theeffectofdifferentlinkersontargetcellcatabolismand ......maytansinoid conjugates hans k. erickson...

11
Preclinical Development The Effect of Different Linkers on Target Cell Catabolism and Pharmacokinetics/Pharmacodynamics of Trastuzumab Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 , Elaine Mai 4 , Holly A. Johnson 1 , Bert Gunter 5 , Charlene A. Audette 1 , Manish Gupta 3 , Jan Pinkas 1 , and Jay Tibbitts 3 Abstract Trastuzumab emtansine (T-DM1) is an antibody–drug conjugate consisting of the anti-HER2 antibody trastuzumab linked via a nonreducible thioether linker to the maytansinoid antitubulin agent DM1. T-DM1 has shown favorable safety and efficacy in patients with HER2-positive metastatic breast cancer. In previous animal studies, T-DM1 exhibited better pharmacokinetics (PK) and slightly more efficacy than several disulfide-linked versions. The efficacy findings are unique, as other disulfide-linked antibody–drug conjugates (ADC) have shown greater efficacy than thioether-linked designs. To explore this further, the in vitro and in vivo activity, PK, and target cell activation of T-DM1 and the disulfide-linked T-SPP-DM1 were examined. Both ADCs showed high in vitro potency, with T-DM1 displaying greater potency in two of four breast cancer cell lines. In vitro target cell processing of T-DM1 and T-SPP-DM1 produced lysine-N e -MCC-DM1, and lysine- N e -SPP-DM1 and DM1, respectively; in vivo studies confirmed these results. The in vitro processing rates for the two conjugate to their respective catabolites were similar. In vivo, the potencies of the conjugates were similar, and T-SPP-DM1 had a faster plasma clearance than T-DM1. Slower T-DM1 clearance translated to higher overall tumor concentrations (conjugate plus catabolites), but unexpectedly, similar levels of tumor catabolite. These results indicate that, although the ADC linker can have clear impact on the PK and the chemical nature of the catabolites formed, both linkers seem to offer the same payload delivery to the tumor. Mol Cancer Ther; 11(5); 1133–42. Ó2012 AACR. Introduction An increasing number of antibody–drug conjugates (ADC) are entering clinical trials for the treatment of cancer (1). One of the most advanced and promising, trastuzumab emtansine (T-DM1) has shown favorable efficacy and safety in clinical trials for the treatment of patients with HER2-positive metastatic breast cancer (2– 4). T-DM1 is an ADC that contains the humanized anti- HER2 immunoglobulin G (IgG)1 trastuzumab linked to the maytansinoid antitubulin agent DM1 via a thioether bond. T-DM1 retains the multiple mechanisms of action (MOA) described for trastuzumab, (5) and conjugation with DM1 confers the potential for additional cell killing activity via the delivery of potent antimitotic maytansi- noid catabolites to targeted cancer cells. The efficacy of T-DM1 in patients who progressed on HER2-directed therapies underscores the importance of this additional activity (2–4). T-DM1 is distinct from other clinically tested maytan- sinoid containing ADCs by virtue of its thioether linker. All other such ADCs in clinical testing use disulfide-based linkers (1). The thioether linker is considered more stable because it resists chemical or enzymatic cleavage in bio- logical systems, in contrast to disulfide-based linkers that may be cleaved following thiol-disulfide exchange reactions. T-DM1 was found to be slightly more active in mouse models than T-SPP-DM1 and several other disul- fide-linked ADCs (6). This contrasts with similar studies of other ADCs that led to the selection of disulfide-based linkers for clinical development (7–9). The factors determining the efficacy of an ADC include pharmacokinetics (PK), tumor penetration and accumu- lation, target binding and cellular uptake, release of active catabolic products, and potency of the catabolic products. Previous studies have shown that the plasma concentra- tions of thioether-linked ADCs decrease more slowly than those of disulfide-linked ADCs, likely due to greater linker stability (6, 10); which may account for differences in the efficacy between T-DM1 and disulfide-linked analogues. It has also been shown that other aspects of the MOA Authors' Afliations: 1 ImmunoGen, Inc., Waltham, MA; and 2 Research Oncology, 3 Pharmacokinetics and Pharmacodynamics, 4 Assay and Auto- mation Technologies, and 5 Nonclinical Biostatistics, Genentech, Inc., South San Francisco, CA. Note: Supplementary material for this article is available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Author: Jay Tibbitts, Genentech, 1 DNA Way, MS463A, South San Francisco, CA 94080. Phone: 650-467-3194; Fax: 650-742- 5234; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-11-0727 Ó2012 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 1133 on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727

Upload: others

Post on 19-Jun-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: TheEffectofDifferentLinkersonTargetCellCatabolismand ......Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 ,

Preclinical Development

The Effect of Different Linkers on Target Cell Catabolism andPharmacokinetics/Pharmacodynamics of TrastuzumabMaytansinoid Conjugates

Hans K. Erickson1, Gail D. Lewis Phillips2, Douglas D. Leipold3, Carmela A. Provenzano1, Elaine Mai4,Holly A. Johnson1, Bert Gunter5, Charlene A. Audette1, Manish Gupta3, Jan Pinkas1, and Jay Tibbitts3

AbstractTrastuzumab emtansine (T-DM1) is an antibody–drug conjugate consisting of the anti-HER2 antibody

trastuzumab linked via a nonreducible thioether linker to themaytansinoid antitubulin agentDM1. T-DM1has

shown favorable safety and efficacy in patients with HER2-positive metastatic breast cancer. In previous

animal studies, T-DM1 exhibited better pharmacokinetics (PK) and slightly more efficacy than several

disulfide-linked versions. The efficacyfindings are unique, as other disulfide-linked antibody–drug conjugates

(ADC) have showngreater efficacy than thioether-linkeddesigns. To explore this further, the in vitro and in vivo

activity, PK, and target cell activation of T-DM1 and the disulfide-linked T-SPP-DM1 were examined. Both

ADCs showed high in vitro potency, with T-DM1 displaying greater potency in two of four breast cancer cell

lines. In vitro target cell processing of T-DM1 and T-SPP-DM1 produced lysine-Ne-MCC-DM1, and lysine-

Ne-SPP-DM1 and DM1, respectively; in vivo studies confirmed these results. The in vitro processing rates for

the two conjugate to their respective catabolites were similar. In vivo, the potencies of the conjugates were

similar, and T-SPP-DM1 had a faster plasma clearance than T-DM1. Slower T-DM1 clearance translated to

higher overall tumor concentrations (conjugate plus catabolites), but unexpectedly, similar levels of tumor

catabolite. These results indicate that, although the ADC linker can have clear impact on the PK and the

chemical nature of the catabolites formed, both linkers seem to offer the same payload delivery to the tumor.

