throbber
ANTICANCER RESEARCH 29: 4879-4886 (2009)
`
`Review
`
`Efficacy of Ligand-based Targeting
`for the EGF System in Cancer
`FUSANORI YOTSUMOTO1, AYAKO SANUI2, TATSUYA FUKAMI2, KYOKO SHIROTA2, SHINJI HORIUCHI2,
`HIROSHI TSUJIOKA2, TOSHIYUKI YOSHIZATO2, MASAHIDE KUROKI1 and SHINGO MIYAMOTO1,2
`Departments of 1Biochemistry, and 2Obstetrics and Gynecology, School of Medicine,
`Fukuoka University, 45-1, 7-chome, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
`
`Abstract. Although drugs inhibiting ErbB receptors such
`as epidermal growth factor receptor (EGFR) and HER2 have
`been developed as anticancer agents targeting the EGF
`family, they are not effective for all types of cancer and
`instead target only certain types. We propose the following
`four main reasons for these observations: (i) although seven
`EGFR ligands exist, effective inhibition of specific EGFR
`ligands may occur because their expression levels differ in
`different malignancies; (ii) suppressing EGFR ligands
`inhibits aggregation of EGFR and other ErbB receptors and
`activation of ERK and Akt signals; (iii) EGFR ligands may
`have various combinations for signal transduction through
`the EGFR pathway and other receptor signals; and (iv) the
`intracellular C-terminals of EGFR ligands move into the
`nucleus and strongly regulate cell proliferation. In this
`review, we describe important implications for targeted
`cancer therapy against EGFR ligands and describe the
`current situation in the development of ligand-based
`therapies for cancer.
`Epidermal growth factor (EGF) ligands, comprising EGF-
`related peptides, activate EGF receptor (EGFR, also known as
`ErbB1/HER1) and other ErbB receptors (ErbB3/HER3 and
`ErbB4/HER4). They are broadly classified into EGFR family
`ligands that bind to EGFR (comprising EGF, transforming
`growth factor (TGF)-α, heparin-binding EGF-like growth
`factor (HB-EGF), amphiregulin (AR), epiregulin, betacellulin
`(BTC) and epigen) and neuregulin (NRG) family ligands that
`
`Correspondence to: Shingo Miyamoto, Department of Biochemistry,
`School of Medicine, Fukuoka University, 45-1, 7-chome, Nanakuma,
`Jonan-ku, Fukuoka, 814-0180, Japan. Tel: +81 928011011, Fax: +81
`928013600, e-mail: smiya@cis.fukuoka-u.ac.jp
`Key Words: Cancer, HB-EGF, amphiregulin, targeted therapy,
`review.
`
`bind to ErbB3 or ErbB4 (comprising NRG1, NRG2, NRG3
`and NRG4) (1). Binding of ligands to the extracellular
`domains of ErbB receptors initiates their homodimerization or
`heterodimerization with other ErbB
`receptors
`and
`phosphorylation of tyrosine residues within their cytoplasmic
`domains, which in turn activates downstream growth and
`survival signals such as the mitogen-activated protein kinase
`(MAPK) and phosphoinositol 3-kinase/v-akt murine thymoma
`viral oncogene homolog (PI3K/AKT) pathways (2-4). No
`ligands that bind ErbB2 have been identified, and the kinase
`activity of ErbB3 is defective. These receptors are capable of
`generating intracellular signals by forming heterodimers with
`other ErbB receptors (5, 6). The EGF family members play
`important roles in normal tissue processes including ontogeny,
`morphogenesis, migration, differentiation and proliferation.
`Dysregulation of EGF family members and related signaling
`molecules can contribute to carcinogenesis and is associated
`with tumorigenesis, invasion and metastasis (2).
`EGFR and ErbB receptors have been especially focused
`upon as target molecules for cancer treatments because
`overexpression and mutations of these receptors are
`frequently observed in human malignancies. A variety of
`small molecule kinase inhibitors targeting EGFR (e.g.
`erlotinib: Tarceva™) and monoclonal antibodies targeting
`EGFR
`(e.g.
`cetuximab: Erbitux) and HER2
`(e.g.
