throbber
Mol. Cells, Vol. 9, No.1 , pp. 31-36
`
`Molecules
`and
`Cells
`
`© Springer-Verlag 1999
`
`Luteinizing Hormone Releasing Hormone-RNase A Conjugates
`Specifically Inhibit the Proliferation of LHRH-Receptor-Positive
`Human Prostate and Breast Tumor Cells
`
`Yong Song GhoT and Chi-Born Chae*
`Department of Life Science, Pohang University of Science and Technology, Pohang 790-784, Korea.
`
`(Received on July 11, 1998)
`
`Human prostate and breast tumor cells produce
`luteinizing hormone-releasing hormone (LHRH)
`receptors on their cell surface even when they have lost
`dependency on sex steroid hormones for growth. To
`investigate whether LHRH can be used as a cell(cid:173)
`binding moiety to deliver toxin molecules into prostate
`and breast tumor cells, LHRH-bovine RNase A
`conjugates were constructed using the chemical cross(cid:173)
`linking method. The treatment of the LHRH receptor(cid:173)
`positive cells such as prostate LNCapFGC and breast
`MCF7 tumor cells with LHRH-RNase A conjugates
`resulted in a dose-dependent inhibition of growth. The
`cytotoxic activities of these conjugates were effectively
`reduced by the presence of exogenous LHRH. Either
`free RNase A or LHRH alone did not affect the
`proliferation of these cells. The LHRH-RNase A
`conjugates did not show cytotoxicity against FRTLS
`and TM4 cells which do not express the LHRH
`receptors. These results suggest that LHRH can be used
`as a cell-binding molecule for the specific delivery of
`toxin molecules into the cells which express LHRH
`receptors on their surface. Thus, a new class of
`biomedicines that act as fusion proteins between LHRH
`and toxins will give us a new avenue for the treatment
`of human prostate and breast cancers, regardless of
`their steroid hormone dependency.
`
`Keywords: Breast; LHRH; Prostate; RNase A; Tumor.
`
`Introduction
`
`An immunotoxin (IT) is a cytotoxic agent that consists of
`a cell-binding moiety and a toxin moiety. The cell-binding
`moiety can be an antibody, a growth factor, or a hormone
`that binds selectively to the cell surface molecules of
`certain cell types. The toxin moiety can be a naturally
`occurring Pseudomonas exotoxin, diphtheria toxin, or
`plant toxin . These immunotoxins can kill cells with
`selectivity in tissue culture and in animal models (Aaron
`and Phillip, 1981 ; Ahmann et ai., 1987). However, the
`toxins derived from bacteria are very immunogenic
`molecules and some immunotoxins are toxic to the liver
`because of the nonspecific uptake by the liver (Bajusz et
`ai., 1989; Beintema et ai., 1984). The use of a member of
`the human ribonuclease A superfamily such as serum
`protein angiogenin may alleviate the problems caused by
`the immunogenicity and liver toxicity (Bond, 1988 ;
`Brinkmann et ai., 1993; Denis and Mahler, 1990; Fekete
`et aI., 1989). When angiogenin was fu sed to the transferrin
`or anti-transferrin receptor monoclonal antibody by
`chemical cross-linking or recombinant methods, these
`immunotoxins exhibit cell type-specific cytotoxic activity
`(Denis and Mahler, 1990; Fekete et aI. , 1989). In contrast,
`angiogenin alone does not have any cytotoxicity at
`appropriate concentrations.
`LHRH is a decapeptide hormone released by the
`hypothalamus and acts on gonadotropes at the anterior
`pituitary to stimulate the release of the luteinizing hormone
`(LH) and follicle-stimulating hormone (FSH). Potent
`agonists or antagonists of LHRH have been used for the
`
`* To whom correspondence should be addressed.
`Tel: 82-562-279-2125; Fax: 82-562-279-2199
`E-mail: cbchae@postech.ac.kr
`
`t Present address: Cell Biology Section, National Institute of
`Dental Research, MIH, Bethesda, MD 20892, USA.
`
`Abbreviations : EDC , l-ethyl-3-(3-dimethylamino propyl)
`carbodiimide·HCl; FSH, follicle-stimulating hormone; IT,
`immunotoxin ; LH, luteinizing hormone; LHRH, luteinizing
`hormone-releasing hormone.