Mol Cancer Ther; 11(5); 1133–42. �2012 AACR.

IntroductionAn increasing number of antibody–drug conjugates

(ADC) are entering clinical trials for the treatment ofcancer (1). One of the most advanced and promising,trastuzumab emtansine (T-DM1) has shown favorableefficacy and safety in clinical trials for the treatment ofpatients with HER2-positive metastatic breast cancer (2–4). T-DM1 is an ADC that contains the humanized anti-HER2 immunoglobulin G (IgG)1 trastuzumab linked tothe maytansinoid antitubulin agent DM1 via a thioetherbond. T-DM1 retains the multiple mechanisms of action(MOA) described for trastuzumab, (5) and conjugationwith DM1 confers the potential for additional cell killingactivity via the delivery of potent antimitotic maytansi-

noid catabolites to targeted cancer cells. The efficacy ofT-DM1 in patients who progressed on HER2-directedtherapies underscores the importance of this additionalactivity (2–4).

T-DM1 is distinct from other clinically tested maytan-sinoid containing ADCs by virtue of its thioether linker.All other suchADCs in clinical testing use disulfide-basedlinkers (1). The thioether linker is considered more stablebecause it resists chemical or enzymatic cleavage in bio-logical systems, in contrast to disulfide-based linkersthat may be cleaved following thiol-disulfide exchangereactions. T-DM1 was found to be slightly more active inmouse models than T-SPP-DM1 and several other disul-fide-linked ADCs (6). This contrasts with similar studiesof other ADCs that led to the selection of disulfide-basedlinkers for clinical development (7–9).

The factors determining the efficacy of an ADC includepharmacokinetics (PK), tumor penetration and accumu-lation, target binding and cellular uptake, release of activecatabolic products, and potency of the catabolic products.Previous studies have shown that the plasma concentra-tions of thioether-linkedADCs decreasemore slowly thanthoseofdisulfide-linkedADCs, likelydue togreater linkerstability (6, 10); which may account for differences in theefficacy between T-DM1 and disulfide-linked analogues.It has also been shown that other aspects of the MOA

Authors' Affiliations: 1ImmunoGen, Inc., Waltham, MA; and 2ResearchOncology, 3Pharmacokinetics and Pharmacodynamics, 4Assay and Auto-mation Technologies, and 5Nonclinical Biostatistics, Genentech, Inc.,South San Francisco, CA.

Note: Supplementary material for this article is available at MolecularCancer Therapeutics Online (http://mct.aacrjournals.org/).

Corresponding Author: Jay Tibbitts, Genentech, 1 DNA Way, MS463A,South San Francisco, CA 94080. Phone: 650-467-3194; Fax: 650-742-5234; E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-11-0727

�2012 American Association for Cancer Research.

MolecularCancer

Therapeutics

www.aacrjournals.org 1133

on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727

Page 2: TheEffectofDifferentLinkersonTargetCellCatabolismand ......Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 ,

(i.e., target binding, cellular uptake, and release of cata-bolic products) may be similar for disulfide-linked andthioether-linked ADCs (11). To develop a better under-standing of the MOA of trastuzumab maytansinoid con-jugates and differences in the preclinical activity betweenthese ADCs, we investigated the molecular basis for theanticancer activity in in vitro and in vivo efficacy, PK, andqualitative and quantitative tumor catabolism studies.

Materials and MethodsAll experimental procedures conformed to the princi-

ples of the Guide for the Care and Use of LaboratoryAnimals and the American Physiological Society andwere approved by the Institutional Animal Care and UseCommittees of the respective laboratories.

Cell lines and reagentsThe SK-BR-3 and BT-474 breast carcinoma cell lines

were obtained from The American Type Culture Collec-tion and used within 2 months of receipt. The BT-474EEItrastuzumab-resistant breast carcinoma cell line wasderived at Genentech (6) by subculturing tumors derivedfrom a BT-474 variant line (courtesy of Dr. Jose Baselga).BT-474EEI tumors express approximately 0.25� 106HER2receptors per tumor cell and are insensitive to trastuzu-mab. MCF7-neo/HER2s are HER2-transfected cells madeat Genentech. Cells were cultured in RPMI medium sup-plemented with 10% heat-inactivated FBS. MCF7-neo/HER2, BT-474, and BT-474 in vivo-selected variant (Base-lga) were genotyped and authenticated using IlluminaGolden Gate single-nucleotide polymorphism testing.Although the parent cells of the BT-474EEI cells (BT-474Baselga variant) were genotyped as BT-474, the resultingcells (EEI) after in vivo selection have not been authenti-cated at this point. Porapak extraction cartridges wereobtained from Waters. Ultima Gold scintillation fluidwas obtained from PerkinElmer. N-ethylmaleimide(NEM) and all other chemicals were obtained fromSigma-Aldrich. The 10 mm C-18 column (0.46 � 25 cm;Vydak) was obtained from the Nest Group. The ULTRA-TURRAX T8 dispersing instrument with an S8N dispers-ing tool was obtained from IKA Works Inc. Trastuzumaband the humanized antiglycoprotein D (gD) controlantibody, 5B6, were prepared at Genentech. Antibody-[3H]maytansinoid conjugates were prepared at Immuno-Gen, Inc., as described previously (12). The ratio of linked[3H]maytansinoid per antibody molecule (MAR) foreach conjugate was as follows: T-SPP-[3H]DM1 (3.2),T-[3H]DM1 (4.0), 5B6-SPP-[3H]DM1 (3.5) [SPP ¼ N-succi-nimidyl 4-(2-pyridyldithio)pentanoate], and hu5B6-MCC-[3H]DM1 (3.6) [SMCC ¼ N-succinimidyl 4-(N-maleimido-methyl)cyclohexane-1 carboxylate]. Unlabeled antibody–maytansinoid conjugates with similar MAR values wereprepared at Genentech.

