a novel therapeutic strategy for pancreatic cancer ... · approved in the clinic (8). thus, novel...

14
Large Molecule Therapeutics A Novel Therapeutic Strategy for Pancreatic Cancer: Targeting Cell Surface Glycan Using rBC2LC-N LectinDrug Conjugate (LDC) Osamu Shimomura 1 , Tatsuya Oda 1 , Hiroaki Tateno 2 , Yusuke Ozawa 1 , Sota Kimura 1 , Shingo Sakashita 3 , Masayuki Noguchi 3 , Jun Hirabayashi 2 , Makoto Asashima 2 , and Nobuhiro Ohkohchi 1 Abstract Various cancers, including pancreatic ductal adenocarcinoma (PDAC), remain intractable even with costly tumor-targeting antibody drugs. Because the outermost coatings of cancer cells are composed of cell-specic glycan layers (glycocalyx), lectins, proteins with glycan-binding potential, were evaluated for pos- sible use as drug carriers in PDAC treatment. A human PDAC cell line with well-to-moderately differentiated properties (Capan-1) was subjected to lectin microarray analysis to identify specic lectinglycan pairs. The selected lectin was fused with a bacterial exotoxin for the construction of a lectindrug conjugate (LDC), and its safety and antitumor effects were evaluated. A specic afnity between a recombinant bacterial C-type lectin (rBC2LC- N) and Capan-1 was identied, and its positivity was conrmed in 69 human samples. In contrast to the belief that all lectins mediate harmful hemagglutination, rBC2LC-N did not cause hemagglu- tination with human erythrocytes and was safely administered to mice. The 50% inhibitory concentration of LDC to Capan-1 (1.04 pg/mL ¼ 0.0195 pmol/L) was 1/1,000 lower than that reported for conventional immunotoxins. The intraperitoneal administra- tion of LDC reduced the tumor weight from 390 to 130.8 mg (P < 0.01) in an orthotopic model and reduced the number of nodules from 48 to 3 (P < 0.001) and improved survival from 62 to 105 days in a peritoneal dissemination model (P < 0.0001). In addition, the effect of LDC was reproduced in nodules from patient-derived PDAC xenografts through intravenous injection. Herein, we show the concept of utilizing lectins as drug carriers to target glycans on the cancer cell surface, highlighting new insights into cancer treatments. Mol Cancer Ther; 17(1); 18395. Ó2017 AACR. Introduction Cancer-targeting antibody drugs, including antibody-drug con- jugates (ADC; refs. 1, 2), are attractive therapies, but their clinical success rates have been limited (3). The targets of these drugs are usually peptides of cell transmembrane proteins; however, it is important to note that cell surfaces are covered by a glycan layer that is referred to as the glycocalyx (4). The glycomes of cancer cell surfaces are often unique, with aberrant glycosyl- ation, including sialylation, fucosylation, O-glycan truncation, and N- and O-linked glycan branching (57). Therefore, targeting outer-layer glycans might be a more effective cancer-targeting strategy than targeting the underlying core proteins. While car- bohydrate-binding monoclonal antibodies have now been inves- tigated for tumor targeting for several decades, none have yet been approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based on the principal of antigenantibody afnities should shed light on intractable cancers such as pancreatic ductal adenocarcinoma (PDAC; ref. 9). Lectins, proteins that recognize glycans and thus have glycan- binding potential, are an alternative to antibodies. However, exogenous lectins, such as those collected from plants, fungi, and snake venom, often have erythrocytic agglutination activity, which has discouraged their application in vivo (10). In contrast to the general concept, the extent of lectin hemagglutination activity is quite variable (10). More than 100 putative endogenous lectins, such as selectin and galectin (11), have been identied in humans (12) and contribute to various physiological processes (10). In this report, we tested whether lectins could be a drug carrier for cancer therapy that is applicable in vivo. We rst identied a lectin that possesses specic afnity to the glycans of cancer cell surfaces using a rare and valuable PDAC cell line that has a well-to- moderately differentiated morphology. The selected lectin was then fused to a bacterial toxin to construct a lectindrug conjugate (LDC), and its prominent cytocidal effect was demonstrated using an in vitro assay. The therapeutic effects of LDC on PDAC were tested in vivo using a cell line-based xenograft (cell-xeno) model and a patient-derived xenograft (PDX) model. Materials and Methods Cell lines A total of six human pancreatic cancer cell lines with various cell differentiation states in their origin were used. AsPC-1 (CRL-1682, 1 Department of Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan. 2 Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan. 3 Department of Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Author: Tatsuya Oda, Department of Surgery, Faculty of Med- icine, Clinical Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8575, Japan. Phone: 81-298-53-3221; Fax: þ81-298-53-3222; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-17-0232 Ó2017 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 183 on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Upload: others

Post on 04-Aug-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

Large Molecule Therapeutics

A Novel Therapeutic Strategy for PancreaticCancer: Targeting Cell Surface Glycan UsingrBC2LC-N Lectin–Drug Conjugate (LDC)Osamu Shimomura1, Tatsuya Oda1, Hiroaki Tateno2, Yusuke Ozawa1, Sota Kimura1,Shingo Sakashita3, Masayuki Noguchi3, Jun Hirabayashi2, Makoto Asashima2, andNobuhiro Ohkohchi1

Abstract

Various cancers, including pancreatic ductal adenocarcinoma(PDAC), remain intractable even with costly tumor-targetingantibody drugs. Because the outermost coatings of cancer cellsare composed of cell-specific glycan layers (glycocalyx), lectins,proteins with glycan-binding potential, were evaluated for pos-sible use as drug carriers in PDAC treatment. A human PDAC cellline with well-to-moderately differentiated properties (Capan-1)was subjected to lectin microarray analysis to identify specificlectin–glycan pairs. The selected lectin was fused with a bacterialexotoxin for the construction of a lectin–drug conjugate (LDC),and its safety and antitumor effects were evaluated. A specificaffinity between a recombinant bacterial C-type lectin (rBC2LC-N) andCapan-1was identified, and its positivitywas confirmed in69human samples. In contrast to thebelief that all lectinsmediate

harmful hemagglutination, rBC2LC-N did not cause hemagglu-tination with human erythrocytes and was safely administered tomice. The 50% inhibitory concentration of LDC to Capan-1 (1.04pg/mL ¼ 0.0195 pmol/L) was 1/1,000 lower than that reportedfor conventional immunotoxins. The intraperitoneal administra-tion of LDC reduced the tumor weight from 390 to 130.8mg (P <0.01) in an orthotopicmodel and reduced the number of nodulesfrom 48 to 3 (P < 0.001) and improved survival from 62 to 105days in a peritoneal dissemination model (P < 0.0001). Inaddition, the effect of LDC was reproduced in nodules frompatient-derived PDAC xenografts through intravenous injection.Herein, we show the concept of utilizing lectins as drug carriers totarget glycans on the cancer cell surface, highlighting new insightsinto cancer treatments. Mol Cancer Ther; 17(1); 183–95. �2017 AACR.

IntroductionCancer-targeting antibody drugs, including antibody-drug con-

jugates (ADC; refs. 1, 2), are attractive therapies, but their clinicalsuccess rates have been limited (3). The targets of these drugs areusually peptides of cell transmembrane proteins; however, itis important to note that cell surfaces are covered by a glycanlayer that is referred to as the glycocalyx (4). The glycomesof cancer cell surfaces are often unique, with aberrant glycosyl-ation, including sialylation, fucosylation, O-glycan truncation,andN- andO-linked glycan branching (5–7). Therefore, targetingouter-layer glycans might be a more effective cancer-targetingstrategy than targeting the underlying core proteins. While car-bohydrate-binding monoclonal antibodies have now been inves-tigated for tumor targeting for several decades, none have yet been

approved in the clinic (8). Thus, novel powerful therapeuticmethods targeting cancer cell surface glycans that are not basedon the principal of antigen–antibody affinities should shed lighton intractable cancers such as pancreatic ductal adenocarcinoma(PDAC; ref. 9).

