AU2005200354B2 - N-terminally truncated HER-2/neu protein as a cancer prognostic indicator - Google Patents

N-terminally truncated HER-2/neu protein as a cancer prognostic indicator Download PDF

Info

Publication number
AU2005200354B2
AU2005200354B2 AU2005200354A AU2005200354A AU2005200354B2 AU 2005200354 B2 AU2005200354 B2 AU 2005200354B2 AU 2005200354 A AU2005200354 A AU 2005200354A AU 2005200354 A AU2005200354 A AU 2005200354A AU 2005200354 B2 AU2005200354 B2 AU 2005200354B2
Authority
AU
Australia
Prior art keywords
cancer
p95her
alkyl
neu
cells
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.)
Ceased
Application number
AU2005200354A
Other versions
AU2005200354C1 (en
AU2005200354A1 (en
Inventor
Gail Clinton
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.)
Oregon Health Science University
Original Assignee
Oregon Health Science University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US1999/027010 external-priority patent/WO2000029609A1/en
Application filed by Oregon Health Science University filed Critical Oregon Health Science University
Publication of AU2005200354C1 publication Critical patent/AU2005200354C1/en
Publication of AU2005200354A1 publication Critical patent/AU2005200354A1/en
Application granted granted Critical
Publication of AU2005200354B2 publication Critical patent/AU2005200354B2/en
Assigned to OREGON HEALTH AND SCIENCES UNIVERSITY reassignment OREGON HEALTH AND SCIENCES UNIVERSITY Request for Assignment Assignors: OREGON HEALTH SCIENCES UNIVERSITY
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Hospice & Palliative Care (AREA)
  • Food Science & Technology (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Peptides Or Proteins (AREA)