Clonogenic assaysCells were plated at low density (100 cells per well for

SK-BR-3, MCF7-neo/HER2, and BT-474EEI; 200 cells per

well for BT-474) in Ham’s F-12:DMEM (50:50) þ 10% FBSþ 2 mmol/L L-glutamine and allowed to adhere over-night. Conjugates were added the next day and the cellsincubated until colony formation was determined to bemaximal. The medium was removed and the coloniesstained with crystal violet dye (0.5% in methanol). Colo-nies were quantified with a GelCount (Oxford Optronix).

In vitro catabolism studiesA stable tritium label was incorporated into the C-20

methoxy group of theDM1used toprepare the 3H-labeledconjugates as describedpreviously (12).HER2-dependentprocessing of the 3H-labeled conjugates was investigatedin HER2-overexpressing BT-474EEI, MCF7-neo/HER2,and SK-BR-3. Cells were plated in flat bottom T-75 cellculture flasks in cell culture medium and grown to adensity of approximately 107 cells per flask. The mediumwas exchanged with 10 mL of RPMI containing 10% FBSand 20 to 40 nmol/L of T-[3H]DM1 or T-SPP-[3H]DM1.The cells were placed on ice for 2 to 3 hours or for 30minutes at 37�Cwith 6% CO2. The treated cells were thenwashed 3 times with freshmedium and incubated at 37�Cwith 6% CO2 with 10 mL fresh culture medium for 3 to 24hours. The spent medium was separated and cells wereharvested at selected time intervals and analyzed forcatabolites following acetone extraction as described pre-viously (11, 12). The precipitates from the acetone extrac-tions were solubilized and analyzed for tritium by liquidscintillation counting (LSC) to assess the level of protein-bound maytansinoid associated with the cell pellet, pre-sumed tobenonmetabolized conjugate orDM1still linkedto large protein fragments.

Maytansinoid catabolites were isolated from the medi-um by solid phase extraction followed by reversed-phasehigh-pressure liquid chromatography (HPLC). Mediasamples were applied to 6 mL porapak cartridges equil-ibrated with 3 mL acetone and then with 3 mL water. Thecartridges were washed with 4 mL water and the may-tansinoids eluted with 4 mL acetone. The eluates wereevaporated and analyzed by reversed-phase HPLC andLSCasdescribed for extracts containing the cell catabolites.

Cell volume measurementsCell volumes were calculated from average cell dia-

meters as measured by a cell viability analyzer (BeckmanCoulter). Cell volumes for the SK-BR-3, BT-474EEI, andMCF7/neo-HER2 cells were 3823, 3955, and 3648 mm3,respectively.

AnimalsFemale beige nude XID mice (Charles Rivers Labora-

tory)were used for all in vivo studies.Micewere housed ina clean barrier facility in standard rodent microisolatorcages.

In vivo efficacy studiesTo allow comparison of results obtained in vitro and

in vivo, a mouse xenograft model was selected that

Erickson et al.

Mol Cancer Ther; 11(5) May 2012 Molecular Cancer Therapeutics1134

on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727

Page 3: TheEffectofDifferentLinkersonTargetCellCatabolismand ......Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 ,

was suitable for both systems. Previously, the founder5 (Fo5) mouse allograft transplant model was used forin vivo comparisons of maytansinoid ADC efficacy (6).However, this model was not suitable for these studiesas it cannot be cultured in vitro. Thus, the BT-474EEIxenograft model, suitable for in vitro and in vivo investiga-tions, was chosen (13). BT-474EEI cells (2 � 107) wereinjected into the mammary fat pad of beige nude XIDmice, and tumors were grown to approximately 250mm3.Animals were then randomized into 9 groups (n ¼ 10 pergroup) and treated with a single i.v. bolus dose of 3to 18 mg/kg (�60–360 mg/kg based on DM1 dose) of theconjugates. Tumors were measured with caliperstwice weekly for 24 days following dosing, and thenweekly for 120 days or until tumors reached a volume of3,000 mm3. Tumor volumes were determined with theformula: Tumor volume (mm3) ¼ (longer diameter �shorter diameter)2 � 0.5.

In vivo catabolism studiesMice-bearing BT-474EEI tumors (as described above)

were randomized into 2 groups (n ¼ 15 per group)and treated with a single 300 mg/kg i.v. bolus dose ofT-[3H]DM1 or T-SPP-[3H]DM1 (doses based on DM1,equivalent to 10–12 mg/kg antibody). Three mice pergroup were sacrificed after 8 hours and 1, 2, 4, and7 days. Immediately following sacrifice, the systemiccirculation was flushed by injecting 5.0 mL PBS intothe left ventricle and draining through an incision in theinferior vena cava. Whole tumor tissues were collectedand frozen at �80�C. Two control groups of mice (n ¼3 per group) were treated with matching doses ofnontargeting 5B6-MCC-[3H]DM1 or 5B6-SPP-[3H]DM1conjugates and sacrificed after 2 days. Tumors werehomogenized and analyzed for total radioactivity bysolubilization and LSC, and maytansinoid catabolitesby HPLC and LSC as described previously. To deter-mine the amount of protein-free maytansinoid catabo-lites present in the tumors, the tumor homogenateswere extracted with organic solvent and analyzed asdescribed previously (12).

Analytic methodsAll maytansinoids were separated on an analytic C-18

column equilibrated with 20% aqueous acetonitrile(CH3CN) containing 0.025% trifluoroacetic acid andusing a linear gradient of 2% CH3CN min�1 and a flowrate of 1 mL�min�1. The effluent was collected in 6 mLpolypropylene scintillation vials (1 mL fractions). Countsper minute (CPM) of tritium associated with each frac-tion were determined by mixing each vial with 4 mLUltima Gold liquid scintillation cocktail before countingfor 5 minutes in a Tri-Carb 2900T liquid scintillationcounter (Packard BioScience).The identities of the catabolites were confirmed by

liquid chromatography mass spectrometry by methodssimilar to those described previously (11).

Pharmacokinetic studiesAnimals (n ¼ 20 per group) were administered a 3

mg/kg i.v. bolus dose of T-DM1 or T-SPP-DM1. Atselected intervals up to 42 days following dosing, bloodsamples were collected on a rotational basis, with n ¼ 4animals per time point. Blood sampleswere processed forplasma by centrifugation and plasma decanted to poly-propylene collection tubes. Samples were stored at�60�Cto �80�C until analysis.

