Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using...

14
Cancer Therapy: Preclinical Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective Treatment of Diverse Epithelial Cancers: Preclinical Studies in Human Cancer Xenograft Models and Monkeys Thomas M. Cardillo 1 , Serengulam V. Govindan 1 , Robert M. Sharkey 2 , Preeti Trisal 1 , and David M. Goldenberg 2 Abstract Purpose: Evaluate the efficacy of an SN-38-anti-Trop-2 antibody–drug conjugate (ADC) against several human solid tumor types, and to assess its tolerability in mice and monkeys, the latter with tissue cross- reactivity to hRS7 similar to humans. Experimental Design: Two SN-38 derivatives, CL2-SN-38 and CL2A-SN-38, were conjugated to the anti-Trop-2–humanized antibody, hRS7. The immunoconjugates were characterized in vitro for stability, binding, and cytotoxicity. Efficacy was tested in five different human solid tumor-xenograft models that expressed Trop-2 antigen. Toxicity was assessed in mice and in Cynomolgus monkeys. Results: The hRS7 conjugates of the two SN-38 derivatives were equivalent in drug substitution (6), cell binding (K d 1.2 nmol/L), cytotoxicity (IC 50 2.2 nmol/L), and serum stability in vitro (t/ 1/2 20 hours). Exposure of cells to the ADC demonstrated signaling pathways leading to PARP cleavage, but differences versus free SN-38 in p53 and p21 upregulation were noted. Significant antitumor effects were produced by hRS7-SN-38 at nontoxic doses in mice bearing Calu-3 (P 0.05), Capan-1 (P < 0.018), BxPC-3 (P < 0.005), and COLO 205 tumors (P < 0.033) when compared to nontargeting control ADCs. Mice tolerated a dose of 2 12 mg/kg (SN-38 equivalents) with only short-lived elevations in ALT and AST liver enzyme levels. Cynomolgus monkeys infused with 2 0.96 mg/kg exhibited only transient decreases in blood counts, although, importantly, the values did not fall below normal ranges. Conclusions: The anti-Trop-2 hRS7-CL2A-SN-38 ADC provides significant and specific antitumor effects against a range of human solid tumor types. It is well tolerated in monkeys, with tissue Trop-2 expression similar to humans, at clinically relevant doses, and warrants clinical investigation. Clin Cancer Res; 17(10); 3157–69. Ó2011 AACR. Introduction Human trophoblast cell-surface antigen (Trop-2), also known as GA733-1 (gastric antigen 733-1), EGP-1 (epithe- lial glycoprotein-1), and TACSTD2 (tumor-associated cal- cium signal transducer), is expressed in a variety of human carcinomas and has prognostic significance in some, being associated with more aggressive disease (1–14). Studies of the functional role of Trop-2 in a mouse pancreatic cancer cell line transfected with murine Trop-2 revealed increased proliferation in low serum conditions, migration, and anchorage-independent growth in vitro, and enhanced growth rate with evidence of increased Ki-67 expression in vivo and a higher likelihood to metastasize (15). Trop-2 antigen’s distribution in many epithelial cancers makes it an attractive therapeutic target, but its normal tissue expression profile casts some doubt on Trop-2 as a suitable target (11, 14). Stein and colleagues (4) character- ized an antibody, designated RS7-3G11 (RS7), that bound to EGP-1 (16, 17), which was present in a number of solid tumors, but the antigen was also expressed in some normal tissues, usually in a lower intensity, or in restricted regions. Targeting and therapeutic efficacies were documented in a number of human tumor xenografts using radiolabeled RS7 (18–20), but this internalizing antibody did not show therapeutic activity in unconjugated form (18). However, in vitro it has demonstrated antibody-dependent cellular cytotoxicity (ADCC) activity against Trop-2 positive carci- nomas (21). We reported the preparation of antibody–drug conju- gates (ADC) using an anti-CEACAM5 (CD66e) IgG coupled to several derivatives of SN-38, a topoisomerase- I inhibitor that is the active component of irinotecan, or CPT-11 (22, 23). The derivatives varied in their in vitro Authors' Affiliations: 1 Immunomedics, Inc., Morris Plains; and 2 Center for Molecular Medicine and Immunology, Garden State Cancer Center, Belleville, New Jersey Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). T.M. Cardillo and S.V. Govindan contributed equally to this work. Corresponding Author: David M. Goldenberg, Garden State Cancer Center, 520 Belleville Ave, Belleville, NJ 07109. Phone: 973-844-7010; Fax: 973-844-7020; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-10-2939 Ó2011 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 3157 Research. on April 2, 2020. © 2011 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Transcript of Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using...

Page 1: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

Cancer Therapy: Preclinical

Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective Treatmentof Diverse Epithelial Cancers: Preclinical Studies in Human CancerXenograft Models and Monkeys

Thomas M. Cardillo1, Serengulam V. Govindan1, Robert M. Sharkey2, Preeti Trisal1, andDavid M. Goldenberg2

AbstractPurpose: Evaluate the efficacy of an SN-38-anti-Trop-2 antibody–drug conjugate (ADC) against several

human solid tumor types, and to assess its tolerability in mice and monkeys, the latter with tissue cross-

reactivity to hRS7 similar to humans.

Experimental Design: Two SN-38 derivatives, CL2-SN-38 and CL2A-SN-38, were conjugated to the

anti-Trop-2–humanized antibody, hRS7. The immunoconjugates were characterized in vitro for stability,

binding, and cytotoxicity. Efficacy was tested in five different human solid tumor-xenograft models that

expressed Trop-2 antigen. Toxicity was assessed in mice and in Cynomolgus monkeys.

Results: The hRS7 conjugates of the two SN-38 derivatives were equivalent in drug substitution (�6),

cell binding (Kd � 1.2 nmol/L), cytotoxicity (IC50 � 2.2 nmol/L), and serum stability in vitro (t/1/2 � 20

hours). Exposure of cells to the ADC demonstrated signaling pathways leading to PARP cleavage, but

differences versus free SN-38 in p53 and p21 upregulation were noted. Significant antitumor effects were

produced by hRS7-SN-38 at nontoxic doses in mice bearing Calu-3 (P � 0.05), Capan-1 (P < 0.018),

BxPC-3 (P < 0.005), and COLO 205 tumors (P < 0.033) when compared to nontargeting control ADCs.

Mice tolerated a dose of 2� 12mg/kg (SN-38 equivalents) with only short-lived elevations in ALT and AST

liver enzyme levels. Cynomolgus monkeys infused with 2� 0.96 mg/kg exhibited only transient decreases

in blood counts, although, importantly, the values did not fall below normal ranges.

Conclusions: The anti-Trop-2 hRS7-CL2A-SN-38 ADC provides significant and specific antitumor

effects against a range of human solid tumor types. It is well tolerated in monkeys, with tissue Trop-2

expression similar to humans, at clinically relevant doses, and warrants clinical investigation. Clin Cancer

Res; 17(10); 3157–69. �2011 AACR.

Introduction

Human trophoblast cell-surface antigen (Trop-2), alsoknown as GA733-1 (gastric antigen 733-1), EGP-1 (epithe-lial glycoprotein-1), and TACSTD2 (tumor-associated cal-cium signal transducer), is expressed in a variety of humancarcinomas and has prognostic significance in some, beingassociated with more aggressive disease (1–14). Studies ofthe functional role of Trop-2 in a mouse pancreatic cancercell line transfected with murine Trop-2 revealed increasedproliferation in low serum conditions, migration, and

anchorage-independent growth in vitro, and enhancedgrowth rate with evidence of increased Ki-67 expressionin vivo and a higher likelihood to metastasize (15).

Trop-2 antigen’s distribution in many epithelial cancersmakes it an attractive therapeutic target, but its normaltissue expression profile casts some doubt on Trop-2 as asuitable target (11, 14). Stein and colleagues (4) character-ized an antibody, designated RS7-3G11 (RS7), that boundto EGP-1 (16, 17), which was present in a number of solidtumors, but the antigen was also expressed in some normaltissues, usually in a lower intensity, or in restricted regions.Targeting and therapeutic efficacies were documented in anumber of human tumor xenografts using radiolabeledRS7 (18–20), but this internalizing antibody did not showtherapeutic activity in unconjugated form (18). However,in vitro it has demonstrated antibody-dependent cellularcytotoxicity (ADCC) activity against Trop-2 positive carci-nomas (21).

We reported the preparation of antibody–drug conju-gates (ADC) using an anti-CEACAM5 (CD66e) IgGcoupled to several derivatives of SN-38, a topoisomerase-I inhibitor that is the active component of irinotecan, orCPT-11 (22, 23). The derivatives varied in their in vitro

Authors' Affiliations: 1Immunomedics, Inc., Morris Plains; and 2Center forMolecular Medicine and Immunology, Garden State Cancer Center,Belleville, New Jersey

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

T.M. Cardillo and S.V. Govindan contributed equally to this work.

Corresponding Author: David M. Goldenberg, Garden State CancerCenter, 520 Belleville Ave, Belleville, NJ 07109. Phone: 973-844-7010;Fax: 973-844-7020; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-10-2939

�2011 American Association for Cancer Research.

