the discovery and preclinical development of asg-5me ... · hector avina~ 1, claudia guevara1,...

10
Large Molecule Therapeutics The Discovery and Preclinical Development of ASG-5ME, an AntibodyDrug Conjugate Targeting SLC44A4-Positive Epithelial Tumors Including Pancreatic and Prostate Cancer Michael Mattie 1 , Arthur Raitano 1 , Kendall Morrison 1 , Karen Morrison 1 , Zili An 1 , Linnette Capo 1 , Alla Verlinsky 1 , Monica Leavitt 1 , Jimmy Ou 1 , Rossana Nadell 1 , Hector Avi ~ na 1 , Claudia Guevara 1 , Faisal Malik 1 , Ruth Moser 2 , Steven Duniho 2 , Jeffrey Coleman 1 , Ying Li 1 , Daniel S. Pereira 1 , Fernando Do ~ nate 1 , Ingrid B.J. Joseph 1 , Pia Challita-Eid 1 , Dennis Benjamin 2 , and David R. Stover 1 Abstract Here, we report the development of an antibodydrug conju- gate, ASG-5ME, which targets the solute carrier receptor SLC44A4. SLC44A4 is a member of a family of putative choline transporters that we show to be markedly upregulated in a variety of epithelial tumors, most notably prostate and pancreatic cancer. SLC44A4 is normally expressed on the apical surface of secretory epithelial cells, but in cancer we show expression is not restricted to the luminal surface in advanced and undifferentiated tumors. ASG-5ME consists of a human IgG2 anti-SLC44A4 antibody conjugated through a cleavable linker to the microtubule-disrupt- ing agent monomethylauristatin E. It has potent antitumor activ- ity in both cell lineand patient-derived xenograft models of pancreatic and prostate cancers. Combination studies with ASG- 5ME and nab-paclitaxel demonstrated combination effect in both pancreatic and prostate tumor models. Altogether, the data pre- sented here suggest that ASG-5ME may have the potential to offer a new therapeutic option for the treatment of pancreatic and prostate cancers. Mol Cancer Ther; 15(11); 267987. Ó2016 AACR. Introduction SLC44A4, also known as CTL4, is a member of the family of solute carrier proteins known as SLC44A1-5 or choline transport- er-like proteins (CTL1-5). Although this family of proteins is thought to be involved in choline transport, only SLC44A1 and SLC44A2 have been demonstrated to have choline transport activity (17). SLC44A4 has not been shown to be involved in choline transport, but it has been linked with acetylcholine synthesis and transport (8) as well as uptake of thiamine pyro- phosphate, the phosphorylated form of vitamin B1 (9). Through RNA expression proling and IHC analysis, we have shown SLC44A4 to be selectively expressed in a broad range of epithelial tumors while having restricted expression in normal tissues. This represents the rst thorough evaluation of SLC44A4 expression in both normal and cancer tissues in the literature. A link between SLC44A4 expression and tumorigenesis has not been reported. However, due to the expression observed in prostate and pancreatic cancers, where over 90% of cases are positive, and low expression in normal tissues SLC44A4 is a good target for an antibodydrug conjugate (ADC). Here, we describe the expression of SLC44A4 in pancreatic and prostate cancers and report on the preclinical development of ASG-5ME, an ADC composed of an SLC44A4-specic human IgG2 anti- body conjugated with monomethyl auristatin E (MMAE) through a protease cleavable valine-citruline linker. Data pre- sented here support that ASG-5ME may represent a new treat- ment option for patients with prostate cancer and pancreatic cancer and that SLC44A4 is a novel target in these cancers and others of epithelial origin. Materials and Methods Immunohistochemistry For immunohistochemical (IHC) studies, a monoclonal anti- body (mAb; M5-121.131) against an extracellular domain of human SLC44A4 (largest extracellular loop) was generated using standard mouse hybridoma technology. The antibody was vali- dated for SLC44A4 specicity by correlating staining patterns on formalin-xed, parafn-embedded (FFPE) sections of various xenografts and cell lines with available transcript expression data. The specicity of the antibody was also conrmed by FACS analysis with recombinant cell lines expressing SLC44A4 and control cell lines not expressing the target. SLC44A4-specic IHC was performed on tissue microarrays (TMA) of prostate cancer, pancreatic cancer, and normal tissues (US Biomax, TriStar Technologies, Accumax). After deparafniza- tion and rehydration, tissue sections were treated for antigen retrieval with trypsin (MP Biomedicals) for 10 minutes at 37 C 1 Agensys Inc., an Afliate of Astellas Pharma Inc., Santa Monica, California. 2 Seattle Genetics, Inc., Bothell, Washington. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Author: Michael Mattie, Agensys Inc., an afliate of Astellas Pharma Inc., 1800 Stewart Street, Santa Monica, CA 90404. Phone: 424-280- 5400; Fax: 424-280-5044; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-16-0225 Ó2016 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 2679 on January 11, 2021. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst August 22, 2016; DOI: 10.1158/1535-7163.MCT-16-0225

Upload: others

Post on 20-Sep-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Discovery and Preclinical Development of ASG-5ME ... · Hector Avina~ 1, Claudia Guevara1, Faisal Malik1, Ruth Moser2, Steven Duniho2, Jeffrey Coleman1,Ying Li1, Daniel S. Pereira1,

Large Molecule Therapeutics

The Discovery and Preclinical Development ofASG-5ME, anAntibody–DrugConjugateTargetingSLC44A4-Positive Epithelial Tumors IncludingPancreatic and Prostate CancerMichael Mattie1, Arthur Raitano1, Kendall Morrison1, Karen Morrison1, Zili An1,Linnette Capo1, Alla Verlinsky1, Monica Leavitt1, Jimmy Ou1, Rossana Nadell1,Hector Avi~na1, Claudia Guevara1, Faisal Malik1, Ruth Moser2, Steven Duniho2,Jeffrey Coleman1, Ying Li1, Daniel S. Pereira1, Fernando Do~nate1, Ingrid B.J. Joseph1,Pia Challita-Eid1, Dennis Benjamin2, and David R. Stover1

Abstract

Here, we report the development of an antibody–drug conju-gate, ASG-5ME, which targets the solute carrier receptor SLC44A4.SLC44A4 is a member of a family of putative choline transportersthat we show to bemarkedly upregulated in a variety of epithelialtumors, most notably prostate and pancreatic cancer. SLC44A4 isnormally expressed on the apical surface of secretory epithelialcells, but in cancer we show expression is not restricted tothe luminal surface in advanced and undifferentiated tumors.ASG-5ME consists of a human IgG2 anti-SLC44A4 antibody

conjugated through a cleavable linker to themicrotubule-disrupt-ing agent monomethylauristatin E. It has potent antitumor activ-ity in both cell line– and patient-derived xenograft models ofpancreatic and prostate cancers. Combination studies with ASG-5ME and nab-paclitaxel demonstrated combination effect in bothpancreatic and prostate tumor models. Altogether, the data pre-sented here suggest that ASG-5MEmay have the potential to offera new therapeutic option for the treatment of pancreatic andprostate cancers. Mol Cancer Ther; 15(11); 2679–87. �2016 AACR.

