US20030035804A1 - Drug complex for treatment of metastatic prostate cancer - Google Patents

Drug complex for treatment of metastatic prostate cancer Download PDF

Info

Publication number
US20030035804A1
US20030035804A1 US10/119,417 US11941702A US2003035804A1 US 20030035804 A1 US20030035804 A1 US 20030035804A1 US 11941702 A US11941702 A US 11941702A US 2003035804 A1 US2003035804 A1 US 2003035804A1
Authority
US
United States
Prior art keywords
drug
group
prostate cancer
peptide
drug complex
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
US10/119,417
Inventor
Anthony D'Amico
Glenn Bubley
David Jebaratnam
James Weinberg
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.)
Beth Israel Deaconess Medical Center Inc
Original Assignee
Beth Israel Deaconess Medical Center Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beth Israel Deaconess Medical Center Inc filed Critical Beth Israel Deaconess Medical Center Inc
Priority to US10/119,417 priority Critical patent/US20030035804A1/en
Assigned to BETH ISRAEL DEACONESS MEDICAL CENTER reassignment BETH ISRAEL DEACONESS MEDICAL CENTER ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BUBLEY, GLENN J.
Publication of US20030035804A1 publication Critical patent/US20030035804A1/en
Priority to US11/102,277 priority patent/US20050233948A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • 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
    • A61K47/54Medicinal 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 organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Definitions

