Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational...

12
Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab Emtansine (T-DM1) Matthew Sung 1 , Xingzhi Tan 1 , Bingwen Lu 1 , Jonathan Golas 1 , Christine Hosselet 1 , Fang Wang 1 , Laurie Tylaska 2 , Lindsay King 2 , Dahui Zhou 3 , Russell Dushin 3 , Jeremy S. Myers 1 , Edward Rosfjord 1 , Judy Lucas 1 , Hans-Peter Gerber 4 , and Frank Loganzo 1 Abstract Trastuzumab emtansine (T-DM1) is an antibodydrug con- jugate (ADC) that has demonstrated clinical benet for patients with HER2 þ metastatic breast cancer; however, its clinical activity is limited by inherent or acquired drug resistance. The molecular mechanisms that drive clinical resistance to T-DM1, especially in HER2 þ tumors, are not well understood. We used HER2 þ cell lines to develop models of T-DM1 resistance using a cyclical dosing schema in which cells received T-DM1 in an "on- off" routine until a T-DM1resistant population was generated. T-DM1resistant N87 cells (N87-TM) were cross-resistant to a panel of trastuzumab-ADCs (T-ADCs) with noncleavable- linked auristatins. N87-TM cells do not have a decrease in HER2 protein levels or an increase in drug transporter protein (e.g., MDR1) expression compared with parental N87 cells. Intriguingly, T-ADCs using auristatin payloads attached via an enzymatically cleavable linker overcome T-DM1 resistance in N87-TM cells. Importantly, N87-TM cells implanted into athymic mice formed T-DM1 refractory tumors that remain sensitive to T-ADCs with cleavable-linked auristatin payloads. Comparative proteomic proling suggested enrichment in pro- teins that mediate caveolae formation and endocytosis in the N87-TM cells. Indeed, N87-TM cells internalize T-ADCs into intracellular caveolin-1 (CAV1)positive puncta and alter their trafcking to the lysosome compared with N87 cells. T-DM1 colocalization into intracellular CAV1-positive puncta correlat- ed with reduced response to T-DM1 in a panel of HER2 þ cell lines. Together, these data suggest that caveolae-mediated endocytosis of T-DM1 may serve as a novel predictive biomark- er for patient response to T-DM1. Mol Cancer Ther; 17(1); 24353. Ó2017 AACR. Introduction Antibodydrug conjugates (ADCs) are emerging therapeutic modalities strategically designed to selectively deliver chemo- therapeutic agents to tumors while limiting non-tumor tissue toxicities. ADCs are biotherapeutics compromised of a tumor- targeting antibody and a cytotoxic payload attached via a chemical linker. Once bound to their target antigen, ADCs are internalized and release their cytotoxic payload. Since the early 2000s, three ADCs have received FDA-approval and over 50 more are current- ly in clinical development (1, 2). Despite encouraging signs of early clinical benet, the sustained efcacy of many targeted anticancer therapies is limited by either inherent or acquired tumoral drug resistance. The molecular mechanisms that drive clinical drug resistance are multifactorial and ultimately depend upon the therapeutic modality and cancer indication (3). Modications of the drug target (e.g., kinase mutations; refs. 4, 5), drug efux protein expression (e.g., ABCB1 in AML; refs. 68), and shifts in the expression ratio of pro- versus antiapoptotic proteins (e.g., BCL-2 expression in AML; ref. 9) are examples of tumor alterations that correlate with reduced clinical response to anticancer therapies. However, the molecular mechanisms that drive clinical resistance to ADCs remain largely unknown. Trastuzumab emtansine (T-DM1, Kadcyla Ò ) currently repre- sents the only FDA-approved ADC for the treatment of a solid tumor indication. T-DM1 is composed of the trastuzumab anti- body chemically bound to the microtubule-disrupting DM1 maytansinoid via a thioether linker (10). In the phase III EMILIA clinical trial, T-DM1 showed signicantly improved progression- free and overall survival versus lapatinib plus capecitabine in HER2 þ metastatic breast cancer patients who had failed previous treatment with trastuzumab and a taxane (11). The overall response rate for HER2 þ patients treated with T-DM1 was 43.6% (11). On the basis of these results, T-DM1 was granted FDA approval for the treatment of this patient population. How- ever, these data showed that, despite high tumoral HER2-posi- tivity, over 50% of patients treated with T-DM1 did not show clinical benet. In the phase II/III GATSBY clinical trial in HER2 þ gastric cancer patients, T-DM1 did not show enhanced clinical response versus standard chemotherapy (12). Thus, HER2 1 Pzer Inc., Oncology Research and Development, Pearl River, New York. 2 Pzer Inc., Biomedicine Design, Groton, Connecticut. 3 Pzer Inc., Worldwide Medicinal Chemistry, Groton, Connecticut. 4 Maverick Therapeutics Inc., Brisbane, California. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Current address for Hans-Peter Gerber, Maverick Therapeutics Inc., Brisbane, California. Corresponding Author: Matthew Sung, Pzer, Inc., 401 North Middletown Road, 200/4618, Pearl River, NY 10965. Phone: 845-602-3731; Fax: 845-474-3161; E-mail: matthew.sung@pzer.com doi: 10.1158/1535-7163.MCT-17-0403 Ó2017 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 243 on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Transcript of Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational...

Page 1: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

Cancer Biology and Translational Studies

Caveolae-Mediated Endocytosis as a NovelMechanism of Resistance to TrastuzumabEmtansine (T-DM1)Matthew Sung1, Xingzhi Tan1, Bingwen Lu1, Jonathan Golas1, Christine Hosselet1,Fang Wang1, Laurie Tylaska2, Lindsay King2, Dahui Zhou3, Russell Dushin3,JeremyS.Myers1, EdwardRosfjord1, Judy Lucas1, Hans-PeterGerber4, andFrank Loganzo1

Abstract

Trastuzumab emtansine (T-DM1) is an antibody–drug con-jugate (ADC) that has demonstrated clinical benefit for patientswith HER2þ metastatic breast cancer; however, its clinicalactivity is limited by inherent or acquired drug resistance. Themolecular mechanisms that drive clinical resistance to T-DM1,especially in HER2þ tumors, are not well understood. We usedHER2þ cell lines to develop models of T-DM1 resistance using acyclical dosing schema in which cells received T-DM1 in an "on-off" routine until a T-DM1–resistant population was generated.T-DM1–resistant N87 cells (N87-TM) were cross-resistant to apanel of trastuzumab-ADCs (T-ADCs) with non–cleavable-linked auristatins. N87-TM cells do not have a decrease inHER2 protein levels or an increase in drug transporter protein(e.g., MDR1) expression compared with parental N87 cells.Intriguingly, T-ADCs using auristatin payloads attached via

an enzymatically cleavable linker overcome T-DM1 resistancein N87-TM cells. Importantly, N87-TM cells implanted intoathymic mice formed T-DM1 refractory tumors that remainsensitive to T-ADCs with cleavable-linked auristatin payloads.Comparative proteomic profiling suggested enrichment in pro-teins that mediate caveolae formation and endocytosis in theN87-TM cells. Indeed, N87-TM cells internalize T-ADCs intointracellular caveolin-1 (CAV1)–positive puncta and alter theirtrafficking to the lysosome compared with N87 cells. T-DM1colocalization into intracellular CAV1-positive puncta correlat-ed with reduced response to T-DM1 in a panel of HER2þ celllines. Together, these data suggest that caveolae-mediatedendocytosis of T-DM1 may serve as a novel predictive biomark-er for patient response to T-DM1. Mol Cancer Ther; 17(1); 243–53.�2017 AACR.