ADC plasma concentration analysisPlasma ADC concentrations were determined with an

ELISA that measured any trastuzumab antibody contain-ing one ormore conjugatedDM1molecules. An anti-DM1monoclonal antibody was coated on ELISA plates. Cap-tured ADC was detected with biotinylated HER2 extra-cellular domain (HER2 ECD) followed by streptavidin-horseradish peroxidase. The limit of quantitation (LOQ)was 1.6 ng/mL. Total trastuzumab concentrations weredetermined as described previously (6) using an ELISAthat measured any trastuzumab antibody. In brief, HER2ECD was coated on ELISA plates. Captured total trastu-zumabwas detectedwith goat-anti-human IgG-horserad-ish peroxidase. The LOQ was 1.6 ng/mL.

Pharmacokinetic analysisThe PK analysis of plasma ADC ELISA concentration–

time data was conducted using a na€�ve pool approachwith individual data from all animals. Data were fit to a2-compartmental model with i.v. bolus input, first-order elimination, and macro rate constants (Model 8,WinNonlin Pro, v.5.2.1; Pharsight Corporation). Nominalsample collection times and dose concentrations wereused in the data analysis.

Exposure responseIn vivo response was assessed by calculating the mean

tumor volume for each animal up to day 38 followingdosing. Themean tumor volume is a sumof themeasuredtumor volumes in an individual animal from days 0 to 38,divided by the number of sampling times. Day 38 waschosen to allow for the observation of full ADC responsewithout substantial tumor regrowth. Evaluation of othertime points did not affect the results (data not shown). Toshow exposure response, the individual animal meantumor volume value for each dose group was plottedagainst the estimated plasma ADC exposure [area underthe curve (AUC)] for that dose group.

ResultsHER2-positive breast carcinoma cells display highsensitivity to T-DM1 and T-SPP-DM1

The cytotoxic potencies of T-DM1 and T-SPP-DM1 andthe matching nonbinding conjugates were compared inthe trastuzumab-sensitive breast cancer cell lines SK-BR-3and BT-474, and the trastuzumab-insensitive breast can-cer cell lines BT-474EEI and MCF7-neo/HER2 using a

Effect of Linkers on Trastuzumab Maytansinoid Conjugates

www.aacrjournals.org Mol Cancer Ther; 11(5) May 2012 1135

on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727

Page 4: TheEffectofDifferentLinkersonTargetCellCatabolismand ......Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 ,

clonogenic assay (Fig. 1). Both trastuzumab conjugateswere found to be potent inhibitors of colony formation inthe HER2-positive cell lines, whereas the nontargetingconjugates displayed little cytotoxicity. The chemicalnature of the linker had little influence on the cytotoxicityof trastuzumab–DM1 conjugates toward BT-474 (Fig. 1A)and SK-BR-3 cells (Fig. 1B), consistent with an earlierreport (6). However, the T-DM1 was more cytotoxic thanT-SPP-DM1 toward BT-474EEI (Fig. 1C) and MCF7-neo/HER2 cells (Fig. 1D).

T-DM1 and T-SPP-DM1 display similar efficacy inmice bearing trastuzumab-insensitive BT-474EEItumors

The antitumor activity associated with T-DM1 and T-SPP-DM1 inmice bearing the BT-474EEI tumors is shownin Fig. 2A. Following treatment with trastuzumab con-jugates, tumor volume decreased in proportion to dosewith maximum response at 18 mg/kg of either conjugate.Tumor regression reached its maximum approximately10 to 21 days after dosing with subsequent stasis orregrowth. The exposure-response relationship for T-DM1and T-SPP-DM1 was similar (Fig. 2B).

Pharmacokinetics of T-DM1 and T-SPP-DM1The PK of T-DM1 and T-SPP-DM1 are shown

in Fig. 3 and Supplementary Table S1. Total trastuzu-mab (Tab) PK were similar for both conjugates (Sup-plementary Table S1), indicating that conjugationwith the respective linker-DM1 did not differentiallyaffect the antibody PK behavior. T-SPP-DM1 ADCconcentrations decreased more rapidly than T-DM1,with T-SPP-DM1 exhibiting a faster clearance (40.1 �1.87 vs. 18.9 � 0.29 mL/d/kg) and shorter terminalhalf-life (2.69 � 0.087 vs. 5.72 � 0.150 days), consistentwith a previous report (6). Central compartment vol-ume of distribution was similar between the conju-gates, as expected, based on the general similarity intheir structure.

Characterization of maytansinoid catabolitesfollowing in vitro exposure of breast cancer cellsto T-DM1 and T-SPP-DM1

The radiochromatograms in Fig. 4A show the cata-bolites of T-[3H]DM1 and T-SPP-[3H]DM1 followingtreatment of the BT-474EEI cells. The sole T-[3H]DM1catabolite at each time point was found to be lysine-Ne-MCC-[3H]DM1. The corresponding lysine-Ne-SPP-

Figure 1. In vitro clonogenic survival assays of trastuzumab–DM1 conjugates in HER2-overepressing breast cancer cell lines [IC50 values (ng/mL)]. Colonynumberwasassessedafter 7 (MCF7-neo/HER2) to21 (BT-474) daysof exposure toT-DM1,T-SPP-DM1,or control ADCs.A, T-SPP-DM1¼4.0 , T-DM1¼5.0.B, T-SPP-DM1 ¼ 1.8, T-DM1 ¼ 1.1. C, T-SPP-DM1 ¼ 30.0, T-DM1 ¼ 6.0. D, T-SPP-DM1 ¼ 10.0, T-DM1 ¼ 3.0.

Erickson et al.

Mol Cancer Ther; 11(5) May 2012 Molecular Cancer Therapeutics1136

on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727

Page 5: TheEffectofDifferentLinkersonTargetCellCatabolismand ......Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 ,

[3H]DM1 catabolite of T-SPP-[3H]DM1 was observedalong with a similarly abundant DM1 catabolite. TheDM1 catabolite was alkylated at its free sulfhydrylgroup by NEM in the suspension buffer before extrac-tion and thus was detected as its NEM adduct in theradiograms. No maytansinoid catabolites (<0.1 pmol/106 cells) were detected in the acetone extracts of thespent medium samples indicating that little, if any,efflux of maytansinoids from the cells had occurredover the 24-hour assay.