IntroductionSLC44A4, also known as CTL4, is a member of the family of

solute carrier proteins known as SLC44A1-5 or choline transport-er-like proteins (CTL1-5). Although this family of proteins isthought to be involved in choline transport, only SLC44A1 andSLC44A2 have been demonstrated to have choline transportactivity (1–7). SLC44A4 has not been shown to be involved incholine transport, but it has been linked with acetylcholinesynthesis and transport (8) as well as uptake of thiamine pyro-phosphate, the phosphorylated form of vitamin B1 (9).

Through RNA expression profiling and IHC analysis, we haveshown SLC44A4 to be selectively expressed in a broad range ofepithelial tumors while having restricted expression in normaltissues. This represents the first thorough evaluation of SLC44A4expression in both normal and cancer tissues in the literature. Alink between SLC44A4 expression and tumorigenesis has notbeen reported. However, due to the expression observed inprostate and pancreatic cancers, where over 90% of cases are

positive, and low expression in normal tissues SLC44A4 is agood target for an antibody–drug conjugate (ADC). Here, wedescribe the expression of SLC44A4 in pancreatic and prostatecancers and report on the preclinical development of ASG-5ME,an ADC composed of an SLC44A4-specific human IgG2 anti-body conjugated with monomethyl auristatin E (MMAE)through a protease cleavable valine-citruline linker. Data pre-sented here support that ASG-5ME may represent a new treat-ment option for patients with prostate cancer and pancreaticcancer and that SLC44A4 is a novel target in these cancers andothers of epithelial origin.

Materials and MethodsImmunohistochemistry

For immunohistochemical (IHC) studies, a monoclonal anti-body (mAb; M5-121.131) against an extracellular domain ofhuman SLC44A4 (largest extracellular loop) was generated usingstandard mouse hybridoma technology. The antibody was vali-dated for SLC44A4 specificity by correlating staining patterns onformalin-fixed, paraffin-embedded (FFPE) sections of variousxenografts and cell lines with available transcript expression data.The specificity of the antibody was also confirmed by FACSanalysis with recombinant cell lines expressing SLC44A4 andcontrol cell lines not expressing the target.

SLC44A4-specific IHC was performed on tissue microarrays(TMA) of prostate cancer, pancreatic cancer, and normal tissues(US Biomax, TriStar Technologies, Accumax). After deparaffiniza-tion and rehydration, tissue sections were treated for antigenretrieval with trypsin (MP Biomedicals) for 10 minutes at 37�C

1Agensys Inc., an Affiliate of Astellas Pharma Inc., Santa Monica,California. 2Seattle Genetics, Inc., Bothell, Washington.

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

Corresponding Author: Michael Mattie, Agensys Inc., an affiliate of AstellasPharma Inc., 1800 Stewart Street, Santa Monica, CA 90404. Phone: 424-280-5400; Fax: 424-280-5044; E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-16-0225

�2016 American Association for Cancer Research.

MolecularCancerTherapeutics

www.aacrjournals.org 2679

on January 11, 2021. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 22, 2016; DOI: 10.1158/1535-7163.MCT-16-0225

Page 2: The Discovery and Preclinical Development of ASG-5ME ... · Hector Avina~ 1, Claudia Guevara1, Faisal Malik1, Ruth Moser2, Steven Duniho2, Jeffrey Coleman1,Ying Li1, Daniel S. Pereira1,

and then incubated with the primary antibody M5-131.121 orisotype control antibody mouse IgG1k for 1 hour. Expression ofSLC44A4 was detected with the Biogenex Super Sensitive Poly-mer-HRP IHC Detection Kit (Biogenex Laboratories Inc.).

Expression in patient cancer specimenswas scored qualitativelybased on a combination of average staining intensity of tumorcells: (0) none, (1) weak, (2) mild, (3) moderate, and (4) strong,and approximate percentage of positively staining tumor cells: (0)none; (1) >0–25%; (2) >25%–50%; (3) >50%–75%; and (4)>75%. They were then assigned to one of three different catego-ries: samples where�50% of tumor cells had an average intensityscore of 2, 3, or 4 were considered high/moderate and all otherpositives were considered low and the remainder negative. Nor-mal tissue expression was descriptive.

Generation of recombinant SLC44A4 proteinThe first extracellular domain of SLC44A4, amino acids 50–

227, was generated using a modified version of the pAPTag5vector system (GenHunter Corp.), in which the Alkaline phos-phatase fusion gene was removed. 293T cells were transfectedwith the vector by calcium phosphate precipitation and selectedwith zeocin to derive a stable cell line secreting the recombinantprotein. 293T recombinants were propagated in 293-SFM IImedium (Life Technologies) in a 25-L Wave bag. Medium washarvested, filtered, and the recombinant protein was purified bymetal chelate chromatography on a Ni-NTA column (Qiagen).The protein was eluted with imidazole, dialyzed into PBS pH 7.2,and quantitated by BCA assay.

Cell cultureCell lines were obtained from the indicated vendors, followed

by a limited number of passages after thawing (> 5 passages) togenerate aliquots for freezing down to create a central master cellbank. Cell line authenticationwas performed on banked cell linesby short tandem repeat (STR) profiling carried out by GE Health-care, SeqWright Genomic Services using the Promega PowerPlex16HSkit. All studies describedhere utilized themaster cell bank asthe source of cells for respective studies. The PC-3 cell line wasobtained from ATCC (catalog number CRL-1435) on March 1,2012, and STR profiling performed on June 21, 2012. From thisparental PC-3 cell line, we created the PC3-Neo and PC3-SLC44A4 recombinant lines (human and monkey) which werebanked and STR profiling was performed on September 4, 2013.HT-29 and OVCAR-5 cells were obtained from NCI (catalognumbers 0507335 and 0507337), and STR profiling was per-formed on March 16, 2011. VCaP was obtained from ATCC onJanuary 27, 2009 (catalog number CRL-2876) and 22Rv1 cellswere obtained fromATCC on February 3, 2012, and STR profilingwas performed on July 24, 2013.

Expression of SLC44A4 in cell linesHuman (PC3) and rodent (3T3 clone 7, Rat1, 300.19) recom-

binant cell lines overexpressing SLC44A4 were generated byretroviral transduction using the replication deficient pSRalpha-MSV-TKneo amphotropic retroviral vector system (10). Cells wereinfected with retrovirus-containing media encoding SLC44A4.Stable-expressing cells were derived by selection for neomycinresistance using G418 (Life Technologies). Control cells weregenerated similarly but with retrovirus expressing only the neo-mycin drug resistance gene. Stable SLC44A4 expression wasmonitored by FACS and Western blotting.

