 1999-09-17 2002-03-26 Immunomedics, Inc. Methods and compositions for increasing the target-specific toxicity of a chemotherapy drug
US6355623B2 (en) * 1998-09-24 2002-03-12 Hopital-Sainte-Justine Method of treating IBD/Crohn's disease and related conditions wherein drug metabolite levels in host blood cells determine subsequent dosage
US6284742B1 (en) 1998-09-29 2001-09-04 Uab Research Foundation Immunomodulation by genetic modification of dendritic cells and B cells
WO2000034461A2 (en) 1998-12-10 2000-06-15 Board Of Regents, The University Of Texas System Compositions and methods of modulating cholesterol metabolism
WO2000050457A1 (en) 1999-02-22 2000-08-31 Beckman Coulter, Inc. Prostate-specific membrane antigens and methods of making and using
US20020102208A1 (en) 1999-03-01 2002-08-01 Paul Chinn Radiolabeling kit and binding assay
EP1175616A4 (en) 1999-04-13 2003-08-13 Northwest Biotherapeutics Inc Methods for the diagnosis and treatment of metastatic prostate tumors
ES2223705T3 (en) 1999-04-28 2005-03-01 Board Of Regents, The University Of Texas System COMPOSITIONS AND METHODS FOR THE TREATMENT OF CANCER THROUGH INHIBITION SELECTIVE OF VEGF.
JP2004510683A (en) 1999-06-07 2004-04-08 センター・フォー・モレキュラー・メディシン・アンド・イムノロジー Alpha or beta emitters attached to fragments in radioimmunotherapy
WO2001009303A2 (en) 1999-07-30 2001-02-08 Vical Inc. Flt-3 LIGAND-ENCODING POLYNUCLEOTIDE AS A POLYNUCLEOTIDE-BASED VACCINE ENHANCER
KR20050004240A (en) 1999-08-31 2005-01-12 제넨테크, 인크. Compositions and Methods for the Treatment of Tumor
US7442776B2 (en) * 1999-10-08 2008-10-28 Young David S F Cancerous disease modifying antibodies
US6342587B1 (en) 1999-10-22 2002-01-29 Ludwig Institute For Cancer Research A33 antigen specific immunoglobulin products and uses thereof
US6824780B1 (en) 1999-10-29 2004-11-30 Genentech, Inc. Anti-tumor antibody compositions and methods of use
JP2003514903A (en) 1999-11-24 2003-04-22 イムノージェン インコーポレーテッド Taxane-containing cytotoxic drugs and their therapeutic use
CZ20023203A3 (en) 2000-03-24 2003-08-13 Micromet Ag Multifunctional polypeptides comprising a binding site to an epitope of the NKG2D receptor complex
US6573096B1 (en) 2000-04-01 2003-06-03 The Research Foundation At State University Of New York Compositions and methods for inhibition of cancer invasion and angiogenesis
WO2001077342A1 (en) 2000-04-11 2001-10-18 Genentech, Inc. Multivalent antibodies and uses therefor
CA2405363A1 (en) 2000-04-28 2001-11-08 Ctl Immunotherapies Corp. Epitope synchronization in antigen presenting cells
US6861234B1 (en) 2000-04-28 2005-03-01 Mannkind Corporation Method of epitope discovery
US6375991B1 (en) 2000-09-08 2002-04-23 Albemarle Corporation Production of concentrated biocidal solutions
DE10045591A1 (en) 2000-09-15 2002-04-04 Klaus Pfizenmaier Site-specific, antibody-mediated activation of proapoptotic cytokines - AMAIZe (Antibody-Mediated Apoptosis Inducing Cytokines)
US7229960B2 (en) * 2000-11-03 2007-06-12 University Of Vermont And State Agricultural College Methods and compositions for inhibiting GRB7
US6972324B2 (en) 2001-05-18 2005-12-06 Boehringer Ingelheim Pharmaceuticals, Inc. Antibodies specific for CD44v6