  • Prostate cancer is the second most common malignancy in men and is the third most common cause of cancer death in men older than age 55.
  • Harrison's Principles of Internal Medicine Eds. J. D. Wilson et al., 11th Edition, pp. 1630-1633, McGraw Hill, New York, 1991.
  • New methods of identifying and treating prostate cancer early are extremely important in saving the lives of those afflicted.
  • Presently, only a limited number of treatments are available for treatment of prostate cancer. These treatments include surgery, radiation therapy, and androgen deprivation.
  • the present invention relates to a method of delivering a drug to a specific cell type, such as a cancer cell, in an individual by means of a drug complex which comprises three components and is also the subject of the present invention.
  • the three components of the drug complex include the following: a targeting carrier molecule; a linker which is acted upon by a molecule present at an effective concentration in the environs of the specific cell (referred to as a target cell); and a drug (or agent) to be delivered to the specific cell type.
  • the targeting carrier molecule is one which is delivered specifically to (has a biodistribution which favors) a specific cell type, or tissue containing the specific cell type, to which the drug is to be delivered.
  • the linker can be any type of molecule such as a peptide, provided that it is a substrate for a molecule, such as an enzyme (e.g., a protease), which is found in the environs of the target cell to which the drug is to be delivered and is present in sufficient quantities at the site(s) of the target cell to act upon the linker and release the drug or agent to be delivered, such as by cleaving the linker.
  • the linker can comprise more than one type of molecule.
  • the linker can comprise a component which is a substrate for a molecule which acts upon it and releases the drug to be delivered and an additional component(s), such as a molecule which acts as a good leaving group; additional amino acid residues or other molecule, such as one which acts as a spacer between the targeting carrier molecule and the drug to be delivered.
  • the drug or agent to be delivered can be any therapeutically or diagnostically useful drug or agent, such as a cytotoxic drug or an imaging agent. It remains essentially functionally inactive while it is a component of the drug complex; upon release at the site(s) of the target cells, the drug is functionally active (exerts or displays its therapeutic or diagnostic function(s)).
  • the linker comprises a substrate for a molecule as described previously, and one or more other additional components, such as chemical molecules, e.g. para-amino benzoic acid (PABA) and para-aminobenzyloxycarbonyl (pABOC), which are good leaving groups.
  • PABA para-amino benzoic acid
  • pABOC para-aminobenzyloxycarbonyl
  • amino acid residues there can be amino acid residues in addition to those acted upon by the enzyme or other molecule.
  • Such amino acid residues can be at either or both ends (amino and/or carboxy terminus) of the amino acid residues acted upon.
  • additional amino acid residue (or residues) is/are included between the carboxy terminal amino acid of the substrate and the drug or agent to be delivered.
  • amino acid residues can be, for example, one or more amino acid residues of the substrate (e.g., if the enzyme cleaves at an internal amino acid residue in the substrate), one or more amino acid residues which are not substrate amino acid residues (e.g., if the enzyme cleaves between the carboxyl terminal amino acid residue of the substrate and the amino terminal amino acid residue of additional amino acids) or both.
  • the additional amino acid residue(s) between the substrate carboxyl terminal amino acid residue and the drug or agent to be delivered to the target cell can be linked to the drug or agent in such a manner (e.g., through an amide bond, disulfide bond or ester bond) that cellular enzymes act upon them; resulting in release of the amino acid residue(s) attached to the drug or agent when it enters the cell.
  • the drug or agent to be delivered can be any therapeutically or diagnostically useful drug or agent, such as a cytotoxic drug or an imaging agent.
  • the present invention also relates to a method of delivering a drug to a specific cell type by means of the drug complex described herein.
  • the drug complex is administered to an individual in need of therapy or in whom a diagnostic procedure or assessment is to be conducted.
  • the present invention is a method of killing prostate cancer cells, including metastatic prostate cancer cells or androgen independent prostate cancer cells, in a man with prostate cancer.
  • the method is particularly effective because it makes use of a drug complex whose components provide the basis for localization to and killing prostate cancer cells, particularly androgen-independent cancer cells.
  • the method of killing metastatic prostate cancer cells comprises administering to the man a therapeutically effective amount of a drug complex which comprises three components: a targeting carrier molecule which is selectively delivered to prostate tissue, bone or both; a linker which comprises a component acted upon by prostate specific antigen (PSA) which is present in the microenvironment of malignant prostatic epithelial cells; and a cytotoxic drug which is toxic to prostatic cancer cells.
  • PSA prostate specific antigen
  • the present invention also relates to a method of treating metastatic cancer in an individual, in which the drug complex described above is administered to a man with prostate cancer, in a quantity sufficient to deliver the cytotoxic drug to prostate cancer cells at a concentration sufficient to kill some or all of the cancer cells or inhibit replication or division of the cancer cells.
  • Drug complexes useful in the method of treating prostate cancer and the method of killing prostate cancer cells are also the subject of this invention.
  • the drug complex comprises a targeting carrier molecule which is an imaging agent for prostate and prostate derived tumors, such as fluoropropylputrescine (e.g., N-3 fluoropropylputrescine); a peptide which serves as a linker and comprises amino acid residues acted upon by PSA; and a cytotoxic drug, which is linked to the targeting carrier molecule by the peptide.
  • the linker can be comprised of more than one type of molecule, as described herein.
  • the drug complex is administered to an individual by any route which results in delivery of the cytotoxic drug to prostate cells. In one embodiment, the drug complex is administered intravenously.
  • FIG. 1 is a graphic representation of growth delay of the human DU-145 prostate carcinoma xenograft after treatment with fractionated radiation therapy (RT) on days 7-11 or with thapsigargin (TG) on days 4-18.
  • RT fractionated radiation therapy
  • TG thapsigargin
  • FIG. 2 is a graphic representation of growth delay of the human DU-145 prostate carcinoma xenograft after treatment with fractionated RT on days 7-11 and 14-18 or with TG on days 4-18.
  • FIG. 3 is a schematic representation of one embodiment of a drug complex of the present invention, which comprises a targeting carrier molecule (“carrier”), a peptide linker (“peptide”), and a drug (“drug”); the peptide is a substrate for a molecule whose action at the cleavage site results in cleavage of the drug from the drug complex.
  • carrier a targeting carrier molecule
  • peptide a peptide linker
  • drug drug
  • FIG. 4 is a schematic representation of one embodiment of a drug complex of the present invention in which the carrier is a polyamine, the peptide is a PSA-specific peptide and the drug is a cytotoxic drug.
  • FIG. 5 is a schematic representation of a tyrosine-containing tripeptide (1) to which a succinyl ester group (MeO-Suc) is attached and of a para-nitroanilide (pNA) derivative of the tyrosine-containing peptide (2), with the PSA cleavage site indicated an arrow.
  • a succinyl ester group MeO-Suc
  • pNA para-nitroanilide
  • FIG. 6 is a schematic representation of a tyrosine-containing tripeptide with adriamycin attached directly to the tripeptide via adriamycin's NH 2 group; MeO-Suc represents a succinyl ester group attached to the tyrosine-containing tripeptide.
  • FIG. 7 is a schematic representation of one embodiment of the present invention in which the synthesized compound contains pABA.
  • FIG. 7 shows a model compound (4) that demonstrated that pABA is a good leaving group and that pABA can activate the tyrosine carbonyl for hydrolysis.
  • FIG. 7 further shows a compound (5) comprising a succinyl ester group (MeO-Suc) attached to a tripeptide (Arg-Pro-Tyr), which is attached to pABA, which is attached to adriamycin.
  • FIG. 7 shows a compound (6), which arises as a result of the cleavage of compound (5) by PSA.
  • FIG. 8 is a schematic representation of one embodiment of the present invention in which the synthesized compound contains pABOC.
  • FIG. 8 shows a compound (7) comprising a succinyl ester group (MeO-Suc) attached to a tripeptide (Arg-Pro-Tyr), which is attached to pABOC, which is attached to adriamycin.
  • FIG. 8 further shows how the fluorine-containing pABOC, being electron-withdrawing, activates the tyrosine carbonyl toward hydrolysis (8).
  • FIG. 8 shows how the spontaneous breakdown of 8, results in the release of “native” adriamycin.
  • the present invention relates to a method of selectively delivering a drug or other agent to a particular cell type by means of a drug complex which includes at least three components: a targeting carrier molecule which is selectively distributed to the cell type or tissue to which the drug or agent is to be delivered; a linker which is acted upon (cleaved or otherwise broken or disrupted) by a molecule present at the site to which the drug or agent is to be delivered; and a drug or agent to be delivered.
  • the invention also relates to the drug complex, which, optionally, includes additional components.
  • the drug complex is administered via an appropriate route to an individual in need of the drug or agent.
  • the drug complex moves to the cells or tissues to which the targeting carrier molecule is specifically delivered.
  • the linker is cleaved by the molecule present at the site to which the drug or agent is to be delivered and the drug or agent is made available to enter into or otherwise interact with the specific cell type.
  • the location(s) in the body at which the tissue or cell type(s) to which a drug or agent is to be delivered are referred to as site(s) of the target cell(s).
  • the invention is a method of treating prostate cancer by killing prostate cancer cells or inhibiting replication or division of prostate cancer cells, such as androgen independent prostate cancer cells.
  • the drug complex comprises three components: a targeting carrier molecule which is any agent or molecule which is selectively delivered to prostate, bone or both; a linker which is cleaved by PSA; and a drug which is cytotoxic to cancer cells, particularly androgen independent prostatic cancer cells.
  • the linker may be comprised of more than one type of molecule.
  • the linker may comprise a substrate for a molecule as described previously, and may also include one or more other molecules, such as one or more desirable chemical molecules, e.g. para-amino benzoic acid (pABA) and para-aminobenzyloxycarbonyl (pABOC).
  • a selectively delivered drug is one which is delivered or distributed to the targeted cell or tissue to an extent sufficient for delivery of the drug or agent, such as a cytotoxic drug or agent, in a quantity or concentration sufficient to have the desired therapeutic or diagnostic effects.
  • the desired effect is killing of some or all of the cancer cells (e.g., prostatic cancer cells) in an individual or at least a reduction in replication or division of the cancerous cells.
  • the targeting carrier molecule generally will be a molecule which is distributed to a limited number of sites within the body, but it is not necessary that it be delivered only to those sites.
  • the targeting carrier molecule can be an imaging agent, such as N-3 fluoropropylputrescine which is selectively distributed to prostate and bone, relative to blood and muscle, after administration to a man.
  • the targeting carrier molecule is a polyamine (e.g., putrescine, spermine or spermidine) because of its affinity to prostatic cancer cells through the polyamine surface receptor molecules present on prostatic epithelial cells.