`trastuzumab: Herceptin™) have been developed and some of
`them have already been used for treatment of lung cancer
`and breast cancer (7). However, these medicines have not
`exhibited the expected levels of clinical efficacy thus far,
`despite numerous cases of administration to patients with
`malignant tumors targeting EGFR and HER2 (8). One of the
`reasons for these observations is that EGFR and HER2 form
`complexes with HER3 and other signal receptors. The
`proliferation of cancer cells is subsequently accelerated by
`these complexes, whose formation cannot be inhibited by
`targeted therapies against EGFR and HER2. Another reason
`is that the anti-EGFR drugs can suppress the proliferation of
`
`0250-7005/2009 $2.00+.40
`
`4879
`
`1
`
`EX2129
`Eli Lilly & Co. v. Teva Pharms. Int'l GMBH
`IPR2018-01426
`
`

`

`ANTICANCER RESEARCH 29: 4879-4886 (2009)
`
`extracellular signal-related kinase (ERK) signals located
`downstream of EGFR but cannot suppress protein kinase B
`(AKT) survival signals. This background has given rise to
`an increasing demand for further development of targeted
`medicines against the EGF family. The belief that cancer
`agents targeting EGFR ligands are less effective than those
`targeting receptors has delayed the development of such
`medicines until now, but our research has revealed that this
`notion is not necessarily true.
`In this review, we will discuss the significance of targeting
`EGFR
`ligands for cancer
`therapy and describe
`the
`characteristics and present state of the development of
`anticancer agents targeting EGFR ligands.
`Significance of Targeting EGFR
`Ligands for Cancer Therapy
`Predominant expression of HB-EGF or AR in cancer (Figure
`1a). Enhancement of EGFR ligand expression, an autocrine
`loop mediated by an EGFR ligand itself, is the main
`mechanism
`implicated
`in cancer development and
`progression (9-11). First, in order to identify the EGFR
`ligands forming autocrine loops in cancer, we examined the
`expression levels of EGFR ligands in a variety of cancer cell
`lines (12). HB-EGF expression was dominantly elevated in
`ovarian, gastric and breast cancer, melanoma and
`glioblastoma. In pancreatic, colon and prostate cancer, renal
`cell carcinoma and cholangiocarcinoma, the expression of
`AR was primarily enhanced. Next, in order to examine
`whether inhibition of EGFR ligands exerts antitumor effects,
`we transfected individual small interfering RNAs (siRNAs)
`for these EGFR ligands into the cancer cell lines. In the cell
`lines with dominant expression of HB-EGF, a siRNA for
`HB-EGF increased the number of apoptotic cells and
`suppressed the activation of EGFR and ERK, whereas
`transfection of siRNAs for other EGFR ligands had no
`effects. Similarly, in the cell lines with abundant expression
`of AR, apoptosis and attenuation of EGFR and ERK signals
`were significantly mediated by inhibition of AR, while
`inhibition of the other EGFR ligands had no effects. Taken
`together, these findings suggest that HB-EGF and AR play
`pivotal roles in cancer cell proliferation and should be
`considered as promising target molecules for cancer therapy.
`Increases in the levels of HB-EGF or AR can also
`contribute to oncogenic transformation. In the absence of
`growth factors, Ha-ras-transformed human mammary
`epithelial cells do not form colonies in soft agar, and
`exogenous recombinant human HB-EGF is able to promote
`their anchorage-independent growth (13). AR is not
`expressed in the healthy liver, but is induced in the
`regenerating liver after partial hepatectomy and behaves as
`a pivotal mitogenic and antiapoptotic factor for normal
`hepatocytes (14, 15). Conversely, suppression of AR
`
`4880
`
`production dramatically reduces the aggressiveness of
`hepatocellular carcinoma cancer cells
`in anchorage-
`independent growth (16). Furthermore, EGFR ligands play
`important roles in inflammatory and neoplastic lesions in
`human tissues. Secretion of HB-EGF or AR, a heparin-
`binding EGFR ligand with undetectable expression in normal
`gastric tissues, is stimulated by the hormone gastrin, which
`induces gastric mucosa proliferation, and the inflammatory
`cytokine interleukin-1β. Inflammation of the gastric mucosa
`is itself associated with the proliferation of parietal cells in
`the gastric gland and
`the development of gastric
`malignancies (17). These pieces of evidence indicate that
`among the EGFR ligands, HB-EGF and AR are the main
`contributing growth factors for human carcinomas.
