Site-specific PEGylation of anti-mesothelin recombinant ... · 12/21/2019  · pegylation was...

37
1 Site-specific PEGylation of anti-mesothelin recombinant immunotoxins increases half-life and anti-tumor activity Authors: Zeliang Zheng 1 , Ryuhei Okada 2 , Hisataka Kobayashi 2 , Tadanobu Nagaya 2 , Junxia Wei 1 , Qi Zhou 1 , Fred Lee 1 , Tapan K Bera 1 , Yun Gao 3 , William Kuhlman 3 , Chin-Hsien Tai 1 , and Ira Pastan 1 * Author Affiliations: 1 Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264 2 Laboratory of Molecular Theranostics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264 3 Selecta Biosciences, 480 Arsenal Way, Watertown, MA 02472 *Correspondence: Dr. Ira Pastan, Laboratory of Molecular Biology, National Cancer Institute, 37 Convent Drive, Room 5106, Bethesda, MD 20892-4264 e-mail: [email protected]; Tel: (240) 760-6470; Fax: (240) 541-4501 Running Title: Site specific PEGylation improves immunotoxin activity Keywords: Mesothelin, mesothelioma, cancer therapy, immunotherapy, pegylation Conflict of Interest Statement: I.P. is an inventor on several patents on immunotoxins that have all been assigned to the NIH. on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

Transcript of Site-specific PEGylation of anti-mesothelin recombinant ... · 12/21/2019  · pegylation was...

  • 1

    Site-specific PEGylation of anti-mesothelin recombinant

    immunotoxins increases half-life and anti-tumor activity

    Authors: Zeliang Zheng1, Ryuhei Okada

    2, Hisataka Kobayashi

    2, Tadanobu Nagaya

    2, Junxia

    Wei1, Qi Zhou

    1, Fred Lee

    1, Tapan K Bera

    1, Yun Gao

    3, William Kuhlman

    3, Chin-Hsien Tai

    1, and

    Ira Pastan1*

    Author Affiliations:

    1Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute,

    National Institutes of Health, Bethesda, MD 20892-4264

    2Laboratory of Molecular Theranostics, Center for Cancer Research, National Cancer Institute,

    National Institutes of Health, Bethesda, MD 20892-4264

    3Selecta Biosciences, 480 Arsenal Way, Watertown, MA 02472

    *Correspondence: Dr. Ira Pastan, Laboratory of Molecular Biology, National Cancer Institute,

    37 Convent Drive, Room 5106, Bethesda, MD 20892-4264

    e-mail: [email protected]; Tel: (240) 760-6470; Fax: (240) 541-4501

    Running Title: Site specific PEGylation improves immunotoxin activity

    Keywords: Mesothelin, mesothelioma, cancer therapy, immunotherapy, pegylation

    Conflict of Interest Statement:

    I.P. is an inventor on several patents on immunotoxins that have all been assigned to the NIH.

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 2

    Abstract

    Recombinant Immunotoxins (RITs) are chimeric proteins containing an Fv that binds to tumor

    cells, fused to a fragment of Pseudomonas exotoxin (PE) that kills the cell. Their efficacy is

    limited by their short half-life in the circulation. Chemical modification with polyethylene glycol

    (PEG) is a well-established method to extend the half-lives of biologics. Our goal was to

    engineer RITs with an increase in half-life and high cytotoxic activity. We introduced single

    cysteines at different locations in five anti-mesothelin RITs and employed site-specific

    PEGylation to conjugate them to 20kD PEG. Because our previous PEGylation method using β-

    mercapto-ethanol reduction, gave poor yields of PEG-modified protein, we employed a new

    method using TCEP to reduce the protein, and could PEGylate RITs at ~90% efficiency. The

    new proteins retained 19-65% of cytotoxic activity. Although all proteins are modified with the

    same PEG, the radius of hydration varies from 5.2 to 7.1 showing PEG location has a large effect

    on protein shape. The RIT with the smallest radius of hydration has the highest cytotoxic

    activity. The PEGylated RITs have a 10-30-fold increase in half-life which is related to the

    increase in hydrodynamic size. Biodistribution experiments indicate that the long half-life is due

    to delayed uptake by the kidney. Anti-tumor experiments show that several PEG-RITs are much

    more active than unmodified RIT and the PEG location greatly affects anti-tumor activity. We

    conclude that PEGylation is a useful approach to improve the half-life and anti-tumor activity of

    RITs.

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 3

    Introduction

    Small therapeutic proteins have limited efficacy because they are rapidly removed from

    circulation (1). Two well-known mechanisms of removal are renal filtration and hepatic

    elimination. Increasing the hydrodynamic volume is a widely adopted strategy to increase half-

    life. Other strategies include fusion with albumin, albumin binding domain, Fc domain of an

    antibody, or N-terminal glycosylation (1). Most of these strategies take advantage of the

    naturally occurring FcRn-mediated, pH-dependent recycling of IgG and albumin that is

    responsible for their exceptionally long half-life.

    RITs are small chimeric proteins consisting of the Fv portion of an antibody that serves

    as the targeting moiety, fused to a bacterial toxin that kills the target cells (2). Our lab studies

    RITs that target mesothelin, a cell-surface glycoprotein that is robustly expressed in many

    common solid tumors (3). The bacterial toxin is Pseudomonas exotoxin A (PE). To make RITs

    domain I, the cell-targeting domain of PE is replaced by an anti-mesothelin Fv or Fab and

    unnecessary portions of domain II are deleted. The size of the RITs ranges from 50 to 72kD, and

    they have half-lives of 10 to 20 minutes in mice. Recently we reported that the addition of an

    albumin binding domain to a RIT targeting mesothelin greatly increased half-life and anti-tumor

    activity (4). This finding encouraged us to explore other approaches to increase half-life that do

    not require the addition of a foreign protein domain, which could increase immunogenicity.

    PEGylation is an approach in which polyethylene glycol (PEG) is conjugated to a

    macromolecule. It is a well-established method for half-life extension and does this by increasing

    the hydrodynamic volume and reducing renal filtration (5). Currently, there are 14 FDA

    approved PEGylated drugs on the market and 20 more are in clinical trials. They range from

    proteins to liposomes, indicating the versatile nature of PEGylation (6). Our initial effort at

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 4

    PEGylation was to modify human transforming growth factor- fused to the 38kD PE38; this

    resulted in a protein with enhanced plasma half-life and anti-tumor activity; however, the

    pegylation was lysine-specific that resulted in heterogenous populations of PEG-modified

    immunotoxin because there are multiple lysines presented (7). We subsequently showed that

    PEGylation of the Fv portion of LMB-2, a RIT composed of an Fv that binds to CD25 fused to

    the PE38 also increased half-life, reduced toxicity and immunogenicity (8). The drawback of this

    study was that the method involved reduction of the protein with β-mercaptoethanol resulting in

    variable and poor yields and purity. Therefore, we could only modify a single position in the

    immunotoxin. Our goal in this study was to make multiple PEG-modified immunotoxins, each

    with a single PEG site-specifically conjugated at a unique position in order to find a site that

    retains high cytotoxic activity, a long half-life in the circulation and high anti-tumor activity. In

    order to do this, we needed to develop a new method of PEGylation using TCEP to reduce the

    protein (9).