  • the linker which is acted upon by a molecule which is present at an effective concentration in the environs of the specific cell, may be any entity which is acted upon by a molecule, such as an enzyme, at or within a given target cell. Acted upon is defined as any change in the linker induced by the molecule at or within the given target cell which results in release of the drug.
  • the linker comprises amino acid residues which are acted upon by PSA.
  • the linker used comprises at least those amino acid residues necessary for PSA to act, resulting in cleavage of the linker and release of the cytotoxic drug.
  • the linker may comprise additional amino acid residues, which may be the same as those in the PSA peptide substrate or different (e.g., they can be random or filler amino acid residues, amino acid residues which enhance uptake of the cytotoxic drug into the prostatic cancer cell, amino acid residues which stabilize the cytotoxic drug or enhance its resistance to degradation once inside the cell or amino acid residues which are substrates for cellular enzymes).
  • the linker can comprise more than one type of molecule.
  • the linker may comprise amino acid residues acted upon by PSA as described previously, and may also include one or more other molecules, such as one or more desirable chemical molecules, e.g. para-amino benzoic acid (PABA) and para-aminobenzyloxycarbonyl (pABOC).
  • PABA para-amino benzoic acid
  • pABOC para-aminobenzyloxycarbonyl
  • Examples of possible amino acids that can be used in the subject invention include the following combinations of amino acid residues, as represented in Table 1 of Denmeade et al., Cancer Research 57: 4924-4930 (1997): KGISSQY (SEQ ID NO.: 1); SRKSQQY (SEQ ID NO.: 2); GQKGQHY (SEQ ID NO.: 3); EHSSKLQ (SEQ ID NO.: 4); QNKISYQ (SEQ ID NO.: 5); ENKISYQ (SEQ ID NO.: 6); ATKSKQH (SEQ ID NO.: 7); KGLSSQC (SEQ ID NO.: 8); LGGSQQL (SEQ ID NO.: 9); QNKGHYQ (SEQ ID NO.: 10); TEERQLH (SEQ ID NO.: 11); GSFSIQH (SEQ ID NO.: 12); HSSKLQ (SEQ ID NO.: 13); SKLQ (SEQ ID NO.: 14); KLQ
  • AKFE SEQ ID NO. 17
  • the peptide link between the targeting carrier molecule and the cytotoxic drug will be sustained because there is insufficient unbound PSA to act to a significant extent on the linking peptide that is substrate for unbound PSA.
  • PSA will cleave the linking peptide to a great extent and thereby release the cytotoxic drug into the cell.
  • the targeting carrier molecule is highly target specific for the androgen independent prostate cancer cell. Such a highly target specific targeting carrier molecule will increase the efficacy of and decrease the toxicity of the cytotoxic drug component used.
  • Targeting molecules can be, for example, polyamines, e.g., putrescine, spermine or spermidine, which provide affinity to prostatic cancer cells through the polyamine surface receptor molecules present on prostatic epithelial cells.
  • a specific example of such a targeting carrier molecule is N-3 fluoropropylputrescine. (Hwang, D. et al., J. Nucl. Med., 30:1205-1210 (1989)).
  • the cytotoxic drug which is a component of the drug complex can be any agent effective in killing prostate cancer cells, including metastatic prostate cancer cells.
  • any of the following drugs can be used: adriamycin, amonafide, cisplatin, carboplatin (CBDCA), CHIP, cyclophosphamide, doxorubicin, epirubicin, estramustine, etoposide, 5-fluorouracil, gallium nitrate, idarubicin, ifosfamide+mesna, ketoconazole, liarozole (R85,246), methotrexate, mitomycin C, mitoguazone, mitoxantrone, proscar (finasteride), suramin, taxol, thapsigargin, trimetrexate, vinblastine (CI), and vinblastine and emcyt.
  • Adriamycin is particularly desirable for several reasons. First, it gives a response rate of 15-20% in hormone refractory metastatic prostate cancer with a median survival of 33 weeks. These results are among the best noted for single agents in this disease. (Kreis, W., Cancer Investigation, 13:296-312 (1996)). Second, the chemistry is well established. For example, adriamycin can be easily coupled to peptides through its amino (—NH 2 ) group. (Nogusa, H. et al., Chem. Pharm. Bull. Jpn., 43:1931-1936 (1995). Moreover, several of its analogs, including N-substituted ones, are known to display high anticancer activity. (Israel, A. et al., Cancer Treatment Reviews, 14:163-167 (1987).
  • Thapsigargin is a sesquiterpene lactone extracted from the roots of the umbelliferous plant Thapsia garganica L. (Thastrup, O. et al., Proc. Nat. Acad. Sci., USA, 87:2466-2470 (1990)).
  • This highly lipophilic agent specifically inhibits Ca ++ -ATPase pumps of the endoplasmic reticulum, but not the pumps of erythrocytes, plasma, or mitochondrial membranes.
  • the dose of the drug complex to be used is a sufficient quantity of complex to result in delivery and availability to target cells of the cytotoxic drug at a therapeutically effective level. Dose will be determined empirically and will be determined, for example, by the stage or condition of the disease for which an individual is being treated, the individual's general health, size, age, and sex. In the embodiment of the present invention in which a man is treated for prostate cancer, the dose will be determined by taking into consideration the cytotoxic drug being used, the stage of the cancer, and the man's age, general health and size.
  • the timing and number of doses of the drug complex administered will also be determined empirically.
  • the number of doses may be at any interval sufficient to promote or to result in killing of cells and/or inhibition of replication or division.
  • the drug complex can be administered hourly, daily, weekly, monthly, or any combination thereof.
  • the route of administration can be any route that delivers a therapeutically effective quantity of the drug complex to the target cells, such as to prostate cancer cells.
  • the formulation of the drug complex can be any pharmaceutically effective formulation or carrier, such as any physiologically acceptable buffer, saline solution, or water.
  • the prodrug of the invention has the structure shown below, and is believed to have a prostate specific antigen (PSA) cleavage site as indicated.
  • PSA prostate specific antigen
  • R represents a polyarnine attached through the amino group of the N-terminal amino acid Histidine
  • Adriamycin indicates attachment of Adriamycin (structure given below) through its amino group.
  • PC3 and DU-145 androgen-independent human prostate cancer cell lines obtained from the American Tissue Culture Collection (ATCC) are used for in vitro studies.
  • Cells are grown as monolayer cultures in RPMI 1640 medium supplemented with 10% fetal bovine serum, 2 mM glutamine and antibiotics. Radiation survival curves and radiation-induced apoptotic DNA fragmentation patterns in these cell lines are already established in the laboratory. Cells are treated with this drug, carrier-peptide, or carrier-peptide-drug complex with or without radiation. After 24 hours, the cells are trypsinized and plated for clonogenic survival studies.
  • Apoptotic response to the treatments is determined by: 1) agarose gel electrophoresis analysis of DNA fragmentation at 48 and 72 hours after treatment, 2) morphological observations after staining with DAPI, and 3) terminal deoxytransferase-mediated dUTP Nick End Labeling (TUNEL) assay.
  • Two human prostate carcinoma tumor lines grown as xenografts in male SCID mice are used: the human DU-145 prostate carcinoma which is not androgen dependent and the human LNCaP prostate carcinoma which produces PSA and has a well characterized androgen receptor and response to androgens.
  • the drug complexes are administered up to maximally tolerated doses alone and in conjunction with fractionated radiation therapy ( 137 Cs Gamma Cell 40) delivered to the tumor bearing limb.
  • the progress of each tumor is assessed thrice weekly by caliper measurements until the tumors reach 2000 mm 3 .
  • Tumor growth delay is calculated as the number of days for each tumor to reach a volume of 500 mm 3 as compared to untreated controls.
  • the efficacy of combination treatments is assessed using isobologram analysis for determination of additivity/synergy.
  • a drug complex is chosen for testing in ten patients with hormone refractory metastatic prostate cancer.
  • a single intravenous dose of the drug complex is given on an inpatient basis with cardiorespiratory monitoring.
  • Toxicity evaluation of all major organ systems is evaluated using blood, urine, stool, and, if necessary, bone marrow studies. Doses are escalated in 10% increments per patient until the maximal response with acceptable toxicity is reached. Response is assessed using a combination of a weekly digital rectal examination (DRE), PSA, LDH, hemoglobin, and monthly bone scan.
  • the starting dose is selected on the toxicity data from the preceding animal studies.
  • Table 1 shows the effect of thapsigargin and radiotherapy on proliferation of DU-145 prostate cancer cells. The results indicate that thapsigargin inhibits cell proliferation in vitro. TABLE 1 Effect of TG and RT on proliferation of DU-145 prostate cancer cells* Total Number of Cells per Thapsigargin Dish (10 6 ) (nM) 0 Gy 2 Gy 4 Gy 8 Gy 0 2.1 1.4 1.2 0.5 20 1.0 0.8 0.3 0.4 100 0.7 0.5 0.5 0.4 400 0.7 0.4 0.3 0.3 0.3 0.3 0.3 0.3
  • Table 2 shows the effect of thapsigargin and radiotherapy on clonogenic cell survival of PC3 prostate cancer cells. The results indicate that thapsigargin inhibits clonogenic survival in vitro.
  • TABLE 2 Effect of TG and RT on clonogenic survival of PC3 cells**
  • Surviving Fraction (SF) NET survival
  • Thapsigargin (PE treated/PE control) (SF/TG toxicity) (nM) 0 Gy 2 Gy 4 Gy 2 Gy 4 Gy 0 1 (0.8) 0.57 0.38 0.57 0.38 20 0.69 0.42 0.23 0.61 0.33 50 0.80 0.39 0.23 0.49 0.29 100 0.62 0.25 0.18 0.40 0.30 200 0.58 0.25 0.13 0.43 0.22 400 0.60 0.25 0.13 0.42 0.22
  • mice bearing the human DU-145 prostate carcinoma Xenograft growing subcutaneously in a hind-limb were treated daily with TG (0.5 mg/kg) by IP injection from day 4-18 post tumor cell implantation.
  • Some animal groups also received fractionated RT locally to the tumor bearing region on days 7-11 or days 7-11 and 14-18.
  • FIG. 1 low dose TG (0.5 mg/kg) administration produced a measurable tumor growth delay and, when given in conjunction with RT, the independent effects of TG and RT were additive.
  • the RT regimen was extended to 2 weeks, the additional benefit of the TG administration was maintained.
  • a tyrosine tripeptide (compound 1 in FIG. 5) was prepared using methods known to those in the peptide chemistry art. It is known that PSA cleaves its para-nitroanilide (pNA) derivative at the tyrosine site (compound 2 in FIG. 5). (Christensson, A. et al., Eur. J. Biochem., 194:755-763 (1990)). It is also known that the “released” pNA makes it possible to study this reaction colormetrically. Using this feature, it was demonstrated that the sonicate from a cell line that is known to produce PSA, i.e. LNCaP, cleaves compound 2 in FIG. 5.
  • pNA para-nitroanilide
  • the pNA group was replaced with adriamycin, i.e. by attaching adriamycin directly to the tyrosine (FIG. 6) via adriamycin's —NH 2 group. It was hypothesized that PSA would cleave the molecule of FIG. 6 at the tyrosine site and release adriamycin. However, there was no cleavage and no release of adriamycin.
  • adriamycin to a tripeptide molecule (see compound 1 of FIG. 5) through a para-amino benzoic acid can solve the problems previously discussed for the following reasons: (a) pABA can function as a good leaving group because it contains an electron withdrawing carbonyl group in the para-position, (b) because pABA is electron-withdrawing, it can activate the tyrosine carbonyl hydrolysis, and (c) PSA activity may not be sterically hindered as the pABA linker can keep adriamycin away from the tyrosine cleavage site.
  • Cleavage of compound 5 can be assessed using the sonicate of LNCaP that exhibits “PSA-like” activity. It is expected that cleavage will occur at the tyrosine site of compound 5. However, what will be “released” is an analog of adriamycin (compound 6 of FIG. 7) and not adriamycin itself. The ability of this analog to intercalate DNA can be assessed and compared with that of native adriamycin using known methods, such as measurement of emissions at 450 nm on ethanol-precipitated DNA. The data is also normalized for efficiency of precipitation by assaying the DNA at 260 nm.