Generation of SLC44A4-specific human mAbsAGS-5M2, the parental antibody used to create ASG-5ME, was

selected from a large panel of SLC44A4-specific human antibo-dies. This panel was generated by immunizing human IgG2producing Xenomice (11) with B300.19 cells engineered toexpress SLC44A4. Lymph node cells from immunized mice werefused with Sp2/0-AG14 B-cell hybridoma cells (ATCC) by electrocell fusion using an ECM2100 electro cell manipulator (BTX).SLC44A4-specific hybridomas were identified by screeninghybridoma supernatants for binding to cells engineered to expressloop one of the multi-transmembrane SLC44A4 protein but notto the nonexpressing parental cells.

Generation of CHO host cells expressing AGS-5M2Total RNA was extracted from the antibody-producing hybrid-

oma cells using TRIzol Reagent (Life Technologies) according tothe manufacturer's suggested protocol. cDNA synthesis was gen-erated from total RNA using the RACE cDNA Amplification kit(Clontech) according to themanufacturer's protocol. The variablehuman heavy and kappa light chains were amplified using RACEprimer and a reverse primer within the constant region of thehuman light and heavy chains, sequenced and cloned into amammalian cell expression vector encoding human IgG2 andkappa constant domains. The plasmid was transfected intoCHOK1SV host cells (Lonza) to generate a stable antibody-pro-ducing cell line according to the manufacturer's protocol.

Generation of ASG-5MEASG-5MEwasmadeby conjugating AGS-5M2withMMAEvia a

protease-cleavable maleimidocaproyl valine citruline p-amino-benzyl alcohol dipeptide linker (Supplementary Fig. S1). MMAEwas attached to reduced cysteines in the antibody hinge region,and linked to reduced cysteines of the antibody as previouslydescribed (12).

Antibody and ADC affinity for SLC44A4FACS analysis was used to determine affinity of antibodies and

ADCs for SLC44A4. Recombinant PC3 cells expressing eitherhuman or cynomolgus SLC44A4 or the neomycin resistance gene(negative control) were maintained at 37�C in 5% CO2 in mediacontaining RPMI-1640 (Gibco) supplemented with 10% fetalbovine serum (Omega Scientific). The cells were reconstitutedin FACS buffer, and the AGS-5M2 and H3-1.4 isotype controlantibodies as well as the ASG-5ME ADC and its isotype controlH3-1.4-vcE (negative control for conjugated mAb) ADCs weremixedwith cells in a dilution curve atfinal concentrations rangingfrom 0.01 to 160 nmol/L. The samples were tested in triplicate.Cells were incubated with antibodies or ADCs at 4�C on a plateshaker overnight. Following a wash, cells were incubated with thedetection antibody for 1 hour, washed, and read on the flowcytometer. Detectable binding of antibodies over the range of testconcentrations (MFI values of samples subtracted from theirrespective negative control antibodies) was analyzed using theone-site binding, nonlinear regression analysis in GraphPadPrism version 4 (Graphpad Software, Inc.). The KD (equilibriumdissociation constant) and Bmax (maximum specific binding) arereported.

Confocal internalization studiesRecombinant PC3 cells expressing SLC44A4 were seeded onto

poly-D-lysine coated 8-well chamber slides (2.5 � 104 cells per

Mattie et al.

Mol Cancer Ther; 15(11) November 2016 Molecular Cancer Therapeutics2680

on January 11, 2021. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 22, 2016; DOI: 10.1158/1535-7163.MCT-16-0225

Page 3: The Discovery and Preclinical Development of ASG-5ME ... · Hector Avina~ 1, Claudia Guevara1, Faisal Malik1, Ruth Moser2, Steven Duniho2, Jeffrey Coleman1,Ying Li1, Daniel S. Pereira1,

well in RPMI media) overnight and non-adherent cells were sub-sequently washed off. Cells were then incubated with 10 mg/mLASG-5ME for 1 hour at 4�C (on ice) or 24 hours at 37�C. Afterthe incubation period, unbound antibody was washed off withPBS, and cells were fixed in 4% paraformaldehyde for 20minutesat room temperature. Cells were then permeabilized in PBS þ0.1% Triton-X-100 for 15 minutes and non-specific labeling wasblocked in PBS þ 10% normal goat serum. Cell surface andintracellular ASG-5ME was visualized by incubating cells withAlexa Fluor 488–labeled goat anti-human IgG (Invitrogen). Lyso-somes were visualized by staining with lysosome-associatedmembrane protein 1, LAMP1 (mouse CD107a clone H4A3, BDBiosciences) and a secondary antibody, Alexa Fluor 568–labeledgoat anti-mouse IgG (Invitrogen). Nuclei were visualized withTOPRO-3 Iodide (Invitrogen) and coverslips weremounted usingProlong Gold anti-fade reagent (Invitrogen) for imaging. High-resolution laser scanning confocal image sections were acquiredusing a Leica TCS SP5-II (63� oil immersion objective; NA¼ 1.4)and Alexa Fluor 488/568 were scanned sequentially to minimizefluorophore cross-talk and false-positive colocalization.

In vitro cytotoxicityExponentially growing recombinant PC3 cells expressing

SLC44A4 or the neomycin drug resistance gene with a viabilityof 95% or greater were plated in fresh RPMI-1640 (Gibco-Invi-trogen) media containing phenol red supplemented with 10%

FBS. Cells were left overnight and treated with ADCs and anisotype control. After 5 days of treatment and incubation at 37�Cand 5% CO2, cell viability was measured following 1-hour incu-bation at 37�C with Presto Blue Reagent (Invitrogen). Sampleswere analyzed using a Synergy Microplate reader. Survival valueswere plotted using Graph Pad Prism to derive EC50 values using acurve-fitting analysis model for nonlinear curve regression, sig-moidal dose response with variable slope formula.

In vivo activity studiesFour- to 5-week-old ICR-SCIDmice (Taconic) weremaintained

and used at Agensys' animal facility using Agensys' InstitutionalAnimal Care and Use Committee (IACUC)–approved protocols.Studies were performed with either cell line–derived xenograftmodels or patient-derived tumor xenografts (PDX) developed atAgensys. Human tumor xenografts were established either sub-cutaneously or orthotopically in immunodeficient ICR SCIDmiceby injectionof human tumor cells or fragments of PDXs. SLC44A4expression in all xenograft models used was confirmed by IHCanalysis. Tumor growth was monitored using caliper measure-ments every 3 to 4 days until the end of study. Tumor volume wascalculated as width2 � length/2, where width is the smallestdimension and length is the largest. Mean tumor volume datafor each group were plotted over time with standard error bars. Astatistical analysis of the tumor volumedata for the last day beforeanimal sacrifice was performed using the Kruskal–Wallis test.

Table 1. Protein expression in prostate and pancreatic cancers

Prostate cancer Pancreatic cancerScore Samples, n Percentage of total Score Samples, n Percentage of total

High/moderate 182 85 High/moderate 138 67Low 22 10 Low 48 24Negative 10 5 Negative 19 9Total positive 204/214 95 Total positive 186/205 91

A

D E

B C

Figure 1.

SLC44A4 expression in prostate and pancreatic adenocarcinoma. A, low-grade prostate adenocarcinoma. B, high-grade prostate adenocarcinoma. C,metastatic prostate adenocarcinoma in bone. D, low-grade pancreatic ductal adenocarcinoma. E, high-grade pancreatic ductal adenocarcinoma. Notethe polarized distribution of SLC44A4 toward the apical surface in low-grade, well-differentiated adenocarcinomas, which is lost in the high-grade,poorly differentiated tumors.