    The mechanism by which RITs kill mesothelin expressing cancer cells consists of several

    steps. First the RIT binds to the cell through recognition of mesothelin, followed by

    internalization through endocytosis. Then the toxin is cleaved from the Fv by the furin protease

    and undergoes retrograde transport through the Golgi to the endoplasmic reticulum (ER). Next

    the toxin is released into the cytosol where it ADP ribosylates and inactivates elongation factor 2

    (EF2); this halts protein synthesis causing cell death (3). Given the complex network of

    interactions, it is important to identify positions on the RIT where PEG can be placed that do not

    decrease the activity of the RIT by sterically hindering mesothelin binding, furin cleavage,

    retrograde transport to the ER, transfer to the cytosol from the ER and ADP-ribosylation of EF2

    (8).

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 5

    To overcome these challenges, we adopted approaches that would give us the best chance

    to produce active protein: First we used a single-chain Fv as the targeting moiety, because it only

    contains buried disulfide bonds, which cannot be PEGylated. Second, we exploited structural

    modeling of the mesothelin-RIT-EF2 complex to locate cysteines at positions distant from the

    mesothelin and EF2-binding sites. Third, we placed cysteines on spatially and functionally

    distinct domains. These were located on the Fv, next to the furin cleavage site, and on domain III

    of the toxin. Fourth, we employed the highly efficient maleimide-based site-specific conjugation

    to prevent random PEGylation.

    Materials and Methods

    Materials: methoxy-PEG-maleimide (molecular weight 20 kD) was obtained from JenKem

    Technology. Other materials were obtained from standard sources.

    Bacterial Strains and Plasmids: Escherichia coli DH5 (High Efficiency) was obtained from

    New England Biolabs for the propagation of plasmids. E. coli BL21(DE3), which carries T7

    RNA polymerase gene under the control of an inducible promoter on a prophage, was used as a

    host to express RIT. Plasmids that express RIT are under the control of T7 promoter and contain

    a single-chain Fv that is genetically fused to a 24kD bacterial toxin PE by a flexible GS linker

    and a furin cleavage site.

    Construction, Expression, and Purification of RIT: Double-stranded gene fragment (gBlock,

    Integrated DNA Technologies) that contains the designated cysteine was ligated onto our

    standard laboratory RIT production vector as described before (10) using Gibson Assembly

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 6

    Master Mix (New England Biolabs) according to the manufacturer’s protocol. The correct

    assembly of plasmids was confirmed by cutting with the appropriate restriction enzymes and

    sequencing analysis. The plasmid was transformed into E. coli. BL21(DE3) and RIT was

    induced with 1mM IPTG at OD.2.5 for 2 hours. All RITs were expressed and purified as

    inclusion body according to the protocol described before (11). Briefly, inclusion body was

    dissolved in GTE buffer (6M guanidine-HCl, 100mM Tris-HCl, 2mM EDTA) for 19 hours,

    followed by refolding in 1000ml 100mM Tris-HCl, 1mM EDTA, 0.5M arginine and 0.5M

    NDSB-201, pH 10.0 for 31 hours and dialysis against 50 liters of 30mM Tris-HCl, 0.1M urea for

    19 hours. The refolded RITs were purified through Q-sepharose and MonoQ ion exchange

    columns (GE Healthcare)(10).

    PEGylation of RIT: RITs were PEGylated in collaboration with Selecta Bioscience. 4mg RIT

    was buffer exchanged into 20mM potassium phosphate, pH 8.0, 2mM EDTA using Vivaspin 20

    (5kD MWCO, GE healthcare) until the pH of the flow through matches that of the buffer. A 3-

    molar excess of tris(2-carboxyethyl)phosphine (TCEP) was added to the final 2mg/ml RIT and

    incubated at room temperature for 1-hour to reduce cysteine. Then a 10-molar excess of

    methoxy-PEG maleimide was added and left to react for overnight. For purification the

    PEGylated RIT was buffer exchanged into 10mM Tris-HCl, pH 8.0 using a Vivaspin column and

    applied to an anion exchange spin column (Pierce). Unreactive methoxy-PEG maleimide was

    washed away with four 10 ml washes of the same buffer and the RIT was eluted with 1XPBS.

    Samples collected before and after TCEP treatment, and after the final elution were analyzed on

    SDS-PAGE gels. For high-performance liquid chromatography (HPLC) analysis, additional

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 7

    samples were collected during column washes to make sure methoxy-PEG maleimide was

    completely gone.

    HPLC Analysis: Reverse phase HPLC was performed to separate PEGylated RIT from the

    parental RIT and unreacted PEG. The column was purchased from Water Corporation and

    features a non-polar C4 stationary phase with 3.5m particle size and 2.1mm I.D. Mobile phases

    A and B contain 5% and 90% acetonitrile, respectively and are supplemented with 0.1%

    trifluoroacetic acid. 5l of ~1mg/ml samples were injected into the gradient run, starting from

    95% buffer A to 100% buffer B in 10min.

    Mesothelin Binding: A 96-well microtiter plate was coated with 50ul of 1g/ml MSLN-hFc, a

    fusion protein consisting of human IgG Fc and mesothelin protein and incubated at 4°C

    overnight. The plate was blocked with blocking buffer (1x PBS supplemented with 25% DMEM,

    25mM HEPES, 0.5% BSA, 0.1% Azide, 5% FBS) and 50l serial dilution of the PEGylated

    RITs was added in triplicate and 50l monoclonal anti-PE antibody (IP12) and HRP-labeled goat

    anti-mouse IgG were added as a primary and secondary antibodies (4). After washing with

    blocking buffer, the plate was developed with TMB substrate kits (Thermos Scientific) and

    subsequently read with the plate reader at 640 and 450nm wavelength. Binding curve was

    generated using GraphPad and 50% binding (IC50) was calculated. Table 1 summarizes average

    IC50 values of two experiments.

    ADP-ribosylation: A 20l reaction was set up to contain 4l buffer (20mM Tris-HCl, pH 7.5,

    1mM EDTA), 1ul 1M DTT, 1ul 20ng/ul RIT, 5g protein lysate (prepared from KLM1 cell) and

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 8

    1l 250uM Biotin-NAD (Trevigen) and incubated at room temperature for 1 hour. Samples were

    analyzed in SDS-PAGE gel and western blot was performed. The PVDF membrane was probed

    with HRP-streptavidin followed by ECL development to visualize ADP-ribosylated EF2.

    Cytotoxicity Assay: Human cell lines KLM1 (provided by Dr. U. Rudloff, National Cancer

    Institute; 12), L55 (provided by Dr. T. Yamori, Pharmaceuticals and Medical Device Agency;

    13) and A431/H9 (A431 human carcinoma cells transfected with mesothelin cDNA; 14) were

    described previously and confirmed by short tandem repeat (STR) testing. MTBM test results

    were all negative. Cells were grown in RPMI media supplemented with 10% FBS and 1%

    penicillin-streptomycin at 37°C until 70-78% confluency before they were trypsinized. Then

    4000 cells were seeded onto each well in a 96-well plate, and serial dilutions of RIT were added

    in triplicate rows and incubated for 72 hours. Cytotoxicity activity was evaluated with a WST8

    assay according to the manufacturer’s protocol (Dojindo Molecular Technologies). Plates were

    read at 640 and 450nm wavelength, and data were plotted using GraphPad to obtain IC50 values.

