Top Banner
Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) Acquired Resistance in HER2-positive Breast Cancer MohammadA Sabbaghi 1 , Gabriel Gil-G omez 1 , Cristina Guardia 1 , Sonia Servitja 1,2 , Oriol Arpí 1 , Sara García-Alonso 3 , Silvia Menendez 1 , Montserrat Arumi-Uria 4 , Laia Serrano 4 , Marta Salido 1,4 , Aura Muntasell 5 , Maria Martínez-García 1,2 , Sandra Zazo 6 , Cristina Chamizo 6 , Paula Gonz alez-Alonso 6 , Juan Madoz-G urpide 6 , Pilar Eroles 7 , Joaquin Arribas 8,9,10 , Ignasi Tusquets 1,2 , Ana Lluch 11 , Atanasio Pandiella 3 , Federico Rojo 6 , Ana Rovira 1,2 , and Joan Albanell 1,2,12 Abstract Purpose: Trastuzumab-emtansine (T-DM1) is a standard treat- ment in advanced HER2-positive breast cancer. However, resis- tance inevitably occurs. We aimed to identify mechanisms of acquired T-DM1 resistance. Experimental Design: HER2-positive breast cancer cells (HCC1954, HCC1419, SKBR3, and BT474) were treated in a pulse-fashion with T-DM1 to induce a resistant phenotype. Cel- lular and molecular effects of T-DM1 in parental versus resistant cells were compared. CDK1 kinase activity and cyclin B1 expres- sion were assayed under various conditions. Genetic modica- tions to up- or downregulate cyclin B1 were conducted. Effects of T-DM1 on cyclin B1 levels, proliferation, and apoptosis were assayed in human HER2-positive breast cancer explants. Results: We obtained three cell lines with different levels of acquired T-DM1 resistance (HCC1954/TDR, HCC1419/TDR, and SKBR3/TDR cells). HER2 remained amplied in the resistant cells. Binding to HER2 and intracellular uptake of T-DM1 were main- tained in resistant cells. T-DM1 induced cyclin B1 accumulation in sensitive but not resistant cells. Cyclin B1 knockdown by siRNA in parental cells induced T-DM1 resistance, while increased levels of cyclin B1 by silencing cdc20 partially sensitized resis- tant cells. In a series of 18 HER2-positive breast cancer fresh explants, T-DM1 effects on proliferation and apoptosis paral- leled cyclin B1 accumulation. Conclusions: Defective cyclin B1 induction by T-DM1 med- iates acquired resistance in HER2-positive breast cancer cells. These results support the testing of cyclin B1 induction upon T-DM1 treatment as a pharmacodynamic predictor in HER2- positive breast cancer. Clin Cancer Res; 23(22); 700619. Ó2017 AACR. Introduction Trastuzumab-emtansine (T-DM1) is an antibodydrug conju- gate (ADC) consisting of the anti-HER2 antibody trastuzumab covalently linked to the antimitotic agent DM1 through a stable linker that potently inhibits growth of both trastuzumab-sensitive and -resistant HER2-amplied cancer cells (1). T-DM1 has mechanisms of action containing of the antitumor effects related to trastuzumab and those associated with intracellular DM1 catabolites (2, 3). DM1 is a derivative of maytansine, a highly potent antimitotic drug (4). Once bound to HER2, T-DM1 enters in the cell by receptor-mediated endocytosis and the HER2T- DM1 complex is processed via degradation of trastuzumab in lysosomes giving rise to the intracellular release of the active catabolite Lys-MCC-DM1. In the cytoplasm, DM1 exerts its func- tions through binding to the beta subunit of tubulin and mod- ifying its assembly properties. By doing so, DM1 is able to disrupt the formation of the mitotic spindle necessary for accurate chro- mosome segregation along mitotic process. Overall, DM1 as other antimitotic drugs elicits a mitotic arrest and cells therefore fail to complete a normal mitosis. This prolonged cell-cycle delay eventually culminates in cell death by mitotic catastrophe, necro- sis, or apoptosis (5). In addition to these DM1-related actions, T-DM1 maintains properties of trastuzumab such as inhibition of HER2-directed signal transduction and activation of antibody- dependent cell-mediated cytotoxicity (2, 3). T-DM1 is a standard second-line treatment for HER2- positive metastatic breast cancer patients based on the results 1 Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barce- lona, Spain. 2 Medical Oncology Department, Hospital del Mar- CIBERONC, Barcelona, Spain. 3 Centro de Investigaci on del C ancer, Universidad de Sala- manca-CSIC-CIBERONC, Salamanca, Spain. 4 Pathology Department, Hospital del Mar, Barcelona, Spain. 5 Immunity and infection Laboratory, IMIM (Hospital del Mar Research Institute), Barcelona, Spain. 6 Pathology Department, IIS- Fundaci on Jim enez Díaz- CIBERONC, Madrid, Spain. 7 INCLIVA Biomedical Research Institute, Valencia, Spain. 8 Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO)-CIBERONC, Barcelona, Spain. 9 Autonomous Uni- versity of Barcelona, Barcelona, Spain. 10 Instituci o Catalana de Recerca I Estudis Avan¸ cats (ICREA), Barcelona, Spain. 11 Oncology and Hematology Department, Hospital Clínico Universitario-CIBERONC, Valencia, Spain. 12 Universitat Pompeu Fabra, Barcelona, Spain. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Joan Albanell, Medical Oncology Department, Hospital del Mar, Passeig Maritim 25-29, Barcelona 08003, Spain. Phone: 349-3248-3137; Fax: 349-3248-3366; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-17-0696 Ó2017 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 23(22) November 15, 2017 7006 on December 14, 2020. © 2017 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696
15

Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

Aug 24, 2020

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

Cancer Therapy: Preclinical

Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance inHER2-positive Breast CancerMohammadA Sabbaghi1, Gabriel Gil-G�omez1, Cristina Guardia1, Sonia Servitja1,2,Oriol Arpí1, Sara García-Alonso3, Silvia Menendez1, Montserrat Arumi-Uria4, Laia Serrano4,Marta Salido1,4, Aura Muntasell5, Maria Martínez-García1,2, Sandra Zazo6,Cristina Chamizo6, Paula Gonz�alez-Alonso6, Juan Madoz-G�urpide6, Pilar Eroles7,Joaquin Arribas8,9,10, Ignasi Tusquets1,2, Ana Lluch11, Atanasio Pandiella3, Federico Rojo6,Ana Rovira1,2, and Joan Albanell1,2,12

Abstract

Purpose: Trastuzumab-emtansine (T-DM1) is a standard treat-ment in advanced HER2-positive breast cancer. However, resis-tance inevitably occurs. We aimed to identify mechanisms ofacquired T-DM1 resistance.

Experimental Design: HER2-positive breast cancer cells(HCC1954, HCC1419, SKBR3, and BT474) were treated in apulse-fashion with T-DM1 to induce a resistant phenotype. Cel-lular and molecular effects of T-DM1 in parental versus resistantcells were compared. CDK1 kinase activity and cyclin B1 expres-sion were assayed under various conditions. Genetic modifica-tions to up- or downregulate cyclin B1 were conducted. Effects ofT-DM1 on cyclin B1 levels, proliferation, and apoptosis wereassayed in human HER2-positive breast cancer explants.

Results: We obtained three cell lines with different levels ofacquired T-DM1 resistance (HCC1954/TDR,HCC1419/TDR, and

SKBR3/TDR cells). HER2 remained amplified in the resistant cells.Binding to HER2 and intracellular uptake of T-DM1 were main-tained in resistant cells. T-DM1 induced cyclin B1 accumulation insensitive but not resistant cells. Cyclin B1 knockdown by siRNAin parental cells induced T-DM1 resistance, while increasedlevels of cyclin B1 by silencing cdc20 partially sensitized resis-tant cells. In a series of 18 HER2-positive breast cancer freshexplants, T-DM1 effects on proliferation and apoptosis paral-leled cyclin B1 accumulation.

Conclusions: Defective cyclin B1 induction by T-DM1 med-iates acquired resistance in HER2-positive breast cancer cells.These results support the testing of cyclin B1 induction uponT-DM1 treatment as a pharmacodynamic predictor in HER2-positive breast cancer. Clin Cancer Res; 23(22); 7006–19. �2017AACR.

IntroductionTrastuzumab-emtansine (T-DM1) is an antibody–drug conju-

gate (ADC) consisting of the anti-HER2 antibody trastuzumab

covalently linked to the antimitotic agent DM1 through a stablelinker that potently inhibits growth of both trastuzumab-sensitiveand -resistant HER2-amplified cancer cells (1). T-DM1 hasmechanisms of action containing of the antitumor effects relatedto trastuzumab and those associated with intracellular DM1catabolites (2, 3). DM1 is a derivative of maytansine, a highlypotent antimitotic drug (4). Once bound to HER2, T-DM1 entersin the cell by receptor-mediated endocytosis and the HER2–T-DM1 complex is processed via degradation of trastuzumab inlysosomes giving rise to the intracellular release of the activecatabolite Lys-MCC-DM1. In the cytoplasm, DM1 exerts its func-tions through binding to the beta subunit of tubulin and mod-ifying its assembly properties. By doing so, DM1 is able to disruptthe formation of the mitotic spindle necessary for accurate chro-mosome segregation alongmitotic process.Overall, DM1as otherantimitotic drugs elicits a mitotic arrest and cells therefore failto complete a normal mitosis. This prolonged cell-cycle delayeventually culminates in cell death by mitotic catastrophe, necro-sis, or apoptosis (5). In addition to these DM1-related actions,T-DM1maintains properties of trastuzumab such as inhibition ofHER2-directed signal transduction and activation of antibody-dependent cell-mediated cytotoxicity (2, 3).

T-DM1 is a standard second-line treatment for HER2-positive metastatic breast cancer patients based on the results

1Cancer Research Program, IMIM (Hospital del Mar Research Institute), Barce-lona, Spain. 2Medical Oncology Department, Hospital del Mar- CIBERONC,Barcelona, Spain. 3Centro de Investigaci�on del C�ancer, Universidad de Sala-manca-CSIC-CIBERONC, Salamanca, Spain. 4Pathology Department, Hospitaldel Mar, Barcelona, Spain. 5Immunity and infection Laboratory, IMIM (Hospitaldel Mar Research Institute), Barcelona, Spain. 6Pathology Department, IIS-Fundaci�on Jim�enez Díaz- CIBERONC, Madrid, Spain. 7INCLIVA BiomedicalResearch Institute, Valencia, Spain. 8Preclinical Research Program,Vall d'HebronInstitute of Oncology (VHIO)-CIBERONC, Barcelona, Spain. 9Autonomous Uni-versity of Barcelona, Barcelona, Spain. 10Instituci�o Catalana de Recerca I EstudisAvancats (ICREA), Barcelona, Spain. 11Oncology and Hematology Department,Hospital Clínico Universitario-CIBERONC, Valencia, Spain. 12Universitat PompeuFabra, Barcelona, Spain.