Description

AUSTRALIA
PATENTS ACT 1990 DIVISIONAL APPLICATION NAME OF APPLICANT: Oregon Health Sciences University ADDRESS FOR SERVICE: DAVIES COLLISON CAVE Patent Attorneys 1 Nicholson Street Melbourne, 3000.
INVENTION TITLE: "N-terminally truncated HER-2/neu protein as a cancer prognostic indicator" The following statement is a full description of this invention, including the best method of performing it known to us: Q:\OPER\jrc\2004-2005 Financial Year\Specification Filing\12561270-div file.doc 28/1/05 P:kOPERljrcSpciricaonsUX4-200Divisjonlas\777803-djv.doc-28/01I/5 -1- N-TERMINALLY TRUNCATED HER-2/NEU PROTEIN AS A CANCER SPROGNOSTIC INDICATOR This is a divisional of Australian patent application No. 16246/00 (777,803), the entire contents of which are incorporated herein by reference.
Technical Field of the Invention SThe present invention provides an N-terminally truncated HER-2/neu polypeptide, t p95HER-2, that is useful as a diagnostic and prognostic indicator for breast cancer. The Spresent invention further provides a 12-15 amino acid "extracellular stub" polypeptide that is also a useful epitope for an immunological assay for diagnosis and prognosis of various adenocarcinomas, particularly breast cancer and ovarian cancer.
The present invention was made with funding from the United States Government under grant CA-71447 from the National Cancer Institute and DAMD17-6204 from the Department of Defence (DOD) Breast Cancer Research Program. The United States Government may have certain rights in this invention.
Background of the Invention The HER-2/neu (erbB-2) gene encodes a receptor-like tyrosine kinase (RTK) which is a member of the epidermal growth factor receptor family (Coussens et al., Science 230:1132-1139, 1985). Overexpression of HER-2/neu has been observed in tumors arising at many sites including non-small cell lung (Kern et al., Cancer Res. 50:5184-5191, 1990), colon (Cohen et al., Oncogene, 4:81-88, 1989), prostate (Arai et al., Prostate 30:195-201, 1997), ovarian, and breast (Slamon et al., Science 244:707-712, 1989). In human breast cancer, where HER-2/neu involvement has been studied, overexpression occurs in 15-30% of the cases (Singleton and Strickler, Pathol. Annual 27 Pt 1:165-198, 1992) and predicts for significantly lower survival rate and shorter time to relapse in patients with lymph node positive disease (Slamon et al., Science 244:707-712, 1989; Singleton and Strickler, Pathol. Annual 27 Pt 1:165-198, 1992; Slamon et al., Science 235:177-182, 1987; and Slamon et al., Science 235:177-182, 1987). The significance of HER-2/neu in node negative patients is controversial and so far its clinical utility as a prognostic indicator is limited (Slamon et al., Science 235:177-182, 1987; and Hynes et al., Biochem. Biophys.
Acta 1198:165-184, 1994). Various approaches are being taken toward HER-2/neu targeted therapeutics many of which are based on antibodies specific to the extracellular P:\OPERjrc\Specificatiors\204-2005 Divisionals\777803-div.doc28/OI/O O -1An.s domain (ECD) of the transmembrane protein, which either down regulate receptor function 0, or target recombinant toxins with the goal of specifically killing HER-2/neu expressing tumor cells (Hynes et al., Biochem. Biophys. Acta 1198:165-184, 1994; Press et al., Progress in Clinical Biological Research 354:209-221, 1990; and Dougall et al., Cc 5 Oncogene 9:2109-2123, 1994).
In addition to the full length transmembrane product, p 1 85, of the HER-2/neu gene, a truncated product corresponding to the extracellular domain (ECD) is released from Sbreast carcinoma cells in culture by regulated proteolysis (Lin and Clinton, Oncogene 6:639-643, 1991; Zabrecky et al., J. Biol. Chem. 266:1716-1720, 1991; and Pupa et al., Oncogene 8:2917-2923, 1993), and is also produced from an alternative transcript (Scott et al., Mol. Cell. Biol.
I I
O
0 13:2247-2257 1993). HER-2/neu ECD is elevated in the serum of patients with breast (Leitzel et al., J. Clin. Oncol. 10:1436-1443, 1992), ovarian (Maden et al., Anticancer Res. 17:757-760, tc 1997), and prostate cancer (Myers et al., Int J. Cancer 69:398-402, 1996). Several studies of 00 breast cancers estimate that 6% or less of early stage breast cancer, about 25% of patients with metastatic and locally advanced disease, and greater than 50% of patients with recurrent metastatic disease have elevated serum ECD (Brandt-Rauf et al., Mutation Res. 333:203-208, S1995). Elevated ECD in serum is associated with overexpression of HER-2/neu in tumor r n tissue and also has been correlated to tumor load (Molina et al., Br. J. of Cancer 4:1126-1131, O 1996; and Brodowicz et al., Oncology 54:475-481, 1997). Serum ECD is a marker of metastatic disease and may predict recurrence (Molina et al., Br. J of Cancer 4:1126-1131, O 1996), shortened survival (Brodowicz et al., Oncology 54:475-481, 1997; Kandl et al., Br. J.
CN Cancer 70:739-742, 1994; Fehm et al., Oncology 55:33-38, 1998 and Mansour et al., Anticancer Res. 17:3101-3105, 1997), and response to antiestrogen therapy in advanced stage patients (Leitzel et al., J. Clitn. Oncol. 13:1129-1135, 1995; and Yamauchi ct al., J.Clin.Oncol.
15:2518-2525, 1997). Serum ECD has also been reported to neutralize the activity of anti HER-2/neu antibodies targeted to the ECD (Baselga et al., J Clin. Oncol. 14:737-744, 1996; and Brodowicz et al., Int. J. Cancer. 73:875-879, 1997) possibly allowing escape of HER-2rich tumors from immunological control.
Cellular fragments created by ectodomain shedding have been described for the colony stimulating factor receptor (CSF-IR) (Downing et al., Mol. Cell .Biol. 9:2890-2896, 1989), the TrkA neurotrophin receptor (Cabrera et al., J Cell. Biol. 132:427-436, 1996), Axl receptor (O'Bryan et al., J. Biol. Chem. 270:551-557, 1995), and HER-4 (Vecchi et al., J. Biol. Chem.
271:18989-18995, 1996). However, a truncated cellular product of HER-2/neu shedding has not been identified. The truncated CSF-IR was found to have in vitro kinase activity (Downing et al., Mol Cell .Biol. 9:2890-2896, 1989), and the cytoplasmic HER-4, induced by phorbol ester tumor promoters, had little or no kinase activity (Vecchi et al., J. Biol. Chem.
271:18989-18995, 1996) while a truncated HER-4 found in cells treated with a proteosome inhibitors was an active kinase (Vecchi et al., J. Cell Biol. 139:995-1003, 1997). Therefore, there is a need in the art to identify a truncated HER-2/neu polypeptide and determine if it has enzymatic activity in general or kinase activity in particular. Moreover, such a truncated polypeptide is likely to be a better marker for tumor diagnosis, screening and prognosis as it will be easier to assay for the polypeptide than to assay for shed ECD, which is present in a much more dilute form.
The ECD of full-length transmembrane receptors often exerts a negative regulatory constraint on their signaling activity. Engineered deletion of a region of the HER-2 ECD was found to enhance its oncogenic potency (DiFiore et al., Science 237:178-182, 1987; Hudziak et al., Proc. Natl. Acad. Sci. USA 84:7159-7163, 1987; Segatto et al., Mol. Cell. Biol. 8:5570- 5574, 1988; and Bargmann and Weinberg, EMBO J. 7:2043-2052, 1988). This has also been illustrated by engineered removal of the ECD from the epidermal growth factor (EGF) receptor
O
O and by the oncogenic potency of viral encoded v-erbB, v-kit, and v-ros, that are missing Sregions of the ECD found in their normal cellular counterparts (Rodrigucs and Park, Cur.
Opin. Genet. Dev. 4:15-24, 1994). Naturally occurring mutant EGF receptors with N-terminal 00 truncations have been identified in several human carcinomas (Moscatello et al., Cancer Res.
Cl 5 55:5536-5539, 1995) and have constitutive signaling activity and enhanced oncogenic transforming activity in cell culture and animal models (Moscatello et al., Oncogene 13:85-96, 1996; and Huang, et al. J. Biol. Chem. 272:2927-2935, 1997).
Cc Therefore, there is a need in the art to better study the HER-2/neu receptor and to S determine if there are better regions of this protein available for using as a more sensitive diagnostic and prognostic indicator for breast cancer. Moreover, there is no procedure o available to monitor for staging and prognosis of various adenocarcinomas, such as breast 0 C cancers, other than physically investigating adjacent tissue, such as regional lymph nodes and then sectioning the tissue by difficult histological techniques. Therefore, there is a need in the art to provide improved means for determining adenocarcinoma staging and further determining prognostic factors to guide appropriate treatment strategies. The present invention was made to address the foregoing needs in the art.
Summary of the Invention The present invention is based upon the initial identification of an N-terminally truncated HER-2/neu product. This product is a polypeptide having approximately a 95 kDa molecular weight and having in vitro kinase activity. Moreover, immunoprecipitation using domain specific antibodies was able to isolate this specific polypeptide from intracellular fragments for use as a diagnostic and prognostic indicator of various carcinomas without the severe dilution effects encountered by measuring ECD in blood/serum. The carcinomas for which the 95 kDa polypeptide will have diagnostic and prognostic value include, for example, carcinomas that overexpress HER-2, including breast, gastric, cervical, non-small cell lung, and prostate carcinomas.
The present invention provides a method for diagnostic and prognostic screening of a metastatic stage carcinoma that overexpresses HER-2, comprising: providing a suspected tissue sample having cells; lysing the cells to expose intracellular contents and form a lysate; and measuring the lysate for the presence of 95HER-2 polypeptide.