SLC44A4, an ADC Target for Pancreatic and Prostate Cancer

www.aacrjournals.org Mol Cancer Ther; 15(11) November 2016 2681

on January 11, 2021. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 22, 2016; DOI: 10.1158/1535-7163.MCT-16-0225

Page 4: The Discovery and Preclinical Development of ASG-5ME ... · Hector Avina~ 1, Claudia Guevara1, Faisal Malik1, Ruth Moser2, Steven Duniho2, Jeffrey Coleman1,Ying Li1, Daniel S. Pereira1,

Pairwise comparisons were made using Tukey test procedures(two-sided) to protect the experiment-wise error rate. This imple-mentation of the Tukey test was performed on the ranks of thedata. The percent tumor growth inhibition in each treated groupversus a control group was calculated as [(Control � Controlbaseline) � (Treated � Treated baseline)] / (Control � Controlbaseline) � 100%. Details on dosing regimens for individualstudies are provided in respective tables orfigures. Choice of dosesgiven for drug combination studies was based on prior dosetitration studies for each drug dosed individually.

ResultsIdentification of SLC44A4 and gene expression profiling

To identify new targets overexpressed in cancer versus normaltissue, we performed discovery experiments using suppressionsubtractive hybridization. SLC44A4 was selected from a subtrac-tion of prostate cancer minus benign prostatic hyperplasia. Geneexpression of SLC44A4 was examined in a wider range of normaland cancer tissue specimens throughmining of public microarraydatasets contained within Oncomine Powertools cancer expres-sion database (https://powertools.oncomine.com). Tumor typeswith expression observed to behigher than corresponding normaltissues include breast, gastric, ovarian, pancreatic, and possibly asubset of lung cancers (Supplementary Fig. S2). SLC44A4 geneexpression in normal tissues is found to be generally low, with theexception of gastrointestinal tract and prostate.

SLC44A4 protein expression in patient cancer specimens andnormal tissues

IHC staining of FFPE confirmed SLC44A4 protein expressionin patient tumors. Most notably, 85% of primary prostatecancer specimens and distant metastases examined showed amedium to high level of staining for SLC44A4 and 67% ofpancreatic tumors had a medium to high level of staining.Greater than 90% of pancreatic and prostate tumors werepositive for SLC44A4 expression (Table 1). We observed polar-ized distribution of SLC44A4 toward the apical surface in lowgrade, well-differentiated tumors, which is lost in high grade,poorly differentiated tumors (Fig. 1). Protein expressionincreased with higher disease grade in both pancreatic andprostate tumors. While the specific focus discussed here is inregard to pancreatic and prostate cancers, we did observeSLC44A4 protein expression in other indications, consistentwith gene expression profiling data mentioned above.

To assess SLC44A4 expression in normal tissues, 225 sampleswere tested representing 36 different human tissues (Table 2).Specific positive staining for SLC44A4was observedmainly in theepithelia of prostate, lung bronchioles, gastro-intestinal tract, asubset of tubules in the kidney cortex, fallopian tube, bladder,ureter, and uterine endometrium. SLC44A4 expression was alsoseen in theductal epitheliumof some samples of liver (bile ducts),breast, salivary gland, esophagus, pancreas, skin (sweat glands).The expression of SLC44A4 was generally seen with a polarizeddistribution toward the apical surface of the cells, with theextracellular portion of SLC44A4 facing toward the lumen andnot immediately adjacent to the intravascular space (Fig. 2).SLC44A4 expression was not observed in adrenal, bladder, bonemarrow, brain heart, ileum, lymph node, ovary, parathyroid,pituitary, rectum, skeletal muscle, spleen, testis, thymus, andthyroid tissues.

SLC44A4 expression in human cancer cell linesWe observed very weak levels of expression across a large panel

of cancer cell lines when looking to identify suitable in vitro cellline models. For some cell lines, SLC44A4 expression increaseswhen implanted and grown as tumor xenografts in vivo. The levelof expression is briefly maintained for several hours after harvest-ing and digestion of tumors and growth in cell culture conditions;however, the level of expression decreases back to initial levelsafter subsequent passages. One example of this phenomenon isillustrated with the OVCAR-5 ovarian cancer cell line (Supple-mentary Data S3). Use of an in vitro cell line model with stablehigh levels of expression required the generation of PC3 cellsengineered to express SLC44A4 recombinantly.

Antibody and ADC characterizationAGS-5M2 was selected among a panel of anti-SLC44A4 mAbs

produced in the Xenomouse screened for specific binding andhigh affinity for SLC44A4 recombinant protein and endogenouslyor recombinantly expressed in human cancer cell lines. Themicrotubule-disrupting agent MMAE was conjugated to theAGS-5M2 antibody as previously described to produce the ADCASG-5ME. Results from affinity binding studies with recombin-ant PC3 cells engineered to express human SLC44A4 demonstrat-ed similar affinities (Kd) for the parental antibody, AGS5-M2

Table 2. Summary of SLC44A4 expression in normal tissues

OrganPositivecases, n

Totalcases

Positivecases, %

Prostate 8 8 100Lung 4 11 36Fallopian tube 3 3 100Bladder, transitional epithelium 3 3 100Ureter 2 3 67Small intestine 9 10 90Colon 9 10 90Rectum 7 7 100Kidney—cortex 11 11 100Stomach 9 10 90Salivary gland 1 1 100Uterus, endometrium 3 4 75Liver, bile ducts 5 9 56Kidney—medulla 2 3 67Pancreas, ductal epithelium 4 8 50Breast 3 10 30Tonsil, epithelium, interfolliculararea

1 4 25

Esophagus 2 8 25Skin 2 10 20Uterine cervix 1 8 13Adrenal gland 0 3 0Blood PBL 0 2 0Bone marrow 0 5 0Brain 0 11 0Eye 0 1 0Heart 0 4 0Lymph node 0 5 0Ovary 0 8 0Parathyroid 0 1 0Pituitary gland 0 2 0Placenta 0 3 0Spinal cord 0 2 0Spleen 0 8 0Striated muscle 0 4 0Testis 0 9 0Thymus gland 0 8 0Thyroid 0 3 0

Mattie et al.

Mol Cancer Ther; 15(11) November 2016 Molecular Cancer Therapeutics2682

on January 11, 2021. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 22, 2016; DOI: 10.1158/1535-7163.MCT-16-0225

Page 5: The Discovery and Preclinical Development of ASG-5ME ... · Hector Avina~ 1, Claudia Guevara1, Faisal Malik1, Ruth Moser2, Steven Duniho2, Jeffrey Coleman1,Ying Li1, Daniel S. Pereira1,

(1.6 nmol/L) and the ADCASG-5ME (1.7 nmol/L; Fig. 3A). UsingPC3 cells engineered to express cynomolgus SLC44A4, we dem-onstrated similar affinities of AGS-5M2 (3.9 nmol/L) and ASG-5ME (1.4 nmol/L; Fig. 3B). The homology between human andcynomolgusmonkey SLC44A4 is 96%. These studies demonstrat-ed that conjugation of AGS-5M2 with vcMMAE did not signifi-cantly impact the binding affinity or Bmax for human or cyno-molgus monkey SLC44A4 and that affinities to human andcynomolgus monkey SLC44A4 are similar.