    Half-Life Assay: For each protein two mice (6-8 weeks old, 23-25g) were I.V. injected with

    25g RIT. Blood was harvested at 5min, 2, 4, 8 and 24 hours for LMB-203-PEG, LMB-244-

    PEG, and LMB-249-PEG and 5min, 1, 4, 24 hours for LMB-163-PEG and LMB-179-PEG

    afterward, and serum was isolated. ELISA was used to determine the amount of RIT at each time

    point as described in Mesothelin Binding experiment. Half-life and AUC was calculated using

    GraphPad, with two-phase decay fitting, except LMB-12 which was fitted with one-phase decay.

    The 5min sample was used as the initial amount of RIT in the blood, and the later time points

    were normalized against this value.

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 9

    Radius of hydration (Rh): The Rh of five PEGylated proteins was measured using Dynamic

    Light Scattering (Wyatt Technology). Proteins in the range of 0.5-1mg/ml were spun down at

    13,000rpm for 5 min to pellet aggregates and 20l was transferred to a quartz cuvette slowly to

    avoid bubbles. Each sample was measured at least two times, and each time constitutes an

    average of 30 measurements. Rh values were extracted and plotted against the half-life,

    cytotoxicity, and anti-tumor activity using GraphPad. Linear regression analysis was performed

    to obtain a line of best-fit and R2 values.

    Biodistribution of RIT: 5.8nmol of LMB-249-PEG and 4.0nmol of SS1P were incubated with

    23.1nmol and 15.9 nmol of FNIR-Z-759 in PBS buffer, pH 8.5, at room temperature for 1 hour,

    respectively. The labeled protein was purified with Sephadex G25 column. 3 mice bearing

    ~100mm3

    A431/H9 tumor were I.V. injected with 41g (580pmol) of labeled LMB-249-PEG or

    25g (400pmol) of labeled SS1P, followed by serial dorsal and ventral 800-nm fluorescence

    imaging immediately and 15, 30min, 1, 2, 3, 4, 5, 6, 9, 12, 24, 48hours afterward. Fluorescence

    accumulation in the kidney, liver, and tumor was monitored. Background fluorescence was also

    calculated to obtain the target-to-background ratio. Plots were generated using GraphPad from

    average results of 3 mice.

    Anti-tumor Experiment: All animal experiments were performed in accordance with NIH

    guidelines and approved by the NCI Animal Care and Use Committee. Female (10 per group)

    nu/nu mice (6-week-old, 20-25g) were injected s.c. in the flank with 5 x 106 KLM1 cells (12).

    When the tumor reached 100mm3, mice were grouped into similar weight and tumor size and

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 10

    RIT in PBS supplemented with 0.2% HSA was I.V. injected at 10g/mouse three times per week

    for one week. Tumor volume and mice weight were monitored every two to three days. Mice

    were euthanized if they lost more than 10% of the body weight. The experiment was repeated for

    LMB-244-PEG and LMB-163-PEG (n = 10) and injected at 20g/mouse, two times per week for

    two weeks. Mouse weight was monitored every two to three days and were euthanized if they

    lost more than 10% of the body weight.

    Results

    Design of RITs

    The RITs used in this study are shown in Figure 1A. SS1P is the first immunotoxin made

    to target mesothelin and contains an anti-mesothelin Fv attached to PE38 that contains domains

    II and III of PE (15). LMB-12 and LMB-84 both have domain II deleted and contain an 11-

    amino acids furin cleavage site that connects the Fv to domain III (PE24). In LMB-12, the Vl

    and Vh are connected by a disulfide bond and in LMB-84 they are connected by a flexible 15-

    residues (G4S)3 linker. LMB-179, LMB-244, LMB-203, LMB-249, and LMB-163 are all

    derived from LMB-84, but have cysteines used for PEGylation. They also contain a 17-residue

    GS linker placed either between the Vl and the furin cleavage site (LMB-179, LMB-244, and

    LMB-203) or at the N-terminus of the Vh (LMB-249). LMB-163 is similar to LMB-84 but

    contains mutations that humanize the Fv portion. Figure 1B shows a cartoon representation of

    the RITs. The 20kD PEG is shown as a red wiggly line.

    We use site directed mutagenesis to insert cysteine residues at various locations in the

    RIT. To choose locations that are less likely to interfere with immunotoxin activity, we modeled

    the RIT using the existing structures of its various components. The model of immunotoxin is

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 11

    shown in Figure 2A. The structural complex of the immunotoxin bound to mesothelin and EF2 is

    shown in Figure 2B, and it was generated by superposing the templates of mesothelin-mAb

    (PDB 4F3F (16)), scFV (PDB 3GKZ (17)), PE-EF2 (PDB 1ZM4 (18)) and PE-NAD-AMP (PDB

    1DMA (19)) in UCSF Chimera (20). Figure 2C shows a model of 20kD PEG, composed of 454

    units of ethylene glycol, conjugated to immunotoxin at D406 of domain III. Five different sites

    were mutated to cysteine for PEG conjugation as indicated by red balls. For all five locations, we

    tried to diminish interference with mesothelin binding, processing by furin protease and ADP-

    ribosylation of EF2.

    LMB-179 and LMB-244 have cysteine residues located in domain III at E522 and D406,

    respectively (Figure 1). The cysteine residues are located on different sides of PE24 and are

    distant from the EF2 binding site (Figure 2A). LMB-203 has the cysteine placed in the middle of

    a 17 amino acid linker connecting the Fv to the furin cleavage peptide (Figure 1). It is 8 residues

    away from the furin cleavage site in order to diminish effects on cleavage or mesothelin binding

    to the Fv or EF2 binding to domain III. In LMB-249 the cysteine residue is located on the amino

    terminus of a 17-residue GS linker attached to the first residue of the heavy chain of the Fv

    (Figure 1 and 2); this location is unlikely to interfere with the binding of mesothelin to the CDRs

    of the Fv. LMB-163 does not contain an extra 17-residues GS linker like the other four RITs.

    The cysteine is placed at S63 in the light chain (Figure 1 and 2). We chose S63 to mutate to

    cysteine because the residue is surface-exposed and distant from the CDRs and the furin

    cleavage peptide so the mutation is less likely to interfere with binding to mesothelin or cleavage

    of the RIT by furin protease. All of RITs were expressed in E. coli in the form of inclusion

    bodies, which were denatured, refolded and purified through Q Sepharose and Mono Q columns.