`
`IMMUNOGE 2156, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`32
`
`Killing Tumor Cells by LHRH-RNase A
`
`treatment of sex steroid hormone-dependent tumors such
`as prostate and breast cancers (Isaacs and Coffey, 1981).
`The suppression of gonadal steroid secretion by hormonal
`therapy with these agonists and antagonists retards the
`growth of these hormone-dependent tumors . However,
`these hormone analogs may also act directly on the tumors.
`Human prostate and breast cancer cells produce LHRH
`receptors on their cell surface even when they have lost
`their dependency on sex steroid hormones for growth, and
`LHRH agonists or antagonists can inhibit the growth of
`some breast cancer cell lines (Jennes et a!., 1988; Miller et
`al., 1985 ; Newton et al., 1994). Highly potent
`metallopeptide analogs of LHRH show selective
`cytotoxicity against human prostate and breast cancer cell
`lines in vitro, and LHRH analogs containing cytotoxic
`radicals inhibit the growth of estrogen-independent MXT
`mouse mammary carcinoma cells in vivo (Newton et a!.,
`1996; Pai et a!., 1992). The LHRH analogs containing
`cytotoxic agents may be useful for the treatment of human
`prostate and breast cancer, especially for the hormone(cid:173)
`independent tumors because hormonal therapy does not
`prevent the growth of hormone-independent cells (Pai et
`al. , 1990). These results implicate that LHRH can be used
`as a cell-binding moiety to deliver toxin molecules such as
`ribonuclease into prostate and breast tumor cells regardless
`of their steroid hormone dependency for growth.
`To investigate this possibility, LHRH-bovine RNase A
`conjugates were constructed using the chemical cross(cid:173)
`linking method. According to our preliminary experiments,
`these conjugates show cell type-specific cytotoxicity, and
`the cytotoxicity of these conjugates is abolished by the
`presence of excess LHRH. The res ults suggest that
`decapeptide LHRH can be successfully used as a cell(cid:173)
`binding molecule for the specific delivery of toxin
`molecules into the cells which express LHRH receptors on
`their surface. Thus, construction of the fusion protein
`between LHRH and a member of the human ribonuclease
`superfamily such as pancreatic RNase A, angiogenin and
`eosinoprul-derived neurotoxin will give us a new avenue to
`treat the human prostate and breast cancers regardless of
`their steroid hormone dependency.
`
`Materials and Methods
`
`Chemical conjugation of LHRH and RNase A Bovine
`pancreatic RNase A (2 mg, Sigma) and LHRH free acid (5 mg,
`Bachem California) were dissolved together in 0.8 mJ of water,
`and the pH of the mixture was adjusted to 7-8. Next, l-ethyl-3-
`(3 -dimethylamino propyl) carbodiimide·HCI (100 mg, EOC,
`Pierce) freshly dissolved in 0.2 ml of water, was added . The
`reaction was permitted to proceed with gentle agitation at room
`temperature for 2 h and terminated by gel filtration on a POlO
`column (Pharmacia) to remove the unreacted EDC and LHRH.
`The sample was further dialyzed against water for 24 h with three
`changes. The resultin g LHRH-RNase A conjugates were
`
`aliquoted and stored at -70°C until use. The amount of
`conjugated protein was quantified by Bio-Rad protein assay
`reagents (Bio-Rad Laboratories) using bovine serum albumin as
`a standard.
`
`Assay of ribonucJeolytic activity The ribonucleolytic activity
`was determined in a reaction mixture (0.1 ml) containing 4 mg/ml
`of yeast tRNA (type X, Sigma), 10 ~ of PMSF, 0.1 mg/ml of
`human serum albumin (Calbiochem), 33 mM HEPES/NaOH pH
`7.5, 33 mM NaCl , and appropriate concentrations of RNase A or
`LHRH-RNase A conjugates. After 15 min incubation at 37°C, the
`reaction was termi nated by the addition of 0.24 ml of 3.4% ice(cid:173)
`cold perchloric acid. The samples were kept on ice for 10 min and
`then centrifuged for 10 min at 4°C. Absorbance of the supernatant
`was determined at 260 nm (Ramakrishnan et al., 1992). Tbe
`activity of LHRH-RNase A conjugates was compared with that of
`non-conjugated RNase A. All experiments were carried out in
`duplicates.