The catabolism rates of trastuzumab conjugatesfollowing exposure to BT-474EEI cells are similarThe radioactivity associated with the maytansinoid

catabolites in acetone extracts (Fig. 4A) and precipitates

from the acetone extractions were converted to pmol/celland nmol/L and plotted versus time (Fig. 4B). Of partic-ular interest for understanding the antimitotic activity ofT-DM1 is the concentration of itsmaytansinoid catabolitesin the cytoplasmof the targeted cells. Themolar values forthe catabolites in Fig. 4B may not accurately reflect theircytoplasmic concentrations if they accumulate in subcel-lular compartments such as lysosomes. However, previ-ous studieswith anti-CanAgmaytansioid conjugates sim-ilar to those described here suggest that maytansinoidcatabolites are retained by targeted cells through bindingto their cytoplasmic tubulin target (11). Therefore, themolar values in Fig. 4 likely approximate the true cyto-plasmic values. The amount of maytansinoids in theprecipitates (representing intact conjugates and any

Figure 2. A, efficacy oftrastuzumab–DM1 conjugates inmice-bearing BT-474EEI tumors.Individual tumor volumes versus timefollowing a single i.v. bolus dose oftrastuzumab–DM1 conjugates onday 0. Solid gray lines representindividual animal tumor volumes. Thegreen line is lower LOQ (8 mm3).Values below this are statisticallyimputed and recorded as 8 to avoidextreme negative or infinite values onlog scale. The blue line is fittedcontrol profile. B, mean tumorvolume measurements for eachanimal through day 38 versus ADCexposure (AUC) with box plotsoverlaid. The horizontal line is themedian of the 10 animal meanvolumes in each group; the ends ofthe box range from the 3rd to 7thhighest of the 10 means. Thisinterquartile range (IQR) is an outlier-resistant estimate of the variability ofeach group of mean volumes. Thedashed whiskers extend 1.5 IQR'sfrom the ends of the box, denoting aGaussian distribution-basedestimate of statistical variability.Values beyond the whiskers aretherefore considered statisticallyaberrant.

Effect of Linkers on Trastuzumab Maytansinoid Conjugates

www.aacrjournals.org Mol Cancer Ther; 11(5) May 2012 1137

on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727

Page 6: TheEffectofDifferentLinkersonTargetCellCatabolismand ......Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 ,

potential protein fragments) was found to decrease atapproximately the same rate for both conjugates. Thisdecrease was matched by a corresponding increase in themaytansinoid catabolites in the extracts, reaching maxi-mal intracellular maytansinoid concentrations of 200nmol/L and 300 nmol/L after 24 hours for cells treatedwith T-SPP-DM1 and T-DM1, respectively. In separateexperiments, the total maytansinoids associated with theextract and the precipitate for each time point were equiv-alent to the amount of conjugate bound at t ¼ 0 (data notshown) indicating that all maytansinoid catabolites hadbeen accounted for (upper curves, Fig. 4B). Approximate-ly 30% more T-DM1 was bound to cells after the incuba-tion and wash steps than T-SPP-DM1. This, coupled witha higherMAR for T-DM1 comparedwith T-SPP-DM1 (4.0vs. 3.2) accounts for the higher maytansinoid valuesassociated with cells treated with T-DM1 (Fig. 4B, a andb). The catabolite levels for the conjugateswere dividedbythe total maytansinoids in the sample (acetone pellet þextract � conjugate bound at t ¼ 0) to determine the

Figure 4. Activation of trastuzumab–DM1 conjugates in HER2-positive breast carcinoma cells. A, HPLC radiograms of the target cellcatabolites following exposure of BT474EEI cells to T-[3H]DM1 and T-SPP-[3H]DM1. Cells were harvested at the indicated time points andanalyzed for maytansinoids. The chromatograms show the fraction number (x-axis) and the CPM (y-axis). B, rates for the catabolism ofconjugates. Panels a and b show the concentration of maytansinoid catabolites of T-DM1 (a) and T-SPP-DM1 (b) formed within cells over time.Concentrations were calculated from the radioactivities in A and plotted versus time. A total of 1 pmol/106 cells is equivalent to 253 nmol/L. Thecorresponding concentration of intact conjugate still associated with the cells (*) was determined from the radioactivity associated with theacetone precipitates. ~, total maytansinoid levels. Panel c shows the percentage of the conjugates processed at each time. The processing of T-DM1 (&) and T-SPP-DM1(o) was calculated from a and b by dividing the catabolites of each conjugate formed at each time by the correspondingtotal maytansinoid levels.

Figure 3. Plasma pharmacokinetics in nontumor-bearing mice.Mean (� SD) ADC and Tab concentrations following i.v.bolus administration of 3 mg/kg of trastuzumab–DM1conjugates.

Erickson et al.

Mol Cancer Ther; 11(5) May 2012 Molecular Cancer Therapeutics1138

on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727

Page 7: TheEffectofDifferentLinkersonTargetCellCatabolismand ......Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 ,

fraction of the conjugates processed to the observed cat-abolites and replotted in Fig. 4B, c. The rates of formationof catabolites from the 2 conjugates are similar when sonormalized. HER2-mediated processing of these conju-gates is similar in other breast carcinoma lines (Supple-mentary Fig. S1).

Catabolism of trastuzumab conjugates within tumortissueThe concentration of the conjugates in plasma (based

on DM1) was calculated from the plasma radioactivityand shown in Fig. 5A. The concentration of T-DM1 after7 days is about 3-fold greater than the concentration ofT-SPP-DM1 (top), consistent with the ELISA PK innontumor-bearing mice (Fig. 3). The plasma conjugateconcentrations (ELISA) in nontumor-bearing mice(Fig. 3) when adjusted for dose, were approximately�2 higher than the estimated plasma conjugateconcentrations derived from the plasma radioactivitymeasurements in tumor bearing mice, consistent withobservations from a separate study (14). The causes ofthese discrepancies are not known, but do not affect theinterpretation of this study. The plasma concentrationsof the nonbinding control conjugates after 2 days wereequivalent to the matched trastuzumab conjugates, asexpected.Maytansinoid concentrations in the tumors of mice

treated with trastuzumab conjugates were found to reacha maximum level at about 1 to 2 days with peak concen-

trations of approximately 700 nmol/L (equivalent to 9%ID/g) or 500 nmol/L, (equivalent to 7% ID/g) for T-DM1or T-SPP-DM1, respectively, followed by a gradualdecline (Fig. 5A, middle). The 7 day AUC for the totalmaytansinoids in the tumors treated with T-DM1 was1.5- fold higher than T-SPP-DM1.