Preferably, the lysing step is followed by an additional step separating soluble from insoluble material of the lysate to remove dense fibrous material. Preferably, the measuring step utilizes an assay procedure selected from the group consisting of Western blotting, immunochemistry, ELISA, and combinations thereof. Preferably, the carcinoma that overexpresses HER-2 is selected from the group consisting of breast cancer, gastric carcinoma, prostate cancer, non-small cell lung carcinoma, and ovarian carcinoma.
The present invention provides a method for diagnostic and prognostic screening of a metastatic stage carcinoma that overexpresses HER-2, comprising:
O
S providing a suspected tissue sample having cells; S(b) providing an antibody that binds to a stub region of HER-2, wherein the stub region is a polypeptide sequence of SEQ ID NO. 1 or a fragment thereof; and 00 determining the percentage of cells that have an exposed extracellular stub region.
Preferably, the means for determining the percentage of cells having an exposed extracellular stub region utilizes an assay procedure, wherein the assay procedure is selected from the group consisting of Western blotting, immunochemistry, red cell agglutination, eM ELISA, affinity chromatography, and combinations thereof. Preferably, the carcinoma that o overexpresses HER-2 is selected from the group consisting of breast carcinoma, gastric carcinoma, prostate cancer, non-small cell lung carcinoma, and ovarian cancer.
O A method for predicting the therapeutic effectiveness to treat a carcinoma that C" overexpresses HER-2 with a therapeutic agent, wherein the therapeutic agent is a HER-2 binding ligand, comprising: providing a tumor tissue sample having tumor cells contained therein; providing an antibody that binds to a stub region of HER-2, wherein the stub region is a polypeptide sequence of SEQ ID NO. 1 or a fragment thereof; and determining the percentage of cells that have an exposed extracellular stub region, wherein a high percentage of tumor cells binding to the antibody indicates that the cancer will likely be resistant to the therapeutic agent.
Preferably, the means for determining the percentage of cells having an exposed extracellular stub region utilizes an assay procedure, wherein the assay procedure is selected from the group consisting of Western blotting, immunochemistry, red cell agglutination, ELISA, affinity chromatography, and combinations thereof. Preferably, the carcinoma that overexpresses HER-2 is selected from the group consisting of breast carcinoma, gastric carcinoma, prostate cancer, non-small lung carcinoma, and ovarian cancer. Preferably, the therapeutic agent is a humanized monoclonal antibody that binds to the extracellular domain of HER-2 (Herceptin).
A method for treating HER-2/neu-positive carcinomas, comprising administering an effective amount of a hydroxamate compound. Preferably, the hydroxamate compound is
TAPI.
The present invention provides a method for determining node status in breast cancer prognosis, comprising: providing a suspected tissue sample having cells; dividing the tissue sample for measuring both p95HER-2 intracellularly and p 85HER-2; lysing the cells to expose intracellular contents and form a lysate for p95HER-2 assay; measuring the tissue sample for p185HER-2; and
I
O
O measuring the lysate for the presence of 95HER-2 polypeptide, wherein tissue g samples that were both p95HER-2 positive and rich with pl85HER-2 predict lymph node or other metastasis.
00 Preferably, the lysing step is followed by an additional step separating soluble from insoluble material of the lysate to remove dense fibrous material. Preferably, the measuring step utilizes an assay procedure selected from the group consisting of Western blotting, immunochemistry, ELISA, and combinations thereof.
C Brief Description of the Drawings SFigure 1 shows an N-terminally truncated HER-2/neu product having kinase enzymatic activity. About 25 jg of protein from 17-3-1 cells were western blotted with anti-neu (C) O diluted 1:10,000 (lane In lanes 2-4, 400 gg protein were immunoprecipitated with anti-neu (lanes 2,4) or with monoclonal antibody against the extracellular domain, anti-neu(N) (lane or depleted ofpl 85HER-2/neu by extracting twice with anti-neu(N) and then immunoprecipitated with anti-neu(C) (lane The immune complexes were phosphorylated with (-3 2 p) ATP and analyzed by SDS-PAGE and autoradiography, demonstrating kinase activity.
Figure 2 shows that human breast carcinoma cell lines contain p95HER-2/neu.
Indicated amounts of cell lysates from BT474, HBL-100, MDA-MB-453, SKBR3, HMEC, and 17-3-1 cells were immunoprecipitated with anti-neu and phosphorylated following the same procedure described for Figure 1 above.
Figure 3 provides the results of an experiment wherein tyrosine phosphorylation of localized in a particulate fraction of BT474 breast carcinoma cells. Particulate and soluble fractions were prepared by incubation of 107 cells in ice for 10 min in 3 ml of homogenization buffer (10 mM Tris pH 7 4 10 mM NaC1, 2 mM MgCl with 2 mM vanadate and protease inhibitors), followed by dounce homogenization, and then centrifugation at 100,000 x g for 1 hr. The pellet was resuspended in 3 ml of homogenization buffer. About 200 Lpg of protein from the particulate fraction and an equal volume of the soluble fraction were immunoprecipitated with anti-neu and analyzed as a Western blot with monoclonal anti-phosphotyrosine antibody (Sigma). These data show that p95HER-2 is located at the plasma membrane (with pl85HER-2) and that p95HER-2 is phosphorylated in vivo, which is an indication of signaling activity.
Figure 4 shows the expression of p95 and ECD in SKOV3 and BT474 cells. Cells were treated for 24 hrs in serum-free medium with control vehicle or with 500 nM of the phorbol ester TPA and 50 pLM chloroquine. In the top panel (Figure 4A). 5 ml of conditioned media was concentrated 100 fold, denatured under nonreducing conditions, and aliquots normalized to cell extract protein were analyzed by western blotting with anti-neu monoclonal antibody at I pg/ml. In the lower panel (Figure 4B), 20 gg of cell proteins were analyzed by Western blotting using anti-neu The data shown in Figure 4 are representative of three replicate experiments.
O
0 Figure 5 shows the results of experiments showing that P95HER-2 and ECD are inhibited by the hydroxamic acid, TAPI. BT474 cells in serum-free medium were treated for S 24 hrs with the control vehicle or with 1, 10, 20, and 40 pM TAPI (a gift from Immunex, OO Seattle, WA). In the top panel (Figure 5A), the concentrated, conditioned media, normalized l 5 to the amount of cell extract, were analyzed by western blotting with anti-neu Similar results were obtained when 5 -g of protein from the conditioned media from each culture were analyzed. In the lower panel (Figure 5B), 20 pg of cell proteins were analyzed by Western CC) blotting using anti-neu o Figure 6 shows a Western blotting analysis of 12 breast cancer tissues. Human intraductal breast cancer tissues were fractionated and 20 jlg of protein from 12 patients were 0 subjected to western blotting with anti-neu as described in Figure 1, lane 1. The control C lane contained 3 4ig protein from transfected 3T3 cells and 17-3-1 cells. The position the top band, and p95HER-2 the lower band, are marked in the control 17-3-1 sample in the lower panel (Figure 6B). The top panel is a photograph of the film that was exposed to the membrane for 20 min and the bottom panel was exposed for 5 min. HER-2/neu immunoassay values were: <100 Units for #60,39,69; 389 U for #40; 258 U for #58; 302 U for #38; 200 U for #53; 2000 U for #04; 10,000 U for #22; 1000 U for #57; 550 U for #17; 674 U for Detailed Description of the Invention The present invention is based upon the initial identification and characterization of a N-terminally truncated HER-2/neu protein (p95HER-2 or simply p95) and a subsequent examination and correlation with ECD shedding and association with breast cancer pathologic factors.
The present invention identified an N-terminally truncated HER-2/neu product of about kDa, which was detected by Western blotting and by immunoprecipitation with anti-peptide antibodies against the C-terminus, but did not react with monoclonal antibodies against the Nterminus ofpl85HER-2/neu. P95HER-2 has kinase activity evidenced by its selfphosphorylation when p185HER-2 was cleared from the cell extract prior to immunoprecipitation with anti-neu (Figure Several controls and extraction procedures were conducted to rule out that p95 was created by an in vitro degradation artifact. Cells' extracted with protease inhibitors had only two major cytoplasmic HER-2/neu proteins, p95HER-2 and p 185HER-2, with no indication of smaller degradation products. P95HER-2 levels were not affected by procedures that would eliminate the activity ofproteases including direct extraction of cells in boiling 10% SDS-containing buffers.
One mechanism previously described for generation of N-terminally truncated receptor tyrosine kinases is by protcolytic release of their ECD (Downing et al., Mol. Cell.Biol. 9:2890- 2896, 1989; Cabrera et al., J. Cell. Biol. 132:427-436, 1996; O'Bryan et al., J. Biol. Chem., 270:551-557, 1995; and Vecchi et al., J. Biol. Chem. 271:18989-18995, 1996). Production of p95HER-2 in cultured cells occurs by endoproteolytic processing. The presence of p95HIER-2 in 17-3-1 cells transfected with HER-2/neu cDNA indicates that p95HER-2 is a proteolytic
O
O product rather than the product of an alternative transcript. Furthermore, the levels of Sp95HER-2 and soluble HER-2 ECD released from cultured cells were correlated. First, both p95HER-2 and ECD levels were low in SKOV3 cells compared to BT474 cells (Figure 4).
00 Secondly, augmentation of both p95HER-2 and ECD by long term (24 hr) treatment with TPA and chloroquine (Figure 4) further indicated that the truncated HER-2 products were generated through a common pathway.