ClinicalCancer

Research

www.aacrjournals.org 3157

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 2: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

serum stability properties, and in vivo studies found 1 form(designated CL2) to be more effective in preventing orarresting the growth of human colonic and pancreaticcancer xenografts than other linkages with more or lessstability. Importantly, these effects occurred at nontoxicdoses, with initial testing failing to determine a dose-limit-ing toxicity (23). These results were encouraging, but alsosurprising, because the CEACAM5 antibody does not inter-nalize, a property thought to be critical to the success of anADC.We speculated that the therapeutic activity of the anti-CEACAM5-SN-38 conjugate might be related to the slowrelease of SN-38 within the tumor after the antibodylocalized. Because irinotecan performs best when cellsare exposed during the S-phase of their growth cycle, asustained release is expected to improve responses (24–26).Indeed, SN-38 coupled to nontargeting, plasma extendingagents, such as polyethylene glycol (PEG) or micelles, hasshown improved efficacy over irinotecan or SN-38 alone(27–33), lending additional support to this mechanism.

Given the RS7 antibody’s broad reactivity with epithelialcancers and its internalization ability, we hypothesized thatan RS7-SN-38 conjugate could benefit not only from thesustained release of the drug, but also from direct intra-cellular delivery. Therefore, we prepared and tested theefficacy of SN-38 conjugates using a humanized versionof the murine RS7 antibody (hRS7). A slight modificationwas made to the SN-38 derivative that was reported pre-viously (22, 23), which improved the quality of the con-jugate without altering its in vitro stability or its efficacy invivo, and thus this new derivative (designated CL2A) iscurrently the preferred agent for SN-38 coupling to anti-bodies. Herein, we show the efficacy of the hRS7-SN-38

conjugate in several epithelial cancer cell lines implanted innudemice at nontoxic dosages, with other studies revealingthat substantially higher doses could be tolerated. Moreimportantly, toxicity studies in monkeys that also expressTrop-2 in similar tissues as humans showed that hRS7-SN-38 was tolerated at appreciably higher amounts than thetherapeutically effective dose in mice, providing evidencethat this conjugate is a promising agent for treating patientswith a wide range of epithelial cancers.

Materials and Methods

Cell lines, antibodies, and chemotherapeuticsAll human cancer cell lines used in this study were

purchased from the American Type Culture Collection.These include Calu-3 (non–small cell lung carcinoma),SK-MES-1 (squamous cell lung carcinoma), COLO 205(colonic adenocarcinoma), Capan-1 and BxPC-3 (pancrea-tic adenocarcinomas), and PC-3 (prostatic adenocarcino-mas). Humanized RS7 IgG and control humanized anti-CD20 (hA20 IgG, veltuzumab) and anti-CD22 (hLL2 IgG,epratuzumab) antibodies were prepared at Immunome-dics, Inc. Irinotecan (20 mg/mL) was obtained fromHospira, Inc.

SN-38 immunoconjugates and in vitro aspectsSynthesis of CL2-SN-38 has been described previously

(22). Its conjugation to hRS7 IgG and serum stability wereperformed as described (22, 23). Preparations of CL2A-SN-38 (M.W. 1480) and its hRS7 conjugate, and stability,binding, and cytotoxicity studies, were conducted asdescribed previously (22), and are presented in the Supple-mental Data. Cell lysates were prepared and immunoblot-ting for p21Waf1/Cip, p53, and PARP (poly-ADP-ribosepolymerase) was done as described in Supplemental Data.Concentrations, timing, and primary antibodies are shownin the figure legends.

In vivo therapeutic studiesFor all animal studies, the doses of SN-38 immunocon-

jugates and irinotecan are shown in SN-38 equivalents.Based on a mean SN-38/IgG substitution ratio of 6, a doseof 500 mg ADC to a 20-g mouse (25 mg/kg) contains0.4 mg/kg of SN-38. Irinotecan doses are likewiseshown as SN-38 equivalents (i.e., 40 mg irinotecan/kg isequivalent to 24 mg/kg of SN-38).

NCr female athymic nude (nu/nu)mice, 4 to 8weeks old,and male Swiss-Webster mice, 10 weeks old, were pur-chased from Taconic Farms. All animal studies wereapproved by the Center for Molecular Medicine andImmunology’s Institutional Animal Care and Use Com-mittee (IACUC). Tolerability studies were performed inCynomolgus monkeys (Macaca fascicularis; 2.5–4 kg maleand female) by SNBL USA, Ltd. after approval by SNBLUSA’s IACUC.

Animals were implanted subcutaneously with differenthuman cancer cell lines as described in the Supplementarydata. Tumor volume (TV) was determined by measure-

Translational Relevance

Successful irinotecan treatment of patients with solidtumors has been limited due in large part to the lowconversion rate of the CPT-11 prodrug into the activeSN-38 metabolite. Others have examined nontargetedforms of SN-38 as a means to bypass the need for thisconversion and to deliver SN-38 passively to tumors.Weconjugated SN-38 covalently to a humanized anti-Trop-2 antibody, hRS7. This antibody–drug conjugate hasspecific antitumor effects in a range of s.c. human cancerxenograft models, including non–small cell lung carci-noma, pancreatic, colorectal, and squamous cell lungcarcinomas, all at nontoxic doses (e.g., �3.2 mg/kgcumulative SN-38 equivalent dose). Trop-2 is widelyexpressed in many epithelial cancers, but also somenormal tissues, and therefore a dose escalation studyin Cynomolgus monkeys was performed to assess theclinical safety of this conjugate. Monkeys tolerated24 mg SN-38 equivalents/kg with only minor, reversi-ble, toxicities. Given its tumor-targeting and safety pro-file, hRS7-SN-38 may provide an improvement in themanagement of solid tumors responsive to irinotecan.

Cardillo et al.

Clin Cancer Res; 17(10) May 15, 2011 Clinical Cancer Research3158

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 3: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

ments in 2 dimensions using calipers, with volumesdefined as: L � w2/2, where L is the longest dimensionof the tumor and w is the shortest. Tumors ranged in sizebetween 0.10 and 0.47 cm3 when therapy began. Treat-ment regimens, dosages, and number of animals in eachexperiment are described in the Results. The lyophilizedhRS7-CL2A-SN-38 and control ADC were reconstitutedand diluted as required in sterile saline. All reagents wereadministered intraperitoneally (0.1 mL), except irinotecan,which was administered intravenously. The dosing regi-men was influenced by our prior investigations, where theADC was given every 4 days or twice weekly for varyinglengths of time (22, 23). This dosing frequency reflected aconsideration of the conjugate’s serum half-life in vitro, toallow a more continuous exposure to the ADC.

StatisticsGrowth curves are shown as percent change in initial TV

over time. Statistical analysis of tumor growth was based onarea under the curve (AUC). Profiles of individual tumorgrowth were obtained through linear-curve modeling. Anf-test was employed to determine equality of variancebetween groups before statistical analysis of growth curves.A 2-tailed t-test was used to assess statistical significancebetween the various treatment groups and controls, exceptfor the saline control, where a 1-tailed t-test was used(significance at P � 0.05). Statistical comparisons ofAUC were performed only up to the time that the firstanimal within a group was euthanized due to progression.

Pharmacokinetics and biodistribution111In-radiolabeled hRS7-CL2A-SN-38 and hRS7 IgG

were injected into nude mice bearing s.c. SK-MES-1 tumors(�0.3 cm3). One group was injected intravenously with 20mCi (250-mg protein) of 111In-hRS7-CL2A-SN-38, whereasanother group received 20 mCi (250-mg protein) of 111In-hRS7 IgG. At various timepoints mice (5 per timepoint)were anesthetized, bled via intracardiac puncture, and theneuthanized. Tumors and various tissues were removed,weighed, and counted by g scintillation to determine thepercentage injected dose per gram tissue (% ID/g). A thirdgroup was injected with 250 mg of unlabeled hRS7-CL2A-SN-38 3 days before the administration of 111In-hRS7-CL2A-SN-38 and likewise necropsied. A 2-tailed t-testwas used to compare hRS7-CL2A-SN-38 and hRS7 IgGuptake after determining equality of variance using thef-test. Pharmacokinetic analysis on blood clearance wasperformed using WinNonLin software (Parsight Corp.).

Tolerability in Swiss-Webster mice and CynomolgusmonkeysDetails of these tolerability studies are given in the

Supplementary data. Briefly, mice were sorted into 4 groupseach to receive 2-mL i.p. injections of either a sodiumacetate buffer control or 3 different doses of hRS7-CL2A-SN-38 (4, 8, or 12 mg/kg of SN-38) on days 0 and 3followed by blood and serum collection, as describedin Results. Cynomolgus monkeys (3 male and 3 female;

2.5–4.0 kg) were administered 2 different doses of hRS7-CL2A-SN-38. Dosages, times, and number ofmonkeys bledfor evaluation of possible hematologic toxicities and serumchemistries are described in the Results.

Results

Stability and potency of hRS7-CL2A-SN-38Two different linkages were used to conjugate SN-38 to

hRS7 IgG. The first is termed CL2-SN-38 and has beendescribed previously (22, 23). Aminor change was made tothe synthesis of our CL2 linker in that the phenylalaninemoiety was removed (Fig. 1A). This change simplified thesynthesis, but did not affect the conjugation outcome (e.g.,both CL2-SN-38 and CL2A-SN-38 incorporated �6 SN-38per IgG molecule). Side-by-side comparisons found nosignificant differences in serum stability, antigen binding,or in vitro cytotoxicity.

