Protein Engineering, Design & Selection, 2016, vol. 29 no. 5, pp. 159–167 doi: 10.1093/protein/gzw002 Advance Access Publication Date: 10 March 2016 Original Article Original Article ASP2408 and ASP2409, novel CTLA4-Ig variants with CD86-selective ligand binding activity and improved immunosuppressive potency, created by directed evolution Shinsuke Oshima1,*, Erik E. Karrer2,*, Madan M. Paidhungat2, Margaret Neighbors3, Steven J. Chapin3, Rong A. Fan3, Margaret A. Reed3, Kuoting Wu3, Clifford Wong3, Yonghong Chen5, Marc Whitlow4, Francisco A. Anderson2, Rujuta A. Bam2, Qian Zhang2, Brent R. Larsen5, Sridhar Viswanathan6, Bruce H. Devens3, Steven H. Bass2, and Yasuyuki Higashi1 1 Pharmacology Research Laboratories, Drug Discovery Research, Astellas Pharma, Inc., Tsukuba, Ibaraki 3058585, Japan, 2Department of Molecular Biology, Perseid Therapeutics, Redwood City, CA 94063, USA, 3Department of Biology and Pharmacology, Perseid Therapeutics, Redwood City, CA 94063, USA, 4Colabrativ, Inc., Orinda, CA 94563, USA, 5Department of Protein Analytics and Pharmaceutics, Perseid Therapeutics, Redwood City, CA 94063, USA, and 6Department of Process Development and Manufacturing, Perseid Therapeutics, Redwood City, CA 94063, USA *To whom correspondence should be addressed. E-mail: [email protected] (S.O.), [email protected] (E.E.K.) Edited by Frances Arnold Received 15 September 2015; Revised 19 January 2016; Accepted 20 January 2016 Abstract The CTLA4-Ig therapeutics abatacept and belatacept inhibit CD28-mediated T cell activation by binding CD80 (B7-1) and CD86 (B7-2) co-stimulatory ligands. Both compounds preferentially bind CD80, yet CD86 has been implicated as the dominant co-stimulatory ligand. Using directed evolution methods, novel CTLA4-Ig variants were created with selective CD86 binding affinity, a property that confers increased immunosuppressive potency and potentially improved efficacy and safety profiles. Relative to abatacept (wild-type CTLA4-Ig), ASP2408 and ASP2409 have 83-fold and 220-fold enhanced binding affinity to CD86 while retaining 1.5-fold and 5.6-fold enhanced binding affinity to CD80, respectively. Improvements in CD86 binding affinity correlates with increased immunosuppressive potency in vitro and in vivo. Our results highlight the power of directed evolution methods to obtain non-intuitive protein engineering solutions and represent the first examples of CD86-selective CTLA4-Ig compounds that have entered clinical trials. Key words: costimulation blockade, next-generation protein therapeutics, phage display, rheumatoid arthritis, transplantation © The Author 2016. Published by Oxford University Press. All rights reserved. For Permissions, please e-mail: [email protected] 159 S.Oshima et al. 160 Introduction Adaptive immune responses are tightly regulated to balance immune activation with homeostasis (Bour-Jordan et al., 2011). During T cell activation, positive co-stimulatory signaling mediated by engagement of CD28 receptors on T cells with CD80/CD86 ligands on antigen-presenting cells (APC) is counterbalanced by down-regulatory signaling via engagement of CTLA-4 receptors on T cells with the same ligands (Salomon and Bluestone, 2001; Walker and Sansom, 2011). This CD80/CD86–CD28/ CTLA-4 regulatory network represents substantial opportunities for therapeutic intervention, either for immunosuppressive approaches blocking positive signaling through CD28 or immunostimulatory approaches blocking down-regulatory functions of CTLA-4. To date, there are three marketed biologics targeting this network: (a) abatacept (http:// www.pdr.net/drug-information/orencia?druglabelid=112), a wild-type CTLA4-Ig fusion protein for rheumatoid arthritis (Keating, 2013); (b) belatacept (http://www.pdr.net/drug-information/nulojix?druglabelid=110), a modified CTLA4-Ig with improved CD80/CD86 ligand binding affinity for kidney transplantation (Wojciechowski