Although the mechanism for this stimulation was not examined directly, long term C exposure of cells to TPA has been found to enhance intemalization of RTKs (receptor tyrosine S kinases) (Seedorfet al. J. Biol. Chem. 270:18953-18960, 1995). Moreover, chloroquine, an agent that alters pH in cellular endosomes and lysosomes, inhibited complete proteolytic 0 breakdown or altered RTK trafficking (Marshall, J. Biol. Chem 260:4136-4144, 1985).
C- Finally, both p95HER-2 and ECD levels from intact cells were inhibited by the hydroxamate compound, TAPI. Inhibition was maximal at a TAPI concentration of 10 pM or less (Figure The strong inhibition by TAPI indicates that most of the ECD and p95HER-2 in BT474 cells were generated by a metalloprotcase (McGeehan et al., Nature 370:561, 1994; and Mohler et al., Nature 370:218-220, 1994) and that this class ofprotease inhibitors is effective in controlling shedding in breast cancer patients. Although p95HER-2 and shedding were modulated under several different conditions, changes in cellular p185HER-2 levels could not be detected. Unlike several transmembrane proteins that only shed when induced by TPA, proteolytic shedding of pl 85HER-2 occurs continually at a low basal level (Lin and Clinton, Oncogene 6:639-643, 1991; and Zabrecky et al, J. Biol. Chem. 266:1716-1720, 1991) with only about 20% converted into soluble ECD in 2 hrs (Pupa et al., Oncogene, 8:2917-2923, 1993).
The truncated cell protein of about 95 kDa described herein was somewhat larger than the expected 75-80 kDa for the cytoplasmic remnant of the -105-110 kDa ECD. ECD is a glycosylated protein with multiple bands on gel migraton. P95HER-2 or the ECD might migrate anomalously in gels, since the site of cleavage for ECD shedding is not known. The ECD and p95HER-2 are coordinately produced in culture by proteolytic activity that is sensitive to a metalloprotease inhibitor.
A HER-2/neu product of the same size, 95kDa, in transfected 3T3 cells, cultured breast carcinoma cells, breast cancer tissue, and ovarian cancer tissue indicates that a similar proteolytic processing event occurs in the different cells. However p95HER-2 was not detected in all cells and tumor tissue that contain pi 85HER-2. Two non-tumorigenic breast epithelial cell lines had no detectable p95HER-2 (Figure In addition, the SKOV3 ovarian carcinoma cells, which overexpress pi 85HER-2, had a disproportionately low amount of p95HER-2 (Figure These observations indicate that production of p95HER-2 is regulated.
The cells with variable levels of truncated HER-2/neu products may differ in the amount of the relevant protease activity or the protein substrate may have an altered conformation affecting sensitivity to proteolytic cleavage.
O
o P95HER-2/neu has kinase enzymatic activity. It is tyrosine phosphorylated and it is truncated from its N-terminus. Oncogenic signaling by HER-2/neu depends upon its level of kinase activity (DiFiore et al., Science 237:178-182, 1987; Hudziak et al-, Proc. Natl. Acad.
00 Sci. USA 84:7159-7163, 1987; and Segatto et al., Moa Cell Biol. 8:5570-5574, 1988). Since
C
5 p95HER-2 was at 100% ofpl85HER-2 in some breast cancer samples, it impacted the amplitude of the kinase signal. Moreover, an N-terminally truncated kinase domain, such as p95HER-2, is expected to emit a constitutive signal by analogy to results with engineered Cn deletions of the ECD from the HER-2/neu product (Vecchi et al., J. Cell Biol. 139:995-1003, S 1997; DiFiore et al., Science 237:178-182, 1987; Hudziak et al., Proc. Natl. Acad. Sci. USA 84:7159-7163, 1987; Segatto et al., Mol Cell. Biol. 8:5570-5574, 1988; and Bargmann and o Weinberg, EMBO J. 7:2043-2052, 1988). Taken together these data (provided herein) indicate that p95HER-2 will elevate the kinase signal in some patients and is thereby associated with more aggressive tumor growth.
Cancer tissues were analyzed by Western blotting and scored for p95HER-2 and for p185HER-2/neu expression. Breast and ovarian cancer tissues were both found to express p95HER-2 in addition to p185HER-2/neu. Of 161 breast cancer tissues studied, 22.4% expressed p95HER-2, 21.7% ovcrexpressed p185HER-2, and 143% were both p95HER-2 positive and overexpressed p185HER-2. A higher proportion of node positive patients (23 of 78) than node negative patients (9 of 63) expressed p95HER-2 in all tumors combined In the group that overexpressed pl85-HER-2, those that contained p95HER-2 were associated with node positive patients (15 of 21) whereas those that were p95 negative were associated with node negative patients (8 of 11) Neither p95HER-2 nor p185HER-2rich patients significantly correlated with tumor size or with hormone receptor status in this study. These data indicate that breast cancers, which express the HER-2/neu oncogene, are heterogeneous with respect to HER-2/neu protein products. Moreover, p95HER-2/neu appeared to distinguish tumors that have metastasized to the lymph nodes from those in node negative patients.
In the following examples, 161 breast cancer tissues were homogenized, fractionated and analyzed by Western blotting, a technique that can distinguish pl85HER-2 from its truncated cytoplasmic protein, p95HER-2. A study conducted by Tandon et al, (Tandon et al., J. Clin. Oncol. 7:1120-1128, 1989) also used Western analysis of breast tissue extracts, but Tandon et al. only evaluated the full length product, pl85HER-2. These data are consistent with the results reported in Tandon et al. These data in the examples also found pl 85HER-2 to be expressed frequently in breast tumors with a subpopulation of 21.7%, compared to Tandon et al's 16% that was scored as highly positive. These results are consistent.
The data in the examples herein show that breast cancers, which express HER-2/neu, are heterogeneous with respect to protein products. The distinct products, p95HER-2 and p185HER-2, were differentially associated with node status. While the group that overexpressed p 185HER-2 did not associate with node status (Table those that were pi
O
0 rich and contained p95HER-2 were significantly associated with lymph node metastasis (Table This may help explain why several previous studies, which have attempted to show association with lymph node metastasis based on assays of p185HER-2 protein overexpression 00 or HER-2/neu gene amplification, have yielded inconsistent results (see, for example, s 5 Singleton and Strickler, Pathol.Annual 27 Pt 1:165-198, 1992). Without being bound by theory, a biological explanation for these data is that loss of the ECD regulatory region from the p95HER-2 kinase, combined with amplified pi 85HER-2 signal in primary breast tumor C cells, promotes their metastasis, such as to the lymph nodes.
SP95HER-2 positive or pl85HER-2 highly positive samples did not correlate with other 10 prognostic markers in these data, including tumor size or hormone receptor status. While no O consistent correlation with tumor size has been detected, other studies have reported C association of HER-2/neu overexpression with ER and PR negativity (Singleton and Strickler, PathoLAnnual 27 Pt 1:165-198, 1992; Tandon et al., J Clin. Oncol. 7:1120-1128, 1989; and Carlomagno et al., J. Clin. Oncol. 14:2702-2708, 1996). Moreover, in contrast to the data reported herein, the relationship between HER-2 overexpression and hormone receptor status were examined in a subgroup of high-risk patients or in groups that were stratified by levels of hormone receptors (Tandon et al., Clin. Oncol. 7:1120-1128, 1989; and Carlomagno et al.; and J. Clin. Oncol. 14:2702-2708, 1996).
In conclusion, HER-2/neu overexpression in tumor tissue is a strong prognostic marker only in node positive patients (Slamon et al., Science 244:707-712, 1989; Singleton and Strickler, Pathol.Annual 27 Pt 1:165-198, 1992; Slamon et al., Science 235:177-182, 1987; Press et al., Progress Clinical Biological Research 354:209-221, 1990; Hynes et al., Biochenz. Biophys. Acta 1198:165-184, 1994; and Tandon et al., J. Clin. Oncol. 7:1120-1128, 1989). The data presented herein indicate that p95HER-2 is preferentially found in HER-2/neu positive patients with lymph node involvement. Higher expression of p95HER-2 is a critical factor that helps explain the increased prognostic significance of HER-2/neu in node positive patients.
Both ECD and p95HER-2 were at about 20 fold lower levels in SKOV3 ovarian carcinoma cells compared to BT474 breast carcinoma cells. Both were stimulated by treatment of cells with the phorbol ester tumor promoter (TPA) and the lysosomotrophic agent, chloroquine. The hydroxamate inhibitor ofmetalloproteases, TAPI, suppressed both 2 and ECD (HER-2/neu extracellular domain) in a dose-dependent fashion with maximal inhibition at 10 pM or less in BT474 cells.
Proteolytic release of the ECD is expected to create an N-terminally truncated, membrane-associated fragment with kinase activity.
P95HER-2 P95HER-2 is the C-terminal polypeptide fragment of p 85HER-2, whose complete sequence was first published in Coussens et al., Science 230:1132-1139, 1985. P185HER-2 is a 1255 amino acid polypeptide ending in Val residue at position 1255. The N-terminus of
O
p95HER-2 begins from about Asp at position 639 to about the Glu residue at position 645.
SMost likely, the N-terminal residue is Pro from position 643.
Hvdroxamate Compounds 00 The present invention further provides a method for treating carcinomas that overexpress HER-2, comprising administering a hydroximate compound, wherein the hydroximate compound is described in formula 1: o0 0 n II
I
X CH C N CH C N B -NH S010 R R2 R3 0 wherein: X is hydroxamic acid, thiol, phosphoryl or carboxyl; m is 0, 1 or 2; Ri, R2, and R3 is independently hydrogen, alkylene(cycloalkyl), OR 4 SRI, halogen, a substituted or unsubstituted C, to C 6 alkyl, C, to C6 alkylenearyl, aryl, a protected or unprotected side chain of a naturally occurring a-amino acid; or the group RR 7 wherein R 6 is substituted or unsubstituted C, to Cg alkyl and R7 is OR 4
SR