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

Corresponding Author: Joan Albanell, Medical Oncology Department, Hospitaldel Mar, Passeig Maritim 25-29, Barcelona 08003, Spain. Phone: 349-3248-3137;Fax: 349-3248-3366; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-17-0696

�2017 American Association for Cancer Research.

ClinicalCancerResearch

Clin Cancer Res; 23(22) November 15, 20177006

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 2: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

of a phase III clinical trial (EMILIA trial), that compared theefficacy and toxicity of T-DM1 versus the combination oflapatinib and capecitabine in patients previously treated withtrastuzumab and chemotherapy. T-DM1 offered superior activ-ity, tolerability, and survival than lapatinib and capecitabine(6). However, resistance to T-DM1 occurs (3, 7, 8). To date, themain T-DM1 clinical resistance mechanisms studies includedan exposure–response and exploratory biomarker analysis,both based on patients from the EMILIA and TH3RESA trial(9–11). In the exposure–response analysis, the data showedthat higher T-DM1 exposure was associated with improvedefficacy. This analysis suggested that there is an opportunity tooptimize T-DM1 dose in the patient subgroup with low expo-sure (10). With regards to the exploratory biomarker study ofthe EMILIA trial, focusing only in the T-DM1 group, univariateanalysis of the data suggested that there were no evidentdifferences in progression-free survival (PFS) related to EGFR,and HER3 median mRNA concentration ratios, PIK3CA muta-tions status, or PTEN expression; patients with HER2 mRNAabove the median had better outcomes on T-DM1 than thoseequal or below the median (11). In the TH3RESA phase IIItrial, performed in heavily pretreated HER2-positive advancedbreast cancer, patients were randomized to T-DM1 or treat-ment of physician's choice. An exploratory biomarker analysisincluded HER2 and HER3 mRNA expression, PIK3CA mutationstatus, and PTEN protein expression. T-DM1 prolonged medianPFS in all the subgroups analyzed. A numerically greater benefitwas reported in patients with tumors expressing HER2 mRNAabove the median (9). More recently, in the ZEPHIR trial, theuse of molecular imaging of HER2 by HER2-PET/CT with(89)Zr-trastuzumab combined with early metabolic responseassessment by FDG-PET/CT, discriminated patients with shortversus long time to T-DM1 treatment failure (12).

The biomarker analyses discussed above were focused on theHER2 signaling pathway. However, given the dual mechanism of

action of T-DM1, we focused on resistance potentially related tothe cell-cycle–modulating effects of DM1. We identified that theG2–M arrest induced by T-DM1 in sensitive HER2 breast cancercells did not occur in their resistant counterparts in a CDK1/cyclinB1–dependent manner. In fresh HER2-positive breast cancerexplants, lack of induction of cyclin B1 correlated with T-DM1failure to induce apoptosis.

Materials and MethodsCell lines and reagents

Breast cancer cell lines BT474, SKBR3, AU565, EFM-192A,HCC1954, and HCC1419 were obtained from the ATCC.Authenticity of the cells was tested by STR DNA Profilinganalysis at the ATCC (June 2013 and December 2014) beforestarting the generation of resistant cells. The number of pas-sages between thawing and use in the described experimentswas five or less. T-DM1 (trastuzumab emtansine, Kadcyla) wasprovided by Genentech under MTA agreement (Sliwkowski MX,Lewis Phillips GD) and trastuzumab by Hospital del Marpharmacy (Barcelona, Spain).

Cell proliferation assaysCells were plated in duplicate into 12-well culture plates at a

density of 8–12� 103 cells per well and left overnight and then onday zero, treatmentwas initiated. At this timeT-DM1(0.1–1mg/mL)was added. On days 3, 7, and 10, cells were washed with PBS,trypsinized, resuspended inmedia, and counted with Scepter Auto-mated Cell Counter (Millipore).

Generation of cell lines with acquired T-DM1 resistanceT-DM1–resistant cell lines were derived from original paren-

tal cell lines by exposure to stepwise increasing concentrationsof T-DM1 in a pulse fashion (13). The protocol is summarizedin Fig. 1A. Three T-DM1 acquired resistance sublines werecollected named SKBR3/TDR, HCC1954/TDR, and HCC1419/TDR. In addition, vehicle-treated parental cell lines were kept inculture during this period as control cell lines. We established anexposure to 0.1 mg/mL of T-DM1 for 3 days as a referenceschedule to define resistance in our model. This schedule wasselected on the basis of experiments performed in MCF7 cellsthat do not overexpress HER2, whose growth was unaffecteduntil higher concentrations and longer exposure to T-DM1were used.

FISH for HER2Formalin-fixed and paraffin-embedded cell pellets were pre-

pared from parental and T-DM1–resistant cells to assess the statusof HER2 gene by scoring its amplification following ASCP/CAPguidelines (14). The commercial PathVysion HER2 DNA probewas used.

T-DM1/HER2 receptor bindingT-DM1 binding to the cell surface was evaluated by indirect

immunofluorescence staining and flow cytometry. Cells (2–5 �105) were incubated with T-DM1 (7.5 mg/mL) for 30 minutesfollowed by R-Phycoerythrin AffiniPure F(ab0)2 Fragment GoatAnti-Human IgG, Fcg Fragment Specific (Jackson ImmunoRe-search; 1:500 dilution) for 30 minutes at 4�C. Trastuzumab(7.5 mg/mL) and rituximab (MabTera, Roche; 7.5 mg/mL) were

Translational Relevance

Trastuzumab-emtansine (T-DM1) is an antibody–drug con-jugate constituted by trastuzumab linked to DM1 (a tubulinpolymerization inhibitor). T-DM1 is a standard treatment inadvanced HER2-positive metastatic breast cancer. However,resistance inevitably occurs. To date, no clinical biomarker ofT-DM1 resistance has been identified. In this study, weobtained three T-DM1 acquired resistance breast cancer invitro models. We report that cyclin B1 induction is a hallmarkof T-DM1 activity in both trastuzumab primary sensitive andresistant cells. In HER2-positive breast cancer cells withacquired T-DM1 resistance, the drug failed to induce cyclinB1, while reducing cyclin B1 degradation in these cellspartially restores sensitivity. In fresh human HER2-positivebreast cancer explants, the induction (but not the baselinelevels) of cyclin B1 by T-DM1 correlated with apoptosis,suggesting that a pharmacodynamic assay to test cyclin B1induction in breast cancer patients treated with T-DM1 mayhelp to identify early the patients more likely to benefit fromthat drug treatment.

Cyclin B1 and T-DM1 Acquired Resistance

www.aacrjournals.org Clin Cancer Res; 23(22) November 15, 2017 7007

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 3: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

used as positive andnegative controls, respectively. Live/dead gatewas set withDAPI counterstaining. Samples were acquired on LSRFortessa flow cytometer (BD Biosciences), and data analyzed withFlowJo software (TreeStar).

T-DM1 internalization assayT-DM1 internalization was evaluated by immunofluores-

cence staining. Cells (1.5 � 105) were seeded on coverslips andtreated with 1.5 mg/mL T-DM1 for 15 minutes. After washingout the drug, cells were cultured for 24 hours with or without5 mmol/L chloroquine (a drug that induced changes in lyso-somal pH) to accumulate intracellular T-DM1. Anti-humanCy3-conjugated antibody was used to detect T-DM1, phalloi-din-FITC (P5282 Sigma) was used for actin staining, andnuclei were counterstained with DAPI. Sample processing wasperformed as reported previously (15).

Cell-cycle assayCells were seeded on 6-well plates and treatedwith compounds

for 24 hours by T-DM1 (0.1 mg/mL). Thereafter, cells were incu-bated with 30 mmol/L bromodeoxyuridine (BrdUrd, B9285,Sigma) for 30 minutes at 37�C, and then subsequently trypsi-nized, washed with PBS, and fixed in ice-cold ethanol for at leastone hour at �20�C. Cells were digested in prewarmed 1 mg/mLpepsin in 30 mmol/L HCl (pH 1.5) for 30 minutes at 37�C withgentle shaking, and then incubated in 2mol/LHCl for 20minutesat room temperature. Next, cells were washed with PBS andantibody buffer (0.5% w/v BSA, 0.5% v/v Tween-20 in PBS) andincubatedwithmouseprimary antibody against BrdUrd (555627,BD Pharmingen) in antibody buffer for one hour at room tem-perature. After washing with PBS, samples were incubated withsecondary goat anti-mouse IgG FITC-conjugated antibody for 30minutes at room temperature in the dark. Finally, cells were

A

B

T-DM1 [μg/mL]

0 0.1 10

40

80

120

HCC1954 HCC1954/TDR

***

***

T-DM1 [μg/mL]

0 0.1 10

40

80

120

HCC1419 HCC1419/TDR

**

Parental cells 3 days

on/off

3 days

on/off

3 days

on/off

3 days

on/off

3 days

on/off

3 days

on/off

3 days

on/off

3 days

on/off

3 days

on/off

T-DM1 [1 μg/mL]18 days

T-DM1 [2 μg/mL]18 days

T-DM1 [4 μg/mL]18 days

T-DM1 Acquired resistant cells

HCC1954 HCC1954/TDR0

40

80

120

Cel

l num

ber

(% o

f con

trol)

Cel

l num

ber

(% o

f con

trol)

HCC1419 HCC1419/TDR0

40

80

120

SKBR3 SKBR3/TDR0

40

80

120ControlTrastuzumab

** ***C

T-DM1 [μg/mL]

0 0.1 10

40

80

120

SKBR3 SKBR3/TDR

*** ***

Figure 1.