To confirm that the change in the SN-38 linker fromCL2 to CL2A did not impact in vivo potency, hRS7-CL2Aand hRS7-CL2-SN-38 were compared in mice bearingCOLO 205 or Capan-1 tumors (Fig. 1B and C, respec-tively), using 0.4 mg or 0.2 mg/kg SN-38 twice weekly� 4weeks, respectively, and with starting tumors of 0.25 cm3

size in both studies. Both the hRS7-CL2A and CL2-SN-38conjugates significantly inhibited tumor growth com-pared to untreated (AUC14days P < 0.002 vs. saline inCOLO 205 model; AUC21days P < 0.001 vs. saline inCapan-1 model), and a nontargeting anti-CD20 controlADC, hA20-CL2A-SN-38 (AUC14days P < 0.003 in COLO-205 model; AUC35days: P < 0.002 in Capan-1 model). Atthe end of the study (day 140) in the Capan-1 model,50% of the mice treated with hRS7-CL2A-SN-38 and 40%of the hRS7-CL2-SN-38 mice were tumor-free, whereasonly 20% of the hA20-ADC-treated animals had novisible sign of disease. Importantly, there were no differ-ences in efficacy between the 2 specific conjugates in boththe tumor models.

Mechanism of actionIn vitro cytotoxicity studies demonstrated that hRS7-

CL2A-SN-38 had IC50 values in the nmol/L range againstseveral different solid tumor lines (Table 1). The IC50 withfree SN-38 was lower than the conjugate in all cell lines.Although there was no correlation between Trop-2 expres-sion and sensitivity to hRS7-CL2A-SN-38, the IC50 ratio ofthe ADC versus free SN-38 was lower in the higher Trop-2-expressing cells, most likely reflecting the enhanced abilityto internalize the drug when more antigen is present.

SN-38 is known to activate several signaling pathways incells, leading to apoptosis (34–37). Our initial studiesexamined the expression of 2 proteins involved in earlysignaling events (p21Waf1/Cip1 and p53) and 1 late apop-totic event [cleavage of poly-ADP-ribose polymerase(PARP)] in vitro (Fig. 2). In BxPC-3 (Fig. 2A), SN-38 ledto a 20-fold increase in p21Waf1/Cip1 expression, whereashRS7-CL2A-SN-38 resulted in only a 10-fold increase, afinding consistent with the higher activity with free SN-38

Anti-TROP-2-SN-38 Conjugate in Cancer Therapy

www.aacrjournals.org Clin Cancer Res; 17(10) May 15, 2011 3159

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 4: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

in this cell line (Table 1). However, hRS7-CL2A-SN-38increased p21Waf1/Cip1 expression in Calu-3 more than 2-fold over free SN-38 (Fig. 2B).

A greater disparity between hRS7-CL2A-SN-38- and freeSN-38-mediated signaling events was observed in p53expression. In both BxPC-3 and Calu-3, upregulation ofp53 with free SN-38 was not evident until 48 hours,whereas hRS7-CL2A-SN-38 upregulated p53 within 24hours. In addition, p53 expression in cells exposed tothe ADC was higher in both cell lines compared to

SN-38. Interestingly, although hRS7 IgG had no appreci-able effect on p21Waf1/Cip1 expression, it did induce theupregulation of p53 in both BxPC-3 and Calu-3, but onlyafter a 48-hour exposure. In terms of later apoptotic events,cleavage of PARP was evident in both cell lines whenincubated with either SN-38 or the conjugate (Fig. 2C).The presence of the cleaved PARP was higher at 24 hours inBxPC-3,which correlateswithhigh expressionof p21and itslower IC50. The higher degree of cleavage with free SN-38over the ADC was consistent with the cytotoxicity findings.

A

B C

hRS7

CL2-SN-38:CL2A-SN-38:

AA = PheAA = None

SN-38

HO 10 7

7

N

NN

N

N

N

N NH H

H

NO

O

OOO

CH2O

O

[AA]-Lys-O

O

O

Short PEGfor solubility

Cathepsin Bcleavable peptidewhen AA = Phe

O

OO

O

pH-dependentcleavage site

Agent

SN-38/lgGSubstitution

Ratio

FreeSN-38

(%)

HumanSerum

Half-Life(h)

Cell Binding:Kd (nM)

(95% C.I.)

Cytotoxicity:IC50 (nM)(95% C.I.)

hRS7-CL2-SN-38

hRS7-CL2A-SN-38

6.2

6.1

0.7 22.1

20.30.6

1.19 (0.89 to 149) 4.12 (2.88 to 5.89)

4.24 (2.99 to 6.01)1.09 (0.97 to 1.21)

Per

cent

cha

nge

in tu

mor

vol

ume

Per

cent

cha

nge

in tu

mor

vol

umeCOLO 205

700

400

800

600

500

300

200

100

900

0

700

400

800

600

500

300

200

100

0 10 20 30 40 50 60

900

00 7 14 21

Time (days)

hRS7-CL2-SN-38 (0.4 mg/kg)

hRS7-CL2A-SN-38 (0.4 mg/kg)

hA20-CL2A-SN-38 (0.4 mg/kg)

Saline control

hRS7 ADC vs. controlsP < 0.003: AUC

hRS7-CL2-SN-38 (0.2 mg/kg)

hRS7-CL2A-SN-38 (0.2 mg/kg)

hA20-CL2A-SN-38 (0.2 mg/kg)

Saline control

AUC hRS7 ADC versus*Saline P < 0.001**hA20 ADC P < 0.001

Time (days)

Capan-1

Figure 1. Structure and characterization of SN-38 conjugates: hRS7-CL2-SN-38 and hRS7-CL2A-SN-38. A, the chemical structure of the linkage ofSN-38 to the hRS7 antibody via the CL-linker. A single amino acid deletion delineates the difference between the CL2-SN-38 and CL2A-SN-38 formsof the hRS7 anti-Trop-2 ADC. Comparisons in chemistry and potency are shown in the table. Comparability of hRS7-CL2-SN-38 versus hRS7-CL2A-SN-38in mice bearing COLO 205 (B) and Capan-1 tumors (C). Animals were treated twice weekly for 4 weeks as indicated by the arrows. COLO 205mice (N ¼ 6) were treated with 0.4 mg/kg ADC and tumors measured twice a week. Capan-1 mice (N ¼ 10) were treated with 0.2 mg/kg ADC and tumorsmeasured weekly.

Cardillo et al.

Clin Cancer Res; 17(10) May 15, 2011 Clinical Cancer Research3160

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 5: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

Efficacy of hRS7-SN-38Because Trop-2 is widely expressed in several human

carcinomas, studies were performed in several differenthuman cancer models, which started with an evaluationof the hRS7-CL2-SN-38 linkage, but later, conjugates withthe CL2A-linkage were used. Calu-3–bearing nude micegiven 0.04 mg SN-38/kg of the hRS7-CL2-SN-38 every 4days � 4 had a significantly improved response comparedto animals administered the equivalent amount of hLL2-CL2-SN-38 (TV ¼ 0.14 � 0.22 cm3 vs. 0.80 � 0.91 cm3,respectively; AUC42days P < 0.026; Fig. 3A). A dose–response was observed when the dose was increased to0.4 mg/kg SN-38. At this higher dose level, all mice giventhe specific hRS7 conjugate were "cured" within 28 days,and remained tumor-free until the end of the study on day147, whereas tumors regrew in animals treated with theirrelevant ADC (specific vs. irrelevant AUC98days: P¼ 0.05).In mice receiving the mixture of hRS7 IgG and SN-38,tumors progressed >4.5-fold by day 56 (TV ¼ 1.10 � 0.88cm3; AUC56days P < 0.006 vs. hRS7-CL2-SN-38).Efficacy also was examined in human colonic (COLO

205) and pancreatic (Capan-1) tumor xenografts. In COLO205 tumor-bearing animals, (Fig. 3B), hRS7-CL2-SN-38(0.4 mg/kg, q4dx8) prevented tumor growth over the28-day treatment period with significantly smaller tumorscompared to control anti-CD20 ADC (hA20-CL2-SN-38),or hRS7 IgG (TV ¼ 0.16 � 0.09 cm3, 1.19 � 0.59 cm3, and1.77 � 0.93 cm3, respectively; AUC28days P < 0.016). TheMTD of irinotecan (24 mg SN-38/kg, q2dx5) was as effec-tive as hRS7-CL2-SN-38, because mouse serum can moreefficiently convert irinotecan to SN-38 (38–41) thanhuman serum, but the SN-38 dose in irinotecan (2,400mg cumulative) was 37.5-fold greater than with the con-jugate (64 mg total). Animals bearing Capan-1 showed nosignificant response to irinotecan alone when given at anSN-38-dose equivalent to the hRS7-CL2-SN-38 conjugate(e.g., on day 35, average tumor size was 0.04 � 0.05 cm3 in

animals given 0.4 mg SN-38/kg hRS7-SN-38 vs. 1.78 �0.62 cm3 in irinotecan-treated animals given 0.4mg/kg SN-38; AUCday35 P < 0.001; Fig. 3C).When the irinotecan dosewas increased 10-fold to 4 mg/kg SN-38, the responseimproved, but still was not as significant as the conjugateat the 0.4 mg/kg SN-38 dose level (TV ¼ 0.17 � 0.18 cm3

vs. 1.69� 0.47 cm3, AUCday49 P < 0.001). An equal dose ofnontargeting hA20-CL2-SN-38 also had a significant anti-tumor effect as compared to irinotecan-treated animals, butthe specific hRS7 conjugate was significantly better than theirrelevant ADC (TV¼ 0.17� 0.18 cm3 vs. 0.80� 0.68 cm3,AUCday49 P < 0.018).