Abstract

A drug complex for delivery of a drug or other agent to a target cell, comprising a targeting carrier molecule which is selectively distributed to a specific cell type or tissue containing the specific cell type; a linker which is acted upon by a molecule which is present at an effective concentration in the environs of the specific cell type; and a drug or an agent to be delivered to the specific cell type. In particular, a drug complex for delivering a cytotoxic drug to prostate cancer cells, comprising a targeting carrier molecule which is selectively delivered to prostate tissue, bone or both; a peptide which is a substrate for prostate specific antigen; and a cytotoxic drug which is toxic to androgen independent prostate cancer cells.

Description

    RELATED APPLICATION
  • This application is a divisional of U.S. application Ser. No. 09/110,822 filed Jul. 6, 1998, which is a continuation-in-part of application Ser. No. 09/003,838 filed Jan. 7, 1998, which is a continuation of application Ser. No. 08/713,114 filed Sep. 16, 1996, now abandoned, the teachings of which are incorporated herein in their entirety.[0001]
  • BACKGROUND OF THE INVENTION
  • Prostate cancer is the second most common malignancy in men and is the third most common cause of cancer death in men older than age 55. [0002] Harrison's Principles of Internal Medicine, Eds. J. D. Wilson et al., 11th Edition, pp. 1630-1633, McGraw Hill, New York, 1991. New methods of identifying and treating prostate cancer early are extremely important in saving the lives of those afflicted. Presently, only a limited number of treatments are available for treatment of prostate cancer. These treatments include surgery, radiation therapy, and androgen deprivation.
  • There is a need for additional methods of treatment of prostate cancer. [0003]
  • SUMMARY OF THE INVENTION
  • The present invention relates to a method of delivering a drug to a specific cell type, such as a cancer cell, in an individual by means of a drug complex which comprises three components and is also the subject of the present invention. The three components of the drug complex include the following: a targeting carrier molecule; a linker which is acted upon by a molecule present at an effective concentration in the environs of the specific cell (referred to as a target cell); and a drug (or agent) to be delivered to the specific cell type. The targeting carrier molecule is one which is delivered specifically to (has a biodistribution which favors) a specific cell type, or tissue containing the specific cell type, to which the drug is to be delivered. The linker can be any type of molecule such as a peptide, provided that it is a substrate for a molecule, such as an enzyme (e.g., a protease), which is found in the environs of the target cell to which the drug is to be delivered and is present in sufficient quantities at the site(s) of the target cell to act upon the linker and release the drug or agent to be delivered, such as by cleaving the linker. The linker can comprise more than one type of molecule. For example, the linker can comprise a component which is a substrate for a molecule which acts upon it and releases the drug to be delivered and an additional component(s), such as a molecule which acts as a good leaving group; additional amino acid residues or other molecule, such as one which acts as a spacer between the targeting carrier molecule and the drug to be delivered. The drug or agent to be delivered can be any therapeutically or diagnostically useful drug or agent, such as a cytotoxic drug or an imaging agent. It remains essentially functionally inactive while it is a component of the drug complex; upon release at the site(s) of the target cells, the drug is functionally active (exerts or displays its therapeutic or diagnostic function(s)). In one embodiment, the linker comprises a substrate for a molecule as described previously, and one or more other additional components, such as chemical molecules, e.g. para-amino benzoic acid (PABA) and para-aminobenzyloxycarbonyl (pABOC), which are good leaving groups. Optionally, there can be amino acid residues in addition to those acted upon by the enzyme or other molecule. Such amino acid residues can be at either or both ends (amino and/or carboxy terminus) of the amino acid residues acted upon. In one embodiment, additional amino acid residue (or residues) is/are included between the carboxy terminal amino acid of the substrate and the drug or agent to be delivered. Cleavage of the substrate results in a product in which an amino acid residue(s) is attached to the drug or agent delivered to the target cell. Such amino acid residues can be, for example, one or more amino acid residues of the substrate (e.g., if the enzyme cleaves at an internal amino acid residue in the substrate), one or more amino acid residues which are not substrate amino acid residues (e.g., if the enzyme cleaves between the carboxyl terminal amino acid residue of the substrate and the amino terminal amino acid residue of additional amino acids) or both. The additional amino acid residue(s) between the substrate carboxyl terminal amino acid residue and the drug or agent to be delivered to the target cell can be linked to the drug or agent in such a manner (e.g., through an amide bond, disulfide bond or ester bond) that cellular enzymes act upon them; resulting in release of the amino acid residue(s) attached to the drug or agent when it enters the cell. The drug or agent to be delivered can be any therapeutically or diagnostically useful drug or agent, such as a cytotoxic drug or an imaging agent. [0004]
  • The present invention also relates to a method of delivering a drug to a specific cell type by means of the drug complex described herein. In the method, the drug complex is administered to an individual in need of therapy or in whom a diagnostic procedure or assessment is to be conducted. [0005]
  • In one embodiment, the present invention is a method of killing prostate cancer cells, including metastatic prostate cancer cells or androgen independent prostate cancer cells, in a man with prostate cancer. The method is particularly effective because it makes use of a drug complex whose components provide the basis for localization to and killing prostate cancer cells, particularly androgen-independent cancer cells. The method of killing metastatic prostate cancer cells comprises administering to the man a therapeutically effective amount of a drug complex which comprises three components: a targeting carrier molecule which is selectively delivered to prostate tissue, bone or both; a linker which comprises a component acted upon by prostate specific antigen (PSA) which is present in the microenvironment of malignant prostatic epithelial cells; and a cytotoxic drug which is toxic to prostatic cancer cells. The present invention also relates to a method of treating metastatic cancer in an individual, in which the drug complex described above is administered to a man with prostate cancer, in a quantity sufficient to deliver the cytotoxic drug to prostate cancer cells at a concentration sufficient to kill some or all of the cancer cells or inhibit replication or division of the cancer cells. Drug complexes useful in the method of treating prostate cancer and the method of killing prostate cancer cells (such as androgen independent cells) are also the subject of this invention. [0006]
  • In a further embodiment, the drug complex comprises a targeting carrier molecule which is an imaging agent for prostate and prostate derived tumors, such as fluoropropylputrescine (e.g., N-3 fluoropropylputrescine); a peptide which serves as a linker and comprises amino acid residues acted upon by PSA; and a cytotoxic drug, which is linked to the targeting carrier molecule by the peptide. The linker can be comprised of more than one type of molecule, as described herein. The drug complex is administered to an individual by any route which results in delivery of the cytotoxic drug to prostate cells. In one embodiment, the drug complex is administered intravenously. [0007]
  • Combining a targeting carrier molecule with specificity for prostate tissue, bone or both with a cytotoxic drug, which are joined by a linking peptide that is a substrate for PSA, provides a treatment for cancer having higher efficacy and lower toxicity than presently available treatments.[0008]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a graphic representation of growth delay of the human DU-145 prostate carcinoma xenograft after treatment with fractionated radiation therapy (RT) on days 7-11 or with thapsigargin (TG) on days 4-18. [0009]
  • FIG. 2 is a graphic representation of growth delay of the human DU-145 prostate carcinoma xenograft after treatment with fractionated RT on days 7-11 and 14-18 or with TG on days 4-18. [0010]
  • FIG. 3 is a schematic representation of one embodiment of a drug complex of the present invention, which comprises a targeting carrier molecule (“carrier”), a peptide linker (“peptide”), and a drug (“drug”); the peptide is a substrate for a molecule whose action at the cleavage site results in cleavage of the drug from the drug complex. [0011]
  • FIG. 4 is a schematic representation of one embodiment of a drug complex of the present invention in which the carrier is a polyamine, the peptide is a PSA-specific peptide and the drug is a cytotoxic drug. [0012]
  • FIG. 5 is a schematic representation of a tyrosine-containing tripeptide (1) to which a succinyl ester group (MeO-Suc) is attached and of a para-nitroanilide (pNA) derivative of the tyrosine-containing peptide (2), with the PSA cleavage site indicated an arrow. [0013]
  • FIG. 6 is a schematic representation of a tyrosine-containing tripeptide with adriamycin attached directly to the tripeptide via adriamycin's NH[0014] 2 group; MeO-Suc represents a succinyl ester group attached to the tyrosine-containing tripeptide.
  • FIG. 7 is a schematic representation of one embodiment of the present invention in which the synthesized compound contains pABA. FIG. 7 shows a model compound (4) that demonstrated that pABA is a good leaving group and that pABA can activate the tyrosine carbonyl for hydrolysis. FIG. 7 further shows a compound (5) comprising a succinyl ester group (MeO-Suc) attached to a tripeptide (Arg-Pro-Tyr), which is attached to pABA, which is attached to adriamycin. Finally, FIG. 7 shows a compound (6), which arises as a result of the cleavage of compound (5) by PSA. [0015]
  • FIG. 8 is a schematic representation of one embodiment of the present invention in which the synthesized compound contains pABOC. FIG. 8 shows a compound (7) comprising a succinyl ester group (MeO-Suc) attached to a tripeptide (Arg-Pro-Tyr), which is attached to pABOC, which is attached to adriamycin. FIG. 8 further shows how the fluorine-containing pABOC, being electron-withdrawing, activates the tyrosine carbonyl toward hydrolysis (8). Finally, FIG. 8 shows how the spontaneous breakdown of 8, results in the release of “native” adriamycin.[0016]
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to a method of selectively delivering a drug or other agent to a particular cell type by means of a drug complex which includes at least three components: a targeting carrier molecule which is selectively distributed to the cell type or tissue to which the drug or agent is to be delivered; a linker which is acted upon (cleaved or otherwise broken or disrupted) by a molecule present at the site to which the drug or agent is to be delivered; and a drug or agent to be delivered. The invention also relates to the drug complex, which, optionally, includes additional components. In the method, the drug complex is administered via an appropriate route to an individual in need of the drug or agent. The drug complex moves to the cells or tissues to which the targeting carrier molecule is specifically delivered. The linker is cleaved by the molecule present at the site to which the drug or agent is to be delivered and the drug or agent is made available to enter into or otherwise interact with the specific cell type. The location(s) in the body at which the tissue or cell type(s) to which a drug or agent is to be delivered are referred to as site(s) of the target cell(s). [0017]
  • In a particular embodiment, the invention is a method of treating prostate cancer by killing prostate cancer cells or inhibiting replication or division of prostate cancer cells, such as androgen independent prostate cancer cells. In this embodiment, the drug complex comprises three components: a targeting carrier molecule which is any agent or molecule which is selectively delivered to prostate, bone or both; a linker which is cleaved by PSA; and a drug which is cytotoxic to cancer cells, particularly androgen independent prostatic cancer cells. Additionally, the linker may be comprised of more than one type of molecule. For example, the linker may comprise a substrate for a molecule as described previously, and may also include one or more other molecules, such as one or more desirable chemical molecules, e.g. para-amino benzoic acid (pABA) and para-aminobenzyloxycarbonyl (pABOC). [0018]
  • A selectively delivered drug is one which is delivered or distributed to the targeted cell or tissue to an extent sufficient for delivery of the drug or agent, such as a cytotoxic drug or agent, in a quantity or concentration sufficient to have the desired therapeutic or diagnostic effects. In the case of treatment of cancer, such as the treatment of prostate cancer, the desired effect is killing of some or all of the cancer cells (e.g., prostatic cancer cells) in an individual or at least a reduction in replication or division of the cancerous cells. In other words, the targeting carrier molecule generally will be a molecule which is distributed to a limited number of sites within the body, but it is not necessary that it be delivered only to those sites. For example, the targeting carrier molecule can be an imaging agent, such as N-3 fluoropropylputrescine which is selectively distributed to prostate and bone, relative to blood and muscle, after administration to a man. In the embodiment in which prostate cancer is treated, the targeting carrier molecule is a polyamine (e.g., putrescine, spermine or spermidine) because of its affinity to prostatic cancer cells through the polyamine surface receptor molecules present on prostatic epithelial cells. [0019]
  • The linker, which is acted upon by a molecule which is present at an effective concentration in the environs of the specific cell, may be any entity which is acted upon by a molecule, such as an enzyme, at or within a given target cell. Acted upon is defined as any change in the linker induced by the molecule at or within the given target cell which results in release of the drug. [0020]
  • In the embodiment in which the drug complex is used to treat prostate cancer, the linker comprises amino acid residues which are acted upon by PSA. The linker used comprises at least those amino acid residues necessary for PSA to act, resulting in cleavage of the linker and release of the cytotoxic drug. The linker may comprise additional amino acid residues, which may be the same as those in the PSA peptide substrate or different (e.g., they can be random or filler amino acid residues, amino acid residues which enhance uptake of the cytotoxic drug into the prostatic cancer cell, amino acid residues which stabilize the cytotoxic drug or enhance its resistance to degradation once inside the cell or amino acid residues which are substrates for cellular enzymes). Optionally, the linker can comprise more than one type of molecule. For example, the linker may comprise amino acid residues acted upon by PSA as described previously, and may also include one or more other molecules, such as one or more desirable chemical molecules, e.g. para-amino benzoic acid (PABA) and para-aminobenzyloxycarbonyl (pABOC). [0021]