`
`Activation of signals-mediated ErbB receptor heterodime-
`rization resistance to anti-EGFR or anti-HER2 therapy
`(Figure 1b). Recently, there have been a large number of
`studies confirming the notion that dimerization of ErbB
`receptors is associated with resistance to anti-EGFR or anti-
`HER2 therapy (18-20). Dimerization is necessary for the
`signaling activity of ErbB receptors. The ligand-induced
`formation of a receptor complex stimulates the intrinsic
`tyrosine kinase activities of the receptors and induces
`autophosphorylation of specific tyrosine residues within their
`cytoplasmic domains. These phosphorylated residues serve
`as docking sites for various adaptor proteins and enzymes
`involved in potent signaling cascades, such as the raf proto-
`oncogene serine/threonine protein kinase/mitogen-activated
`protein kinase kinase/mitogen-activated protein kinase
`(Raf/MEK/MAPK) and phosphoinositide 3 kinase/v-akt
`murine thymoma viral oncogene homolog (PI3K/AKT)
`pathways (21, 22). When one receptor is functionally
`inactivated, its function as a receptor tyrosine kinase can be
`replaced by another receptor among the HER receptors.
`Gefinitib down-regulates the signaling pathway via EGFR,
`but does not block dimer formation between EGFR and other
`HER receptors. EGFR/HER2 (and EGFR/HER3) complex
`formation is increased in PC-9/ZD cells, a non-small cell
`lung cancer cell line with acquired gefinitib resistance (23).
`In addition, we tested the abovementioned hypothesis using
`MDA-MB-468 cells, a breast cancer cell line that secretes
`abundant soluble HB-EGF (12). In serum-free medium
`supplemented with HB-EGF, MDA-MB-468 cells formed
`EGFR/HER2 complexes, and this complex formation was
`enhanced by trastuzumab but reduced by CRM197, a
`specific HB-EGF
`inhibitor
`(unpublished
`data).
`Correspondingly, CRM197 attenuated the phosphorylation of
`ERK as well as AKT and led to significant apoptotic cell
`death compared with trastuzumab (unpublished data). Taken
`together, it is assumed that ligand-induced dimerization of
`ErbB receptors plays important roles in retrieving the
`intracellular signaling for cell survival against targeted
`
`2
`
`

`

`Yotsumoto et al: EGFR Ligands in Targeted Cancer Therapy (Review)
`
`Figure 1. Multidirectional functions of ligands in the EGF family. (a) Expression of a specific EGFR ligand (HB-EGF or AR) is enhanced in fourteen
`types of carcinomas and associated with cancer progression. (b) Binding of a ligand induces heterodimerization of ErbB receptors and subsequently
`activates resistance signals to targeted therapies for ErbB receptors. (c) Ligand-bound EGFR interacts with other growth factor receptors and
`SGLT1. (d) The shed extracellular domain and the CTF of HB-EGF transmit cell proliferation signals.
`
`4881
`
`3
`
`

`

`ANTICANCER RESEARCH 29: 4879-4886 (2009)
`
`therapy against EGFR or HER2 alone, and that targeting of
`a dominantly expressed EGFR ligand is an available strategy
`for the development of cancer therapies.
`
`Combinations of EGFR and other signaling pathways
`(Figure 1c). EGFR signaling can be modulated by several
`mechanisms that include transactivation by or crosstalk with
`other growth factor receptors such as insulin-like growth
`factor-I receptor (IGF-IR) (24) or steroid hormone receptor
`(25). Enhancement of IGF-IR expression was reported to be
`associated with
`trastuzumab
`resistance
`in HER2-
`overexpressing breast cancer cells. Inhibition of IGF-IR
`signaling rescued trastuzumab sensitivity (26). In recent
`studies, IGF-I was identified as another ligand that uses
`EGFR for MAPK and AKT activation (27, 28). IGF-I
`binding to IGF-IR activates matrix metalloproteinase
`(MMP)-2 and MMP-9, which cleave HB-EGF and release it
`to bind to EGFR, thereby leading to stimulation of MAPK.
`In MCF-7 breast cancer cells, IGF-IR activation induced by
`17β-estradiol/estrogen receptor α complexes on the cell
`membrane can also trigger a downstream signaling cascade
`through MMP-2, MMP-9, HB-EGF and EGFR and finally
`active MAPK (29). MDA-MB-468 cells are also a model for
`gefinitib resistance via constitutive activation of the
`intracellular signaling downstream of EGFR (i.e. the PI3K-
`AKT and MEK-MAPK pathways) (30-32). Under gefinitib
`treatment, MDA-MB-468 cells exhibit significant up-
`regulation (by up to 21-fold) of the EGFR ligands EGF, AR
`and BTC (33).