Generation and growth characteristics of HER2-positive breast cancer cells with T-DM1 acquired resistance. A, Pulsed treatment strategy for the in vitroestablishment of acquired resistant cells to T-DM1. The procedure consisted of three consecutive cycles of 3 days on treatment followed by 3 days off treatment foreach T-DM1 concentration of 1, 2, and 4 mg/mL. The entire 54-day protocol resulted in the generation of cells with varying levels of T-DM1 resistance. B, Growthcharacteristics of T-DM1–resistant cells. Each resistant cell line was compared with the corresponding age and passage-matched parental cell line. HCC1954-HCC1954/TDR, HCC1419-HCC1419/TDR, and SKBR3-SKBR3/TDR pairs of sensitive and resistant cells were seeded in duplicate in 12-well plates at a densityof 8–12� 103 cells perwell and allowed to adhere overnight. Then, cellswere treatedwithout (white columns) orwith two concentrations of T-DM1 (black columns) for3 days. Cells were collected by trypsinization and counted using a Scepter cell counter (Millipore). Experiments were performed in triplicate and data representthe mean � SD cell number relative to control. Error bars, SD. C, T-DM1–resistant cells retain a similar sensitivity to trastuzumab. Each paired sensitive/resistant cell lineswere seeded as inB and treated in duplicatewith orwithout 15mg/mL trastuzumab for 7 days. Medium changeswere performed every 3 days. Cellswere trypsinized, collected, and counted using Scepter. The results are presented as mean percent of viability in treated versus nontreated controls � SD.

Sabbaghi et al.

Clin Cancer Res; 23(22) November 15, 2017 Clinical Cancer Research7008

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 4: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

washed one more time with PBS and then stained with 25 mg/mLpropidium iodide. Flow cytometry was performed using a BectonDickinson FACScan operated by the CELLQuest software.

Cdc2/CDK1 activity assayCdc2/CDK1 kinase activity was measured with the nonradio-

active MESACUP Cdc2/CDK1 Kinase Assay Kit (MBL, Interna-tional Corporation) following the manufacturer's instructions.Themethod is based on an ELISA assay that utilizes a biotinylatedsynthetic peptide as a substrate for the Cdc2/CDK1 kinasespresent in the samples and a mAb conjugated to horseradishperoxidase (HRP) recognizing the phosphorylated form of thepeptide. The HRP substrate is then added and the intensity of thecolor was measured at 492 nm. The results are reported as foldinduction of OD 492 nm in arbitrary units (RLA) of treatedsample using untreated condition as reference set at 1.

ImmunocytochemistryCells were seeded on glass coverslips and cultured as indicated

in the figure legends. Cells washed by PBS, and then fixed in 4%paraformaldehyde for 10minutes at room temperature. Then thesamples permeabilized by PBS containing 0.25% Triton X-100,and then, incubated with 1% BSA in PBST for 30 minutesfor blocking unspecific binding of the antibodies. In the nextstep, samples incubated for 1 hour with anti-a-tubulin FITC-conjugated antibody (F2168, Sigma). Cell nuclei were stainedwith DAPI (1:1,000; Sigma) for 15minutes at room tempera-ture. After washing, coverslips were mounted with a drop ofmounting medium and viewed using a Leica SP5 uprightconfocal microscope.

ImmunoblottingCellular protein lysateswere prepared in lysis buffer [50mmol/L

Tris-HCl, pH8.0, 100mmol/LNaCl, 1% (v/v)Nonidet P-40, 0.1%(w/v) SDS] containing protease inhibitors and quantified byuse ofthe Bradford assay (Bio-Rad Laboratories). Equivalent proteinamounts of each sample were analyzed. Protein detection onWestern blots was performed according to standard protocols.The antibodies used were: HER2 (Biogenex), cyclin B1 (sc-245),and CDC2 p34 (CDK1; sc-54) purchased from Santa Cruz Bio-technology and b-actin (A-5316) was purchased from Sigma.

Apoptosis and cell death analysisFor measuring apoptosis, the Annexin V and Dead Cell Assay

Kit (Millipore) was used according to the manufacturer's instruc-tions. Briefly, after treatment, the cells were incubated withAnnexin V and Dead Cell Reagent (7-AAD) for 20 minutes atroom temperature in the dark, and the events for dead, lateapoptotic, early apoptotic, and live cells were counted with theMuse Cell Analyzer (Millipore) and analyzed with MuseSoft1.4.0.0 (Millipore).

Cyclin B1 and cdc20 silencingFor transient transfection experiments, cells were transfected by

use of the Amaxa 4D-Nucleofector device, according to manu-facturer's instruction. For each electroporation reaction, 100 mL ofcomplete Nucleofector solution combined with 300 nmol/LsiRNA against Cyclin B1, cdc20, and scrambled siRNA as a control(duplex siRNAs were purchased from GE Dharmacon) under a

specific optimized program for siRNA delivery with the Nucleo-fector (SKBR3 E-009).

Exposure of fresh human breast cancer explants to T-DM1 exvivo

The studywas approvedby the ethics committee of theHospitaldel Mar and conducted following institutional guidelines. Freshtumor specimens from women with HER2-positive breast cancerundergoing routine cancer surgery, which were not needed fordiagnostic purposes, were collected to add ex vivo T-DM1 andassess its molecular effects according to our experience (16).Samples were sliced and cultured in RPMI1640 medium supple-mented with 10% FBS, 2 mmol/L L-glutamine, and 100 U/mLpenicillin–streptomycin for 120 hours in the absence (control) orpresence of T-DM1 (0.1 mg/mL). Specimens were fixed in 10%neutral-buffered formalin for 16 hours and embedded in paraffinthen assayed by IHC.

IHCThree-micrometer–thick paraffin sections from tissue blocks of

the tumors were stained for HER2 (Herceptest P980018/S010,Dako), cyclin B1(sc-245, Santa Cruz Biotechnology), Ki-67(GA62661-2, MIB1 clone, Dako), phosphorylated (Ser10) His-tone 3 (9701, Cell Signaling Technology), and active caspase-3(9664, Cell Signaling Technology) followed by incubation withan anti-rabbit Ig dextran polymer (Flexþ, Dako) and 3,30-diami-nobenzidine as chromogen in a Dako Link platform. HER2staining was scored following ASCP/CAP guidelines (14). For theother markers, the percentage of positive tumor cells was scored.

Statistical analysisData are presented as mean � SD. The GraphPad Prism soft-

ware was used to construct graphs and statistical analysis. Statis-tical significance was determined using Student t test or one-wayANOVA. P � 0.05 was considered as significant.

ResultsGeneration of T-DM1–resistant HER2-positive breast cancercells

We assessed the half maximal inhibitory concentration (EC50)values of T-DM1 in a panel of HER2-positive breast cancer cells.The median EC50 values for BT474, SKBR3, AU565, EFM-192A,HCC1954, andHCC1419 cell lines at 72 hourswere 0.025, 0.002,0.005, 0.009, 0.020, and 0.018 mg/mL, respectively. Four celllines, two sensitive to trastuzumab (SKBR3 and BT474) andtwo with primary resistance to trastuzumab (HCC1954 andHCC1419) were selected to generate T-DM1 resistance (TDR) byapplying a pulsed administration strategy of the drug, often usedfor the development of chemotherapy drug resistance (Fig. 1A;ref. 13). This protocol encompasses short pulses of drug treatmentfollowed by rest periods in drug-free media to allow the cells torecover from toxicity between treatments until a stable resistantphenotype is observed. Specifically, the procedure consisted ofthree consecutive cycles of 3 days on treatment followed by 3 daysoff treatment for each T-DM1 concentration of 1, 2, and 4 mg/mL.The entire 54-day protocol resulted in the generation of cells withvarying levels of T-DM1 resistance.

We established an exposure to0.1mg/mLof T-DM1 for 3days asa reference schedule to define resistance in our model. Thisschedule was selected on the basis of experiments performed in

Cyclin B1 and T-DM1 Acquired Resistance

www.aacrjournals.org Clin Cancer Res; 23(22) November 15, 2017 7009

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 5: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

D

A

HER2

β-Actin

B

HCC1954 HCC1954/TDRT-DM1

0hT-DM1

24hT-DM1 +

Chloroquine-24hT-DM1

0hT-DM1

24h

T-D

M1

Pha

lloid

inD

AP

IM

erge

d

SKBR3 SKBR3/TDRT-

DM

1P

hallo

idin

DA

PI

Mer

ged

HCC1419 HCC1419/TDR

T-D

M1

Phal

loid

inD

AP

IM

erge

d

T-DM1 +Chloroquine-24h

Parental (+Trastuzumab)

Parental (rituximab) Parental (rituximab)

HC

C14

19

% o

f Max

T-DM1 Trastuzumab

HC

C19

54SK

BR3

Resistant (+T-DM1)

Resistant (rituximab) Parental (+T-DM1)

Resistant (+Trastuzumab)

Resistant (rituximab)

% o

f Max

% o

f Max

C

10μm

Parental Resistant H

CC

1954

HC

C14

19S

KB

R3

T-DM10h

T-DM124h

T-DM1 +Chloroquine-24h

T-DM10h

T-DM124h

T-DM1 +Chloroquine-24h

T-DM10h

T-DM124h

T-DM1 +Chloroquine-24h

T-DM10h

T-DM124h

T-DM1 +Chloroquine-24h

Figure 2.

Pharmacodynamic characterization of T-DM1–resistant HER2-positive breast cancer cells. A, FISH assay for HER2 gene amplification. Paraffin-embedded cellpellets were prepared from each resistant cell line (HCC1419/TDR, SKBR3/TDR, and HCC1954/TDR; right) and the corresponding age and passage-matched parentalcells (HCC1419, SKBR3, and HCC1954; left). The level of HER2 amplification in cell nuclei was determined as the ratio HER2/CEP17. Nuclei are highlighted bythe blue fluorescent counter stain [40 , 6-diamidino-2-phenylindole (DAPI)]. Nuclei with high levels of HER2 gene amplification (red signal) are shown. Green signal(CEP17): centromere of chromosome 17. (scale bar: 10 mm). B, HER2 protein expression in parental and T-DM1–resistant cells. Expression of HER2 in whole-celllysates from the cell pairs under basal growth. b-Actin served as internal loading control. Western blot is from a representative experiment. C, Flow cytometricanalysis of T-DM1-receptor binding in parental and T-DM1–resistant cells. Cell line pairs were incubated with 7.5 mg/mL rituximab (a humanized anti-CD20IgG1 mAb, as a negative isotype control) or 7.5 mg/mL of T-DM1, for 30 minutes on ice. Trastuzumab at 7.5 mg/mL was used as positive control. Phycoerythrin (PE)-conjugated goat F(ab0)2 anti-human immunoglobulin was used as a secondary reagent. Representative flow cytometry histograms are shown. D, T-DM1internalization analysis. Cells were incubated with 1.5 mg/mL T-DM1 at 37�C for 15 minutes. In some conditions, cells were cotreated with chloroquine (50 mmol/L) toaccumulate T-DM1 intracellularly. Cells were fixed and stained with Cy3-conjugated anti-human (for T-DM1 detection; red), FITC-phalloidin (for distributionof the F actin filaments; green), and DAPI (a nuclear DNA stain; blue). Scale bar, 7.5 mm.