Studies with the hRS7-CL2A-SN-38 ADC were thenextended to 2 other models of human epithelial cancers.In mice bearing BxPC-3 human pancreatic tumors(Fig. 3D), hRS7-CL2A-SN-38 again significantly inhibitedtumor growth in comparison to control mice treated withsaline or an equivalent amount of nontargeting hA20-CL2A-SN-38 (TV ¼ 0.24 � 0.11 cm3 vs. 1.17 � 0.45cm3 and 1.05 � 0.73 cm3, respectively; AUCday21 P <0.001), or irinotecan given at a 10-fold higher SN-38equivalent dose (TV ¼ 0.27 � 0.18 cm3 vs. 0.90 � 0.62cm3, respectively; AUCday25 P < 0.004). Interestingly, inmice bearing SK-MES-1 human squamous cell lung tumorstreated with 0.4 mg/kg of the ADC (Fig. 3E), tumor growthinhibition was superior to saline or unconjugated hRS7 IgG(TV ¼ 0.36 � 0.25 cm3 vs. 1.02 � 0.70 cm3 and 1.30 �1.08 cm3, respectively; AUC28 days, P < 0.043), but non-targeting hA20-CL2A-SN-38 or the MTD of irinotecanprovided the same antitumor effects as the specific hRS7-SN-38 conjugate.

In all murine studies, the hRS7-SN-38 ADC was welltolerated in terms of body weight loss (not shown).

Biodistribution of hRS7-CL2A-SN-38The biodistributions of hRS7-CL2A-SN-38 or unconju-

gated hRS7 IgG were compared in mice bearing SK-MES-1

Table 1. Expression of Trop-2 and in vitro cytotoxicity of SN-38 and hRS7-SN-38 in several solid tumorlines

Trop-2 expression viaFACS

Cytotoxicity results

Cell line Medianfluorescence(background)

Percentpositive

SN-38 95% CI hRS7-SN-38a 95% CI ADC/freeSN-38 ratio

IC50 (nmol/L) IC50 (nmol/L) IC50 (nmol/L) IC50 (nmol/L)

Calu-3 282.2 (4.7) 99.6% 7.19 5.77–8.95 9.97 8.12–12.25 1.39COLO 205 141.5 (4.5) 99.5% 1.02 0.66–1.57 1.95 1.26–3.01 1.91Capan-1 100.0 (5.0) 94.2% 3.50 2.17–5.65 6.99 5.02–9.72 2.00PC-3 46.2 (5.5) 73.6% 1.86 1.16–2.99 4.24 2.99–6.01 2.28SK-MES-1 44.0 (3.5) 91.2% 8.61 6.30–11.76 23.14 17.98–29.78 2.69BxPC-3 26.4 (3.1) 98.3% 1.44 1.04–2.00 4.03 3.25–4.98 2.80

aIC50-value is shown as SN-38 equivalents of hRS7-SN-38.

Anti-TROP-2-SN-38 Conjugate in Cancer Therapy

www.aacrjournals.org Clin Cancer Res; 17(10) May 15, 2011 3161

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 6: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

human squamous cell lung carcinoma xenografts (Supple-mentary Table S1), using the respective 111In-labeled sub-strates. A pharmacokinetic analysis was performed todetermine the clearance of hRS7-CL2A-SN-38 relative tounconjugated hRS7 (Fig. 4A). The ADC cleared faster thanthe equivalent amount of unconjugated hRS7, with theADC exhibiting�40% shorter half-life and mean residencetime. Nonetheless, this had a minimal impact on tumoruptake (Fig. 4B). Although there were significant differ-ences at the 24- and 48-hour timepoints, by 72 hours (peakuptake) the amounts of both agents in the tumor were

similar. Among the normal tissues, hepatic (Fig. 4C) andsplenic (Fig. 4D) differences were the most striking. At 24hours postinjection, there was >2-fold more hRS7-CL2A-SN-38 in the liver than hRS7 IgG. Conversely, in the spleenthere was 3-fold more parental hRS7 IgG present at peakuptake (48-hour timepoint) than hRS7-CL2A-SN-38.Uptake and clearance in the rest of the tissues generallyreflected differences in the blood concentration.

Because twice-weekly doses were given for therapy,tumor uptake in a group of animals that first received apredose of 0.2 mg/kg (250 mg protein) of the hRS7 ADC

A B

C

BxPC-3

BxPC-3

Calu-3

Calu-3

Treatment Treatment

Treatment Treatment

Treatment

Treatment

Time (hrs) Time (hrs)

Time (hrs) Time (hrs)

Time (hrs)

Time (hrs)

p21 p21

p21 p21

p53 p53

p53 p53

β-actin β-actin

Changes in expression levels Changes in expression levels

0

0

0

0

24 24 24 24

24

24 24 24

24 24

24

48 48 4848

48

48 48 48

48 48

48

24

24 24

2448

48 48

48

SN-38 SN-38

SN-38

SN-38

SN-38

hRS7-SN-38 hRS7-SN-38

hRS7-SN-38

hRS7-SN-38

hRS7-SN-38

hRS7 lgG hRS7 lgG

24 24 2448 4848

SN-38 hRS7-SN-38 hRS7 lgG

hRS7 lgG

hRS7 lgG

hRS7 lgG

1 1

0

0

6.2 1.1

0.8 3.2 9.8 1.22.5 2.6

1.35.8 11.3 14.4

11

20.8

0.7 3.9 5.1 6.1 1.2 1.7

20.9 11.3 9.5 1.3 1.0

PARP (116 kDa)

PARP (116 kDa)

Cleaved PARP (89 kDa)

Cleaved PARP (89 kDa)

Figure 2. Western blot analysis for early and late signaling events mediated by hRS7-CL2A-SN-38. Cells were plated overnight in 6-well plates beforethe addition of hRS7-CL2A-SN-38 (1 mmol/L SN-38 equivalents), free SN-38 (1 mmol/L), or the protein equivalent of hRS7 IgG (25 mg/mL). At the indicatedtimes, cells were lysed and 20 mg protein from these lysates subjected to SDS-PAGE (4%–20%) followed by blotting with antibodies to p21, p53, b-actin, orPARP. Time 0 lanes are from untreated cells grown in the plates for 48 hours in growth media alone and represent basal expression levels. Changes inexpression are normalized to b-actin loading controls and are relative to levels in the untreated cells.

Cardillo et al.

Clin Cancer Res; 17(10) May 15, 2011 Clinical Cancer Research3162

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 7: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

3 days before the injection of the 111In-labeled antibodywas examined. Tumor uptake of 111In-hRS7-CL2A-SN-38in predosed mice was substantially reduced at every time-point in comparison to animals that did not receive thepredose (e.g., at 72 hours, predosed tumor uptake was12.5% � 3.8% ID/g vs. 25.4% � 8.1% ID/g in animals notgiven the predose; P ¼ 0.0123; Fig. 4E). Predosing had no

appreciable impact on blood clearance or tissue uptake(Supplementary Table S2). These studies suggest that insome tumor models, tumor accretion of the specific anti-body can be reduced by the preceding dose(s), which likelyexplains why the specificity of a therapeutic response couldbe diminished with increasing ADC doses and why furtherdose escalation is not indicated.

900

A BCOLO 205

BxPC-3

D

Calu-3

Capan-1 *

**

**

*

**

C

ESK-MES-1

700

400

800

600

500

300

200

0

–100

1000

–200

Per

cent

cha

nge

in tu

mor

vol

ume

Per

cent

cha

nge

in tu

mor

vol

ume

Per

cent

cha

nge

in tu

mor

vol

ume

Per

cent

cha

nge

in tu

mor

vol

ume

Per

cent

cha

nge

in tu

mor

vol

ume 1000

900800700600500400300200100

1100

–100 7 14 21 28 35

0

1200

1000

800

600

400

200

1400

–200 7 14 21 28 35 42 49 56 63

0

1200

1000

800

600

400

200

1400

–200

700

600

500

400

300

200

100

800

–100 5 10 15 20 25 30

0

7 14 21 28 35Time (day)

Time (days)

Time (days)

Time (days)

0

10 20 30 40 50 60 70 80 90 100 110

hRS7-CL2-SN-38 (0.4 mg/kg)

hRS7-CL2-SN-38 (0.04 mg/kg)

hRS7-CL2A-SN-38 (0.4 mg/kg)

hRS7-CL2-SN-38 (0.4 mg/kg)

hA20-CL2A-SN-38 (0.04 mg/kg)

hA20-CL2A-SN-38 (0.04 mg/kg)

hRS7 IgG (25 mg/kg)

hRS7 IgG (50 mg/kg)