`Although the significance of EGFR, an oncogenic protein,
`has been sufficiently proven, as described above, this review
`begins to clarify a new role of EGFR in malignancy. The
`Authors have demonstrated that the extracellular domain of
`EGFR associates with and stabilizes the sodium/glucose co-
`transporter SGLT1 to promote glucose uptake into cancer
`cells (34). In PC-3MM2 prostate cancer cells, a siRNA for
`EGFR induced autophagic cell death with a decrease in the
`intracellular glucose level, whereas inhibition of EGFR
`kinase did not exert these effects. Furthermore, EGFR
`increased its complex formation with SGLT1 and glucose
`uptake following stimulation of EGFR by exposure to EGF
`in serum-free medium. These novel insights into EGFR
`functions could widen its potential in the development of
`anticancer agents for EGF family members.
`Transfer of HB-EGF-CTF to the nucleus (Figure 1d). Recent
`studies have reported that the intracellular HB-EGF
`carboxyl-terminal fragment (CTF) translocates from the
`plasma membrane to the nucleus and regulates the cell cycle
`when membrane-anchored HB-EGF
`is proteolytically
`cleaved by a disintegrin and metalloprotease (35, 36). BAG-
`1, promyelocytic leukemia zinc finger (PLZF) and B-cell
`leukemia 6 (Bcl6) have been identified as binding proteins
`
`4882
`
`for HB-EGF-CTF by yeast two-hybrid screening with the
`cytoplasmic region of HB-EGF (amino acids 185-208).
`Interactions between BAG-1, a prosurvival co-chaperone,
`and HB-EGF-CTF lead to attenuation of cell adhesion,
`resistance to apoptosis and enhancement of soluble HB-EGF
`expression (37). PLZF and Bcl6 are
`transcriptional
`repressors, and function as negative regulators within the cell
`cycle. Internalized HB-EGF-CTF co-localizes with PLZF or
`Bcl6 at the nuclear periphery, which releases suppression of
`the cell cycle. In addition, inhibition of HB-EGF-CTF
`nuclear translocation has been shown to reduce gastric
`cancer cell growth (38). Treatment using KB-R7785, an
`inhibitor of HB-EGF shedding, with or without EGFR
`activation by cetuximab interferes with the transfer of HB-
`EGF-CTF from the plasma membrane to the nucleus. As a
`result, KB-R7785 induces cell cycle arrest and increases the
`subG1 DNA content because PLZF remains in the nucleus
`and suppresses the cell cycle. Investigation of these functions
`of HB-EGF-CTF will lead to further understanding of the
`actions of EGFR ligands as growth factors and also provide
`new aspects for targeted therapies against EGFR ligands.
`Present State of the Development of Anticancer
`Agents Targeting EGFR Ligands
`Development of targeted therapeutic agents using CRM197.
`As discussed earlier, HB-EGF may be a promising target for
`ovarian cancer (39-41). Diphtheria toxin secreted by
`Corynebacterium diphtheriae binds to the EGF domain of
`HB-EGF and inhibits cell proliferation activity. Diphtheria
`toxin cannot be used as an HB-EGF inhibitor owing to its
`strong
`toxicity. However, cross-reacting material 197
`(CRM197), a mutated diphtheria toxin, can be used because it
`is a non-toxic protein with a variation in the active site and
`binds to HB-EGF more strongly than, or at least as strongly
`as, diphtheria toxin. We investigated the anticancer effects of
`CRM197 on ovarian cancer by evaluating the proliferation of
`human ovarian cancer cell lines (namely SKOV3, RMG1 and
`OVMG1) subcutaneously
`implanted
`into nude mice.
`CRM197 significantly suppressed peritoneal dissemination in
`the nude mice peritoneally injected with RMG1 or high HB-
`EGF-expressing SKOV3 cells (42). Furthermore, concomitant
`administration of CRM197 and paclitaxel induced complete
`disappearance of tumors at concentrations that showed no
`satisfactory antitumor effects in single treatments with either
`agent (43). The above findings suggest that CRM197 should
`be considered as a promising antineoplastic agent against
`ovarian cancer because it shows synergistic antitumor effects
`with a conventional chemotherapeutic agent and exerts effects
`on peritoneal dissemination. We have already started phase 1
`of a clinical trial of CRM197 administration for intractable
`advanced or recurrent ovarian cancer patients under the
`approval of an ethical committee.