IntroductionAntibody–drug conjugates (ADCs) are emerging therapeutic

modalities strategically designed to selectively deliver chemo-therapeutic agents to tumors while limiting non-tumor tissuetoxicities. ADCs are biotherapeutics compromised of a tumor-targeting antibody anda cytotoxic payload attached via a chemicallinker. Once bound to their target antigen, ADCs are internalizedand release their cytotoxic payload. Since the early 2000s, threeADCs have received FDA-approval and over 50 more are current-ly in clinical development (1, 2).

Despite encouraging signs of early clinical benefit, the sustainedefficacy of many targeted anticancer therapies is limited by either

inherent or acquired tumoral drug resistance. The molecularmechanisms that drive clinical drug resistance are multifactorialand ultimately depend upon the therapeutic modality and cancerindication (3). Modifications of the drug target (e.g., kinasemutations; refs. 4, 5), drug efflux protein expression (e.g., ABCB1in AML; refs. 6–8), and shifts in the expression ratio of pro- versusantiapoptotic proteins (e.g., BCL-2 expression in AML; ref. 9) areexamples of tumor alterations that correlate with reduced clinicalresponse to anticancer therapies. However, the molecularmechanisms that drive clinical resistance to ADCs remain largelyunknown.

Trastuzumab emtansine (T-DM1, Kadcyla�) currently repre-sents the only FDA-approved ADC for the treatment of a solidtumor indication. T-DM1 is composed of the trastuzumab anti-body chemically bound to the microtubule-disrupting DM1maytansinoid via a thioether linker (10). In the phase III EMILIAclinical trial, T-DM1 showed significantly improved progression-free and overall survival versus lapatinib plus capecitabine inHER2þ metastatic breast cancer patients who had failed previoustreatment with trastuzumab and a taxane (11). The overallresponse rate for HER2þ patients treated with T-DM1 was43.6% (11). On the basis of these results, T-DM1 was grantedFDA approval for the treatment of this patient population. How-ever, these data showed that, despite high tumoral HER2-posi-tivity, over 50% of patients treated with T-DM1 did not showclinical benefit. In the phase II/III GATSBY clinical trial in HER2þ

gastric cancer patients, T-DM1 did not show enhanced clinicalresponse versus standard chemotherapy (12). Thus, HER2

1Pfizer Inc., Oncology Research and Development, Pearl River, New York.2Pfizer Inc., Biomedicine Design, Groton, Connecticut. 3Pfizer Inc., WorldwideMedicinal Chemistry, Groton, Connecticut. 4Maverick Therapeutics Inc.,Brisbane, California.

Note: Supplementary data for this article are available at Molecular CancerTherapeutics Online (http://mct.aacrjournals.org/).

Current address for Hans-Peter Gerber, Maverick Therapeutics Inc., Brisbane,California.

Corresponding Author:Matthew Sung, Pfizer, Inc., 401 NorthMiddletown Road,200/4618, Pearl River, NY 10965. Phone: 845-602-3731; Fax: 845-474-3161;E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-17-0403

�2017 American Association for Cancer Research.

MolecularCancerTherapeutics

www.aacrjournals.org 243

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Page 2: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

expression does not solely predict clinical response to T-DM1 andthe need exists to more accurately define the patient sub-popu-lation most likely to respond to T-DM1.

Although data are still emerging from T-DM1–treated patientsamples, multiple pre-clinical models of T-DM1 resistance havebeen generated to characterize the changes at the tumor cell levelthat may mediate clinical resistance to T-DM1. Although themodels of T-DM1 resistance in these studies were generated withdifferent dosing schemas, the following are mechanisms respon-sible for T-DM1 resistance seen across multiple models: (i)decreased HER2 expression (13, 14), (ii) increased expressionand activity of drug efflux proteins (13, 14), and (iii) increasedheregulin expression (15).

Herein, we used an ADC structurally similar to T-DM1 (13) togenerate T-DM1–resistant cells in vitro. We report the generationand characterization of threemodels of T-DM1 resistance, includ-ing one with a unique trafficking defect that has not been previ-ously associatedwith T-DM1acquired resistance andmayprovidea novel biomarker for clinical response to T-DM1.

Materials and MethodsCell lines

NCI-N87 gastric carcinoma, HCC1954 breast carcinoma, andBT474 breast carcinoma cells were obtained from the ATCC andmaintained in RPMI media supplemented with 10% FBS, 1%L-glutamine, and 1% sodium pyruvate. H69 andH69AR cell lineswere obtained from the ATCC and were maintained in RPMI/25 mmol/L HEPES media supplemented with 10% FBS, 1%L-glutamine, and 1% sodium pyruvate. KB, KB8.5, and KBVI cellswere generously provided by Dr. Michael Gottesman (NationalCancer Institute). Parental N87 cells were purchased from theATCC (CRL-5822, lot 58033347) in June 2010. N87 andN87-TMcells were authenticated as NCI-N87 in 2014 (IDEXX Bio-Research), when the cells were being characterized for this work.

Mycoplasma testing was conducted approximately quarterlyusing MycoAlert (Lonza), and results were consistently negative;the most recent testing was conducted in October 2016.

BioconjugationsADCs and chemical structures of disclosed payloads described

herein were prepared as previously described (13). The synthesesand structures of the novel auristatin linker-payloads as well aspreparation and characterization of ADCs presented herein aredescribed in the Supplementary Methods.

Generation of T-DM1–resistant cellsCells were passaged into two separate flasks and each flask was

treated identically with respect to the resistance generation pro-tocol to enable biological duplicates. Cells were exposed to fivecycles of T-DM1 conjugate at 10-fold IC50 concentrations (10nmol/L payload concentration; �388 ng/mL antibody concen-tration) for 3 days, followed by approximately 4 to 11 daysrecovery without treatment. After five cycles at 10 nmol/L of theT-DM1 conjugate, the cellswere exposed to six additional cycles of100 nmol/L T-DM1 in a similar fashion. The procedure wasintended to simulate the chronic, multi-cycle (on/off) dosing atmaximally tolerated doses typically used for cytotoxic therapeu-tics in the clinic, followed by a recovery period. Parental cellsderived fromNCI-N87 are referred to asN87, and cells chronicallyexposed to T-DM1 are referred to as N87-TM. Moderate- to high-

level drug resistance developedwithin 4months forN87-TMcells.Drug selection pressure was removed after approximately 3 to 4months of cycle treatments when the level of resistance no longerincreased after continued drug exposure. Responses and pheno-types remained stable in the cultured cell lines for approximately3 to 6 months. Thereafter, a reduction in the magnitude of theresistance phenotype as measured by cytotoxicity assays wasoccasionally observed, in which case early passage cryo-preservedT-DM1–resistant cells were thawed for additional studies. Allreported characterizations were conducted after removal ofT-DM1 selection pressure for at least 2 to 8 weeks to ensurestabilization of the cells. Data were collected from various thawedcryopreserved populations derived from a single selection, overapproximately 1 to 2 years after model development to ensureconsistency in the results. Clonal populations were derived fromsingle N87-TM cells using a limited dilution technique. Briefly,single cells were plated into each well of a 96-well plate andcultured in the presence of 100 nmol/L T-DM1 until clonalpopulations were established.

Cytotoxicity assayCellular responses to ADCs and free drugs were measured with

a cytotoxicity assay as previously described (13). Briefly, cells wereseeded into96-well plates and treatedwith varying concentrationsof ADCs or free drugs for 96 hours. Cell viability was measuredusing CellTiter 96 AQueous One MTS Solution (Promega). IC50

values were calculated using a four-parameter logistic model withXLfit (IDBS).