US7666414B2 (en) 2001-06-01 2010-02-23 Cornell Research Foundation, Inc. Methods for treating prostate cancer using modified antibodies to prostate-specific membrane antigen
JP4619651B2 (en) * 2001-06-01 2011-01-26 コーネル・リサーチ・ファンデーション・インコーポレイテッド Modified antibodies against prostate-specific membrane antigen and uses thereof
US7514078B2 (en) * 2001-06-01 2009-04-07 Cornell Research Foundation, Inc. Methods of treating prostate cancer with anti-prostate specific membrane antigen antibodies
KR20040054669A (en) 2001-08-03 2004-06-25 글리카트 바이오테크놀로지 아게 Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
EP2287186B1 (en) 2001-09-06 2014-12-31 Agensys, Inc. Nucleic acid and corresponding protein entitled STEAP-1 useful in treatment and detection of cancer
US7494646B2 (en) 2001-09-06 2009-02-24 Agensys, Inc. Antibodies and molecules derived therefrom that bind to STEAP-1 proteins
WO2003024388A2 (en) 2001-09-20 2003-03-27 Cornell Research Foundation, Inc. Methods and compositions for treating and preventing skin disorders using binding agents specific for psma
HUP0600342A3 (en) 2001-10-25 2011-03-28 Genentech Inc Glycoprotein compositions
GB0126378D0 (en) 2001-11-02 2002-01-02 Oxford Biomedica Ltd Antigen
ES2358642T3 (en) 2001-11-07 2011-05-12 Mannkind Corporation EXPRESSION VECTORS CODING ANTIGENS EPITHOPES AND METHODS FOR THEIR DESIGN.
US20040018519A1 (en) 2001-11-16 2004-01-29 Wright ,Jr. George L Methods and devices for quantitative detection of prostate specific membrane antigen and other prostatic markers
AU2002351353A1 (en) 2001-12-19 2003-09-02 Genentech, Inc. Stabilizing polypeptides which have been exposed to urea
AU2003210802B2 (en) 2002-02-05 2009-09-10 Genentech Inc. Protein purification
EP1499353A4 (en) 2002-04-15 2006-04-05 Human Genome Sciences Inc Antibodies that specifically bind to tl5
ES2545067T3 (en) 2002-04-26 2015-09-08 Genentech, Inc. Protein purification based on non-affinity
EP2365004B1 (en) 2002-06-21 2016-01-06 Johns Hopkins University School of Medicine Membrane associated tumor endothelium markers
US9809654B2 (en) 2002-09-27 2017-11-07 Vaccinex, Inc. Targeted CD1d molecules
US7820166B2 (en) 2002-10-11 2010-10-26 Micromet Ag Potent T cell modulating molecules
PL224786B1 (en) 2003-01-22 2017-01-31 Glycart Biotechnology Ag Fusion constructs and use of same to produce antibodies with increased fc receptor binding affinity and effector function
US20050026178A1 (en) 2003-03-28 2005-02-03 Marit Nilsen-Hamilton Allosteric probes and methods
GB0308988D0 (en) 2003-04-17 2003-05-28 Univ Singapore Molecule
US7595379B2 (en) 2003-05-30 2009-09-29 Agensys, Inc. Antibodies and related molecules that bind to PSCA proteins
AU2004242845B2 (en) 2003-05-31 2011-06-02 Amgen Research (Munich) Gmbh Human-anti-human CD3 binding molecules
CA2529945A1 (en) 2003-06-27 2005-01-06 Biogen Idec Ma Inc. Use of hydrophobic-interaction-chromatography or hinge-region modifications for the production of homogeneous antibody-solutions
AU2004265253B2 (en) 2003-07-28 2011-09-01 Genentech, Inc. Reducing protein A leaching during protein A affinity chromatography
GB0321805D0 (en) 2003-09-18 2003-10-15 Univ Wales Medicine Human tumour growth patterns