`
`4
`
`

`

`Yotsumoto et al: EGFR Ligands in Targeted Cancer Therapy (Review)
`
`Pan-ligand trapping as a targeted therapy for cancer. In
`2008, a new strategy was reported that targets multiple
`ligands in the EGF family using a bispecific ligand trap,
`RB200 (44). RB200 was designed as a chimeric molecule
`composed of the full-length extracellular domains (ECDs) of
`EGFR and HER3 fused with the Fc domain of human
`immunoglobulin G1. cDNAs for EGFR and HER3 linked to
`the Fc domain (EGFR/Fc and HER3/Fc, respectively) were
`transiently co-transfected into HEK293T cells. RB200 was
`obtained by purification of conditioned medium harvested
`from the co-transfected cells. The purified RB200 bound
`EGFR ligands including EGF, TGFα and HB-EGF as well
`as HER3 ligands including NRG1-α, NRG1-β1 and NRG1-
`β3. Experiments were carried out to elucidate whether
`RB200 could inhibit ligand-stimulated phosphorylation of
`ErbB receptors and cell proliferation in a variety of cancer
`cells. Compared with C225 (the murine parent of cetuximab)
`and trastuzumab, RB200 successfully competed with the
`receptors for binding to ligands and suppressed both EGF-
`and NRG1-β1-induced tyrosine phosphorylation of EGFR,
`HER2 and HER3. Furthermore, RB200 exhibited inhibitory
`effects on cell growth in monolayer cultures of nine tumor
`cell lines and in vivo antitumor efficacy in two xenograft
`models (A431 human epidermoid carcinoma cells and H1437
`non-small cell lung cancer cells). Furthermore, mutants with
`amino acid substitutions in the EGFR and HER3 ECDs
`showed enhanced ligand-binding affinities and were more
`powerful than RB200 for inhibition of cell proliferation (45).
`In conclusion, EGFR and HER3 ligand traps may be potent
`tools for cancer therapy.
`Future Directions
`treatments with conventional
`Limitations of cancer
`anticancer agents, such as nucleic acid analogs and cell
`division inhibitors, developed so far have already been
`proven, making it all the more crucial to develop molecular
`targeted medicines on the basis of cancer characteristics.
`The targeted therapies that have achieved certain treatment
`outcomes to date have involved either plasma membrane
`molecules (receptors: EGFR, HER2; ligands: vascular
`endothelial growth factor, HB-EGF) or nuclear receptors
`(estrogen
`receptor,
`androgen
`receptor, peroxisome
`proliferator-activated receptor γ). We have demonstrated
`through non-clinical basic science investigations that
`CRM197 targets HB-EGF, blocks the autocrine loop created
`by HB-EGF and shows efficacy exceeding that of targeted
`therapies for EGFR. As a consequence, CRM197 is
`currently undergoing a clinical trial for administration to
`cancer patients.
`In future studies, we aim to elucidate the transcriptional
`mechanism involved in the acceleration of the autocrine loop
`mechanism by HB-EGF and identify novel target molecules
`
`that can inhibit nuclear signal transduction. The targeted
`medicines developed based on the results of these studies are
`expected to possess synergistic antitumor effects when used
`concomitantly with CRM197. Since these studies aim to
`identify transcription factors that induce the expression of
`HB-EGF, they appear to represent research into creating a
`novel concept targeting the molecules controlling the EGF
`system. According to our understanding, introduction of the
`HER2 gene or a mutated K-ras gene accelerates HB-EGF
`expression in breast cancer. These observations suggest that
`the transcription factor controlling the expression of HB-
`EGF is likely to be a target molecule not only for breast
`cancer but also for stomach and pancreatic cancer with
`abnormalities in the K-ras gene. Consequently, we presume
`that these findings will help to improve the prognosis of
`cancer patients as well as clarify how EGF family members
`promote cancer progression.
`Acknowledgements
`The Authors would like to thank Professor Eisuke Mekada
`(Department of Cell Biology, Research Institute for Microbial
`Diseases, Osaka University) for helpful discussions. This work was
`supported in part by a Grant-in-Aid for Scientific Research on
`Priority Areas (No. 19591947) and Research Promotion for
`Innovative Therapies against Cancers from the Ministry of
`Education, Culture, Sports, Science and Technology to E.M.
`References
`1 Citri A and Yarden Y: EGF-ERBB signaling: towards the
`systems level. Nat Rev Mol Cell Biol 7: 505-516, 2006.
