characterization of an antibody that can detect an activated igf-i receptor in human cancers

11
Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers Michele Rubini,* Consuelo D’Ambrosio,² Sabrina Carturan,* Gladys Yumet,‡ Edison Catalano,§ Simei Shan,‡ Ziwei Huang,‡ Mario Criscuolo,² Micol Pifferi,² and Renato Baserga‡ ,1 *University of Ferrara, Via L. Borsari 46, 44100 Ferrara, Italy; ²University of Modena, Via del Pozzo 71, 41100 Modena, Italy; Kimmel Cancer Center, Thomas Jefferson University, 233 S. 10th Street, 624 B.L.S.B., Philadelphia, Pennsylvania 19107; and §Cooper Hospital University Medical Center, One Cooper Plaza, Camden, New Jersey 08103 The type 1 insulin-like growth factor receptor (IGF- IR) plays an important role in malignant transforma- tion and in apoptosis. Its role in human cancer has now been firmly established. IGF-IR signaling occurs only when the receptor is activated by its ligands, which induce autophosphorylation of the receptor at several tyrosine residues. Although the IGF-IR (phos- phorylated or not) can be detected in human cancers with conventional antibodies, it would be desirable to obtain antibodies that can detect the IGF-IR only when activated by its ligands. We describe and char- acterize in this paper such an antibody and show that it can be used in sections of human cancers to detect an autophosphorylated IGF-IR. This antibody will be useful in detecting autocrine or paracrine influences on normal and tumor cells and could eventually be also useful in diagnostic and prognostic studies of hu- man primary and metastatic cancer. © 1999 Academic Press INTRODUCTION The insulin-like growth factor receptor (IGF-IR) is known to play an important role in the transformation and survival of cells, both in vivo and in vitro [1]. Over-expression and/or constitutive activation of IGF-IR in a variety of cell types leads to ligand-depen- dent growth in serum-free medium and to the estab- lishment of a transformed phenotype; i.e., ability to form colonies in soft agar and/or to produce tumors in mice [2, 3]. When the function of the IGF-IR is de- creased or otherwise impaired by antisense strategies or by dominant negative mutants, or by triple-helix formation, there is a dramatic inhibition of tumor growth [4 –11] and metastases [12–14] in experimental animals. The inhibition of tumor growth is largely due to the fact that the IGF-IR protects cells from a variety of apoptotic injuries, and, as a consequence, when the receptor is impaired, tumor cells are more susceptible to cell death. Thus, addition of IGF-I and/or overex- pression of the IGF-IR protect hemopoietic cells from apoptosis caused by interleukin-3 (IL-3) withdrawal [15–19]. IGF-I also protects cells overexpressing c-myc, which undergo apoptosis in serum-free medium and in the presence of other growth factors [20]. Finally, an activated IGF-IR protects human cancer cells from cell death induced by diverse anti-cancer drugs [21], etopo- side [22], tumor necrosis factor a [23], transforming growth factor b [24], p53 [25], ionizing and non-ioniz- ing radiation [26 –28], okadaic acid [29], and serum deprivation [30]. The IGF-IR is ubiquitous, and the only two cell types that, so far, are known to be devoid of IGF-IR are hepatocytes and B lymphocytes [1]. Although there are substantial amounts of IGF-I and IGF-II in the plasma [31], it is now generally accepted that these ligands are mostly bound to a number of IGF-binding proteins [32] and that the activation of the IGF-IR is usually due to autocrine or paracrine influences [1]. The IGF-IR can be detected in tissue sections by the use of appropriate antibodies, and in fact, it has already been used to show that it is a significant prognostic factor in human breast cancer [27]. Other reports have confirmed the importance of the IGF system in human cancer [33, 34], especially in breast cancer [35–38], and prostate cancer [39]. Because of the importance of the IGF sys- tem in cancer, it would be desirable to develop antibod- ies against the IGF-IR that could become useful tools in the diagnosis and/or prognosis of malignant diseases. The antibodies presently available detect the IGF-IR in cells regardless of its state of activation. In immu- noblots, the activated receptor can be visualized, after specific immunoprecipitation, with an anti-phosphoty- rosine antibody, but this procedure is not feasible in tissue sections. We therefore decided to develop an antibody that could specifically detect a tyrosyl phos- phorylated IGF-IR, i.e., an IGF-IR activated by its li- gands, in tissue sections from normal or cancerous human tissues. There are certain limitations to this 1 To whom correspondence and reprint requests should be ad- dressed. Fax: (215) 923-0249. E-mail: [email protected]. 22 0014-4827/99 $30.00 Copyright © 1999 by Academic Press All rights of reproduction in any form reserved. Experimental Cell Research 251, 22–32 (1999) Article ID excr.1999.4562, available online at http://www.idealibrary.com on

Upload: michele-rubini

Post on 17-Oct-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers

Itnowspwowaiauoam

kaOIdlfmcofgato

d

0CA

Experimental Cell Research 251, 22–32 (1999)Article ID excr.1999.4562, available online at http://www.idealibrary.com on

Characterization of an Antibody That Can Detect an Activated IGF-IReceptor in Human Cancers

Michele Rubini,* Consuelo D’Ambrosio,† Sabrina Carturan,* Gladys Yumet,‡ Edison Catalano,§ Simei Shan,‡Ziwei Huang,‡ Mario Criscuolo,† Micol Pifferi,† and Renato Baserga‡,1

*University of Ferrara, Via L. Borsari 46, 44100 Ferrara, Italy; †University of Modena, Via del Pozzo 71, 41100 Modena, Italy;‡Kimmel Cancer Center, Thomas Jefferson University, 233 S. 10th Street, 624 B.L.S.B., Philadelphia, Pennsylvania 19107;

and §Cooper Hospital University Medical Center, One Cooper Plaza, Camden, New Jersey 08103

rtpa[wtadsgid

ths[maabasbi3ctit

insrtapg

The type 1 insulin-like growth factor receptor (IGF-R) plays an important role in malignant transforma-ion and in apoptosis. Its role in human cancer hasow been firmly established. IGF-IR signaling occursnly when the receptor is activated by its ligands,hich induce autophosphorylation of the receptor at

everal tyrosine residues. Although the IGF-IR (phos-horylated or not) can be detected in human cancersith conventional antibodies, it would be desirable tobtain antibodies that can detect the IGF-IR onlyhen activated by its ligands. We describe and char-cterize in this paper such an antibody and show thatt can be used in sections of human cancers to detectn autophosphorylated IGF-IR. This antibody will beseful in detecting autocrine or paracrine influencesn normal and tumor cells and could eventually belso useful in diagnostic and prognostic studies of hu-an primary and metastatic cancer. © 1999 Academic Press

INTRODUCTION

The insulin-like growth factor receptor (IGF-IR) isnown to play an important role in the transformationnd survival of cells, both in vivo and in vitro [1].ver-expression and/or constitutive activation of

GF-IR in a variety of cell types leads to ligand-depen-ent growth in serum-free medium and to the estab-ishment of a transformed phenotype; i.e., ability toorm colonies in soft agar and/or to produce tumors inice [2, 3]. When the function of the IGF-IR is de-

reased or otherwise impaired by antisense strategiesr by dominant negative mutants, or by triple-helixormation, there is a dramatic inhibition of tumorrowth [4–11] and metastases [12–14] in experimentalnimals. The inhibition of tumor growth is largely dueo the fact that the IGF-IR protects cells from a varietyf apoptotic injuries, and, as a consequence, when the

1 To whom correspondence and reprint requests should be ad-

hressed. Fax: (215) 923-0249. E-mail: [email protected].