`
`Cell culture LNCap·FGC human prostate adenocarcinoma
`cells (ATCC CRL 1740) were grown in -RPMI 1640 with 10%
`fetal bovine serum. MCF7 human breast adenocarcinoma cells
`(ATCC HTB 22) were cultured in Eagle's MEM with 10% fetal
`bovine serum. TM3 mouse Leydig cells (ATCC CRL 1714) and
`TM4 mouse Sertoli cells (ATCC CRL 1715) were grown in a 1: 1
`mixture of Ham's F12 medium with Oulbecco's modified Eagle's
`medium containing 5% horse serum and 2.5% fetal bovine serum.
`FRTL-5 rat tbyroid cells (ATCC CRL 8305) were cultured in
`Coon 's modified Ham's F12 medium with 5% bovine calf serum.
`Cells were subcultured after trypsinization , and the medium was
`changed every 2-3 d.
`
`Cytotoxicity assay Each well of a tissue culture plate (96 well,
`Nunc) was plated with 104 cells. After incubation at 37°C for 2 d,
`100 III of varying amounts of LHRH-RNase A conjugates, LHRH
`or RNase A samples in culture medium was added to each well.
`After incubation for two additional days, the number of viable
`ce ll s was determined usi ng Cell Titer 96 Non-Radioactive
`Proliferation assay kit (Promega) according to the manufacturer's
`instructions. Each experiment was carried out in triplicates, and
`an average value was used to determine percent viability.
`A time course of the action of LHRH-RNase A conjugates
`against LNCapFGC and MCF7 cells was investigated as follows .
`Cells were placed in a 96-well tissue culture plate at 103 cells per
`well and incubated for 2 d. The cells were grown for 2, 5, and 9
`additional days in the presence of 111M of LHRH-RNase A
`conjugates.
`
`Results
`
`Preparation of LHRH-RNase A conjugates Natural
`LHRH does not have any free carboxyl or amino groups
`(pGl u- His-Trp-Ser-Tyr-Gly-Leu- Arg-Pro-Gly-NH 2) .
`Therefore, bovine pancreatic ribonuclease A (RNase A)
`was coupled to LHRH free acid which contains a Gly-free
`carboxyl group at the C-terminus instead of Gly-arnide
`using the heterobifunctional crosslinking agent EDC. Due
`to the difficulty of separation of LHRH-RNase A
`
`IMMUNOGE 2156, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Yong Song Gbo & Chi-Born Cbae
`
`33
`
`conjugates from RNase, RNase A was reacted with a
`3S-fold molar excess of LHRR free acid. The unreacted
`LHRH and EDC were separated from the conj ugates by gel
`fi ltration on a PD 10 desalting column. Successful
`conjugation between the reactants was confirmed by SDS(cid:173)
`PAGE and Western blotting using monoclonal antiLHRH
`antibody (data not shown). The ribonucleolytic activity of
`LHRH-RNase A was lO-fold lower than that of RNase A.
`This reduction of enzymatic activity of the conjugates may
`be due to the modification of some lysine at the active
`center of RNase A (Rodriguez et aI., 1993).
`
`The cell type-specific cytotoxic effect of LHRH-RNase
`A conjugates To investigate whether LHRH-RNase A
`conjugates can inhibit proliferation of the cells which
`retain LHRH receptors on their surface, human prostate
`adenocarcinoma LNCapFGC cells and human breast
`adenocarcinoma MCF7 cells were treated with varying
`amounts of conjugates for 2 d. As shown in Figs. 1 and 2,
`the conj ugate s inhibit the proliferation of these
`cell s in a concentration -dependent manner, and the
`concentrations of conj ugates giving a SO% inhibition of
`cell proliferation were found to be -O.S )lM and -0.7 )lM
`for LNCapFGC and MCF7 cells, respectively. However,
`RNase A and LHRH alone do not affect the growth of both
`cells at up to 10 )lM. Therefore, the chemical attachment of
`LHRH to RNase A is needed for the cytotoxicity of these
`conjugates.