`2 Yarden Y and Sliwkowski MX: Untangling the ErbB signalling
`network. Nat Rev Mol Cell Biol 2: 127-137, 2001.
`3 Sharma SV, Bell DW, Settleman J and Harver DA: Epidermal
`growth factor receptor mutations in lung cancer. Nat Rev Cancer
`7: 169-181, 2007.
`4 Burgess AW, Cho HS, Eigenbrot C, Ferguson KM, Garrett TP,
`Leahy DJ, Lemmon MA, Sliwkowski MX, Ward CW and
`Yokoyama S: An open-and-shut case? Recent insights into the
`activation of EGF/ErbB receptors. Mol Cell 12: 541-552, 2003.
`5 Klapper LN, Glathe S, Vaisman N, Hynes NE, Andrews GC,
`Sela M and Yarden: The ErbB-2/HER2 oncoprotein of human
`carcinomas may function solely as a shared co-receptor for
`multiple stroma-derived growth factors. Proc Natl Acad Sci USA
`96: 4995-5000, 1999.
`6 Guy PM, Platko JV, Cantley LC, Cerione RA and Carraway KL
`3rd: Insect cell-expressed p180erbB3 possesses an impaired
`tyrosine kinase activity. Proc Natl Acad Sci USA 91: 8132-8136,
`1994.
`7 Imai K and Takaoka A: Comparing antibody and small-molecule
`therapies for cancer. Nat Rev Cancer 6: 714-727, 2006.
`8 Morgillo F and Lee HY: Resistance to epidermal growth factor
`receptor-targeted therapy. Drug Resist Update 8: 298-310, 2005.
`9 Gangarosa LM, Sizemore N, Graves-Deal R, Oldham SM, Der
`CJ and Coffey RJ: A raf-independent epidermal growth factor
`receptor autocrine loop is necessary for Ras transformation of rat
`intestinal epithelial cells. J Biol Chem 272: 18926-18931, 1997.
`
`4883
`
`5
`
`

`

`ANTICANCER RESEARCH 29: 4879-4886 (2009)
`
`10 Roberts PJ and Der CJ: Targeting the Raf-MEK-ERK mitogen-
`activated protein kinase cascade for the treatment of cancer.
`Oncogene 26: 3291-3310, 2007.
`11 Normanno N, De Luca A, Bianco C, Strizzi L, Mancino M,
`Maiello MR, Carotenuto A, De Feo G, Caponigro F and
`Salomon DS: Epidermal growth factor receptor (EGFR)
`signaling in cancer. Gene 366: 2-16, 2006.
`12 Yotsumoto F, Yagi H, Suzuki SO, Oki E, Tsujioka H, Hachisuga
`T, Sonoda K, Kawarabayashi T, Mekada E and Miyamoto S:
`Validation of HB-EGF and amphiregulin as targets for human
`cancer therapy. Biochem Biophys Research Commun 365: 555-
`561, 2008.
`13 Martínez-Lacaci I, De Santis M, Kannan S, Bianco C, Kim N,
`Wallace-Jones B, Wechselberger C, Ebert AD and Salomon DS:
`Regulation of heparin-binding EGF-like growth
`factor
`expression in Ha-ras transformed human mammary epithelial
`cells. J Cell Physiol 186: 233-242, 2001.
`14 Berasain C, Garcia-Trevijano ER, Castillo J, Erroba E,
`Santamaria M, Lee DC, Prieto J and Avila MA: Novel role for
`amphiregulin in protection from liver injury. J Biol Chem 280:
`19012-19020, 2005.
`15 Berasain C, Garcia-Trevijano ER, Castillo J, Erroba E, Lee DC,
`Prieto J and Avila MA: Amphiregulin: an early trigger of liver
`regeneration in mice. Gastroenterology 128: 424-432, 2005.
`16 Castillo J, Erroba E, Perugorría MJ, Santamaría M, Lee DC,
`Prieto J, Avila MA and Berasain C: Amphiregulin contributes to
`the transformed phenotype of human hepatocellular carcinoma
`cells. Cancer Res 66: 6129-6138, 2006.
`17 Beales IL: Gastrin and interleukin-1beta stimulate growth factor
`secretion from cultured rabbit gastric parietal cells. Life Sci 75:
`2983-2995, 2004.
`18 Olayioye MA, Neve RM, Lane HA and Hynes NE: The ErbB
`signaling network: receptor heterodimerization in development
`and cancer. EMBO J 19: 3159-3167, 2000.