  • Examples of possible amino acids that can be used in the subject invention include the following combinations of amino acid residues, as represented in Table 1 of Denmeade et al., [0022] Cancer Research 57: 4924-4930 (1997): KGISSQY (SEQ ID NO.: 1); SRKSQQY (SEQ ID NO.: 2); GQKGQHY (SEQ ID NO.: 3); EHSSKLQ (SEQ ID NO.: 4); QNKISYQ (SEQ ID NO.: 5); ENKISYQ (SEQ ID NO.: 6); ATKSKQH (SEQ ID NO.: 7); KGLSSQC (SEQ ID NO.: 8); LGGSQQL (SEQ ID NO.: 9); QNKGHYQ (SEQ ID NO.: 10); TEERQLH (SEQ ID NO.: 11); GSFSIQH (SEQ ID NO.: 12); HSSKLQ (SEQ ID NO.: 13); SKLQ (SEQ ID NO.: 14); KLQ (SEQ ID NO.: 15) and LQ (SEQ ID NO.: 16).
  • Another example of a possible combination of amino acids that can be used in the subject invention is AKFE (SEQ ID NO.: 17). [0023]
  • In the bloodstream, the peptide link between the targeting carrier molecule and the cytotoxic drug will be sustained because there is insufficient unbound PSA to act to a significant extent on the linking peptide that is substrate for unbound PSA. In the microenvironment of the androgen independent prostate cancer cells, where unbound PSA is found in large concentrations, PSA will cleave the linking peptide to a great extent and thereby release the cytotoxic drug into the cell. By having the features of specificity for prostate and bone and the required presence of unbound PSA for drug release, the targeting carrier molecule is highly target specific for the androgen independent prostate cancer cell. Such a highly target specific targeting carrier molecule will increase the efficacy of and decrease the toxicity of the cytotoxic drug component used. Targeting molecules can be, for example, polyamines, e.g., putrescine, spermine or spermidine, which provide affinity to prostatic cancer cells through the polyamine surface receptor molecules present on prostatic epithelial cells. A specific example of such a targeting carrier molecule is N-3 fluoropropylputrescine. (Hwang, D. et al., [0024] J. Nucl. Med., 30:1205-1210 (1989)).
  • The cytotoxic drug which is a component of the drug complex can be any agent effective in killing prostate cancer cells, including metastatic prostate cancer cells. For example, any of the following drugs can be used: adriamycin, amonafide, cisplatin, carboplatin (CBDCA), CHIP, cyclophosphamide, doxorubicin, epirubicin, estramustine, etoposide, 5-fluorouracil, gallium nitrate, idarubicin, ifosfamide+mesna, ketoconazole, liarozole (R85,246), methotrexate, mitomycin C, mitoguazone, mitoxantrone, proscar (finasteride), suramin, taxol, thapsigargin, trimetrexate, vinblastine (CI), and vinblastine and emcyt. [0025]
  • Adriamycin is particularly desirable for several reasons. First, it gives a response rate of 15-20% in hormone refractory metastatic prostate cancer with a median survival of 33 weeks. These results are among the best noted for single agents in this disease. (Kreis, W., [0026] Cancer Investigation, 13:296-312 (1996)). Second, the chemistry is well established. For example, adriamycin can be easily coupled to peptides through its amino (—NH2) group. (Nogusa, H. et al., Chem. Pharm. Bull. Jpn., 43:1931-1936 (1995). Moreover, several of its analogs, including N-substituted ones, are known to display high anticancer activity. (Israel, A. et al., Cancer Treatment Reviews, 14:163-167 (1987).
  • Another cytotoxic drug, Thapsigargin, is a sesquiterpene lactone extracted from the roots of the umbelliferous plant [0027] Thapsia garganica L. (Thastrup, O. et al., Proc. Nat. Acad. Sci., USA, 87:2466-2470 (1990)). This highly lipophilic agent specifically inhibits Ca++-ATPase pumps of the endoplasmic reticulum, but not the pumps of erythrocytes, plasma, or mitochondrial membranes.
  • In vitro treatment with 500 nm thapsigargin in a series of both rat and human androgen independent prostate cancer cell lines, which either express or completely lack p53 protein expression, results in the elevation of intracellular Ca[0028] ++. Within 72 to 96 hours, these cells undergo apoptosis and lose their clonogenic potential.
  • The dose of the drug complex to be used is a sufficient quantity of complex to result in delivery and availability to target cells of the cytotoxic drug at a therapeutically effective level. Dose will be determined empirically and will be determined, for example, by the stage or condition of the disease for which an individual is being treated, the individual's general health, size, age, and sex. In the embodiment of the present invention in which a man is treated for prostate cancer, the dose will be determined by taking into consideration the cytotoxic drug being used, the stage of the cancer, and the man's age, general health and size. [0029]
  • The timing and number of doses of the drug complex administered will also be determined empirically. The number of doses may be at any interval sufficient to promote or to result in killing of cells and/or inhibition of replication or division. For example, the drug complex can be administered hourly, daily, weekly, monthly, or any combination thereof. [0030]
  • The route of administration can be any route that delivers a therapeutically effective quantity of the drug complex to the target cells, such as to prostate cancer cells. The formulation of the drug complex can be any pharmaceutically effective formulation or carrier, such as any physiologically acceptable buffer, saline solution, or water. [0031]
  • In one embodiment, the prodrug of the invention has the structure shown below, and is believed to have a prostate specific antigen (PSA) cleavage site as indicated. [0032]
    Figure US20030035804A1-20030220-C00001
  • Where R represents a polyarnine attached through the amino group of the N-terminal amino acid Histidine, and [0033]
    Figure US20030035804A1-20030220-C00002
  • Adriamycin, indicates attachment of Adriamycin (structure given below) through its amino group. [0034]
    Figure US20030035804A1-20030220-C00003
  • One method of forming this prodrug is shown below: [0035]
    Figure US20030035804A1-20030220-C00004
  • The present invention is illustrated by the following examples, which are not intended to be limiting in any way. [0036]
  • EXAMPLE 1
  • In Vitro Testing of Drug Complexes [0037]
  • PC3 and DU-145 androgen-independent human prostate cancer cell lines obtained from the American Tissue Culture Collection (ATCC) are used for in vitro studies. Cells are grown as monolayer cultures in RPMI 1640 medium supplemented with 10% fetal bovine serum, 2 mM glutamine and antibiotics. Radiation survival curves and radiation-induced apoptotic DNA fragmentation patterns in these cell lines are already established in the laboratory. Cells are treated with this drug, carrier-peptide, or carrier-peptide-drug complex with or without radiation. After 24 hours, the cells are trypsinized and plated for clonogenic survival studies. Apoptotic response to the treatments is determined by: 1) agarose gel electrophoresis analysis of DNA fragmentation at 48 and 72 hours after treatment, 2) morphological observations after staining with DAPI, and 3) terminal deoxytransferase-mediated dUTP Nick End Labeling (TUNEL) assay. [0038]
  • EXAMPLE 2
  • In Vivo Testing of Drug Complexes [0039]
  • Two human prostate carcinoma tumor lines grown as xenografts in male SCID mice are used: the human DU-145 prostate carcinoma which is not androgen dependent and the human LNCaP prostate carcinoma which produces PSA and has a well characterized androgen receptor and response to androgens. The drug complexes are administered up to maximally tolerated doses alone and in conjunction with fractionated radiation therapy ([0040] 137Cs Gamma Cell 40) delivered to the tumor bearing limb.
  • The progress of each tumor is assessed thrice weekly by caliper measurements until the tumors reach 2000 mm[0041] 3. Tumor growth delay is calculated as the number of days for each tumor to reach a volume of 500 mm3 as compared to untreated controls. The efficacy of combination treatments is assessed using isobologram analysis for determination of additivity/synergy.
  • Phase I Clinical Trial: (Maximal Tolerable Dose Assessment) [0042]
  • Based on the results of the in vitro and in vivo studies of the drug complexes, a drug complex is chosen for testing in ten patients with hormone refractory metastatic prostate cancer. A single intravenous dose of the drug complex is given on an inpatient basis with cardiorespiratory monitoring. Toxicity evaluation of all major organ systems is evaluated using blood, urine, stool, and, if necessary, bone marrow studies. Doses are escalated in 10% increments per patient until the maximal response with acceptable toxicity is reached. Response is assessed using a combination of a weekly digital rectal examination (DRE), PSA, LDH, hemoglobin, and monthly bone scan. The starting dose is selected on the toxicity data from the preceding animal studies. [0043]
  • EXAMPLE 3
  • Effect of Thapsigargin (TG) on Cell Proliferation in vitro [0044]
  • Preliminary in vitro data indicate that TG inhibits cell proliferation (Table 1, [0045] DU 145 cells) clonogenic cell survival (Table 2, PC3 cells) of androgen independent human prostate cancer cells.
  • Table 1 shows the effect of thapsigargin and radiotherapy on proliferation of DU-145 prostate cancer cells. The results indicate that thapsigargin inhibits cell proliferation in vitro. [0046]
    TABLE 1
    Effect of TG and RT on proliferation of DU-145 prostate cancer cells*
    Total Number of Cells per
    Thapsigargin Dish (106)
    (nM) 0 Gy 2 Gy 4 Gy 8 Gy
     0 2.1 1.4 1.2 0.5
     20 1.0 0.8 0.3 0.4
    100 0.7 0.5 0.5 0.4
    400 0.7 0.4 0.3 0.3
  • Table 2 shows the effect of thapsigargin and radiotherapy on clonogenic cell survival of PC3 prostate cancer cells. The results indicate that thapsigargin inhibits clonogenic survival in vitro. [0047]
    TABLE 2
    Effect of TG and RT on clonogenic survival of PC3 cells**
    Surviving Fraction (SF) NET survival
    Thapsigargin (PE treated/PE control) (SF/TG toxicity)
    (nM) 0 Gy 2 Gy 4 Gy 2 Gy 4 Gy
    0 1 (0.8) 0.57 0.38 0.57 0.38
    20 0.69 0.42 0.23 0.61 0.33
    50 0.80 0.39 0.23 0.49 0.29
    100 0.62 0.25 0.18 0.40 0.30
    200 0.58 0.25 0.13 0.43 0.22
    400 0.60 0.25 0.13 0.42 0.22
  • EXAMPLE 4
  • Effect of Thapsigargin (TG) on Cell Proliferation in vivo [0048]
  • Male SCID mice bearing the human DU-145 prostate carcinoma Xenograft growing subcutaneously in a hind-limb were treated daily with TG (0.5 mg/kg) by IP injection from day 4-18 post tumor cell implantation. Some animal groups also received fractionated RT locally to the tumor bearing region on days 7-11 or days 7-11 and 14-18. FIG. 1, low dose TG (0.5 mg/kg) administration produced a measurable tumor growth delay and, when given in conjunction with RT, the independent effects of TG and RT were additive. Moreover, when the RT regimen was extended to 2 weeks, the additional benefit of the TG administration was maintained. [0049]
  • EXAMPLE 5
  • PSA Cleaving of pNA Derivative [0050]
  • A tyrosine tripeptide (compound 1 in FIG. 5) was prepared using methods known to those in the peptide chemistry art. It is known that PSA cleaves its para-nitroanilide (pNA) derivative at the tyrosine site ([0051] compound 2 in FIG. 5). (Christensson, A. et al., Eur. J. Biochem., 194:755-763 (1990)). It is also known that the “released” pNA makes it possible to study this reaction colormetrically. Using this feature, it was demonstrated that the sonicate from a cell line that is known to produce PSA, i.e. LNCaP, cleaves compound 2 in FIG. 5. It was also shown that a negative control, i.e. CV-1 cell line, that does not produce PSA did not cleave compound 2 in FIG. 5 and that neither newborn calf serum, nor pooled human serum cleaved compound 2 in FIG. 5. These results indicate that a “PSA-like” activity exists in the sonicate of LNCaP (and not in human or fetal calf serum) that can cleave compound 1 in FIG. 5 at the tyrosine site and release pNA.
  • Based on the above results, the pNA group was replaced with adriamycin, i.e. by attaching adriamycin directly to the tyrosine (FIG. 6) via adriamycin's —NH[0052] 2 group. It was hypothesized that PSA would cleave the molecule of FIG. 6 at the tyrosine site and release adriamycin. However, there was no cleavage and no release of adriamycin.
  • There are several reasons why cleavage and release did not occur. First, pNA, because of its electron withdrawing para-NO[0053] 2 group, is a better leaving group than adriamycin. Second, because pNA is electron-withdrawing, it can greatly activate the tyrosine carbonyl group toward hydrolysis relative to adriamycin. Finally, since adriamycin is sterically bulkier than the pNA group, the drug may have prevented PSA from approaching the tyrosine site.
  • EXAMPLE 6
  • Construction of Molecules Containing para-Amino Benzoic Acid and para-Aminobenzyloxycarbonyl [0054]
  • Attaching adriamycin to a tripeptide molecule (see compound 1 of FIG. 5) through a para-amino benzoic acid can solve the problems previously discussed for the following reasons: (a) pABA can function as a good leaving group because it contains an electron withdrawing carbonyl group in the para-position, (b) because pABA is electron-withdrawing, it can activate the tyrosine carbonyl hydrolysis, and (c) PSA activity may not be sterically hindered as the pABA linker can keep adriamycin away from the tyrosine cleavage site. [0055]
  • The results with this molecule indicate that pABA is a good leaving group and that it can activate the tyrosine carbonyl for hydrolysis. That is, the sonicate of LNCaP that showed “PSA-like” activity cleaved [0056] compound 4 of FIG. 7 at the tyrosine site and released pABA.
  • Since it is established, based on the above results, that there is a linker suitable for the attachment of adriamycin, [0057] compound 5 of FIG. 7 can be synthesized. Furthermore, it can be examined to determine if it can also be cleaved at the tyrosine site to release an active form of adriamycin, namely compound 6 of FIG. 7.
  • Cleavage of [0058] compound 5 can be assessed using the sonicate of LNCaP that exhibits “PSA-like” activity. It is expected that cleavage will occur at the tyrosine site of compound 5. However, what will be “released” is an analog of adriamycin (compound 6 of FIG. 7) and not adriamycin itself. The ability of this analog to intercalate DNA can be assessed and compared with that of native adriamycin using known methods, such as measurement of emissions at 450 nm on ethanol-precipitated DNA. The data is also normalized for efficiency of precipitation by assaying the DNA at 260 nm.
  • An alternative approach takes advantage of a compound that is similar to pABA. This compound is para-aminobenzyloxycarbonyl (pABOC), which offers the advantage of breaking down spontaneously (compound 8 of FIG. 8) to release “native” adriamycin (i.e. adriamycin lacking N-substituents; [0059] compound 9 of FIG. 8). Because of the similarity between pABA and pABOC, the latter offers many of the same advantages. First, since the physical size of pABOC is similar to that of pABA, it can keep the “bulky” adriamycin away from the tyrosine cleavage site. Second, electron-withdrawing fluorine substituents to be added at the two ortho-positions (shown in compounds 7 and 8 of FIG. 8) should make pABOC a good leaving group without introducing any steric bulk, since fluorine and hydrogen atoms are essentially the same size. Finally, the fluorine-containing pABOC is electron-withdrawing and, thus, can also activate the tyrosine carbonyl toward hydrolysis. Based on this information, it can be expected that PSA will cleave compound 7 of FIG. 8 at the tyrosine site and release compound 8 of FIG. 8. Then, as described above, compound 8 of FIG. 8 should break down spontaneously under physiological conditions to release “native” adriamycin. This can be assessed using known methods.
  • Equivalents [0060]
  • Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims. [0061]