Sabbaghi et al.

Clin Cancer Res; 23(22) November 15, 2017 Clinical Cancer Research7010

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 6: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

MCF7 cells that do not overexpress HER2, whose growth wasunaffected until higher concentrations and longer exposure to T-DM1 were used. After approximately 2 months, three cell linesdeveloped resistance to T-DM1 (called HCC1954/TDR,HCC1419/TDR and SKBR3/TDR; Fig. 1B). We were unsuccessfulto generate BT474-resistant cells even after repeated attempts.HCC1954/TDR cells were completely resistant after 10 days ofexposure to T-DM1 at 0.1 mg/mL. HCC1419/TDR and SKBR3/

TDR cells decreased the sensitivity to the antiproliferative effectsof T-DM1 by 20%–30% at 0.1 mg/mL after 3 days. This level ofresistance, albeit modest, has been helpful to identify mechan-isms of resistance to several agents (13). T-DM1–resistant cellshad a growth rate similar to parental cells in vitro, as assessed bycell counting after 7 days of culture under the same conditions.The relative cell numbers of resistant versus parental cells were110% � 8.3% in HCC1954/TDR, 93% � 4% in HCC1419/TDR

BA

C

HCC1419Control T-DM1

HCC1419/TDRControl T-DM1

HCC1954Control T-DM1

HCC1954/TDRControl T-DM1

SKBR3Control T-DM1

SKBR3/TDRControl T-DM1

% C

ells

HCC1419 HCC1419/TDR

ControlT-DM1

ControlT-DM1

% C

ells

HCC1954 HCC1954/TDR

DNA content

% C

ells

SKBR3

DNA content

SKBR3/TDR

ControlT-DM1

ControlT-DM1

ControlT-DM1

ControlT-DM1

ResistantParental

T-D

M1

Con

trol

HC

C19

54T-

DM

1C

ontro

lH

CC

1419

T-D

M1

Con

trol

SKBR

3

Prop

idiu

mIo

dide

Pr

opid

ium

Iodi

de

Prop

idiu

mIo

dide

AnnexinV AnnexinV AnnexinV AnnexinV

HCC1954 HCC1954/TDR0

20

40

60

80

100

Cel

l dea

th (%

)

*

HCC1419 HCC1419/TDR0

20

40

60

80

100

Cel

l dea

th (%

)

*

SKBR3 SKBR3/TDR0

20

40

60

80

100

Cel

l dea

th (%

)

Control T-DM1

***

*

HCC1419 HCC1419/TDR0

200

400

600

800

*

HCC1954 HCC1954/TDR0

200

400

600

800

***

SKBR3 SKBR3/TDR0

200

400

600

800

Control T-DM1

**

***

Contro

l

T-DM1

TDR/C

ontro

l

TDR/T-

DM1 0

20406080

100120

% C

ells

Sub-G1

G1

SG2−MMultinucleated cells

Sub-G1

G1

SG2−MMultinucleated cells

Sub-G1

G1

SG2−MMultinucleated cells

Contro

l

T-DM1

TDR/C

ontro

l

TDR/T-

DM1 0

20406080

100120

% C

ells

Contro

l

T-DM1

TDR/C

ontro

l

TDR/T-

DM1 0

20406080

100120

% C

ells

Contro

l

T-DM1

TDR/C

ontro

l

TDR/T-

DM1 0

20406080

100120

% C

ells

LiveEarly apoptosisLate apoptosis/DeadDead cells

Contro

l

T-DM1

TDR/C

ontro

l

TDR/T-

DM1 0

20406080

100120

% C

ells

LiveEarly apoptosisLate apoptosis/DeadDead cells

Contro

l

T-DM1

TDR/C

ontro

l

TDR/T-

DM1 0

20406080

100120

% C

ells

LiveEarly apoptosisLate apoptosis/DeadDead cells

Fold

cha

nge

in G

2−M

(%)

Fold

cha

nge

in G

2−M

(%)

Fold

cha

nge

in G

2−M

(%)

Figure 3.

Cellular effects of T-DM1 in parental and resistant cells. A, The action of T-DM1 on cell-cycle distribution. Cell-cycle status was analyzed using BrdUrdincorporation and propidium iodide (PI) to assess DNA content by flow cytometry. Sensitive (HCC1954 HCC1419 and SKBR3) and T-DM1–resistant (HCC1954/TDR,HCC1419/TDR, and SKBR3/TDR) cells were seeded into 6-well plates at a density of 2 � 105 cells/well for 24 hours. Cells were then treated without (Control) orwith T-DM1 (0.1 mg/mL) for 24 hours and pulsed for an hour with BrdUrd prior to cell harvest and analysis. Left, representative flow cytometry plots from anexperiment performed in triplicate that is consistent with other biological replicates. Right, the bar chart shows the percentage of cells in the G0–G1, S,and G2–M phases of the cell cycle following T-DM1 treatment. A bar chart with statistics for each pair of cell lines showing the percentage of change in G2–Mpopulation after T-DM1 treatment assigning a value of 100% to the untreated condition is shown. � , P < 0.05; �� , P < 0.01; ��� , P < 0.001. B, Effects of T-DM1 inmicrotubules. The three pairs of sensitive and T-DM1–resistant cells were grown at low density on cover slips, and incubated with T-DM1 (0.1 mg/mL) for 24 hours.Cells were washed, fixed, and permeabilized before incubation with anti-a tubulin FITC-conjugated antibody. The nuclei were counterstained with DAPI.Representative confocal images of cells visualizing the organization of tubulin filaments (green) are shown. Right, a magnification of the picture shown and a phasecontrast image of the same area. Scale bar, 24 mm.C, T-DM1 is able to induce apoptosis in responsive cells. The three pairs of sensitive and resistant cellswere treatedwith or without T-DM1 (0.1 mg/mL) for 48 hours. The cells were harvested, washed with PBS, and subsequently stained with Annexin-V conjugated to FITC. Viable(Annexin-V�/PI�) preapoptotic (Annexin-Vþ/PI�), apoptotic (Annexin-Vþ/PIþ), and the residual damaged (Annexin-V�/PIþ) cells were quantified by flowcytometric analysis using the Muse Cell Analyzer. Representative flow cytometry dot plots of three independent experiments are shown. Annexin V incombination with PI can discriminate between among early apoptotic cells, late apoptotic cells, and cells that are in either the very late stages of apoptosis ornecrosis. Graphs show the average percentage of indicated subsets of cells from three experiments. A bar chart with the average percentage of total celldeath from three experiments with statistics is shown. � , P < 0.05; ��� , P < 0.001.

Cyclin B1 and T-DM1 Acquired Resistance

www.aacrjournals.org Clin Cancer Res; 23(22) November 15, 2017 7011

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 7: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

A

B

SKBR3 SKBR3/TR

+ +

CDK1

Cyclin B1

β-Actin

EFM-192A EFM-192A/TR

+ +

AU565 AU565/TR

+ +T-DM1 BT474 BT474/TR

+ +

D

BT474 SKBR3 AU565 EFM-192A HCC1954 HCC1419T-DM1 + + + + + +

Cyclin B1

β-Actin

CDK1

HCC1954 HCC1954/TDR0

1

2

3

4

CD

K A

ctiv

ity (A

492)

(rela

tive

to c

ontro

l)

***

HCC1419 HCC1419/TDR0

1

2

3 *

SKBR3 SKBR3/TDR0

1

2

3ControlT-DM1

*

BT474 BT474/TR0

1

2

3

CD

K A

ctiv

ity (A

492)

(rela

tive

to c

ontro

l)

*

SKBR3 SKBR3/TR0

1

2

3*

*

EFM-192A EFM-192A/TR0

1

2

3 ControlT-DM1*

**

Cyclin B1

β-Actin

HCC1419 Control T-DM1

P TDR P TDR

CDK1

SKBR3Control T-DM1P TDR P TDR

HCC1954 Control T-DM1

P TDR P TDR

AU565 AU565/TR0

1

2

3**

*

C HCC1954 HCC1419 SKBR3

Cyclin B1

β-Actin

Figure 4.

Effects of T-DM1 on CDK1/cyclin B1 kinase activity and expression in different HER2-positive breast cancer preclinical models. A, T-DM1 is able to upregulatethe expression of cyclin B1 in a number of T-DM1 sensitive HER2-positive breast cancer cell lines. Cells were treated with T-DM1 (0.1 mg/mL) for 24 hours.Protein expression levels of cyclin B1 andCDK1were evaluated byWestern blot analysis using 25 mg of protein cell lysate. The anti-b-actin antibodywas used to verifyequal protein loading. Representative image of three separate experiments is shown. B, Effects of T-DM1 on CDK1/cyclin B1 kinase activity and expressionin HER2-positive breast cancer cell with acquired resistance to trastuzumab. (Continued on the following page.)

Sabbaghi et al.

Clin Cancer Res; 23(22) November 15, 2017 Clinical Cancer Research7012

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 8: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

and 103% � 7% in SKBR3 (no statistical differences). The mor-phology of parental versus resistant cells was similar by opticmicroscopic observation. All resistant cell lines exhibited resis-tance features for at least one year after their generation. Of note,SKBR3/TDR cells retained a similar antiproliferative response totrastuzumab alone than parental cells (50% growth inhibition),suggesting that the resistance was associated to DM1 (Fig. 1C).Both parental and resistant cells showed similar EC50 valuesfor paclitaxel [HCC1954 (4.5 nmol/L), HCC1954/TDR (6.5nmol/L), HCC1419 (6.0 nmol/L), HCC1419/TDR (5.6 nmol/L),SKBR3 (3.2 nmol/L), SKBR3/TDR (3.1 nmol/L)].