4 N(R4)(R 5 or halogen, wherein R4 and Rs are independently hydrogen or substituted or unsubstituted C, to Cs alkyl; wherein n is 0, 1 or 2; with a first proviso that when n is 1, A is a protected or an unprotected a-amino acid radical; and with a second proviso that when n is 2, A is the same or different protected or unprotected a-amino acid radical; and wherein B is an unsubstituted or substituted C2 to Ca alkylene.
Methods for synthesizing compounds of formula 1 are disclosed in U.S. Patent 5,629,285, the disclosure of which is incorporated by reference herein. Pharmaceutical formulations are compositions are also disclosed in U.S. Patent 5,629,285.
Example 1 This example illustrates the identification ofN-terminally truncated HER-2/neu protein with kinase activity. 3T3 cells were transfected with HER-2/neu cDNA (17-3-1 cells) (Applied BioTechnololgy, Inc. Cambridge, MA) and release soluble ECD by proteolytic processing of pl85HER-2/neu (Zabrecky et al., J. Biol. Chem. 266:1716-1720, 1991). To detect truncated cytoplasmic products, 17-3-1 extracts were resolved in gels and immunoblotted with antibodies against the C-terminus of the HER-2/neu product (anti-neu 17-3-1 Cells, were cultured in Dulbecco's modified Eagles medium (DMEM) supplemented with 5% fetal bovine serum containing 0.4 mg/ml geneticin (G418 GIBCO- BRL). Briefly, anti-neu has been described (Lin et al., Mol. Cell. Endocrin. 69:111-119, 1990). Monoclonal antibody against the extracellular domain of HER-2/neu was prepared as described (McKenzie et al., Oncogene 4:543-548, 1989) and was provided by Applied BioTechnology Inc. Briefly, freshly prepared cell lysates in TEDG buffer (50 mM Tris, mM EDTA, 0.5 mM dithiothreitol, 10% glycerol pH 7.5 with 1% aprotinin, 2 mM PMSF, and 2 mM vanadate) containing 1% Nonidet P-40 were immunoprecipitated by incubation with antibody for 2 hrs with continuous shaking at 4 'C as described (Lin et al., Mol. Cell Endocrin.
69:111-119, 1990). The inunune complexes, bound to Protein G Sepharose (Pharmacia), were
O
o washed twice with TEDG buffer and incubated 10 min on ice in a kinase reaction mixture F^ containing 20 mM HEPES pH 8.0, 2 mM dithiothreitol, 25 pM vanadate, 0.5% Nonidet mM MnCI2, 1 p.M ATP, and 15 pCi (y- 32 P) ATP (New England Nuclear). The immune 00 complexes were washed 3 times with buffer and the proteins were released by boiling for 2 C 5 min in SDS-PAGE sample buffer.
Two major protein products were detected in cell extracts; the full length p185 HER- -2/neu and a truncated protein of about 95 kDa (Figure 1, lane The extracts were cn immunoprecipitated and the 95 kDa protein, as well as p 185HER-2/neu, were phosphorylated S in the immune complex with (y-3P)ATP (Figure 1, lane A monoclonal antibody specific for the N-terminal region ofpl85HER-2/neu (anti-neu did not immunoprecipitate O p95HER-2, indicating that the N-terminal region was missing (Figure 1, lane Therefore, Sp95HER-2 is a fragment of p 85HER-2 and is no an N-terminal fragment.
Example 2 This example illustrates that p95HER-2 has self-phosphorylating activity and was not the substrate of the full length receptor tyrosine kinase. P 85HER-2 was first removed from the cell lysate with anti-neu and then p95HER-2 was immunoprecipitated with anti-neu as described in example 1. P95HER-2 was phosphorylated when pl85HER-2 levels were greatly depleted (Figure I lane These data indicate that p95HER-2 has kinase enzymatic activity.
Moreover, p95HER-2 kinase activity is in human breast carcinoma cells but not in nontumorigenic breast epithelial cells. The human breast carcinoma cell line, BT474, known to release soluble ECD (Lin and Clinton, Oncogene 6:639-643, 1991), also contains two autophosphorylated HER-2/neu products, pi 85HER-2 and p95HER-2. The human breast carcinoma cell line BT474 was cultured in RPMI medium supplemented with 10% FBS and 10g/ml insulin. Both were found at elevated levels compared to the nontumorigenic breast epithelial cell line HBL-100 (Figure It was possible that p95 could not be detected in the small amount of HBL-100 cells, since they express low levels ofpl85HER-2 (Kraus et al., EMBOJ. 6:605-610, 1987). To compensate for different levels of HER-2/neu expression, the amounts of extract from HBL-100, human mammary epithelial cells, (HMEC), and three breast carcinoma cell lines were adjusted and proteins were phosphorylated with (y- 32 P) ATP.
P95HER-2 was detected in the low (MDA-MB-453) and high (BT474 and SKBR3) HER- 2 /neu expressing breast carcinoma cells, but not in the HBL-100 nor HMEC cells, despite a robust signal from the HER-2/neu receptor which migrated as a slightly smaller protein in the breast epithelial cells (Figure 2).
Example 3 This example illustrates that p95HER-2 is a tyrosine phosphorylated polypeptide with kinase enzymatic activity and is located in the membrane fraction from BT474 cells. Tyrosine phosphorylation of tyrosine kinase receptors generally indicates their activation in signaling (Hynes et al., Biochem. Biophys. Acta 1198:165-184, 1994; and Dougall et al., Oncogene
I
O
O 9:2109-2123, 1994). The tyrosine phosphorylation of p95HER-2, and its subcellular location were examined by fractionation of BT474 cell extracts into a soluble fraction and a particulate fraction. Each fraction was immunoprecipitated with anti-neu and then subjected to 00 Western blot analysis using monoclonal antibodies against phosphotyrosine. Briefly, CN 5 following SDS-PAGE, cell lysates or proteins from concentrated, conditioned medium were electroblotted onto nitrocellulose (Trans-Blot, Bio-Rad) using a semi-dry transfer unit (Bio- Rad) at 15 volts for 20 min per mini gel of 0.75 mm thickness (Mini-PROTEAN II rn electrophoresis cell, BioRad) equilibrated with 25 mM Tris pH 8.3, 192 mM glycine, 50 mM S NaC, 20% methanol. Binding sites were blocked by incubating the membrane with 5% nonfat CN 10 dry milk. After incubation with the primary antibody, the blot was washed twice for 15 min O and 4 times for 5 min with Tris-buffered saline (TBS) containing 0.05% Tween and then S incubated for 40 min with goat anti-rabbit or goat anti-mouse antibody conjugated to horseradish peroxidase (HRP) (Bio-Rad) diluted in TBS-Twecn. After incubation with secondary antibody, the blot was washed as described above with TBS-Tween and developed IS with chemiluminescent reagent (Pierce).
Figure 3 illustrates that a tyrosine phosphorylated p95HER-2 fractionated with p185HER-2 in the particulate fraction. The particulate fraction contains the plasma membranes. P95HER-2 was further shown to be tyrosine phosphorylated by first immunoprecipitating with anti-phosphotyrosine antibodies and then probing the Western blot with anti-neu (data not illustrated).
Example 4 This example illustrates that p95HER-2 polypeptide intracellular levels corresponded to levels of soluble ECD released from different cells. To examine the relationship ofp95HER-2 to. soluble ECD, their levels were compared in different cells under varied conditions. The basal levels of ECD and cellular p95HER-2/neu were first examined in two cell lines that overexprss HER-2/neu, BT474 and the ovarian carcinoma cell line SKOV-3. Both cell lines were reported to produce low levels of ECD (Pupa et al., Oncogene 8:2917-2923, 1993).
The amount ofp95HER-2, relative to p 85HER-2 and to cell protein, was greatly elevated in BT474 cells. Correspondingly, the ECD in the extracellular medium from BT474 cells, detected with anti-neu was enhanced by greater than 10 fold compared to the SKOV3 cells (Figure 4).
Shedding of several membrane proteins is rapidly and transiently induced by phorbol ester tumor promoters (Ehlers and Riordan, Biochem. J. 321:265-279, 1997). While short term treatment with tumor promoters does not induce HER-2 shedding (Vecchi et al., J. BioL Chem.
271:18989-18995, 1996), chronic administration of the phorbol ester TPA synergized with chloroquine to stimulate release of soluble HER-2.
To determine whether p95HER-2 and ECD were coordinately regulated, TPA (500 nM) and chloroquine (50 IM) or the control vehicle were added to the culture media of BT474 and SKOV3 cells. SKOV3 cells were grown in DMEM supplemented with 10% FBS and the
O
O antibiotic gentamicin at 0.05%. At 24 hrs, the ECD levels in the extracellular media and p95HER-2 levels in the cell extract were analyzed- Soluble ECD was elevated several fold in the conditioned medium from stimulated BT474 cells and SKOV3 cells, while p95HER-2 was 00 upregulated about three-fold in BT474 cells (Figure Overexposure of the immunoblot Cl 5 revealed that p95HER-2 in SKOV3 cell extracts was also stimulated about three-fold by TPA and chloroquine (data not illustrated in figures).
Example C This example illustrates that a metalloprotease inhibitor depressed levels of p95HER-2 O and ECD from BT474 cells. Shedding of diverse transmembrane proteins is inhibited by l 10 hydroxamic acid-based compounds, which are potent metalloproteinase inhibitors (McGeehan S et al., Nature 370:561, 1994; Mohlcr et al., Nature 370:218-220, 1994; and Arribas et al., J 0 Biol. Chem. 271:11376-11382, 1996). Therefore, effects of different concentrations of the hydroxamic acid, TAPI (Mohler et al., Nature 370:218-220, 1994) was tested on shedding of HER-2/neu ECD and on cell levels of p95. TAPI (0 to 40 pM) was added to cultured BT474 cells for 24 hrs, the ECD in concentrated conditioned media was analyzed by immunoblotting with anti-neu and p95HER-2 and p185HER-2 polypeptides were examined in cell extracts using an anti-neu monoclonal antibody. The results in Figure 5 show that production of ECD was partially inhibited at a 1pM TAPI concentration and maximally inhibited at a 10 uM TAPI concentration. A residual amount of about 10% of the ECD resisted inhibition by even 40 pJM TAPI. The level of truncated p95HER-2 in the cytoplasm was also inhibited by TAPI, with little or no effect at a 1 pM concentration and maximal inhibition at a 10 ipM concentration (Figure These data were reproducible in another cell line.