Proteomic profiling and immunoblot analysesProteomic profiling ofmembrane fractions was performed and

analyzed as previously described (13). Whole-cell lysates wereprepared in RIPA buffer supplemented with protease inhibitorcocktail (Sigma). Lysates were fractionated on 4% to 20% SDS-PAGE gels (Bio-Rad), transferred onto membranes and probedwith primary and secondary antibodies further detailed in theSupplementary Methods.

Immunofluorescence and live-cell imagingFor immunofluorescence experiments, cells were seeded onto

#1.5 coverslips (Electron Microscopy Services) and fixed with 4%paraformaldehyde. Cells were stained with primary antibodies atroom temperature (RT) for 1 hour and secondary antibodies at RTfor 30 minutes. Coverslips were mounted onto slides with Pro-Long Diamond Antifade Mountant (Thermo Fisher Scientific).Images were acquired with a Zeiss LSM 710 (Carl Zeiss Inc.). Forimaging cytometry, cells were incubated with PE-labeled trastu-zumab (2 mg/mL) on ice for 90minutes or at 37�C for time pointsindicated. At designated time points, internalization was stoppedby addition of cold PBS and cells were fixed with Cytofix/Cyto-perm solution (BD Biosciences) for 20 minutes. Cells wereimmunostained with LAMP1-AF647 (BD Biosciences; cat#562622) for 30 minutes or CAV1 antibody for 30 minutesfollowed by an anti-rabbit IgG Fab2 AF647 secondary antibody(ThermoFisher Scientific; cat# A-21246) for 30 minutes. Cellswere analyzed on the ImageStreamX Mark II Imaging Flow Cyt-ometer (AMNIS, EMD Millipore). For live-cell imaging experi-ments, cells were seeded into 4-well CELLview dishes (GrenierBio-One) and imaged with an ImagEM camera (HamamatsuPhotonics K.K.,HamamatsuCity, Japan)mounted onto aCSU-X1spinning disk head (Yokogawa Electric Corporation, Inc.) on an

Sung et al.

Mol Cancer Ther; 17(1) January 2018 Molecular Cancer Therapeutics244

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Page 3: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

Eclipse Ti microscope stand (Nikon Instruments Inc.) equippedwith a 60x oil objective (NA1.4, Nikon Instruments Inc.). Imageanalysis was performed using Volocity 3D Image software(PerkinElmer).

In vivo efficacy studiesAll procedures performed on animals in this study were in

accordance with established guidelines and regulations, and werereviewed and approved by the Pfizer Institutional Animal Careand Use Committee. Pfizer animal care facilities that supportedthis work are fully accredited by AAALAC International. FemaleNOD scid gamma (NSG) immunodeficient mice (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) were obtained from The Jackson Lab-oratory. Mice were injected subcutaneously in the right flank withsuspensions of either N87 or N87-TM cells (7.5 � 106 cells perinjection, with 50% Matrigel). Mice were randomized into studygroups when tumors reached approximately 0.3 g (�250 mm3).ADCs or vehicle were administered intravenously in saline on day0 and repeated for a total of four doses, 4 days apart (q4d � 4).Tumors were measured with calipers weekly and tumor masscalculated as volume ¼ (width � width � length)/2.

ImmunohistochemistryFive micrometer–thick formalin-fixed, paraffin-embedded tis-

sue sections were stained immunohistochemically. Detailed sam-ple preparations and staining procedures are provided in theSupplementary Methods.

ResultsGeneration and characterization of T-DM1–resistant cells

To identify potential mechanisms that may drive clinical resis-tance to T-DM1 ADC, we generated T-DM1–resistant versions oftheHER2-expressing cancer cell linesN87, BT474, andHCC1954.We used a cyclical dosing schema of a 3-day exposure to high doseT-DM1ADC followed by awashout and recovery period tomimicpatient exposure and pharmacokinetics when they receive bolus-chemotherapy treatment cycles (Fig. 1A).

N87 cells were inherently sensitive to T-DM1 (IC50 ¼ 1.6nmol/L payload concentration; 62 ng/mL antibody concentra-tion; Fig. 1B). Two populations of N87 cells were exposed to thetreatment cycles and, after approximately four months of cyclicaldosing, these two populations (henceforth namedN87-TM-1 andN87-TM-2) became refractory to the ADC by 109-fold comparedwith parental N87 cells (Fig. 1B, Table 1). Interestingly, minimalcross-resistance (1.8x) to the corresponding unconjugated may-tansinoid free drug, DM1-SMe, was observed (SupplementaryFig. S1, Table 1) which suggests the alterations in N87-TM cellsthat lead to T-DM1 resistance are specific to the whole ADCbiomolecule and not solely to the payload.

Previous reports of T-DM1 resistance in other cell models haveimplicated ATP-binding cassette (ABC) transporter protein over-expression (e.g., ABCB1/MDR1, ABCC1/MRP1) or target antigendownregulation as key drivers of T-DM1 resistance (13, 14).However, proteomics and immunoblots demonstrate that N87-TM cells do not overexpress MDR1 (Fig. 1C) or MRP1 (Fig. 1D)and retain similar levels of HER2 as compared with parental N87cells (Fig. 1E).Despite retaininghigh levels ofHER2,N87-TMcellsdisplay approximately 50% reduced binding to the unconjugatedantibody of T-DM1 (i.e., trastuzumab) as measured by flowcytometry (Supplementary Fig. S2). In contrast, T-DM1–resistant

populations of BT474 (BT474-TM) and HCC1954 (HCC1954-TM), selected in a similar manner as N87-TM, show significantlydecreased HER2 expression (Supplementary Fig. S3A and S3B),suggesting that reduced antigen is the primary mode of resistancein these two cell models. Hence, we focused additional charac-terization on the N87-TM model.

Taking advantage of the modular nature of ADCs, we hypoth-esized that altering key components of the biomolecule wouldresult in an agent able to overcome T-DM1 resistance in N87-TMcells. N87-TM cells displayed cross-resistance to several trastuzu-mab-based ADCs (T-ADCs; Fig. 1F and Table 1) which wereconjugated to auristatins via a non-cleavable maleimide linker.Remarkably, N87-TM cells were sensitive to T-ADCs conjugatedto auristatins using a protease-cleavable linker sequence (ValCit-PAB) in an antigen-dependent manner (Fig. 1G, SupplementaryFig. S4 and Table 1, Supplementary Table S1). The sensitivity ofN87-TM cells to other trastuzumab-based ADCs suggests thatreduced levels of HER2 in these cells is not a primary mode ofresistance.

To determine whether the ADC-resistant N87-TM cells werebroadly resistant to other standard-of-care chemotherapeutics,N87 and N87-TM cells were treated with DNA-damaging agents(e.g., cisplatin, doxorubuicin), kinase inhibitors (e.g., rapamycin,dacomitinib), and microtubule inhibitors (e.g., docetaxel, vin-blastine). Importantly, N87-TM cells remained sensitive to thesesmall molecule inhibitors when compared with the parentalN87 cells, discounting general growth defects as a mechanismof resistance to T-DM1 ADC (Supplementary Fig. S4, Table 1).