T-DM1 binds to cell surface HER2 and is internalized inresistant cells

HCC1419/TDR and SKBR3/TDR cells retained the same level ofHER2 amplification and protein expression than parental cells(Fig. 2A and B). However, HCC1954/TDR cells displayeddecreased amplification ofHER2 and reduced HER2 protein (Fig.2A and B), and mRNA levels (6% of relative mRNA expression)comparedwithparental cells. ParentalHCC1954 cells had ameanof 40HER2 copies but they contained two additional subpopula-tions; 17% of the cells had less than 6 copies (some scored asamplified for having only one copy of CEP17) and 11.4% had6–10 HER2 copies. During the generation of the resistant cells,T-DM1 eradicated the cells with strongHER2 amplification after ashort exposure, allowing the emergence of subdominant cloneswith lowHER2 amplification (average number of 8HER2 copies,HER2 1þ by IHC). The emergence of HER2 low resistant cells inHCC1954 was confirmed in three independent experiments.Despite this important change in the population, the resistantcells still met the criteria for scoring them as HER2 amplified asthey havemore than 6HER2 copies (14). In addition, other genessuch as ORMDL3, STARD3, PPP1R1B, MIEN1 located in theminimal common regionof 17q12 amplificationwere also down-modulated as assayed by expression arrays. SKBR3/TDR andHCC1419/TDR did not have any significant differences in thelevels of HER2, ORMDL3, STARD3, PPP1R1B andMIEN1mRNAin comparisonwithparental cells, as assayedbyqRT-PCR.Wenextassayed the ability of T-DM1 to bind to cell surface HER2 by flowcytometric analysis. Although surface HER2 levels might vary inHCC1954/TDR compared with parental cells, T-DM1-HER2binding was preserved in all TDR cells (Fig. 2C).

We assayed whether T-DM1 was capable of inducing internal-ization upon binding to HER2 in paired sensitive and resistantcells (15). Cells were treated with 10 nmol/L T-DM1 for 15minutes at 37�C. After washing out the drug, cells were culturedfor 24 hours with or without 50 mmol/L chloroquine to accumu-late T-DM1 intracellularly. As shown in Fig. 2D, T-DM1 (red dots)

was internalized into both parental and resistant cells. The mag-nitude of T-DM1 internalization and intracellular pattern weresimilar in parental and resistant SKBR3 and HCC1419 cells. InHCC1954/TDR cells we also detected T-DM1 intracellularly, butto a lesser extent than in their parentalHER2-positive counterpart,in agreement with their lower HER2 amplification level and thesurface expression.

Effects of T-DM1 on G2–M arrest and mitotic catastrophe inparental versus resistant cells

T-DM1 increased significantly the percentage of cells in G2–

M phase while it induced a decrease in the S and G0–G1 phasesin the three parental cell lines (Fig. 3A). In the resistant cells, theeffects of T-DM1 on G2–M cell-cycle arrest were less pro-nounced than in parental cells. In HCC1954/TDR andHCC1419/TDR, there was no increase in G2–M. In SKBR3 therewas a significant increase following T-DM1 but to a much lesserdegree than in parental cells (Fig. 3A). Next, we evaluated theeffects of T-DM1 onmicrotubule arrangement. In parental cells,T-DM1 caused the formation of multinucleated giant cells withseverely defective microtubule arrangements (Fig. 3B). Previ-ously, it has been reported that T-DM1 causes tumor growthinhibition by mitotic catastrophe (17). These morphologicalterations suggestive of mitotic catastrophe were undetectedin T-DM1–resistant cells. As most cells undergoing mitoticcatastrophe are destined to die by apoptosis, we evaluated theeffects of T-DM1 on apoptosis induction. T-DM1 inducedapoptosis in HCC1954 and SKBR3 parental cells at 48 hoursbut the effect on the resistant counterparts was much lesspronounced under the same conditions (Fig. 3C).

T-DM1–resistant cells failed to induce CDK1/cyclin B1We hypothesized that resistant cells may have a defective cell-

cycle–regulatory machinery that does not allow T-DM1 to induceG2–M arrest and consequently, mitotic catastrophe. We focusedon the potential involvement of cyclin dependent kinase 1(CDK1) and cyclin B1 in T-DM1 resistance. Activation of theCDK1–cyclin B1 complexes are essential for progression into M-phase, but prolonged mitotic arrest (for example because of theinability of forming the mitotic spindle in cells treated withmicrotubule-affecting agents such as DM1) leads tomitotic catas-trophe (18). The activity of CDK1–cyclin B1 complex is regulatedmainly by the expression of cyclin B1 and by the phosphorylationstatus of the catalytic subunit CDK1. Degradation of cyclin B1 bythe proteasome after ubiquitination by themulti-subunit ubiqui-tin E3-ligase APC/Ccdc20 is essential for exiting mitosis. T-DM1exposure augmented cyclin B1 expression in a panel of HER2-positive parental breast cancer cells (except BT-474; Fig. 4A).

(Continued.) Acquired trastuzumab-resistant cells (BT474/TR, SKBR3/TR, AU565/TR, and EFM-192A/TR) have been generated by our group and used inthese experiments together with the corresponding age and passage-matched parental cell line (19). Cells were treated with T-DM1 (0.1 mg/mL) and harvested at24 hours. Whole-cell extracts were prepared from each experimental condition and CDK1/cyclin B1 kinase activity was measured with the nonradioactiveMESACUP Cdc2/CDK1 Kinase Assay. The results are reported as fold induction of OD 492 nm in arbitrary units (RLA) of treated sample using untreated condition asreference set at 1. Data are presented as mean � SD for three experiments. Cells treated as in the CDK1/cyclin B1 kinase activity assay experiment were lysedand Western blots were performed with equal amounts of cell lysate (25 mg protein). Expression of cyclin B1 and CDK1 was evaluated. b-Actin was used asan internal control. Shown are representative images from one of experiments. � , P < 0.05; �� , P < 0.01. C, Trastuzumab is not able to upregulate the expression ofcyclin B1 in a number of T-DM1–sensitive HER2-positive breast cancer cell lines. Cells were treated with trastuzumab (15 mg/mL) and T-DM1 (0.1 mg/mL) for24 hours and analyzed as inA. Shownare representative images fromoneof experiments.D,Effects of T-DM1 onCDK1/cyclin B1 kinase activity andexpression in pairsof HER2-positive sensitive and T-DM1–resistant breast cancer cells. HCC1954 and HCC1954/TDR, HCC1419-HCC1419/TDR, and SKBR3-SKBR3/TDR cells weretreated with T-DM1 (0.1 mg/mL) for 24 hours and analyzed for CDK1 kinase activity using the MESACUP Cdc2/CDK1 Kinase Assay Kit and cyclin B1 levels byWestern blot analysis as in Fig. 3C. Data are expressed as fold induction versus control arbitrarily set at 1. � , P < 0.05; ��� , P < 0.001.

Cyclin B1 and T-DM1 Acquired Resistance

www.aacrjournals.org Clin Cancer Res; 23(22) November 15, 2017 7013

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 9: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

We analyzed the effects of T-DM1 on the activity of CDK1.We first tested this effect in four parental HER2 positive celllines and in their trastuzumab resistant (TR) counterparts thatwe recently reported (19). These TR cell lines were included inthis analysis to confirm whether acquired trastuzumab resis-tance (that corresponds to the clinical setting in which T-DM1is approved) affects T-DM1 ability to induce CDK1 activity. Theresults showed a significant increase in CDK1 Kinase activity 24hours posttreatment with T-DM1 (0.1 mg/mL) in both sensitiveand trastuzumab-resistant cells by using the MESACUP Cdc2/Cdk1 Kinase Assay Kit with exception of the BT-474 cell pairs(Fig. 4B). In all the cell lines with acquired trastuzumabresistance, T-DM1 exposure decreased viable cells (cell numberrelative to control at 3 days of 20% � 3.2% in AU565/TR, 40%� 6% in EFM-192A, 17% � 3% in SKBR3/TR and 72% � 3% inBT474/TR) and cyclin B1 was induced following drug exposure(Fig. 4B). Trastuzumab alone did not induce cyclin B1 in thesecell lines (Fig. 4C).

We then assessed T-DM1 effects on the CDK1/cyclin B1complex kinase activity and cyclin B1 levels in the paired celllines parental and resistant to T-DM1. We performed a time–course study the HCC1954 and HCC1954/TDR cells. Theactivation of the mitotic kinase CDK1/cyclin B1 was detectedas early as 12 hours post-T-DM1 in parental HCC1954 cells (2-fold) but not in HCC1954/TDR cells (P < 0.05). Protein levelsof total CDK1 were unchanged in parental and resistant cells.Cyclin B1 protein levels were moderately increased (2-fold) at12 hours following T-DM1 treatment in parental cells in par-allel with CDK1 activity and they were maintained elevated upto 24 hours afterwards. This increase in the cyclin B1 levels wasundetected in resistant cells (Fig. 4D). A similar pattern ofcyclin B1 expression and appearance of CDK1/cyclin B1 kinaseactivity were observed with the other T-DM1–sensitive/resistantcell pairs after 24 hours of T-DM1 exposure (Fig. 4C). Theanalysis of the levels of cyclin B1 mRNA by qRT-PCR showed anonsignificant increase (Supplementary Fig. S1), suggestingpostranscriptional regulatory mechanisms.

Cyclin B1 mediates T-DM1 resistanceWe tested whether cyclin B1 knockdown could mediate

resistance to T-DM1 in parental cells. Cell lines were transfectedwith siRNA directed against cyclin B1 mRNA or with a suitablecontrol. Cyclin B1 levels were measured 24 and 48 hours afterthe transfection by Western blot confirming that they weresignificantly lower in the siRNA-transfected cells than in thesiControl (Fig. 5A). The paired cell lines transfected with thecyclin B1 siRNA or the siControl were exposed to T-DM1 andcell viability was assayed after 48 hours. Silencing of cyclin B1induced a significant resistance to T-DM1 in the three parentalcell lines (Fig. 5A).

We next tested whether increasing the levels of cyclin B1 inresistant cells might sensitize them to T-DM1. Cdc20 is a regu-latory subunit of the multi-subunit ubiquitin E3-ligase APC/Cthat is responsible of cyclin B1 degradation at the end of mitosis.We silenced cdc20 to promote cyclin B1 accumulation (20). Asshown in Fig. 5B, cyclin B1 protein level were increased followingcdc20 silencing in two of the three resistant cells. In HCC1954/TDR cells, no differences in T-DM1 sensitivity between control(scrambled siRNA) and cdc20-silenced cells were observed. How-ever, T-DM1 resistance was partially reverted in cdc20-silencedSKBR3/TDR and HCC1419/TDR cells (Fig. 5B).