Lectins, proteins that recognize glycans and thus have glycan-binding potential, are an alternative to antibodies. However,exogenous lectins, such as those collected from plants, fungi, andsnake venom, often have erythrocytic agglutination activity, whichhas discouraged their application in vivo (10). In contrast to thegeneral concept, the extent of lectin hemagglutination activity isquite variable (10). More than 100 putative endogenous lectins,such as selectin and galectin (11), have been identified in humans(12) and contribute to various physiological processes (10).

In this report, we tested whether lectins could be a drug carrierfor cancer therapy that is applicable in vivo. We first identified alectin that possesses specific affinity to the glycans of cancer cellsurfaces using a rare and valuable PDACcell line that has awell-to-moderately differentiated morphology. The selected lectin wasthen fused to a bacterial toxin to construct a lectin–drug conjugate(LDC), and its prominent cytocidal effect was demonstrated usingan in vitro assay. The therapeutic effects of LDC on PDAC weretested in vivo using a cell line-based xenograft (cell-xeno) modeland a patient-derived xenograft (PDX) model.

Materials and MethodsCell lines

A total of six humanpancreatic cancer cell lineswith various celldifferentiation states in their originwere used. AsPC-1 (CRL-1682,

1Department of Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba,Ibaraki, Japan. 2Biotechnology Research Institute for Drug Discovery, NationalInstitute of Advanced Industrial Science and Technology (AIST), Tsukuba,Ibaraki, Japan. 3Department of Pathology, Faculty of Medicine, University ofTsukuba, Tsukuba, Ibaraki, Japan.

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

Corresponding Author: Tatsuya Oda, Department of Surgery, Faculty of Med-icine, Clinical Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki305-8575, Japan. Phone: 81-298-53-3221; Fax: þ81-298-53-3222; E-mail:[email protected]

doi: 10.1158/1535-7163.MCT-17-0232

�2017 American Association for Cancer Research.

MolecularCancerTherapeutics

www.aacrjournals.org 183

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 2: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

September 19, 2013), BxPC-3 (CRL-1687, Aug. 4, 2015), Capan-1(HTB-79, July 26, 2013), MIAPaCa-2 (CRL-1420, September 5,2013), and PANC-1 (CRL-1469, March 4, 2015) were purchasedfrom ATCC, and SUIT-2 (JCRB1094, July 28, 2015) was obtainedfrom the National Institute of Biomedical Innovation. Themorphologies of their clinical PDAC origin were as follows: welldifferentiated for Capan-1; and those of the other five lines werepoorly or poorly moderately differentiated (13). Capan-1, MIA-PaCa-2, and AsPC-1 cells were verified by STR-PCR inMarch 2016(report number: KBN0366, National Institute of BiomedicalInnovation). The remaining cell lines were used within 6 monthsof purchase and were tested for contamination with mycoplasma.

Patient samples and tissue collectionFreshhumanpancreatic cancer tissuewas obtainedwithpatient

consent as approved by the Research Ethics Board of the Univer-sity of Tsukuba. A total of 69 resected pancreatic adenocarcinomaspecimens (7 poorly differentiated (P/D) cases, 53 moderatelydifferentiated (M/D) cases, and 9 well-differentiated cases) wereused in this study. For the use of these clinical samples for researchpurposes, written informed consent was obtained from allpatients, and approval was obtained from the Tsukuba ClinicalResearch&DevelopmentOrganization (T-CReDOprotocol num-ber: H28-90).

Lectin microarrayThe high-density lectin microarray was prepared as previously

described (14). Briefly, proteins from each cell line were preparedfrom whole-cell lysates of six different wells and fluorescentlylabeled with the monoreactive dye Cy3 (GE Healthcare). Then,0.5 mg/mL Cy3-labelled lysate was added to each well of amicroarray plate, and the plate was incubated at 20�C overnight.Fluorescence imageswere then acquiredusing an evanescentfield-activated fluorescence scanner (GlycoStation Reader; GlycoTech-nica Ltd.). The fluorescence signal of each spot was quantifiedusing Array-Pro Analyzer version 4.5 (Media Cybernetics), mean-normalized, log-transformed, and analyzed via the average link-age method using Cluster 3.0 (yellow: high; black: intermediate;blue: low).

Lectin stainingLectin histochemistry. Antigen retrieval in 2-mm slide sections offormalin-fixed and paraffin-embedded (FFPE) tissues was per-formed by autoclaving, endogenous peroxidase activity wasblocked with 3% H2O2 with methanol, and horseradish perox-idase-labeled rBC2 was applied and visualized by applying thechromogen diaminobenzidine. The clinical cases were judged as0, 1þ, 2þ, 3þ with regard to rBC2 reactivity, and the correlationwith cell differentiation was assessed.

Live-cell staining by rBC2 lectin. Live cells were incubated for12 hours in medium containing FITC-conjugated rBC2LC-N(1 mg/mL); images were captured using a BIOREVO BZX-710fluorescence microscope (KEYENCE).

Production of rBC2-PE38The recombinant N-terminal domain of BC2L-C (rBC2LC-N;

156 amino acids) was fused to a truncated form of the catalyticdomain of pseudomonas exotoxin (PE) A (399–613 residues; 215amino acids) carrying a C-terminal 6�-His tag (HHHHHH) and aKDEL sequence via a ten-amino-acid linker (GSG3)2 (Fig. 2A).

The generated rBC2-PE38 (371 amino acids) was expressedin E. coli and purified via one-step affinity chromatography onan L-fucose-Sepharose column. The yieldwas 10mg/L of bacterialculture. rBC2-PE38 was detected as a major band at the predictedmolecular weight of 54 kDa by SDS-PAGE in the presence andabsence of 2-mercaptoethanol (Supplementary Fig. S1).

Cell assaysLectin binding analysis. Cells were harvested and stained withvarious concentrations of rBC2-FITC for 30 minutes on ice. TheMFI of the cells was determined using an LSR Fortessa X-20 flowcytometer. The data were analyzed using a nonlinear regressionfitting program in GraphPad Prism 6 (GraphPad Software Inc.).

MTT assay. The cytotoxicity of rBC2-PE38 was measured in a cellviability assay using WST-8 (Dojindo Molecular Technologies) asrecommended by themanufacturer. Cells were seeded on 96-wellplates at 5� 103 cells/well. After incubation for 24 hours, rBC2 orrBC2-PE38 was applied to the medium at the indicated concen-trations (0.1–10 mg/mL). After further incubation for 48 hours,the culture medium was replaced with the appropriate freshmedium, and the cells were incubated for an additional 24 hours.We used 48 þ 24 hours and not a continuous 72 hours becausemany cells died after 48hours due to the action of LDC,whichwasinternalized into cells during the first 48 hours. Cell viability wasthen assessed via an MTT assay (WST-8) according to the man-ufacturer's instructions. After solubilization of the purple forma-zan crystals, absorbance was measured at 450 nm (backgroundwavelength, 650 nm) using a plate spectrophotometer. The IC50

was calculated using GraphPad Prism 6.