US7524813B2 (en) 2003-10-10 2009-04-28 Novo Nordisk Health Care Ag Selectively conjugated peptides and methods of making the same
ATE493441T1 (en) 2003-10-16 2011-01-15 Micromet Ag MULTI-SPECIFIC DEIMMUNIZED CD3-BINDING MOLECULES
ES2428358T3 (en) 2003-10-17 2013-11-07 Novo Nordisk A/S Combination therapy
ITPD20030264A1 (en) 2003-10-30 2005-04-30 Xeptagen Spa HIGHLY SPECIFIC DIAGNOSIS METHOD FOR NEOPLASIA
CA2543888C (en) 2003-11-06 2016-01-19 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
US7235641B2 (en) 2003-12-22 2007-06-26 Micromet Ag Bispecific antibodies
CA2553261C (en) 2004-01-16 2014-03-18 Stefan Barth Immunokinases
CA2555503C (en) 2004-02-16 2018-03-27 Micromet Ag Humanized anti-cd3 bispecific binding molecules having reduced immunogenicity, uses and compositions relating thereto
EP1718667B1 (en) 2004-02-23 2013-01-09 Genentech, Inc. Heterocyclic self-immolative linkers and conjugates
WO2005094882A1 (en) 2004-03-03 2005-10-13 Millennium Pharmaceuticals, Inc. Modified antibodies to prostate-specific membrane antigen and uses thereof
JP4861308B2 (en) 2004-04-07 2012-01-25 ジェネンテック, インコーポレイテッド Mass spectrometry of antibody conjugates
PT1742966E (en) 2004-04-22 2014-02-05 Agensys Inc Antibodies and molecules derived therefrom that bind to steap-1 proteins
NZ551180A (en) 2004-06-01 2009-10-30 Genentech Inc Antibody drug conjugates and methods
WO2005118802A2 (en) 2004-06-03 2005-12-15 The Regents Of The University Of California Targeting pseudotyped retroviral vectors
US20060159689A1 (en) 2004-06-17 2006-07-20 Chih-Sheng Chiang Combinations of tumor-associated antigens in diagnostics for various types of cancers
NZ552745A (en) 2004-07-16 2009-01-31 Micromet Ag Expression-enhanced polypeptides
CN101065151B (en) 2004-09-23 2014-12-10 健泰科生物技术公司 Cysteine engineered antibodies and conjugates
US20100111856A1 (en) 2004-09-23 2010-05-06 Herman Gill Zirconium-radiolabeled, cysteine engineered antibody conjugates
US7947839B2 (en) 2004-12-01 2011-05-24 Genentech, Inc. Heterocyclic-substituted bis-1,8 naphthalimide compounds, antibody drug conjugates, and methods of use
US20070134243A1 (en) 2004-12-01 2007-06-14 Gazzard Lewis J Antibody drug conjugates and methods
CA2491067A1 (en) 2004-12-24 2006-06-24 Stichting Katholieke Universiteit Mrna rations in urinary sediments and/or urine as a prognostic marker for prostate cancer
GB2426581A (en) 2005-05-27 2006-11-29 Univ Nottingham Immunoassay methods
HUE026303T2 (en) 2005-07-25 2016-06-28 Emergent Product Dev Seattle B-cell reduction using CD37-specific and CD20-specific binding molecules
US20080279850A1 (en) 2005-07-25 2008-11-13 Trubion Pharmaceuticals, Inc. B-Cell Reduction Using CD37-Specific and CD20-Specific Binding Molecules
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
WO2007100385A2 (en) 2005-10-31 2007-09-07 Genentech, Inc. Macrocyclic depsipeptide antibody-drug conjugates and methods
US20070212331A1 (en) 2006-03-07 2007-09-13 Baldassare Joseph J Methods and compositions for selectively killing cells
ES2363891T3 (en) 2006-03-20 2011-08-18 The Regents Of The University Of California ANTIBODIES AGAINST THE ANTIGEN OF TRONCAL CELLS OF THE PROSTATE (PSCA) GENETICALLY MODIFIED FOR ADDRESSING TO CANCER.