On the basis of HPLC radiochromatograms, the cata-bolites of the trastuzumab conjugates in tumor tissueswere identical to those identified in vitro—lysine-Ne-MCC-DM1 for T-DM1 and lysine-Ne-SPP-DM1 and DM1for T-SPP-DM1 (Fig. 5B). The concentrations of the may-tansinoid catabolites for the 2 conjugates (Fig. 5A, bottom)were very similar despite the differences in total maytan-sinoid levels (Fig. 5A, middle), with maximal maytansi-noid catabolite concentration for both conjugates ofapproximately 150 nmol/L.

The lysine-Ne-MCC-DM1 catabolite observed withT-DM1 was also observed for the control 5B6-MCC-DM1 conjugate at substantially reduced levels (Fig.6A). Similarly, the lysine-Ne-SPP-DM1 and DM1 cata-bolites observed for T-SPP-DM1 were also observedfor the 5B6-SPP-DM1 conjugate—again at reducedlevels. Concentrations of maytansinoid catabolites ofthe nontargeting conjugates were approximately 5-foldand 3-fold lower, respectively, than the concentrationsof the catabolites of T-DM1 and T-SPP-DM1 (Fig. 6B).These results show the effect of HER2 targeting onthe maytansinoid delivery of trastuzumab–DM1conjugates.

Figure 5. Tumor localization andcatabolism of trastuzumab–DM1conjugates. A, plasma clearance ofconjugate (top) and accumulation oftotal maytansinoids (middle) andmaytansinoid catabolites (bottom) intumors following administration of asingle i.v. dose of 200 mg/kg (basedon DM1 concentration) of T-[3H]DM1(~), T-SPP-[3H]DM1 (*), 5B6-MCC-[3H]DM1 (&), and 5B6-SPP-[3H]DM1(*) to mice. Concentrations ofconjugate in plasma (DM1concentration) and totalmaytansinoid (conjugate þcatabolites) in tumor were calculatedfrom the total radioactivity in plasmaand tumors samples, respectively,expressed as concentration. Themaytansinoid catabolite levels in thetumors were determined by theradioactivity associated with HPLCcatabolites shown in B. The countsper minute (CPM) of tritium in eacheffluent fraction was determined byLSC. The radiograms show thefraction number on the x-axis andCPM of tritium on the y-axis.

Effect of Linkers on Trastuzumab Maytansinoid Conjugates

www.aacrjournals.org Mol Cancer Ther; 11(5) May 2012 1139

on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727

Page 8: TheEffectofDifferentLinkersonTargetCellCatabolismand ......Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 ,

DiscussionInterest in ADCs has grown in response to the favor-

able safety and efficacy associated with T-DM1 andSGN-35—the latter, an antibody-auristatin conjugatetargeting CD30-positive tumors (2, 15). AdditionalADCs are in early clinical trials and more are expectedto follow (1, 16). Efforts to understand the molecularbasis for the anticancer activities of ADCs have inten-sified; with studies of maytansinoid and auristatin con-jugates providing a mechanistic basis for their clinicalactivity (11, 12, 17–20).

The aims of these studies were to investigate the MOAand anticancer activity of T-DM1 and the role of the linkerin these processes. To explore the underlyingmechanismsdetermining trastuzumab–maytansinoid ADC activity, invitro studies were conducted assessing ADC potency andthe kinetics of ADC uptake into tumor cells, and theidentity and accumulation of the catabolic products. Bothtrastuzumab conjugates were highly, and similarly,potent against HER2-expressing cell lines (Fig. 1; SK-BR-3, BT-474), consistent with a previous report (6). How-ever, T-DM1 exhibited slightly greater potency in theBT474EEI and MCF7-neo/HER2 cell lines. It is not clearwhy these cell lines behave differently than the other celllines tested.

In vitro tumor cell uptake and catabolism studiesshowed that the uptake and catabolism occurred at acomparable rate for both ADCs (Fig. 4B). This findingwas expected, based on the similarity in HER2 bindingaffinity of the ADCs (data not shown), and the assump-tion that target-mediated cellular trafficking and catab-olism of the ADCs are independent of linker type. Thehalf-life of cellular catabolism of the ADCs was found tobe approximately 19 hours, consistent with datareported for 125I-trastuzumab; indicating that the may-tansinoid conjugation does not alter catabolism rate(21). These studies also showed that lysine-Ne-MCC-DM1 is the sole T-DM1 catabolite accumulating in thetumor cells tested (Fig. 4A). The high in vitro activationrate for T-DM1 within cancer cells is consistent with its

potent cytotoxicity (Fig. 1) and is likely an importantfactor in its antitumor activity.

To explore the in vitro–in vivo correlation of these find-ings, PK, efficacy, in vivo tumor uptake, and catabolismstudies were conducted with the HER2þ, trastuzumabinsensitive, xenograft tumor BT474EEI. In vivo tumorcatabolism data confirmed the in vitro data identifyinglysine-Ne-MCC-DM1 as the sole catabolic product(Fig. 5B).

Previous nonclinical studies reported that T-DM1 dis-played slightly greater efficacy than T-SPP-DM1, whencomparedbydose (6). Itwashypothesized that the greaterefficacy of T-DM1, which uses a thioether to link DM1 tothe antibody, comparedwith T-SPP-DM1,which containsa disulfide linker, may be related to improved PK ordifferences in the tumor accumulation of active catabolicproducts. The plasma clearance of T-DM1 is approximate-ly 2 times slower than T-SPP-DM1 (Supplementary TableS1), likely due to greater stability of the thioether linker.This results in greater plasma exposure (AUC) per unitdose of T-DM1, but does not result in a substantiveincrease in efficacy (Fig. 2A). Indeed, comparing ADCefficacy as a function of plasma ADC exposure indicatessimilar in vivo potency (Fig. 2B).