Hemagglutination assayBlood samples were obtained from four healthy volunteers of

each blood type, i.e., A, B, O, and AB. A 5-mL aliquot of freshlywhole blood was washed 3 times with 20 to 25 mL of PBS(centrifugation at 500 �g for 5 minutes, followed by removal ofthe plasma and white cell ghost layer at the top of the pellet). Thesuspension of untreated (without using trypsin) erythrocytes wasthenmixedwith an equal volume of incubation buffer (0.1mol/Lacetate buffer containing 1 mmol/L CaCl2, pH 5.5) containing1 unit/mL neuraminidase (from C. welchii Sigma N2876)and incubated at 37�C for 1 hours with occasional shaking. Allsamples were againwashed 3 timeswith PBS and then prepared asa 2% (v/v) erythrocyte suspension in PBS. ConcanavalinAor rBC2lectin solution (50 mg/mL) was applied to the left well of a U-shaped 96-well plate, and a series of 2-fold dilutionswas preparedin a horizontal line. The same volume of erythrocyte suspensionwas applied to each well, and agglutination was observed.

Toxicology study of rBC2-PE38 (LDC) in wild-type miceTo evaluate the LD50 (50% lethal dose) of LDC, female wild-

type mice (5–6 weeks old, 18–20 g, Charles River LaboratoriesInternational, Japan) were given an intraperitoneal (i.p.) or intra-venous (i.v.) injectionwith a single dose of 1.0 to 15.0 mg of rBC2/mouse or rBC2-PE38/mouse (50 to 750 mg/kg) in 300 mL of PBS.The mice were observed for survival until day 14.

Animal models of PDACAnimal experiments were conducted in compliance with the

ethical regulations approved by the Animal Care Committee,

Shimomura et al.

Mol Cancer Ther; 17(1) January 2018 Molecular Cancer Therapeutics184

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 3: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

University Health Network, University of Tsukuba, Tsukuba,Ibaraki, Japan.

Subcutaneous cell xenograft model. A total of 3 � 106 Capan-1 orSUIT-2 cells were subcutaneously injected into female nude mice(BALB/c nu/nu, 6–8 weeks old, CLEA Japan).

Subcutaneous PDX model. Fresh human pancreatic cancer tissueoriginated from a nodule of liver metastasis of clinical PDAC thatshowed moderately-to-poorly differentiated morphology wasimplanted into subcutaneous pockets on the back skin ofCB17/Icr-scid/SCIDmice (female, 6 to 8 weeks old, CLEA Japan).After the establishment of first-generation subcutaneous nodules,the tumor nodules were minced into 2-mm cubic fragments, and3 pieces were implanted into subcutaneous pockets on the backskin of SCID mice. These PDX nodules show moderately differ-entiation in morphology.

Pancreatic orthotopic model. Nude mice were anesthetized withisoflurane, a laparotomy was created using a left lateral abdom-inal incision, and thepancreaswas exteriorized. Four tumorpieces(approximately 2 mm in diameter) obtained from peritonealdisseminated Capan-1 nodules were transplanted to the body ofthe pancreas and secured with absorbable surgical sutures.

Peritoneal dissemination model. Nude mice were inoculated withCapan-1 or SUIT-2 cells by i.p. injection of 2.0 � 106 cellssuspended in 100 mL of PBS. On day 14, 2 mice were sacrificedto confirm the establishment of disseminated cancer nodules. Theremaining mice were then randomly allocated to four groups.

rBC2-PE38 (LDC) treatment for in vivo PDAC mouse modelsDirect injection (d.i.) to subcutaneous cell xenograft models and aPDXmodel.A d.i. of 40 ng, 1 mg, or 5 mg of LDC in 100 mL of PBSwas performed a total of 4 times near the subcutaneous tumorson days 14, 18, 22, and 26. For the control, 100 mL of normalsaline was injected. The tumor volume was determined everyday for 2 weeks using the following formula: (width)2 �(length)/2. On day 34, the subcutaneous tumors were excisedfor weight measurements.

i.p. and i.v. injection of LDC to subcutaneous PDXmodels.A total of1 mg LDCwas administered in 100 mL of PBS via i.p. injection (n¼4 each) or i.v. injection via the tail vein for a total of 8 times ondays 14, 17, 19, 22, 24, 26, 29, and 31. On day 36, the subcu-taneous nodules were excised for weight measurements.

i.p. and i.v. injection of LDC to orthotopic models. The mice weretreated and evaluated on a treatment schedule identical to that ofthe cell xenograft models, using 1 or 5 mg of LDC (n¼ 5 each). Ondays 21, 24, 28, and 31, 1 mg of LDC was administered i.p. or i.v.via the tail vein. On day 45, the mice were sacrificed for theobservation of abdominal status and tumorweightmeasurement.

i.p. and i.v. injection to peritoneal dissemination models. TheCapan-1 peritoneal dissemination mouse models were treatedby injection of 1 mg of rBC2 lectin without PE38, 40 ng of LDC, or1mg of LDC in 100mLof PBSby i.p. or i.v. injection via the tail veina total of 4 times on days 14, 18, 22, and 26 (n¼ 4 each). On day30, the mice were sacrificed for manual counting of the dissem-

inated nodules. The Kaplan–Meier survival curves were calculatedusing 36 nude mice. At 14 days after the Capan-1 cell injection, 4groups of mice (n ¼ 9 each) were treated on days 1, 5, 9, and 13with control, 40 ng of LDC via i.p. injection, 1 mg of LDC via i.p.injection, or 1 mg of LDC via i.v. injection.

Statistical analysesA heat map of the clustering analysis results was constructed

using Java TreeView. Differences in the lectin signal between thetwo arbitrary data sets were evaluated using Student t test in SPSSStatistics 21.0 (SPSS Inc.). Significantly different lectin signals orglycosyltransferase expression levels were selected if they satisfieda familywise error rate (FWER) of <0.001 according to the Bon-ferronimethod. Applied statistics included the unpaired Student ttest and the Mann–Whitney U test. P < 0.05 was considered toindicate significance. Kaplan–Meier curves (statistically analyzedby the log-rank test) and LDC IC50 values were calculated usingGraphPad Prism 6; other analyses were performed using SPSS21.0 (SPSS Inc.).

ResultsSelection of PDAC cell lines that display clinical PDAC andcancer stem-cell-like characteristics

For cancer cell glycome analysis, clinical PDAC samples arelimited in terms of sample availability and quality due to thetypical histological characteristics of PDAC, which include thepresence of epithelial cancer cells forming ductal glands (gþþ)surrounded by abundant stromal components (sþþ; Fig. 1A).Wetherefore began the experiments by selecting a cell line thatrepresents the dominant morphological properties of clinicalPDAC when grown in vivo. Only Capan-1 cells strongly exhibitedorganized gland formation (gþþ) and dense stromal prolifera-tion (sþþ; Fig. 1B). The expressionof the knownpancreatic cancerstem cell markers CD24, CD44, EpCAM, and CD133 (15, 16), asrevealed by flow cytometry analysis, indicated that the Capan-1line was positive for 3 markers and was the only cell line toshow CD 133 positivity (Supplementary Fig. S2). Therefore, weconsideredCapan-1 to be a rare PDAC line thatmaintainswell-to-moderately differentiated tumor features and thus represents thedominant morphology of clinical PDAC characteristics. In fact,although most clinical human PDACs are well or well-to-mod-erately differentiated cancers according tohistology, andpoorly ormoderate-to-poorly differentiated cancers represent only 10% to15% of cases, most available PDAC cell lines are derived frompoorly or moderate-to-poorly differentiated cancers (13).

High-density lectin microarray analysis of six types ofPDAC cell lines

Glycan expression in Capan-1 cells, in which we have focusedas the tester cell line that may represent PDAC glycan expressions,was compared with that in five other cell lines using high-densitylectin microarrays (14). A representative heat map demonstratedthat Capan-1 and BxPC-3 cells cluster on the same branch of a treediagram, separate from the branch that includes the other four celllines (Fig. 1C). Of the 96 lectins tested, the top 10 lectins withsignificantly different signal intensity in the Capan-1 cell linewereidentified; these included 8 lectins with increased reactivity and 2lectins with decreased reactivity (Supplementary Fig. S3 and TableS1). The most prominent difference was found in the case of therBC2LC-N (recombinant N-terminal domain of BC2L-C) lectin

rBC2LC-N Lectin–Drug Conjugate (LDC) for Pancreatic Cancer

www.aacrjournals.org Mol Cancer Ther; 17(1) January 2018 185

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 4: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

Shimomura et al.