RU2008134135A (en) 2006-03-21 2010-04-27 Вайет (Us) COMPOUNDS-ANTAGONISTS OF THE PROTEIN RAGE AND WAYS OF THEIR APPLICATION
ES2776100T3 (en) 2006-03-31 2020-07-29 Massachusetts Inst Technology System for targeted delivery of therapeutic agents
US7566276B2 (en) 2006-04-14 2009-07-28 Dogleg Right Corporation Multi-piece putter head having an insert
WO2007137117A2 (en) 2006-05-17 2007-11-29 Massachusetts Institute Of Technology Aptamer-directed drug delivery
CN101632020B (en) 2006-09-13 2013-11-27 昂西免疫有限公司 Improved immunoassay methods
BRPI0717395A2 (en) 2006-10-25 2013-10-15 Koninkl Philips Electronics Nv COMPOUND FOR DIAGNOSING PROSTATE CANCER IN A HUMAN OR ANIMAL INDIVIDUAL, PHARMACEUTICAL COMPOSITION, AND, USE OF A COMPOUND OR COMPOSITION
WO2008079302A2 (en) 2006-12-21 2008-07-03 Millipore Corporation Purification of proteins
US8362217B2 (en) 2006-12-21 2013-01-29 Emd Millipore Corporation Purification of proteins
EP2144930A1 (en) 2007-04-18 2010-01-20 ZymoGenetics, Inc. Single chain fc, methods of making and methods of treatment
US20090252729A1 (en) 2007-05-14 2009-10-08 Farrington Graham K Single-chain Fc (scFc) regions, binding polypeptides comprising same, and methods related thereto
US9364557B2 (en) 2007-08-01 2016-06-14 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Fold-back diabody diphtheria toxin immunotoxin and methods of use
CA2698343C (en) 2007-09-04 2018-06-12 The Regents Of The University Of California High affinity anti-prostate stem cell antigen (psca) antibodies for cancer targeting and detection
WO2009039854A2 (en) 2007-09-27 2009-04-02 Dako Denmark A/S Mhc multimers in tuberculosis diagnostics, vaccine and therapeutics
RU2499606C2 (en) 2007-10-18 2013-11-27 Бавэариан Нордик Инк. Using mva (modified vaccinia ankara) for treating prostate cancer
US8273550B2 (en) 2007-10-25 2012-09-25 Viventia Biotechnologies Inc. Antibodies against a cancer-associated epitope of variant HnRNPG and uses thereof
US9107858B2 (en) 2007-12-05 2015-08-18 Wisconsin Alumni Research Foundation Dendritic cell targeting compositions and uses thereof
WO2009097128A1 (en) 2008-01-29 2009-08-06 Ludwig Institute For Cancer Research Ltd. Membrane transporter napi2b (slc34a2) epitope for antibody therapy, antibodies directed thereto, and target for cancer therapy
WO2009099741A1 (en) 2008-02-01 2009-08-13 Genentech, Inc. Nemorubicin metabolite and analog reagents, antibody-drug conjugates and methods
US8043830B2 (en) 2008-02-01 2011-10-25 The Regents Of The University Of California Biotin-ligase system for secretion of biotinylated protein
TW200947474A (en) 2008-05-05 2009-11-16 Ren-Huan Pan Manufacturing equipment and method of quadratic arc alignment dua-turret formation center, and miniature milliohm current sensors manufactured thereof
AU2009246516B2 (en) 2008-05-13 2015-03-05 Genentech, Inc. Analysis of antibody drug conjugates by bead-based affinity capture and mass spectrometry
MX2011000509A (en) 2008-07-15 2011-04-05 Genentech Inc Anthracycline derivative conjugates, process for their preparation and their use as antitumor compounds.