Claims (22)

We claim:
1. A drug complex comprising:
a) a targeting carrier molecule which, when introduced into an individual, is selectively distributed to a specific cell type or tissue containing the specific cell type;
b) a linker which is acted upon by a molecule present at an effective concentration in the environs of the specific cell type; and
c) a drug or an agent to be delivered to the specific cell type.
2. A drug complex comprising:
a) a targeting carrier molecule which, when introduced into an individual, is selectively delivered to prostate tissue, bone or both;
b) a peptide which is a substrate for prostate specific antigen; and
c) a cytotoxic drug which is toxic to androgen independent prostate cancer cells,
wherein the peptide links the targeting molecule and the cytotoxic drug.
3. The drug complex of claim 2, wherein the targeting carrier molecule is selected from the group consisting of polyamines; the peptide linker is selected from the group consisting of SEQ ID NOS. 1-14 and alanine-lysine-phenylalanine-glutamate; and the cytotoxic drug is selected from the group consisting of: adriamycin, amonafide, cisplatin, carboplatin (CBDCA), CHIP, cyclophosphamide, doxorubicin, epirubicin, estramustine, etoposide, 5-fluorouracil, gallium nitrate, idarubicin, ifosfamide+mesna, ketoconazole, liarozole (R85,246), methotrexate, mitomycin C, mitoguazone, mitoxantrone, proscar (finasteride), suramin, taxol, thapsigargin, trimetrexate, vinblastine (CI), and vinblastine and emcyt.
4. The drug complex of claim 3, wherein the polyamine is selected from the group consisting of putrescine, spermine, and spermidine.
5. The drug complex of claim 4, wherein the putrescine is fluoropropylputrescine.
6. The drug complex of claim 2, wherein the peptide linker additionally comprises a chemical selected from the group consisting of para-amino benzoic acid and para-aminobenzyloxycarbonyl.
7. A method of killing androgen independent prostate cancer cells in a man with prostate cancer, comprising administering to the man a therapeutically effective amount of a drug complex which comprises:
a) a targeting carrier molecule which is selectively delivered to prostate tissue and bone;
b) a peptide which is a substrate for prostate specific antigen; and
c) a cytotoxic drug which is toxic to androgen independent prostate cancer cells,
wherein the peptide links the targeting molecule and the cytotoxic drug and wherein the drug complex is administered to the man in such a manner that it is delivered to androgen independent prostate cancer cells and the cytotoxic drug enters androgen independent prostate cancer cells, thereby killing the cells.
8. The method of claim 7, wherein the peptide which is a substrate for prostate specific antigen is selected from the group consisting of SEQ ID NOS. 1-14 and alanine-lysine-phenylalanine-glutamate.
9. The method of claim 7, wherein the targeting carrier molecule is selected from the group consisting of polyamines.
10. The method of claim 9, wherein the polyamine is selected from the group consisting of putrescine, spermine, and spermidine.
11. The method of claim 10, wherein the putrescine is fluoropropylputrescine.
12. The method of claim 7, wherein the peptide linker additionally comprises a chemical selected from the group consisting of para-amino benzoic acid and para-aminobenzyloxycarbonyl.
13. The method of claim 12, wherein the drug complex is administered intravenously.
14. The method of claim 13, wherein the cytotoxic drug is selected from the group consisting of: adriamycin, amonafide, cisplatin, carboplatin (CBDCA), CHIP, cyclophosphamide, doxorubicin, epirubicin, estramustine, etoposide, 5-fluorouracil, gallium nitrate, idarubicin, ifosfamide+mesna, ketoconazole, liarozole (R85,246), methotrexate, mitomycin C, mitoguazone, mitoxantrone, proscar (finasteride), suramin, taxol, thapsigargin, trimetrexate, vinblastine (CI), and vinblastine and emcyt.
15. A method of treating metastatic prostate cancer in an man, comprising administering to the man a therapeutically effective amount of a drug complex which comprises:
a) a targeting carrier molecule which is selectively delivered to prostate tissue and bone;
b) a peptide which is a substrate for prostate specific antigen; and
c) a cytotoxic drug which is toxic to metastatic prostate cancer cells,
wherein the peptide links the targeting molecule and the cytotoxic drug and wherein the drug complex is administered to the man in such a manner that it is delivered to prostate tissue and bone and the cytotoxic drug enters metastatic prostate cancer cells, thereby killing the cells.
16. The method of claim 15, wherein the peptide which is a substrate for prostate specific antigen is selected from the group consisting of SEQ ID NOS. 1-14 and alanine-lysine-phenylalanine-glutamate.
17. The method of claim 15, wherein the targeting carrier molecule is selected from the group consisting of polyamines.
18. The method of claim 17, wherein the polyamine is selected from the group consisting of putrescine, spermine, and spermidine.
19. The method of claim 18, wherein the putrescine is fluoropropylputrescine.
20. The method of claim 15, wherein the peptide linker additionally comprises a chemical selected from the group consisting of para-amino benzoic acid and para-aminobenzyloxycarbonyl.
21. The method of claim 20, wherein the drug complex is administered intravenously.
22. The method of claim 21, wherein the cytotoxic drug is selected from the group consisting of: adriamycin, amonafide, cisplatin, carboplatin (CBDCA), CHIP, cyclophosphamide, doxorubicin, epirubicin, estramustine, etoposide, 5-fluorouracil, gallium nitrate, idarubicin, ifosfamide+mesna, ketoconazole, liarozole (R85,246), methotrexate, mitomycin C, mitoguazone, mitoxantrone, proscar (finasteride), suramin, taxol, thapsigargin, trimetrexate, vinblastine (CI), and vinblastine and emcyt.
US10/119,417 1996-09-16 2002-04-09 Drug complex for treatment of metastatic prostate cancer Abandoned US20030035804A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/119,417 US20030035804A1 (en) 1996-09-16 2002-04-09 Drug complex for treatment of metastatic prostate cancer
US11/102,277 US20050233948A1 (en) 1996-09-16 2005-04-08 Drug complex for treatment of metastatic prostate cancer

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US71311496A 1996-09-16 1996-09-16
US383898A 1998-01-07 1998-01-07
US09/110,822 US6368598B1 (en) 1996-09-16 1998-07-06 Drug complex for treatment of metastatic prostate cancer
US10/119,417 US20030035804A1 (en) 1996-09-16 2002-04-09 Drug complex for treatment of metastatic prostate cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/110,822 Division US6368598B1 (en) 1996-09-16 1998-07-06 Drug complex for treatment of metastatic prostate cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/102,277 Division US20050233948A1 (en) 1996-09-16 2005-04-08 Drug complex for treatment of metastatic prostate cancer

Publications (1)

Publication Number Publication Date
US20030035804A1 true US20030035804A1 (en) 2003-02-20

Family

ID=22335136

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/110,822 Expired - Fee Related US6368598B1 (en) 1996-09-16 1998-07-06 Drug complex for treatment of metastatic prostate cancer
US10/119,417 Abandoned US20030035804A1 (en) 1996-09-16 2002-04-09 Drug complex for treatment of metastatic prostate cancer
US11/102,277 Abandoned US20050233948A1 (en) 1996-09-16 2005-04-08 Drug complex for treatment of metastatic prostate cancer

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/110,822 Expired - Fee Related US6368598B1 (en) 1996-09-16 1998-07-06 Drug complex for treatment of metastatic prostate cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/102,277 Abandoned US20050233948A1 (en) 1996-09-16 2005-04-08 Drug complex for treatment of metastatic prostate cancer

Country Status (3)

Country Link
US (3) US6368598B1 (en)
AU (1) AU5089799A (en)
WO (1) WO2000001419A1 (en)