N87-TM tumors maintain T-DM1 resistance in vivoA problem that often plagues in vitro drug resistance models is

their lack of translatability into the in vivo setting. To determinewhether the resistance observed in cell culturewas recapitulated invivo, N87 cells and N87-TM cells were injected into the flanks offemale NOD scid gamma (NSG) immunodeficient mice andtreated with T-DM1 ADC [at doses of 6 mg/kg (mpk) and 10mpk)] and a T-ADC with a cleavable-linked auristatin (T-vc-Aur0101; at 3 mpk), on a q4d � 4 schedule. The N87 tumorsretained high expression of HER2 in vivo and were quite sensitiveto all three T-ADC treatment conditions (Fig. 2A). Remarkably,N87-TM tumors also retained high expression ofHER2 in vivo andwere refractory to both regimens of T-DM1 ADC, yet tumorsregressed with T-vc-Aur0101 (Fig. 2B). Thus, N87-TM tumorsretain a similar response profile to T-DM1 and T-vc-Aur0101ADCs in vitro and in vivo.

To better understand how N87-TM tumors are resistant toT-DM1 and sensitive to T-vc-Aur0101, we first interrogated thetumoral distribution of the ADCs. N87-TM tumors, from thepreviously described efficacy study, were collected 24 hoursfollowing the last ADC dose, and ADC distribution was evaluatedthrough immunohistochemistry stainingwith an anti-human IgGantibody. N87-TM tumors treated with 6 mpk T-DM1 ADCdisplayed almost complete tumor coverageof IgGbinding,where-as those treated with 3 mpk T-vc-Aur0101 only had ADC bindingat themost vessel-proximal regions of the tumor (SupplementaryFig. S5). Thus, tumoral ADCdistribution is unlikely to explain theefficacy response of these ADCs in N87-TM tumors.

Next, we evaluated a biomarker of downstream response to ananti-microtubule–targeted agent to determine how T-DM1 andT-vc-Aur0101 ADCs differentially impacted N87 and N87-TMtumors. Phosphorylation of histone H3 (pHH3) is a marker

Altered Trafficking as Mechanism of ADC Resistance

www.aacrjournals.org Mol Cancer Ther; 17(1) January 2018 245

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Page 4: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

Figure 1.

N87-TM model generation and characterization. A, Schematic model of cyclical dosing paradigm used to generate T-DM1 resistant cells. Parental cells were dosedwith T-DM1 for 3 days, followed by a washout and recovery period. This process was cycled multiple times until a T-DM1 resistant population emerged.B, Parental N87 (closed circle) and two T-DM1–resistant populations (open square and open circle), selected simultaneously (N87-TM-1 andN87-TM-2), were treatedwith T-DM1 for 4 days. Cell viability is plotted against ADC payload concentration. C–E, Whole-cell lysates from parental and T-DM1–resistant N87 cells wereseparated by SDS-PAGE and levels of MDR1 (C), MRP1 (D), and HER2 (E) were determined by immunoblot analyses. Whole-cell lysates from positive and negativecontrol cell lines for MDR1 (KB85 and KBV1) andMRP1 (A549 andH69AR) are shown. F,Parental and T-DM1–resistant N87 cellswere treatedwith a trastuzumab-ADCconjugated to a non-cleavable linker and auristatin payload for 4 days. Cell viability is plotted against ADC payload concentration. G, Parental and T-DM1–resistantN87 cells were treated with a trastuzumab–ADC conjugated to a cleavable-linked auristatin payload for 4 days. Cell viability is plotted against ADC payloadconcentration.

Sung et al.

Mol Cancer Ther; 17(1) January 2018 Molecular Cancer Therapeutics246

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Page 5: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

ofmitotic cells and canbeused to evaluate the activity of antimito-tic agents. To determine whether N87-TM tumors had reducedpHH3 staining following T-DM1 ADC treatment as comparedwith N87 tumors, mice were administered a single-dose ofT-DM1 ADC (6 mpk) and tumors were collected 24 hours later.Co-immunostaining revealed ADC and pHH3 double–positivecells in the N87 tumors (Fig. 2C, top left), whereas many of theADC-bound N87-TM tumor cells were pHH3 negative (Fig. 2C,bottom left). These data are consistent with the efficacy datashowing N87, but not N87-TM, tumors are sensitive to T-DM1ADC. In a similar study conducted with T-vc-Aur0101 ADC(3 mpk), N87 and N87-TM tumors had ADC and pHH3 doublepositive cells (Fig. 2C, top and bottom middle, respectively).Intriguingly, both T-vc-Aur0101 ADC treated tumors displaymany pHH3-positive cells without ADC-bound to them, a prop-erty known as bystander effect whereby antigen negative tumorcells neighboring antigen-positive tumor cells are susceptible tomembrane permeable payloads. Thus, it is clear from this in vivobiomarker study that T-ADCs utilizing a cleavable linker thatreleases a membrane permeable cytotoxic payload can overcomeT-DM1 resistance in N87-TM tumors.

The active released species from a catabolized T-DM1moleculeis the amino acid capped linker payload, Lysine-MCC-DM1 (16,17). The structural and ionic nature of Lysine-MCC-DM1 in thetumor microenvironment renders the molecule membraneimpermeable and is likely why T-DM1 does not show strongbystander effect (18). To determine whether amino-acid cappingof the released ADC species dictates differential ADC cytotoxicresponse in N87-TM as compared with N87 cells, we prepared aseries of ADCs using the auristatin analog (Aur8261) conjugatedto the same targeting antibody (trastuzumab) via different linkers

(19). As shown above, N87-TM cells are cross-resistant to a T-ADCwith a non–cleavable-linked payload with a released species ofcysteine-mc-Aur8261. However, N87-TM cells are sensitive to aT-ADC with Aur8261 attached via a cleavable linker where thereleased species is the unmodified, membrane-permeable auris-tatin compound. N87-TM cells were cross-resistant to a T-ADCwith a cleavable linker but with a released species of cysteine-mc-Aur8261 (Supplementary Fig. S6). Thus, T-ADCswhose activecatabolite is an amino acid-capped linker payload are effective inkilling N87, but not N87-TM, cells. Taken together, these datashow that N87-TM tumors maintain T-DM1 resistance in vivo andT-ADCs with cleavable linkers and permeable payloads overcomeT-DM1 resistance in vitro and in vivo.

The role of CAV1 in ADC biology and resistanceTo further characterize the mechanism of T-DM1 resistance in

N87-TM cells in an unbiased approach, we performed a proteo-mic evaluation of membrane fractions to profile proteins differ-entially expressed in the N87-TM cells as compared with theparental N87 cells. Significant changes in expression level of523 proteins between the cell lines were observed (Fig. 3A).Immunoblot analysis validated a number of the proteomic hits(Fig. 3B). Of interest was the overexpression of caveolin-1 (CAV1)and polymerase I and transcript release factor (PTRF) in N87-TMcells. As CAV1 and PTRF are essential protein constituents ofcaveolae (20),wehypothesized thatN87-TM cells are enriched forcaveolar endocytic compartments. Caveolae-mediated endocyto-sis is thought to be induced by Src-mediated phosphyloration ofCAV1 at Tyr 14 (21). Indeed, N87-TM cells have enhancedphospho-Src and phospho-CAV1 as compared with N87 cells(Fig. 3C). Immunofluorescence for CAV1 in N87 and N87-TM

Table 1. Resistance profiles of N87 and N87-TM cells to ADCs, standard-of-care chemotherapeutics and other unconjugated drugs

N87: Parental versus TM-resistantCompound Linker N87 N87-TM-2 RelativeName Type (IC50, nmol/L) (IC50, nmol/L) Resistance