Internalization of ASG-5MELaser scanning confocal fluorescence microscopy was used to

visualize internalized ASG-5ME (Fig. 4). Recombinant PC3 cells

expressing SLC44A4 cells incubated at 4�C showed distinct cellsurface localization of ASG-5ME (Fig. 4A, green), with minimalevidence of ASG-5ME internalization (Fig. 4C). Lysosomes visu-alized using an antibody to lysosome-associated membrane pro-tein 1 (LAMP1; Fig. 4B–C, red) appeared as discrete punctatecytosolic staining. At 37�C for 24 hours, surface membranelocalization of ASG-5ME ADC was dramatically reduced and thepredominant distribution of ADC localization appeared as dis-crete intracellular punctate staining throughout the cytosol (Fig.4D–F, green). The ASG-5MEwas shown to colocalizewith LAMP1(Fig. 4F, arrow, yellow), indicating that within 24 hours, ASG-5ME was trafficked to the lysosomes in the prostate cancer cellsand targeted for subsequent degradation.

Figure 3.

Binding affinity of AGS-5M2 and ASG-5ME to human and cynomolgusmonkey SLC44A4. Cells were stained with serially diluted amounts of AGS-5M2 and ASG-5MEand detected with PE-labeled anti-human IgG detection antibodies to determine the affinity for cell surface SLC44A4. A, binding of AGS-5M2 and ASG-5MEto human SLC44A4 on PC3 cells engineered to express human SLC44A4. B, binding of AGS-5M2 and ASG-5ME on PC3 cells engineered to expresscynomolgus SLC44A4.

A

C D

B

Figure 2.

SLC44A4 expression in normaltissues. A, bronchiolar epithelium. B,colonic mucosal epithelium. C,prostate glandular epithelium. D,subset of tubules within renal cortex.Note the polarized distribution ofSLC44A4 toward the apical surface inall normal epithelium.

SLC44A4, an ADC Target for Pancreatic and Prostate Cancer

www.aacrjournals.org Mol Cancer Ther; 15(11) November 2016 2683

on January 11, 2021. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 22, 2016; DOI: 10.1158/1535-7163.MCT-16-0225

Page 6: The Discovery and Preclinical Development of ASG-5ME ... · Hector Avina~ 1, Claudia Guevara1, Faisal Malik1, Ruth Moser2, Steven Duniho2, Jeffrey Coleman1,Ying Li1, Daniel S. Pereira1,

Cytotoxicity studies with ASG-5METhe potency and efficacy of ASG-5ME to induce cell death was

evaluated in PC3 cells engineered to express the SLC44A4 antigen.We observed potent dose-dependent cytotoxicity in PC3-SLC44A4 cells treated with ASG-5ME with an EC50 of �2nmol/L (Fig. 5). ASG-5ME was not cytotoxic on mock PC3-Neocells that do not express the SLC44A4 antigen. Moreover, thenative antibody AGS-5M2, and the control ADC did not causecytotoxicity in either PC3-SLC44A4 or PC3-Neo cells, indicatingthat the activity observed with the ADC was solely attributable tothe released MMAE after specific binding to the SLC44A4-expres-sing cells.

In vivo activity studiesHuman cancer xenograftmodels were selected for in vivo testing

on the basis of having favorable SLC44A4 expression. A summaryof selected ASG-5ME in vivo activity studies in prostate, pancreas,colon, andovarian xenograftmodels is shown inTable 3.Notably,we observed significant tumor growth inhibition in both cell line–

and patient-derived tumor xenografts when implanted eitherorthotopically or subcutaneously in immunodeficient mice, andwithin androgen-dependent and castration-resistant prostate can-cer models. While activity was also observed in colon and ovariancancer models, here we focus on pancreatic and prostate cancermodels due to the high percentages of positivity of SLC44A4expression in the tumors of these two patient populations. IHCstaining of SLC44A4 expression in untreated tumors is shown inthe panels for the selectedmodels (Fig. 6A–C). In all of our single-agent activity studies ASG-5ME was administered biweekly for atotal of four doses. Treatment of established LAPC-9AI patient-derived prostate tumor xenografts with 1 or 3 mg/kg ASG-5MEresulted in statistically significant (P < 0.0001) tumor growthinhibition (TGI) of 88.3% and 93.3%, respectively, at the end ofthe study (day 27) compared with the control ADC (Fig. 6A). Inthe VCaP prostate xenograft model, treatment with 5mg/kg ASG-5ME significantly inhibited tumor growth by 95% at day 38 (P <0.0001) at the end of the study (day 52) compared with thecontrol ADC(Fig. 6B). In theAG-Panc4patient-derived pancreatic

Figure 4.

Trafficking of ASG-5ME to thelysosomes in PC3-SLC44A4 cells.Recombinant PC3-SLC44A4 cells wereincubated with ASG-5ME for 1 hour at4�C to inhibit endocytosis (A–C) or 24hours at 37�C (D–F). Cell surface andintracellular ASG-5ME were visualizedby confocal fluorescence microscopy.ASG-5ME is shown in green; lysosomalmarker LAMP1 is shown in red; TOPRO-3-stained nuclei are shown in blue.Colocalization of signals for internalizedantibodywith LAMP1 (F, arrow) is shownin yellow. Magnification bar, 10 mm.

125

100

75

50

25

0

125

100

75

50

25

0

% S

urvi

val

% S

urvi

val

PC3-SLC44A4 PC3-Neo

ASG-5ME ASG-5MEAGS-5M2 AGS-5M2Control ADC Control ADC

ASG-5ME IC50: 290 ng/mL1.9 nmol/L

0.01 0.1 1 10 100

1,00

010

,000

0.01 0.1 1 10 100

1,00

010

,000

ng/mL ng/mL

Figure 5.

Cytotoxicity of ASG-5ME on PC3-SLC44A4 and PC3-Neo cells. Cytotoxicity was measured after treatment with serial dilutions of AGS-5M2, ASG-5ME, and anisotype control ADC for 3 days in PC3-SLC44A4 and PC3-Neo cells.

Mattie et al.

Mol Cancer Ther; 15(11) November 2016 Molecular Cancer Therapeutics2684

on January 11, 2021. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 22, 2016; DOI: 10.1158/1535-7163.MCT-16-0225

Page 7: The Discovery and Preclinical Development of ASG-5ME ... · Hector Avina~ 1, Claudia Guevara1, Faisal Malik1, Ruth Moser2, Steven Duniho2, Jeffrey Coleman1,Ying Li1, Daniel S. Pereira1,

tumor xenograft model (Fig. 6C), treatment with 4 mg/kg ASG-5ME significantly inhibited tumor growth by 87.1% (P < 0.0001)at the end of the study (day 68) compared with the control ADC.