22014-4827/99 $30.00opyright © 1999 by Academic Pressll rights of reproduction in any form reserved.

eceptor is impaired, tumor cells are more susceptibleo cell death. Thus, addition of IGF-I and/or overex-ression of the IGF-IR protect hemopoietic cells frompoptosis caused by interleukin-3 (IL-3) withdrawal15–19]. IGF-I also protects cells overexpressing c-myc,hich undergo apoptosis in serum-free medium and in

he presence of other growth factors [20]. Finally, anctivated IGF-IR protects human cancer cells from celleath induced by diverse anti-cancer drugs [21], etopo-ide [22], tumor necrosis factor a [23], transformingrowth factor b [24], p53 [25], ionizing and non-ioniz-ng radiation [26–28], okadaic acid [29], and serumeprivation [30].The IGF-IR is ubiquitous, and the only two cell types

hat, so far, are known to be devoid of IGF-IR areepatocytes and B lymphocytes [1]. Although there areubstantial amounts of IGF-I and IGF-II in the plasma31], it is now generally accepted that these ligands are

ostly bound to a number of IGF-binding proteins [32]nd that the activation of the IGF-IR is usually due toutocrine or paracrine influences [1]. The IGF-IR cane detected in tissue sections by the use of appropriatentibodies, and in fact, it has already been used tohow that it is a significant prognostic factor in humanreast cancer [27]. Other reports have confirmed themportance of the IGF system in human cancer [33,4], especially in breast cancer [35–38], and prostateancer [39]. Because of the importance of the IGF sys-em in cancer, it would be desirable to develop antibod-es against the IGF-IR that could become useful tools inhe diagnosis and/or prognosis of malignant diseases.

The antibodies presently available detect the IGF-IRn cells regardless of its state of activation. In immu-oblots, the activated receptor can be visualized, afterpecific immunoprecipitation, with an anti-phosphoty-osine antibody, but this procedure is not feasible inissue sections. We therefore decided to develop anntibody that could specifically detect a tyrosyl phos-horylated IGF-IR, i.e., an IGF-IR activated by its li-ands, in tissue sections from normal or cancerous

uman tissues. There are certain limitations to this
Page 2: Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers

eho[1cifpthdc

w4aMNdwtdcgtpCm0aoci

mUspQ

ikws4tacp

auapbi

gg(

s2t

pIpwmNnc

HmmaS(fmHsbsdwb[4saI1eidmr

wCpbaEn

odp(biDis

tsa

23IGF-I RECEPTOR AND CANCER

ndeavor: the IGF-IR and the insulin receptor (IR)ave extensive homologies in the tyrosine-rich regionsf the kinase domain and the juxtamembrane domain40]. In the kinase domain, the homology is close to00%, making it very difficult to obtain antibodies thatould distinguish between the two receptors. However,n the C-terminus, homology between the two receptorsalls to 44%, and we therefore selected this region toroduce an antibody to the tyrosyl phosphorylated C-erminus of the IGF-IR. We describe and characterizeere this antibody, and we demonstrate its ability toetect an activated IGF-IR in tissue sections of humanancers.

MATERIALS AND METHODS

Peptide synthesis. As described previously [41, 42], the peptidesere prepared by solid-phase synthesis using Fmoc strategy on a30A peptide synthesizer (Applied Biosystems, Foster City, CA) and

9050 Pepsynthesizer Plus (Perseptive Biosystems, Cambridge,A). A 4-fold excess of Na-Fmoc amino acid, O-benzotriazol-1-yl-,N,N9,N9-tetramethyluronium hexafluorophosphate, and 1-hy-roxybenzotriazole and a 10-fold excess of diisopropylethylamineere used in every coupling reaction step. Removal of the NH2-

erminal Fmoc group was accomplished by 20% piperidine in N,N-imethylformamide. The cleavage of peptides from the resin wasarried out with reagent K [43] for 2 h at room temperature withentle stirring. Crude peptides were precipitated in ice-cold methyl--butyl ether, centrifuged, and lyophilized. Crude peptides were thenurified by preparative reverse-phase HPLC using a Dynamax-300Å18 column (25 cm 3 21.4 mm, inner diameter) with a flow rate of 9l/min and two solvent systems of 0.1% trifluoroacetic acid/H2O and

.1% trifluoroacetic acid/acetonitrile. The fractions containing theppropriate peptide were pooled together and lyophilized. The purityf the final products was assessed by analytical reverse-phase HPLC,apillary electrophoresis, and matrix-assisted laser desorption/ion-zation time-of-flight mass spectrometry.

Pep922 was a 19-amino-acid peptide corresponding to the C-ter-inus residues 1319–1337 of the human IGF-IR (numbering ofllrich et al., ref. 40). Pep1046 was a 28-amino-acid peptide corre-

ponding to residues 1310-1337 of the human IGF-IR, including ahosphotyrosyl residue at position 1316. Its sequence is NH2, FDER-PpYAHMNGGRKNERALPLPQSSTCCOOH.Immunization protocol and antibodies. Peptides were dissolved

n 0.1 M sodium phosphate buffer (pH 7.0) and coupled to activatedeyhole limped hemocyanin (Pierce) by glutaradehyde. Immunogensere diluted 1:1 with complete (first shot) or incomplete (subsequent

hots) Freund’s adjuvant and injected intradermally in rabbits at-week intervals. The anti-peptide serum antibody level was moni-ored by ELISA. To get rid of unwanted anti-C terminus antibodies,nti-pep1046 serum was cleared by reverse immunoaffinity purifi-ation using pep922-bound Sepharose. The antibody againstep1046 is hereafter referred to as anti-pY1316.For detection of the IGF-IR b-subunit, an anti-C-terminus IGF-IR

ntibody (Santa Cruz) was used. We also used an antibody obtainedsing anti-pep922 antiserum; it behaved exactly as the commercialntibody against the C-terminus. For detection of the overall tyrosylhosphorylation of immunoprecipitated receptors, the PY20 anti-ody (Transduction Labs.) was used. Other antibodies are describedn the section on immunoblots.

Cell cultures. Cells were maintained in Dulbecco’s modified Ea-le’s medium (DMEM) supplemented with 10% bovine calf serum,lutamine (20 mg/ml) MEM, penicillin (10 U/ml), and streptomycin

0.1 mg/ml). Incubations were at 37°C in a CO2 incubator under o

tandard conditions. To make cells quiescent, they were cultured fordays in serum-free medium (DMEM supplemented with 50 mg/ml

ransferrin and 0.1% bovine serum albumin).Immunoprecipitation and immunoblotting. To induce receptor

hosphorylation, quiescent cells were stimulated for 5 min withGF-I (3.3 nM) or with insulin (6.5 nM). Culture dishes were thenlaced on ice and cells were rinsed three times in cold Hanks’. Cellsere lysed in lysis buffer [50 mM Hepes (pH 7.5), 150 mM NaCl, 1.5M MgCl2, 1 mM EGTA, 10% glycerol, 1% Triton X-100, 100 mMaF, 0.2 mM Na3VO4, 10 mM Na4P2O7, 1 mM phenylmethylsulpho-yl fluoride, 10 mg/ml aprotinin] for 4 min at 4°C. Lysates wereollected and cleared from nuclei by centrifugation.Lysates (150 mg of protein) were diluted with HNTG buffer [20 mMepes (pH 7.5), 150 mM NaCl, 0.1% Triton X-100, 10% glycerol, 100M NaF, 0.2 mM Na3VO4, 5 mM phenylmethysulphonyl fluoride, 10g/ml aprotinin] containing 15 ml of protein A–agarose (Calbiochem)nd 10 ml of anti-IGF-IR (Ab-1) monoclonal antibody (Oncogenecience) or 10 ml of anti-insulin receptor (Ab-3) monoclonal antibody