In three separate experiments, 1 M TAPI inhibited ECD and p95HER-2 levels by or less, and in all cases, maximum inhibition was achieved by a 10 tM concentration of TAPI.
No change in pl85HER-2/neu levels could be detected in cells treated with TAPI or when shedding was stimulated by TPA and chloroquine (Figure Without being bound by theory, but these results are because proteolytic processsing ofpl85HER-2 is constitutive and limited with about 20% converted into soluble HER-2/neu in 2 hrs (Pupa et al., Oncogene 8:2917- 2923, 1993). TAPI also increased p95HER-2 in a cell line. However different mechanisms of action may apply.
Example 6 This example illustrates the detection ofpl 85HER-2 and p95HER-2 in breast cancer tissue. Tumor tissues were homogenized, fractionated, and examined for HER-2/neu proteins by Western analysis. Briefly, about 0.1 gm of tumor tissue, which had been fresh-frozen and stored at -70 OC, was minced on dry ice and suspended in TEDG buffer. Tissues were homogenized using a Brinkman polytron for 5-10 second bursts repeated 2-3 times with a chilled probe. Homogenates were centrifuged at 1500 x g for 10 min at 4 OC. The lipid layer was removed with a wooden stick and the supernatant was centrifuged for 20 min at 40,000 x g at 4 The lipid layer was collected with a wooden stick, the supernatant decanted, and the
I
O
O pellet containing the membranes was solubilized in TEDG buffer containing 0.1 %SDS for min with intermittent vortexing and clarified by centrifugation at 15,000 x g for 15 min. The t protein concentration in the superatant was determined by the Bio-Rad protein assay reagent S and aliquots were frozen at -80 °C.
Cl 5 P95HER-2 and p 185HER-2 in breast cancer tissue were analyzed according to the following method. About twenty pg of protein from the membrane fraction prepared from each tumor sample was resolved under denaturing and reducing conditions by SDS-PAGE in rn, 10% gels. Each gel also contained 3 pg of protein from extracts of 17-3-1 cells to mark the S migration ofpl85 and p95 and to provide a standard for the entire study. Proteins were CN 10 electro-transferred onto membranes as described above, which were incubated with anti-neu 0 diluted 1:10,000 in TBS-Tween at 4 °C overnight with shaking and then incubated with a 0 1:10,000 dilution of goat anti-rabbit HRP conjugated antibody (Bio-Rad) for 40 min at room temperature. To develop the blot, the membranes were incubated with chemilumenescent reagent (Pierce) for 5 min and then exposed to Kodak X-OMAT AR film for 1, 5, 20, and 120 min. To define the samples that overexpressed pl85HER-2/neu, specimens with HER-2 immunoassay values that were considered HER-2/neu-rich (400 units or greater) compared to samples with low HER-2/neu levels (less than 400 units) were characterized for their p1 85HER-2 signal relative to the control 17-3-1 cells by western analysis. Samples were scored as highly positive with a p 85HER-2 signal that could be detected by 1 min exposure of the membrane to film and that was equal to or greater than the pl85HER-2 levels found in 3 lg of 17-3-1 cells, as revealed by laser densitometric analysis of the film.
A HER-2/neu tissue extract ELISA assay was run on the extracted samples. Briefly, aliquots of membrane-rich fractions prepared from breast cancer tissue, as described above, were assayed using the Triton Diagnostics c-erbB-2 Tissue Extract EIA kit (Ciba Corning) according to manufacturer's instructions. This assay employs two monoclonal antibodies against the HER-2/neu ECD. The HER-2/neu units/mg protein in the specimens was calculated from a calibration curve generated by plotting the HER-2/neu concentration of the calibration standards versus the absorbance obtained from the immunoassay.
Clinical information on tissue from each patient included information for age, nodal status, size of the primary tumor, age of the patient, stage of disease at diagnosis, estrogen receptor (ER) levels and progesterone receptor (PR) receptor levels. Specimens were considered ER positive and PR positive if they contained at least 10 fmol specific binding sites permg of cytosolic proteins. The stage of the specimens included 1 at stage 0, 32 at stage I, 56 stage II, 45 stage III and 13 stage IV. Fourteen specimens were of unknown stage. The average age of the patients was 60. The 8 ovarian cancer tissues included 3 that were grade III and 5 that were grade IV.
Using this method, 21.7% of the samples overexpressed p 185HER-2. This proportion was comparable to the 15-30% of breast cancers found to overexpress HER-2/neu in numerous clinical studies. In the samples that had detectable p95HER-2, its level ranged from 10% to
I
O
O 100% ofp 85. In this study, specimens were scored as positive ifp95HER-2 was detected at a or greater proportion ofpI85HER-2 by 2 hrs of exposure of the membrane to film.
Because of the high titer of the primary antibody, anti-neu there were rarely background bands, even when the immunoblots were exposed to film for 2 hrs.
c 5 The membrane-enriched but not the soluble fraction (data not shown) from some tumor tissues contained the full-length product, p185HER-2, and the truncated p95HER-2/neu protein that co-migrated with HER-2/neu proteins from the control 17-3-1 cells (Figure In tt" addition, p95HER-2, along with pl85HER-2, was detected in 2 of 8 ovarian cancer tissues O (raw data not illustrated). Initial analyses of several breast cancer tissues revealed distinct C 10 expression patterns ofp95HER-2 and p 85HER-2. One group had no delectable pl85HER-2 S or p95HER-2 (see 39 and 69 in Figure A second category of tumor specimens O expressed both pl85HER-2 and p95HER-2 polypeptides 60, 53, 04, and 22). An additional group contained pl85HER-2 with relatively little or no p95HER-2 polypeptide expression 40, 58, 38, 57, 17, and 75). As observed in previous studies by others, some samples were pl85HER-2-rich 04, 22, 57, 17, and 75). The samples that were characterized as highly positive for pl85HER-2 were initially identified by imnnunoassay values of greater than 400 units. The results of the Western analysis indicated that the tumors were heterogeneous with respect to HER-2/neu protein products and that they can be subdivided based on the presence or absence of p95HER-2.
Western analysis of 161 breast cancer samples revealed that 22.4% were p95HER-2 positive. The p185HER-2 positive samples were further subdivided into "highly positive" or HER-2-rich specimeni based on comparisons with HER-2/neu overexpressing samples identified by immunoassay and comparisons with the control 17-3-1 extract. The "highly positive" pi 85HER-2 tumor samples represented 21.7% of the total. All of the tumor samples that expressed p95HER-2 were also positive for pl85HER-2, although 65% ofp185 positive tumor samples did not contain detectable levels ofp95HER-2 polypeptide. Of the p95HER-2 positive tumor samples, 63.9% were also highly positive for p185HER-2 and 36% had low p 185HER-2 levels. Therefore, intracellular p95HER-2 polypeptide detection appears to be a reliable prognosticator indicator.
Example 8 This example illustrates a relationship as between p95HER-2 positive tumor samples, pl85HER-2 highly positive tumor samples, and other prognostic factors of breast or ovarian cancer. Of 78 node-positive breast cancer patients, a higher proportion expressed p95HER-2 polypeptide in intracelluar tumor samples, than for the node negative patients (P=.032).
Moreover, pl85HER-2 rich samples had no significant association with node status (Table I).
Neither p95HER-2 positive nor pi 85HER-2 rich samples correlated significantly with other factors known to predict poor prognosis (McGuire et al., N. Engl. J. Med. 326:1756-1761, 1992) including estrogen receptor and progesterone receptor negativity or tumor size of 3 cm or greater (Table 1).
I I 0 o Table 1 Relationship between p95 positive, p185 highlypositive, and otherprognostic Cl factors' C %p 9 5 Factor Positive P value High Positive P value 00 Nodes .032 NSb Pos(78) 29.5 24.4 Neg(63) 14.3 22.2 S Tumor Size NS NS O _3cm(54) 27.8 22.2 C <3cm(79) 17.7 21.5 0 o 15 ER NS NS N Neg(37) 32.0 29.7 Pos(117) 19.7 17.9 PR NS NS Neg(59) 23.7 20.3 22.1 23.2 2 161 samples were examined by western analysis. Not all samples had information for the factors examined.
b NS=not significant.
Example 9 This example illustrates an influence ofp95HER-2 in the p185HER-2 highly positive group. This experiment began by asking the question why a similar percentage of node positive and node negative patients were pl 85HER-2-rich (24.4% versus 22.2%, Table 1), while p95HER-2 was associated with node positive patients, since 65.7% of the pl85HER-2rich samples contained p95HER-2. The experiment examined whether the presence or absence of p95HER-2 in the specimens that overexpressed p 185HER-2/neu affected the relationship with lymph node status (Table The p1 85HER-2 highly positive samples that contained p95HER-2 (n=21) had a significantly higher association with metastasis to the lymph nodes, while the p185HER-2 highly positive samples that were negative for p95HER-2 (n=l 1) were associated with lymph node negative patients (P=.017).
Table 2 Relationship between pl85 highly positive samples that are p95 negative versus positive with node status.
p185 highly positive" p95 positive p 9 5 negative n=21 n=1l node positive 71.4% b 27.3% node negative 28.6% 72.7% a The p185 highly positive group (n=32) was divided into those that contained p95 (n=21) and those that were p95 negative (n=l 1).
P:\OPER\jrc\Spccifaikon\2(X)4.Z(X)5 Diviiona\77v03-di,.d28/Al/() 0 -17- S°bThe samples that contained p95 had a significantly higher association with node 0, positive patients (15 of 21), and those that were p95 negative correlated with node negative patients (8 of 11) (P=.017).
n 5 The reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that that prior art forms part of the Scommon general knowledge in Australia.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.