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 12

    PEGylation of RITs

    Because a free thiol can lead to dimer formation, we stopped the purification after the

    Mono Q column, where we usually observed a peak of monomer and another of dimer. Figure

    3A shows Mono Q fractions of LMB-179, LMB-244, LMB-203 and LMB-249 that are mostly

    monomer (lanes 2, 5, 6, and 8) or dimer (lanes 3, 4, 7, and 9, respectively). LMB-163 only has

    monomer fractions (lanes 10 and 11). Monomer runs at ~50kD, and dimer runs slightly above

    100kD, consistent with the predicted molecular weight of each species. Since both monomer and

    dimer can be reduced by TCEP and used for PEGylation, we did not carry out final purification

    on size exclusion chromatography as used for other RITs.

    The PEGylation reaction was based on modifying a free thiol group of a surface-exposed

    cysteine to form a thioester bond with the maleimide functional group of a 20kD linear PEG

    (Figure 3B). To determine the best conditions for conjugation, we examined different conditions

    and found that the reaction was most efficient in PBS at pH 8.0. TCEP has been shown to

    enhance PEGylation efficiency compared to the standard reducing agents such as dithiothreitol

    and β-mercaptoethanol, used in our previous studies (9). Since TCEP does not contain a free

    thiol, there is no need to remove it prior to PEGylation, and the yields of reduced protein are

    much higher than using traditional reducing reagents. A 10-fold molar excess of methoxy PEG-

    maleimide was added to the TCEP-treated protein for PEGylation overnight. We performed

    HPLC analysis of the reaction mixture to determine the PEGylation efficiency; a typical HPLC

    run is shown for LMB-203 and its PEGylated counterpart LMB-203-PEG in Figure 3C. We

    quantified the peaks and found that the PEGylation was ~90 percent complete. The unreacted

    methoxy-PEG-maleimide was removed on an anion exchange column and the PEGylated protein

    was eluted with PBS.

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 13

    Eluted proteins were analyzed on the SDS-PAGE gel along with pre and post-TCEP

    treated samples (Figure 3D). For LMB-179 and LMB-244, the starting materials were mostly

    dimer (lanes 2 and 5). For LMB-203 and LMB-249, there is a mixture of monomer and dimer

    (lanes 8 and 11). LMB-163 was all monomer (lane 14) perhaps because the SH group in is not

    readily available for dimer formation. After reduction with TCEP, all became monomers (lanes

    3, 6, 9, 12 and 15). After addition of PEG, 90% or more of the TCEP-treated monomer was

    PEGylated (lanes 4, 7, 10, 13 and 16). The calculated molecular weight of a PEGylated RIT is

    about 72kD; however, they all run at ~100kD indicating the molecules are asymmetrical. The

    PEGylated proteins were quantified using Image J. The PEGylation efficiencies were ~90%

    overall, with LMB-203/LMB-203-PEG being 93-95%, which is consistent with the HPLC

    analysis. The PEG-modified proteins were frozen at -70ºC in small aliquots and thawed as

    needed for assays.

    About 10% of each RIT could not be modified even when we added more methoxy-PEG-

    maleimide and incubated for an extended period of time. We also tried removing TCEP before

    the addition of methoxy-PEG-maleimide, but this did not improve the yield. We assume the

    cysteine is modified during protein purification so that it cannot be reduced to generate a free

    thiol by TCEP.

    Cytotoxicity Assays

    To determine whether PEGylation hampers the activity of the RITs, we performed

    cytotoxicity assays on three mesothelin-expressing cancer cell lines. LMB-12 and LMB-84 have

    nearly identical cytotoxic activity (Figure S1A); we picked LMB-12 as un-PEGylated control

    since it has been widely studied. PEGylated and control RITs were incubated with cancer cells

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 14

    for three days, and WST assays were performed. The results are shown in Figure 4A. The cell-

    killing data is best fit to a sigmoidal curve. In general, the range of linearity in which the cell-

    killing activity is dependent on the concentration of RIT and occurs between 0.1-1ng/ml. Using

    KLM1 cells, all of the cysteine containing proteins are as active as the LMB-12 except for LMB-

    244 which is somewhat more active (Table 1). For KLM1 (pancreatic) cell line, parental RITs

    LMB-249, LMB-203, LMB-163 and LMB-179 have IC50 of 0.22 ± 0.06, 0.29 ± 0.02, 0.22 ±

    0.03, and 0.21 ± 0.01ng/ml, respectively. LMB-244 has an IC50 of 0.16 ± 0.03 ng/ml. The PEG

    modified immunotoxins were 4-5-fold less active except for LMB-244-PEG, which lost only

    35% activity. After PEGylation the IC50s for LMB-249-PEG, LMB-203-PEG, LMB-163-PEG

    and LMB-179-PEG were 1.18 ± 0.03, 1.10 ± 0.19, 1.13 ± 0.19, and 1.09 ± 0.13 ng/ml, and

    LMB-244-PEG was 0.25 ± 0.05 ng/ml. We also tested activities of LMB-244-PEG and LMB-

    163-PEG on A431/H9 epidermoid carcinoma cells and MKN28 stomach cancer cells. Consistent

    with KLM1 cell line, LMB-244 was the most active and lost less than 50% of its activity. To

    show that PEG doesn’t affects mesothelin-specific cell killing, we tested a few of PEG-modified

    RITs on the A431 epidermoid carcinoma cell line that was not transfected with mesothelin

    cDNA. The result in Figure S1B shows no specific killing.

    Figure 4 B-D shows bar graphs of the activity of PEG-modified RITs relative to the

    parental control, which is set to 100%. It is evident that LMB-244-PEG is the most active. In

    three tumor cell lines, KLM1 (Figure 4B), A431/H9 (Figure 4C), and MKN28 (Figure 4D),

    LMB-244-PEG retains 65, 62 and 70% of the parental activity.

    Mesothelin Binding and ADP Ribosylation are not affected by PEG

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 15

    The first step in the action of RIT is binding of the mesothelin on the cell surface of the

    cancer cell by the Fv. To test if binding is affected by the PEG, we used serial dilutions of each

    PEGylated RIT and tested the binding to mesothelin using ELISA. The results are shown in

    Figure S2 and 50% maximum binding is indicated. The binding of the PEGylated RITs is close

    to that of un-PEGylated LMB-12. The values are: LMB-12 (0.022 ± 0.001g/ml), LMB-249-

    PEG (0.027 ± 0.001g/ml), LMB-203-PEG (0.024 ± 0.004g/ml), LMB-163-PEG (0.029 ±

    0.006g/ml), LMB-179-PEG (0.023 ± 0.002g/ml) and LMB-244-PEG (0.013 ± 0.001g/ml).

    Another crucial step in RIT action is the ADP-ribosylation and inactivation of EF2. This

    step can be measured by incubating EF2-containing cell lysate with the ADP precursor NAD-

    biotin and PEG-modified RIT and measuring the amount of biotin incorporated into EF2 using

    western blots and Strep-HRP antibody. Figure S3 shows an ADP ribosylation assay, in which the

    PEGylated RIT were incubated with the EF2-containing cell lysate. The resulting blots show a

    RIT-dependent modification of EF2, with only one major band that is consistent with the

    molecular weight of EF2 (indicated by arrow). The band is not detected in the negative control

    where RIT was omitted (lane 16). We quantified the EF2-ADP bands and normalized each to

    LMB-12 positive control. For accuracy, we loaded 5, 10 and 15 l of each reaction. Interestingly,

    all RITs retain ~100% of the original activity. This indicates the loss of cytotoxic activity is not

    due to the inability of the immunotoxin to inactivate EF2.