T-MCC_DM1 NC 1.6 174 109xT_mc_Aur8261 NC 5.3 >1,000 >188xT_mc_MMAD NC 16 492 31xT_mc_Aur0101 NC 138 >1,000 >7xT_vc_Aur8261 C 0.43 0.49 1.1xT-vc_MMAD C 0.4 0.88 2.2xT_vc_Aur0101 C 1.1 0.8 0.73xT_vc_Cys-mc-Aur8261 C 3.9 136 35xDM1-Sme — 19 35 1.8xAur-8254 (8261) — 0.61 0.92 1.5xMMAD — 0.14 0.49 3.5xAur-0101 — 1.5 1.9 1.3xCys_mc-Aur8261 — 2,334 4,392 1.9xVinblastine — 3.6 4.8 1.3xDocetaxel — 2.9 3.8 1.3xDoxorubicin — 38 37 1.0x5-fluorouracil — 1,611 3,135 1.9xCamptothecin — 21 9.8 0.47xGemcitabine — 7.3 8.4 1.2xOxaliplatin — 623 1,511 2.4xTemsirolimus — 2,131 6,706 3.1xRapamycin — 4,682 12,403 2.6xDacomitinib — 7.8 33 4.2xPelitinib — 42 119 2.8xNeratinib — 2 11 5.5x

NOTE: IC50 values of ADCs reported here represent payload concentrations. Data are mean of multiple determinations. These relative resistance values arerepresented graphically in Supplementary Fig. S4. Relative resistance is the ratio of the mean IC50 value for the TM-resistant cell line versus the parental cell line.Abbreviations: C, cleavable; NC, noncleavable.

Altered Trafficking as Mechanism of ADC Resistance

www.aacrjournals.org Mol Cancer Ther; 17(1) January 2018 247

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Page 6: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

cells demonstrates that N87-TM cells contain many more intra-cellular CAV1þ puncta than N87 cells (Fig. 3D).

Wenext hypothesized thatN87-TM cells can internalize T-DM1into these caveolar endocytic compartments. We treated N87 andN87-TM cells with T-DM1 for 24 hours and co-immunostainedfor anti-human IgG andCAV1 to interrogate the co-localization ofthe ADC with CAV1þ puncta. Indeed, T-DM1 ADC co-localizedwith CAV1 in N87-TM, but not N87, cells (Fig. 3E). To determinewhether this positive correlation between T-DM1 ADC and CAV1occurred at the early phases of ADC endocytosis, we analyzedtrastuzumab and CAV1 co-localization at multiple early timepoints using imaging cytometry. N87-TM, but not N87, cellsshowed enhanced trastuzumab and CAV1 co-localization overtime (Fig. 3F). Because the internalization of trastuzumab wasinto differential compartments in N87 and N87-TM cells, we next

sought to determine whether N87-TM cells displayed impairedlysosomal trafficking of trastuzumab because T-DM1 is catabo-lized in the lysosome. Lysosomal co-localization of trastuzumabwas decreased in N87-TM as compared with N87 cells (Fig. 3F).

Caveolar endocytic compartments are phenotypically distinctfrom the endo-lysosomal pathway (22). For example, the luminaof caveolar endocytic compartments have a neutral pH, whereasthe lumina of endo-lysosomal compartments are increasinglyacidic during pathway maturation (23). To determine whetherT-ADCs were internalized into compartments with different pH,we generated a T-ADC conjugated to two fluorescent moieties: (i)a pH-insensitive AlexaFluor-modified auristatin non-cleavablelinker payload and (ii) a pH-sensitive dye, pHRodo, wherebythe fluorescence signal is proportional to the acidity of themicro-environment. We performed live-cell imaging with this

Figure 2.

N87-TM cells retain T-DM1 resistanceand T-vc-Aur0101 sensitivity in vivo.A and B, Parental (A) and T-DM1–resistant (B) N87 cells were implantedinto NOD-SCID mice and treated withADCswhen the average tumor volumeof 250 mm3 was achieved. Mice weredosed q4dx4 with vehicle (blackcircles), T-DM1 at 6 mg/kg (mpk; redsquares), T-DM1 at 10 mpk (redtriangles), T-vc-Aur0101 at 3 mpk(blue diamonds), or a negative control,isotype ADC with "vc-Aur0101" at 3mpk (hashed blue line withdownward-pointing triangles).Tumors at staging size for each cell linewere collected, formalin-fixed andimmunostained for HER2 byimmunohistochemistry (insets). C,N87 and N87-TM tumors were treatedwith a single dose of T-DM1 at 6 mpk,T-vc-Aur0101 at 3 mpk or a negativecontrol, isotype ADC with "vc-Aur0101" at 3 mpk. After 24 hours,mice were perfused and tumorsformalin fixed and co-immunostainedfor anti-human IgG (brown color) andphospho-histone H3 (pHH3; purplecolor). Bystander effect is denoted bythe prevalence of pHH3þ cells devoidof ADC binding.

Sung et al.

Mol Cancer Ther; 17(1) January 2018 Molecular Cancer Therapeutics248

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Page 7: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

Figure 3.

N87-TM cells are enriched for intracellular caveolar compartments and internalize trastuzumab-ADCs via caveolae-mediated endocytosis. A, Membranefractions from N87 and N87-TM cells were prepared for proteomic profiling. A volcano plot is shown representing the relative fold change for each protein(x-axis) by the significance level for each protein. Points in the upper right quadrant of the plot represent proteins enriched in N87-TM cells as compared with N87cells. B, Whole-cell lysates of N87 and N87-TM cells were prepared, separated by SDS-PAGE and immunoblotted for proteins identified in the proteomicprofiling to validate the hits. C,Whole-cell lysates from N87 and N87-TM cells were separated by SDS-PAGE and levels of total SRC, phospho-SRC, total CAV1, andphospho-CAV1 were determined by immunoblot analysis. D, CAV1þ puncta were identified via immunofluorescence staining of N87 and N87-TM cells. Average ofnumber of puncta calculated as total number of CAV1þ puncta normalized by number of nuclei for each field of view. E, N87 and N87-TM cells were treatedwith T-DM1 overnight at 37�C, fixed and co-immunostainedwith anti-human IgG (green color) and anti-CAV1 (red color). Image analysiswas performed to determineco-localization between T-DM1 and CAV1 in N87 and N87-TM cells. F, N87 and N87-TM cells were incubated with fluorescently labeled trastuzumab andkinetic co-localization analysis between T-DM1 and CAV1 and LAMP1 markers were performed via image cytometry.

Altered Trafficking as Mechanism of ADC Resistance

www.aacrjournals.org Mol Cancer Ther; 17(1) January 2018 249

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Page 8: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

fluorescent T-ADC monitoring ADC internalization intopHRodo-positive areas within the cell. As expected, T-ADCshowed increasingly higher co-localizationwith low pH compart-ments through the time course in N87 cells (Supplementary Fig.S7A, black line). However, the T-ADC did not traffic into low pHcompartments in N87-TM cells during the same time course(Supplementary Fig. S7A, red lines). Using the pH-insensitiveAlexaFluor signal intensity as a normalization factor, the distri-bution of T-ADC into low pH compartments was decreasedin N87-TM cells as compared with N87 cells (SupplementaryFig. S7B).

Caveolae-mediated ADC internalization is likely to be thepredominant mechanism by which T-ADCs enter N87-TM, butnot N87, cells. To determine whether we could induce caveolae-mediated ADC internalization in N87 cells, we transfectedCAV1-GFP into N87 cells and performed live-cell imaging.CAV1-GFP introduction resulted in the formation of similarintracellular CAV1þ puncta as N87-TM cells (SupplementaryFig. S8). Interestingly, T-ADC containing an AlexaFluor-mod-ified auristatin non-cleavable linker payload internalized intothe induced caveolar endocytic compartments in N87 cells(Supplementary Fig. S8).

These data suggest that T-ADC internalization in N87-TM cellsis mediated via caveolae endocytosis. We next hypothesized thatthis caveolae-mediated endocytosis drives resistance to T-DM1 inN87-TM cells. To determine whether CAV1 knockdown re-sensi-tized N87-TM cells to T-DM1, we performed doxycycline (DOX)-controlled shRNA-mediated CAV1 knockdown in N87-TM cells.Following DOX withdrawal, CAV1 targeted shRNAs wereexpressed and resulted in reduced CAV1 protein levels as com-pared with a non-targeted control sequence (Supplementary Fig.S9). However, CAV1 knockdown was not sufficient to re-sensitizeN87-TM cells to T-DM1 (Supplementary Fig. S9). Taken together,these data suggest that N87-TM cells are enriched for caveolarendocytic compartments capable of internalizing T-DM1 anddifferentially providing an alternative route for ADC endocytosisand trafficking.

Caveolae-mediated T-DM1 endocytosis across HER2þ cell linesCaveolae-mediated endocytosis has been implicated as a

possible mechanism of inherent insensitivity to ADCs targetingother antigens (e.g., melanotransferrin; ref. 24). We investigat-ed a panel of HER2þ cell lines to determine their inherentsensitivity to T-DM1 and whether CAV1 protein level correlatedwith the magnitude of T-DM1 response. We did not observe apositive correlation between CAV1 protein level and decreasedT-DM1 sensitivity in this panel of HER2þ cell lines (Fig. 4A).Given that high levels of CAV1 protein alone do not necessarilyinduce the formation of caveolar endocytic compartments (25),we next determined the propensity for caveolae-mediatedT-DM1 internalization across this cell line panel. Interestingly,some CAV1 high cell lines (e.g., JIMT1, N87-TM) showedcolocalization of T-DM1 and CAV1þ puncta whereas otherCAV1 high cell lines (e.g., SKOV3, HCC1954) did not (Fig.4B; similar data observed with T-vc-Aur0101, SupplementaryFig. S10). Intriguingly, the amount of ADC colocalization withCAV1þ puncta positively correlated with reduced response toT-DM1 across this cell line panel (Fig. 4C). Collectively, thesedata suggest that the inherent proclivity of tumor cells tointernalize T-DM1 via caveolae-mediated endocytosis may pre-dict their response to the ADC.

DiscussionIn this study, we generated and characterized multiple in vitro

models of T-DM1 resistance using a drug selection paradigmdesigned to mimic the clinical dosing regimen of T-DM1. Usingthis dosing schema, two models (BT474-TM and HCC1954-TM)acquired T-DM1 resistance via a previously reported mechanism(i.e., decreased HER2 expression) and another model (N87-TM)acquired T-DM1 resistance likely via a novel mechanism ofdifferential ADC internalization and trafficking. Herein, we reportthe first model of acquired T-DM1 resistance that implicatescaveolae-mediated endocytosis of T-DM1 as a novel mechanismof T-DM1 resistance.

Despite using the same approach to generate resistance, thesethree models and our two previously reported models of T-DM1resistance (13) highlight the diversity of molecular mechanismsthat cells may adopt to overcome the challenge of chronic drugexposure. Although the background genetics of each model maydictate the eventual mechanism of resistance to a drug, onemightexpect a myriad of changes that may impact a tumor's sensitivityto T-DM1 given the complexity of its mechanism of action. ForT-DM1 to effectively kill a tumor cell, itmust (i) bind toHER2; (ii)internalize into the intracellular portion of the cell; (iii) transitthrough the endosomalmaturation pathwaywithfinal delivery tothe lysosome; (iv) be catabolized within the lysosomal milieureleasing a membrane impermeable payload that is likely trans-ported out of the lysosome via a lysosomalmembrane transporterto interact with tubulin (the target of DM1). Indeed, there areexamples of potential alterations to some of these steps inexperimental models that may ultimately lead to T-DM1 resis-tance. As previously mentioned, decreased HER2 expressioncould prevent sufficient T-DM1 binding but other factors havebeen shown to influence trastuzumabbinding toHER2, includingthe accumulation of the truncated p95-HER2 isoform that lacks atrastuzumab binding site (26). Efficient internalization of T-DM1may not always be achieved because trastuzumab–HER2 com-plexes have been shown to have a rapid rate of plasmamembranerecycling once they are internalized (27). It remains to be seenwhether a similar phenomenon occurs with T-DM1-HER2 com-plexes. Lysosomal delivery of T-DM1 is another key event and theresults of this study show that alternative T-DM1 internalizationand trafficking parameters can influence lysosomal accumulationof T-DM1. The mechanism of lysosomal escape of lysine-MCC-DM1 (the released species of T-DM1) has long been debated butthe recent description of SLC46A3 as a lysosomal membranetransporter protein that mediates the lysosome-to-cytoplasmtransfer of this metabolite has provided insights into potentialmechanisms of transport (28).

Despite the complexity of these biomolecules, the modularnature of ADCs allows for the interchanging of various compo-nents to assess their influence on the properties of the ADC. TheT-DM1–resistant N87-TM cells were cross-resistant to trastuzu-mab-ADCs delivering other non-cleavable linked tubulin inhibi-tors. By switching the linker to an enzymatically cleavable linker,ADCs carrying other microtubule-targeting payloads (e.g., ValCit-linked auristatins) were able to overcome T-DM1 resistance inN87-TM cells. In a similar fashion, cell linemodelsmade resistantto CD22- or CD79b-targeting ADCs employing the vcMMAElinker-payload remained sensitive to the respective ADCs deliv-ering a different payload (e.g., PNU-159682; ref. 29). Thus, byswapping each module of an ADC with a different functional

Sung et al.

Mol Cancer Ther; 17(1) January 2018 Molecular Cancer Therapeutics250

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Page 9: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

group, ADC activity can be modulated. This "component switch-ing" property of ADCs implies that patients with ADC-refractorytumors may still derive benefit from a similar ADC, targeting thesame antigen, but with altered composition.

This report implicates caveolae-mediated endocytosis ofT-DM1 as a potential mechanism of acquired resistance, yet

trastuzumab-ADCs delivering tubulin inhibitors with an enzy-matically cleavable linker overcame T-DM1 resistance. It is impor-tant to note that caveolae contain enzymatically-active cysteinecathepsins capable of processing the "ValCit" linker utilizedherein (30). Thus, although the caveolar microenvironment maynot favor the antibody catabolism necessary to release the non-

Figure 4.

Caveolae-mediated endocytosis of T-DM1 predicts reduced sensitivity to T-DM1. A, A panel of breast and gastric cancer cell lines were evaluated for their relativeHER2 and CAV1 at basal state by immunoblot analysis. CAV1 score was assigned on the basis of basal CAV1 expression from this experiment. The samepanel of cell lines was tested for sensitivity to T-DM1 when treated for 4 days in a cell viability assay. The correlation of CAV1 score versus the T-DM1 IC50 valueis represented. B, The panel of cell lines were incubated with T-DM1 and analyzed for co-localization to CAV1þ puncta as previously described in Fig. 3.Representative images from each cell line show T-DM1 (green color) and CAV1 (red color) immunostaining. White arrows indicate examples of T-DM1 co-localizedwith CAV1þ puncta (yellow/orange color). C, The T-DM1 and CAV1 co-localization scores from (B) for each cell line were plotted against the IC50 value ofeach cell line to T-DM1. The correlation between T-DM1 and CAV1 colocalization score versus the T-DM1 IC50 value is represented.

Altered Trafficking as Mechanism of ADC Resistance

www.aacrjournals.org Mol Cancer Ther; 17(1) January 2018 251

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Page 10: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

cleavable linker-payload of T-DM1, it may be suitable to processenzymatically cleavable linkers. The impact of caveolae-mediatedendocytosis of other ADCs has been reported as well. Melano-transferrin- and CD133-targeting ADCs showed potent cytotoxicactivity in antigen-positive cell lines where the ADC preferentiallyaccumulated in lysosomes; however, the conjugateswerenot activein antigen-positive cell lines in which the ADC colocalized withcaveolin-1 (24, 31). Bothof theseADCsused the "vcMMAF" linker-payload which contains the same enzymatically cleavable dipep-tide linker used here but releases a different payload (monomethylauristatin F, MMAF). Because of its carboxylic acid functionalgroup, MMAF is a charged species at neutral pH; thus, althoughthe linker may be processed in caveolae, the charged nature ofMMAF renders it membrane impermeable and not able to enterinto the cytoplasmof the cell. It is this property that prevents ADCsusing MMAF from having substantial bystander effect (2). Alongsimilar lines, a CD20-targeting ADC using a "vcMMAE" linker wasreported to be internalized via both clathrin- and caveolae-medi-ated endocytosis; however, the nature of endocytosis did not affectthe sensitivity of antigen-positive cell lines to theADC (32). UnlikeMMAF, monomethyl auristatin E (MMAE) does not exist as achargedmolecule at neutral pH and ismembrane permeable. Dataherein implicate caveolae-mediated endocytosis in T-DM1 resis-tance; however, another group has suggested that caveolae-medi-ated endocytosismay sensitize cells to T-DM1 (33). Although thesedata implicate a possible role for CAV1, the inherent differences insensitivity to free DM1 (18) may explain the differences in basalsensitivity to T-DM1 seen across the highHER2 cell lines evaluatedin that study.

The functional studies in this report implicate a role forcaveolae-medicated endocytosis in the mechanism of resistanceto T-DM1 in N87-TM cells. However, CAV1 knockdown was notsufficient to re-sensitize N87-TM cells to T-DM1. IncompleteCAV1 protein loss may prevent re-sensitization, or as eluded topreviously, the formation of caveolae is not strictly dictated bythe level of CAV1 expression, but rather the interplay of caveolar-membrane coat proteins (caveolin family) interacting withcaveolar-membrane stabilizing scaffold proteins (cavin family;ref. 20). N87-TM cells are enriched for proteins from bothfamilies (i.e., CAV1 and PTRF). Thus, it may be that the driverof caveolae formation in N87-TM cells is a combination ofaltered proteins. Indeed, another caveolar coat protein may becompensating for the loss of CAV1 in the CAV1-knockdownN87-TM cells which, in conjunction with the enhanced expres-sion of PTRF, allows for the continued formation of caveolae.Along similar lines, CAV1 expression solely did not correlatewith T-DM1 sensitivity across a panel of HER2þ cell lines; rather,the functional aptitude for cells to internalize/traffic T-DM1 intocaveolar compartments correlated with T-DM1 sensitivity. Thefunction of caveolae in cancer biology remains largely unknown.However, this report adds to the growing body of literature thatsuggests caveolae may dictate sensitivity of cancer cells to ADCsthat use certain types of linker-payloads.

It has become increasingly clear that "precision medicine" willonly be as effective as our ability to definepatient sub-populationsthrough the use of reliable biomarkers. For many targeted ther-apies, such asADCs, theparamount biomarker is the expressionofthe target for that agent (e.g., HER2 for T-DM1). However, giventumoral genetic complexity and heterogeneity, target expressiondoes not solely predict a patient's tumor sensitivity to a targetedtherapy, particularly in solid tumor indications. For example, inthe EMILIA trial, only 43% of breast cancer patients with highHER2-expressing tumors responded to T-DM1 (11). Thus, therecontinues to be an unmet need in our ability to better identifypatients most likely to respond to T-DM1 and other targetedtherapies. Although alterations in some genes (e.g., EGFR, HER3,and PTEN protein expression and PIK3CA mutations; ref. 34)have been found to not correlate with T-DM1 response, data inthis report warrant the investigation of caveolae-mediated endo-cytosis of T-DM1 in patient tumors as a potential predictivebiomarker for response to T-DM1 and other non–cleavable-linked ADCs.

Disclosure of Potential Conflicts of InterestRussellDushin has ownership Interest (includingpatents) inPfizer Stock and

Stock Options. No other potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: M. Sung, R. Dushin, H.-P. Gerber, F. LoganzoDevelopment of methodology:M. Sung, X. Tan, J. Golas, C. Hosselet, F. Wang,L. Tylaska, L. King, D. Zhou, E. Rosfjord, J. Lucas, F. LoganzoAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.):M. Sung, X. Tan, B. Lu, J. Golas, C. Hosselet, L. Tylaska,L. King, J. LucasAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): M. Sung, B. Lu, F. Wang, L. Tylaska, J.S. Myers,E. Rosfjord, J. Lucas, F. LoganzoWriting, review, and/or revision of the manuscript: M. Sung, H.-P. Gerber,F. LoganzoAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases):M. Sung, F.Wang,D. Zhou, R.Dushin, J.S.MyersStudy supervision: M. Sung, J.S. Myers, J. Lucas, F. Loganzo

AcknowledgmentsThe authors appreciate the contributions of Pfizer Oncology colleagues

Elwira Muszynska, Nadira Prashad, Kiran Khandke, Manoj Charati, WilliamHu, My-Hanh Lam, Sylvia Musto, Xiang Zheng, Anton Xavier, Andreas Gian-nakou, Mike Cinque, Erik Upeslacis, AnnaMaria Barbuti, and Katherine Yao fortechnical assistance. The authors also thank Pfizer Medicinal Chemistry collea-gues Jeff Casavant, Zecheng Chen, Matthew Doroski, Gary Filzen, AndreasMaderna, Ludivine Moine, Alexander Porte, Hud Risley, and ChristopherO'Donnell for design and synthesis of the payloads, linker-payloads andconjugates used in this study.

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received May 5, 2017; revised September 7, 2017; accepted September 27,2017; published OnlineFirst October 20, 2017.

References1. Peng L, Chen X. Antibody-drug conjugates. Bioconjugate Chem 2015;26:

2169.2. Kim EG, Kim KM. Strategies and advancement in antibody-drug con-

jugate optimization for targeted cancer therapeutics. Biomol Thera2015;23:493–509.

3. Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancerdrug resistance: an evolving paradigm. Nat Rev Cancer 2013;13:714–26.

4. Engelman JA, Settleman J. Acquired resistance to tyrosine kinase inhibitorsduring cancer therapy. Curr Opin Genet Develop 2008;18:73–9.

Mol Cancer Ther; 17(1) January 2018 Molecular Cancer Therapeutics252

Sung et al.

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Page 11: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

5. Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinaseinhibitors. Nat Rev Cancer 2009;9:28–39.

6. de Jonge-Peeters SD, Kuipers F, de Vries EG, Vellenga E. ABC transporterexpression in hematopoietic stem cells and the role in AML drug resistance.Crit Rev Oncol Hematol 2007;62:214–26.

7. Fletcher JI,HaberM,HendersonMJ,NorrisMD.ABC transporters in cancer:more than just drug efflux pumps. Nat Rev Cancer 2010;10:147–56.

8. Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM.Targeting multidrug resistance in cancer. Nat Rev Drug Discov 2006;5:219–34.

9. Lauria F, Raspadori D, Rondelli D, VenturaMA, FiacchiniM, Visani G, et al.Highbcl-2 expression in acutemyeloid leukemia cells correlateswithCD34positivity and complete remission rate. Leukemia 1997;11:2075–8.

10. Lambert JM, Chari RV. Ado-trastuzumab Emtansine (T-DM1): an anti-body-drug conjugate (ADC) for HER2-positive breast cancer. J Med Chem2014;57:6949–64.

11. Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, et al. Trastu-zumab emtansine for HER2-positive advanced breast cancer. N Eng Med2012;367:1783–91.

12. Kang Y-K, ShahMA,Ohtsu A, VanCutsemE, Ajani JA, Van derHorst T, et al.A randomized, open-label, multicenter, adaptive phase 2/3 study oftrastuzumab emtansine (T-DM1) versus a taxane (TAX) in patients (pts)with previously treated HER2-positive locally advanced or metastaticgastric/gastroesophageal junction adenocarcinoma (LA/MGC/GEJC).[abstract]. J Clin Oncol 2016;34:5.

13. Loganzo F, Tan X, Sung M, Jin G, Myers JS, Melamud E, et al. Tumor cellschronically treated with a trastuzumab-maytansinoid antibody-drug con-jugate develop varied resistance mechanisms but respond to alternatetreatments. Mol Cancer Ther 2015;14:952–63.

14. Phillips G.Mechanisms of acquired resistance to Trastuzumab emtansine(T-DM1). World ADC Summit;2011 Oct 25-28: San Francisco, CA: Lon-don: Hanson Wade Limited.

15. Phillips GD, Fields CT, Li G, Dowbenko D, Schaefer G, Miller K, et al. Dualtargeting of HER2-positive cancer with trastuzumab emtansine and pertu-zumab: critical role for neuregulin blockade in antitumor response tocombination therapy. Clin Cancer Res 2014;20:456–68.

16. Erickson HK, Park PU, Widdison WC, Kovtun YV, Garrett LM, Hoffman K,et al. Antibody-maytansinoid conjugates are activated in targeted cancercells by lysosomal degradation and linker-dependent intracellular proces-sing. Cancer Res 2006;66:4426–33.

17. Chari RV.Targeted cancer therapy: conferring specificity to cytotoxic drugs.Accounts Chem Res 2008;41:98–107.

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

19. MadernaA,DoroskiM, SubramanyamC, Porte A, Leverett CA, Vetelino BC,et al. Discovery of cytotoxic dolastatin 10 analogues with N-terminalmodifications. J Med Chem 2014;57:10527–43.

20. KovtunO, Tillu VA,AriottiN, PartonRG,Collins BM.Cavin family proteinsand the assembly of caveolae. J Cell Sci 2015;128:1269–78.

21. Shajahan AN, Timblin BK, Sandoval R, Tiruppathi C, Malik AB, MinshallRD. Role of Src-induced dynamin-2 phosphorylation in caveolae-mediat-ed endocytosis in endothelial cells. J Biol Chem 2004;279:20392–400.

22. Parton RG, Simons K. Themultiple faces of caveolae. Nat RevMol Cell Biol2007;8:185–94.

23. Pelkmans L, Kartenbeck J,Helenius A. Caveolar endocytosis of simian virus40 reveals a new two-step vesicular-transport pathway to the ER. Nat CellBiol 2001;3:473–83.

24. Smith LM, Nesterova A, Alley SC, Torgov MY, Carter PJ. Potent cyto-toxicity of an auristatin-containing antibody-drug conjugate targetingmelanoma cells expressing melanotransferrin/p97. Mol Cancer Ther2006;5:1474–82.

25. SowaG, PypaertM, FultonD, SessaWC. Thephosphorylationof caveolin-2on serines 23 and36modulates caveolin-1-dependent caveolae formation.Proc Nat Acad Sci U S A 2003;100:6511–6.

26. Scaltriti M, Rojo F, Ocana A, Anido J, GuzmanM, Cortes J, et al. Expressionof p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer. J Nat Cancer Inst 2007;99:628–38.

27. Austin CD, De Maziere AM, Pisacane PI, van Dijk SM, Eigenbrot C,Sliwkowski MX, et al. Endocytosis and sorting of ErbB2 and the site ofactionof cancer therapeutics trastuzumab and geldanamycin.Mol BiolCell2004;15:5268–82.

28. Hamblett KJ, Jacob AP, Gurgel JL, Tometsko ME, Rock BM, Patel SK, et al.SLC46A3 is required to transport catabolites of noncleavable antibodymaytansine conjugates from the lysosome to the cytoplasm. Cancer Res2015;75:5329–40.

29. Yu SF, Zheng B, Go M, Lau J, Spencer S, Raab H, et al. A novel anti-CD22anthracycline-based antibody-drug conjugate (ADC) that overcomes resis-tance to auristatin-based ADCs. Clin Cancer Res 2015;21:3298–306.

30. Mohamed MM, Sloane BF. Cysteine cathepsins: multifunctional enzymesin cancer. Nat Rev Cancer 2006;6:764–75.

31. Smith LM, Nesterova A, Ryan MC, Duniho S, Jonas M, Anderson M, et al.CD133/prominin-1 is a potential therapeutic target for antibody-drugconjugates in hepatocellular and gastric cancers. Br J Cancer 2008;99:100–9.

32. Law CL, Cerveny CG, Gordon KA, Klussman K, Mixan BJ, Chace DF, et al.Efficient elimination of B-lineage lymphomas by anti-CD20-auristatinconjugates. Clin Cancer Res 2004;10:7842–51.

33. Chung YC, Kuo JF, Wei WC, Chang KJ, Chao WT. Caveolin-1 dependentendocytosis enhances the chemosensitivity of HER-2 positive breastcancer cells to trastuzumab emtansine (T-DM1). PloS One 2015;10:e0133072.

34. Baselga J, Lewis Phillips GD, Verma S, Ro J, Huober J, Guardino AE, et al.Relationship between tumor biomarkers and efficacy in EMILIA, a phase IIIstudy of trastuzumab emtansine inHER2-positivemetastatic breast cancer.Clin Cancer Res 2016;22:3755–63.

www.aacrjournals.org Mol Cancer Ther; 17(1) January 2018 253

Altered Trafficking as Mechanism of ADC Resistance

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

Page 12: Caveolae-Mediated Endocytosis as a Novel Mechanism of … · Cancer Biology and Translational Studies Caveolae-Mediated Endocytosis as a Novel Mechanism of Resistance to Trastuzumab

2018;17:243-253. Published OnlineFirst October 20, 2017.Mol Cancer Ther   Matthew Sung, Xingzhi Tan, Bingwen Lu, et al.   Resistance to Trastuzumab Emtansine (T-DM1)Caveolae-Mediated Endocytosis as a Novel Mechanism of

  Updated version

  10.1158/1535-7163.MCT-17-0403doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2017/10/20/1535-7163.MCT-17-0403.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/17/1/243.full#ref-list-1

This article cites 33 articles, 13 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/17/1/243.full#related-urls

This article has been cited by 10 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/17/1/243To request permission to re-use all or part of this article, use this link

on February 9, 2021. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403