Saline

hRS7-CL2-SN-38 (0.4 mg/kg)

hRS7-SN-38 versus

*Saline and Irinotecan P < 0.001

**hA20-SN-38 P < 0.018

hRS7-SN-38 versusSaline and hRS7 IgGP < 0.043

hRS7-SN-38 versus

*Saline and hA20-SN-38 P < 0.005

**Irinotecan P < 0.004

P < 0.033 hRS7-SN-38 and Irinotecanversus all other groups

hA20-CL2-SN-38 (0.4 mg/kg)

hA20-CL2-SN-38 (0.4 mg/kg)

Irinotecan (4 mg/kg)

Irinotecan (24 mg/kg)

Irinotecan (0.4 mg/kg)

Saline Control

Irinotecan (24 mg/kg)

hRS7-CL2A-SN-38 (0.4 mg/kg)

Irinotecan (4 mg/kg)

Saline

Saline Control

hLL2-CL2-SN-38 (0.4 mg/kg)

hLL2-CL2-SN-38 (0.04 mg/kg)

hRS7 IgG + SN-38 (25 mg/kg + 0.4 mg/kg)

P ≤ 0.05 for hRS7-SN38(0.4 mg/kg) vs. allgroups

Saline

100

Time (days)

Figure 3. Therapeutic efficacy of hRS7-SN-38 ADC in several solid tumor-xenograft disease models. Efficacy of hRS7-CL2-SN-38 and hRS7-CL2A-SN-38 ADC treatment was studied in mice bearing human non–small cell lung, colorectal, pancreatic, and squamous cell lung tumor xenografts. All the ADCsand controls were administered in the amounts indicated (expressed as amount of SN-38 per dose; long arrows ¼ conjugate injections, short arrows¼ irinotecan injections). A, mice bearing Calu-3 tumors (N ¼ 5–7) were injected with hRS7-CL2-SN-38 every 4 days for a total of 4 injections (q4dx4). B,COLO 205 tumor-bearing mice (N¼ 5) were injected 8 times (q4dx8) with the ADC or every 2 days for a total of 5 injections (q2dx5) with the MTD of irinotecan.Capan-1 (C; N ¼ 10) or BxPC-3 tumor-bearing mice (D; N ¼ 10) were treated twice weekly for 4 weeks with the agents indicated. E, in addition to ADC giventwice weekly for 4 week, SK-MES-1 tumor-bearing (N ¼ 8) mice received the MTD of CPT-11 (q2dx5).

Anti-TROP-2-SN-38 Conjugate in Cancer Therapy

www.aacrjournals.org Clin Cancer Res; 17(10) May 15, 2011 3163

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 8: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

A B

D

100

1

10

0 25 50 75 100 125 150 175 200Time (hrs) 0 25 50 75 100 125 150 175 200

Time (hrs)

hRS7 lgG

hRS7 lgGhRS7 lgG

*

*

**

**

*

*

**

*

***

**

hRS7 lgG

hRS7 lgG

hRS7-CL2A-SN-38

hRS7-CL2A-SN-38

hRS7-CL2A-SN-38

hRS7-CL2A-SN-38

hRS7-CL2A-SN-38

hRS7-CL2A-SN-38 after Pre dose

hRS7-CL2A-SN-38

Pharmacokinetics

161

161117

1064 230

447

T1/2 AUC0.08 MRT0.08(h) (day∗mg/L) (h)

Per

cent

ID/g

(m

ean

± S

D)

Per

cent

ID/g

(m

ean

± S

D)

100

1

10

Per

cent

inje

cted

dos

e pe

r gr

am (

mea

n ±

SD

)

Per

cent

ID/g

(m

ean

± S

D)

C

E

25

15

35

30

25

20

15

10

5

0

Per

cent

ID/g

(m

ean

± S

D)

35

30

25

20

15

10

5

0

4 24 48 72Time (hrs)

5

20

10

00.083 2 4 168724824 0.083 2 4 168724824

Time (hrs) Time (hrs)

*P < 0.001

*P < 0.0132

*P < 0.046

*P < 0.025 Figure 4. Biodistribution and pharmacokinetics of hRS7-CL2A-SN-38 in SK-MES-1 tumor-bearing mice. A–D, mice wereinjected with either 111In-DTPA-hRS7-CL2A-SN-38 or111In-DTPA-hRS7 IgG (N ¼ 5/observation). Data are expressedas % ID/g. A, blood clearance and pharmacokineticsparameters; B, tumor; C, liver; D, spleen are shown. E, predosingwith 250 mg of hRS7-CL2A-SN-38 3 days before receiving111In-DTPA-hRS7-CL2A-SN-38 effect on tumor uptake.

Cardillo et al.

Clin Cancer Res; 17(10) May 15, 2011 Clinical Cancer Research3164

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 9: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

Tolerability of hRS7-CL2A-SN-38 in Swiss-Webstermice and Cynomolgus monkeysSwiss-Webster mice tolerated 2 doses over 3 days, each of

4, 8, and 12 mg SN-38/kg of the hRS7-CL2A-SN-38, withminimal transient weight loss (Supplementary Fig. S2). Nohematopoietic toxicity occurred and serumchemistries onlyrevealed elevated aspartate transaminase (AST) and alaninetransaminase (ALT; Fig. 5). Seven days after treatment, ASTrose above normal levels (>298 U/L) in all 3 treatmentgroups (Fig. 5A), with the largest proportion of mice beingin the 2 � 8 mg/kg group. However, by 15 days posttreat-ment, most animals were within the normal range. ALTlevels were also above the normal range (>77 U/L) within

7 days of treatment (Fig. 5B) and with evidence of normal-ization by Day 15. Livers from all these mice did not showhistologic evidence of tissue damage (not shown). In termsof renal function, only glucose and chloride levels weresomewhat elevated in the treated groups. At 2 � 8 mg/kg,5 of 7 mice had slightly elevated glucose levels (range of273–320 mg/dL, upper end of normal 263 mg/dL) thatreturned to normal by 15 days postinjection. Similarly,chloride levels were slightly elevated, ranging from 116 to127 mmol/L (upper end of normal range 115 mmol/L) inthe 2 highest dosage groups (57% in the 2� 8mg/kg groupand 100% of the mice in the 2 � 12 mg/kg group), andremained elevated out to 15 days postinjection. This also

A B

C D

day 7 day 7day 15 day 15

Per

cent

of m

ice

with

ele

vate

d A

ST

(>2

98 U

/L)

Per

cent

of m

ice

with

ele

vate

d A

LT (

>77

U/L

)

75 75

50 50

25 25

100

0

100

0

Control

Control

2 x 4 m

g/kg

2 x 4 m

g/kg

2 x 8 m

g/kg

2 x 8 m

g/kg

2 x 12 m

g/kg

2 x 12 m

g/kg

Control

Control

2 x 4 m

g/kg

2 x 4 m

g/kg

2 x 8 m

g/kg

2 x 8 m

g/kg

2 x 12 m

g/kg

2 x 12 m

g/kg

Platelet

Lowerlimit ofnormalrange

Lowerlimit ofnormalrange

*P < 0.004 vs. control*P < 0.006 vs. control

Mea

n co

unt (

103 /

μL)

Mea

n ce

ll co

unt (

103 /

μL) 450

350

250

15010050

550

0

300

200

500

400

Time (day)–1 3 6 11

Time (day)–1 3 6 11

8

7

6

5

4

3

2

1

9

0

Neutrophil

Control0.96 mg/kg

1.92 mg/kg

***

Figure 5. Tolerability of hRS7-CL2A-SN-38 in Swiss-Webster mice and Cynomolgus monkeys. A and B, fifty-six Swiss-Webster mice were administered 2 i.p.doses of buffer or the hRS7-CL2A-SN-38 3 days apart (4, 8, or 12 mg/kg of SN-38 per dose; 250, 500, or 750 mg conjugate protein/kg per dose).Seven and 15 days after the last injection, 7 mice from each group were euthanized, with blood counts and serum chemistries performed. Graphs showthe percent of animals in each group that had elevated levels of AST (A) and ALT (B). Six monkeys per group were injected twice 3 days apart withbuffer (control) or hRS7-CL2A-SN-38 at 0.96 mg/kg or 1.92 mg/kg of SN-38 equivalents per dose (60 and 120 mg/kg conjugate protein). All animals were bledon day �1, 3, and 6. Four monkeys were bled on day 11 in the 0.96 mg/kg group, 3 in the 1.92 mg/kg group. Changes in neutrophil (C) and plateletcounts (D), respectively in Cynomolgus monkeys.

Anti-TROP-2-SN-38 Conjugate in Cancer Therapy

www.aacrjournals.org Clin Cancer Res; 17(10) May 15, 2011 3165

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 10: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

could be indicativeof gastrointestinal toxicity, becausemostchloride is obtained through absorption by the gut; how-ever, at termination, there was no histologic evidence oftissue damage in any organ system examined (not shown).

Because mice do not express Trop-2 identified by hRS7, amore suitable model was required to determine the poten-tial of the hRS7 conjugate for clinical use. Immunohistol-ogy studies revealed binding in multiple tissues in bothhumans and Cynomolgus monkeys (breast, eye, gastroin-testinal tract, kidney, lung, ovary, fallopian tube, pancreas,parathyroid, prostate, salivary gland, skin, thymus, thyroid,tonsil, ureter, urinary bladder, and uterus; not shown).Based on this cross-reactivity, a tolerability study wasperformed in monkeys.