    Half-life Measurements

    To determine the half-lives of the PEGylated RITs in the circulation, we injected 25 g

    of each protein IV into nu/nu mice and collected serum at 5-min and 2, 4, 8 and 24 hours for

    LMB-203-PEG, LMB-244-PEG, and LMB-249-PEG, and at 5-min and 1, 4 and 24 hours for

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 16

    LMB-163-PEG and LMB-179-PEG. ELISA was used to determine the amount of remaining

    RIT. To normalize the data, it is plotted relative to the 5-min time point, which is set as 100%

    (Figure 5A). LMB-12, which has no cysteine added and is not PEGylated was used as an un-

    PEGylated control. The decay for LMB-12 is rapid and mono-exponential. The curves for the

    PEG-modified proteins best fit a two-phase decay function; therefore, we calculated two half-life

    values for each protein, a fast (⍺) and a slow (β) decay (Table 1). We also calculated Area Under

    Curve (AUC) to represent the amount in the blood present over time so that we have a single

    value to compare for each protein. Figure 5A shows that all PEGylated proteins have much

    longer half-lives than LMB-12. The AUC for LMB-12 is 0.2, while the PEGylated proteins have

    AUCs that are at least 10-fold higher and range from 2.2 for LMB-244-PEG to 5.9 for LMB-

    249-PEG. LMB-203-PEG has the second largest AUC of 5.1. LMB-163-PEG and LMB-179-

    PEG have AUCs of 4.3 and 3.9. Based on the AUC values, the order from the longest to the

    shortest half-life is LMB-249-PEG > LMB-203-PEG > LMB-163-PEG > LMB-179-PEG >

    LMB-244-PEG. The difference between LMB-244-PEG and LMB-249-PEG is about 3-fold

    even though the mass of each protein is identical.

    The PEGylated proteins have much longer β than ⍺ decay. LMB-249-PEG has the

    longest half-life with an ⍺ of 144-min and a β of 74400-min. LMB-163-PEG has an ⍺ of 29-min

    and a β of 62200-min. LMB-244-PEG has the shortest half-life, with an ⍺ of 25-min and a β of

    256-min. In summary, PEGylation extends the half-life of RITs significantly, particularly the β

    phase. Because protein removal from the blood is controlled by glomerular filtration with

    subsequent degradation in the kidney, we postulated that the proteins could have different

    hydrodynamic radii that affected their rate of filtration by the glomerulus in the kidney.

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 17

    Hydrodynamic Radius

    To determine if the addition of PEG at different locations changed the hydrodynamic

    radius, we employed the Dynamic Light Scattering. Each sample was measured at least two

    times and representative measurements are shown in Figure S4 and summarized in Table 1. The

    %Mass is plotted against the radius. Overall, the results show a monomodal size distribution for

    LMB-163-PEG, LMB-244-PEG, LMB-12 and LMB-203. For LMB-249-PEG, LMB-203-PEG,

    and LMB-179-PEG, we observed polymodal size distributions, which is due to the presence of

    trace amount of aggregate that also diffract laser light. LMB-12 and LMB-203 were included as

    un-PEGylated controls. LMB-12 has the smallest Rh of 3.4nm. LMB-203 is slightly larger, with

    Rh of 3.7nm (Figure S4). With addition of 20kD PEG, the Rhs increase substantially to 7.1, 6.9,

    5.9, 5.6, and 5.2nm for LMB-249-PEG, LMB203-PEG, LMB-179-PEG, LMB-163-PEG, and

    LMB-244-PEG, respectively. Comparing LMB-203 and LMB-203-PEG, the increase in Rh is

    almost 2-fold.

    Biodistribution Assay

    It is widely-stated that PEG can extend the serum half-life by increasing the

    hydrodynamic volume of biologics and reducing their renal filtration (5). To determine how

    quickly the PEGylated RITs were removed by the kidney and liver, we performed biodistribution

    studies with LMB-249-PEG labelled with FNIR-Z-759, a near-infrared fluorophore that allows

    external imaging of living mice (21). Labeled LMB-249-PEG (41g, 580pmol) was injected I.V

    into tumor-bearing mice and accumulation in the kidney, liver, and tumor was imaged over 12

    hours. The images in the upper panel of Figure S5A are from a dorsal view, showing uptake by

    the kidneys and by tumor that are close to the dorsal side of the mouse. The images in the lower

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 18

    panel of Figure S5A show uptake by liver which is close to the ventral side. To compare the

    biodistribution with an immunotoxin without PEG, we performed the same experiment with

    SS1P, which has a MW of 63kD, close to 72kD for the PEG modified RITs. The upper panel of

    Figure S5B shows accumulation of SS1P in kidney and tumor and the lower panel shows liver

    uptake.

    Quantification of Uptake

    To determine the amount of LMB-249-PEG and SS1P taken up by kidney, liver, and

    tumor, the images were scanned and quantified. Accumulation as a function of time for kidney is

    plotted in Figure 5 C, for liver in Figure 5D, and for tumor in Figure 5E. The graphs show that

    uptake of LMB-249-PEG in kidney peaks at 6 hours whereas uptake of SS1P peaks much earlier

    at 2 hours. Liver uptake of LMB-249-PEG peaks late at 9 hours, while SS1P uptake much earlier

    at 2 hours. The results for tumor follow a different pattern. Accumulation of LMB-249-PEG by

    tumor increases steadily over many hours and peaks at 9-12 hours, whereas uptake of SS1P is

    much smaller and peaks at 2-3 hours.

    Anti-tumor experiments

    To do an initial assessment of the anti-tumor activity of the various PEGylated RITs, we

    treated tumor-bearing mice IV with 10g of PEG-modified RIT given every other day x3, when

    the tumors reached about ~100 mm3 in size. In the PBS treatment group, the tumors grew

    rapidly (Figure 6A). All treatment groups responded to PEGylated RITs. LMB-244-PEG and

    LMB-163-PEG were the most active and the tumors decreased in size to 69 mm3 and 79 mm

    3 on

    day 14 and the p-values are both 0.001, respectively. LMB-179-PEG, LMB-203-PEG and LMB-

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 19

    249-PEG slowed tumor growth; on day 14, the tumors reached 259 mm3 in the control group and

    only 152, 117 and 143 mm3 in LMB-179-PEG, LMB-203-PEG and LMB-249-PEG,

    respectively. The weights of the treated mice decreased by less than 10% (Figure S6A), but two

    mice in the LMB-179-PEG group and one in the LMB-249-PEG died.