In vivo combination studies were performed to assess whetherASG-5ME can enhance activity of standard-of-care agents forprostate or pancreatic cancer. Agents tested included enzaluta-mide, gemcitabine, erlotinib, and nab-paclitaxel. Analysis ofcombination data revealed that ASG-5ME only significantlyenhanced antitumor activity of nab-paclitaxel. Antitumor activityof ASG-5ME, nab-paclitaxel, or the combination was evaluated inAG-Panc4 pancreatic and LAPC-9AI prostate xenograft models. Inthe AG-Panc4 model (Fig. 6D), ASG-5ME (1 mg/kg) or nab-paclitaxel (100 mg/kg) administered every 4 days for a total of3 doses both demonstrated modest antitumor activity comparedwith the control, resulting in 40.3% (P¼ 0.0100) and 32.6% (P¼0.0384) tumor growth inhibition, respectively, on day 29. Whengiven in combination, a significant improvement in activity wasobserved resulting in 72.7% (P < 0.0001) tumor growth inhibi-tion. The combination activity was superior to single-agent ASG-5ME (P ¼ 0.0043) or nab-paclitaxel (P ¼ 0.0009). In the LAPC-9AI model (day 13), neither ASG-5ME (0.5 mg/kg) nor nab-paclitaxel (60 mg/kg), both administered every four days for atotal of four doses, demonstrated antitumor activity as singleagents, while nab-paclitaxel at 120 mg/kg demonstrated signifi-cant antitumor activity compared with the control (40.3% TGI,p ¼ 0.0476) (Fig. 6E). However, in combination, ASG-5MEsignificantly enhanced activity of nab-paclitaxel dosed at 60mg/kg resulting in 84% tumor growth inhibition (P < 0.0001).Additionally, tumor regression (76.8%, P < 0.0001) was observedwhen ASG-5ME (0.5 mg/kg) was combined with the higher dose(120 mg/kg) of nab-paclitaxel. Pairwise comparisons of single-agent ASG-5ME or nab-paclitaxel at 60 or 120 mg/kg to eithercombination were significantly different (P < 0.0001 for allcomparisons).

DiscussionOur studies describe the identification of SLC44A4 as a novel

differentially expressed target in pancreatic and prostate cancers.Relatively little literature has been published regarding the endog-enous function or expression of SLC44A4. Based on sequence and

structural similarity to other SLC44 family members, it wasthought to play a role in choline transport. In functional experi-ments, not shown here, our results did not support a role forSLC44A4 in choline transport, nor was it involved in the transportof a number of other substrates we tested. Thiamine (vitamin B1)serves an essential role as a co-factor of enzymes involved incellular metabolic pathways (13–14); however, it cannot besynthesized de novo in mammalian cells and must be obtainedthrough dietary and microbiota-generated sources. A recentlypublished report demonstrated that SLC44A4 is involved intransport of the phosphorylated form of thiamine (thiaminepyrophosphate) produced by microbiota in the intestine (9).Such a functional role would be consistent with the expressionwe observed within normal tissues of the gastrointestinal tract,although thiamine pyrophosphate was not one of the substrateswe tested for uptake.

Additional studies performed in our lab did not identify afunctional role for SLC44A4 in tumorigenesis, and there are noreports linking SLC44A4 to cancer. Furthermore, treatment withAGS-5M2 or any of our other SLC44A4-specific mAbs did nothave antitumor effects. Due to this, we focused on the suitabilityof SLC44A4 as an ADC target.

We describe here for the first time an extensive analysis ofexpression of this novel target across normal and cancer speci-mens and identify cancer indications with overexpression ofSLC44A4. IHC analysis of the expression of SLC44A4 in normalgastrointestinal tract revealed that the expression in epithelial cellsis largely confined to the apical, or luminal, side of secretoryepithelial cells. This apical polarity of expression is lost inadvanced or poorly differentiated tumors. This may favor target-ing tumor cells with an ADC and sparing normal cells that expressSLC44A4. This is because tight junctions that form the epithelialbarrier between systemic circulation and luminal spaces greatlyreduce diffusion of large macromolecules into the lumen (15).This provides a rationale for lower exposure of apical surfaces ofepithelial cells to circulating IgGs. However, several mechanismsfor specifically transporting immunoglobulins across the epithe-lial barrier are known. The polymeric immunoglobulin receptor(pIgR) acts as a transporter of IgA and IgM molecules acrossepithelial barriers, but does not bindmonomeric IgGs (16). FcRn,on the other hand, can transcytose IgGs across the epithelialbarrier but the serum concentration of IgG1 has been reportedto be 500- to 1,000-fold higher than luminal concentrations (17).This analysis suggests that the exposure of tumor cells to injectedIgG is likely to be greater than the exposure at the apical side ofnormal epithelial cells, thus a therapeutic with anti-SLC44A4targeting antibody conjugated to a non-selective cytotoxic mol-ecule could be very tumor cell specific. Toxicology will be neededto validate this hypothesis.

In the process of identifying suitable in vitro and in vivo tumormodels, we observed that SLC44A4 is expressed at very low levelsin most tumor cell lines when grown in vitro. However, inOVCAR5 cells and additional cell lines implanted and grown astumor xenografts in vivo, the expression of SLC44A4 increasedconsiderably, suggesting that there might be factors or contribu-tions from the tumor microenvironment that upregulated theexpression of SLC44A4.

While the RNA levels observed in normal prostate and prostatetumors did not indicate differential expression, many patientsdiagnosed with prostate cancer undergo radical prostatectomyduring treatment, which removes concern for on-target toxicity for

Table 3. Summary of selected in vivo efficacy studies with ASG-5ME

Model name Cancer type ASG-5ME dosing TGI (%)

LAPC-9AI (orthotopic) Prostate 5 mg/kg q4d�4 95.7LAPC-9AI Prostate 5 mg/kg q4d�4 93.3LAPC-9AD Prostate 5 mg/kg q4d�4 83.4LAPC-4AD Prostate 5 mg/kg q4d�4 73PC3-SLC44A4 Prostate 3 mg/kg single dose 99.2VCaP Prostate 5 mg/kg q4d�4 99.122Rv1 (orthotopic) Prostate 5 mg/kg q4d�4 24.9–47.322Rv1 Prostate 5 mg/kg q4d�4 46.4CWR-22R Prostate 6 mg/kg q4d�3 29.8MDA-PCa-2b Prostate 5 mg/kg q4d�4 85.6AG-Panc4 Pancreatic 5 mg/kg q4d�4 87.3–95.4AG-Panc3 (orthotopic) Pancreatic 5 mg/kg q4d�4 69.8AG-Panc3 Pancreatic 5 mg/kg q4d�4 42AG-Panc2 Pancreatic 5 mg/kg q4d�4 80AG-C4 Colon 3 mg/kg q4d�4 77.4HT-29 Colon 3 mg/kg q4d�4 90.5–92.1OVCAR-5 Ovarian 3 mg/kg q4d�4 45.6