`19 Anido J, Matar P, Albanell J, Guzmán M, Rojo F, Arribas J,
`Averbuch S and Baselga J: ZD1839, a specific epidermal growth
`factor receptor (EGFR) tyrosine kinase inhibitor, induces the
`formation of inactive EGFR/HER2 and EGFR/HER3 heterodimers
`and prevents heregulin signaling in HER2-overexpressing breast
`cancer cells. Clin Cancer Res 9: 1274-1283, 2003.
`20 Knowlden JM, Hutcheson IR, Jones HE, Madden T, Gee JM,
`Harper ME, Barrow D, Wakeling AE and Nicholson RI: Elevated
`levels of epidermal growth factor receptor/c-erbB2 heterodimers
`mediate an autocrine growth regulatory pathway in tamoxifen-
`resistant MCF-7 cells. Endocrinology 144: 1032-1044, 2003.
`21 Massague J and Pandiella A: Membrane-anchored growth
`factors. Annu Rev Biochem 62: 515-541, 1993.
`22 Riese DJ II and Stern DF: Specificity within the EGF
`family/ErbB receptor family signaling network. Bioessays 20:
`41-48, 1998.
`23 Koizumi F, Shimoyama T, Taguchi F, Saijo N and Nishio K:
`Establishment of a human non-small cell lung cancer cell line
`resistant to gefitinib. Int J Cancer 116: 36-44, 2005.
`24 Adams TE, McKern NM and Ward CW: Signalling by the type 1
`insulin-like growth factor receptor: interplay with the epidermal
`growth factor receptor. Growth Factors 22: 89-95, 2004.
`25 Arpino G, Wiechmann L, Osborne CK and Schiff R: Crosstalk
`between the estrogen receptor and the HER tyrosine kinase
`receptor family: molecular mechanism and clinical implications
`for endocrine therapy resistance. Endocr Rev 29: 217-233, 2008.
`
`4884
`
`26 Lu Y, Zi X, Zhao Y, Mascarenhas D and Pollak M: Insulin-like
`growth factor-I receptor signaling and resistance to trastuzumab
`(Herceptin™). J Natl Cancer Inst 93: 1852-1857, 2001.
`27 Hallak H, Seiler AE, Green JS, Ross BN and Rubin R:
`Association of heterotrimeric G(i) with the insulin-like growth
`factor-I receptor. Release of G(betagamma) subunits upon
`receptor activation. J Biol Chem 275: 2255-2258, 2000.
`28 El-Shewy HM, Kelly FL, Barki-Harrington L and Luttrell LM:
`Ectodomain shedding-dependent transactivation of epidermal
`growth factor receptors in response to insulin-like growth factor
`type I. Mol Endocrinol 18: 2727-2739, 2004.
`29 Song RX, Zhang Z, Chen Y, Bao Y and Santen RJ: Estrogen
`signaling via a linear pathway involving insulin-like growth
`factor I receptor, matrix metalloproteinases, and epidermal
`growth factor receptor to activate mitogen-activated protein
`kinase in MCF-7 breast cancer cells. Endocrinology 148: 4091-
`4101, 2007.
`30 Moasser MM, Basso A, Averbuch SD and Rosen N: The tyrosine
`kinase inhibitor ZD1839 (‘Iressa™’) inhibits HER2-driven
`signaling and suppresses the growth of HER2-overexpressing
`tumor cells. Cancer Res 61: 7184-7188, 2001.
`31 Bianco R, Shin I, Ritter CA, Yakes FM, Basso A, Rosen N,
`Tsurutani J, Dennis PA, Mills GB and Arteaga CL: Loss of
`PTEN/MMAC1/TEP in EGF receptor-expressing tumor cells
`counteracts the antitumor action of EGFR tyrosine kinase
`inhibitors. Oncogene 22: 2812-2822, 2003.
`32 She QB, Solit D, Basso A and Moasser MM: Resistance to
`gefitinib in PTEN-null HER-overexpressing tumor cells can be
`overcome through restoration of PTEN function or pharmacologic
`modulation of constitutive phosphatidylinositol 3’-kinase/Akt
`pathway signaling. Clin Cancer Res 9: 4340-4346, 2003.
`33 Ferrer-Soler L, Vazquez-Martin A, Brunet J, Menendez JA, De
`Llorens R and Colomer R: An update of the mechanisms of
`resistance to EGFR-tyrosine kinase inhibitors in breast cancer:
`Gefitinib (Iressa™)-induced changes in the expression and
`nucleo-cytoplasmic trafficking of HER-ligands (Review). Int J
`Mol Med 20: 3-10, 2007.