Oncogene Science). Antibody–antigen complexes were allowed toorm for 4 h at 4°C and then collected by centrifugation at 4°C for 4

in. Immunoprecipated complexes were washed three times inNTG and resuspended in 20 ml of Laemmli buffer (20% glycerol, 3%

odium dodecyl sulfate, 3% b-mercaptoethanol, 10 mM EDTA, 0.05%romophenol blue). Samples were boiled for 4 min and proteins wereeparated on 4–20% polyacrylamide gradient gels (Bio-Rad) by so-ium dodecyl sulfate–polyacrylamide gel electrophoresis. Proteinsere electroblotted onto nitrocellulose filters. Membranes werelocked with 5% bovine serum albumin or 5% nonfat milk in TBST10 mM Tris (pH 8.0), 150 mM NaCl, 0.1% Tween 20] overnight at°C, probed with 10 ml of the indicated antibodies in 20 ml TBSTolution for 1 h. When secondary antibody interaction was needed,fter five washes in TBST, filters were probed with 2 ml of anti-rabbitgG-HRP antibody (Oncogene Science) in 20 ml TBST solution forh. After washing five times in TBST, blots were developed with the

nhanced chemiluminescence detection system (Amersham) accord-ng to the manufacturer’s instructions. Immunoblot reprobing wasone after filters were incubated for 30 min in stripping buffer [62.5M Tris (pH 6.8), 2% SDS, 100 mM b-mercaptoethanol] at 50°C and

eblocking overnight at 4°C in 5% nonfat milk–TBST.Peptide competition test. Anti-pep1046 (anti-pY1316) antibodyas challenged with pep922, or the commercial antibody to the-terminus (Santa Cruz), or pep1046 itself. Five microliters of anti-ep1046 antibody was incubated with 8 ml of 0.1 M Na-phosphateuffer (PB), pH 7.0, and 10 ml of 10 mg/ml pep922, commercialntibody, pep1046, or just PB. Incubation was for 30 min at 37°C.ach reaction was used to probe immunoprecipitated IGF-IR immu-oblots.Staining of tissue sections. Tissue sections (5 mm thick) were

btained from paraffin-embedded tumor specimens. After standardeparaffinization, the sections were probed either with an anti-Y1316 antibody or with an antibody to the C-terminus of the IGF-IRanti-pep922 antibody). After incubation with goat anti-rabbit IgGiotinylated antibody and peroxidase-conjugated streptavidin, themmune complex was visualized with the chromogenic substrateAB (diaminobenzidine tetrahydrochloride). Several controls were

ncluded (preimmune serum, peptide competition), and consecutiveections were examined.

RESULTS

For clarity, we designate as antibody anti-pY1316,he antibody developed against the 1046 peptide de-cribed under Materials and Methods, which containsphosphorylated tyrosine 1316. We shall refer to the

ther antibodies that recognize the C-terminus of the

Page 3: Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers

Ia

T

ptuwb[TIwhtlbMbCetTbomfgta

abcdlada

S

ptcdit(oanat(iIedtas

RRRRRRRRRRRR

i[mu

tawtwupI

24 RUBINI ET AL.

GF-IR (regardless of the status of its phosphorylation)s anti-C-terminus antibodies.

he Anti-pY1316 Antibody Recognizes an Epitopein the C-terminus of the IGF-IR

The first test was to determine whether this anti-Y1316 antibody recognized the intended epitope inhe C-terminus of the IGF-IR. For this purpose, wesed three cell lines, all derived from R2 cells [44, 45],hich are 3T3-like cells originating from mouse em-ryos with a targeted disruption of the IGF-IR genes46, 47]. The three cell lines were the parental R2 cells,c-4 cells, which are R2 cells stably transfected with anGF-IR truncated at residue 1229 [48], and R1 cells,hich are R2 cells stably transfected with a wild-typeuman IGF-IR cDNA [44]. Lysates were prepared fromhese cells stimulated or not by IGF-I. In Fig. 1, allysates were first immunoprecipitated with an anti-ody to the a subunit of the IGF-IR (see Materials andethods), which precipitates both subunits. In Fig. 1,

ottom, the blot was stained with an antibody to the-terminus of the IGF-IR (anti-pep922 antibody): asxpected, only lysates from R1 cells gave a signal, sincehis antibody will not recognize R2 cells (no receptor) orc-4 cells (missing the C-terminus). In Fig. 1, top, thelot was stained with the anti-pY1316 antibody. Again,nly R1 cells give a positive signal, since the Y1316 isissing from the receptor of Tc-4 cells (and obviously

rom R2 cells). The antibody against the C-terminusives bands of similar intensity, regardless of the ac-ivity status of the receptor, while the anti-pY1316

FIG. 1. Specificity of the anti-py1316 antibody. In all instances,he IGF-I receptor was immunoprecipitated from lysates with anntibody to the a subunit of the IGF-IR (Oncogene Science). Lysatesere prepared from three types of cells: R2 (no IGF-IR); Tc-4 (IGF-IR

runcated at residue 1229); and R1 (full-length IGF-IR). The lysatesere prepared from cells that were unstimulated (2) or were stim-lated for 5 min with IGF-I (3.3 nM). (Top) Blot with an antibody toY1316. (Bottom) Blot with an antibody to the C-terminus of theGF-IR.

ntibody gives a much stronger signal when the lysates e

re prepared from IGF-I-stimulated cells. The faintand in unstimulated cells is due to the fact that R1

ells secrete a small amount of IGF-I [44], which pro-uces a background activation of the receptor (see be-ow). Therefore, the anti-pY1316 antibody can detectn autophosphorylated wild-type receptor, but does notetect an IGF-IR lacking the C-terminus (the last 108mino acids).

pecificity of the Anti-pY1316 Antibody

The anti-pY1316 antibody was then tested on aanel of R2-derived cells expressing a number of mu-ants of the IGF-IR. These mutant receptors and theell lines derived from them have been described inetail in previous papers [49–53] and are summarizedn Table 1. The cells were left unstimulated (see Ma-erials and Methods) or were stimulated with IGF-I3.3 nM) for 10 min. Figure 2 summarizes the resultsbtained by immunoprecipitating the lysates with anntibody to the a subunit of the IGF-IR (which immu-oprecipitates both subunits) and blotting with thenti-pY1316 antibody (top), or an anti-C-terminus an-ibody (middle), or an anti-phosphotyrosine antibodybottom). R2 cells are negative with all three antibod-es, as expected. An antibody to the C-terminus of theGF-IR (middle) detects the IGF-IR in all cell lysates,xcept those of R2 cells and the cells expressing the1245 mutant. The R2/d1245 cells [53] have a receptorruncated at residue 1245, and the epitopes for the twontibodies used to detect the b subunit are both down-tream from residue 1245. This mutant receptor,