The group receiving 2 � 0.96 mg SN-38/kg of hRS7-CL2A-SN-38 had no significant clinical events followingthe infusion and through the termination of the study.Weight loss did not exceed 7.3% and returned to acclima-tion weights by day 15. Transient decreases were noted inmost of the blood count data (neutrophil and platelet datashown in Fig. 5C and D), but values did not fall belownormal ranges. No abnormal values were found in theserum chemistries. Histopathology of the animals necrop-sied on day 11 (8 days after last injection) showed micro-scopic changes in hematopoietic organs (thymus,mandibular and mesenteric lymph nodes, spleen, andbone marrow), gastrointestinal organs (stomach, duode-num, jejunum, ileum, cecum, colon, and rectum), femalereproductive organs (ovary, uterus, and vagina), and at theinjection site. These changes ranged from minimal tomoderate and were fully reversed at the end of the recoveryperiod (day 32) in all tissues, except in the thymus andgastrointestinal tract, which were trending towards fullrecovery at this later timepoint.

At the 2 � 1.92 mg SN-38/kg dose level of the conjugate,there was 1 death arising from gastrointestinal complica-tions and bone marrow suppression, and other animalswithin this group showed similar, but more severe adverseevents than the 2 � 0.96 mg/kg group. These data indicatethat dose-limiting toxicities were identical to that of irino-tecan; namely, intestinal and hematologic. Thus, the MTDfor hRS7-CL2A-SN-38 lies between 2 � 0.96 and 1.92 mgSN-38/kg, which represents a human equivalent dose of2 � 0.3 to 0.6 mg/kg SN-38.

Discussion

Trop-2 is a protein expressed on many epithelial tumors,including lung, breast, colorectal, pancreas, prostate, andovarian cancers, making it a potentially important target fordelivering cytotoxic agents (7–9, 11, 13). The RS7 antibodyinternalizes when bound to Trop-2 (18), which enablesdirect intracellular delivery of cytotoxics.

Conjugation of chemotherapeutic drugs to antibodieshas been explored for over 30 years (42, 43). Because asubstantial portion of an ADC is not processed by thetumor, but by normal tissues, there is a risk that theseagents will be too toxic to normal organ systems before

reaching the therapeutic level in tumors. As with anytherapeutic, the therapeutic window is a key factor deter-mining the potential of an ADC, and thus rather thanexamining "ultratoxic" drugs, we chose SN-38 as the drugcomponent of the Trop-2-targeted ADC.

SN-38 is a potent topoisomerase-I inhibitor, with IC50

values in the nanomolar range in several cell lines. It is theactive form of the prodrug, irinotecan, that is used for thetreatment of colorectal cancer, and which also has activityin lung, breast, and brain cancers. We reasoned that adirectly targeted SN-38, in the form of an ADC, wouldbe a significantly improved therapeutic over CPT-11, byovercoming the latter’s low and patient-variable bioconver-sion to active SN-38 (26).

The Phe-Lys peptide inserted in the original CL2 deriva-tive allowed for possible cleavage via cathepsin B (44). Inan effort to simplify the synthetic process, in CL2A, phe-nylalanine was eliminated, and thus the cathepsin B clea-vage site was removed. Interestingly, this product had abetter-defined chromatographic profile compared to thebroad profile obtained with CL2 (not shown), but moreimportantly, this change had no impact on the conjugate’sbinding, stability, or potency in side-by-side testing. Thesedata suggest that SN-38 in CL2 was released from theconjugate primarily by the cleavage at the pH-sensitivebenzyl carbonate bond to SN-38’s lactone ring and notthe cathepsin B cleavage site (Fig. 1A).

In vitro cytotoxicity of hRS7 ADC against a range of solidtumor cell lines consistently had IC50 values in the nmol/Lrange. However, cells exposed to free SN-38 demonstrateda lower IC50 value compared to the ADC. This disparitybetween free and conjugated SN-38 was also reported forENZ-2208 (28, 29) and NK012 (27). ENZ-2208 utilizes abranched PEG to link about 3.5 to 4 molecules of SN-38per PEG, whereas NK012 is a micelle nanoparticle contain-ing 20% SN-38 by weight. With our ADC, this disparity(i.e., ratio of potency with free vs. conjugated SN-38)decreased as the Trop-2 expression levels increased inthe tumor cells, suggesting an advantage to targeted deliv-ery of the drug. In terms of in vitro serum stability, both theCL2- and CL2A-SN-38 forms of hRS7-SN-38 yielded a t/1/2of �20 hours, which is in contrast to the short t/1/2 of 12.3minutes reported for ENZ-2208 (29), but similar to the57% release of SN-38 from NK012 under physiologicalconditions after 24 hours (27).

Treatment of tumor-bearing mice with hRS7-SN-38(either with CL2-SN-38 or CL2A-SN-38) significantlyinhibited tumor growth in 5 different tumor models. In4 of them, tumor regressions were observed, and in the caseof Calu-3, all mice receiving the highest dose of hRS7-SN-38 were tumor-free at the conclusion of study. Unlike inhumans, irinotecan is very efficiently converted to SN-38by a plasma esterase in mice, with a greater than 50%conversion rate (38), and yielding higher efficacy in micethan in humans (39–41). When irinotecan was adminis-tered at 10-fold higher or equivalent SN-38 levels, hRS7-SN-38 was significantly better in controlling tumor growth.Only when irinotecan was administered at its MTD of

Cardillo et al.

Clin Cancer Res; 17(10) May 15, 2011 Clinical Cancer Research3166

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 11: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

24 mg/kg q2dx5 (37.5-fold more SN-38) did it equal theeffectiveness of hRS7-SN-38. In patients, we would expectthis advantage to favor hRS7-CL2A-SN-38 even more,because the bioconversion of irinotecan would be substan-tially lower.We also showed in some antigen-expressing cell lines,

such as SK-MES-1, that using an antigen-binding ADC doesnot guarantee better therapeutic responses than a nonbind-ing, irrelevant conjugate. This is not an unusual or unex-pected finding. Indeed, the nonbinding SN-38 conjugatesmentioned earlier enhance therapeutic activity when com-pared to irinotecan, and so an irrelevant IgG-SN-38 con-jugate is expected to have some activity. This is related tothe fact that tumors have immature, leaky vessels that allowthe passage of macromolecules better than normal tissues(45).With our conjugate, 50%of the SN-38will be releasedin �13 hours when the pH is lowered to a level mimickinglysosomal levels (e.g., pH 5.3 at 37�C; data not shown),whereas at the neutral pH of serum, the release rate isreduced nearly 2-fold. If an irrelevant conjugate enters anacidic tumor microenvironment, it is expected to releasesome SN-38 locally. Other factors, such as tumor physiol-ogy and innate sensitivities to the drug, will also play a rolein defining this "baseline" activity. However, a specificconjugate with a longer residence time should haveenhanced potency over this baseline response as long asthere is ample antigen to capture the specific antibody.Biodistribution studies in the SK-MES-1 model alsoshowed that if tumor antigen becomes saturated as aconsequence of successive dosing, tumor uptake of thespecific conjugate is reduced, which yields therapeuticresults similar to that found with an irrelevant conjugate.Although it is challenging to make direct comparisons

between our ADC and the published reports of other SN-38delivery agents, some general observations can be made.In our therapy studies, the highest individual dose was0.4 mg/kg of SN-38. In the Calu-3 model, only 4 injectionswere given for a total cumulative dose of 1.6 mg/kg SN-38or 32 mg SN-38 in a 20 gmouse. Multiple studies with ENZ-2208 were done using its MTD of 10 mg/kg � 5 (28, 31),and preclinical studies with NK012 involved its MTD of30mg/kg� 3 (27). Thus, significant antitumor effects wereobtained with hRS7-SN-38 at 30-fold and 55-fold less SN-38 equivalents than the reported doses in ENZ-2208 andNK012, respectively. Even with 10-fold less hRS7 ADC(0.04 mg/kg), significant antitumor effects were observed,whereas lower doses of ENZ-2208 were not presented, andwhen the NK012 dose was lowered 4-fold to 7.5 mg/kg,efficacy was lost (27). Normal mice showed no acutetoxicity with a cumulative dose over 1 week of 24 mg/kgSN-38 (1,500 mg/kg of the conjugate), indicating that theMTD was higher. Thus, tumor-bearing animals were effec-tively treated with 7.5- to 15-fold lower amounts of SN-38equivalents.As a topoisomerase-I inhibitor, SN-38 induces significant

damage to a cell’s DNA, with upregulation of p53 andp21WAF1/Cip1 resulting in caspase activation and cleavage ofPARP (34–37). When we exposed BxPC-3 and Calu-3 cells

to our ADC, both p53 and p21WAF1/Cip1 were upregulatedabove basal levels. In addition, PARP cleavage was alsoevident in both cell lines, confirming an apoptotic event inthese cells. Of interest was the higher upregulation ofp21WAF1/Cip1 in BxPC-3 and Calu-3 relative to p53 byboth free SN-38 and our hRS7-SN-38. This may be indi-cative of the mutational status of p53 in these 2 cell lines(46–48) and the use of a p53-independent pathway forp21WAF1/Cip1-mediated apoptosis (49).