`34 Weihua Z, Tsan R, Huang WC, Wu Q, Chiu CH, Fidler IJ and
`Hung MC: Survival of cancer cells is maintained by EGFR
`independent of its kinase activity. Cancer Cell 13: 385-393, 2008.
`35 Nanba D, Mammoto A, Hashimoto K and Higashiyama S:
`Proteolytic release of the carboxy-terminal fragment of proHB-EGF
`causes nuclear export of PLZF. J Cell Biol 163: 489-502, 2003.
`36 Kinugasa Y, Hieda M, Hori M and Higashiyama S: The
`carboxyl-terminal fragment of pro-HB-EGF reverses Bcl6-
`mediated gene repression. J Biol Chem 282: 14797-14806, 2007.
`37 Lin J, Hutchinson L, Gaston SM, Raab G and Freeman MR:
`BAG-1 is a novel cytoplasmic binding partner of the membrane
`form of heparin-binding EGF-like growth factor: a unique role
`for proHB-EGF in cell survival regulation. J Biol Chem 276:
`30127-30132, 2001.
`38 Shimura T, Kataoka H, Ogasawara N, Kubota E, Sasaki M,
`Tanida S and Joh T: Suppression of proHB-EGF carboxy-
`terminal fragment nuclear translocation: a new molecular target
`therapy for gastric cancer. Clin Cancer Res 14: 3956-3965, 2008.
`39 Miyamoto S, Hirata M, Yamazaki A, Kageyama T, Hasuwa H,
`Mizushima H, Tanaka Y, Yagi H, Sonoda K, Kai M, Kanoh H,
`Nakano H and Mekada E: Heparin-binding EGF-like growth
`factor is a promising target for ovarian cancer therapy. Cancer
`Res 64: 5720-5727, 2004.
`
`6
`
`

`

`Yotsumoto et al: EGFR Ligands in Targeted Cancer Therapy (Review)
`
`40 Yagi H, Miyamoto S, Tanaka Y, Sonoda K, Kobayashi H,
`Kishikawa T, Iwamoto R, Mekada E and Nakano H: Clinical
`significance of heparin-binding epidermal growth factor-like
`growth factor in peritoneal fluid of ovarian cancer. Br J Cancer
`92: 1737-1745, 2005.
`41 Tanaka Y, Miyamoto S, Suzuki SO, Oki E, Yagi H, Sonoda K,
`Yamazaki A, Mizushima H, Maehara Y, Mekada E and Nakano
`H: Clinical significance of heparin-binding epidermal growth
`factor-like growth factor and a disintegrin and metalloprotease-
`17 expression in human ovarian cancer. Clin Cancer Res 11:
`4783-4792, 2005.
`42 Yagi H, Yotsumoto F and Miyamoto S: Heparin-binding
`epidermal growth
`factor-like growth
`factor promotes
`transcoelomic metastasis in ovarian cancer through epithelial
`mesenchymal transition. Mol Cancer Ther 7: 3441-3451, 2008.
`43 Yagi H, Yotsumoto F, Sonoda K, Kuroki M, Mekada E and
`Miyamoto S: Synergistic antitumor effect of paclitaxel with
`CRM197, an inhibitor of HB-EGF, in ovarian cancer. Int J
`Cancer 124: 1429-1439, 2009.
`
`44 Sarup J, Jin P, Turin L, Bai X, Beryt M, Brdlik C, Higaki JN,
`Jorgensen B, Lau FW, Lindley P, Liu J, Ni I, Rozzelle J, Kumari
`R, Watson SA, Zhang J and Shepard HM: Human epidermal
`growth factor receptor (HER-1:HER-3) Fc-mediated heterodimer
`has broad antiproliferative activity in vitro and in human tumor
`xenografts. Mol Cancer Ther 7: 3223-3236, 2008.
`45 Jin P, Zhang J, Beryt M, Turin L, Brdlik C, Feng Y, Bai X, Liu
`J, Jorgensen B and Shepard HM: Rational optimization of a
`bispecific ligand trap targeting EGF receptor family ligands. Mol
`Med 15: 11-20, 2009.
`
`Received April 28, 2009
`Revised July 28, 2009
`Accepted September 16, 2009
`
`4885
`
`7
`
`

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