`To demonstrate that the cytotoxicity of LHRH-RNase A
`conj ugates was due to the binding to the LHRH receptor,
`two types of experiments were carried out. First, the
`cytotoxic activity of varying concentrations of conjugates
`against LNCapFGC and MCF7 cells was investigated in
`the prese nce of 10 nM of LHRH. Exogenous LHRH
`effectively reduces the cytotoxicity of LHRH-RNase A
`conjugates against these cells (Figs. 1 and 2). Secondly, we
`investigated whether LHRH-RNase A conjugates could be
`delivered into specific cells which express LHRH receptors
`on their surface. The conj ugates had no cytotoxic activities
`against LHRH-receptor-negative Sertoli TM4 cells and
`thyroid FRTLS cells (Fig. 3). However, the proliferation of
`Leydig TM3 cells which express LHRH receptors on their
`cell surface was also inhibited by the presence of
`conjugates.
`
`__ LHRH-RNase A
`-0-- RNase A
`_____ LHRH
`---'V- LHRH-RNase A + LHRH (10 nM)
`
`120
`
`100
`
`80
`
`60
`
`40
`
`20
`
`<a
`>
`.~
`
`:::l
`(/)
`~
`
`-10
`
`-9
`
`-8
`
`-7
`
`-6
`
`-5
`
`-4
`
`Log [Concent rat ion]
`
`Fig_ L Cytotoxicity of LHRH-RNase A conju gates against
`LNCapFGC prostate cancer cells. LNCapFGC cells were treated
`with varying concentrations of LHRH-RNase A, RNase A and
`LHRH for 2 d and percentage viability was determined as
`described in Materials and Methods. The cytotoxic activity of
`varying concentrations of LHRH-RNase A conj ugates against
`LNCapFGC cells was also investigated in the presence of 10 nM
`of LHRH.
`
`- - LHRH-RNase A
`-0-- RNaseA
`_____ LHRH
`---'V- LHRH-RNase A + LHRH (10nM)
`
`T
`
`120
`
`100
`
`80
`
`60
`
`40
`
`20
`
`<a
`>
`.~
`
`:::l
`(/)
`~
`
`Prolonged inhibition of cell proliferation by LHRH(cid:173)
`RNase A conjugates We investigated a time course of
`the cytotoxic activity of LHRH-RNase A conj ugates
`against LNCapFGC and MCF7 cells by growing the cells
`for 2, S, and 9 d in the presence of conj ugates. The
`inhibition of the growth of LNCapFGC and MCF7 cells
`continued for 9 d. This effect is not due to the possible
`deterioration of culture medium since the control cells
`grew continuously in the absence of conj ugates for 9 d in
`the culture medium without replacement (Fig. 4).
`
`·10
`
`-9
`
`-8
`
`-7
`
`-6
`
`-5
`
`-4
`
`Log [Concentration]
`
`Fig. 2. Cytotoxicity of LHRH-RNase A conjugates against
`MCF7 breast cancer cells. MCF7 cells grown in culture were
`treated with varying concentrations of LHRH-RNase A, RNase A
`and LHRH for 2 d and percentage viability was determined as
`described in Materials and Methods. The cytotoxic activity of
`varying concentrations of LHRH-RNase A conjugates against
`MCF7 cells was also investigated in the presence of 10 nM of
`LHRH.
`
`IMMUNOGE 2156, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`34
`
`120
`
`100
`
`80
`
`60
`
`40
`
`20
`
`IV
`>
`.~
`
`:::l en
`<f!.
`
`·10
`
`·9
`
`·8
`
`·7
`
`·6
`
`·5
`
`-4
`
`Log [Concentratio n]
`
`Fig. 3. Cell type-specific cytotoxic activities of LHRH-RNase A
`conjugates . Leydig TM3 , Sertoli TM4, and thyroid FRTL5 cells
`grown in culture were treated with varying amounts of LHRH(cid:173)
`RN ase A conjugates for 2 d and percentage viability was
`determined as described in Materials and Methods.
`
`---- LNCapFGC
`-0- LNCapFGC + LHRH-RNase A
`--.- MCF7
`-<r- MCF7 + LHRH-RNase A
`
`T
`
`0 .... OJ) <
`
`2.0
`
`1.6
`
`1.2
`
`.8
`
`.4
`
`0.0
`
`2
`
`4
`
`8
`
`10
`
`Incubation Days
`
`Fig. 4. Time course of the action of LHRH-RNase A conjugates.