Induction of cyclin B1 by T-DM1 in HER2-positive humanbreast cancer explants associates with apoptosis

To overcome, at least in part, the issue of the limited number ofcell lines, we tested the association between cyclin B1 inductionand T-DM1 antiproliferative effect in a panel of 18 fresh humanHER2-positive breast cancer explants. The results confirmed suchassociation. We assayed the basal levels of cyclin B1 in 18 HER2-positive breast cancers according to our experience in freshexplants (16). In 7 cases, cyclin B1 was undetected and in theremaining 11, the percentage of tumor cells with detected cyclinB1 staining ranged from 2% to 10% (Fig. 6A). A fraction of thesetissues were cultured ex vivo for 5 days in the presence of T-DM1 orcontrol and assayed formodulation in cyclin B1 expression. Therewere two main observations; first, cyclin B1 was induced in themajority of explants exposed to T-DM1 (12 of 18, 66.6% of thetumors), including those with undetected baseline levels; second,the percentage of tumor cells that stained positive for cyclin B1was dramatically increased inmany specimens (61.1%of cases), afinding consistent with the induction of a G2–M arrest by T-DM1(Fig. 6A and B). These findings support the effect of T-DM1 oninducing a persistent accumulation of cyclin B1 in tumor, whichwas a hallmark of T-DM1 effects in our panel of sensitive breastcancer cells. Of note, in 4 explants with cyclin B1 detected atbaseline, T-DM1 exposure did not further increase the percentageof expressing cells. We also assayed induction of apoptosis byexpression of active caspase-3, proliferation (Ki67), and activa-tion of theM-phase checkpoint kinases (phospho-Histone H3, p-H3; Fig. 6A). With regards to proliferation, Ki67 staining was notmodulated by T-DM1 after 5 days while reduction in p-H3staining paralleled the induction of apoptosis by T-DM1. Thelack of an effect on Ki67 is consistent with the fact that cellsarrested in G2–M also stain positive to Ki67 (Fig. 6A; ref. 21). Wefound two main patterns of cyclin B1 and apoptosis changesfollowing T-DM1 exposure: no/very low cyclin B1 accumulationand very low upregulation of apoptosis versus strong cyclin B1(accumulation) and high induction of apoptosis. Trastuzumabalone did not induce cyclin B1 in ex vivo explants (Fig. 6B). Thetumor cell areaswith cyclin B1 staining and the oneswith caspase-3 active staining did not frankly overlapped in the tissue sections,suggesting different stages of T-DM1 effects.

Fifteen explants were from diagnostic specimens derivedfrom patients that received neoadjuvant treatment without T-DM1. The other three were from metastatic patients. Tworeceived T-DM1 and had cyclin B1 and apoptosis inductionex vivo. One had de novometastatic disease, the explant was fromthe diagnostic breast cancer biopsy, and received T-DM1 assecond line achieving a partial response. The second patienthad bone and liver disease and after several lines of treatmentreceived T-DM1. The explant was obtained from a liver metas-tasis just before T-DM1 (Supplementary Fig. S2) and subse-quently had a partial response.

DiscussionWe have generated three different HER2-positive breast cancer

cell lines with various levels of acquired T-DM1 resistance byapplying a pulsed administration strategy of the drug, anapproach commonly used for the development of chemotherapydrug-resistant cancer cells (13). The resistant phenotype wasachieved within the first twomonths of drug exposure, suggestingthe emergence of a chemotherapy-driven mechanism. We chose

Sabbaghi et al.

Clin Cancer Res; 23(22) November 15, 2017 Clinical Cancer Research7014

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 10: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

initially four cells lines to develop resistance, but in one of them(BT474) we were unsuccessful. The three remaining cell lines,albeit not sufficient to establish the generalizability of the find-ings, were sufficient to test mechanistically the role of cyclin B1.Similarly, the degree of acquired resistance varied in the 3 cell linesbut a role of cyclin B1 in T-DM1 resistance was observed in all ofthem. The modest resistance observed in two of the cell linesclearly suggested that cyclin B1-independent mechanisms ofaction of T-DM1 remain active.

The generation of resistant cell lines in a short timeframe maybe caused by several mechanisms and may vary between celllines. In HCC1954, a specific finding was a marked reduction ofHER2 gene amplification after the first round of exposure to T-DM1. In parental HCC1954 cells, there was a predominant

subpopulation (�93% of cells) with high Her2 amplificationand a minority subpopulation (�7%) with low, but amplifiedHER2 gene. An early clonal selection of the subpopulation withlowerHER2 amplification following T-DM1 exposure appears tocontribute to resistance. Regardless of this, the rapid emergenceof resistance, also in cell lines that retain the same level of HER2amplification, suggests a mechanistic link with the cytotoxicDM1 component rather than to trastuzumab, by as yet unknownmechanisms such as epigenetic and/or cellular pathway rewir-ing. SKBR3 T-DM1–resistant cells retained sensitivity to trastu-zumab, whereas the other two remained trastuzumab resistant.This suggests that multiple mechanisms of resistance may coex-ist in the same cell line. Additional studies are needed tounderstand if these mechanisms coexist in a single cell or

A

B

cdc20

β-Actin

Cyclin B1

0 0.10

40

80

120

Cel

l num

ber

(% o

f con

trol)

Cel

l num

ber

(% o

f con

trol)

HCC1954/TDR

T-DM1 [μg/mL]0 0.1

0

40

80

120

T-DM1 [μg/mL]

siControlsicdc20

SKBR3/TDR

*

24h 48h

SKBR3

24h 48h24h 48h

HCC1954 HCC1419

Cyclin B1

β-Actin

0 0.10

40

80

120

T-DM1 [μg/mL]

***

HCC1419

0 0.10

40

80

120HCC1419/TDR

T-DM1 [μg/mL]

*

0 0.10

40

80

120siControlsiCyclin B1

**

SKBR3

T-DM1 [μg/mL]

0 0.10

40

80

120

T-DM1 [μg/mL]

***

HCC1954

48h 72h

HCC1419/TDR48h 72hHCC1954/TDR

48h 72h

SKBR3/TDR

Figure 5.

Geneticmodulation of cyclin B1 affectsthe viability of cancer cells treatedwith T-DM1.A,Depletion of cyclin B1 inthe parental drug-sensitive cellsreduced T-DM1 sensitivity. ParentalT-DM1–sensitive cells weretransfected with target-specific-cyclinB1 siRNA and scrambled control siRNAby using a Nucleofector. Aftertransfection, cells were recovered andused for experiments. Viabilityexperiments were performed on analiquot of transfected cells afterseeding in 12-well plates at aconcentration of 10–15� 103 cells/welland allowed to adhere overnight. Cellswere then incubated with T-DM1 (0.1mg/mL). Duplicated samples wereharvested and counted at 48 hourswith the Scepter automated cellcounter. The results are presented asthe mean � SD from experimentsreplicated five times (�� , P < 0.01;��� , P < 0.001). The remainingtransfected cells were analyzed byWestern blot analysis. Total cellularprotein was extracted from cells at 48hours after siRNA transfection.Western blots confirmed that cyclin B1was downregulated. b-Actin wasblotted for loading control. B, Increaseof cyclin B1 in the drug-resistant cellsaugments their response to T-DM1.T-DM1–resistant cellswere transfectedwith target-specific cdc20 siRNA andscrambled control siRNA by using aNucleofector. After transfection, cellswere recovered and used for cellviability and Western blotexperiments. Cells were incubatedwith T-DM1 (0.1 mg/mL). Cell numberwas determined after 72 hoursposttransfection with Scepter. Theresults are presented as the mean �SD from triplicate experiments(� , P < 0.05; ��� , P < 0.001). Lysateswere prepared 48 and 72 hoursposttransfection and analyzed for thelevels of cyclin B1 and cdc20 byWestern blot.

Cyclin B1 and T-DM1 Acquired Resistance

www.aacrjournals.org Clin Cancer Res; 23(22) November 15, 2017 7015

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 11: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

whether this represents tumor cell heterogeneity (as inHCC1954 relative to HER2 amplification). Nevertheless, theantimitotic essence of DM1 should be considered as a sourceof heterogeneity generation.

The cytotoxic effect of T-DM1 might be impaired by inefficientinternalizationor enhancedpercentage ofHER2–T-DM1complexthat is recycled back to the cell surface (3). It is believed that theHER2/T-DM1 conjugate enters cancer cells via the clathrin-depen-dent endocytosis pathway. However, a clathrin-independent

mechanism, such as caveolae membranes composed mainly bycaveolin-1 has also been demonstrated (22). Furthermore, it hasbeen shown that the high endocytic activity of stem cell–likebreast cancer cells make them particularly sensitive to T-DM1(23).Wehave shown that T-DM1was internalized and exhibited asimilar intracellular pattern in parental and resistant cells, albeitHCC1954/TDR cells had less T-DM1 detected intracellularly.Overall, it appeared that the pathway mediating HER2-T-DM1endocytosis was intact in resistant cell lines.

CB

Baseline

100

80

60

40

20

0

T-DM1 Baseline T-DM1 Baseline T-DM1 Baseline T-DM1

Cyclin B1 c-casp3 Ki-67 p-H3A

Per

cent

age

of tu

mor

ce

lls

Cyc

linB

1c-

casp

3

Ki6

7

Cas

e 11

Cas

e 4

Control T-DM1

Annotation416

1718

62

75810129143131119

−5 +50

1

Trastuzumab

Cyc

lin B

1c-

casp

3C

yclin

B1

c-ca

sp3

50 μm

50 μm

60

40

20

0

60

40

20

0

40

20

0

Figure 6.

Effects of T-DM1 added ex vivo to fresh human breast cancer explants. Formalin-fixed paraffin-embedded (FFPE) blocks were prepared for control andtreated breast tumor at five days after the T-DM1 treatment.A,Graphs showing the percentage of cells positive for eachmarker. Cyclin B1, active caspase-3, Ki67, andp-H3 were determined in basal control condition and after treatment with T-DM1 for 5 days. B, Representative IHC images of control and T-DM1–treated tumors.Two representative examples of staining for cyclin B1 and caspase-3 active (apoptosis) are shown. Scale bar, 50 mm. These tumors were also treated withtrastuzumab (15 mg/mL) and the results are also shown.C,Hierarchical cluster analysis of T-DM1 effects on cyclin B1 and active caspase-3 in a total of 18 human breastcancers. Each row represents an effect on expression of the paired control- and T-DM1–treated samples, and each column, a single IHC marker, includingcyclin B1 and active caspase-3. Upregulation of expression is displayed in red, and no effect in white.

Sabbaghi et al.