Mol Cancer Ther; 17(1) January 2018 Molecular Cancer Therapeutics186

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 5: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

(Fig. 1D), a TNF-like lectin that was originally identified in thegram-negative bacterium Burkholderia cenocepacia and that specif-ically binds to fucosylated glycan epitopes of H type 1/3/4trisaccharides (17).

The rBC2LC-N lectin exhibited specific affinity forCapan-1 cellsand clinical PDAC samples

The affinity between the rBC2LC-N lectin and Capan-1 cellglycan was further confirmed by live-cell staining (Fig. 1E),flow cytometry analysis using FITC-labeled rBC2LC-N (Fig. 1F;Supplementary Fig. S4), and histochemical staining of anin vivo mouse cell line-derived xenograft model with labeledrBC2LC-N (Fig. 1G; Supplementary Fig. S5). Each experimentrevealed similar strong reactivity of the rBC2LC-N lectin toCapan-1 cells (Fig. 1E–G). To addresswhether reactivitywith the rBC2LC-N lectin is specifically associated with Capan-1 cells or whether itcan be expanded to general clinical PDAC, we analyzed 69humanclinical PDAC specimens by rBC2LC-N lectin histochemical stain-ing. The representative staining patterns of three clinical PDAC ofpoorly (P/D), moderately (M/D) and well-differentiated (W/D)specimens are shown in Fig. 1H. In 7 P/D cases, 6 were weaklypositive (1þ) for rBC2 staining, whereas in 9 W/D cases, 7 werestrongly (2þ) or very strongly (3þ) positive.M/D cases comprisedthe majority (53 cases) of the 69 clinical PDAC cases, and theirrBC2 staining was as follows: 1þ in 18 cases, 2þ in 25 cases, and3þ in 10 cases. The surrounding stromal components werenegative for rBC2 staining (Fig. 1H).

For the top 10 lectins with the highest differences in specificaffinity, lectin histochemical analysis of serial sections of clinicalPDAC specimens demonstrated that rBC2LC-N showed the mostprominent contrast in terms of its intensive affinity for cancer cellsand its lack of affinity for stromal fibroblast cells (SupplementaryFig. S6). Our data indicate that a fucosylated glycan epitope (Htype 1/3/4) that is recognized by the rBC2LC-N lectinmay serve asa promising therapeutic target for PDAC.

Construction of LDC and its cytocidal activity in vitroWith the aim of utilizing the rBC2LC-N lectin (rBC2) as a

therapeutic drug carrier, a 38-kDa region of the catalytic domainof Pseudomonas aeruginosa exotoxin A (PE A) was fused with thelectin sequence to construct LDC(rBC2-PE38; ref. 18; Fig. 2A). Thedissociation constant (Kd) values for rBC2 indicated high-affinitybinding of the conjugate only to Capan-1 cells (Kd: 171.4 nmol/L, Fig. 2B). The cytocidal effect of LDCwasmeasuredusing anMTTassay; IC50 of LDC against Capan-1 cells was 1.04 pg/mL (0.0195

pmol/L, Fig. 2C), nearly 1,000 times lower than that of conven-tional immunotoxins, which generally have IC50 values on theorder of ng/mL (Supplementary Table S2). The other five cell linesthat displayed no affinity for the rBC2LC-N lectin showed mod-erate to negative cytocidal effects (Fig. 2D). Because internaliza-tion into the cell cytoplasm is a key indispensable step for thecytocidal toxicity of PE toxin (19), the endocytosis of rBC2 lectinswas observed.One hour after application of the conjugate, the cellsurfaces of theCapan-1 cellswere surrounded by rBC2 lectins (Fig.2E). Endocytic vesicles containing rBC2 lectin were visible insideCapan-1 cells at 24 hours and more conspicuous at 48 hours. Incontrast to Capan-1 cells that possess positive affinity for rBC2lectins, the negative control cell line (SUIT-2), which has noaffinity for rBC2 lectins, showed no fluorescence intensity on thecell membrane at 24 hours and no endocytic vesicles inside thecytoplasm at 48 hours.

Safety of intravital administration of LDC (rBC2-PE38)We questioned whether our selected lectin would be a safe and

effective drug carrier that could be administered to cancer patientsbecause lectins are often defined by their cell agglutinationactivities. We evaluated the reactivity of the rBC2LC-N lectin onthe surface of noncancerous abdominal organs in mice (Fig. 3A)and confirmed that rBC2LC-N exhibited negative reactivity for theentire parietal peritoneum and the serosal membranes of visceralorgans. The disseminated cancer nodules of Capan-1 cells wereconfirmed to be strongly positive for rBC2LC-N reactivity. Impor-tantly, an in vitro hemagglutination test of erythrocytes frommiceand a human volunteer (blood type A) demonstrated that therBC2LC-N lectin did not induce hemagglutination even at a highconcentration of 50 mg/mL, in contrast to the obvious aggregationcaused by the concanavalin A (Con A) lectin at 1.56 mg/mL inmouse and human erythrocytes (Fig. 3B). LDC (rBC2-PE38)slightly affected hemagglutination at very high concentrations,as demonstrated by a positive result at 25 mg/mL in mouseerythrocytes and 12.5 mg/mL in human erythrocytes (Fig. 3B).Identical results were obtained for human erythrocytes of otherblood types, including type B, AB, andO (Supplementary Fig. S7).In addition, the rBC2LC-N lectin staining of human red bloodcells contaminating clinical PDAC slides revealed that all redblood cells were negative, indicating that human red blood cellsare likely to be negative for the H type 1/3/4 motif (Fig. 3C;Supplementary Fig. S8). Notably, white blood cells (� with bluenuclei) were also negative for rBC2LC-N reactivity.When the LDC50% lethal dose (LD50) was analyzed by i.p. and i.v.

Figure 1.Screening for lectins that specifically bind to PDAC.A, The pathology of a typical PDAC specimen consists of a cluster of ductal spreading cancer cells with abundantstromal components, defined as gland formation (gþþ) and stromal induction (sþþ). B, The morphologies of the 6 pancreatic cancer cell lines in mousexenograftswith respect to gland formation (gþþ,þor�) and stromal induction (sþþ,þor�). Capan-1 cells exhibited strong, organized gland formation (gþþ) anddense stromal proliferation (sþþ). C, High-density lectin microarray analysis of 96 various lectins was performed on 6 PDAC cell lines (n ¼ 6 protein samplesfor each cell line, technical replicates). A heat map of the clustering analysis (yellow: high; black: intermediate; blue: low) placed BxPC-3 and Capan-1 cells on anidentical branch of the tree diagram (above the line). The position of the rBC2LC-N lectin is indicated by an arrow. D, For each of the 96 lectins assessed,the signal intensity in the Capan-1 cell line was compared with that of the other 5 cell lines. The rBC2LC-N lectin exhibited the most robust difference (two-tailedunpaired Student t test). The top 10 lectins that showed specific affinity differences in Capan-1 cells are described in Supplementary Fig. S3 and Table S1.E, The specific affinity of rBC2LC-N for Capan-1 cells in the lectin microarray was further confirmed by live-cell staining using FITC-labeled rBC2LC-N (scale bar, 50mm). F, rBC2LC-N reactivity analysis by flow cytometry confirmed that Capan-1 cells displayed the highest affinity for rBC2LC-N, as indicated by an MFIof 61300. G, Lectin histochemistry of cell-based xenograft models demonstrated strong staining for rBC2LC-N in Capan-1 xenografts (brown signal and bluecounterstaining with hematoxylin) and mosaic staining in BxPC-3 xenografts. The remaining 4 xenografts were negative for rBC2LC-N (scale bar, 100 mm).H, rBC2LC-N lectin histochemistry in human clinical PDAC specimens. Representative micrographs of 3 cases, including P/D with weak (1þ) staining, M/D withmoderate (2þ) staining, and W/D with strong (3þ) staining. I, Among the 69 human clinical PDAC cases, P/D cases tended to show weak staining withrBC2, and W/D cases showed moderate to strong staining. The primary differentiation was M/D (53/69), which demonstrated similar distributionas W/D cases.