US20100069616A1 (en) 2008-08-06 2010-03-18 The Regents Of The University Of California Engineered antibody-nanoparticle conjugates
RU2509968C2 (en) 2008-09-08 2014-03-20 Конокофиллипс Компани System for separation of non-condensed component at natural gas liquefaction plant
WO2010037397A1 (en) 2008-10-01 2010-04-08 Dako Denmark A/S Mhc multimers in cmv immune monitoring
US20110318380A1 (en) 2008-10-01 2011-12-29 Dako Denmark A/S MHC Multimers in Cancer Vaccines and Immune Monitoring
EP2356457B1 (en) 2008-10-13 2018-05-16 Xeptagen SPA Method for the preparation of immunoconjugates and use thereof
CA2742324A1 (en) 2008-10-30 2010-06-03 Caris Life Sciences Luxembourg Holdings, S.A.R.L. Methods for assessing rna patterns
WO2010056337A2 (en) 2008-11-12 2010-05-20 Caris Mpi, Inc. Methods and systems of using exosomes for determining phenotypes
KR20110083730A (en) 2008-11-13 2011-07-20 이머전트 프로덕트 디벨롭먼트 시애틀, 엘엘씨 Cd37 immunotherapeutic combination therapies and uses thereof
SG171764A1 (en) 2008-12-16 2011-07-28 Millipore Corp Purification of proteins
WO2010073462A1 (en) 2008-12-25 2010-07-01 東海ゴム工業株式会社 Fluid-sealed vibration damping device
RU2636046C2 (en) 2009-01-12 2017-11-17 Сайтомкс Терапьютикс, Инк Modified antibodies composition, methods of production and application
WO2010102195A2 (en) 2009-03-06 2010-09-10 The Johns Hopkins University Annexin a11 and associated genes as biomarkers for cancer
EP2411808B1 (en) 2009-03-24 2015-11-11 Biocept, Inc. Devices and methods of cell capture and analysis
CN102459346B (en) 2009-04-27 2016-10-26 昂考梅德药品有限公司 The method manufacturing heteromultimers molecule
US20100303821A1 (en) 2009-05-28 2010-12-02 Glaxo Group Limited Immunoglobulins
WO2011000054A1 (en) 2009-07-03 2011-01-06 Avipep Pty Ltd Immuno-conjugates and methods for producing them
US8416206B2 (en) 2009-07-08 2013-04-09 Smart Technologies Ulc Method for manipulating a graphic widget in a three-dimensional environment displayed on a touch panel of an interactive input system
TWM383852U (en) 2009-07-13 2010-07-01 Speedtech Corp An improvement of the universal serial bus connector
JPWO2011043061A1 (en) 2009-10-05 2013-03-04 キヤノン株式会社 Contrast agent for photoacoustic imaging and photoacoustic imaging method using the same
EP3135302A1 (en) 2009-12-02 2017-03-01 Imaginab, Inc. J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use
WO2011075786A1 (en) 2009-12-23 2011-06-30 Avipep Pty Ltd Immuno-conjugates and methods for producing them 2
EA201492253A1 (en) 2009-12-29 2015-06-30 Эмерджент Продакт Дивелопмент Сиэтл, Ллс CONSTRUCTORS CONNECTING WITH RON AND METHODS OF THEIR USE
CN103237901B (en) 2010-03-01 2016-08-03 卡里斯生命科学瑞士控股有限责任公司 For treating the biomarker of diagnosis
US9812638B2 (en) 2010-03-19 2017-11-07 Globalfoundries Inc. Backend of line (BEOL) compatible high current density access device for high density arrays of electronic components

Patent Citations (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4454106A (en) * 1982-06-07 1984-06-12 Gansow Otto A Use of metal chelate conjugated monoclonal antibodies
US4472509A (en) * 1982-06-07 1984-09-18 Gansow Otto A Metal chelate conjugated monoclonal antibodies
US4814275A (en) * 1985-05-13 1989-03-21 E.I. Dupont De Nemours And Company Monoclonal hybridoma antibody specific for a human epithelial antigen
US6331175B1 (en) * 1985-07-05 2001-12-18 Immunomedics, Inc. Method and kit for imaging and treating organs and tissues
US5697902A (en) * 1985-07-05 1997-12-16 Immunomedics, Inc. Method for imaging and treating organs and tissues