OVCAR-5 Ovarian 5 mg/kg q4d�4 72.3

SLC44A4, an ADC Target for Pancreatic and Prostate Cancer

www.aacrjournals.org Mol Cancer Ther; 15(11) November 2016 2685

on January 11, 2021. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 22, 2016; DOI: 10.1158/1535-7163.MCT-16-0225

Page 8: The Discovery and Preclinical Development of ASG-5ME ... · Hector Avina~ 1, Claudia Guevara1, Faisal Malik1, Ruth Moser2, Steven Duniho2, Jeffrey Coleman1,Ying Li1, Daniel S. Pereira1,

this organ. Based on the high prevalence of SLC44A4 expressionobserved in pancreatic and prostate cancer tissue microarrays, weevaluated the activity of SLC44A4-specific ADCs in in vitro and invivomodels of these indications. From thesewe selected ASG-5MEas our lead candidate and we report here for the first time thedevelopment of this molecule for the treatment of pancreaticcancer, prostate cancer, and other SLC44A4-positive tumors. Wehave shown that this ADC has high affinity for SLC44A4 and thatit is cytotoxic and able to inhibit in vivo growth of several tumormodels, including patient-derived pancreatic and prostate tumorxenografts.

A number of antibody-based therapeutics are being evaluat-ed in the clinic but none have been approved for use in prostateor pancreatic cancer. Although newer therapies have beenapproved for advanced pancreatic cancer (Abraxane, nab-pac-litaxel) and prostate cancer (Zytiga and Xtandi), resistance tothese treatments means that additional therapeutic options areneeded. We evaluated whether ASG-5ME can enhance antitu-

mor activity when combined with standard-of-care agents forpancreatic and prostate cancers in xenograft models. Our dataindicated that a combination of ASG-5ME and nab-paclitaxel atsub-optimal doses showed a marked increase in antitumoractivity compared with the single agents alone. Although tax-ane-based chemotherapy has activity in advanced prostatecancer, Abraxane has not been approved for use in prostatecancer and has not been extensively evaluated in that setting.Our combination studies provide an initial proof of conceptfor combining ASG-5ME with Abraxane in pancreatic andprostate cancers. Of further interest are the enhanced antitumoreffects with a combination of two anti-tubulin agents. Furtherpreclinical efficacy and safety evaluation would be necessary tojustify clinical evaluation of this combination approach. Takentogether, our preclinical data generated with ASG-5ME high-lights its therapeutic potential in pancreatic and prostate cancerand supports its clinical development as an anti-SLC44A4therapeutic.

1,200.0

1,000.0

800.0

600.0

400.0

200.0

0.0

1,400

1,200

1,000

800

600

400

200

0

0 5 10 15 20 25 30 0 5 10 15 20 25 30 35 40 45 50 55 0 10 20 30 40 50 60 70

1,200

1,000

800

600

400

200

0

Time (day)

Time (day) Time (day)

Time (day) Time (day)

LAPC-9AI

AG-Panc4

VCaP AG-Panc4

Control ADC 3 mg/kgControl ADCASG-5ME 1 mg/kg

ASG-5ME 3 mg/kg ASG-5ME

Control ADCASG-5ME

Mea

n tu

mor

vol

ume

(mm

3 )M

ean

tum

or v

olum

e (m

m3 )

Mea

n tu

mor

vol

ume

(mm

3 )

Mea

n tu

mor

vol

ume

(mm

3 )

Mea

n tu

mor

vol

ume

(mm

3 )

LAPC-9AIVehicle control

Vehicle controlASG-5ME 1 mg/kgASG-5ME 0.5 mg/kg

ASG-5ME (0.5) + nab-paclitaxel (60)ASG-5ME (0.5) + nab-paclitaxel (120)

Nab-paclitaxel 100 mg/kgNab-paclitaxel 60 mg/kgNab-paclitaxel 120 mg/kg

ASG-5ME + nab-paclitaxel

1,6001,5001,4001,3001,2001,1001,000

900800700600500400300200100

0 3 6 9 12 15 18 21 24 27 30 33 0 2 4 6 8 10 12 14

1,200.01,100.01,000.0

900.0800.0700.0600.0500.0400.0300.0200.0100.0

0.0

A

D E

B C

Figure 6.

In vivo efficacy of ASG-5ME alone or in combination with nab-paclitaxel. Tumor fragments were implanted or cells were injected s.c. into the flank of ICR-SCIDmice, and animals were randomized into treatment groups (n ¼ 10) when tumors reached a size of approximately 150 mm3. IHC staining of SLC44A4expression is shown in A–C. A, in vivo efficacy of ASG-5ME in an androgen-independent patient-derived prostate tumor xenograft model LAPC-9AI. Treatmentwas initiated i.v. with 1 or 3 mg/kg of ASG-5ME or a control ADC (3 mg/kg) administered on days 0, 4, 8, and 12 for a total of four doses. B, in vivo efficacyof ASG-5ME in the VCaP prostate cell line xenograft model. Treatment was initiated i.v. on day 0 with 5 mg/kg of either ASG-5ME or a non-binding controlADC administered every 4 days for a total of 3 doses. C, in vivo efficacy of ASG-5ME in a patient-derived pancreatic tumor xenograft model (AG-Panc4).Treatment was initiated i.v. on day 0 with 4 mg/kg of ASG-5ME or a control ADC administered on days 0, 4, 8, and 12 for a total of four doses. D, ASG-5ME,nab-paclitaxel, or combination of both on tumor growth inhibition in the AG-Panc4 model. Treatment was initiated i.v. on day 0 with 1 mg/kg of ASG-5ME,nab-paclitaxel at 100 mg/kg, or combinations of ASG-5ME at 1 mg/kg with 100 mg/kg nab-paclitaxel administered every 4 days for a total of 3 doses. E, ASG-5ME,nab-paclitaxel, or combination of both on tumor growth inhibition in the LAPC-9AI model. Treatment was initiated i.v. with 0.5 mg/kg of ASG-5ME, nab-paclitaxelat 60 mg/kg, nab-paclitaxel at 120 mg/kg, and combinations of 0.5 mg/kg ASG-5ME with nab-paclitaxel at either 60 mg/kg or 120 mg/kg administeredevery 4 days for a total of 3 doses.

Mattie et al.