rBC2LC-N Lectin–Drug Conjugate (LDC) for Pancreatic Cancer

www.aacrjournals.org Mol Cancer Ther; 17(1) January 2018 187

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 6: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

administration, acceptable values were estimated to be 7.14 mg/mouse (424.8mg/kg) for i.p. and7.22mg/mouse (429.6mg/kg) fori.v. (Fig. 3D). Administration of the BC2LC-N lectin withoutconjugated toxin did not cause death at any dose tested up to15 mg/mouse (892.5 mg/kg), either i.p. or i.v. Based on theseobservations, wewere confident that at least the i.p. injection, andlikely also the i.v. injection, of LDC would be safe for mousemodels in vivo.

Therapeutic effect of LDC in various mouse tumor modelsSubcutaneous xenografts treated by local injection. In cell line-basedmodels, Capan-1 xenografts (positive for the H type 1/3/4 glycan

epitope) were significantly diminished by LDC d.i. (Fig. 4A1–3).However, regarding SUIT-2 xenografts, which exhibit no affinityto the rBC2LC-N lectin, LDC injection did not reduce the tumorsizes except when the dose reached 5 mg/mouse (Fig. 4B1–3). In amore clinically relevant PDXmodel, tumors decreased in size afterLDC administration in a dose-dependent manner (Fig. 4C1–3).

Subcutaneous PDX xenografts treated by i.p. or i.v. injection. Theantitumor effects of LDC administration via the i.p. or i.v. routesfor subcutaneous tumors of the PDX model are shown in Fig.4D1–3. Although these tumors grow rapidly in the control group(normal saline i.p.), LDC treatment groups that received a total of

Figure 2.

LDC (rBC2-PE38) and its in vitro cytotoxicity against PDAC cells. A, The 54-kDa LDC (rBC2-PE38) molecule contains a 16-kDa rBC2LC-N (recombinant N-terminaldomain of BC2 L-C lectin) region and a 38-kDa region representing the catalytic domain of PE A (PE38). B, Binding affinity of the rBC2LC-N lectin in6 PDAC cell lines. Only Capan-1 cells (Kd¼ 171.4 nmol/L) demonstrated high affinity for rBC2LC-N,whereas Kd values could not be estimated for the other cell lines.C,MTT assays using WST-8 were performed to more precisely evaluate the in vitro cytotoxic effect of LDC (black circle). The IC50 value against Capan-1cells (0.0195 pmol/L ¼ 1.04 pg/mL) was substantially lower than that against SUIT-2 (16,600 pmol/L). D, Summary of LDC IC50 values against 6 PDAC cell lines.Capan-1 showed marked sensitivity, BxPC3 showed intermediate sensitivity (IC50 ¼ 9.24 pmol/L ¼ 520 pg/mL), whereas the remaining 4 cell lines had IC50

values on the order of 10,000 pmol/L, indicating no significant affinity for rBC2LC-N. E, Live Capan-1 cells after incubationwith fluorescently labeled rBC2. At 1 hour,confocal laser microscopy showed that the cell surface membranes were covered with rBC2-FITC, but rBC2-FITC had not been ingested by the cells. After 24 hours,fluorescent deposits were observed inside the cells, and these becamemore conspicuous at 48 hours (scale bar, 25 mm). A negative control cell line (SUIT-2) that hasno affinity for rBC2 lectins showed no fluorescence intensity on the cell membrane at 24 hours and no endocytic vesicles inside the cytoplasm at 48 hours.

Shimomura et al.

Mol Cancer Ther; 17(1) January 2018 Molecular Cancer Therapeutics188

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 7: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

Figure 3.

Safety of LDC administration.A, rBC2LC-N lectin reactivity tomouse organ surfaces thatmight be exposed to LDC administered via the i.p. route. Negative reactivityto the entire parietal peritoneum and the visceral peritoneum of hollow, solid and genitourinary organs (arrows) were confirmed, in contrast to the strongpositivity in the Capan-1 disseminated cancer nodules. Positive reactivity to rBC2LC-N in epithelial cells on the internal surface of the hollow organs (strongly in thestomach, duodenum and fallopian tube and weakly in the small intestine and colon) can be ignored in i.p. administration because these cells do not encounter LDCwhen injected via the i.p. route. (scale bar, 100 mm) B, Hemagglutination assay of rBC2LC-N and LDC. The rBC2LC-N lectin alone did not induce erythrocyteaggregation from mice and a human volunteer, even at an extremely high concentration of 50 mg/mL. A positive control with concanavalin A (Con A) lectindemonstrated aggregation at 1.56mg/mL. LDC (rBC2-PD38) slightly affected hemagglutination at the very high concentrations of 25mg/mL inmouse and 12.5mg/mLin human samples. Each test contained n ¼ 3 technical replicates. Other blood types are shown in Supplementary Fig. S7. C, rBC2LC-N lectin stainingof human red blood cells (Type A) contaminating clinical PDAC slides revealed that all red blood cells were negative, indicating that the rBC2LC-N lectin should notinduce the aggregation of red blood cells. Notably, white blood cells (� with blue nucleus) were also negative for rBC2LC-N reactivity (scale bar, 100 mm).Other blood types are shown in Supplementary Fig. S8. D, The 50% lethal dose (LD50) of rBC2LC-N alone and LDC by i.p (n ¼ 10 wild-type mice for eachconcentrations) and i.v. (n ¼ 7) administration were evaluated. The LD50 of a single i.p. and i.v. administration of LDC was calculated to be 7.14 mg/mouseand 7.22 mg/mouse, respectively. By contrast, rBC2LC-N alone did not induce death at any dose tested up to 15 mg/mouse by both i.p. and i.v. administrations.

rBC2LC-N Lectin–Drug Conjugate (LDC) for Pancreatic Cancer

www.aacrjournals.org Mol Cancer Ther; 17(1) January 2018 189

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 8: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

Mol Cancer Ther; 17(1) January 2018 Molecular Cancer Therapeutics190

Shimomura et al.

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 9: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

eight treatments showed reduced growth with either i.v. or i.p.administration. The average tumorweights at day 36were 991mgin the control group, whereas those of i.p. and i.v. administrationgroups were 540.6 and 686.6 mg, respectively (P < 0.01 and 0.05,respectively, relative to control).

Pancreatic orthotopic xenografts treated by i.p. or i.v. injection. Theeffects of 4 LDC treatments (1 mg/mouse) administered via i.p. ori.v. for orthotopic tumors are shown in Fig. 4E1–4. The averagetumorweights after i.p. and i.v. administrationwere 130.8mg and203 mg, respectively (P < 0.001 and 0.05, respectively, relative tothe control value of 390mg). Macroscopic metastases to the liver,spleen, or abdomen occurred frequently in the control groups (3–4/5 mice) but were rare in the LDC treatment groups (0–2/5mice).