An interesting observation was the early upregulation ofp53 in both BxPC-3 and Calu-3 at 24 hours mediated bythe hRS7-ADC relative to free SN-38. Even the naked hRS7IgG could upregulate p53 in these cell lines, although onlyafter a 48-hour exposure. Trop-2 overexpression and cross-linking by antibodies has been linked to several MAPK-related signaling events (11), as well as intracellular cal-cium release (5). While binding of hRS7 was not sufficientto induce apoptosis in BxPC-3 and Calu-3, as evidenced bythe lack of PARP cleavage, it may be enough to prime a cell,such that the inclusion of SN-38 conjugated to hRS7 maylead to a greater effect on tumor growth inhibition. Studiesare currently underway to understand which pathways areinvolved with hRS7-delivery of SN-38 and how they maydiffer from free SN-38, and what effect p53 status may playin this signaling.

Biodistribution studies revealed the hRS7-CL2A-SN-38had similar tumor uptake as the parental hRS7 IgG, butcleared substantially faster with 2-fold higher hepaticuptake, which may be due to the hydrophobicity of SN-38. With the ADC being cleared through the liver, hepaticand gastrointestinal toxicities were expected to be doselimiting. Although mice had evidence of increased hepatictransaminases, gastrointestinal toxicity was mild at best,with only transient loss in weight and no abnormalitiesnoted upon histopathologic examination. Interestingly, nohematological toxicity was noted. However, monkeysshowed an identical toxicity profile as expected for irino-tecan, with gastrointestinal and hematological toxicitybeing dose-limiting.

Because Trop-2 recognized by hRS7 is not expressed inmice, it was critically important to perform toxicity studiesin monkeys that have a similar tissue expression ofTrop-2 as humans. Monkeys tolerated 0.96 mg/kg/dose(�12 mg/m2) with mild and reversible toxicity, whichextrapolates to a human dose of �0.3 mg/kg/dose(�11 mg/m2). In a Phase I clinical trial of NK012, patientswith solid tumors tolerated 28 mg/m2 of SN-38 every3 weeks with Grade 4 neutropenia as dose-limiting toxicity(DLT; ref. 30). Similarly, Phase I clinical trials with ENZ-2208 revealed dose-limiting febrile neutropenia, with arecommendation to administer 10 mg/m2 every 3 weeksor 16 mg/m2 if patients were administered G-CSF (50, 51).Becausemonkeys tolerated a cumulative human equivalentdose of 22 mg/m2, it is possible that even though hRS7binds to a number of normal tissues, the MTD for a singletreatment of the hRS7 ADC could be similar to that of theother nontargeting SN-38 agents. Indeed, the specificity ofthe anti–Trop-2 antibody did not appear to play a role in

Anti-TROP-2-SN-38 Conjugate in Cancer Therapy

www.aacrjournals.org Clin Cancer Res; 17(10) May 15, 2011 3167

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 12: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

defining the DLT, because the toxicity profile was similarto that of irinotecan. More importantly, if antitumoractivity can be achieved in humans as in mice thatresponded with human equivalent dose of just at0.03 mg SN-38 equivalents/kg/dose, then significant anti-tumor responses could be realized clinically.

In conclusion, toxicology studies in monkeys, combinedwith in vivo human cancer xenograft models in mice, haveindicated that this ADC targeting Trop-2 is an effectivetherapeutic in several tumors of different epithelial origin,supporting future clinical testing.

Disclosure of Potential Conflicts of Interest

T.M. Cardillo, S.V. Govindan, P. Trisal, and D.M. Goldenberg, employ-ment and other financial relationships with Immunomedics, Inc., includingpatent inventions. R.M. Sharkey has no financial disclosures.

Acknowledgments

We thank Dr. Sung-Ju Moon, Fatma Tat, and Agatha Sheerin for con-tributions to synthetic and conjugation chemistries, Anju Nair, MariaZalath, Lou Osorio, and Ashraf Gomaa for their assistance with the animalstudies, and Roberto Arrojo for technical assistance in performing the in vitrostudies.

Grant Support

The study was supported in part by a grant from the National CancerInstitute of the NIH (CA114802-02; PI: S.V. Govindan).

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received November 3, 2010; revised December 22, 2010; acceptedFebruary 4, 2011; published OnlineFirst March 3, 2011.

References1. Lipinski M, Parks DR, Rouse RV, Herzenberg LA. Human trophoblast

cell-surface antigens defined by monoclonal antibodies. Proc NatlAcad Sci U S A 1981;78:5147–50.

2. Stein R, Chen S, Sharkey RM, Goldenberg DM. Murine monoclonalantibodies raised against human non-small cell carcinoma of the lung:specificity and tumor targeting. Cancer Res 1990;50:1330–6.

3. Alberti S, Miotti S, Stella M, Klein CE, Fornaro M, Menard S, et al.Biochemical characterization of Trop-2, a cell surface moleculeexpressed by human carcinomas: formal proof that the monoclonalantibodies T16 and MOv-16 recognize Trop-2. Hybridoma 1992;11:539–45.

4. Stein R, Basu A, Chen S, Shih LB, Goldenberg DM. Specificity andproperties of MAb RS7–3G11 and the antigen defined by this pan-carcinoma monoclonal antibody. Int J Cancer 1993;55:938–46.

5. Ripani E, Sacchetti A, Corda D, Alberti S. Human Trop-2 is atumor-associated calcium signal transducer. Int J Cancer 1998;76:671–6.

6. Shimada A, Kano J, Ishiyama T, Okubo C, Iijima T, Morishita Y, et al.Establishment of an immortalized cell line from a precancerous lesionof lung adenocarcinoma, and genes highly expressed in the earlystages of lung adenocarcinoma development. Cancer Sci 2005;96:668–75.

7. Ohmachi T, Tanaka F, Mimori K, Inoue H, Yanaga K, Mori M. Clinicalsignificance of TROP2 expression in colorectal cancer. Clin CancerRes 2006;12:3057–63.

8. Wang J, Day R, Dong Y,Weintraub SJ, Michel L. Identification of Trop-2 as an oncogene and an attractive therapeutic target in coloncancers. Mol Cancer Ther 2008;7:280–5.

9. Fong D, Moser P, Krammel C, Gostner JM, Margreiter R, Mitterer M,et al. High expression of TROP2 correlates with poor prognosis inpancreatic cancer. Br J Cancer 2008;99:1290–5.

10. Fong D, Spizzo G, Gostner JM, Gastl G, Moser P, Krammel C, et al.TROP2: a novel prognostic marker in squamous cell carcinoma of theoral cavity. Mod Pathol 2008;21:186–91.

11. Cubas R, Li M, Chen C, Yao Q. Trop2: a possible therapeutic target forlate stage epithelial carcinomas. Biochim Biophys Acta 2009;1796:309–14.

12. Fang YJ, Lu ZH, Wang GQ, Pan ZZ, Zhou ZW, Yun JP, et al. Elevatedexpressions of MMP7, TROP2, and survivin are associated withsurvival, disease recurrence, and liver metastasis of colon cancer.Int J Colorectal Dis 2009;24:875–84.

13. Kobayashi H, Minami Y, Anami Y, Kondou Y, Iijima T, Kano J, et al.Expression of the GA733 gene family and its relationship to prognosisin pulmonary adenocarcinoma. Virchows Arch 2010;457:69–76.

14. Trerotola M, Guerra E, Alberti S. Letter to the editor: efficacy andsafety of anti-Trop antibodies. Biochim Biophys Acta 2010;1805:119–20.

15. Cubas R, Zhang S, Li M, Chen C, YaoQ. Trop2 expression contributesto tumor pathogenesis by activating the ERK MAPK pathway. MolCancer 2010;9:253.

16. Stein R, Basu A, Goldenberg DM, Lloyd KO, Mattes MJ. Character-ization of cluster 13: the epithelial/carcinoma antigen recognized byMAb RS7. Int J Cancer Suppl 1994;8:98–102.

17. Basu A, Goldenberg DM, Stein R. The epithelial/carcinoma antigenEGP-1, recognized by monoclonal antibody RS7–3G11, is phos-phorylated on serine 303. Int J Cancer 1995;62:472–9.

18. Shih LB, Xuan H, Aninipot R, Stein R, Goldenberg DM. In vitro and invivo reactivity of an internalizing antibody, RS7, with human breastcancer. Cancer Res 1995;55:5857s–63s.

19. Stein R, Chen S, Haim S, Goldenberg DM. Advantage of yttrium-90-labeled over iodine-131-labeled monoclonal antibodies in the treat-ment of a human lung carcinoma xenograft. Cancer 1997;80:2636–41.

20. Govindan SV, Stein R, Qu Z, Chen S, Andrews P, Ma H, et al.Preclinical therapy of breast cancer with a radioiodinated humanizedanti-EGP-1 monoclonal antibody: advantage of a residualizing iodineradiolabel. Breast Cancer Res Treat 2004;84:173–82.