TABLE 1

Cell lines Mutant human IGF-IR

Densitometricratios

pY pY1316

2 Null-/-IGF-IR 0 02/950F Y950F 11.9 4.12/KA K1003A 0 02/YF1 Y1131F, Y1135F, Y1136F 6.1 1.92/1245D Truncation at 1245 6.0 02/Y1250F Y1250F 4.9 2.22/Y1251F Y1251F 1.5 1.52/Y1250/1F Y1250, Y1251F2/S1280-3A S1280-83 to A 29.6 12.52/1316F Y1316F 4.0 01 wt 68.7 30.1-IR9 Null-/-IGF-IR, overexpressing

human insulin receptor

Note. The development of the mutant receptors and the character-zation of the cell lines have been described in previous papers49–54]. The densitometric ratios give the fold increase in densito-etric intensities of the autophosphorylated IGF-IR bands over thenstimulated cells. The higher the increase in ratio, the higher is the

xtent of autophosphorylation of the activated receptor.
Page 4: Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers

tweb

pc[tpiscRmawas

ntfialemkdCY1gdsa

gb

D

pbIdIltciw(ssibpatpbropp

T

c

uwa ibed

25IGF-I RECEPTOR AND CANCER

hough, is visible when the membranes are blottedith an anti-phosphotyrosine antibody. The band is, asxpected, lower than the bands of the other full-lengthsubunits.When the cells are stimulated with IGF-I, the anti-

hosphotyrosine antibody detects the receptor in allell lines, except R2 cells and R2/KA cells. R2/KA cells54] have an IGF-IR with a mutation at lysine 1003,he ATP-binding site. This receptor does not autophos-horylate after addition of IGF-I and is essentially annactive receptor. With the anti-pY1316 antibody, noignal is detectable in R2 cells, in R2/KA cells, d1245ells, and the Y1316F cells. The crucial cell line is the

2/Y1316F cells [53], which have a receptor with autation at Y1316 from tyrosine to phenylalanine and

re therefore missing Y1316. No signal is detectableith the anti-pY1316 antibody, while the other twontibodies (phosphotyrosine and b subunit) give a goodignal.Therefore, from Fig. 2, we can conclude that one

eeds a phosphorylated Y1316 for the anti-pY1316 an-ibody to be able to recognize the IGF-IR. This is con-rmed by the finding that in all other mutants, thenti-pY1316 antibody recognized an autophosphory-ated receptor. These include, besides R1 cells, cellsxpressing the following mutant receptors: a Y950Futant [49]; a mutant at the three tyrosines of the

inase domain [51], in which autophosphorylation isecreased but not abolished; mutant receptors of the-terminus, like the Y1250F, the Y1251F, or both1250 and Y1251 [50]; and a mutant at serines 1280–283 [52]. In some cell lines, the anti-pY1316 antibodyives a faint band even in unstimulated cells, which isue, as explained above, to residual IGF-I or IGF-IIecreted by the cell lines in question [44, 54]. Indeed, in

FIG. 2. Detection of mutant receptors by the anti-pY1316 annstimulated (2) or stimulated with IGF-I (1). The lysates were immere stained with either the anti-pY1316 antibody (first row), ornti-phosphotyrosine antibody (third row). The antibodies are descr

ll those cases, the anti-phosphotyrosine antibody also a

ave a positive signal, indicating that the receptor hadeen activated.

ensitometric Analysis

The intensity of the bands obtained with the anti-hosphotyrosine antibody, with the anti-pY1316 anti-ody, and with the antibody to the C-terminus of theGF-IR were quantitated by densitometry. The ratio ofensitometric values between unstimulated cells andGF-I-stimulated cells (after correction for receptorevel) is given in Table 1, with the list of mutant recep-ors. The ratios vary, depending in part on how quies-ent the cells were, but Table 1 clearly shows that: (1)n R2 and R2/KA cells, the ratios do not changehether the cells are or are not stimulated with IGF-I;

2) the d1245 receptor and the Y1316F receptor show aharp increase in autophosphorylation, when mea-ured with the anti-phosphotyrosine antibody, but noncrease when measured with the anti-pY1316 anti-ody; and (3) with the other mutant receptors, auto-hosphorylation increases are detectable with eitherntibody. It is true that the increases are larger withhe anti-phosphotyrosine antibody, but this can be ex-lained by the fact that the anti-phosphotyrosine anti-ody recognizes all the phosphorylated tyrosines of theeceptor, while the anti-pY1316 antibody recognizesnly one tyrosine. It seems therefore that the anti-Y1316 antibody specifically recognizes an autophos-horylated IGF-IR with a phosphorylated Y1316.

he Anti-pY1316 Antibody Does Not Cross-reactwith the Insulin Receptor

In the Introduction, we have given the rationale forhoosing the peptide used to generate the anti-pY1316

dy. For every cell line, lysates were prepared from cells eitherprecipitated with an antibody anti-IGF-IR (a subunit), and the blots

antibody to the C-terminus of the IGF-IR (second row), or anunder Materials and Methods, and the cell lines in Table 1.

tibouno

an

ntibody. One of the main criteria was that the se-

Page 5: Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers

qstpFdtcTTsItpaeaicwtutw

P

cws

atpcmitpYI

T

n4tswssta

D

ta

((wppwmast

rsIfcwpwI

26 RUBINI ET AL.

uence chosen should be sufficiently different from anyequence in the insulin receptor [40]. This is impor-ant, especially in view of the report that some anti-eptide antibodies can recognize similar receptors [55].igure 3 shows that the anti-pY1316 antibody canistinguish between the activated IGF-IR and the ac-ivated insulin receptor (IR). The cells used were R2

ells expressing a very high level of insulin receptors.his cell line has been previously described [50, 56].he cells were stimulated or not with insulin, the ly-ates were immunoprecipitated with an antibody to theR, and the blots stained with either an anti-phospho-yrosine antibody (Fig. 3, bottom) or with the anti-Y1316 antibody (top). It is clear that the anti-pY1316ntibody cannot recognize the autophosphorylated IR,ven in cells that express roughly 5 3 105 IR/cell [56],very large number of IR, about 5 log above the phys-

ological levels of IR in mouse embryo fibroblasts. For aomparison, we show again lysates from R1 cells,here the IGF-IR is detected by both the anti-phospho-

yrosine antibody and the anti-pY1316 antibody. Thepper band (with either the IR or the IGF-IR) is knowno be the proreceptor form, which is frequently visiblehen cells are overexpressing the receptors.

eptide Competition

To confirm that the anti-pY1316 antibody specifi-ally recognizes the sequence used for immunization,e tried competition experiments. In Fig. 4A, the ly-

FIG. 3. The anti-pY1316 antibody does not recognize the insulineceptor. Preparation of lysates, antibodies, and blotting are de-cribed under Materials and Methods. The cell line examined here isR9 cells (no IGF-IR, about 5 3 103 insulin receptors/cell). Lysatesrom cells stimulated (1) or not (2) with insulin were immunopre-ipitated with an antibody anti-insulin receptor. (Top) Blot stainedith an antibody anti-pY1316. (Bottom) Blot stained with an anti-hosphotyrosine antibody. For control, we used R1 cells (see above),here the lysates were immunoprecipitated with an antibody to the

GF-IR a subunit.