US4863854A (en) * 1985-08-12 1989-09-05 Sloan-Kettering Institute For Cancer Research Monoclonal antibodies to mucin-like human differentiation antigens
US6143274A (en) * 1986-01-23 2000-11-07 Tweedle; Michael F. Method for imaging and radiopharmaceutical therapy using 1-substituted-4,7,10-tricarboxymethyl-1,4,7,10-tetraazacyclododecane and analogs
US5846519A (en) * 1986-01-23 1998-12-08 Bracco Diagnostics Inc. Method for imaging mammalian tissue using 1-substituted-1,4,7-tricarboxymethyl-1,4,7,10-tetraazacyclododecane and analogs
US5674470A (en) * 1986-01-23 1997-10-07 Bracco Diagnostics Inc. Method for imaging mammalian tissue using 1-substituted- 4,7,10-tricarboxymethyl-1,4,7,10-tetraazacyclododecane and analogs
US5474756A (en) * 1986-01-23 1995-12-12 Bracco International B.V. Method for imaging mammalian tissue using 1-substituted-1,4,7-tricarboxymethyl-1,4,7,10-tetraazacyclododecane and analogs
US4855353A (en) * 1986-02-14 1989-08-08 Nihon Medi-Physics Co., Ltd. High molecular compounds having amino groups, and their utilization
US4863851A (en) * 1986-10-20 1989-09-05 The Upjohn Company Monoclonal antibody to prostate secretory protein
US5057302A (en) * 1987-02-13 1991-10-15 Abbott Laboratories Bifunctional chelating agents
US5013645A (en) * 1987-04-14 1991-05-07 Abbott Laboratories Immunological methods and materials for detection of tumor associated antigens
US4885363A (en) * 1987-04-24 1989-12-05 E. R. Squibb & Sons, Inc. 1-substituted-1,4,7-triscarboxymethyl-1,4,7,10-tetraazacyclododecane and analogs
US5198208A (en) * 1987-07-16 1993-03-30 Nycomed Imaging As Aminopolycarboxylic acids and derivatives thereof
US5419893A (en) * 1987-07-16 1995-05-30 Nycomed Imaging As Aminopolycarboxylic acids and derivatives thereof for magnetic resonance imaging
US5531978A (en) * 1987-07-16 1996-07-02 Nycomed Imaging As Aminopolycarboxylic acids and derivatives thereof
US5217704A (en) * 1987-11-06 1993-06-08 Abbott Laboratories Methods and materials for the preparation of metal labelled antibody solutions
US5130118A (en) * 1987-11-06 1992-07-14 Abbott Laboratories Methods and materials for the preparation of metal labelled antibody solutions
US5342924A (en) * 1987-12-31 1994-08-30 Tanox Biosystems, Inc. Extracellular segments of human ε immunoglobulin anchoring peptides and antibodies specific therefor
US5208324A (en) * 1988-01-26 1993-05-04 Nycomed Imaging As Paramagnetic compounds
US5229289A (en) * 1988-03-11 1993-07-20 The Biomembrane Institute Monoclonal antibodies and vaccine development directed to human cancer-associated antigens by immunization with animal and human and with synthetic carbohydrate-carrier conjugates
US5162504A (en) * 1988-06-03 1992-11-10 Cytogen Corporation Monoclonal antibodies to a new antigenic marker in epithelial prostatic cells and serum of prostatic cancer patients
US5763202A (en) * 1988-06-03 1998-06-09 Cytogen Corporation Methods of detecting prostate carcinoma using a monoclonal antibody to a new antigenic marker in epithelial prostatic cells alone or with a monoclonal antibody to an antigen of LNCaP cells
US5578484A (en) * 1988-06-03 1996-11-26 Cytogen Corporation Hybridoma and monoclonal antibody 9H10-A4 specific to an antigen of LNCaP cells
US5118611A (en) * 1988-07-25 1992-06-02 Adeza Biomedical Corporation Adenocarcinoma antigen binding methods and reagents
US5565562A (en) * 1989-02-10 1996-10-15 Celltech Therapeutics Limited Triaza macrocycles
US5053503A (en) * 1989-02-17 1991-10-01 Centocor Chelating agents