`LNCapFGC and MCF7 cells were treated with 1 ~M of LHRH(cid:173)
`RNase A for 2, 5, and 9 d and viable cells were determined as
`described in Material s and Methods. For control experiments,
`LNCapFGC and MCF7 cells were grown in the absence of
`LHRH-RNase A conjugates.
`
`Killing Tumor Cells by LHRH-RNase A
`
`---- TMJ
`-0- TM4
`--.- FRTL5
`
`Discussion
`
`Prostate and breast cancers are the major cancers leading to
`death in American men and women, respectively. The
`functions of the prostate and breast are regulated by steroid
`hormones such as androgen and estrogen, respectively.
`Furthermore, these sex hormones are necessary for the
`growth of hormone-dependent human prostate and breast
`tumors. Available options for the treatment of prostate and
`breast cancers are surgical orchiectomy, radiation therapy,
`hormone treatment and cytotoxic chemotherapy.
`Administration of analogs of the natural decapeptide
`LHRH stimulates the secretion of LH and FSH by
`gonadotropes. When chronic administration is continued,
`LH and FSH secretion is decreased by desensitization of
`the LHRH receptors in the gonadotropes, leading to a
`decrease of circulating steroid hormones and thus
`inhibiting the growth of prostate and breast cancers
`(FitzGerald and Gordon, 1992; Isaacs et al., 1981).
`However, surgical and hormonal combination therapy to
`eliminate the testicular testosterone or the ovarian estrogen
`leaves an opportunity for the adrenal androgen and
`estrogen secreted by adrenal glands to stimulate the growth
`of prostate and breast cancers. Furthermore, hormone(cid:173)
`independent prostate cancer (androgen-independent) and
`breast cancer (estrogen-independent) can still grow after
`these types of combinational treatments have been applied.
`In the case of cytotoxic chemotherapy, the standard
`cytotoxic agents that are normally used against other
`cancers primarily act against rapidly growing tumor cells,
`but prostate cancer is not a rapidly growing tumor. It is not
`clear whether some combination of chemotherapeutic
`agents which is effective for the treatment of hormone(cid:173)
`independent tumors will be found. It seems that it is time
`to try some new avenues. In this sense, the hormone-toxin
`approach is intriguing.
`The purpose for the construction of LHRH-RNase A
`conjugates by chemical conjugation was to evaluate the
`potential of these conjugates to kill human prostate and
`breast cancer cells which express LHRH-receptors. We
`found that the LHRH-RNase A conjugates can selectively
`kill the LHRH receptor-positive cells such as Leydig TM3 ,
`prostate LNCapFGC and breast MCF7 tumor cells but not
`FRTLS and TM4 cells which do not express the LHRH
`receptors. Extracellular RNase A or LHRH alone is not
`cytotoxic toward LNCapFGC and MCF7 cells. Also, the
`cytotoxic activities of these conjugates can be effectively
`reduced by the presence of exogenous LHRH. These
`results clearly indicate that LHRH-RNase A conjugates can
`kill the specific cell types which express the LHRH(cid:173)
`receptors and that the cell type-specific cytotoxic activities
`are due to the delivery of RNase A to the cytosol via
`LHRH receptor-mediated endocytosis.