Claims (29)

1. A method of treating cancer, comprising: administering to a patient determined to have a cancer characterized by over- Cc 5 expression of p185HER-2 an effective amount of a metalloprotease inhibitor, whereby Sproduction of p95HER-2 is decreased.
2. The compound of claim 1, wherein the metalloprotease inhibitor is a 0 hydroxamate compound.
3. The method of claim 1 or claim 2, wherein the cancer is selected from the group consisting of breast cancer, ovarian cancer, prostate cancer, cervical cancer, non- small lung cancer, gastric cancer, and combinations thereof.
4. The method of any of claims 1-3, wherein the patient has elevated serum levels of extracellular domain (ECD) shed from p 85HER-2. The method of any of claims 1-4, wherein the cancer also expresses p95HER-2.
6. The method of any of claims 1-5, wherein the metalloprotease inhibitor is a compound of formula 1: o o H II H H II H X-[CH]m-C-C-N-C-C -[A]n-N-B--NH 2 Formula 1 R 1 R 2 R 3 wherein: X is hydroxamic acid, thiol, phosphoryl or carboxyl; m is 1 or 2; R 2 and R 3 are independently hydrogen, alkylene(cycloalkyl), OR 4 SR 4 N(R 4 )(R 5 halogen, substituted or unsubstituted C 1 to C 6 alkyl, Ci to C 6 alkylenearyl, aryl, a protected or unprotected side chain of a naturally occurring a-amino acid; or the group -R 6 R 7 wherein R 6 is CI to C 8 alkyl and R 7 is OR 4 SR 4 N(R 4 )(R 5 or halogen, wherein R 4 and R 5 are independently hydrogen or substituted or unsubstituted CI to C 8 alkyl; n is 0, 1 or 2; provided that when n is 1, A is a protected or an unprotected a-amino acid radical; and provided that when n is 2, A is the same or different protected or unprotected a-amino acid radical; and PAOPER\RASWCI.inI256127O 2d Vpa 265 dm.31062O6 IO -19- O wherein B is an unsubstituted or substituted C 2 to Cg alklylene; and the IN pharmaceutically acceptable salts thereof.
7. The method of claim 6, wherein B is C 2 to C 6 alkylene.
8. The method of claim 7, wherein B is dimethylene.
9. The method of claim 6, wherein X is hydroxamic acid. The method of claim 6, wherein R' is hydrogen or CI to C 6 alkyl. t 11. The method of claim 8, wherein R' is hydrogen.
12. The method of claim 6, wherein R 2 is hydrogen or Ci to C 6 alkyl.
13. The method of claim 10, wherein R 2 is isobutyl.
14. The method of claim 6, wherein R 3 is selected from the group consisting of Ci to C 6 alkyl, C 1 to C 6 alkylenephenol, C 1 to C 6 alkylene(cycloalkyl) and C 1 to C 6 alkylenearyl. The method of claim 12, wherein R 3 is C 1 to C 6 alkyl.
16. The method of claim 13, wherein R 3 is t-butyl.
17. The method of claim 12, wherein R 3 is Ci to C 6 alkylenearyl.
18. The method of claim 15, wherein R 3 is methylene-(2'-naphthyl).
19. The method of claim 6, wherein A is an alanyl or seryl radical, and n is 1. The method of claim 17, wherein A is alanyl, and n is 0 or 1.
21. The method of any of claim 6, wherein the inhibitor is selected from among N- {D,L-2-(hydroxyaminocarbonyl)methyl-4-methylpentanoyl}-L-3-(2'-naphthyl)alanyl-L- alanine, 2-(amino)ethyl amide; N-{D,L-2-(hydroxyaminocarbonyl)methyl-4- methylpentanoyl}-L-3-amino-2-dimethylbutanoyl-L-alanine, 2-(amino)ethyl amide; and combinations thereof.
22. Use of a metalloprotease inhibitor for the formulation of a medicament for treatment of a cancer characterized by over-expression of p185HER-2, wherein the metalloprotease inhibitor decreases production of p95HER-2 from p185HER-2.
23. The use of claim 22, wherein the metalloprotease inhibitor is a hydroxamate compound.
24. The use of claim 22 or claim 23, wherein the cancer is selected from the group consisting of breast cancer, ovarian cancer, prostate cancer, cervical cancer, non- small lung cancer, gastric cancer, and combinations thereof. P.AOERRASCIaimsI265627O Ud pa 263 dmo.I2006 IO -q- O 25. The use of any of claims 22-24, wherein the patient has elevated serum ND levels of extracellular domain (ECD) shed from p185HER-2.
26. The use of any of claims 22-25, wherein the cancer also expresses 1 p95HER-2. m 5 27. The use of any of claims 22-26, wherein the metalloprotease inhibitor is a Scompound of formula 1: S0 0 H II H H II H X-[CH]m-C--CN-C-C-[A]n-N-B-NH2 Formula 1 R 1 R 2 R 3 wherein: X is hydroxamic acid, thiol, phosphoryl or carboxyl; m is 0, 1 or 2; R 2 and R 3 are independently hydrogen, alkylene(cycloalkyl), OR 4 SR 4 N(R 4 )(R 5 halogen, substituted or unsubstituted Ci to C 6 alkyl, Ci to C 6 alkylenearyl, aryl, a protected or unprotected side chain of a naturally occurring a-amino acid; or the group -R 6 R 7 wherein R 6 is Ci to C 8 alkyl and R 7 is OR 4 SR 4 N(R 4 )(R 5 or halogen, wherein R 4 and R 5 are independently hydrogen or substituted or unsubstituted C 1 to C 8 alkyl; n is 1, 0 or 2; provided that when n is 1, A is a protected or an unprotected a-amino acid radical; and provided that when n is 2, A is the same or different protected or unprotected a-amino acid radical; and B is an unsubstituted or substituted C 2 to C 8 alkylene; and the pharmaceutically acceptable salt thereof.
28. The use of claim 27, wherein B is C 2 to C 6 alkylene.
29. The use of claim 28, wherein B is dimethylene. The use of claim 27, wherein X is hydroxamic acid.
31. The use of claim 27, wherein R' is hydrogen or Ci to C 6 alkyl.
32. The use of claim 29, wherein R' is hydrogen.
33. The use of claim 27, wherein R 2 is hydrogen or C 1 to C 6 alkyl.
34. The use of claim 31, wherein R 2 is isobutyl. The use of claim 27, wherein R 3 is selected from the group consisting of C PAOPMRRASTCI.inu0I256I27O 2.d "p 265 doc.5II012(X6 -21- Q to C 6 alkyl, C 1 I to C 6 alkylenephenol, C 1 I to C 6 alkylene(cycloalkyl) and C, to C 6 IND alkylenearyl.
36. The use of claim 33, wherein R 3 is C 1 to C 6 alkyl.
37. The use of claim 34, wherein R 3 is t-butyl.
538. The use of claim 33, wherein R 3 sC to C 6 alkylenearyl. 39. The use of claim 36, wherein R 3 is methylene-(2'-naphthyl). The use of claim 27, wherein A is an alanyl or seryl radical, and n is 1. 41. The use of claim 38, wherein A is alanyl, and n isO0 or 1. 42. The use of claim 27, wherein the inhibitor is selected from among N-{D,L- 2-(hydroxyaminocarbonyl)methyl-4-methylpentanoyl }-L-3-(2'-naphthyl)alanyl-L-alanine, 2-(amino)ethyl amide; N- {D,L-2-(hydroxyaminocarbonyl)methyl-4-methylpentanoyl)}-L- 3-amino-2-dimethylbutanoyl-L-alanine, 2-(amino)ethyl amide; and combinations thereof. 43. A method according to any one of claims 1-21 or use according to any one of claims 22-42 substantially as hereinbefore described with reference to the Figures and/or Examples.
AU2005200354A 1998-11-13 2005-01-28 N-terminally truncated HER-2/neu protein as a cancer prognostic indicator Ceased AU2005200354B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/192206 1998-11-13
PCT/US1999/027010 WO2000029609A1 (en) 1998-11-13 1999-11-13 N-terminally truncated her-2/neu protein as a cancer prognostic indicator
AU16246/00A AU777803B2 (en) 1998-11-13 1999-11-13 N-terminally truncated HER-2/neu protein as a cancer prognostic indicator