The greater exposure of T-DM1 compared with T-SPP-DM1 corresponds with increased total tumor maytansi-noid concentrations (Fig. 5A) for T-DM1, a logical sequelabased on the similarity in the factors determining tumoruptake and binding for both ADCs. However, despitedifferences in total tumor maytansinoid concentrations,the tumor catabolite exposure levels for the 2 conjugatesare similar (Fig. 5A). This was not expected, as the highertotal maytansinoid concentrations for T-DM1, coupledwith themore residualizing nature of its catabolic productpredictedhigher catabolite concentrations forT-DM1.Thereasons for the similarity in tumor catabolite levels areunclear. One possibility is that the primary accumulationof conjugate in the tumor driving the subsequent HER2-mediated catabolism occurs within the first day or sowhen the plasma concentrations for the 2 conjugates are

Figure 6. HER2 dependence ofmaytansinoid delivery to tumor. A,HPLC radiograms associated withthe 2-day tumor catabolites of T-[3H]DM1, T-SPP-[3H]DM1, and thenontargeting 5B6-MCC-[3H]DM1and 5B6-SPP-[3H]DM1 conjugate.The radiograms show the fractionnumber (x-axis) and CPM of tritium(y-axis). B, concentrations of the 2-day tumor catabolites werecalculated from the peaks ofradioactivity in the radiogramsfrom A.

Erickson et al.

Mol Cancer Ther; 11(5) May 2012 Molecular Cancer Therapeutics1140

on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727

Page 9: TheEffectofDifferentLinkersonTargetCellCatabolismand ......Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 ,

similar. Another possibility is that the in vivo cleavage ofthe disulfide-linked T-SPP-DM1 was more efficient thanT-DM1 despite similar in vitro processing rates. For exam-ple, direct cleavage of DM1 from T-SPP-DM1 withintumor tissue via thiol-disulfide exchange reactions couldincrease the processing rate by complementing the lysine-Ne-SPP-DM1 released via the lysosomal degradationroute.It is postulated that the cellular catabolic products of

ADCs are responsible for their cytotoxic activity. Theobservation of similar tumor catabolite concentrationsfor T-DM1 and T-SPP-DM1 when both conjugates areadministered at the same ADC dose coupled with theslightly greater in vitro potency of T-DM1 in BT474EEIcells would predict greater efficacy for T-DM1; how-ever, no substantive difference in efficacy was observed(Fig. 2). This is a unique observation as, in studies withADCs targeting the CanAg antigen antibody maytan-sioid conjugate, thioether-linked conjugates were lessactive than disulfide-linked conjugates, even whentumor catabolite concentrations for the thioether-linkedADC were somewhat higher than those of the disul-fide-linked ADC (12), an observation attributed tobystander killing (11–13). Both the disulfide-linked andthioether-linked anti-CanAg conjugates were efficientlydegraded in the lysosomes of cancer cells to yield thecorresponding lysine-linker-maytansinoid catabolites.For the thioether-linked conjugate, no further catabo-lism was observed. However, the catabolites of the 2disulfide-linked conjugates huC242-SPP-DM1 andhuC242-SPDB-DM4 were both further processed toyield catabolites capable of diffusing throughout tumortissue, thereby enhancing the activity of the disulfide-linked conjugates through bystander killing mechan-isms. Although this may also apply to the trastuzumabADCs in this study, the greater in vitro potency of T-DM1 may compensate for the bystander effect associ-ated with T-SPP-DM1 to explain the similarity in in vivoefficacy.In summary, the studies described herein provide a

detailed understanding of mechanistic aspects of the PK,tumor uptake, and pharmacologic activity of T-DM1 andcontrast those properties with a more labile disulfidelinker. These studies confirm that T-DM1 andT-SPP-DM1

have potent antitumor activity in HER2-expressing celllines and show that T-DM1 is rapidly activated by HER2-positive cancer cells to lysine-Ne-MCC-DM1. Improvedlinker stability with T-DM1 leads to greater plasma ADCexposure and tumor uptake of total maytansinoid than T-SPP-DM1. Surprisingly, the increase in total tumor uptakewith T-DM1 does not translate into greater tumorcatabolite concentrations or improved efficacy, whencompared with T-SPP-DM1. Sustained exposure of thelysine-Ne-MCC-DM1 catabolite in tumors of treated miceprovides a mechanistic basis for the preclinical activity ofT-DM1. The efficacy and tumor-targeting characteristicsof T-DM1 and T-SPP-DM1 suggest that both linker for-mats are effective in payload delivery and antitumoractivity.

Disclosure of Potential Conflicts of InterestC.Audette has an ownership interest in ImmunoGen, Inc. J. Pinkas held

the role of Director (Pharmacology) in ImmunoGen, Inc.; also has own-ership interest in ImmunoGen, Inc. J. Tibbitts has ownership interest(including patents) in Roche. No potential conflicts of interest were dis-closed by the other authors.

Authors' ContributionsConception and design: H.K. Erickson, G.D. Lewis Phillips, C.A.Provenzano, B. Gunter, J. Pinkas, J. TibbittsDevelopment ofmethodology:H.K. Erickson, G.D. Lewis Phillips, E.Mai,M. Gupta, J. TibbittsAcquisition of data: H.K. Erickson, G.D. Lewis Phillips, D.D. Leipold, E.Mai, H.A. Johnson, J. Pinkas, J. TibbittsAnalysis and interpretation of data: H.K. Erickson, G.D. Lewis Phillips,D.D. Leipold, E. Mai, C.A. Provenzano, B. Gunter, M. Gupta, J. Pinkas,J. TibbittsWriting, review, and/or revision of the manuscript: H.K. Erickson, G.D.Lewis Phillips, D.D. Leipold, E. Mai, B. Gunter, C.A. Audette, M. Gupta,J. Pinkas, J. TibbittsAdministrative, technical, or material support: C.A. Audette, E. Mai,J. TibbittsStudy supervision: H.K. Erickson, D.D. Leipold, J. Pinkas, J. Tibbitts

AcknowledgmentsThe authors thank John Lambert and Ravi Chari for their many helpful

suggestions and careful reading of the manuscript.

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 October 4, 2011; revised January 24, 2012; accepted January 31,2012; published OnlineFirst March 9, 2012.

References1. Lambert JM. Antibody-maytansinoid conjugates: a new strategy for

the treatment of cancer. Drugs Fut 2010;35:471–80.2. Krop IE, BeeramM, Modi S, Jones SF, Holden SN, YuW, et al. Phase I

study of trastuzumab-DM1, an HER2 antibody-drug conjugate, givenevery 3 weeks to patients with HER2-positive metastatic breast can-cer. J Clin Oncol 2010;28:2698–704.