Peritoneal dissemination models treated by i.p. or i.v. injection. Byday 14, peritoneal disseminated nodules had grown to a visiblesize and measured 1–3 mm (Fig. 5A). In the Capan-1 group,significantly fewer disseminated nodules were observed upontreatment with either 40 ng of LDC/mouse or 1 mg of LDC/mouse(Fig. 5B). In the SUIT-2 group, for which rBC2 exhibited noaffinity, LDC had no effect (Fig. 5C). In the control groups, theperitoneal cavities of the mice contained abundant cancer cells,whereas there were no cancer cells in the peritoneal cavities of themice in the 1 mg of LDC/mouse treatment group (Fig. 5D). At day45, theweights of themicewere significantly improved both in i.v.and i.p. administrations (Fig. 5E). The effect of LDC administra-tion occurred even after i.v. administration, as shown by theabsence of disseminated nodules (Fig. 5F-1), ascites, and livermetastasis (Fig. 5F-2). Finally, we addressed whether glycan-targeting LDC therapy could cure PDAC in the mouse model ofPDAC (Fig. 5G). The median survival of the control group was 62days, with a 90-day survival rate of 0% (0/9). In contrast, admin-istration of four i.p. injections of 1 mg of LDC resulted in a 90-daysurvival rate of 78% (7/9) and improved themedian survival timesignificantly (105 days; P < 0.0001). The effects of four i.v.injections of 1 mg of LDC were comparable to those of i.p.injection, resulting in a 56% (5/9) 90-day survival rate and amedian survival time of 90 days.

DiscussionThus far, the potential of lectins for in vivo cancer treatment has

not been well tested, likely due to the common belief that alllectins mediate harmful hemagglutination. In this study, wefound a specific affinity between the rBC2LC-N lectin and PDAC

cell surface glycans (fucosylated glycan epitopes of H type 1/3/4trisaccharide), and this lectin was shown to be a safe and efficientdrug carrier in cancer treatment.

The identification of rBC2LC-N as the most highly reactivelectin in PDAC was an unexpected coincidence because we pre-viously reported that rBC2LC-N specifically interactswith inducedpluripotent stem (iPS) and embryonic stem (ES) cells but notwithdifferentiated somatic cells (20). The serendipitousfinding that anrBC2LC-N fucose-binding lectin displays affinity for PDAC andiPS/ES cells indicates that consensus glycan modification ofmembrane proteins or lipids containing H type 1/3/4 glycan onPDAC cells plays a crucial role in the formation and developmentof cancerous tissue. The specific affinity between LDC and cancercell surface H type 1/3/4 glycan was confirmed by competitiveinhibition by adding free rBC2LC-N lectin and free glycan(¼fucose; Supplementary Fig. S9).

One critical questionhas beenwhether the rBC2LC-N lectin cansafely be administered to cancer patients because lectins areinitially defined by their blood agglutination activities (10). Thisconcern was addressed using an in vitro hemagglutination assay;the results showed that the rBC2LC-N lectin did not induce theaggregation of human erythrocytes (Fig. 3B; Supplementary Fig.S7), and the negative rBC2LC-N staining of human red blood cellscontaminating 69 clinical PDAC specimens is shown (Fig. 3C;Supplementary Fig. S8). This finding might be explained by thefact that the ABO blood type glycans on red blood cells containpredominantly H type 2 trisaccharide motifs in which Gal andGlcNAc are connected via beta-1-4 glycosidic bonds (21).rBC2LC-N is known to bind only H type 1/3/4 trisaccharidemotifs and not H type 2 motifs (17). The biosynthesis of H type1 trisaccharidemotif is completed via fucose addition to galactosein type 1 (Gal-GalNAc), or H type 3/4 are generated by fucosyla-tion of type 3/4 disaccharide (Gal-GalNAc) by fucosyltransferases(Fig. 6A; ref. 17). Moreover, the rBC2LC-N lectin was adminis-tered to mice without notable adverse events, further indicatingthat this lectin alone does not induce blood coagulation in vivo(Fig. 3D). Based on our experimental data and the literature, weare confident that the rBC2LC-N lectin could be administered viathe intravenous route without inducing hemagglutination.

The next issue should be the potential nonspecific attachmentof rBC2LC-N to noncancerous normal tissues other than redblood cells. Fucose modification of glycans is one of the repre-sentative cancer-specific changes in glycosylation (5); however,fucosylated type 1/3/4 glycan epitopes are also found in noncan-cerous epithelial cells of the gastrointestinal, respiratory, andreproductive tracts (22). Our results obtained using rBC2LC-Nlectin staining also revealed positive staining of epithelial cells in

Figure 4.In vivo effect of LDC (rBC2-PE38) on subcutaneous tumors in a mouse model after local injection. LDC was directly injected (i.v.) near the subcutaneous tumor4 times, with one injection every 4 days (arrows in A-1, B-1). A-1, Subcutaneous tumor growth curves of Capan-1 xenografts. LDC significantly diminishedthe size of the xenograft tumors in a dose-dependentmanner (n¼6biological replicates).A-2, 3, The objectiveweight of enucleated nodules obtained at day30 alsoconfirmed the prominent reduction of the tumour weight of Capan-1 xenografts by LDC. B-1, 2, 3, Subcutaneous SUIT-2 xenograft model (negative for theH type 1/3/4glycan epitope). LDCd.i. did not induce a tumor reduction effect except at a doseof 5mg/mouse.C-1, 2, 3, In themore clinically relevant PDXmodelwith amoderately differentiatedmorphology, LDCd.i. also decreased the size of the subcutaneous nodules in a dose-dependentmanner (n¼ 5 biological replicates).D-1, 2,3, A PDX model with a moderately differentiated morphology was tested for the effect of LDC via various injection root including i.p and i.v. The growthcurves and average tumor weights after a total of 8 i.p. or i.v. administrations were significantly reduced. E-1-4, The effects of LDC (1 mg/mouse) administeredvia either the i.p. or the i.v. route for orthotopically transplanted Capan-1 tumors. H&E staining and rBC2CL-N lectin histochemistry demonstrated thatCapan-1 cells (rBC2LC-N: positive) had invaded the mouse pancreas parenchyma (negative; scale bars, 100 mm). After 4 injections of LDC by either i.p. or the i.v., thetumor weights were significantly reduced (n ¼ 5 biological replicates.). The number of mice with metastases, including peritoneal dissemination, liverand spleen metastases, was decreased in the treatment groups (0–2/5 mice) relative to the control groups (3–4/5 mice). The results of statistical analysis bytwo-tailed unpaired Student t tests relative to control are indicated as follows: � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; ���� , P < 0.0001; n.s., not significant.

www.aacrjournals.org Mol Cancer Ther; 17(1) January 2018 191

rBC2LC-N Lectin–Drug Conjugate (LDC) for Pancreatic Cancer

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 10: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

Figure 5.

Effectiveness of LDC for the treatment of PDACperitoneal dissemination in amousemodel.A,At day 14 after i.p. injection of Capan-1 or SUIT-2 cells, many opal-colorednodules known as milky spots (arrows) were distributed on the peripheral margin of the small intestine (28). LDC was i.p. or i.v. injected 4 times. B, The mesenteriumafter treatment onday 30. In theCapan-1model, numerousnodules (arrow) remained in the control and rBC2LC-Nalonegroups,whereas noduleswere significantly anddose-dependently reduced in LDC-treated groups (n ¼ 4). C, The number of nodules was not altered in the SUIT-2 model. (Continued on the following page.)

Mol Cancer Ther; 17(1) January 2018 Molecular Cancer Therapeutics192

Shimomura et al.