Mol Cancer Ther; 15(11) November 2016 Molecular Cancer Therapeutics2686

on January 11, 2021. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 22, 2016; DOI: 10.1158/1535-7163.MCT-16-0225

Page 9: The Discovery and Preclinical Development of ASG-5ME ... · Hector Avina~ 1, Claudia Guevara1, Faisal Malik1, Ruth Moser2, Steven Duniho2, Jeffrey Coleman1,Ying Li1, Daniel S. Pereira1,

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: M. Mattie, A. Raitano, K. Morrison, Z. An, F. Do~nate,I.B.J. Joseph, P. Challita-Eid, D. Benjamin, D.R. StoverDevelopment of methodology: M. Mattie, A. Raitano, K. Morrison, K. Morri-son, Z. An, C. Guevara, Y. Li, P. Challita-EidAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): M. Mattie, A. Raitano, K. Morrison, L. Capo,A. Verlinsky, M. Leavitt, J. Ou, R. Nadell, H. Avi~na, C. Guevara, F. Malik,R. Moser, S. Duniho, J. Coleman, Y. Li, F. Do~nateAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis):M. Mattie, A. Raitano, K. Morrison, Z. An, M. Leavitt,J. Ou, H. Avi~na, C. Guevara, Y. Li, F. Do~nate, I.B.J. Joseph, P. Challita-Eid,D. Benjamin, D.R. StoverWriting, review, and/or revision of the manuscript: M. Mattie, A. Raitano,K. Morrison, K. Morrison, Z. An, J. Ou, R. Nadell, Y. Li, D.S. Pereira, F. Do~nate,I.B.J. Joseph, P. Challita-Eid, D.R. Stover

Administrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): M. Mattie, Y. Li, D.R. StoverStudy supervision: M. Mattie, A. Raitano, Z. An, I.B.J. Joseph, P. Challita-EidOther (cloned hybridoma heavy and light sequences into CHO cell lineproduction plasmids, and then confirmed those constructs): F. Malik

AcknowledgmentsThe authors thank Jean Gudas, Aya Jakobovits, and the hardworking and

thoughtfulmembers of theAgensys researchdepartments that didnot appear onthis article, but that significantly contributed to previous conference presenta-tions or significantly contributed to historical data that ultimately led to thisfinal article. ASG-5ME was co-developed in conjunction with Agensys' partner,Seattle Genetics, Inc.

The costs of publication of this article were defrayed in part by the paymentof page charges. This article must therefore be hereby marked advertisementin accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received April 12, 2016; revised July 19, 2016; accepted August 7, 2016;published OnlineFirst August 22, 2016.

References1. Inazu M, Takeda H, Matsumiya T. Molecular and functional characteriza-

tion of an Naþ-independent choline transporter in rat astrocytes.J Neurochem 2005;94:1427–37.

2. Traiffort E, Ruat M, O'Regan S, Meunier FM. Molecular characterization ofthe family of choline transporter-like proteins and their splice variants.J Neurochem 2005;92:1116–25.

3. Uchida Y, Inazu M, Takeda H, Yamada T, Tajima H, Matsumiya T.Expression and functional characterization of choline transporter inhuman keratinocytes. J Pharmacol Sci 2009;109:102–9.

4. Kouji H, Inazu M, Yamada T, Tajima H, Aoki T, Matsumiya T. Molecularand functional characterization of choline transporter in human coloncarcinoma HT-29 cells. Arch Biochem Biophys 2009;483:90–8.

5. Yamada T, Inazu M, Tajima H, Matsumiya T. Functional expression ofcholine transporter-like protein 1 (CTL1) in human neuroblastoma cellsand its link to acetylcholine synthesis. Neurochem Int 2010;58:354–65.

6. Nakamura T, Fujiwara R, Ishiguro N, Oyabu M, Nakanishi T, Shirasaka Y,et al. Involvement of choline transporter-like proteins, CTL1 and CTL2, inglucocorticoid-induced acceleration of phosphatidylcholine synthesis viaincreased choline uptake. Biol Pharm Bull 2010;33:691–6.

7. Kommareddi PK, Nair TS, Thang LV, Galano MM, Babu E, Ganapathy V,et al. Isoforms, expression, glycosylation, and tissue distribution of CTL2/SLC44A2. Protein J 2010;29:417–26.

8. Song P, Rekow SS, Singleton CA, Sekhon HS, Dissen GA, Zhou M, et al.Choline transporter-like protein 4 (CTL4) links to non-neuronal acetyl-choline synthesis. J Neurochem 2013;126:451–61.

9. Nabokina SM, Inoue K, Subramanian VS, Valle JE, Yuasa H, Said HM.Molecular identification and functional characterization of the human

colonic thiamine pyrophosphate transporter. J Biol Chem 2014;289:4405–16.

10. Muller AJ, Young JC, Pendergast AM, Pondel M, Landau NR, Littman DR,et al. BCR first exon sequences specifically activate the BCR/ABL tyro-sine kinase oncogene of Philadelphia chromosome-positive leukemias.Mol Cell Biol 1991;4:1785–92.

11. Green L.Antibody engineering via genetic engineering of the mouse:XenoMouse strains are vehicles for the facile generation of therapeutichuman monoclonal antibodies. J Immunol Methods 1999;231:11–23.

12. Doronina SO, Toki BE, Torgov MY, Mendelsohn BA, Cerveny CG, ChaceDF, et al. Development of potent monoclonal antibody auristatin con-jugates for cancer therapy. Nat Biotechnol 2003;21:778–84.

13. Berdanier CD. Advanced nutrition micronutrients. Boca Raton, FL: CRCPress; 1998.

14. Bettendorff L, Wins P. Thiamin diphosphate in biological chemistry. Newaspects of thiamin metabolism, especially triphosphate derivatives actingother than as cofactors. FEBS J 2009;276:2917–25.

15. Gumbiner B. The structure, biochemistry, and assembly of epithelial TJs.Am J Physiol 1987;253:C749–C758.

16. Brandtzaeg P. Mucosal and glandular distribution of immunoglobulincomponents: differential localization of free and bound SC in secretoryepithelial cells. J Immunol 1974;112:1553–9.

17. Yoshida M, Claypool SM, Wagner JS, Mizoguchi E, Mizoguchi A, Roope-nian DC, et al. Human neonatal Fc receptor mediates transport of IgG intoluminal secretions for delivery of antigens to mucosal dendritic cells.Immunity 2004;20:769–83.

www.aacrjournals.org Mol Cancer Ther; 15(11) November 2016 2687

SLC44A4, an ADC Target for Pancreatic and Prostate Cancer

on January 11, 2021. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 22, 2016; DOI: 10.1158/1535-7163.MCT-16-0225

Page 10: The Discovery and Preclinical Development of ASG-5ME ... · Hector Avina~ 1, Claudia Guevara1, Faisal Malik1, Ruth Moser2, Steven Duniho2, Jeffrey Coleman1,Ying Li1, Daniel S. Pereira1,

2016;15:2679-2687. Published OnlineFirst August 22, 2016.Mol Cancer Ther   Michael Mattie, Arthur Raitano, Kendall Morrison, et al.   Tumors Including Pancreatic and Prostate Cancer

Drug Conjugate Targeting SLC44A4-Positive Epithelial−Antibody The Discovery and Preclinical Development of ASG-5ME, an

  Updated version

  10.1158/1535-7163.MCT-16-0225doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2016/08/20/1535-7163.MCT-16-0225.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/15/11/2679.full#ref-list-1

This article cites 16 articles, 2 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/15/11/2679.full#related-urls

This article has been cited by 5 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://mct.aacrjournals.org/content/15/11/2679To request permission to re-use all or part of this article, use this link

on January 11, 2021. © 2016 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst August 22, 2016; DOI: 10.1158/1535-7163.MCT-16-0225