    Since LMB-244-PEG and LMB-163-PEG are the most active, we performed additional

    studies with them using a dose and schedule that did not cause weight loss. We compared their

    activity with that of LMB-12, which is very similar to LMB-84 in structure and activity and is

    not PEG modified. Figure 6B shows that tumors in mice receiving five 20g doses given over 2

    weeks of PEG modified RIT regressed to 46mm3 and 48mm

    3, whereas tumors in mice treated

    with LMB-12 increased in size but grew more slowly than tumors treated with PBS. The p-

    values for LMB-244-PEG and LMB-163-PEG compared to LMB-12 are 0.0049 and 0.0048,

    respectively. At this schedule the weight of the mice decreased by less than 10% and no mice

    died (Figure S6B). This data establishes that increasing half-life greatly increases anti-tumor

    activity.

    Discussion

    We have developed a method to perform site specific PEGylation at cysteine residues

    present at different locations in an anti-mesothelin RIT and identified 2 locations that produced

    PEG modified RITs with high cytotoxic and anti-tumor activity and very prolonged half-lives in

    the circulation. Our strategy utilizes structural information to identify PEGylation sites that are

    least likely to interfere with binding to mesothelin or to EF2 or to affect furin cleavage. Since

    there are many lysine residues in the RIT, we could not achieve specificity using lysine-specific

    PEGylation and chose the highly efficient site-specific PEGylation of cysteine. Although there

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 20

    are two cysteine residues in Fv, they are buried and not available for reduction. Therefore, we

    used site-directed mutagenesis to introduce cysteines at locations exposed and distant from

    known functional sites in the RIT. We were able to PEGylate these mutant RITs at high

    efficiency. The key to this success is the use of TCEP to reduce the protein followed by treating

    with a 20 kDa maleimide derivative of PEG at pH 8.0. This that had not been achievable with β-

    mercaptoethanol in our previous study (8).

    The most active of the PEG modified RITs is LMB-244-PEG, which has a PEG attached

    to residue 406C in domain III of the toxin (Figure 1A). We tested LMB-244-PEG on 3 cell lines

    and found it retained more than 50% of its cytotoxic activity on all lines. When tested in mice, it

    had an 11-fold increase in residence time (AUC) in the circulation compared with an un-

    PEGylated protein LMB-12 and this change resulted in a very large increase in anti-tumor

    activity (Figure 6). To account for differences in activity, we measured its radius of hydration

    and found that LMB-244-PEG had the smallest Rh. This finding suggests that one or more steps

    in immunotoxin action are negatively affected by larger Rh of the protein and a smaller Rh is

    better. We also observed that LMB-244-PEG has a shorter half-life and AUC than the other

    PEGylated RITs indicating it is more efficiently filtered by the kidney glomerulus than RITs

    with higher Rh values.

    Because we used the same 20 kDa PEG to modify all the RITs, we were surprised to

    observe that the Rh values varied widely from 5.2 to 7.1 indicating that the derivatized proteins

    adopted different conformations. Since the Rh values correlate with the half-life in the

    circulation (Figure 5B), and since kidney is the major organ responsible for removal of RITs, we

    assume that the glomerulus in the kidney is sensitive to the size and shape of the proteins and

    filters them at different rates. In addition, the PEG-modified proteins with the smallest Rh

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 21

    (LMB-244-PEG) had excellent anti-tumor activity, suggesting that entry into tumors is also

    affected by Rh. We plan to use external tumor imaging to analyze the entry into tumors of all 5

    PEGylated RITs in a future study.

    The mechanism by which RITs kill cells is complex, beginning with binding to

    mesothelin on the cell surface and ending with inactivation of EF2 in the cytosol. We are unable

    to assess each step in the pathway but did assess two crucial steps and found that binding to

    mesothelin was similar for all PEG modified RITs, although there was a small loss of binding to

    mesothelin by the two that had PEG added to the Fv (Figure S2). We also found no change in the

    ability of the modified proteins to ADP-ribosylate EF2 (Figure S3). We are currently unable to

    measure how efficiently RITs are processed by other steps in the pathway.

    The half-life data of PEGylated RITs fits well to biexponential decay curves (Figure 5A).

    We think it is likely that the ⍺ phase is due to rapid equilibration with the extracellular

    compartment, and the β phase is due to the metabolism by the kidney and liver (22). We used

    external imaging to determine which organs in mice take up SS1P, a small un-PEGylated RIT

    with a half-life of 19 min (AUC 0.2) and LMB-249-PEG with a long half-life and an AUC of

    5.9. The data in Figure 5C and 5E show that SS1P is rapidly taken up by kidney and by liver,

    whereas the uptake of LMB-249-PEG is delayed. This delay leads to the greatly increased half-

    life and AUC. The initial uptake by tumors is the same for both RITs, but the uptake of SS1P is

    arrested as blood levels fall (Figure 5D), whereas the uptake of LMB-249-PEG increases for

    many hours and must be responsible for the enhanced anti-tumor activity of PEGylated RITs.

    It is widely-stated that PEG increases the half-life by increasing the hydrodynamic

    volume, and it has been shown increasing the size of PEG attached to mmTrail increases serum

    half-life (23). Because a change in PEG size does not account for the different biological

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 22

    properties of our proteins, we employed dynamic light scattering to assess the hydrodynamic

    volume of the proteins and found for the first time in our knowledge that the hydrodynamic size

    of protein can be fine-tuned depending on the position of the PEG. In Figure 5B, the Rh values

    are plotted against AUC. The result shows a good correlation between AUC and Rh with the R2

    value of 0.8. In general, larger Rh corresponds to longer half-life. The un-PEGylated LMB-12 is

    smallest protein and has a Rh of ~3.4. LMB-203 is slightly larger (RH 3.7) having a 15 amino

    acid linker connecting the light and heavy chains and 17 amino acid linker connecting the Fv to

    the furin cleavage site (Figure S4). The PEGylated proteins are much larger, with average Rhs of

    7.1, 6.9, 5.6, 5.9, 5.2 nm for LMB-249-PEG, LMB-203-PEG, LMB-163-PEG, LMB-179-PEG,

    and LMB-244-PEG, respectively. Therefore, our results indicate that the PEGylated RITs adopt

    distinct conformations governed by the position of the PEG, and such conformation influences

    the half-life. Since Rh is proportional to the hydrodynamic volume, we also concluded that PEG

    increases the hydrodynamic volume, which leads to slower renal filtration and longer half-life,

    and this is consistent with the reported properties of PEG (5).

    The anti-tumor activity of the PEGylated-RITs varied widely. Two of them, LMB-244-

    PEG and LMB-163-PEG, produced substantial tumor regressions, but the others were less active

    (Figure 6A). The high anti-tumor activity of LMB-244-PEG reflects several factors. The starting

    protein, LMB-244, is almost 2-fold more active than the other proteins and PEG addition caused

    less than a 2-fold loss of activity. Also, LMB-164-PEG has the smallest Rh indicating it is more

    compact and probably enters tumors better than the other PEG modified proteins. However, the

    small Rh probably contributes to its low AUC of 2.2. LMB-163-PEG also has good tumor

    activity despite being 3-fold less cytotoxic than LMB-264-PEG. The basis of the variable anti-

    tumor activity is not understood and will be the subject of further studies.

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 23

    Conclusion

    We have developed a new approach to carry out site specific modification of RITs with

    PEG and identified two RITs that have a long half-life and high anti-tumor activity. We have

    shown that the location of the PEG affects the hydrodynamic size of the protein and contributes

    to anti-tumor activity. PEG modified RITs merit further preclinical development for cancer

    therapy.