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 11: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

the exocrine glands of the stomach, duodenum, small intestine,colon and genitourinary tract, presumably associated with secret-ed mucin (Fig. 3A). However, we considered the risk of LDCaffinity for these cells tobenegligible after administration via the i.p. route because mucin secretion predominantly occurs on theinternal luminal surface of halo organs, and the outer serosalsurfaces of these organs are negative for rBC2LC-N reactivity. As

expected, i.p. administration of LDC resulted in a prominenttumor elimination effect in a mouse model of peritoneal dissem-ination of PDAC without adverse events. Though the rBC2 reac-tivity to human organs showed similar distribution with those ofmouse (Supplementary Fig. S10), the different statuses of glyco-sylation and fucosylation between species, i.e., human andmouse, may result in the potential toxicity of LDC in the human

Figure 6.

A, Schema of rBC2LC-N lectin bindingsites. rBC2LC-N is a TNF-like lectinmolecule identified from the gram-negative bacteriumB. cenocepacia, andthis molecule is revealed to be a fucose-binding lectin (17). Precise survey usinga glycan array containing 377 glycansrevealed that the promising high-affinity ligands of rBC2LC-N are the Htype 1 trisaccharide (Fuca1-2 Galb1-3GlcNAc) and H type 3/4 trisaccharide(Fuca1-2 Galb1-3GalNAc),corresponding to two forms of thehuman blood group O determinant. TheH type 1 trisaccharidemotifs are presentat the end of branchedN-linked glycansand O-linked glycans. The proximateGalNAc in O-linked glycan could be asource of the H Type III trisaccharidemotif. The end glycolipid, i.e., bGalNAcattached to the Globo H construct,could also be a source of the H type IVglycan motif. B, Schema of rBC2LC-Nlectin binding on the cancer cellsurface. Right, The previous failure ofADC strategies may be due to the one-to-one relationship between the cancercell surface peptide antigen (blue star inbrown glycoprotein) and the antibody,which results in the delivery of aninsufficient amount of the drug tocancer cells. Left, In contrast, LDCbinding sites could consist of multipleglycans on various glycoproteins andglycolipids, resulting in the delivery ofmore drug to the surface of target cells.In addition, the relatively large size ofimmunoglobulins tends to preventeffective internalization of antibody-based drugs; however, the relativelysmall (16-kDa) size of rBC2LC-N wouldfacilitate effective endocytosis and theprominent cytocidal effect of LDC.

(Continued.) D, Microscopic observation of the resected abdominal wall (�, skin surface; †, peritoneum) revealed numerous disseminated cancer cells (arrows) in thecontrol, whereas cancer cells were completely absent after i.p. injection of 1 mg of LDC (H&E staining, scale bar, 500 mm). E, Body weight at day 45 wassignificantly reduced in the control and rBC2-lectin-without-toxin cohorts (n¼ 9 biological replicates); n.s., not significant; � , P < 0.05; ��� , P < 0.001; ���� , P < 0.0001(two-tailedunpaired Student t test). Intraperitoneal injection of LDC resulted ingoodweight gain, andbodyweightwas comparable between the i.v. administration andi.p. administration of LDC cohorts. All mice were healthy and did not exhibit damage to noncancerous epithelial cells of hollow or genitourinary organs, andthe body weight of the mice was not affected by this treatment. F-1, Intravenous injection of LDC decreased peritoneal dissemination nodules of PDAC in the mousemodel. F-2, Laparotomy in the 1 mg i.v. LDC cohort revealed no ascites and the mice appeared slim and normal. F-3, The multiple peritoneal disseminatednodules (arrows) observed in the control. F-4, Laparotomy of controls revealed massive bloody ascites due to active disseminated cancer nodules. F-5, Disseminatednodules at the surface of the liver and nodules at the subdiaphragmatic space (arrows). G, Kaplan–Meier survival curves after LDC treatment in the PDACdisseminationmousemodel (n¼ 9 for each group). Themedian number of days of survival in the control group (62 days)was significantly improvedby i.p. injection of 1mg of LDC, resulting in 90-day survival of 7/9 mice (median survival ¼ 105 days). Notably, i.v. injection of 1 mg of LDC conferred a comparable effect, with 90-daysurvival of 5/9 mice (median survival ¼ 90 days; � , P < 0.05; ���� , P < 0.0001; n.s., not significant by log-rank test).

www.aacrjournals.org Mol Cancer Ther; 17(1) January 2018 193

rBC2LC-N Lectin–Drug Conjugate (LDC) for Pancreatic Cancer

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 12: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

setting. Even after i.v. administration, distribution to noncancer-ous organs shouldbe very limitedwith the enhanced permeabilityand retention (EPR) effect, which results in predominant deliveryof large molecules to cancer tissues via the damaged endothelialcell linings of capillaries in the region of the tumor (23). In fact,the i.v. injection of 1 mg of LDC/mouse also shrank tumors in theorthotopic (Fig. 4E) and peritoneal dissemination model ofCapan-1 cells (Fig. 5F) and subcutaneous PDX tumor (Fig. 4D)with an efficacy identical to that of i.p. injection without notableadverse events. However, it should also be noted that the EPReffect showing the predominant accumulation of LDC in PDXtumors in mouse models may not be reproduced in clinicalPDAC, because human PDACs are known to be hypoperfusedand poorly vascularized compared with xenotransplanted tumors(24, 25).

Compared with the affinity of antibodies for peptide antigens(Kd ¼ 0.1–10 nmol/L), the affinity of lectins for their targetglycans is typically weaker (Kd ¼ 100–1,000 nmol/L). However,LDC possesses the following notable advantages (shown sche-matically in Fig. 6B): (i) the target glycans are located at theoutermost coatings of cancer cells, known as glycocalyx, confer-ring easy access for targeting drugs; (ii) the cancer cells possessabundant glycan sites for interaction with LDC, includingbranched glycan chains attached to multiple different core pro-teins; (iii) the small size of the lectin should be beneficial for drugendocytosis; and (iv) the glycolipids are also the binding target ofLDC, and the very close proximity of glycolipids to the cellmembrane should be favorable for drug endocytosis. As a com-prehensive result, the lectin drug displayed a remarkable IC50

valueof 0.0195pmol/L, nearly 1,000 times lower than the IC50s ofconventional immunotoxins, which are on the order of ng/mL(Supplementary Table S2); thus, the lectin drug provides a prom-inent cancer therapeutic effect. As lectins can be generated in aneconomical bacterial system, lectin-based drugs should be advan-tageous from the viewpoint of medical economics in comparisonto antibody-based drugs that require costly eukaryotic cells fortheir production (26).

rBC2LC-N may be a unique lectin that lacks typical hemagglu-tination activity. However, the rapid improvement in lectin engi-neering technologies (27) will provide control over the harmfuladverse effects of some lectins, and synthetic lectin technologieswill permit the development of de novo lectins with ideal char-

acteristics as drug carriers. This study provides new insight intocancer treatments that might serve as alternatives to expensiveantibody-based drugs. Moreover, the application of tumor-target-ing lectins might be expanded by coupling them to small-mol-ecule agents or to nanoparticles, potentially leading to break-throughs in cancer treatment.

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

Authors' ContributionsConception and design: O. Shimomura, T. Oda, H. Tateno, N. OhkohchiDevelopment of methodology: O. Shimomura, T. Oda, H. TatenoAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): O. Shimomura, T. Oda, H. Tateno, Y. Ozawa,S. Kimura, S. SakashitaAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): O. Shimomura, T. Oda, H. TatenoWriting, review, and/or revision of the manuscript: O. Shimomura, T. Oda,H. Tateno, J. HirabayashiAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): H. Tateno, M. NoguchiStudy supervision: T. Oda, J. Hirabayashi, M. Asashima, N. Ohkohchi

AcknowledgmentsWe thank Dr. Sho Tachino, Dr. Jyunji Matsui (Biology Tsukuba Oncology

PCU, Eisai Product Creation Systems, Eisai Co., Ltd.), and Ako Takahashi(Department of Surgery, University of Tsukuba, Tsukuba, Ibaraki, Japan) fortechnical assistance; Professor Susumu Nishimura (Laboratory AnimalResource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan), ProfessorEric O'Neill (CRUK/MRC Oxford Institute for Radiation Oncology, Oxford,UK), andDr. KazukiN. Sugahara (Department of Surgery, ColumbiaUniversity,New York, NY, USA) for valuable scientific discussions and strengthening themanuscript.