Clin Cancer Res; 23(22) November 15, 2017 Clinical Cancer Research7016

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 12: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

In parental cells, T-DM1 induced G2–M arrest and mitoticcatastrophe. The phenomenon of "mitotic catastrophe" consid-ered a mode of cell death per se resulting from prolonged mitoticarrest is now believed to represent a prestage of apoptosis or evennecrosis or senescence (5). In two of the three cell lines sensitive toT-DM1, the mitotic arrest induced by T-DM1 preceded apoptosis.Mitotic catastrophe did not occur in resistant cells suggesting thatthemachinery for inducing cell-cycle arrest at theG2–Mphasewasdisrupted.

This led us to hypothesize a potential involvement of CDK1and cyclin B1, themembers of mitotic promoting factor complex,in T-DM1 resistance (18). Entry into mitosis is initiated by CDK1and binding of CDK1 to cyclin B1 is essential for its activation.CDK1/cyclin B1 complex and the kinase activity of CDK1 iscontrolled by cyclin B1 accumulation. During mitosis, chromo-some segregation is facilitated by the kinetochore, an assembly ofproteins built on centromeric DNA that attach chromosomes tospindle microtubules. After the last unattached kinetochore isattached to microtubules and the chromosomes are properlyaligned, the mitotic checkpoint is switched off. CDK1 is theninactivated as cyclin B is rapidly degraded, and cells progressthrough anaphase, undergo cytokinesis, and exit mitosis (24).However, in the presence of an antimitotic agents (such as DM1),the impossibility of assembling the mitotic spindle activates themitotic checkpoint arresting the cells in mitosis for a prolongedperiod and theymay undergomitotic catastrophe followed by celldeath (25).

In the three parentalHER2-positive breast cancer cells, cyclinB1induction and CDK1/cyclin B1 activation were observed follow-ing T-DM1 treatment and were maintained elevated for up to 24hours. However, T-DM1 failed to raise cyclin B1 levels and,consequently, CDK1/Cyclin B1 activity in T-DM1–resistant cells.Furthermore, silencing of cyclin B1 induced resistance to T-DM1 inparental cell lines while increasing the levels of cyclin B1 inresistant cells sensitized them to T-DM1 in two out of the threecell lines. The failure to generate T-DM1 BT474–resistant cellsunder our experimental protocolmay be related, at least in part, tothe inability of T-DM1 to upregulate cyclin B1.

Other CDK–cyclin complexes have been previously reported asimplicated in resistance to anti-HER2 therapies. For instance,cyclin E has a role in trastuzumab resistance and treatment withCDK2 inhibitors has been proposed for tumors displaying cyclinE amplification/overexpression (26, 27). A relationship of CDK4/cyclin D1 activity in resistance to trastuzumab and lapatinib hasbeen also reported and high levels of cyclin D1 predicted poorresponse to trastuzumab (28). Preclinical studies have also shownthat residual cells surviving within 48 hours following T-DM1treatment began to reenter the cell cycle and the sequentialtreatment with CDK4/6 inhibitors suppressed the proliferationof these residual/resistant clones (29). A clinical trial with theCDK4/6 inhibitor palbociclib in combination with T-DM1(NCT01976169) in HER2-positive patients is underway. On theother hand, our results showing that CDK1/Cyclin B1 activity isneeded for T-DM1 action suggests a note of caution regardingpossible combinations of T-DM1 with pan-CDKs inhibitors orselective CDK1 inhibitors (30).

In human breast cancer, cyclin B1 expression has been associ-ated with poor survival (31) and is a prognostic proliferationmarker in lymphnode–negative breast cancer cohorts (32). In ourseries of fresh HER2-positive breast tumor explants, the basallevels of cyclin B1 were not significantly related to T-DM1 apo-

ptotic or antiproliferative effects. Instead, we found that theinduction of cyclin B1 following T-DM1 exposure ex vivo washighly associated to induction of apoptosis and reduced tumorcell proliferation.

Interestingly, two patients with metastastic breast cancer had aresponse to T-DM1 and in both of them cyclin B1was upregulatedby T-DM1 ex vivo. These anecdotal data suggest that cyclin B1induction in explants may be associated to clinical response. Onthe other hand, the lack of a predictive effect of baseline cyclin B1expression is in linewith anobservation in 7HER2-positive breastcancer patients that had been treated at Hospital del Mar (Barce-lona, Spain) with T-DM1 as part of a neoadjuvant clinical trial.Four of them had detectable levels of cyclin B1, of whom 3achieved a pathologic complete response (pCR). In the 3 patientswith undetected baseline cyclin B1, one pCR was achieved. Albeitpatients received additional anti-HER2 agents or systemic che-motherapy as part of their neoadjuvant treatment, these few casessuggest that baseline cyclin B1 expression is not a prerequisite toachieve a pCR with a neoadjuvant regimen including T-DM1. Weplan to prospectively test cyclin B1 induction as a pharmacody-namics assay (i.e., serial biopsies comparing baseline cyclin B1expression with expression at an early time point after T-DM1treatment) to predict T-DM1 clinical benefit in a GEICAM (Span-ish Breast Cancer Research Group) study in the near future.

A few additional mechanisms of T-DM1 resistance havebeen proposed. For instance, those factors that reduce theintracellular DM1 load per cell, namely the overexpression ofmultidrug resistance (MDR) proteins or an impaired lysosom-al degradation of trastuzumab that might limit the subsequentrelease of DM1 into the cytoplasm (3, 33). Although we cannotrule out a potential role of these mechanisms in our models,we believe that MDR did not play a significant role sincepaclitaxel, a microtubule-stabilizing agents and a substrate ofMDR, exhibited the same cytotoxicity in parental and inresistant cells.

With regards to alterations in the endosome pathways, webelieve that the detection of intracellular T-DM1 in the resistantcells, as well as the effects of cdc20 silencing on restoring T-DM1response in resistant cells, limits the potential role of this putativemechanism of resistance. Nonetheless, a biparatopic HER2-tar-geting ADC containing the tubulysin variant AZ13599185 dem-onstrated superior antitumor activity over T-DM1 in varioustumor models including T-DM1 refractory. The new ADC bytargeting two nonoverlapping epitopes on HER2 can induceHER2 receptor clustering, which in turn promotes robust inter-nalization, lysosomal trafficking, and degradation (34). Also, ithas been suggested a combination of inhibitors of the chaperoneHSP90 that promote HER2 targeting to lysosomes and its degra-dation suggested as a new strategy to improve therapywith T-DM1(35). In addition to this, other novel anti-HER2 antibody–drugconjugate offer promising activity in T-DM1–pretreated breastcancer (36, 37). Other potential mechanisms of resistance thatcome from the trastuzumab part of the T-DM1molecule have notproven its clinical utility when assayed in patient samples fromclinical trials (9, 11). A mechanism of T-DM1 resistance that hasbeen reported preclinically is the presence of the HER3 ligand,heregulin, that reduced the activity of T-DM1 in breast cancer cellsby causing HER2/HER3 dimerization thus strongly activating thePI3K pathway; and this effect was reversed by the addition ofpertuzumab, aHER2–HER3dimerization inhibitor.However, thecombination of T-DM1 and pertuzumab in the clinic has not

www.aacrjournals.org Clin Cancer Res; 23(22) November 15, 2017 7017

Cyclin B1 and T-DM1 Acquired Resistance

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 13: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

proven sufficiently superior to T-DM1 alone, suggesting thismechanism operates at most in a small proportion of patients(38). Finally, T-DM1 is able to alter genes related with immuneresponse and this can influence the response in patients (39).

In short, the studies presented here show that cyclin B1 induc-tion allowed to differentiate T-DM1 sensitive parental cells fromtheir counterparts with acquired resistance. The induction ofcyclin B1 in T-DM1–sensitive cells was independent of their priortrastuzumab sensitivity. The mitotic arrest consequence of thefailure in assembling themitotic spindle leads to sustainedCDK1/Cyclin B1 kinase activity, a hallmark of mitotic catastrophe that isresolved by apoptosis. In fresh human HER2-positive breastcancer explants, the induction of cyclin B1 by T-DM1 correlateswith apoptosis, suggesting that a pharmacodynamic assay to testcyclin B1 induction in breast cancer patients treated with T-DM1may help to identify early the patients more likely to benefit fromthis drug.

Disclosure of Potential Conflicts of InterestJ. Albanell reports receiving speakers bureau honoraria from and is a

consultant/advisory board member for Roche. No potential conflicts of interestwere disclosed by the other authors.

Authors' ContributionsConception and design: M. Sabbaghi, G. Gil-G�omez, P. Eroles, J. Arribas,F. Rojo, A. Rovira, J. AlbanellDevelopment of methodology: M. Sabbaghi, G. Gil-G�omez, C. Guardia,O. Arpi, S. Menendez, S. Zazo, C. Chamizo, P. Gonz�alez-Alonso, F. Rojo,A. Rovira, J. AlbanellAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): M. Sabbaghi, G. Gil-G�omez, S. Servitja, O. Arpi,S. García-Alonso, S. Menendez, M. Arumi-Uria, L. Serrano, A. Muntasell,M. Martínez-García, A. Lluch, F. Rojo

Analysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis):M. Sabbaghi, G. Gil-G�omez, C. Guardia, S. Servitja, S.García-Alonso, S. Menendez, M. Salido, A. Muntasell, P. Gonz�alez-Alonso,J. Madoz-G�urpide, I. Tusquets, A. Pandiella, F. Rojo, J. AlbanellWriting, review, and/or revision of the manuscript: M. Sabbaghi,G. Gil-G�omez, S. Servitja, S. García-Alonso, L. Serrano, A. Muntasell,M. Martínez-García, P. Eroles, J. Arribas, I. Tusquets, A. Lluch, A. Pandiella,F. Rojo, A. Rovira, J. AlbanellAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): O. Arpi, J. Madoz-G�urpide, F. Rojo, J. AlbanellStudy supervision: I. Tusquets, F. Rojo, A. Rovira, J. Albanell

AcknowledgmentsThe authors would like to thank Marc Bataller and Raul Pe~na for technical

assistance and comments. Flow cytometry analysiswas technically supported byOscar Fornas, head of Universitat Pompeu Fabra (UPF) flow cytometry corefacility. We thank Fundaci�o Cellex (Barcelona) for a generous donation to theHospital del MarMedical Oncology Service. We also thank the patients for theirgenerous participation in the study.