`The relatively high ICso value of these conjugates might
`be due to the fact that the chemically linked LHRH-RNase
`
`IMMUNOGE 2156, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Yong Song Gho & Chi-Born Chae
`
`35
`
`A conjugates are heterogeneous and that the ribonuclease
`activity of the conjugate was 10-fold lower than that of
`natural RNase A . According to prev ious reports, the
`recombinant fusion protein between ribonuclease and the
`monoclonal antibody to the tran sferrin receptor was a
`thousand times as potent as the chemically linked hybrid
`(Bond, 1988; Denis and Mahler, 1990). Thus, construction
`of the fusion protein between LHRH and a member of the
`human ribonuclease superfamily such as pancreatic RNase
`A , angiogenin and eosinophil-derived neurotoxin
`eliminates the heterogeneity of chemically cross-linked
`LHRH-RNase A conjugates and may increase the cell type(cid:173)
`s pec ifi c cytotoxicity. Although seve ral kind s of
`immunotoxins which show the cytotoxic activity against
`human prostate and breast cancer in vitro and in vivo have
`been developed as a fusion protein between the specific
`antibody for target cells and bacterial or plant toxin as a
`toxin moiety (Rybak et aI. , 1991 ; 1992; Sharoni et aI.,
`1989), recombinant LHRH-RNase toxins may have several
`potential therapeutic advantages to immunotoxins for the
`following reasons. In contrast to previously developed
`immunotoxins, these new biomedicines may not cause
`immunogenesis and liver toxicity. These LHRH-RNase
`toxins may penetrate easily into the solid tumor due to
`their small size. Both Leydig cells in the testis and
`Granulosa cel ls in the ovary al so express the LHRH(cid:173)
`receptor on their surfaces (Isaacs et aI., 1981). These cells
`are major sources of androgen and estrogen which are
`needed for the growth of prostate and breast cancer cells,
`respectively. Thus, LHRH-RNase toxins may not only
`selectively kill the prostate and breast cancer cells, but also
`effectively eliminate the circulating androgen and ovarian
`estrogen by eliminating either the Leydig cells in men, or
`the Granulosa cells in women. LHRH receptors are also
`present in the central nervous system (Szepeshazi et aI.,
`1992). However, LHRH agonist has been used to treat
`human prostate and breast cancers and LHRH-cytotoxic
`radicals produce significant inhibition of tumor growth
`without visible effect on the brain function (Pai et aI. ,
`1992; Vitetta et aI., 1983). This means that the circulating
`LHRH agonist and LHRH-toxin may not penetrate the
`brain-blood barrier. Similarly, LHRH-RNase toxins may
`not reach the brain nor cause any side effect on the brain
`function.
`In conclusion, LHRH-RNase A conjugates which may
`not have immunogenic problems or liver toxicity will give
`us a new avenue for the treatment of human prostate and
`breast cancers regardless of their steroid hormone(cid:173)
`dependency. In addition, its combination with surgical and
`honnonal therapy which have been used for the treatment
`of these cancers may increase their therapeutic potency.
`
`Acknowledgments This work was supported in part by
`Pohang University of Science and Technology and the Korea
`Green Cross, Inc.
`
`References
`
`Aaron, 1. W. and Phillip, B. C. (1981) Extrapituitary actions of
`gonadotropin-releasing hormone. Endocrinology 2, 437-46l.
`Ahmann, F. R. , Citrin, D. L., and DeHaan, H. A. (1987) Zoladex:
`a sustained release luteinizing hormone-releasing hormone
`analog for the treatment of advanced prostate cancer. 1. Clin.
`Oncol. 5, 912-917.
`Bajusz, S., Janaky, T. , Csemus, V. J., Bokser, L., Fekete, M. ,
`Srkalovic, G. , Redding, T. w., and Schally, A. V. (1989)
`Highly potent metallopeptide ana logues of luteini zin g
`hormone-releasing hormone. Proc. Natl. Acad. Sci. USA 86,
`6313-6317.
`Beintema, J. J. , Wietzes, P., Weickman, J. L., and Glitz, D. G.
`(1984) The amino acid sequen ce of hum an pan creati c
`ribonuclease. Anal. Biochem. 136, 48-64.
`Bond, M. D. (1988) An in vitro binding assay for angiogenin
`using placental ribonuclease inhibitor. Anal. Biochem. 173,
`166- 173.
`Brinkmann, U., Gallo, M., Brinkmann , E. , Kunwar, S., and
`Pastan , 1. (1993) A recombinant active on prostate cancer cell s
`and that is composed of the Fv region of monoclonal antibody
`PRI and a truncated form of Pseudomonas exotoxin. Proc.
`Natl. Acad. Sci. USA 90, 547-55l.
`Denis, L. and Mahler, C. ( 1990) Prostatic cancer: An overview.
`Rev. Oncol. 3, 665-677.
`Fekete, M., Redding, T. w., Comaru-Schally, A. M., Pontes, 1. E.,
`Connelly, R. w., Srkalovi c, G., and Schally, A. V. (1989)
`Luteini zin g hormon e-releas ing horm one, so matostatin ,
`prolactin , and epidermal growth factor in rat and human
`prostate cancers and in benign prostate hyperplasia. Prostate
`14, 191-208.