    Acknowledgements

    Molecular graphics images were produced using the UCSF Chimera package from the Resource

    for Biocomputing, Visualization, and Informatics at the University of California, San Francisco

    (supported by NIH P41 RR-01081). We thank the Biophysics Resource in the Structural

    Biophysics Laboratory, Center for Cancer Research, NCI at Frederick for assistance with light-

    scattering studies. This research was supported by the Intramural Research Program of the NIH,

    the National Cancer Institute, and the Center for Cancer Research (I.P.).

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 24

    References

    1. Kontermann RE. Strategies for extended serum half-life of protein therapeutics. Curr

    Opin Biotechnol 2011;22:868-76

    2. Pastan I, Hassan R, Fitzgerald DJ, Kreitman RJ. Immunotoxin therapy of cancer. Nat Rev

    Cancer 2006;6:559-65

    3. Alewine C, Hassan R, Pastan I. Advances in anticancer immunotoxin therapy. Oncologist

    2015;20:176-85

    4. Wei J, Bera TK, Liu XF, Zhou Q, Onda M, Ho M, et al. Recombinant immunotoxins

    with albumin-binding domains have long half-lives and high antitumor activity. Proc Natl

    Acad Sci U S A 2018;115:E3501-E8

    5. Damodaran V, J. Fee C. Protein PEGylation: An overview of chemistry and process

    considerations. 2010. 18-26 p.

    6. Swierczewska M, Lee KC, Lee S. What is the future of PEGylated therapies? Expert

    Opinion on Emerging Drugs 2015;20:531-6

    7. Wang QC, Pai LH, Debinski W, FitzGerald DJ, Pastan I. Polyethylene glycol-modified

    chimeric toxin composed of transforming growth factor alpha and Pseudomonas

    exotoxin. Cancer Res 1993;53:4588-94

    8. Tsutsumi Y, Onda M, Nagata S, Lee B, Kreitman RJ, Pastan I. Site-specific chemical

    modification with polyethylene glycol of recombinant immunotoxin anti-Tac(Fv)-PE38

    (LMB-2) improves antitumor activity and reduces animal toxicity and immunogenicity.

    Proc Natl Acad Sci U S A 2000;97:8548-53

    9. Humphreys DP, Heywood SP, Henry A, Ait-Lhadj L, Antoniw P, Palframan R, et al.

    Alternative antibody Fab' fragment PEGylation strategies: combination of strong

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 25

    reducing agents, disruption of the interchain disulphide bond and disulphide engineering.

    Protein Eng Des Sel 2007;20:227-34

    10. Pastan I, Beers R, Bera TK. Recombinant immunotoxins in the treatment of cancer.

    Methods Mol Biol 2004;248:503-18

    11. Mazor R, Eberle JA, Hu X, Vassall AN, Onda M, Beers R, et al. Recombinant

    immunotoxin for cancer treatment with low immunogenicity by identification and

    silencing of human T-cell epitopes. Proc Natl Acad Sci U S A 2014;111:8571-6

    12. Hollevoet K, Mason-Osann E, Liu XF, Imhof-Jung S, Niederfellner G, Pastan I. In vitro

    and in vivo activity of the low-immunogenic antimesothelin immunotoxin RG7787 in

    pancreatic cancer. Mol Cancer Ther 2014;13:2040-9

    13. Liu XF, Zhou Q, Hassan R, Pastan I. Panbinostat decreases cFLIP and enhances killing

    of cancer cells by immunotoxin LMB-100 by stimulating the extrinsic apoptotic pathway.

    Oncotarget 2017;8:87307-16

    14. Ho M, Hassan R, Zhang J, Wang QC, Onda M, Bera T, et al. Humoral immune response

    to mesothelin in mesothelioma and ovarian cancer patients. Clin Cancer Res

    2005;11:3814-20

    15. Chowdhury PS, Pastan I. Improving antibody affinity by mimicking somatic

    hypermutation in vitro. Nat Biotechnol 1999;17:568-72

    16. Ma J, Tang WK, Esser L, Pastan I, Xia D. Recognition of mesothelin by the therapeutic

    antibody MORAb-009: structural and mechanistic insights. J Biol Chem

    2012;287:33123-31

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 26

    17. Celikel R, Peterson EC, Owens SM, Varughese KI. Crystal structures of a therapeutic

    single chain antibody in complex with two drugs of abuse-Methamphetamine and 3,4-

    methylenedioxymethamphetamine. Protein Sci 2009;18:2336-45

    18. Jorgensen R, Merrill AR, Yates SP, Marquez VE, Schwan AL, Boesen T, et al. Exotoxin

    A-eEF2 complex structure indicates ADP ribosylation by ribosome mimicry. Nature

    2005;436:979-84

    19. Li M, Dyda F, Benhar I, Pastan I, Davies DR. The crystal structure of Pseudomonas

    aeruginosa exotoxin domain III with nicotinamide and AMP: conformational differences

    with the intact exotoxin. Proc Natl Acad Sci U S A 1995;92:9308-12

    20. Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, et al.

    UCSF Chimera--a visualization system for exploratory research and analysis. J Comput

    Chem 2004;25:1605-12

    21. Sato K, Gorka AP, Nagaya T, Michie MS, Nani RR, Nakamura Y, et al. Role of

    Fluorophore Charge on the In Vivo Optical Imaging Properties of Near-Infrared Cyanine

    Dye/Monoclonal Antibody Conjugates. Bioconjug Chem 2016;27:404-13

    22. Shancer Z, Liu XF, Nagata S, Zhou Q, Bera TK, Pastan I. Anti-BCMA immunotoxins

    produce durable complete remissions in two mouse myeloma models. Proc Natl Acad Sci

    U S A 2019

    23. Nie Q, Jia D, Yang H, Feng Y, Fan Q, Shi Q, et al. Conjugation to 10 kDa Linear PEG

    Extends Serum Half-Life and Preserves the Receptor-Binding Ability of mmTRAIL with

    Minimal Stimulation of PEG-Specific Antibodies. Mol Pharm 2017;14:502-12

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 27

    Table 1. Summary of properties of RITs

    RIT names

    IC50 (ng/ml) in

    KLM 1

    Half Max

    Mesothelin

    Binding Half Life (min) AUC Rh

    without PEG

    with PEG (%

    Activity) (g/ml) Fast () Slow ()

    (arbitrary unit,

    x104)

    (nm)