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004108083A3 (en) * 2003-05-30 2005-05-19 Charles E Zeilig Methods for the selective treatment of tumors by calcium-mediated induction of apoptosis
US20080045589A1 (en) * 2006-05-26 2008-02-21 Susan Kelley Drug Combinations with Substituted Diaryl Ureas for the Treatment of Cancer
US20080268063A1 (en) * 2004-11-04 2008-10-30 Sangyong Jon Coated Controlled Release Polymer Particles as Efficient Oral Delivery Vehicles for Biopharmaceuticals
US20090298710A1 (en) * 2005-12-15 2009-12-03 Farokhzad Omid C System for Screening Particles
US20100092425A1 (en) * 2008-10-12 2010-04-15 Von Andrian Ulrich Nicotine Immunonanotherapeutics
US20100129439A1 (en) * 2008-10-12 2010-05-27 Frank Alexis Adjuvant Incorporation in Immunonanotherapeutics
US20100144845A1 (en) * 2006-08-04 2010-06-10 Massachusetts Institute Of Technology Oligonucleotide systems for targeted intracellular delivery
US20100183727A1 (en) * 2008-10-12 2010-07-22 Matteo Iannacone Immunonanotherapeutics that Provide IgG Humoral Response Without T-Cell Antigen
US20100233251A1 (en) * 2007-10-12 2010-09-16 Massachusetts Institute of Technology Massachusetts Vaccine Nanotechnology
US20100266491A1 (en) * 2006-03-31 2010-10-21 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
US20100297233A1 (en) * 2007-02-09 2010-11-25 Massachusetts Institute Of Technology Oscillating cell culture bioreactor
US20100303723A1 (en) * 2006-11-20 2010-12-02 Massachusetts Institute Of Technology Drug delivery systems using fc fragments
US20110020388A1 (en) * 2009-05-27 2011-01-27 Selecta Biosciences, Inc. Targeted synthetic nanocarriers with ph sensitive release of immunomodulatory agents
US20110052697A1 (en) * 2006-05-17 2011-03-03 Gwangju Institute Of Science & Technology Aptamer-Directed Drug Delivery
US20110098232A1 (en) * 2003-05-30 2011-04-28 Zeilig Charles E Methods For The Selective Treatment Of Tumors By Calcium-Mediated Induction Of Apoptosis
US20110110965A1 (en) * 2009-08-26 2011-05-12 Selecta Biosciences, Inc. Compositions that induce t cell help
US20110223201A1 (en) * 2009-04-21 2011-09-15 Selecta Biosciences, Inc. Immunonanotherapeutics Providing a Th1-Biased Response
EP2436376A1 (en) 2007-09-28 2012-04-04 Bind Biosciences, Inc. Cancer cell targeting using nanoparticles
US8193334B2 (en) 2007-04-04 2012-06-05 The Brigham And Women's Hospital Polymer-encapsulated reverse micelles
US8323698B2 (en) 2006-05-15 2012-12-04 Massachusetts Institute Of Technology Polymers for functional particles
US8343497B2 (en) 2008-10-12 2013-01-01 The Brigham And Women's Hospital, Inc. Targeting of antigen presenting cells with immunonanotherapeutics
US9333179B2 (en) 2007-04-04 2016-05-10 Massachusetts Institute Of Technology Amphiphilic compound assisted nanoparticles for targeted delivery
US9381477B2 (en) 2006-06-23 2016-07-05 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
US9636413B2 (en) 2012-11-15 2017-05-02 Endocyte, Inc. Conjugates for treating diseases caused by PSMA expressing cells
US9951324B2 (en) 2010-02-25 2018-04-24 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
US10039822B2 (en) 2011-04-29 2018-08-07 Selecta Biosciences, Inc. Method for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responses
US10046054B2 (en) 2007-08-17 2018-08-14 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US10188759B2 (en) 2015-01-07 2019-01-29 Endocyte, Inc. Conjugates for imaging
WO2022074152A1 (en) 2020-10-08 2022-04-14 Targimmune Therapeutics Ag Immunotherapy for the treatment of cancer
WO2023079142A2 (en) 2021-11-05 2023-05-11 Targimmune Therapeutics Ag Targeted linear conjugates comprising polyethyleneimine and polyethylene glycol and polyplexes comprising the same
US11951190B2 (en) 2013-10-18 2024-04-09 Novartis Ag Use of labeled inhibitors of prostate specific membrane antigen (PSMA), as agents for the treatment of prostate cancer

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6368598B1 (en) * 1996-09-16 2002-04-09 Jcrt Radiation Oncology Support Services, Inc. Drug complex for treatment of metastatic prostate cancer
TWI253935B (en) * 1998-05-22 2006-05-01 Daiichi Seiyaku Co Drug complex
CN1191093C (en) * 1998-10-30 2005-03-02 第一制药株式会社 DDS compounds and method for assaying the same
WO2000066175A2 (en) * 1999-04-30 2000-11-09 Slil Biomedical Corporation Conjugates as therapies for cancer and prostate diseases
US6482943B1 (en) 1999-04-30 2002-11-19 Slil Biomedical Corporation Quinones as disease therapies
GB0012718D0 (en) * 2000-05-24 2000-07-19 Angeletti P Ist Richerche Bio Conjugates of aminodrugs
EP2518142B1 (en) * 2001-08-24 2015-07-15 UVic Industry Partnerships Inc. Proaerolysin containing protease activation sequences and methods of use for treatment of prostate cancer
AU2003256572A1 (en) * 2002-07-16 2004-02-23 Deltagen Proteomics Inc. Negative selections assays, and compositions thereof
ATE292144T1 (en) * 2002-08-02 2005-04-15 Inst Curie SHIGA TOXIN SUBUNIT B AS A VECTOR FOR DIAGNOSIS OF TUMORS AND FOR DRUG ADMINISTRATION TO GB3-EXPRESSING TUMORS
WO2004021861A2 (en) * 2002-09-03 2004-03-18 Vit Lauermann Targeted release
US7691905B2 (en) * 2002-12-24 2010-04-06 New York University Inhibition of melanogenesis and melanoma metastasis with p-aminobenzoic acid (PABA)
US8198328B2 (en) * 2004-01-21 2012-06-12 New York University Treatment of cancer using benzoic acid derivatives
CA2583389A1 (en) * 2004-10-07 2006-04-20 Emory University Multifunctional nanoparticles conjugates and their use
US20060093639A1 (en) * 2004-10-29 2006-05-04 Starkebaum Warren L Method and device for destroying body tissue
KR101365538B1 (en) 2005-06-14 2014-02-21 프로톡스 테라페유틱 인코포레이티드 Method of treating or preventing benign prostatic hyperplasia using modified pore-forming proteins
PL1912675T3 (en) 2005-07-25 2014-10-31 Emergent Product Dev Seattle B-cell reduction using cd37-specific and cd20-specific binding molecules
DE602007013405D1 (en) 2006-07-14 2011-05-05 Us Government METHOD FOR DETERMINING THE PROGNOSIS OF ADENOCARCINOMA
AU2008289441A1 (en) 2007-08-22 2009-02-26 Cytomx Therapeutics, Inc. Activatable binding polypeptides and methods of identification and use thereof
US20100048914A1 (en) 2008-03-14 2010-02-25 Angela Brodie Novel C-17-Heteroaryl Steroidal Cyp17 Inhibitors/Antiandrogens, In Vitro Biological Activities, Pharmacokinetics and Antitumor Activity
US8785423B2 (en) * 2008-04-14 2014-07-22 University Of Maryland, Baltimore Compositions and methods of inducing endoplasmic reticulum stress response for the treatment of cell proliferative diseases
US20100189651A1 (en) 2009-01-12 2010-07-29 Cytomx Therapeutics, Llc Modified antibody compositions, methods of making and using thereof
EP3023433A1 (en) 2009-02-05 2016-05-25 Tokai Pharmaceuticals, Inc. Novel prodrugs of steroidal cyp17 inhibitors/antiandrogens
US9884067B2 (en) 2013-03-14 2018-02-06 University Of Maryland, Baltimore Androgen receptor down-regulating agents and uses thereof
US9993460B2 (en) 2013-07-26 2018-06-12 Race Oncology Ltd. Compositions to improve the therapeutic benefit of bisantrene and analogs and derivatives thereof
WO2015023710A1 (en) 2013-08-12 2015-02-19 Tokai Pharmaceuticals, Inc. Biomarkers for treatment of neoplastic disorders using androgen-targeted therapies
US10525139B2 (en) 2014-11-05 2020-01-07 Dana-Farber Cancer Institute, Inc. Folate-conjugated molecules for delivery of toxic small molecule inhibitors to cancer cells and methods of use
WO2016183176A1 (en) * 2015-05-12 2016-11-17 Drexel University Compounds and compositions useful for treating or preventing cancer metastasis, and methods using same

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
ES2149768T3 (en) 1992-03-25 2000-11-16 Immunogen Inc CONJUGATES OF BINDING AGENTS OF CELLS DERIVED FROM CC-1065.
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
US5599686A (en) 1994-06-28 1997-02-04 Merck & Co., Inc. Peptides
US5866679A (en) 1994-06-28 1999-02-02 Merck & Co., Inc. Peptides
CA2177644A1 (en) 1995-05-31 1996-12-01 Peter D. Senter Polymeric prodrugs for beta-lactamase and uses thereof
US6368598B1 (en) * 1996-09-16 2002-04-09 Jcrt Radiation Oncology Support Services, Inc. Drug complex for treatment of metastatic prostate cancer