US5314996A (en) * 1992-01-30 1994-05-24 Eastern Virginia Medical School Of Medical College Of Hampton Roads Isolated nucleotide sequences encoding an: antigen binding site of monoclonal antibody PD41; and antigen associated with prostate adenocarcinomas
US5227471A (en) * 1992-01-30 1993-07-13 Eastern Virginia Medical School Of The Medical College Of Hampton Roads Monoclonal antibody PD41 that binds to a prostate mucin antigen that is expressed in human prostatic carcinoma
US5877289A (en) * 1992-03-05 1999-03-02 The Scripps Research Institute Tissue factor compositions and ligands for the specific coagulation of vasculature
US5965132A (en) * 1992-03-05 1999-10-12 Board Of Regents, The University Of Texas System Methods and compositions for targeting the vasculature of solid tumors
US5776427A (en) * 1992-03-05 1998-07-07 Board Of Regents, The University Of Texas System Methods for targeting the vasculature of solid tumors
US6036955A (en) * 1992-03-05 2000-03-14 The Scripps Research Institute Kits and methods for the specific coagulation of vasculature
US6004555A (en) * 1992-03-05 1999-12-21 Board Of Regents, The University Of Texas System Methods for the specific coagulation of vasculature
US5855866A (en) * 1992-03-05 1999-01-05 Board Of Regenis, The University Of Texas System Methods for treating the vasculature of solid tumors
US5863538A (en) * 1992-03-05 1999-01-26 Board Of Regents, The University Of Texas System Compositions for targeting the vasculature of solid tumors
US6261535B1 (en) * 1992-03-05 2001-07-17 The University Of Texas System Board Of Regents Diagnostic methods for targeting the vasculature of solid tumors
US5660827A (en) * 1992-03-05 1997-08-26 Board Of Regents, The University Of Texas System Antibodies that bind to endoglin
US6093399A (en) * 1992-03-05 2000-07-25 Board Of Regents, The University Of Texas System Methods and compositions for the specific coagulation of vasculature
US6051230A (en) * 1992-03-05 2000-04-18 Board Of Regents, The University Of Texas System Compositions for targeting the vasculature of solid tumors
US6004554A (en) * 1992-03-05 1999-12-21 Board Of Regents, The University Of Texas System Methods for targeting the vasculature of solid tumors
US5489525A (en) * 1992-10-08 1996-02-06 The United States Of America As Represented By The Department Of Health And Human Services Monoclonal antibodies to prostate cells
US5538866A (en) * 1992-11-05 1996-07-23 Sloan-Kettering Institute For Cancer Research Prostate-specific membrane antigen
US5639879A (en) * 1993-04-02 1997-06-17 Associated Universities, Inc. Macrocyclic polyaminocarboxylates for stable radiometal antibody conjugates for therapy, spect and pet imaging
US5599677A (en) * 1993-12-29 1997-02-04 Abbott Laboratories Immunoassays for prostate specific antigen
US5935818A (en) * 1995-02-24 1999-08-10 Sloan-Kettering Institute For Cancer Research Isolated nucleic acid molecule encoding alternatively spliced prostate-specific membrane antigen and uses thereof
US6150508A (en) * 1996-03-25 2000-11-21 Northwest Biotherapeutics, Inc. Monoclonal antibodies specific for the extracellular domain of prostate-specific membrane antigen
US7163680B2 (en) * 1996-05-06 2007-01-16 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
US6136311A (en) * 1996-05-06 2000-10-24 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
US5804602A (en) * 1996-06-17 1998-09-08 Guilford Pharmaceuticals Inc. Methods of cancer treatment using naaladase inhibitors
US5958474A (en) * 1996-08-21 1999-09-28 Nestec S.A. Process for preparing food flavor precursors