21. Varughese J, Cocco E, Bellone S, Bellone M, Todeschini P, Carrara L,et al. High-grade, chemotherapy-resistant primary ovarian carcinomacell lines overexpress human trophoblast cell-surface marker (Trop-2)and are highly sensitive to immunotherapy with hRS7, a humanizedmonoclonal anti-Trop-2 antibody. Gynecol Oncol 2011 Mar 29. [Epubahead of print]

22. Moon SJ, Govindan SV, Cardillo TM, D’Souza CA, Hansen HJ, Gold-enberg DM. Antibody conjugates of 7-ethyl-10-hydroxycamptothecin(SN-38) for targeted cancer chemotherapy. J Med Chem 2008;51:6916–26.

23. Govindan SV, Cardillo TM, Moon SJ, Hansen HJ, Goldenberg DM.CEACAM5-targeted therapy of human colonic and pancreatic cancerxenografts with potent labetuzumab-SN-38 immunoconjugates. ClinCancer Res 2009;15:6052–61.

24. Garcia-Carbonero R, Supko JG. Current perspectives on the clinicalexperience, pharmacology, and continued development of the camp-tothecins. Clin Cancer Res 2002;8:641–61.

25. O’Leary J, Muggia FM. Camptothecins: a review of their develop-ment and schedules of administration. Eur J Cancer 1998;34:1500–8.

26. Mathijssen RH, van Alphen RJ, Verweij J, LoosWJ, Nooter K, Stoter G,et al. Clinical pharmacokinetics and metabolism of irinotecan (CPT-11). Clin Cancer Res 2001;7:2182–94.

27. Koizumi F, Kitagawa M, Negishi T, Onda T, Matsumoto S, HamaguchiT, et al. Novel SN-38-incorporating polymeric micelles, NK012, era-dicate vascular endothelial growth factor-secreting bulky tumors.Cancer Res 2006;66:10048–56.

Cardillo et al.

Clin Cancer Res; 17(10) May 15, 2011 Clinical Cancer Research3168

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 13: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

28. Sapra P, Zhao H, Mehlig M, Malaby J, Kraft P, Longley C, et al. Noveldelivery of SN38 markedly inhibits tumor growth in xenografts, includ-ing a camptothecin-11-refractory model. Clin Cancer Res 2008;14:1888–96.

29. Zhao H, Rubio B, Sapra P, Wu D, Reddy P, Sai P, et al. Novel prodrugsof SN38 using multiarm poly(ethylene glycol) linkers. Bioconjug Chem2008;19:849–59.

30. Hamaguchi T, Doi T, Eguchi-Nakajima T, Kato K, Yamada Y, ShimadaY, et al. Phase I Study of NK012, a novel SN-38-incorporating micellarnanoparticle, in adult patients with solid tumors. Clin Cancer Res2010;16:5058–66.

31. Pastorino F, Loi M, Sapra P, Becherini P, Cilli M, Emionite L, et al.Tumor regression and curability of preclinical neuroblastoma modelsby PEGylated SN38 (ENZ-2208), a novel topoisomerase I inhibitor.Clin Cancer Res 2010;16:4809–21.

32. Sumitomo M, Koizumi F, Asano T, Horiguchi A, Ito K, Asano T, et al.Novel SN-38-incorporated polymeric micelle, NK012, strongly sup-presses renal cancer progression. Cancer Res 2008;68:1631–5.

33. Nagano T, Yasunaga M, Goto K, Kenmotsu H, Koga Y, Kuroda J-I,et al. Antitumor activity of NK012 combined with cisplatin againstsmall cell lung cancer and intestinal mucosal changes in tumor-bearing mouse after treatment. Clin Cancer Res 2009;15:4348–55.

34. Cusack JC Jr, Liu R, Houston M, Abendroth K, Elliott PJ, Adams J,et al. Enhanced chemosensitivity to CPT-11 with proteasome inhibitorPS-341: implications for systemic nuclear factor-kB inhibition. CancerRes 2001;61:3535–40.

35. Liu Y, Xing H, Weng D, Song X, Qin X, Xia X, et al. Inhibition of Aktsignaling by SN-38 induces apoptosis in cervical cancer. Cancer Lett2009;274:47–53.

36. Lagadec P, Griessinger E, Nawrot MP, Fenouille N, Colosetti P, ImbertV, et al. Pharmacological targeting of NF-kB potentiates the effect ofthe topoisomerase inhibitor CPT-11 on colon cancer cells. Br JCancer 2008;98:335–44.

37. Whitacre CM, Zborowska E,Willson JKV, Berger NA. Detection of poly(ADP-ribose) polymerase cleavage in response to treatment withtopoisomerase I inhibitors: a potential surrogate end point to assesstreatment effectiveness. Clin Cancer Res 1999;5:665–72.

38. Morton CL, Wierdl M, Oliver L, Ma MK, Danks MK, Stewart CF, et al.Activation of CPT-11 in mice: identification and analysis of a highlyeffective plasma esterase. Cancer Res 2000;60:4206–10.

39. Furman WL, Stewart CF, Poquette CA, Pratt CB, Santana VM, Zam-boni WC, et al. Direct translation of a protracted irinotecan schedulefrom a xenograft model to a phase I trial in children. J Clin Oncol1999;17:1815–24.

40. Zamboni WC, Houghton PJ, Thompson J, Cheshire PJ, Hanna SK,Richmond LB, et al. Altered irinotecan and SN-38 disposition afterintravenous and oral administration of irinotecan in mice bearinghuman neuroblastoma xenografts. Clin Cancer Res 1998;4:455–62.

41. Zamboni WC, Stewart CF, Cheshire PJ, Richmond LB, Hanna SK, LuoX, et al. Studies of the efficacy and pharmacology of irinotecan againsthuman colon tumor xenograft models. Clin Cancer Res 1998;4:743–53.

42. Teicher BA. Antibody-drug conjugate targets. Curr Cancer DrugTargets 2009;9:982–1004.

43. Alley SC, Okeley NM, Senter PD. Antibody-drug conjugates: targeteddrug delivery for cancer. Curr Opin Chem Biol 2010;14:529–37.

44. Dubowchik GM, Firestone RA, Padilla L, Willner D, Hofstead SJ,Mosure K, et al. Cathepsin B-labile dipeptide linkers for lysosomalrelease of doxorubicin from internalizing immunoconjugates: modelstudies of enzymatic drug release and antigen-specific in vitro antic-ancer activity. Bioconjug Chem 2002;13:855–69.

45. Jain RK. Barriers to drug delivery in solid tumors. Sci Am 1994;271:58–61.

46. DiGiuseppe JA, Redston MS, Yeo CJ, Kern SE, Hruban RH. P53-independent expression of the cyclin-dependent kinase inhibitor p21in pancreatic carcinoma. Am J Pathol 1995;4:884–8.

47. Barton CM, Staddon SL, Hughes CM, Hall PA, O’Sullivan C, Kl€oppelG, et al. Abnormalities of the p53 tumour suppressor gene in humanpancreatic cancer. Br J Cancer 1991;64:1076–82.

48. Pratesi G, Perego P, Polizzi D, Righetti SC, Supino R, Caserini C, et al.A novel charged trinuclear platinum complex effective against cis-platin-resistant tumours: hypersensitivity of p53-mutant humantumour xenografts. Br J Cancer 1999;80:1912–9.

49. McDonald AC, Brown R. Induction of p53-dependent and p53-inde-pendent cellular responses by topoisomerase I inhibitors. Br J Cancer1998;78:745–51.

50. Kurzrock R, Wheler J, Hong DS, Guo Z, Mulcahy MF, Benson AB III,et al. Phase 1, first-in-human, dose-escalation study of ENZ-2208, anovel anticancer agent, in patients with advanced malignancies[abstract]. AACR-NCI-EORTC International Conference on MolecularTargets and Cancer Therapeutics; 2009 Nov 15–19; Boston, MA;Poster No C216.

51. Patnaik A, Papadopoulos KP, Beeram M, Kee D, Tolcher AW, SchaafLJ, et al. ENZ-2208, a novel anticancer agent, in patients withadvanced malignancies: a phase 1 dose-escalation study [abstract].AACR-NCI-EORTC International Conference on Molecular Targetsand Cancer Therapeutics. 2009 Nov 15–19; Boston, MA. PosterNo. C221.

Anti-TROP-2-SN-38 Conjugate in Cancer Therapy

www.aacrjournals.org Clin Cancer Res; 17(10) May 15, 2011 3169

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939

Page 14: Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective … · ments in 2 dimensions using calipers, with volumes defined as: L w2/2, where L is the longest dimension of the tumor

2011;17:3157-3169. Published OnlineFirst March 3, 2011.Clin Cancer Res   Thomas M. Cardillo, Serengulam V. Govindan, Robert M. Sharkey, et al.   in Human Cancer Xenograft Models and MonkeysTreatment of Diverse Epithelial Cancers: Preclinical Studies Humanized Anti-Trop-2 IgG-SN-38 Conjugate for Effective

  Updated version

  10.1158/1078-0432.CCR-10-2939doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2011/05/18/1078-0432.CCR-10-2939.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/17/10/3157.full#ref-list-1

This article cites 48 articles, 20 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/17/10/3157.full#related-urls

This article has been cited by 16 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  SubscriptionsReprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://clincancerres.aacrjournals.org/content/17/10/3157To request permission to re-use all or part of this article, use this link

Research. on April 2, 2020. © 2011 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst March 3, 2011; DOI: 10.1158/1078-0432.CCR-10-2939