This studywas supported by grants to T. Oda from theMinistry of Education,Culture, Sports, Science, and Technology of Japan (KAKENHI, 15H04924) andfrom University of Tsukuba and AIST, Tsukuba-city, Japan (the "Awasewazafund").

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.

ReceivedMarch13, 2017; revised July 12, 2017; accepted September 14, 2017;published OnlineFirst September 22, 2017.

References1. Teicher BA, Doroshow JH. The promise of antibody-drug conjugates. N

Engl J Med 2012;367:1847–8.2. Sharkey RM, McBride WJ, Cardillo TM, Govindan SV, Wang Y, Rossi EA,

et al. Enhanced delivery of SN-38 to human tumor xenografts with an Anti-Trop-2-SN-38 antibody conjugate (Sacituzumab Govitecan). Clin CancerRes 2015;21:5131–8.

3. Zhu Y, Choi SH, Shah K. Multifunctional receptor-targeting antibodies forcancer therapy. Lancet Oncol 2015;16:e543–54.

4. Tarbell JM, Cancel LM. The glycocalyx and its significance in humanmedicine. J Intern Med 2016;280:97–113.

5. Pinho SS, Reis CA. Glycosylation in cancer: mechanisms and clinicalimplications. Nat Rev Cancer 2015;15:540–55.

6. Christiansen MN, Chik J, Lee L, Anugraham M, Abrahams JL, Packer NH.Cell surface protein glycosylation in cancer. Proteomics 2014;14:525–46.

7. Reticker-FlynnNE, Bhatia SN. Aberrant glycosylation promotes lung cancermetastasis through adhesion to galectins in the metastatic niche. CancerDiscov 2015;5:168–81.

8. Dingjan T, Spendlove I, Durrant LG, Scott AM, Yuriev E, Ramsland PA.Structural biology of antibody recognition of carbohydrate epitopes andpotential uses for targeted cancer immunotherapies. Mol Immunol2015;67:75–88.

9. Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, MatrisianLM. Projecting cancer incidence anddeaths to 2030: theunexpected burdenof thyroid, liver, and pancreas cancers in the United States. Cancer Res2014;74:2913–21.

10. Sharon N, Lis H. History of lectins: from hemagglutinins to biologicalrecognition molecules. Glycobiology 2004;14:53r–62r.

11. Rabinovich GA, Croci DO. Regulatory circuits mediated by lectin-glycaninteractions in autoimmunity and cancer. Immunity 2012;36:322–35.

12. Belardi B, Bertozzi CR. Chemical lectinology: tools for probing the ligandsand dynamics of mammalian lectins in vivo. Chem Biol 2015;22:983–93.

13. Deer EL, Gonzalez-Hernandez J, Coursen JD, Shea JE, Ngatia J, Scaife CL,et al. Phenotype and genotype of pancreatic cancer cell lines. Pancreas2010;39:425–35.

Mol Cancer Ther; 17(1) January 2018 Molecular Cancer Therapeutics194

Shimomura et al.

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 13: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

14. Kuno A,UchiyamaN, Koseki-Kuno S, Ebe Y, Takashima S, YamadaM, et al.Evanescent-field fluorescence-assisted lectin microarray: a new strategy forglycan profiling. Nat Methods 2005;2:851–6.

15. Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, et al.Distinct populations of cancer stem cells determine tumor growth andmetastatic activity in human pancreatic cancer. Cell Stem Cell 2007;1:313–23.

16. Li C,Heidt DG,Dalerba P, Burant CF, Zhang L, Adsay V, et al. Identificationof pancreatic cancer stem cells. Cancer Res 2007;67:1030–7.

17. SulakO,CiociG,DeliaM, LahmannM,Varrot A, Imberty A, et al. A TNF-liketrimeric lectin domain from Burkholderia cenocepacia with specificity forfucosylated human histo-blood group antigens. Structure 2010;18:59–72.

18. Tateno H, Onuma Y, Ito Y, Minoshima F, Saito S, Shimizu M, et al.Elimination of tumorigenic human pluripotent stem cells by a recombi-nant lectin-toxin fusion protein. Stem Cell Rep 2015;4:811–20.

19. Alewine C, Hassan R, Pastan I. Advances in anticancer immunotoxintherapy. Oncologist 2015;20:176–85.

20. Tateno H, Toyota M, Saito S, Onuma Y, Ito Y, Hiemori K, et al. Glycomediagnosis of human induced pluripotent stem cells using lectinmicroarray.J Biol Chem 2011;286:20345–53.

21. ZhangW, Zhu ZY. Structural modification of H histo-blood group antigen.Blood Transfus 2015;13:143–9.

22. Stanley P, Cummings RD. Structures Common toDifferentGlycans. In: AjitVarki, Cummings RD, Esko JD, et al. , editors. Essentials of glycobiology,2nd ed. New York: Cold Spring Harbor Laboratory Press; 2009.

23. Matsumura Y,MaedaH. Anew concept formacromolecular therapeutics incancer chemotherapy: mechanism of tumoritropic accumulation of pro-teins and the antitumor agent smancs. Cancer Res 1986;46:6387–92.

24. Akashi Y, Oda T, Ohara Y, Miyamoto R, Hashimoto S, Enomoto T, et al.Histological advantages of the tumor graft: a murine model involvingtransplantation of human pancreatic cancer tissue fragments. Pancreas2013;42:1275–82.

25. Akashi Y, Oda T, Ohara Y, Miyamoto R, Kurokawa T, Hashimoto S, et al.Anticancer effects of gemcitabine are enhanced by co-administered iRGDpeptide inmurine pancreatic cancermodels that overexpressed neuropilin-1. Br J Cancer 2014;110:1481–7.

26. Bach PB. New Math on Drug Cost-Effectiveness. N Engl J Med 2015;373:1797–9.

27. Hu D, Tateno H, Hirabayashi J. Lectin engineering, a molecular evolu-tionary approach to expanding the lectin utilities. Molecules 2015;20:7637–56.

28. Hagiwara A, Takahashi T, Sawai K, Taniguchi H, ShimotsumaM, Okano S,et al. Milky spots as the implantation site for malignant cells in peritonealdissemination in mice. Cancer Res 1993;53:687–92.

www.aacrjournals.org Mol Cancer Ther; 17(1) January 2018 195

rBC2LC-N Lectin–Drug Conjugate (LDC) for Pancreatic Cancer

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232

Page 14: A Novel Therapeutic Strategy for Pancreatic Cancer ... · approved in the clinic (8). Thus, novel powerful therapeutic methods targeting cancer cell surface glycans that are not based

2018;17:183-195. Published OnlineFirst September 22, 2017.Mol Cancer Ther   Osamu Shimomura, Tatsuya Oda, Hiroaki Tateno, et al.  

Drug Conjugate (LDC)−Surface Glycan Using rBC2LC-N Lectin A Novel Therapeutic Strategy for Pancreatic Cancer: Targeting Cell

  Updated version

  10.1158/1535-7163.MCT-17-0232doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2017/09/22/1535-7163.MCT-17-0232.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/17/1/183.full#ref-list-1

This article cites 27 articles, 8 of which you can access for free at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://mct.aacrjournals.org/content/17/1/183To request permission to re-use all or part of this article, use this link

on October 20, 2020. © 2018 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst September 22, 2017; DOI: 10.1158/1535-7163.MCT-17-0232