ates were immunoprecipitated with anti-a subunit a

ntibody to the IGF-IR and then immunoblotted withhe anti-pY1316 antibody. A band corresponding to theosition of the IGF-IR is clearly evident in lysates of R1

ells stimulated with IGF-I (lane A1). When the im-unoblot is pretreated with the same peptide used for

mmunization, the band is markedly decreased in in-ensity (lane B1). For control, we have used the sameeptide sequence, but without a phosphorylated1316. The band is now again visible in lysates of

GF-I-stimulated R1 cells (lane C1).

he Anti-pY1316 Antibody Can Immunoprecipitatethe IGF-IR

We next tested the ability of this antibody to immu-oprecipitate the IGF-IR. The results are shown in Fig.B, where lysates from R1 cells were immunoprecipi-ated with the anti-pY1316 antibody, before or aftertimulation with IGF-I. The blots were then stainedith an anti-phosphotyrosine antibody. A band corre-

ponding to the b subunit of the IGF-IR is visible aftertimulation with IGF-I (lane 2). The highest band ishe proreceptor. We have not identified the intermedi-te band, whose phosphorylation increases after IGF-I.

etection of the IGF-I Receptor in Human Cancers

We tested our anti-pY1316 antibody on tissue sec-ions of human cancers. The sections were preparednd stained as described under Materials and Meth-

FIG. 4. Further characterization of the anti-pY1316 antibody.A) Peptide competition. IGF-IR immunoblots from IGF-I-stimulated1) or unstimulated R1 cells. The receptor was immunoprecipitatedith an antibody anti-a subunit and immunoblotted with the anti-Y1316 antibody. (A) No competition. (B) Competition with the phos-horylated peptide pep1046 used for immunization. (C) Competitionith control peptide, pep922 (see Materials and Methods). (B) Im-unoprecipitation with the anti-pY1316 antibody. The anti-pY1316

ntibody was used to precipitate the receptor from lysates of IGF-I-timulated (1) or unstimulated (2) R1 cells. The immunoprecipi-ated proteins were immunodetected using an anti-phosphotyrosine

ntibody.
Page 6: Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers

ocwco(pmwapob5sdroasacctaw

sTfwslpIIptTsdgcotrCI1MI

c

acpwtcIitrtftrtls

psitaIlmcirlmm1d[RwFii

uprhcHaTpwgybt

27IGF-I RECEPTOR AND CANCER

ds. We examined sections of both breast cancers andolon cancers, but, for simplicity (see also below), weill limit ourselves to the documentation of breast

ancers. We ran numerous controls, including the usef a preimmune serum (Fig. 5A), or peptide competitionnot shown, since the specificity of the antibody to theeptide used for immunization has already been docu-ented with a more sensitive method in Fig. 4). Also,e examined consecutive sections, stained with eitherntibody to the C-terminus or an antibody anti-Y1316. For obvious reasons, we are presenting herenly selected pictures, and the selection is admittedlyased on the best and most convincing pictures. FigureB shows a photograph of a human mammary cancer,tained with the anti-pY1316 antibody. Many intra-uctal cancer cells are strongly positive, while the sur-ounding connective tissue is essentially negative. An-ther section of human breast cancer stained with thenti-pY1316 antibody is shown in Fig. 6A. The pictureshown were selected as the best illustrations of thebility of the anti-pY1316 antibody to stain cancerells. In other sections of human cancers, positive can-er cells alternated with cancer cells that were nega-ive or weakly positive. The same can be said of thentibody against the C-terminus of the IGF-IR, ofhich we give an illustration in Fig. 6B.

DISCUSSION

We have developed an antibody, anti-pY1316, thatpecifically recognizes an autophosphorylated IGF-IR.he evidence for this statement can be summarized as

ollows: (1) the antibody recognizes the IGF-IR onlyhen the receptor is phosphorylated, while the corre-

ponding antibody to the C-terminus (nonphosphory-ated) recognizes the IGF-IR regardless of its phos-horylation status; (2) using a panel of several mutantGF-IR, the anti-pY1316 antibody visualizes theGF-IR only when the Y1316 residue is present andhosphorylated; and (3) the anti-pY1316 antibody failso recognize the IR, even after appropriate stimulation.he IR is the only tyrosine kinase receptor that hasome homology to the IGF-IR; the fact that it cannot beetected by the anti-pY1316 antibody even when it isrossly overexpressed rules out any cross-reaction inells with a physiological number of IR. There is an-ther receptor that has substantial homologies withhe IGF-IR (and the IR): the insulin receptor-relatedeceptor (IRRR). However, the IRRR is missing the-termini of the IGF-IR and IR [57]. To be precise, the

RRR terminates at a residue corresponding to residue290 of the IGF-IR. As stated under Materials andethods, our peptide begins at residue 1319 of the

GF-IR.As explained in the Introduction, there are certain

onstraints in developing an antibody specific for an e

utophosphorylated IGF-IR. In the first place, ofourse, one needs a tyrosine (Y) residue that is phos-horylated in the peptide used for immunization. Itould have been better if we could have used the

yrosine kinase domain, which has three Y residues inlose proximity, all of which are autophosphorylated byGF-I. However, the homology with the IR, in this area,s 98%, which makes it very difficult to obtain an an-ibody that will not cross-react with the IR. The Yesidues at 1250/1251 of the IGF-IR could also behought desirable, but the area around 1250/1251 wasound on examination of the sequence to be, at leastheoretically, a poorly antigenic sequence. For theseeasons, we eventually selected a peptide sequence inhe C-terminus of the IGF-IR that has a phosphory-ated tyrosine and little homology to the correspondingequence of the IR.We have shown that the antibody we call anti-

Y1316 is due to immunization with the peptide weynthesized for the purpose. Competition with the orig-nal peptide almost abrogated the immunodetection ofhe autophosphorylated receptor. The properties of thenti-pY1316 antibody are clear. It will detect theGF-IR only when Y1316 is present and phosphory-ated. A mutant receptor in which Y1316 has been

utated is not recognized by the antibody. The samean be said of the d1245 receptor, in which the IGF-IRs truncated at residue 1245. The antibody will notecognize a receptor that cannot be autophosphory-ated, like the KA receptor, in which the IGF-IR has a

utation at lysine 1003, the ATP-binding site [54]. Theutant receptor with mutations at 1131, 1135, and

136 (the tyrosine kinase domain) is known to have aecreased, but not abolished, autophosphorylation51], and this is confirmed in the present experiments.

2 cells are negative for the anti-pY1316 antibody asell as any other antibody raised against the IGF-IR.inally, the anti-pY1316 antibody, besides being useful

n Western blots, is also capable of immunoprecipitat-ng the IGF-IR.

Although the anti-pY1316 antibody will be usefulnder experimental conditions in detecting an auto-hosphorylated antibody directly in Western blots, theeason we developed it is for use on tissue sections ofuman cancers. The IGF-IR is present in many humanancers [33, 34] and sometimes it is overexpressed.owever, the IGF-IR, to transmit its mitogenic andnti-apoptotic signals, must be activated by its ligands.he concentrations of these ligands in the immediateroximity of tumor cells undoubtedly varies, and oneould like to know how many of the cancer cells, at aiven time, actually have an activated (autophosphor-lated) IGF-IR. Our experiments confirm that an anti-ody to the C-terminus of the IGF-IR can easily detecthe receptor in sections of human cancers (we have

lected, in this paper, to show sections of breast cancer,
Page 7: Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers

pc

28 RUBINI ET AL.

FIG. 5. Staining of human breast cancer sections with the anti-pY1316 antibody. (A) A section from a human breast cancer treated withreimmune serum as a negative control. (B) A section from a human breast cancer stained with an anti-pY1316 antibody. The intraductal

ancer cells are intensely positive, while the interstitial tissue is essentially negative.
Page 8: Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers

Aptc

29IGF-I RECEPTOR AND CANCER

FIG. 6. Staining of tissue sections with antibodies against the IGF-I receptor. The antibody used in A was the anti-pY1316 antibody.gain, many intraductal cancer cells are intensely positive, although groups of tumor cells can also be seen that are negative or weaklyositive. The antibody used for the section in B was instead the one obtained with peptide 922 (see Materials and Methods) that recognizeshe C-terminus of the IGF-I receptor, regardless of its phosphorylation status. In both cases, the sections were from cases of human breast

ancer.
Page 9: Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers

bBaTptnpahsaTpbaoewmgdtTactmtastet

tsspdopdiesrpt

aIhspm

atso

t

1

1

1

30 RUBINI ET AL.

ut we could also detect it in metastatic colon cancer).ut we show now that the anti-pY1316 antibody canlso detect the receptor in human breast cancer cells.he next step is to investigate whether the anti-Y1316 antibody can be used for diagnostic or prognos-ic studies, for instance to determine the aggressive-ess of a tumor, or to determine differences betweenrimary tumors and metastases. Prospective studieslong this line have been initiated. Several reportsave emphasized a role of IGF-IR levels in the progno-is of cancer, especially breast cancer [27, 35, 37], andt least two reports have indicated that treatment withamoxifen causes a significant decrease in IGF-Ilasma levels [58, 59]. The anti-pY1316 antibody coulde very useful in following the development of diseasend the efficacy of therapeutic treatment. However,ne should be very careful in studies that have to bextended to a variety of situations. For instance, itould be a mistake to look only at cancers and theiretastases. An activated IGF-IR simply means that

rowth factors (in our case, IGF-I or IGF-II) are pro-uced in relative abundance in the immediate vicini-ies of the positive cells or by the cells themselves.hese growth factors are often produced by fibroblasts,nd the presence of a strong fibroblastic componentould activate the IGF-IR even in benign tumors. Fur-hermore, the IGF-IR is quasi-obligatory for transfor-ation, but, per se, is not an oncogene [1]: the impor-

ance of IGF-I (or IGF-II) in tumors is that it canccelerate their growth. For these reasons, an objectivetudy of the diagnostic and prognostic importance ofhis antibody must await an extended and exhaustivexamination of various forms of benign and malignantumors.

An antibody recognizing an activated PDGF recep-or has recently been published [60] and shown totain meningioma cells in humans. No extensivetudies on this antibody were presented in that pa-er. A similar antibody to the c-erbB-2 receptor wasescribed by Epstein et al. [61]. Antibodies that rec-gnized only an autophosphorylated IR were re-orted by Perlman et al. [62], although these authorsid not investigate the applicability of these antibod-es to sections of human tissues. Our antibody hasssentially the same properties as the antibody de-cribed by Shamah et al. [60] against the PDGFeceptor, but it is directed against the IGF-IR, whichlays a central role in the establishment and main-enance of the transformed phenotype [1].

In conclusion, we present here the development of anntibody that specifically recognizes an activated IGF-R, both in cells in tissue cultures and in sections ofuman cancers. This antibody should be very useful intudies of human cancers and their metastases. In areliminary survey, we have noticed that not all hu-

an breast cancers scored positive for the anti-pY1316

ntibody. However, the eventual usefulness of this an-ibody can be determined only with careful prospectivetudies in which several sections from different areasf benign and malignant tumors can be examined.

This work was supported by Grants CA 56309 and CA 53424 fromhe National Institutes of Health.

REFERENCES

1. Baserga, R., Hongo, A., Rubini, M., Prisco, M., and Valentinis,B. (1997). The IGF-I receptor in cell growth, transformation andapoptosis. Biochim. Biophys. Acta 1332, 105–126.

2. Kaleko, M., Rutter, W. G., and Miller, A. D. (1990). Overexpres-sion of the human insulin-like growth factor 1 receptor pro-motes ligand-dependent neoplastic transformation. Mol. Cell.Biol. 10, 464–473.

3. Pietrzkowski, Z., Lammers, R., Carpenter, G., Soderquist,A. M., Limardo, M., Phillips, P. D., Ullrich, A., and Baserga, R.(1992). Constitutive expression of insulin-like growth factor 1and insulin-like growth factor 1 receptor abrogates all require-ments for exogenous growth factors. Cell Growth Differ. 3, 199–205.

4. Resnicoff, M., Coppola, D., Sell, C., Rubin, R., Ferrone, S., andBaserga, R. (1994). Growth inhibition of human melanoma cellsin nude mice by antisense strategies to the type 1 insulin-likegrowth factor receptor. Cancer Res. 54, 4848–4850.

5. Resnicoff, M., Sell, C., Rubini, M., Coppola, D., Ambrose, D.,Baserga, R., and Rubin, R. (1994). Rat glioblastoma cells ex-pressing an antisense RNA to the insulin-like growth factor I(IGF-I) receptor are non-tumorigenic and induce regression ofwild-type tumors. Cancer Res. 54, 2218–2222.

6. Shapiro, D. N., Jones, B. G., Shapiro, L. H., Dias, P., andHoughton, P. J. (1994). Antisense-mediated reduction in insu-lin-like growth factor 1 receptor expression suppresses the ma-lignant phenotype of a human alveolar rhabdomyosarcoma.J. Clin. Invest. 94, 1235–1242.

7. Lee, C. T., Wu, S., Gabrivolich, D., Chen, H., Nadaf-Rahrov, S.,Ciernick, I. F., and Carbone, D. P. (1996). Antitumor effect of anadenovirus expressing antisense insulin-like growth factor Ireceptor on human lung cancer cell lines. Cancer Res. 56, 3038–3041.

8. Pass, H. L., Mew, D. J. Y., Carbone, M., Matthews, W. A.,Domington, J. S., Baserga, R., Walker, C. L., Resnicoff, M., andSteinberg, S. M. (1996). Inhibition of hamster mesotheliomatumorigenesis by an antisense expression plasmid to the insu-lin-like growth factor I receptor. Cancer Res. 56, 4044–4048.

9. Rininsland, F., Johnson, T. R., Chernicky, C. L., Schulze, E.,Burfeind, B., Ilan, J., and Ilan, J. (1997). Suppression of insulin-like growth factor I receptor by a triple-helix strategy inhibitsIGF-I transcription and tumorigenic potential of rat C6 glio-blastoma cells. Proc. Natl. Acad. Sci. USA 94, 5854–5859.

0. D’Ambrosio, C., Ferber, A., Resnicoff, M., and Baserga, R.(1996). A soluble insulin-like growth factor I receptor that in-duces apoptosis of tumor cells in vivo and inhibits tumorigen-esis. Cancer Res. 56, 4013–4020.

1. Prager, D., Li, H. L., Asa, S., and Melmed, S. (1994). Dominantnegative inhibition of tumorigenesis in vivo by human insulin-like growth factor receptor mutant. Proc. Natl. Acad. Sci. USA91, 2181–2185.

2. Long, L., Rubin, R., Baserga, R., and Brodt, P. (1995). Loss of

the metastatic phenotype in murine carcinoma cells expressing
Page 10: Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers

1

1

1

1

1

1

1

2

2

2

2

2

2

2

2

2

2

3

3

3

3

3

3

3

3

3

3

4

4

4

31IGF-I RECEPTOR AND CANCER

an antisense RNA to the insulin-like growth factor I receptor.Cancer Res. 55, 1006–1009.

3. Burfeind, P., Chernicky, C. L., Rininsland, F., Ilan, J., and Ilan,J. (1996). Antisense RNA to the type I insulin-like growth factorreceptor suppresses tumor growth and prevents invasion by ratprostate cancer cells in vivo. Proc. Natl. Acad. Sci. USA 93,7263–7268.

4. Dunn, S. E., Ehrlich, M., Sharp, N. J. H., Reiss, K., Solomon, G.,Hawkins, R., Baserga, R., and Barrett, J. C. (1998). A dominantnegative mutant of the insulin-like growth factor-1 receptorinhibits the adhesion, invasion and metastasis of breast cancer.Cancer Res. 58, 3353–3361.

5. McCubrey, J. A., Stillman, L. S., Mayhew, M. W., Algate, P. A.,Dellow, R. A., and Kaleko, M. (1991). Growth promoting effectsof insulin-like growth factor 1 (IGF-1) on hematopoietic cells.Overexpression of introduced IGF-1 receptor abrogates inter-leukin-3 dependency of murine factor dependent cells by liganddependent mechanism. Blood 78, 921–929.

6. Rodriguez-Tarduchy, G., Collins, M. K. L., Garcia, I., andLopez-Rivas, A. (1992). Insulin-like growth factor I inhibitsapoptosis in IL-3 dependent hemopoietic cells. J. Immunol. 149,535–540.

7. O’Connor, R., Kauffmann-Zeh, A., Liu, Y., Lehar, S., Evan, G. I.,Baserga, R., and Blattler, W. A. (1997). The IGF-I receptordomains for protection from apoptosis are distinct from thoserequired for proliferation and transformation. Mol. Cell. Biol.17, 427–435.

8. Zhou-Li, F., Xu, S., Dews, M., and Baserga, R. (1997). Co-operation of simian virus 40 T antigen and insulin receptorsubstrate-1 in protection from apoptosis induced by Interleu-kin-3 withdrawal. Oncogene 15, 961–970.

9. Dews, M., Nishimoto, I., and Baserga, R. (1997). IGF-I receptorprotection from apoptosis in cells lacking the IRS proteins.Receptors Signal Transduct. 7, 231–239.

0. Harrington, E. A., Bennett, M. R., Fanidi, A., and Evan, G. I.(1994). c-myc-induced apoptosis in fibroblasts is inhibited byspecific cytokines. EMBO J. 13, 3286–3295.

1. Dunn, S. E., Hardman, R. A., Kari, F. W., and Barrett, J. C.(1997). Insulin-like growth factor 1 (IGF-I) alters drug sensitiv-ity of HBL100 human breast cancer cells by inhibition of apo-ptosis induced by diverse anticancer drugs. Cancer Res. 57,2687–2693.

2. Sell, C., Baserga, R., and Rubin, R. (1995). Insulin-like growthfactor I (IGF-I) and the IGF-I receptor prevent etoposide-in-duced apoptosis. Cancer Res. 55, 303–306.

3. Wu, Y., Tewari, M., Cui, S., and Rubin, R. (1996). Activation ofthe insulin-like growth factor I receptor inhibits tumor necrosisfactor-induced cell death. J. Cell. Physiol. 168, 499–509.

4. Hsing, A. Y., Kadomatsu, K., Bonham, N. J., and Danielpour, D.(1996). Regulation of apoptosis induced by transforming growthfactor b1 in nontumorigenic and tumorigenic rat prostatic epi-thelial cell lines. Cancer Res. 56, 5146–5149.

5. Prisco, M., Hongo, A., Rizzo, M. G., Sacchi, A., and Baserga, R.(1997). The IGF-I receptor as a physiological relevant target ofp53 in apoptosis caused by interleukin-3 withdrawal. Mol. Cell.Biol. 17, 1084–1092.

6. Kulik, G., Klippel, A., and Weber, M. J. (1997). Antiapoptoticsignaling by the insulin-like growth factor I receptor, phospha-tidylinositol 3-kinase, and Akt. Mol. Cell. Biol. 17, 1595–1606.

7. Turner, B. C., Haffty, B. G., Narayanan, L., Yuan, J., Havre,P. A., Gumbs, A. A., Kaplan, L., Burgaud, J. L., Carter, D.,Baserga, R., and Glazer, P. M. (1997). Insulin-like growth factor

1 receptor overexpression mediates cellular resistance and local

breast cancer recurrence after lumpectomy and radiation. Can-cer Res. 57, 3079–3083.

8. Nakamura, S., Watanabe, H., Miura, M., and Sasaki, T. (1997).Effect of the insulin-like growth factor I receptor on ionizingradiation-induced cell death in mouse embryo fibroblasts. Exp.Cell Res. 235, 287–294.

9. D’Ambrosio, C., Valentinis, B., Prisco, M., Reiss, K., Rubini, M.,and Baserga, R. (1997). Protective effect of the IGF-I receptoron apoptosis induced by okadaic acid. Cancer Res. 57, 3264–3271.

0. Jung, Y. K., Miura, M., and Yuan, J. (1996). Suppression ofinterleukin-1 beta converting enzyme-mediated cell death byinsulin-like growth factor. J. Biol. Chem. 271, 5112–5117.

1. Nystrom, F. H., Ohman, P. K., Ekman, B. A., Osterlund, M. K.,Karlberg, B. E., and Arnqvist, H. J. (1997). Population-basedreference values for IGF-I and IGF-binding protein-1: relationswith metabolic and anthropometric variables. Eur. J. Endocr.135, 165–172.

2. Bereket, A., Wilson, T. A., Blethen, S. L., Fan, J., Frost, R. A.,Gelato, M. C., and Lang, C. H. (1996). Effect of short termfasting on free-dissociable insulin-like growth factor I concen-trations in normal human serum. J. Clin. Endocrinol. Metab.81, 4379–4384.

3. Cullen, K. J., Yee, D., and Rosen, N. (1991). Insulin-like growthfactors in human malignancy. Cancer Invest. 9, 443–454.

4. Macaulay, V. M. (1992). Insulin-like growth factors and cancer.Br. J. Cancer 65, 311–320.

5. Peyrat, J. P., Bonneterre, J., Beuscart, R., Djiana, J., and De-maille, A. (1988). Insulin-like growth factor I receptors in hu-man breast cancer and their relation to estradiol and proges-terone receptors. Cancer Res. 48, 6429–6433.

6. Peyrat, J. P., Bonneterre, J., Hecquet, B., Vennin, P., Louchez,M. M., Fournier, C., Lefebvre, J., and Demaille, A. (1993).Plasma insulin-like growth factor I (IGF-I) concentrations inhuman breast cancer. Eur. J. Cancer 29A, 492–497.

7. Railo, M. J., Smitten, K. V., and Pekonen, F. (1994). The prog-nostic value of insulin-like growth factor I in breast cancerpatients. Results of a follow-up study on 126 patients. Eur. J.Cancer 30A, 307–311.

8. Giani, C., Cullen, K. J., Campani, D., and Rasmussen, A.(1996). IGF-II mRNA and protein are expressed in the stromaof invasive breast cancers: An in situ hybridization and immu-nohistochemistry study. Breast Cancer Res. Treat. 41, 43–50.

9. Chan, J. M., Stampfer, M. J., Giovannucci, E., Gann, P. H., Ma,J., Wilkinson, P., Hennekens, C. H., and Pollak, M. (1998).Plasma insulin-like growth factor-1 and prostate cancer risk: Aprospective study. Science 279, 563–566.

0. Ullrich, A., Gray, A., Tam, A. W., Yang-Feng, T., Tsubokawa,M., Collins, C., Henzel, W., Le Bon, T., Kahuria, S., Chen, E.,Jakobs, S., Francke, U., Ramachandran, J., and Fujita-Yamaguchi, Y. (1986). Insulin-like growth factor I receptorprimary structure: comparison with insulin receptor suggestsstructural determinants that define functional specificity.EMBO J. 5, 503–512.

1. Satoh, T., Aramini, J. M., Li, S., Friedman, T., Gao, J., Edling,A., Townsend, R., Germann, M. W., Korngold, R., and Huang, Z.(1997). Bioactive peptide design based on protein surfaceepitopes: A cyclic heptapeptide mimics CD4 domain 1 CC9 loopand inhibits CD4 biological function. J. Biol. Chem. 272,12175–12180.

2. Li, S., Choksi, S., Shan, S., Hu, X., Gao, J., Korngold, R., andHuang, Z. (1998). Identification of the CD8 DE loop as a surfacefunctional epitope: implications for MHC class I binding and

CD8 inhibitor design. J. Biol. Chem. 273, 16442–16445.
Page 11: Characterization of an Antibody That Can Detect an Activated IGF-I Receptor in Human Cancers

4

4

4

4

4

4

4

5

5

5

5

5

5

5

5

5

5

6

6

6

RR

32 RUBINI ET AL.

3. King, D. S., Fields, C. G., and Fields, G. B. (1990). A cleavagemethod which minimizes side reactions following Fmoc solidphase peptide synthesis. Int. J. Peptide Protein Res. 36, 255–266.

4. Sell, C., Dumenil, G., Deveaud, C., Miura, M., Coppola, D.,DeAngelis, T., Rubin, R., Efstratiadis, A., and Baserga, R.(1994). Effect of a null mutation of the insulin-like growthfactor I receptor gene on growth and transformation of mouseembryo fibroblasts. Mol. Cell. Biol. 14, 3604–3612.

5. Sell, C., Rubini, M., Rubin, R., Liu, J. P., Efstratiadis, A., andBaserga, R. (1993). Simian virus 40 large tumor antigen isunable to transform mouse embryonic fibroblasts lacking type 1insulin-like growth factor receptor. Proc. Natl. Acad. Sci. USA90, 11217–11221.

6. Liu, J-P., Baker, J., Perkins, A. S., Robertson, E. J., and Efstra-tiadis, A. (1993). Mice carrying null mutations of the genesencoding insulin-like growth factor I (igf-1) and type 1 IGFreceptor (Igflr). Cell 75, 59–72.

7. Baker, J., Liu, J-P., Robertson, E. J., and Efstratiadis, A.(1993). Role of insulin-like growth factors in embryonic andpostnatal growth. Cell 75, 73–82.

8. Surmacz, E., Sell, C., Swantek, J., Kato, H., Roberts, C. T., Jr.,LeRoith, D., and Baserga, R. (1995). Dissociation of mitogenesisand transforming activity by C-terminal truncation of the in-sulin-like growth factor I receptor. Exp. Cell Res. 218, 370–380.

9. Miura, M., Li, S., and Baserga, R. (1995). Effect of a mutation attyrosine 950 of the insulin-like growth factor I receptor on thegrowth and transformation of cells. Cancer Res. 55, 663–667.

0. Miura, M., Surmacz, E., Burgaud, J. L., and Baserga, R. (1995).Different effects on mitogenesis and transformation of a muta-tion at tyrosine 1251 of the insulin-like growth factor I receptor.J. Biol. Chem. 270, 22639–22644.

1. Li, S., Ferber, A., Miura, M., and Baserga, R. (1994). Mitoge-nicity and transforming activity of the insulin-like growth fac-tor I receptor with mutations in the tyrosine kinase domain.J. Biol. Chem. 269, 32558–32564.

2. Li, S., Resnicoff, M., and Baserga, R. (1996). Effects of muta-tions at serines 1280–1283 on the mitogenic and transformingactivities of the insulin-like growth factor I receptor. J. Biol.Chem. 271, 12254–12260.

3. Hongo, A., D’Ambrosio, C., Miura, M., Morrione, A., and

Baserga, R. (1996). Mutational analysis of the mitogenic and

transforming activities of the insulin-like growth factor I recep-tor. Oncogene 12, 1231–1238.

4. Coppola, D., Ferber, A., Miura, M., Sell, C., D’Ambrosio, C.,Rubin, R., and Baserga, R. (1994). A functional insulin-likegrowth factor I receptor is required for the mitogenic and trans-forming activities of the epidermal growth factor receptor. Mol.Cell. Biol. 14, 4588–4595.

5. Panneerselvan, K., Reitz, H., Khan, S. A., and Bishayee, S.(1996). A conformational-specific anti-peptide antibody to the btype platelet-derived growth factor receptor also recognizes theactivated epidermal growth factor receptor. J. Biol. Chem. 270,7975–7979.

6. Morrione, A., Valentinis, B., Xu, S. Q., Yumet, G., Louvi, A.,Efstratiadis, A., and Baserga, R. (1997). Insulin-like growthfactor II stimulates cell proliferation through the insulin recep-tor. Proc. Natl. Acad. Sci. USA 94, 3777–3782.

7. Shier, P., and Watt, V. M. (1989). Primary structure of a puta-tive receptor for a ligand of the insulin family. J. Biol. Chem.264, 14605–14608.

8. Colletti, R. B., Roberts, J. D., Devlin, J. T., and Copeland, K. C.(1989). Effect of Tamoxifen on plasma-insulin growth factor 1 inpatients with breast cancer. Cancer Res. 49, 1882–1884.

9. Pollak, M., Costantino, J., Polychronakos, C., Blauer, S.,Guyda, H., Redmond, C., Fisher, B., and Margolese, R. (1990).Effect of Tamoxifen on serum insulin-like growth factor 1 levelsin stage 1 breast cancer patients. J. Natl. Cancer Inst. 82,1693–1697.

0. Shamah, S. M., Alberta, J. A., Giannobile, W. V., Guha, A.,Kwon, Y. K., Carroll, R. S., Black, P. M., and Stiles, C. D. (1997).Detection of activated Platelet-derived growth factor receptorsin human meningioma. Cancer Res. 57, 4141–4147.

1. Epstein, R. J., Druker, B. J., Roberts, T. M., and Stiles, C. D.(1992). Synthetic phosphopeptide immunogens yield activation-specific antibodies to the c-erbB-2 receptor. Proc. Natl. Acad.Sci. USA 89, 10435–10439.

2. Perlman, R., Bottaro, D. P., White, M. F., and Kahn, C. R.(1989). Conformational changes in the a- and b-subunits of theinsulin receptor identified by anti-peptide antibodies. J. Biol.

Chem. 264, 8946–8950.

eceived March 5, 1999evised version received May 14, 1999