`Isaacs, J. T. and Coffey, D. S. (198 1) Adaptation versus selecti on
`as the mechanism responsible for the relapse of prostatic
`cancer to androgen ablation therapy as studied in the dunning
`R-3327-H adenocarcinoma. Cancer Res. 41, 5070-5075.
`Jennes, L., Dalati, B., and Conn, P. M. (1988) Distribution of
`gonadotropin releasing hormone agonist binding sites in the
`rat central nervous system. Brain Res. 452, 156--164.
`Miller, W. R., Scott, W. N. , and Morris, R. (1985) Growth of
`human breast cancer ceLIs inhibited by a luteini zing hormone(cid:173)
`releasing hormone agoni st. Nature 313, 23 1-233.
`Newton, D. L., Nicholls, P. 1., Rybak, S. M. , and Youle, R. 1.
`(1994) Expression and characterization of recombinant human
`eos in ophil -derived neurotox in and eos inophil-derived
`neurotoxin-anti-transferrin receptor sFv. 1. Bioi. Chem. 269,
`26739-26745.
`Newton, D. L., Xue, Y., Olson, K. A., Fett, J. w., and Rybak, S.
`M. (1996) Angiogenin single-chain immunofusions: Influence
`of peptide linkers and spacers between fusion protein domains.
`Biochemistry 35, 545-553.
`Pai, L. H. , Batra, 1. K. , Fitzgerald, D. J., Willingham, M. c., and
`Pastan, 1. (1992) Antitumor effects of B3-PE and B3-LysPE40
`in a nude mouse model of human breast cancer and the
`evaluation of B3-PE toxicity in monkeys. Cancer Res. 52,
`3189-3193.
`Pai, L. H., FitzGerald, D. J. , Tepper, M., Schacter, B., and Pastan,
`1. (1990) Inhibition of antibody response to Pseudomonas
`exotoxin and immunotoxin containing Pseudomonas exotoxin
`by 15-Deoxyspergualin in mice. Cancer Res. 50, 7750-7753.
`Ramakrishnan, S., FryxeU, D., Mohanraj, D., Olson, M. , and Li,
`B.-Y. (1992) Cytotoxic co njugates containing ribosomal
`inhibitory proteins. Annu. Rev. Pharmacol. Tox ico. 32,
`579-621 .
`Rodriguez, G. c., Boente, M. P. , Berchuck, A. , Whitaker, R. S.,
`
`IMMUNOGE 2156, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`36
`
`Killing Tumor Cells by LHRH-RNase A
`
`O 'Briant, K. c., Xu, F. , and Bast, R. C. Jr. ( 1993) The effect
`of antibodies and immunotox ins reactive with HER-2/neu on
`growth of ovarian and breast cancer cell lines. Am. 1. Obstet.
`Gynecol. 168, 228-232.
`Rybak, S. M. , Hoogenboom, H . R. , Meade, H . M ., Raus, J. C. ,
`Schwrtz, D. , and Youle, R . J. (1992) Humani zation of
`immunotoxins. Proc. Natl. A cad. Sci. USA 89, 3165-3169.
`Rybak, S. M. , Saxena, S. K., Ackerman, E . J., and Youle, R . J.
`(1991) Cytotox ic potential of ribonuclease and ribonuclease
`hybrid proteins. 1. BioI. Chern.. 266, 21202-2 1207.
`Sharoni , Y. , Bosin, E., Minster, A. , Levy, J. , and Schally, A. V.
`
`(1989) Inhibition of growth of human- mammary tumor cells
`by potent antago ni sts of luteini z ing hormone -re leasing
`honnone. Proc. Natl. Acad. Sci. USA 86, 1648-165 l.
`Szepeshazi, K., Scally, A. v., Juhasz, A. , Nagy, A. , and Janaky, T.
`(1992) Effect of lute ini z ing hormone- re leasi ng hormone
`analogs containing cytotoxic radicals on growth of estrogen(cid:173)
`independent MXT mouse mammary carcinoma in vivo. Anti(cid:173)
`Cancer Drugs 3, 109-116.
`Vitetta, E . S. , Krolick, K. A. , Miyama-lnaba, M. , Cuche\y, w.,
`and Uhr, J. W. (1983) Imunotoxins: A new approach to cancer
`therapy. Science 219, 644-650.
`
`IMMUNOGE 2156, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket