32
1 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 , Frank Loganzo 1 1 Pfizer Inc., Oncology Research & Development, Pearl River, NY 2 Pfizer Inc., Biomedicine Design, Groton, CT 3 Pfizer Inc., Worldwide Medicinal Chemistry, Groton, CT 4 Maverick Therapeutics Inc., Brisbane, CA Running title: Altered trafficking as mechanism of ADC resistance Keywords: T-DM1, Drug Resistance, Biomarker, Caveolin-1, Antibody-Drug Conjugate Abbreviations: T-DM1, trastuzumab emtansine; ADC, antibody-drug conjugate; T, trastuzumab; T-ADC, trastuzumab antibody-drug conjugate; mc, maleimidocaproyl; vc, mcValCitPABC; N87-TM, T-DM1-resistant N87 cells; CAV1, caveolin-1; Aur0101, auristatin analog; Aur8261, auristatin analog; mg/kg, milligrams per kilogram; Financial Support: No public funds were used in support of this work. Corresponding author: Matthew Sung, Pfizer Oncology, 401 North Middletown Road, 200/4714A, Pearl River, NY 10965. Phone: 845-602-3731; Email: [email protected] on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

1 2 3 1 , Hans-Peter Gerber , Frank Loganzo...2017/10/20  · 1 Caveolae-mediated endocytosis as a novel mechanism of resistance to trastuzumab emtansine (T-DM1) Matthew Sung1, Xingzhi

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

  • 1

    Caveolae-mediated endocytosis as a novel mechanism of resistance to trastuzumab emtansine (T-DM1)

    Matthew Sung1, Xingzhi Tan1, Bingwen Lu1, Jonathan Golas1, Christine Hosselet1, Fang Wang1, Laurie

    Tylaska2, Lindsay King2, Dahui Zhou3, Russell Dushin3, Jeremy S. Myers1, Edward Rosfjord1, Judy

    Lucas1, Hans-Peter Gerber4, Frank Loganzo1

    1Pfizer Inc., Oncology Research & Development, Pearl River, NY 2Pfizer Inc., Biomedicine Design, Groton, CT 3Pfizer Inc., Worldwide Medicinal Chemistry, Groton, CT 4Maverick Therapeutics Inc., Brisbane, CA

    Running title:

    Altered trafficking as mechanism of ADC resistance

    Keywords:

    T-DM1, Drug Resistance, Biomarker, Caveolin-1, Antibody-Drug Conjugate

    Abbreviations:

    T-DM1, trastuzumab emtansine;

    ADC, antibody-drug conjugate;

    T, trastuzumab;

    T-ADC, trastuzumab antibody-drug conjugate;

    mc, maleimidocaproyl;

    vc, mcValCitPABC;

    N87-TM, T-DM1-resistant N87 cells;

    CAV1, caveolin-1;

    Aur0101, auristatin analog;

    Aur8261, auristatin analog;

    mg/kg, milligrams per kilogram;

    Financial Support:

    No public funds were used in support of this work.

    Corresponding author:

    Matthew Sung, Pfizer Oncology, 401 North Middletown Road, 200/4714A, Pearl River, NY 10965.

    Phone: 845-602-3731; Email: [email protected]

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    mailto:[email protected]://mct.aacrjournals.org/

  • 2

    ABSTRACT

    Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate (ADC) that has demonstrated clinical

    benefit for patients with HER2+ 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+ tumors, are not well understood. We used HER2+ cell lines to develop models of T-

    DM1 resistance utilizing a cyclical 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 a panel of trastuzumab-ADCs (T-ADCs) with non-cleavable-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 to parental N87 cells. Intriguingly, T-ADCs utilizing 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 which remain sensitive to T-

    ADCs with cleavable-linked auristatin payloads. Comparative proteomic profiling suggested enrichment

    in proteins 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 trafficking to the

    lysosome compared to N87 cells. T-DM1 colocalization into intracellular CAV1 positive puncta

    correlated with reduced response to T-DM1 in a panel of HER2+ cell lines. Together, these data suggest

    caveolae-mediated endocytosis of T-DM1 may serve as a novel predictive biomarker for patient response

    to T-DM1.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 3

    INTRODUCTION

    Antibody-drug conjugates (ADCs) are emerging therapeutic modalities strategically designed to

    selectively deliver chemotherapeutic 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 2000’s, three ADCs have received FDA-approval and over 50 more are currently

    in clinical development (1,2).

    Despite encouraging signs of early clinical benefit, the sustained efficacy of many targeted anti-

    cancer therapies is limited by either inherent or acquired tumoral drug resistance. The molecular

    mechanisms that drive clinical drug resistance are multi-factorial and ultimately depend upon the

    therapeutic modality and cancer indication (3). Modifications of the drug target (e.g. kinase mutations)

    (4,5), drug efflux protein expression (e.g. ABCB1 in AML) (6-8), and shifts in the expression ratio of

    pro- versus anti-apoptotic proteins (e.g. BCL-2 expression in AML) (9) are examples of tumor alterations

    that correlate with reduced clinical response to anti-cancer therapies. However, the molecular

    mechanisms that drive clinical resistance to ADCs remain largely unknown.

    Trastuzumab emtansine (T-DM1, Kadcyla®) currently represents the only FDA-approved ADC

    for the treatment of a solid tumor indication. T-DM1 is composed of the trastuzumab antibody

    chemically bound to the microtubule-disrupting DM1 maytansinoid via a thioether linker (10). In the

    phase III EMILIA clinical trial, T-DM1 showed significantly improved progression free and overall

    survival versus lapatinib plus capecitabine in HER2+ metastatic breast cancer patients who had failed

    previous treatment with trastuzumab and a taxane (11). The overall response rate for HER2+ patients

    treated with T-DM1 was 43.6% (11). On the basis of these results, T-DM1 was granted FDA approval for

    treatment of this patient population. However, these data showed that, despite high tumoral HER2-

    positivity, over 50% of patients treated with T-DM1 did not show clinical benefit. In the phase II/III

    GATSBY clinical trial in HER2+ gastric cancer patients, T-DM1 did not show enhanced clinical response

    versus standard chemotherapy (12). Thus, HER2 expression does not solely predict clinical response to

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 4

    T-DM1 and the need exists to more accurately define the patient sub-population most likely to respond to

    T-DM1.

    While data are still emerging from T-DM1-treated patient samples, multiple pre-clinical models

    of T-DM1 resistance have been generated to characterize the changes at the tumor cell level that may

    mediate clinical resistance to T-DM1. While the models of T-DM1 resistance in these studies were

    generated with different dosing schemas, the following are mechanisms responsible for T-DM1 resistance

    seen across multiple models: (1) decreased HER2 expression (13,14), (2) increased expression and

    activity of drug efflux proteins (13,14), and (3) increased heregulin expression (15).

    Herein, we used an ADC structurally similar to T-DM1 (13) to generate T-DM1 resistant cells

    in vitro. We report the generation and characterization of three models of T-DM1 resistance, including

    one with a unique trafficking defect that has not been previously associated with T-DM1 acquired

    resistance and may provide a novel biomarker for clinical response to T-DM1.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 5

    MATERIALS & METHODS

    Cell Lines

    NCI-N87 gastric carcinoma, HCC1954 breast carcinoma, and BT474 breast carcinoma cells were

    obtained from ATCC (Manassas, VA) and maintained in RPMI media supplemented with 10% fetal

    bovine serum (FBS), 1% L-glutamine, and 1% sodium pyruvate. H69 and H69AR cell lines were

    obtained from ATCC and were maintained in RPMI/25mM HEPES media supplemented with 10% FBS,

    1% L-glutamine, and 1% sodium pyruvate. KB, KB8.5, and KBVI cells were generously provided by Dr.

    Michael Gottesman (National Cancer Institute). Parental N87 cells were purchased from ATCC (CRL-

    5822, lot 58033347) in June 2010. N87 and N87-TM cells were authenticated as NCI-N87 in 2014

    (IDEXX BioResearch, Columbia, MO), when the cells were being characterized for this work.

    Mycoplasma testing was conducted approximately quarterly using MycoAlert (Lonza, Allendale, NJ), and

    results were consistently negative; the most recent testing was conducted in October 2016.

    Bioconjugations

    ADCs and chemical structures of disclosed payloads described herein were prepared as previously

    described (13). The syntheses and structures of the novel auristatin linker-payloads as well as preparation

    and characterization of ADCs presented herein are described in the supplementary methods.

    Generation of T-DM1 Resistant Cells

    Cells were passaged into two separate flasks and each flask was treated identically with respect to the

    resistance-generation protocol to enable biological duplicates. Cells were exposed to five cycles of T-

    DM1 conjugate at 10-fold IC50 concentrations (10 nmol/L payload concentration; ~388 ng/mL antibody

    concentration) for 3 days, followed by approximately 4 to 11 days recovery without treatment. After five

    cycles at 10 nmol/L of the T-DM1 conjugate, the cells were exposed to six additional cycles of 100

    nmol/L T-DM1 in a similar fashion. The procedure was intended to simulate the chronic, multi-cycle

    (on/off) dosing at maximally tolerated doses typically used for cytotoxic therapeutics in the clinic,

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 6

    followed by a recovery period. Parental cells derived from NCI-N87 are referred to as N87, and cells

    chronically exposed to T-DM1 are referred to as N87-TM. Moderate- to high-level drug resistance

    developed within 4 months for N87-TM cells. Drug selection pressure was removed after ~3 - 4 months

    of cycle treatments when the level of resistance no longer increased after continued drug exposure.

    Responses and phenotypes remained stable in the cultured cell lines for approximately 3 – 6 months.

    Thereafter, a reduction in the magnitude of the resistance phenotype as measured by cytotoxicity assays

    was occasionally observed, in which case early passage cryo-preserved T-DM1 resistant cells were

    thawed for additional studies. All reported characterizations were conducted after removal of T-DM1

    selection pressure for at least 2 - 8 weeks to ensure stabilization of the cells. Data were collected from

    various thawed cryopreserved populations derived from a single selection, over approximately 1 – 2 years

    after model development to ensure consistency in the results. Clonal populations derived from single

    N87-TM cells using a limited dilution technique. Briefly, single cells were plated into each well of a 96-

    well plate and cultured in the presence of 100 nmol/L T-DM1 until clonal populations were established.

    N87 and N87-TM cells were genetically authenticated in 2014 (Idexx Bioresearch, Columbia, MO).

    Cytotoxicity Assay

    Cellular responses to ADCs and free drugs were measured with a cytotoxicity assay as previously

    described (13). Briefly, cells were seeded into 96-well plates and treated with varying concentrations of

    ADCs or free drugs for 96 hours. Cell viability was measured using CellTiter® 96 AQueous One MTS

    Solution (Promega, Madison, WI). IC50 values were calculated using a four-parameter logistic model

    with XLfit (IDBS, Bridgewater, NJ).

    Proteomic profiling and immunoblot analyses

    Proteomic profiling of membrane fractions was prepared and analyzed as previously described (13).

    Whole cell lysates were prepared in RIPA buffer supplemented with protease inhibitor cocktail (Sigma,

    St. Louis, MO). Lysates were fractionated on 4-20% SDS-PAGE gels (Bio-Rad, Hercules, CA),

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 7

    transferred onto membranes and probed with primary and secondary antibodies further detailed in the

    supplementary methods.

    Immunofluorescence and Live-Cell Imaging

    For immunofluorescence experiments, cells were seeded onto #1.5 coverslips (Electron Microscopy

    Services, Hatfield, PA) and fixed with 4% paraformaldehyde. Cells were stained with primary antibodies

    at room temperature (RT) for 1 hour and secondary antibodies at RT for 30 min. Coverslips were

    mounted onto slides with ProLong® Diamond Antifade Mountant (ThermoFisher Scientific). Images

    were acquired with a Zeiss LSM 710 (Carl Zeiss Inc., Thornwood, NY). For imaging cytometry, cells

    were incubated with PE-labeled trastuzumab (2 µg/mL) on ice for 90 minutes or at 37°C for time points

    indicated. At designated time points, internalization was stopped by addition of cold PBS and cells were

    fixed with Cytofix/Cytoperm solution (BD Biosciences) for 20 minutes. Cells were immunostained with

    LAMP1-AF647 (BD Biosciences; cat# 562622) for 30 minutes or CAV1 antibody for 30 minutes

    followed by an anti-rabbit IgG Fab2 AF647 secondary antibody (ThermoFisher Scientific; cat# A-21246)

    for 30 minutes. Cells were analyzed on the ImageStreamX Mark II Imaging Flow Cytometer (AMNIS,

    EMD Millipore, Seattle, WA). For live-cell imaging experiments, cells were seeded into 4-well

    CELLview dishes (Grenier Bio-One, Austria) and imaged with an ImagEM camera (Hamamatsu

    Photonics K.K., Hamamatsu City, Japan) mounted onto a CSU-X1 spinning disk head (Yokogawa

    Electric Corporation, Inc., Tokyo, Japan) on an Eclipse Ti microscope stand (Nikon Instruments Inc.,

    Melville, NY) equipped with a 60x oil objective (NA1.4, Nikon Instruments Inc.). Image analysis was

    performed using Volocity 3D Image software (PerkinElmer, Waltham, MA).

    In vivo efficacy studies

    All procedures performed on animals in this study were in accordance with established guidelines and

    regulations, and were reviewed and approved by the Pfizer Institutional Animal Care and Use Committee.

    Pfizer animal care facilities that supported this work are fully accredited by AAALAC International.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 8

    Female NOD scid gamma (NSG) immunodeficient mice (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) were

    obtained from The Jackson Laboratory (Bar Harbor, ME). Mice were injected subcutaneously in the right

    flank with suspensions of either N87 or N87-TM cells (7.5 x 106 cells per injection, with 50% Matrigel).

    Mice were randomized into study groups when tumors reached ~0.3 g (~250 mm3). ADCs or vehicle

    were administered intravenously in saline on day 0 and repeated for a total of four doses, four days apart

    (q4d x 4). Tumors were measured with calipers weekly and tumor mass calculated as volume = (width x

    width x length)/2.

    Immunohistochemistry

    Five micrometer thick formalin fixed, paraffin embedded tissue sections were stained

    immunohistochemically. Detailed sample preparations and staining procedures are provided in the

    supplementary methods.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 9

    RESULTS

    Generation and Characterization of T-DM1 Resistant Cells

    To identify potential mechanisms that may drive clinical resistance to T-DM1 ADC, we

    generated T-DM1 resistant versions of the HER2-expressing cancer cell lines N87, BT474 and HCC1954.

    We utilized a cyclical dosing schema of a 3-day exposure to high dose T-DM1 ADC followed by a

    washout and recovery period to mimic patient exposure and pharmacokinetics when they receive bolus-

    chemotherapy treatment cycles (Figure 1A).

    N87 cells were inherently sensitive to T-DM1 (IC50 = 1.6 nmol/L payload concentration; 62 ng/ml

    antibody concentration) (Figure 1B). Two populations of N87 cells were exposed to the treatment cycles

    and, after approximately four months of cyclical dosing, these two populations (henceforth named N87-

    TM-1 and N87-TM-2) became refractory to the ADC by 109-fold compared with parental N87 cells

    (Figure 1B, Table 1). Interestingly, minimal cross-resistance (1.8x) to the corresponding unconjugated

    maytansinoid free drug, DM1-SMe, was observed (Supplementary Figure S1, Table 1) which suggests the

    alterations in N87-TM cells that lead to T-DM1 resistance are specific to the whole ADC biomolecule and

    not solely to the payload.

    Previous reports of T-DM1 resistance in other cell models have implicated ATP-binding cassette

    (ABC) transporter protein overexpression (e.g. ABCB1/MDR1, ABCC1/MRP1) or target antigen down-

    regulation as key drivers of T-DM1 resistance (13,14). However, proteomics and immunoblots

    demonstrate that N87-TM cells do not overexpress MDR1 (Figure 1C) or MRP1 (Figure 1D) and retain

    similar levels of HER2 as compared to parental N87 cells (Figure 1E). Despite retaining high levels of

    HER2, N87-TM cells display ~50% reduced binding to the unconjugated antibody of T-DM1 (i.e.

    trastuzumab) as measured by flow cytometry (Figure S2). In contrast, T-DM1 resistant populations of

    BT474 (BT474-TM) and HCC1954 (HCC1954-TM), selected in a similar manner as N87-TM, show

    significantly decreased HER2 expression (Supplementary Figures S3A & S3B), suggesting that reduced

    antigen is the primary mode of resistance in these two cell models. Hence, we focused additional

    characterization on the N87-TM model.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 10

    Taking advantage of the modular nature of ADCs, we hypothesized that altering key components

    of the biomolecule would result in an agent able to overcome T-DM1 resistance in N87-TM cells. N87-

    TM cells displayed cross-resistance to several trastuzumab-based ADCs (T-ADCs) (Figure 1F and Table

    1) which were conjugated to auristatins via a non-cleavable maleimide linker. Remarkably, N87-TM

    were sensitive to T-ADCs conjugated to auristatins utilizing a protease-cleavable linker sequence (ValCit-

    PAB) in an antigen-dependent manner (Figure 1G, Supplementary Figure S4 and Table 1, Supplementary

    Table S1). The sensitivity of N87-TM cells to other trastuzumab-based ADCs suggests that the reduced

    level of HER2 in these cells is not a mode of resistance.

    To determine if the ADC-resistant N87-TM cells were broadly 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, vinblastine). Importantly, N87-TM cells remained sensitive to these small molecule inhibitors

    when compared to the parental N87 cells, discounting general growth defects as the mechanism of

    resistance to T-DM1 ADC (Supplementary Figure S4, Table 1).

    N87-TM tumors maintain T-DM1 resistance in vivo

    A problem that often plagues in vitro drug resistance models is their lack of translatability into the

    in vivo setting. In order to determine if the resistance observed in cell culture was recapitulated in vivo,

    N87 cells and N87-TM cells were injected into the flanks of female NOD scid gamma (NSG)

    immunodeficient mice and treated with T-DM1 ADC (at doses of 6 mg/kg (mpk) and 10 mpk) and a T-

    ADC with a cleavable-linked auristatin (T-vc-Aur0101) (at 3 mpk), on a q4d X 4 schedule. The N87

    tumors retained high expression of HER2 in vivo and were quite sensitive to all three T-ADC treatment

    conditions (Figure 2A). Remarkably, N87-TM tumors also retained high expression of HER2 in vivo and

    were refractory to both regimens of T-DM1 ADC, yet tumors regressed with T-vc-Aur0101 (Figure 2B).

    Thus, N87-TM tumors retain a similar response profile to T-DM1 and T-vc-Aur0101 ADCs in vitro and

    in vivo.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 11

    In order to better understand how N87-TM tumors are resistant to T-DM1 and sensitive to T-vc-

    Aur0101, we first interrogated the tumoral distribution of the ADCs. N87-TM tumors, from the

    previously described efficacy study, were collected 24 hours following the last ADC dose, and ADC

    distribution was evaluated through immunohistochemistry staining with an anti-human IgG antibody.

    N87-TM tumors treated with 6 mpk T-DM1 ADC displayed almost complete tumor coverage of IgG

    binding, whereas those treated with 3 mpk T-vc-Aur0101 only had ADC binding at the most vessel-

    proximal regions of the tumor (Supplementary Figure S5). Thus, tumoral ADC distribution is unlikely to

    explain the efficacy response of these ADCs in N87-TM tumors.

    Next, we evaluated a biomarker of downstream response to an anti-microtubule targeted agent to

    determine how T-DM1 and T-vc-Aur0101 ADCs differentially impacted N87 and N87-TM tumors.

    Phosphorylation of histone H3 (pHH3) is a marker of mitotic cells and can be used to evaluate the activity

    of anti-mitotic agents. To determine if N87-TM tumors had reduced pHH3 staining following T-DM1

    ADC treatment as compared to N87 tumors, mice were administered a single-dose of T-DM1 ADC (6

    mpk) and tumors were collected 24 hours later. Co-immunostaining revealed ADC and pHH3 double

    positive cells in the N87 tumors (Figure 2C, top left panel), whereas the majority of the ADC-bound N87-

    TM tumor cells were pHH3 negative (Figure 2C, bottom left panel). These data are consistent with the

    efficacy data showing N87, but not N87-TM, tumors are sensitive to T-DM1 ADC. In a similar study

    conducted with T-vc-Aur0101 ADC (3 mpk), N87 and N87-TM tumors had ADC and pHH3 double

    positive cells (Figure 2C, top and bottom right panels, respectively). Intriguingly, both T-vc-Aur0101

    ADC treated tumors display many pHH3 positive cells without ADC-bound to them, a property known as

    bystander effect whereby antigen negative tumor cells neighboring antigen positive tumor cells are

    susceptible to membrane permeable payloads. Thus, it is clear from this in vivo biomarker study that T-

    ADCs utilizing a cleavable linker that releases a membrane permeable cytotoxic payload can overcome T-

    DM1 resistance in N87-TM tumors.

    The active released species from a catabolized T-DM1 molecule is the amino acid capped linker

    payload, Lysine-MCC-DM1 (16,17). The structural and ionic nature of Lysine-MCC-DM1 in the tumor

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 12

    microenvironment renders the molecule membrane impermeable and is likely why T-DM1 does not show

    strong bystander effect (18). To determine if amino-acid capping of the released ADC species dictates

    differential ADC cytotoxic response in N87-TM as compared to N87 cells, we prepared a series of ADCs

    utilizing the auristatin analog (Aur8261) conjugated to the same targeting antibody (trastuzumab) via

    different linkers (19). As shown above, N87-TM cells are cross-resistant to a T-ADC with a non-

    cleavable-linked payload with a released species of cysteine-mc-Aur8261. However, N87-TM cells are

    sensitive to a T-ADC with Aur8261 attached via a cleavable linker where the released species is the

    unmodified, membrane-permeable auristatin compound. N87-TM cells were cross-resistant to a T-ADC

    with a cleavable linker but with a released species of cysteine-mc-Aur8261 (Supplementary Figure S6).

    Thus, T-ADCs whose active catabolite is an amino-capped linker payload are effective in killing N87, but

    not N87-TM, cells. Taken together, these data show that N87-TM tumors maintain T-DM1 resistance in

    vivo and T-ADCs with cleavable linkers and permeable payloads overcome T-DM1 resistance in vitro and

    in vivo.

    The role of CAV1 in ADC biology and resistance

    To further characterize the mechanism of T-DM1 resistance in N87-TM cells in an unbiased

    approach, we performed a proteomic evaluation of membrane fractions to profile proteins differentially

    expressed in the N87-TM cells as compared to the parental N87 cells. Significant changes in expression

    level of 523 proteins between the cell lines were observed (Figure 3A). Immunoblot analysis validated a

    number of the proteomic hits (Figure 3B). Of interest was the over-expression of caveolin-1 (CAV1) and

    polymerase I and transcript release factor (PTRF) in N87-TM cells. As CAV1 and PTRF are essential

    protein constituents of caveolae (20), we hypothesized that N87-TM cells are enriched for caveolar

    endocytic compartments. Caveolae-mediated endocytosis is thought to be induced by Src-mediated

    phosphyloration of CAV1 at Tyr 14 (21). Indeed, N87-TM cells have enhanced phospho-Src and

    phospho-CAV1 as compared to N87 cells (Figure 3C). Immunofluorescence for CAV1 in N87 and N87-

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 13

    TM cells demonstrates that N87-TM cells contain many more intracellular CAV1+ puncta than N87 cells

    (Figure 3D).

    We next hypothesized that N87-TM cells can internalize T-DM1 into these caveolar endocytic

    compartments. We treated N87 and N87-TM cells with T-DM1 for 24 hours and co-immunostained for

    anti-human IgG and CAV1 to interrogate the co-localization of the ADC with CAV1+ puncta. Indeed, T-

    DM1 ADC co-localized with CAV1 in N87-TM, but not N87, cells (Figure 3E). To determine if this

    positive correlation between T-DM1 ADC and CAV1 occurred at the early phases of ADC endocytosis,

    we analyzed trastuzumab and CAV1 co-localization at multiple early time points using imaging

    cytometry. N87-TM, but not N87, cells showed enhanced trastuzumab and CAV1 co-localization over

    time (Figure 3F). Since the internalization of trastuzumab was into differential compartments in N87 and

    N87-TM cells, we next sought to determine if N87-TM cells displayed impaired lysosomal trafficking of

    trastuzumab because T-DM1 is catabolized in the lysosome. Lysosomal co-localization of trastuzumab

    was decreased in N87-TM as compared to N87 cells (Figure 3F).

    Caveolar endocytic compartments are phenotypically distinct from the endo-lysosomal pathway

    (22). For example, the lumina of caveolar endocytic compartments have a neutral pH, whereas the

    lumina of endo-lysosomal compartments are increasingly acidic during pathway maturation (23). To

    determine if T-ADCs were internalized into compartments with different pH, we generated a T-ADC

    conjugated to two fluorescent moieties: (1) a pH-insensitive AlexaFluor-modified auristatin non-

    cleavable linker payload and (2) a pH-sensitive dye, pHRodo, whereby the fluorescence signal is

    proportional to the acidity of the micro-environment. We performed live-cell imaging with this

    fluorescent T-ADC monitoring ADC internalization into pHRodo-positive areas within the cell. As

    expected, T-ADC showed increasingly higher co-localization with low pH compartments through the

    time course in N87 cells (Supplementary Figure S7A, black line). However, the T-ADC did not traffic

    into low pH compartments in N87-TM cells during the same time course (Supplementary Figure S7A, red

    lines). Using the pH-insensitive AlexaFluor signal intensity as a normalization factor, the distribution of

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 14

    T-ADC into low pH compartments was decreased in N87-TM cells as compared to N87 cells

    (Supplementary Figure S7B).

    Caveolae-mediated ADC internalization is likely to be the predominant mechanism by which T-

    ADCs enter N87-TM, but not N87, cells. To determine if we could induce caveolae-mediated ADC

    internalization in N87 cells, we transfected CAV1-GFP into N87 cells and performed live-cell imaging.

    CAV1-GFP introduction resulted in the formation of similar intracellular CAV1+ puncta as N87-TM cells

    (Supplementary Figure S8). Interestingly, T-ADC containing an AlexaFluor-modified auristatin non-

    cleavable linker payload internalized into the induced caveolar endocytic compartments in N87 cells

    (Supplementary Figure S8).

    These data suggest T-ADC internalization in N87-TM cells is mediated via caveolae endocytosis.

    We next hypothesized that this caveolae-mediated endocytosis drives resistance to T-DM1 in N87-TM

    cells. To determine if CAV1 knockdown re-sensitized 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 were expressed and resulted in reduced CAV1 protein levels as

    compared to a non-targeted control sequence (Supplementary Figure S9). However, CAV1 knockdown

    was not sufficient to re-sensitize N87-TM cells to T-DM1 (Supplementary Figure S9). Taken together,

    these data suggest that N87-TM cells are enriched for caveolar endocytic compartments capable of

    internalizing T-DM1 and differentially providing an alternative route for ADC endocytosis and

    trafficking.

    Caveolae-mediated T-DM1 endocytosis across HER2+ cell lines

    Caveolae-mediated endocytosis has been implicated as a possible mechanism of inherent

    insensitivity to ADCs targeting other antigens (e.g. melanotransferrin) (24). We investigated a panel of

    HER2+ cell lines to determine their inherent sensitivity to T-DM1 and whether CAV1 protein level

    correlated with the magnitude of T-DM1 response. We did not observe a positive correlation between

    CAV1 protein level and decreased T-DM1 sensitivity in this panel of HER2+ cell lines (Figure 4A).

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 15

    Given that high levels of CAV1 protein alone do not necessarily induce the formation of caveolar

    endocytic compartments (25), we next determined the propensity for caveolae-mediated T-DM1

    internalization across this cell line panel. Interestingly, some CAV1 high cell lines (e.g. JIMT1, N87-

    TM) showed colocalization of T-DM1 and CAV1+ puncta whereas other CAV1 high cell lines (e.g.

    SKOV3, HCC1954) did not (Figure 4B; similar data observed with T-vc-Aur0101, Supplementary Figure

    S10). Intriguingly, the amount of ADC colocalization with CAV1+ puncta positively correlated with

    reduced response to T-DM1 across this cell line panel (Figure 4C). Collectively, these data suggest that

    the inherent proclivity of tumor cells to internalize T-DM1 via caveolae-mediated endocytosis may

    predict their response to the ADC.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 16

    DISCUSSION

    In this study, we generated and characterized multiple in vitro models of T-DM1 resistance

    utilizing a drug selection paradigm designed to mimic the clinical dosing regimen of T-DM1. Using this

    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 of differential ADC internalization and trafficking. Herein, we

    report the first model of acquired T-DM1 resistance that implicates caveolae-mediated endocytosis of T-

    DM1 as a novel mechanism of T-DM1 resistance.

    Despite using the same approach to generate resistance, these three models and our two

    previously reported models of T-DM1 resistance (13) highlight the diversity of molecular mechanisms

    that cells may adopt to overcome the challenge of chronic drug exposure. While the background genetics

    of each model may dictate the eventual mechanism of resistance to a drug, one might expect a myriad of

    changes that may impact a tumor’s sensitivity to T-DM1 given the complexity of its mechanism of action.

    For T-DM1 to effectively kill a tumor cell, it must (1) bind to HER2; (2) internalize into the intracellular

    portion of the cell; (3) transit through the endosomal maturation pathway with final delivery to the

    lysosome; (4) be catabolized within the lysosomal milieu releasing a membrane impermeable payload that

    is likely transported out of the lysosome via a lysosomal membrane transporter to interact with tubulin

    (the target of DM1). Indeed, there are examples of potential alterations to some of these steps in

    experimental models that may ultimately lead to T-DM1 resistance. As previously mentioned, decreased

    HER2 expression could prevent sufficient T-DM1 binding but other factors have been shown to influence

    trastuzumab binding to HER2, including the accumulation of the truncated p95-HER2 isoform that lacks

    a trastuzumab binding site (26). Efficient internalization of T-DM1 may not always be achieved since

    trastuzumab-HER2 complexes have been shown to have a rapid rate of plasma membrane recycling once

    they are internalized (27). It remains to be seen if a similar phenomenon occurs with T-DM1-HER2

    complexes. Lysosomal delivery of T-DM1 is another key event and the results of this study show that

    alternative T-DM1 internalization and trafficking parameters can influence lysosomal accumulation of T-

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 17

    DM1. The mechanism of lysosomal escape of lysine-MCC-DM1 (the released species of T-DM1) has

    long been debated but the recent description of SLC46A3 as a lysosomal membrane transporter protein

    that mediates the lysosome-to-cytoplasm transfer of this metabolite has provided insights into potential

    mechanisms of transport (28).

    Despite the complexity of these biomolecules, the modular nature of ADCs allows for the

    interchanging of various components to assess their influence on the properties of the ADC. The T-DM1-

    resistant N87-TM cells were cross-resistant to trastuzumab-ADCs delivering other non-cleavable linked

    tubulin inhibitors. 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

    in N87-TM cells. In a similar fashion, cell line models made resistant to CD22- or CD79b-targeting

    ADCs employing the vcMMAE linker-payload remained sensitive to the respective ADCs delivering a

    different payload (e.g. PNU-159682) (29). Thus, by swapping each module of an ADC with a different

    functional group, ADC activity can be modulated. This “component switching” property of ADCs

    implies that patients with ADC-refractory tumors may still derive benefit from a similar ADC, targeting

    the same antigen, but with altered composition.

    This report implicates caveolae-mediated endocytosis of T-DM1 as a potential mechanism of

    acquired resistance, yet trastuzumab-ADCs delivering tubulin inhibitors with an enzymatically cleavable

    linker overcame T-DM1 resistance. It is important to note that caveolae contain enzymatically-active

    cysteine cathepsins capable of processing the ‘ValCit’ linker utilized herein (30). Thus, while the

    caveolar microenvironment may not favor the antibody catabolism necessary to release the non-cleavable

    linker-payload of T-DM1, it may be suitable to process enzymatically cleavable linkers. Thus, while the

    caveolar microenvironment may not favor the antibody catabolism necessary to release the non-cleavable

    linker-payload of T-DM1, it may be suitable to process enzymatically cleavable linkers. The impact of

    caveolae-mediated endocytosis of other ADCs has been reported as well. Melanotransferrin- and CD133-

    targeting ADCs showed potent cytotoxic activity in antigen-positive cell lines where the ADC

    preferentially accumulated in lysosomes; however, the conjugates were not active in antigen-positive cell

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 18

    lines in which the ADC colocalized with caveolin-1 (24,31). Both of these ADCs employed the

    ‘vcMMAF’ linker-payload which contains the same enzymatically cleavable dipeptide linker used here

    but releases a different payload (monomethyl auristatin F, MMAF). Due to its carboxylic acid functional

    group, MMAF is a charged species at neutral pH; thus, while the linker may be processed in caveolae, the

    charged nature of MMAF renders it membrane impermeable and not able to enter into the cytoplasm of

    the cell. It is this property that prevents ADCs utilizing MMAF from having substantial bystander effect

    (2). Along similar lines, a CD20-targeting ADC utilizing a ‘vcMMAE’ linker was reported to be

    internalized via both clathrin- and caveolae-mediated endocytosis; however, the nature of endocytosis did

    not impact sensitivity of antigen-positive cell lines to the ADC (32). Unlike MMAF, monomethyl

    auristatin E (MMAE) does not exist as a charged molecule at neutral pH and is membrane permeable.

    Data herein implicate caveolae-mediated endocytosis in T-DM1 resistance; however, another group has

    suggested that caveolae-mediated endocytosis may sensitize cells to T-DM1 (33). While these data

    implicate a possible role for CAV1, the inherent differences in sensitivity to free DM1 (18) may explain

    the differences in basal sensitivity to T-DM1 seen across the high HER2 cell lines evaluated in that study.

    The functional studies in this report implicate a role for caveolae-medicated endocytosis in the

    mechanism of resistance to T-DM1 in N87-TM cells. However, CAV1 knockdown was not sufficient to

    re-sensitize N87-TM cells to T-DM1. Incomplete CAV1 protein loss may prevent re-sensitization, or as

    eluded to previously, the formation of caveolae is not strictly dictated by the level of CAV1 expression,

    but rather the interplay of caveolar-membrane coat proteins (caveolin family) interacting with caveolar-

    membrane stabilizing scaffold proteins (cavin family) (20). N87-TM cells are enriched for proteins from

    both families (i.e. CAV1 and PTRF). Thus, it may be that the driver of caveolae formation in N87-TM

    cells is a combination of altered proteins. Indeed, another caveolar coat protein may be compensating for

    the loss of CAV1 in the CAV1-knockdown N87-TM cells which, in conjunction with the enhanced

    expression of PTRF, allows for the continued formation of caveolae. Along similar lines, CAV1

    expression solely did not correlate with T-DM1 sensitivity across a panel of HER2+ cell lines; rather, the

    functional aptitude for cells to internalize/traffic T-DM1 into caveolar compartments correlated with T-

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 19

    DM1 sensitivity. The function of caveolae in cancer biology remains largely unknown. However, this

    report adds to the growing body of literature that suggests caveolae may dictate sensitivity of cancer cells

    to ADCs that utilize certain types of linker-payloads.

    It has become increasingly clear that ‘precision medicine’ will only be as effective as our ability

    to define patient sub-populations through the use of reliable biomarkers. For many targeted therapies,

    such as ADCs, the paramount biomarker is the expression of the target for that agent (e.g., HER2 for T-

    DM1). However, given tumoral genetic complexity and heterogeneity, target expression does not solely

    predict a patient’s tumor sensitivity to a targeted therapy, particularly in solid tumor indications. For

    example, in the EMILIA trial, only 43% of breast cancer patients with high HER2-expressing tumors

    responded to T-DM1 (11). Thus, there continues to be an unmet need in our ability to better identify

    patients most likely to respond to T-DM1 and other targeted therapies. While alterations in some genes

    (e.g. EGFR, HER3, and PTEN expression and PIK3CA mutations) (34) have been found to not correlate

    with T-DM1 response, data in this report warrant the investigation of caveolae-mediated endocytosis of

    T-DM1 in patient tumors as a potential predictive biomarker for response to T-DM1 and other non-

    cleavable linked ADCs.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 20

    ACKNOWLEDGEMENTS

    The authors appreciate the contributions of Pfizer Oncology colleagues Elwira Muszynska,

    Nadira Prashad, Kiran Khandke, Manoj Charati, William Hu, My-Hanh Lam, Sylvia Musto, Xiang

    Zheng, Anton Xavier, Andreas Giannakou, Mike Cinque, Eric Upeslacis, Anna Maria Barbuti, and

    Katherine Yao for technical assistance. The authors also thank Pfizer Medicinal Chemistry colleagues

    Jeff Casavant, Zecheng Chen, Matthew Doroski, Gary Filzen, Andreas Maderna, Ludivine Moine,

    Alexander Porte, Hud Risley, and Christopher O’Donnell for design and synthesis of the payloads, linker-

    payloads and conjugates utilized in this study.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 21

    REFERENCES

    1. Peng L, Chen X. Antibody-Drug Conjugates. Bioconjugate chemistry 2015;26(11):2169. 2. Kim EG, Kim KM. Strategies and Advancement in Antibody-Drug Conjugate Optimization for

    Targeted Cancer Therapeutics. Biomolecules & therapeutics 2015;23(6):493-509. 3. Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving

    paradigm. Nature reviews Cancer 2013;13(10):714-26. 4. Engelman JA, Settleman J. Acquired resistance to tyrosine kinase inhibitors during cancer

    therapy. Current opinion in genetics & development 2008;18(1):73-9. 5. Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nature

    reviews Cancer 2009;9(1):28-39. 6. de Jonge-Peeters SD, Kuipers F, de Vries EG, Vellenga E. ABC transporter expression in

    hematopoietic stem cells and the role in AML drug resistance. Critical reviews in oncology/hematology 2007;62(3):214-26.

    7. Fletcher JI, Haber M, Henderson MJ, Norris MD. ABC transporters in cancer: more than just drug efflux pumps. Nature reviews Cancer 2010;10(2):147-56.

    8. Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nature reviews Drug discovery 2006;5(3):219-34.

    9. Lauria F, Raspadori D, Rondelli D, Ventura MA, Fiacchini M, Visani G, et al. High bcl-2 expression in acute myeloid leukemia cells correlates with CD34 positivity and complete remission rate. Leukemia 1997;11(12):2075-8.

    10. Lambert JM, Chari RV. Ado-trastuzumab Emtansine (T-DM1): an antibody-drug conjugate (ADC) for HER2-positive breast cancer. Journal of medicinal chemistry 2014;57(16):6949-64.

    11. Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. The New England journal of medicine 2012;367(19):1783-91.

    12. Kang Y-K, Shah MA, Ohtsu A, Van Cutsem E, Ajani JA, Van der Horst T, et al. A randomized, open-label, multicenter, adaptive phase 2/3 study of trastuzumab emtansine (T-DM1) versus a taxane (TAX) in patients (pts) with previously treated HER2-positive locally advanced or metastatic gastric/gastroesophageal junction adenocarcinoma (LA/MGC/GEJC). [abstract]. J Clin Oncol 2016;34(suppl 4S):abstr 5.

    13. Loganzo F, Tan X, Sung M, Jin G, Myers JS, Melamud E, et al. Tumor cells chronically treated with a trastuzumab-maytansinoid antibody-drug conjugate develop varied resistance mechanisms but respond to alternate treatments. Molecular cancer therapeutics 2015;14(4):952-63.

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

    15. Phillips GD, Fields CT, Li G, Dowbenko D, Schaefer G, Miller K, et al. Dual targeting of HER2-positive cancer with trastuzumab emtansine and pertuzumab: critical role for neuregulin blockade in antitumor response to combination therapy. Clinical cancer research : an official journal of the American Association for Cancer Research 2014;20(2):456-68.

    16. Erickson HK, Park PU, Widdison WC, Kovtun YV, Garrett LM, Hoffman K, et al. Antibody-maytansinoid conjugates are activated in targeted cancer cells by lysosomal degradation and linker-dependent intracellular processing. Cancer research 2006;66(8):4426-33.

    17. Chari RV. Targeted cancer therapy: conferring specificity to cytotoxic drugs. Accounts of chemical research 2008;41(1):98-107.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 22

    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 antibody-cytotoxic drug conjugate. Cancer research 2008;68(22):9280-90.

    19. Maderna A, Doroski M, Subramanyam C, Porte A, Leverett CA, Vetelino BC, et al. Discovery of cytotoxic dolastatin 10 analogues with N-terminal modifications. Journal of medicinal chemistry 2014;57(24):10527-43.

    20. Kovtun O, Tillu VA, Ariotti N, Parton RG, Collins BM. Cavin family proteins and the assembly of caveolae. Journal of cell science 2015;128(7):1269-78.

    21. Shajahan AN, Timblin BK, Sandoval R, Tiruppathi C, Malik AB, Minshall RD. Role of Src-induced dynamin-2 phosphorylation in caveolae-mediated endocytosis in endothelial cells. The Journal of biological chemistry 2004;279(19):20392-400.

    22. Parton RG, Simons K. The multiple faces of caveolae. Nature reviews Molecular cell biology 2007;8(3):185-94.

    23. Pelkmans L, Kartenbeck J, Helenius A. Caveolar endocytosis of simian virus 40 reveals a new two-step vesicular-transport pathway to the ER. Nature cell biology 2001;3(5):473-83.

    24. Smith LM, Nesterova A, Alley SC, Torgov MY, Carter PJ. Potent cytotoxicity of an auristatin-containing antibody-drug conjugate targeting melanoma cells expressing melanotransferrin/p97. Molecular cancer therapeutics 2006;5(6):1474-82.

    25. Sowa G, Pypaert M, Fulton D, Sessa WC. The phosphorylation of caveolin-2 on serines 23 and 36 modulates caveolin-1-dependent caveolae formation. Proceedings of the National Academy of Sciences of the United States of America 2003;100(11):6511-6.

    26. Scaltriti M, Rojo F, Ocana A, Anido J, Guzman M, Cortes J, et al. Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer. Journal of the National Cancer Institute 2007;99(8):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 of action of cancer therapeutics trastuzumab and geldanamycin. Molecular biology of the cell 2004;15(12):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 Antibody Maytansine Conjugates from the Lysosome to the Cytoplasm. Cancer research 2015;75(24):5329-40.

    29. Yu SF, Zheng B, Go M, Lau J, Spencer S, Raab H, et al. A Novel Anti-CD22 Anthracycline-Based Antibody-Drug Conjugate (ADC) That Overcomes Resistance to Auristatin-Based ADCs. Clinical cancer research : an official journal of the American Association for Cancer Research 2015;21(14):3298-306.

    30. Mohamed MM, Sloane BF. Cysteine cathepsins: multifunctional enzymes in cancer. Nature reviews Cancer 2006;6(10):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-drug conjugates in hepatocellular and gastric cancers. British journal of cancer 2008;99(1):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-auristatin conjugates. Clinical cancer research : an official journal of the American Association for Cancer Research 2004;10(23):7842-51.

    33. Chung YC, Kuo JF, Wei WC, Chang KJ, Chao WT. Caveolin-1 Dependent Endocytosis Enhances the Chemosensitivity of HER-2 Positive Breast Cancer Cells to Trastuzumab Emtansine (T-DM1). PloS one 2015;10(7):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 III Study of Trastuzumab Emtansine in HER2-

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 23

    Positive Metastatic Breast Cancer. Clinical cancer research : an official journal of the American Association for Cancer Research 2016;22(15):3755-63.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 24

    TABLES

    N87: Parental versus TM-resistant

    Compound Linker N87 N87-TM-2 Relative

    name type

    (IC50,

    nmol/L)

    (IC50,

    nmol/L) resistance

    T-MCC_DM1 NC 1.6 174 109x

    T_mc_Aur8261 NC 5.3 >1,000 >188x

    T_mc_MMAD NC 16 492 31x

    T_mc_Aur0101 NC 138 >1,000 >7x

    T_vc_Aur8261 C 0.43 0.49 1.1x

    T-vc_MMAD C 0.4 0.88 2.2x

    T_vc_Aur0101 C 1.1 0.8 0.73x

    T_vc_Cys-mc-

    Aur8261 C 3.9 136 35x

    DM1-Sme -- 19 35 1.8x

    Aur-8254 (8261) -- 0.61 0.92 1.5x

    MMAD -- 0.14 0.49 3.5x

    Aur-0101 -- 1.5 1.9 1.3x

    Cys_mc-Aur8261 -- 2,334 4,392 1.9x

    Vinblastine -- 3.6 4.8 1.3x

    Docetaxel -- 2.9 3.8 1.3x

    Doxorubicin -- 38 37 1.0x

    5-fluorouracil -- 1,611 3,135 1.9x

    Camptothecin -- 21 9.8 0.47x

    Gemcitabine -- 7.3 8.4 1.2x

    Oxaliplatin -- 623 1,511 2.4x

    Temsirolimus -- 2,131 6,706 3.1x

    Rapamycin -- 4,682 12,403 2.6x

    Dacomitinib -- 7.8 33 4.2x

    Pelitinib -- 42 119 2.8x

    Neratinib -- 2 11 5.5x

    NOTE: IC50 values of ADCs reported here represent payload concentrations.

    Data are mean of multiple determinations. These relative resistance values are

    represented graphically in Supplementary Figure S4. Relative resistance is the

    ratio of the mean IC50 for the TM-resistant cell line versus the parental cell

    line. NC = non-cleavable, C = cleavable

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

    chemotherapeutics and other unconjugated drugs.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 25

    FIGURE LEGENDS

    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 dosed with T-DM1 for three 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 and N87-TM-2), were treated with 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 were separated by SDS-PAGE

    and levels of MDR1 (C), MRP1 (D), and HER2 (E) were determined by immunoblot analyses. Whole

    cell lysates from positive and negative control cell lines for MDR1 (KB85 and KBV1) and MRP1 (A549

    and H69AR) are shown.

    (F) Parental and T-DM1-resistant N87 cells were treated with a trastuzumab-ADC conjugated to a non-

    cleavable linker and auristatin payload for 4 days. Cell viability is plotted against ADC payload

    concentration.

    (G) Parental and T-DM1-resistant N87 cells were treated with a trastuzumab-ADC conjugated to a

    cleavable-linked auristatin payload for 4 days. Cell viability is plotted against ADC payload

    concentration.

    Figure 2: N87-TM cells retain T-DM1 resistance and T-vc-Aur0101 sensitivity in vivo.

    (A-B) Parental (A) and T-DM1 resistant (B) N87 cells were implanted into NOD-SCID mice and treated

    with ADCs when the average tumor volume of 250 mm3 was achieved. Mice were dosed q4dx4 with

    vehicle (black circles), T-DM1 at 6 mg/kg (mpk) (red squares), T-DM1 at 10 mpk (red triangles), T-vc-

    Aur0101 at 3 mpk (blue diamonds), or a negative control, isotype ADC with ‘vc-Aur0101’ at 3 mpk

    (hashed blue line with downward-pointing triangles). Tumors at staging size for each cell line were

    collected, formalin fixed and immunostained for HER2 by immunohistochemistry (insets).

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 26

    (C) N87 and N87-TM tumors were treated with a single dose of T-DM1 at 6 mpk, T-vc-Aur0101 at 3

    mpk or a negative control, isotype ADC with ‘vc-Aur0101’ at 3 mpk. After 24 hours, mice were perfused

    and tumors formalin fixed and co-immunostained for anti-human IgG (brown color) and phospho-histone

    H3 (pHH3) (purple color). Bystander effect is denoted by the prevalence of pHH3+ cells devoid of ADC

    binding.

    Figure 3: N87-TM cells are enriched for intracellular caveolar compartments and internalize

    trastuzumab-ADCs via caveolae-mediated endocytosis.

    (A) Membrane fractions 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 to N87 cells.

    (B) Whole cell lysates of N87 and N87-TM cells were prepared, separated by SDS-PAGE and

    immunoblotted for proteins identified in the proteomic profiling 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, and phospho-CAV1 were determined by immunoblot analysis.

    (D) CAV1+ puncta were identified via immunofluorescence staining of N87 and N87-TM cells.

    Average of number of puncta calculated as total number of CAV1+ puncta normalized by number of

    nuclei for each field of view.

    (E) N87 and N87-TM cells were treated with T-DM1 overnight at 37°C, fixed and co-immunostained

    with anti-human IgG (green color) and anti-CAV1 (red color). Image analysis was performed to

    determine co-localization between T-DM1 and CAV1 in N87 and N87-TM cells.

    (F) N87 and N87-TM cells were incubated with fluorescently-labelled trastuzumab and kinetic co-

    localization analysis between T-DM1 and CAV1 and LAMP1 markers were performed via image

    cytometry.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 27

    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 relative HER2 and CAV1 at

    basal state by immunoblot analysis. CAV1 score was assigned based on basal CAV1 expression from

    this experiment. The same panel 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 vs. T-DM1 IC50 is represented.

    (B) The panel of cell lines were incubated with T-DM1 and analyzed for co-localization to CAV1+

    puncta as previously described in Figure 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-

    localized with CAV1+ puncta (yellow/orange color).

    (C) The T-DM1 and CAV1 co-localization scores from (B) for each cell line were plotted against IC50 of

    each cell line to T-DM1. The correlation between T-DM1 and CAV1 colocalization score vs. T-DM1

    IC50 is represented.

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • A

    Figure 1

    Naïve Cells

    “N87”

    ON OFF

    T-DM1-resistant cells

    “N87-TM”

    Treatment: T-DM1 ADC @ 10X IC50

    ON OFF ON OFF

    B

    C

    N8

    7

    N8

    7-T

    M-1

    N8

    7-T

    M-2

    IB: HER2

    IB: Actin

    D

    E F

    G

    0 .0 1 0 .1 1 1 0 1 0 0 1 0 0 0 1 0 0 0 0

    0

    2 5

    5 0

    7 5

    1 0 0

    1 2 5

    C o n c (n M )

    % S

    urv

    iva

    l

    N 87

    N 8 7 -T M -1

    N 8 7 -T M -2

    T -D M 1

    0 .0 0 1 0 .0 1 0 .1 1 1 0 1 0 0 1 0 0 0

    0

    2 5

    5 0

    7 5

    1 0 0

    1 2 5

    C o n c (n M )

    % S

    urv

    iva

    l

    N 87

    N 8 7 -T M -1

    N 8 7 -T M -2

    T -v c -A u r 0 1 0 1

    0 .0 0 1 0 .0 1 0 .1 1 1 0 1 0 0 1 0 0 0

    0

    2 5

    5 0

    7 5

    1 0 0

    1 2 5

    C o n c (n M )

    % S

    urv

    iva

    l

    N 87

    N 8 7 -T M -1

    N 8 7 -T M -2

    T -m c -A u r 8 2 6 1

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • IHC: HER2

    IHC: HER2

    A

    B

    C T-vc-Aur-0101 T-DM1

    N87

    N87-TM

    IHC: Hu IgG, pHH3

    0 2 0 4 0 6 0 8 0

    0

    2 0 0

    4 0 0

    6 0 0

    8 0 0

    1 0 0 0

    1 2 0 0

    1 4 0 0

    1 6 0 0

    1 8 0 0

    2 0 0 0

    T im e (D a y s )

    Tu

    mo

    r v

    ol

    (mm

    3)

    V e h ic le (P B S )

    T -D M 1 @ 6 m p k

    T -D M 1 @ 1 0 m p k

    T -v c -A u r0 1 0 1 @ 3 m p k

    N 8 7

    N e g C tl-v c -A u r0 1 0 1 @ 3 m p k

    0 2 0 4 0 6 0 8 0

    0

    2 0 0

    4 0 0

    6 0 0

    8 0 0

    1 0 0 0

    1 2 0 0

    1 4 0 0

    1 6 0 0

    1 8 0 0

    2 0 0 0

    T im e (D a y s )

    Tu

    mo

    r v

    ol

    (mm

    3)

    V e h ic le (P B S )

    T -D M 1 @ 6 m p k

    T -D M 1 @ 1 0 m p k

    T -v c -A u r0 1 0 1 @ 3 m p k

    N 8 7 -T M

    N e g C tl-v c -A u r0 1 0 1 @ 3 m p k

    NegCtl-vc-Aur-0101

    Figure 2

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • A B

    C

    E

    Down in N87-TM

    Up in N87-TM

    Fold Change (log2)

    Sign

    ific

    ance

    Sco

    re

    IB: IGF2R

    IB: LAMP1

    IB: Tubulin

    N87 N87-TM

    IB: CAV1

    IB: PTRF

    IB: Tubulin

    IB: Clathrin

    N87 N87-TM

    N87

    N87-T

    M

    0

    2 0

    4 0

    6 0

    8 0

    1 0 0

    # o

    f C

    AV

    1 p

    un

    cta

    pe

    r c

    ell

    N87

    N87-T

    M

    0 .0

    0 .2

    0 .4

    0 .6

    0 .8

    A D C C o lo c a liz a t io n w ith C A V 1 + p u n c ta

    a fte r 2 4 h o u r A D C tr e a tm e n t

    Ov

    erla

    p C

    orre

    lati

    on

    R

    0 1 0 0 2 0 0 3 0 0 4 0 0

    0 .2

    0 .4

    0 .6

    0 .8

    T im e (m in )

    Co

    loc

    ali

    za

    tio

    n S

    co

    re

    N 87

    N 87-T M

    C A V 1 C o lo c a liz a t io n w ith T r a s tu z u m a b

    0 1 0 0 2 0 0 3 0 0

    0 .2

    0 .4

    0 .6

    0 .8

    T im e (m in )

    Co

    loc

    ali

    za

    tio

    n S

    co

    re

    N 87

    N 87-T M

    L y s o s o m e C o lo c a liz a t io n w ith T ra s tu z u m a b

    IB: pSRC (Y416)

    IB: CAV1

    IB: pCAV1 (Y14)

    IB: Tubulin

    N87 N87-TM

    IB: SRC

    D

    F

    Figure 3

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • A B N87 N87-TM

    BT474 HCC1954

    SKBR3 SKOV3

    JIMT1

    C

    0 .1 1 1 0 1 0 0 1 0 0 0

    0 .0

    0 .5

    1 .0

    1 .5

    [T -D M 1 ] (P a y lo a d n M )

    AD

    C:C

    AV

    1 C

    olo

    ca

    liz

    ati

    on

    N 8 7

    N 8 7 -T M

    B T 4 7 4H C C 1 9 5 4

    J IM T 1

    S K O V 3S K B R 3

    R2

    = 0 .7 4

    p = 0 .0 1

    IB: HER2

    IB: CAV1

    IB: Tubulin

    0 .1 1 1 0 1 0 0 1 0 0 0

    0

    1

    2

    3

    [T -D M 1 ] (P a y lo a d n M )

    CA

    V1

    Sc

    ore

    (im

    mu

    no

    blo

    t)

    N 8 7

    N 8 7 -T M

    B T 4 7 4

    H C C 1 9 5 4

    J IM T 1S K O V 3

    S K B R 3

    R2

    = 0 .3 0

    p = 0 .2 0

    Figure 4

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/

  • 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:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

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

    Subscriptions

    Reprints and

    [email protected] at

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

    Permissions

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

    .http://mct.aacrjournals.org/content/early/2017/10/20/1535-7163.MCT-17-0403To request permission to re-use all or part of this article, use this link

    on July 2, 2021. © 2017 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on October 20, 2017; DOI: 10.1158/1535-7163.MCT-17-0403

    http://mct.aacrjournals.org/lookup/doi/10.1158/1535-7163.MCT-17-0403http://mct.aacrjournals.org/content/suppl/2017/10/20/1535-7163.MCT-17-0403.DC1http://mct.aacrjournals.org/cgi/alertsmailto:[email protected]://mct.aacrjournals.org/content/early/2017/10/20/1535-7163.MCT-17-0403http://mct.aacrjournals.org/

    Figure 1Figure 2Figure 3Figure 4