Cited By (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004108083A3 (en) * 2003-05-30 2005-05-19 Charles E Zeilig Methods for the selective treatment of tumors by calcium-mediated induction of apoptosis
US20070054874A1 (en) * 2003-05-30 2007-03-08 Zeilig Charles E Methods for the selective treatment of tumors by calcium-mediated induction of apoptosis
US20110098232A1 (en) * 2003-05-30 2011-04-28 Zeilig Charles E Methods For The Selective Treatment Of Tumors By Calcium-Mediated Induction Of Apoptosis
US20080268063A1 (en) * 2004-11-04 2008-10-30 Sangyong Jon Coated Controlled Release Polymer Particles as Efficient Oral Delivery Vehicles for Biopharmaceuticals
US9492400B2 (en) 2004-11-04 2016-11-15 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
US20090298710A1 (en) * 2005-12-15 2009-12-03 Farokhzad Omid C System for Screening Particles
US9267937B2 (en) 2005-12-15 2016-02-23 Massachusetts Institute Of Technology System for screening particles
US20100266491A1 (en) * 2006-03-31 2010-10-21 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
US8709483B2 (en) 2006-03-31 2014-04-29 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
US8802153B2 (en) 2006-03-31 2014-08-12 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
US9080014B2 (en) 2006-05-15 2015-07-14 Massachusetts Institute Of Technology Polymers for functional particles
US8367113B2 (en) 2006-05-15 2013-02-05 Massachusetts Institute Of Technology Polymers for functional particles
US8323698B2 (en) 2006-05-15 2012-12-04 Massachusetts Institute Of Technology Polymers for functional particles
US9688812B2 (en) 2006-05-15 2017-06-27 Massachusetts Institute Of Technology Polymers for functional particles
US20110052697A1 (en) * 2006-05-17 2011-03-03 Gwangju Institute Of Science & Technology Aptamer-Directed Drug Delivery
US20080045589A1 (en) * 2006-05-26 2008-02-21 Susan Kelley Drug Combinations with Substituted Diaryl Ureas for the Treatment of Cancer
US9381477B2 (en) 2006-06-23 2016-07-05 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
US20100144845A1 (en) * 2006-08-04 2010-06-10 Massachusetts Institute Of Technology Oligonucleotide systems for targeted intracellular delivery
US20100303723A1 (en) * 2006-11-20 2010-12-02 Massachusetts Institute Of Technology Drug delivery systems using fc fragments
US9217129B2 (en) 2007-02-09 2015-12-22 Massachusetts Institute Of Technology Oscillating cell culture bioreactor
US20100297233A1 (en) * 2007-02-09 2010-11-25 Massachusetts Institute Of Technology Oscillating cell culture bioreactor
US9333179B2 (en) 2007-04-04 2016-05-10 Massachusetts Institute Of Technology Amphiphilic compound assisted nanoparticles for targeted delivery
US8193334B2 (en) 2007-04-04 2012-06-05 The Brigham And Women's Hospital Polymer-encapsulated reverse micelles
US10646581B2 (en) 2007-08-17 2020-05-12 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US11717514B2 (en) 2007-08-17 2023-08-08 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US10517956B2 (en) 2007-08-17 2019-12-31 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US10517957B2 (en) 2007-08-17 2019-12-31 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US10624970B2 (en) 2007-08-17 2020-04-21 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US10406240B2 (en) 2007-08-17 2019-09-10 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US10624969B2 (en) 2007-08-17 2020-04-21 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US11504357B2 (en) 2007-08-17 2022-11-22 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US10485878B2 (en) 2007-08-17 2019-11-26 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US10624971B2 (en) 2007-08-17 2020-04-21 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US10046054B2 (en) 2007-08-17 2018-08-14 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US10828282B2 (en) 2007-08-17 2020-11-10 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US11083710B2 (en) 2007-08-17 2021-08-10 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US11298341B2 (en) 2007-08-17 2022-04-12 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US11318121B2 (en) 2007-08-17 2022-05-03 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US11369590B2 (en) 2007-08-17 2022-06-28 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
EP2436376A1 (en) 2007-09-28 2012-04-04 Bind Biosciences, Inc. Cancer cell targeting using nanoparticles
EP2644192A1 (en) 2007-09-28 2013-10-02 Bind Therapeutics, Inc. Cancer Cell Targeting Using Nanoparticles
EP2644594A1 (en) 2007-09-28 2013-10-02 Bind Therapeutics, Inc. Cancer Cell Targeting Using Nanoparticles
US11547667B2 (en) 2007-10-12 2023-01-10 Massachusetts Institute Of Technology Vaccine nanotechnology
US20100233251A1 (en) * 2007-10-12 2010-09-16 Massachusetts Institute of Technology Massachusetts Vaccine Nanotechnology
US9474717B2 (en) 2007-10-12 2016-10-25 Massachusetts Institute Of Technology Vaccine nanotechnology
US10736848B2 (en) 2007-10-12 2020-08-11 Massachusetts Institute Of Technology Vaccine nanotechnology
US9526702B2 (en) 2007-10-12 2016-12-27 Massachusetts Institute Of Technology Vaccine nanotechnology
US9539210B2 (en) 2007-10-12 2017-01-10 Massachusetts Institute Of Technology Vaccine nanotechnology
US20100129439A1 (en) * 2008-10-12 2010-05-27 Frank Alexis Adjuvant Incorporation in Immunonanotherapeutics
US8906381B2 (en) 2008-10-12 2014-12-09 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IGG humoral response without T-cell antigen
US9233072B2 (en) 2008-10-12 2016-01-12 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US8343497B2 (en) 2008-10-12 2013-01-01 The Brigham And Women's Hospital, Inc. Targeting of antigen presenting cells with immunonanotherapeutics
US8343498B2 (en) 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US8932595B2 (en) 2008-10-12 2015-01-13 Massachusetts Institute Of Technology Nicotine immunonanotherapeutics
US9308280B2 (en) 2008-10-12 2016-04-12 Massachusetts Institute Of Technology Targeting of antigen presenting cells with immunonanotherapeutics
US20100092425A1 (en) * 2008-10-12 2010-04-15 Von Andrian Ulrich Nicotine Immunonanotherapeutics
US8637028B2 (en) 2008-10-12 2014-01-28 President And Fellows Of Harvard College Adjuvant incorporation in immunonanotherapeutics
US8591905B2 (en) 2008-10-12 2013-11-26 The Brigham And Women's Hospital, Inc. Nicotine immunonanotherapeutics
US8562998B2 (en) 2008-10-12 2013-10-22 President And Fellows Of Harvard College Targeting of antigen presenting cells with immunonanotherapeutics
US9439859B2 (en) 2008-10-12 2016-09-13 Massachusetts Institute Of Technology Adjuvant incorporation in immunoanotherapeutics
US20100183727A1 (en) * 2008-10-12 2010-07-22 Matteo Iannacone Immunonanotherapeutics that Provide IgG Humoral Response Without T-Cell Antigen
US8277812B2 (en) 2008-10-12 2012-10-02 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IgG humoral response without T-cell antigen
US20110223201A1 (en) * 2009-04-21 2011-09-15 Selecta Biosciences, Inc. Immunonanotherapeutics Providing a Th1-Biased Response
US9006254B2 (en) 2009-05-27 2015-04-14 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US20110020388A1 (en) * 2009-05-27 2011-01-27 Selecta Biosciences, Inc. Targeted synthetic nanocarriers with ph sensitive release of immunomodulatory agents
US8629151B2 (en) 2009-05-27 2014-01-14 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US9884112B2 (en) 2009-05-27 2018-02-06 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US20110110965A1 (en) * 2009-08-26 2011-05-12 Selecta Biosciences, Inc. Compositions that induce t cell help
US10557128B2 (en) 2010-02-25 2020-02-11 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US9951324B2 (en) 2010-02-25 2018-04-24 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US11155800B2 (en) 2010-02-25 2021-10-26 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
US10039822B2 (en) 2011-04-29 2018-08-07 Selecta Biosciences, Inc. Method for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responses
US10912840B2 (en) 2012-11-15 2021-02-09 Endocyte, Inc. Conjugates for treating diseases caused by PSMA expressing cells
US9782493B2 (en) 2012-11-15 2017-10-10 Endocyte, Inc. Conjugates for treating diseases caused by PSMA expressing cells
US9636413B2 (en) 2012-11-15 2017-05-02 Endocyte, Inc. Conjugates for treating diseases caused by PSMA expressing cells
US11951190B2 (en) 2013-10-18 2024-04-09 Novartis Ag Use of labeled inhibitors of prostate specific membrane antigen (PSMA), as agents for the treatment of prostate cancer
US10898596B2 (en) 2015-01-07 2021-01-26 Endocyte, Inc. Conjugates for imaging
US10188759B2 (en) 2015-01-07 2019-01-29 Endocyte, Inc. Conjugates for imaging
WO2022074152A1 (en) 2020-10-08 2022-04-14 Targimmune Therapeutics Ag Immunotherapy for the treatment of cancer
WO2023079142A2 (en) 2021-11-05 2023-05-11 Targimmune Therapeutics Ag Targeted linear conjugates comprising polyethyleneimine and polyethylene glycol and polyplexes comprising the same

Also Published As

Publication number Publication date
US6368598B1 (en) 2002-04-09
WO2000001419A1 (en) 2000-01-13
US20050233948A1 (en) 2005-10-20
AU5089799A (en) 2000-01-24

Similar Documents

Publication Publication Date Title
US6368598B1 (en) Drug complex for treatment of metastatic prostate cancer
JP4675028B2 (en) Trimethyl lock type tetrapartate prodrug
US6720306B2 (en) Tetrapartate prodrugs
US9283279B2 (en) Targeted polymeric conjugates and uses thereof
JP2023098946A (en) Drug delivery conjugates, and methods for treating diseases caused by psma expressing cells
EP0769967B1 (en) Conjugates comprising an antitumour agent and their use
JP6854759B2 (en) Small molecule target conjugates specifically activated by the tumor microenvironment and their use
JPH11500108A (en) Methods and compositions for lipidation of hydrophilic molecules
US20080161245A1 (en) Protein-Binding Anthracycline Peptide Derivatives and Drugs Containing Them
JP2004505009A (en) Combinations for treating neoplasms
WO1988007378A1 (en) Improvements relating to drug delivery systems
CA2081308A1 (en) Method for increasing blood-brain barrier permeability
JPH07506113A (en) Pharmaceutical formulations for inhibiting tumors associated with prostate cancer, gastric cancer and breast cancer
EA026870B1 (en) Combination and pharmaceutical composition for treating tumors
Miyazaki et al. Targeted cytotoxic analog of luteinizing hormone–releasing hormone AN-207 inhibits growth of OV-1063 human epithelial ovarian cancers in nude mice
Radulovic et al. Cytotoxic analog of somatostatin containing methotrexate inhibits growth of MIA PaCa-2 human pancreatic cancer xenografts in nude mice
JP2004518776A (en) Tetrapartate prodrug
WO2021013131A1 (en) Hemoglobin-based therapeutic agents
US20060193917A1 (en) Radiosensitizer formulations and methods for use
KR102400031B1 (en) Cancer-specific self-assembled nanoparticles for enhancing antitumor immunity
US11524078B2 (en) Water-soluble macromolecular derivative of Venetoclax
JP5341314B2 (en) Drug delivery system
Keller et al. Growth inhibition of experimental non‐Hodgkin's lymphomas with the targeted cytotoxic somatostatin analogue AN‐238
WO1995012414A1 (en) Novel modified pf4 compositions and methods of use
EP2041173A2 (en) Peptides with anti-proliferative activity

Legal Events

Date Code Title Description
AS Assignment

Owner name: BETH ISRAEL DEACONESS MEDICAL CENTER, MASSACHUSETT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BUBLEY, GLENN J.;REEL/FRAME:013426/0199

Effective date: 20021007

STCB Information on status: application discontinuation

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