3. Burris HA, Rugo HS, Vukelja SJ, Vogel CL, Borson RA, et al. Phase IIstudy of the antibody drug conjugate trastuzumab-DM1 for the treat-ment of human epidermal growth factor receptor 2 (HER2)-positivebreast cancer after prior HER2-directed therapy. J Clin Oncol2011;29:398–405.

4. Burris HA. Trastuzumab emtansine: a novel antibody-drug conjugatefor HER2-positive breast cancer. Expert Opin Biol Ther 2011;11:807–19.

5. Junttila TT, Li G, Parsons K, PhillipsGL, Sliwkowski MX. Trastuzumab-DM1 (T-DM1) retains all the mechanisms of action of trastuzumab andefficiently inhibits growth of lapatinib insensitive breast cancer. BreastCancer Res Treat 2011;128:347–56.

6. Lewis Phillips GD, Li G, Dugger DL, Crocker LM, Parsons KL, Mai E,et al. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res 2008;68:9280–90.

Effect of Linkers on Trastuzumab Maytansinoid Conjugates

www.aacrjournals.org Mol Cancer Ther; 11(5) May 2012 1141

on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727

Page 10: TheEffectofDifferentLinkersonTargetCellCatabolismand ......Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 ,

7. Al-Katib AM, Aboukameel A, Mohammad R, Bissery MC, Zuany-Amorim C. Superior antitumor activity of SAR3419 to rituximab inxenograft models for non-Hodgkin's lymphoma. Clin Cancer Res2009;15:4038–45.

8. Chen Q, Millar HJ, McCabe FL, Manning CD, Steeves R, Lai K, et al.Alpha V integrin-targeted immunoconjugates regress establishedhuman tumors in xenograft models. Clin Cancer Res 2007;13:3689–95.

9. Ikeda H, Hideshima T, Fulciniti M, Lutz RJ, Yasui H, Okawa Y, et al.The monoclonal antibody nBT062 conjugated to cytotoxic maytan-sinoids has selective cytotoxicity against CD138-positive multiplemyeloma cells in vitro and in vivo. Clin Cancer Res 2009;15:4028–37.

10. Kellogg BA, Garrett L, Kovtun Y, Lai KC, Leece B, Miller M, et al.Disulfide-linked antibody-maytansinoid conjugates: optimizationof in vivo activity by varying the steric hindrance at carbonatoms adjacent to the disulfide linkage. Bioconjugate Chem 2011;22:717–27.

11. Erickson HK, Park PU,WiddisonWC, Kovtun YV, Garrett LM, HoffmanK, et al. Antibody-maytansinoid conjugates are activated in targetedcancer cells by lysosomal degradation and linker-dependent intracel-lular processing. Cancer Res 2006;66:4426–33.

12. Erickson HK, Widdison WC, Mayo MF, Whiteman K, Audette C,Wilhelm SD, et al. Tumor delivery and in vivo processing of disul-fide-linked and thioether-linked antibody-maytansinoid conjugates.Bioconjug Chem 2010;21:84–92.

13. Kovtun YV, Audette CA, Ye Y, Xie H, Ruberti MF, Phinney SJ, et al.Antibody-drug conjugates designed to eradicate tumors with homo-

geneous and heterogeneous expression of the target antigen. CancerRes 2006;66:3214–21.

14. Xie H, Audette C, Hoffee M, Lambert JM, Blattler WA. Pharmacoki-netics and biodistribution of the antitumor immunoconjugate, cantu-zumab mertansine (huC242-DM1), and its two components in mice.J Pharmacol Exp Ther 2004;308:1073–82.

15. Younes A, Bartlett NL, Leonard JP, Kennedy DA, Lynch CM, SieversEL, et al. Brentuximab vedotin (SGN-35) for relapsed CD30-positivelymphomas. N Engl J Med 2010;363:1812–21.

16. Senter PD. Potent antibody drug conjugates for cancer therapy. CurrOpin Chem Biol 2009;13:235–44.

17. Jumbe NL, Xin Y, Leipold DD, Crocker L, Dugger D, Mai E, et al.Modeling the efficacy of trastuzumab-DM1, an antibody drug conju-gate, in mice. J Pharmacokinet Pharmacodyn 2010;37:221–42.

18. Alley SC, Zhang X, Okeley NM, Anderson M, Law CL, Senter PD, et al.The pharmacologic basis for antibody-auristatin conjugate activity.J Pharmacol Exp Ther 2009;330:932–8.

19. Doronina SO, Mendelsohn BA, Bovee TD, Cerveny CG, Alley SC,Meyer DL, et al. Enhanced activity of monomethylauristatin F throughmonoclonal antibody delivery: effects of linker technology on efficacyand toxicity. Bioconjug Chem 2006;17:114–24.

20. Okeley NM, Miyamoto JB, Zhang X, Sanderson RJ, Benjamin DR,Sievers EL, et al. Intracellular activation of SGN-35, a potent anti-CD30antibody-drug conjugate. Clin Cancer Res 2010;16:888–97.

21. Austin CD, De Maziere AM, Pisacane PI, van Dijk SM, Eigenbrot C,Sliwkowski MX, et al. Endocytosis and sorting of ErbB2 and the site ofaction of cancer therapeutics trastuzumabandgeldanamycin.Mol BiolCell 2004;15:5268–82.

Erickson et al.

Mol Cancer Ther; 11(5) May 2012 Molecular Cancer Therapeutics1142

on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727

Page 11: TheEffectofDifferentLinkersonTargetCellCatabolismand ......Maytansinoid Conjugates Hans K. Erickson 1 , Gail D. Lewis Phillips 2 , Douglas D. Leipold 3 , Carmela A. Provenzano 1 ,

2012;11:1133-1142. Published OnlineFirst March 9, 2012.Mol Cancer Ther   Hans K. Erickson, Gail D. Lewis Phillips, Douglas D. Leipold, et al.   Maytansinoid ConjugatesPharmacokinetics/Pharmacodynamics of Trastuzumab The Effect of Different Linkers on Target Cell Catabolism and

  Updated version

  10.1158/1535-7163.MCT-11-0727doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2012/03/09/1535-7163.MCT-11-0727.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/11/5/1133.full#ref-list-1

This article cites 21 articles, 12 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/11/5/1133.full#related-urls

This article has been cited by 22 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://mct.aacrjournals.org/content/11/5/1133To request permission to re-use all or part of this article, use this link

on June 26, 2020. © 2012 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst March 9, 2012; DOI: 10.1158/1535-7163.MCT-11-0727