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU16246/00A Division AU777803B2 (en) 1998-11-13 1999-11-13 N-terminally truncated HER-2/neu protein as a cancer prognostic indicator

Publications (3)

Publication Number Publication Date
AU2005200354C1 AU2005200354C1 (en) 2005-02-24
AU2005200354A1 AU2005200354A1 (en) 2005-02-24
AU2005200354B2 true AU2005200354B2 (en) 2006-11-16

Family

ID=39367006

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2005200354A Ceased AU2005200354B2 (en) 1998-11-13 2005-01-28 N-terminally truncated HER-2/neu protein as a cancer prognostic indicator

Country Status (1)

Country Link
AU (1) AU2005200354B2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993021942A2 (en) * 1992-05-01 1993-11-11 British Biotech Pharmaceuticals Limited Use of mmp inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993021942A2 (en) * 1992-05-01 1993-11-11 British Biotech Pharmaceuticals Limited Use of mmp inhibitors

Also Published As

Publication number Publication date
AU2005200354C1 (en) 2005-02-24
AU2005200354A1 (en) 2005-02-24

Similar Documents

Publication Publication Date Title
US8093216B2 (en) Method of treating cancer by inhibition of p95HER-2 production
Christianson et al. NH2-terminally truncated HER-2/neu protein: relationship with shedding of the extracellular domain and with prognostic factors in breast cancer
Price‐Schiavi et al. Rat Muc4 (sialomucin complex) reduces binding of anti‐ErbB2 antibodies to tumor cell surfaces, a potential mechanism for herceptin resistance
Jones HER4 intracellular domain (4ICD) activity in the developing mammary gland and breast cancer
Shaoyan et al. Downregulation of EIF4A2 in non–small-cell lung cancer associates with poor prognosis
Zhou et al. VEGF and EMMPRIN expression correlates with survival of patients with osteosarcoma
EP2097754A1 (en) Activated her3 as a marker for predicting therapeutic efficacy
CA2764386A1 (en) P95-her2 antibodies and uses thereof
Ghedini et al. Shed HER2 extracellular domain in HER2‐mediated tumor growth and in trastuzumab susceptibility
Kim et al. Caveolin-1 phosphorylation in human squamous and epidermoid carcinoma cells: dependence on ErbB1 expression and Src activation
Kabbage et al. Expression of the molecular chaperone αB-crystallin in infiltrating ductal breast carcinomas and the significance thereof: an immunohistochemical and proteomics-based strategy
Maramotti et al. Soluble epidermal growth factor receptor isoforms in non-small cell lung cancer tissue and in blood
WO2003073909A2 (en) Assays to monitor levels of epidermal growth factor receptor (egfr) extracellular domain (ecd) in cancer patients
Ocaña et al. Trastuzumab and antiestrogen therapy: focus on mechanisms of action and resistance
Chahed et al. Detection of protein alterations in male breast cancer using two dimensional gel electrophoresis and mass spectrometry: the involvement of several pathways in tumorigenesis
Hudelist et al. Serum EGFR levels and efficacy of trastuzumab-based therapy in patients with metastatic breast cancer
AU2005200354B2 (en) N-terminally truncated HER-2/neu protein as a cancer prognostic indicator
AU2001256353A1 (en) Cancer diagnosis and assays for screening anti-cancer agents
Daponte et al. L1 (CAM)(CD171) in ovarian serous neoplasms.
KR20180013878A (en) Biological markers for combination therapy including lenbactinib and everolimus
US20140106375A1 (en) Secreted Tumor-Associated Cytochrome as a Blood-Based Biomarker for Cancer
US20170010287A1 (en) Diagnosis of cancer by detecting auto-antibodies against epidermal growth factor (egf)
US6995243B2 (en) Antibodies that recognize and bind phosphorylated human glucocorticoid receptor and methods of using same
EP3102947B1 (en) Diagnosis of cancer by detecting auto-antibodies against par1
KR101127225B1 (en) Biomarker of Urothelial Cancer and the Diagnostic Method of the Same

Legal Events

Date Code Title Description
DA2 Applications for amendment section 104

Free format text: THE NATURE OF THE AMENDMENT IS AS SHOWN IN THE STATEMENT(S) FILED 23 JAN 2007.

FGA Letters patent sealed or granted (standard patent)
DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS AS SHOWN IN THE STATEMENT(S) FILED 23 JAN 2007

TC Change of applicant's name (sec. 104)

Owner name: OREGON HEALTH AND SCIENCES UNIVERSITY

Free format text: FORMER NAME: OREGON HEALTH SCIENCES UNIVERSITY

MK14 Patent ceased section 143(a) (annual fees not paid) or expired