US6022524A (en) * 1997-04-18 2000-02-08 Dibra S.P.A. Process for the conjugation of chelants with molecules containing amino groups
US6683162B2 (en) * 2000-09-15 2004-01-27 Sloan Kettering Institute Of Cancer Research Targeted alpha particle therapy using actinium-255 conjugates

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8114965B2 (en) 2001-10-23 2012-02-14 Psma Development Company, Llc Compositions of PSMA antibodies
US9695248B2 (en) 2001-10-23 2017-07-04 Psma Development Company, Llc PSMA antibodies and uses thereof
US8470330B2 (en) 2001-10-23 2013-06-25 Psma Development Company, Llc PSMA antibodies and uses thereof
US20040033229A1 (en) * 2001-10-23 2004-02-19 Maddon Paul J. PSMA antibodies and protein multimers
US7850971B2 (en) 2001-10-23 2010-12-14 Psma Development Company, Llc PSMA antibodies and protein multimers
US8461308B2 (en) 2005-02-18 2013-06-11 Medarex, Inc. Monoclonal antibodies against prostate specific membrane antigen (PSMA) lacking in fucosyl residues
US20110028696A1 (en) * 2005-02-18 2011-02-03 Medarex, Inc. Monoclonal antibodies against prostate specific membrane antigen (psma) lacking in fucosyl residues
US7875278B2 (en) 2005-02-18 2011-01-25 Medarex, Inc. Monoclonal antibodies against prostate specific membrane antigen (PSMA) lacking in fucosyl residues
US20090060908A1 (en) * 2005-02-18 2009-03-05 Medarex, Inc. Monoclonal Antibodies Against Prostate Specific Membrane Antigen (PSMA) Lacking in Fucosyl Residues
US8124738B2 (en) 2005-09-26 2012-02-28 Medarex, Inc. Human monoclonal antibodies to CD70
US20090028872A1 (en) * 2005-09-26 2009-01-29 Jonathan Alexander Terret Human monoclonal antibodies to cd70
US20100150950A1 (en) * 2006-12-14 2010-06-17 Medarex, Inc. Human antibodies that bind cd70 and uses thereof
EP2740490A1 (en) * 2007-10-03 2014-06-11 Cornell University Treatment of proliferative disorders using antibodies to PSMA
EP3714906A1 (en) 2007-10-03 2020-09-30 Cornell University Treatment of proliferative disorders using radiolabelled antibodies to psma
US9242012B2 (en) 2008-09-08 2016-01-26 Psma Development Company, Llc Methods for killing PSMA-expressing, taxane-resistant cancer cells

Also Published As

Publication number Publication date
US6649163B1 (en) 2003-11-18
US20150316553A1 (en) 2015-11-05
US20030031673A1 (en) 2003-02-13
US7163680B2 (en) 2007-01-16
US20090238755A1 (en) 2009-09-24
US8951737B2 (en) 2015-02-10
US6770450B1 (en) 2004-08-03
US6136311A (en) 2000-10-24

Similar Documents

Publication Publication Date Title
US8951737B2 (en) Treatment and diagnosis of cancer
US7112412B1 (en) Treatment and diagnosis of prostate cancer
US6767711B2 (en) Treatment and diagnosis of prostate cancer
WO1998003873A9 (en) Treatment and diagnosis of cancer
AU2006210589B2 (en) ADAM-9 modulators
US5525337A (en) Monoclonal antibody binding cell surface antigens for diagnosing cancer
US20030086929A1 (en) Treatment of prostate cancer by inhibitors of ATP synthase
JP2008508904A (en) Transferrin receptor antibody
KR101380057B1 (en) Diagnostic agent and therapeutic agent for pancreatic cancer
CA2391450A1 (en) Targeted delivery of therapeutic and diagnostic moieties
MXPA99000642A (en) Derivatized rodamine tint and its copolime

Legal Events

Date Code Title Description
AS Assignment

Owner name: CORNELL RESEARCH FOUNDATION, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BANDER, NEIL H.;REEL/FRAME:019870/0063

Effective date: 19970922

STCB Information on status: application discontinuation

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