Grant SupportThis work was supported by ISCiii (CIBERONC CB16/12/00481, RD12/

0036/0051, RD12/0036/0070, RD12/0036/0003, PIE15/00008, PI13/00864,PI15/00146, PI15/00934, PI15/01617, PT13/0010/0005), Generalitat de Cat-alunya (2014 SGR 740), and the "Xarxa de Bancs de tumors sponsored by PlaDirector d'Oncologia de Catalunya (XBTC). MINECO through BFU2015-71371-R grant supported work in A. Pandiella's laboratory. Our work wassupported by the EU through the regional funding development program(FEDER). P. Gonz�alez-Alonso was supported by Fundaci�on Conchita R�abagode Jim�enez Díaz grant.

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 March 10, 2017; revised June 29, 2017; accepted August 11, 2017;published OnlineFirst August 18, 2017.

References1. Lewis Phillips GD, Li G, Dugger DL, Crocker LM, Parsons KL, Mai

E, et al. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res 2008;68:9280–90.

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

3. Barok M, Joensuu H, Isola J. Trastuzumab emtansine: mechanisms ofaction and drug resistance. Breast Cancer Res 2014;16:209.

4. Lopus M. Antibody-DM1 conjugates as cancer therapeutics. Cancer Lett2011;307:113–8.

5. Vitale I, Galluzzi L, Castedo M, Kroemer G. Mitotic catastrophe: a mech-anism for avoiding genomic instability. Nat Rev Mol Cell Biol 2011;12:385–92.

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

7. deMeloGagliatoD, JardimDL,MarchesiMS,HortobagyiGN.Mechanismsof resistance and sensitivity to anti-HER2 therapies inHER2þbreast cancer.Oncotarget 2016;7:64431–46.

8. LoRusso PM, Weiss D, Guardino E, Girish S, Sliwkowski MX. Trastuzumabemtansine: a unique antibody-drug conjugate in development for humanepidermal growth factor receptor 2-positive cancer. Clin Cancer Res2011;17:6437–47.

9. Kim SB, Wildiers H, Krop IE, Smitt M, Yu R, Lysbet de Haas S, et al.Relationship between tumor biomarkers and efficacy in TH3RESA, a phaseIII study of trastuzumab emtansine (T-DM1) vs. treatment of physician'schoice in previously treated HER2-positive advanced breast cancer. Int JCancer 2016;139:2336–42.

10. Wang J, Song P, Schrieber S, Liu Q, Xu Q, Blumenthal G, et al. Exposure-response relationship of T-DM1: insight into dose optimization forpatients with HER2-positive metastatic breast cancer. Clin Pharmacol Ther2014;95:558–64.

11. 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.

12. Gebhart G, Lamberts LE, Wimana Z, Garcia C, Emonts P, Ameye L, et al.Molecular imaging as a tool to investigate heterogeneity of advancedHER2-positive breast cancer and to predict patient outcome under trastu-zumab emtansine (T-DM1): the ZEPHIR trial. Ann Oncol 2016;27:619–24.

13. McDermott M, Eustace AJ, Busschots S, Breen L, Crown J, Clynes M,et al. In vitro development of chemotherapy and targeted therapy drug-resistant cancer cell lines: a practical guide with case studies. FrontOncol 2014;4:40.

14. Wolff AC,HammondME,HicksDG,DowsettM,McShane LM, Allison KH,et al. Recommendations for human epidermal growth factor receptor 2testing in breast cancer: American Society of Clinical Oncology/College ofAmerican Pathologists clinical practice guideline update. J Clin Oncol2013;31:3997–4013.

15. Montero J, Garcia-Alonso S, Ocana A, Pandiella A. Identification oftherapeutic targets in ovarian cancer through active tyrosine kinase pro-filing. Oncotarget 2015;6:30057–71.

16. Rojo F, Gonzalez-Navarrete I, Bragado R, Dalmases A, MenendezS, Cortes-Sempere M, et al. Mitogen-activated protein kinasephosphatase-1 in human breast cancer independently predictsprognosis and is repressed by doxorubicin. Clin Cancer Res 2009;15:3530–9.

Sabbaghi et al.

Clin Cancer Res; 23(22) November 15, 2017 Clinical Cancer Research7018

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 14: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

17. Barok M, Tanner M, Koninki K, Isola J. Trastuzumab-DM1 causes tumourgrowth inhibition by mitotic catastrophe in trastuzumab-resistant breastcancer cells in vivo. Breast Cancer Res 2011;13:R46.

18. CastedoM, Perfettini JL, Roumier T, Kroemer G. Cyclin-dependent kinase-1: linking apoptosis to cell cycle andmitotic catastrophe. Cell Death Differ2002;9:1287–93.

19. Zazo S, Gonzalez-Alonso P, Martin-Aparicio E, Chamizo C, Cristobal I,Arpi O, et al. Generation, characterization, and maintenance of trastuzu-mab-resistant HER2þ breast cancer cell lines. Am J Cancer Res 2016;6:2661–78.

20. Wan L, Tan M, Yang J, Inuzuka H, Dai X, Wu T, et al. APC(Cdc20)suppresses apoptosis through targeting Bim for ubiquitination anddestruction. Dev Cell 2014;29:377–91.

21. Gerdes J, Lemke H, Baisch H, Wacker HH, Schwab U, Stein H. Cell cycleanalysis of a cell proliferation-associated human nuclear antigen definedby the monoclonal antibody Ki-67. J Immunol 1984;133:1710–5.

22. 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.

23. Diessner J, Bruttel V, Stein R, Horn E, H€ausler S, Dietl J, et al. Targeting ofpreexisting and induced breast cancer stem cells with trastuzumab andtrastuzumab emtansine (T-DM1). Cell Death Dis 2014;5:e1149.

24. Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changingparadigm. Nat Rev Cancer 2009;9:153–66.

25. Weaver BA, Cleveland DW. Decoding the links between mitosis, cancer,and chemotherapy: the mitotic checkpoint, adaptation, and cell death.Cancer Cell 2005;8:7–12.

26. ScaltritiM, Eichhorn PJ, Cortes J, Prudkin L, AuraC, Jimenez J, et al. Cyclin Eamplification/overexpression is a mechanism of trastuzumab resistance inHER2þ breast cancer patients. Proc Natl Acad Sci U S A 2011;108:3761–6.

27. Tormo E, Adam-Artigues A, Ballester S, Pineda B, Zazo S, Gonzalez-AlonsoP, et al. The role of miR-26a and miR-30b in HER2þ breast cancertrastuzumab resistance and regulation of the CCNE2 gene. Sci Rep2017;7:41309.

28. Tanioka M, Sakai K, Sudo T, Sakuma T, Kajimoto K, Hirokaga K, et al.Transcriptional CCND1 expression as a predictor of poor response toneoadjuvant chemotherapy with trastuzumab in HER2-positive/ER-posi-tive breast cancer. Breast Cancer Res Treat 2014;147:513–25.

29. Witkiewicz AK, Cox D, Knudsen ES. CDK4/6 inhibition provides a potentadjunct to Her2-targeted therapies in preclinical breast cancer models.Genes Cancer 2014;5:261–72.

30. AsgharU,Witkiewicz AK, TurnerNC, Knudsen ES. The history and future oftargeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov2015;14:130–46.

31. AaltonenK, Amini R-M,Heikkil€a P, Aittom€aki K, TamminenA,NevanlinnaH, et al. High cyclin B1 expression is associated with poor survival in breastcancer. Br J Cancer 2009;100:1055–60.

32. Koliadi A, Nilsson C, Holmqvist M, Holmberg L, de La Torre M, WarnbergF, et al. Cyclin B is an immunohistochemical proliferation marker whichcan predict for breast cancer death in low-risk node negative breast cancer.Acta Oncol 2010;49:816–20.

33. 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.

34. Li JY, Perry SR,Muniz-Medina V,Wang X,Wetzel LK, Rebelatto MC, et al. Abiparatopic HER2-targeting antibody-drug conjugate induces tumorregression in primary models refractory to or ineligible for HER2-targetedtherapy. Cancer Cell 2016;29:117–29.

35. Raja SM, Desale SS, Mohapatra B, Luan H, Soni K, Zhang J, et al. Markedenhancement of lysosomal targeting and efficacy of ErbB2-targeted drugdelivery by HSP90 inhibition. Oncotarget 2016;7:10522–35.

36. DillonRL, Chooniedass S, PremsukhA, AdamsGP, Entwistle J,MacDonaldGC, et al. Trastuzumab-deBouganin conjugate overcomes multiplemechanisms of T-DM1 drug resistance. J Immunother 2016;39:117–26.

37. Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, et al. DS-8201a, a novel HER2-targeting ADC with a novel DNA topoisomerase Iinhibitor, demonstrates a promising antitumor efficacy with differentia-tion from T-DM1. Clin Cancer Res 2016;22:5097–108.

38. Perez EA, Barrios C, EiermannW, ToiM, ImYH,Conte P, et al. Trastuzumabemtansinewith orwithout pertuzumab versus trastuzumab plus taxane forhuman epidermal growth factor receptor 2-positive, advanced breastcancer: primary results from the phase III MARIANNE study. J Clin Oncol2017;35:141–8.

39. Muller P, Kreuzaler M, Khan T, Thommen DS, Martin K, Glatz K, et al.Trastuzumab emtansine (T-DM1) renders HER2þ breast cancer highlysusceptible to CTLA-4/PD-1 blockade. Sci Transl Med 2015;7:315ra188.

www.aacrjournals.org Clin Cancer Res; 23(22) November 15, 2017 7019

Cyclin B1 and T-DM1 Acquired Resistance

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696

Page 15: Defective Cyclin B1 Induction in Trastuzumab- emtansine (T-DM1) … · Cancer Therapy: Preclinical Defective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1) Acquired Resistance

2017;23:7006-7019. Published OnlineFirst August 18, 2017.Clin Cancer Res   MohammadA Sabbaghi, Gabriel Gil-Gómez, Cristina Guardia, et al.   Acquired Resistance in HER2-positive Breast CancerDefective Cyclin B1 Induction in Trastuzumab-emtansine (T-DM1)

  Updated version

  10.1158/1078-0432.CCR-17-0696doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2017/08/18/1078-0432.CCR-17-0696.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/23/22/7006.full#ref-list-1

This article cites 39 articles, 10 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/23/22/7006.full#related-urls

This article has been cited by 6 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://clincancerres.aacrjournals.org/content/23/22/7006To request permission to re-use all or part of this article, use this link

on December 14, 2020. © 2017 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 18, 2017; DOI: 10.1158/1078-0432.CCR-17-0696