    SS1P 0.19 n/a nt 19 n/a nt nt

    LMB-12 0.22 n/a 0.022 0.001 11 n/a 0.2 3.4

    LMB-84 0.19 n/a nt nt n/a nt nt

    LMB-249 0.22 0.06

    1.18

    0.03(18%) 0.027 0.001 144 74400 5.9 7.1

    LMB-203 0.29 0.02

    1.10 0.19

    (26%) 0.024 0.004 59 51600 5.1 6.9

    LMB-163 0.22 0.03

    1.13 0.19

    (19%) 0.029 0.006 29 62284 4.3 5.6

    LMB-179 0.21 0.01

    1.09 0.13

    (19%) 0.023 0.002 38 26600 3.9 5.9

    LMB-244 0.16 0.03

    0.25 0.05

    (64%) 0.013 0.001 25 256 2.2 5.2

    nt: not tested; n/a: not applicable

    Bolded numbers refer to PEGylated RITs

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 28

    Figure Legends

    Figure 1. Schematics of anti-mesothelin RITs. A. SS1P contains an anti-mesothelin disulfide

    linked Fv connected to PE38 (domain II and III of PE). LMB-12 is derived from SS1P with

    domain II deleted and replaced with an 11 amino acid furin cleavge peptide connecting the Fv to

    domain III. LMB-84 contains a single-chain Fv instead of a dsFv and is the parental RIT from

    which the cysteine containing PEG-modified proteins were prepared. LMB-179, LMB-244, and

    LMB-203 have an additional 17-GS linker that separates Vl from the furin cleavage site. LMB-

    249 has GS linker placed at the N-terminus of Vh. LMB-163 contains mutations that humanize

    the Fv portion. For each construct, the engineered cysteine for site-specific PEGylation is

    indicated by a bolded red C. B. Cartoon representations of RITs shown in A. PEGs on cysteines

    are represented by red lines. Domains are not drawn to scale.

    Figure 2. Ribbon diagrams of anti-mesothelin RITs. A. The ribbon diagram was generated

    with UCSF Chimera. Various domains are colored as: N terminal linker = grey, Vh = blue, Vl =

    cyan, CDR = orange, furin site = green, and PE24 = yellow. Sites that were mutated to cysteine

    are shown as red spheres, and they were chosen to avoid interfering with functional binding to

    mesothelin, furin protease, and EF2. B. A structural model of the complex of the RIT in A bound

    to mesothelin (olive green) and EF2 (dark gray). C. A hypothetical 20kD linear PEG composed

    of 454 repeating units of ethylene glycol is connected to the cysteine at D406 of PE24 (LMB-

    244). The structure of PEG is random, and the overall size of the molecule is modeled to

    approximate the average radius of hydration measured for all five PEGylated RITs.

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 29

    Figure 3. PEGylation of RITs. A. RITs eluted from Mono Q anion exchange column. Fractions

    A, B, and C correspond to separate peaks in the Mono Q chromatogram and were pooled

    separately. B. Schematic showing a hypothetical site-specific PEGylation reaction, which occurs

    by the formation of a thioester bond between a free thiol of a surface-exposed cysteine and a

    maleimide functional group of a PEG. C. Representative HPLC analysis of LMB-203/LMB-203-

    PEG. D. Non-reducing SDS-PAGE gel analysis of parental, TCEP-reduced, and PEGylated

    RITs. Gels were visualized by Coomassie Blue staining. The efficiencies of PEGylation were

    quantified by counting the number of pixels using Image J. PEGylated RIT is indicated by the

    blue square. White lines indicate the point of merge between two separate gels.

    Figure 4. WST assays of parental and PEGylated RITs. A. Representative cytotoxicity assays

    after 3 days incubation of mesothelin-positive cancer cell lines with different RITs. KLM1,

    pancreatic cancer cell line; A431/H9, epidermoid carcinoma cell line transfected with mesothelin

    cDNA; MKN28, gastric cancer cell line. B-D. The IC50 values of PEGylated RITs were

    normalized against the un-PEGylated controls which were set to 100%. The percent remaining

    activity is indicated.

    Figure 5. Biological properties of RITs. A. Pharmacokinetics of PEGylated RITs. 25g of

    LMB-203-PEG, LMB-244-PEG, LMB-163-PEG, LMB-179-PEG, and LMB-249-PEG were

    injected into nu/nu mice and blood was collected at 5 min, 1 (LMB-163-PEG and LMB-179-

    PEG), 2, 4, 8 (LMB-203-PEG, LMB-244-PEG, and LMB-249-PEG), and 24 hour time points.

    ELISA was performed to determine the level of RIT and plotted in two-phase decay using

    GraphPad. Amount of RIT at 5 min was set to 100% and used to normalize the remaining time

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • 30

    points. LMB-12 was data from our previous study and plotted as one-phase decay. The AUC

    values are summarized. B. Rh of PEGylated protein was measured using Dynamic Light

    Scattering. 20l aliquots of proteins (0.5-1mg/ml) were measured at least two times and Rh was

    plotted against half-life using GraphPad. The same color scheme was used as in A. The line of

    best fit was generated with R2 value indicated. C-E. Biodistribution of LMB-249-PEG and SS1P

    in mice. Fluorescently labeled LMB-249-PEG and SS1P was I.V. injected into A431/H9 tumor-

    bearing mice (n=3, tumor ~100mm3), respectively. Serial dorsal and ventral 800nm fluorescence

    images were obtained with focus placed on the tumor, kidney, and liver. The fluorescence

    intensity at each time point was quantified and plotted to show the accumulation in the

    kidney(C), tumor(D) and liver(E). Plots were generated from the average results of 3 imaged

    mice.

    Figure 6. Anti-tumor activity of parental and PEGylated RITs. A. Antitumor activity of

    PEGylated RITs in nude mice (n = 10). Mice were implanted with mesothelin-expressing KLM1

    tumor cells. When tumors reached ~100mm3, mice were I.V. injected with 10g/mouse

    PEGylated RITs qod x3 as indicated by arrows. Tumor burden was monitored over 2 weeks. B.

    Comparison of anti-tumor activities of LMB-12, LMB-163-PEG and LMB-244-PEG. Groups of

    10 mice were treated with 20 g of each agent as shown by arrows and tumor burden was

    monitored over 2 weeks.

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/

  • Published OnlineFirst December 23, 2019.Mol Cancer Ther Zeliang Zheng, Ryuhei Okada, Hisataka Kobayashi, et al. immunotoxins increases half-life and anti-tumor activitySite-specific PEGylation of anti-mesothelin recombinant

    Updated version

    10.1158/1535-7163.MCT-19-0890doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://mct.aacrjournals.org/content/suppl/2019/12/21/1535-7163.MCT-19-0890.DC1

    Access the most recent supplemental material at:

    Manuscript

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

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

    Subscriptions

    Reprints and

    [email protected] at

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

    Permissions

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

    .http://mct.aacrjournals.org/content/early/2019/12/21/1535-7163.MCT-19-0890To request permission to re-use all or part of this article, use this link

    on April 4, 2021. © 2019 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 23, 2019; DOI: 10.1158/1535-7163.MCT-19-0890

    http://mct.aacrjournals.org/lookup/doi/10.1158/1535-7163.MCT-19-0890http://mct.aacrjournals.org/content/suppl/2019/12/21/1535-7163.MCT-19-0890.DC1http://mct.aacrjournals.org/cgi/alertsmailto:[email protected]://mct.aacrjournals.org/content/early/2019/12/21/1535-7163.MCT-19-0890http://mct.aacrjournals.org/

    Article FileFigure 1Figure 2Figure 3Figure 4Figure 5Figure 6