and Vincenti, 2012); and (c) ipilimumab, an anti-CTLA-4 monoclonal antibody for melanoma (Trinh and Hwu, 2012). The primary mechanism of action for CTLA4-Ig therapeutics is binding to CD80/CD86 and thus blocking co-stimulatory signaling through CD28 (Carman et al., 2009). Although both CD80 and CD86 can engage CD28, structural (Stamper et al., 2001), biophysical (Pentcheva-Hoang et al., 2004; Bhatia et al., 2005) and theoretical modeling (Jansson et al., 2005) observations implicate CD86 as the dominant co-stimulatory ligand for signaling through CD28. Cellbased assays in vitro confirm that immunosuppressive potency of CTLA4-Ig variants correlates more with binding affinity to CD86 than CD80 (Latek et al., 2009; Xu et al., 2012). As both abatacept and belatacept are selective for CD80 binding (Larsen et al., 2005; Bernett et al., 2013), we employed DNA shuffling directed evolution methods to create CTLA4-Ig variants with specific improvements in CD86 binding affinity. Here, we provide the first description of CD86-selective CTLA4-Igs with potential for improved clinical effectiveness. Materials and methods Library creation Libraries of CTLA-4 ECD variants were created by synthetic DNA shuffling (Ness et al., 2002) where overlapping oligonucleotides (60-mers) encoding the desired diversity are assembled into full-length genes by polymerase chain reaction (PCR) amplification using flanking primers and cloned via SfiI/NotI into phage display vector pSB0124. pSB0124 is a derivative of pBR322 (New England Biolabs, #N3033S) encoding CTLA-4 ECD-phage M13 pIII fusion proteins targeted for periplasmic secretion by the STII signal peptide (Picken et al., 1983) and tightly regulated expression driven by the pBAD promoter (Guzman et al., 1995) to facilitate monovalent phage display. Library ligations in pSB0124 were transformed into TOP10 Escherichia coli cells (Invitrogen, Inc., #C4040-50) by electroporation, and the cells were cultured in Luria Broth (LB) media containing 50 µg/ml carbenicillin to make stocks of library DNA. Libraries of phage-displayed CTLA-4-ECD proteins were created by transforming of E. coli TG-1 cells (Stratagene, #200123) with library DNA by electroporation. Transformed cells were cultured in LB medium, infected with helper phage M13KO7 (Invitrogen, #18311-019) at a multiplicity of infection of 5–10 under dual selection for phagemid (carbenicillin at 50 µg/ml) and helper phage (kanamycin at 70 µg/ml). Procedure in detail is described in the Supplementary data. Library screening Phage libraries were panned for five rounds against soluble biotinylated ligands (Round 1) or immobilized ligands (Rounds 2–5). In the first round, library phage were incubated in capture reactions containing biotinylated hCD80-Ig or hCD86-Ig. Ligand-bound phage were captured by streptavidin-coated Dynal beads (Life Technologies, #11205D). Bound phage were eluted by incubation with elution buffer (0.2 M glycine pH 2.2 + 1% BSA) and neutralized by neutralization buffer (1 M Tris pH 9.0). Escherichia coli TG-1 cells were infected with eluted phage to generate enriched library phage for the next round of selection. In Rounds 2–5, library phage were panned against ligand immobilized on immunotubes (NUNC Maxi-Sorp, Thermo Scientific, #470319) coated with CD80-Ig or CD86-Ig at 10 µg/ml. Phage enriched from panning were characterized by phage ELISA. Phage were captured by coated CD80-Ig or CD86-Ig on the plate and detected by HRP-conjugated anti-M13 monoclonal antibody (GE Healthcare, #27-9421-01). Phage displaying variant CTLA-4 ECDs with improved ligand binding affinity were identified by further incubating with soluble competitor protein (0.3 µM abatacept for CD80-Ig-coated plates or 1 µM abatacept for CD86-Ig-coated plates). Differences in the ratio of ELISA signal in the presence or absence of competitor protein were used to identify CTLA-4 variants with higher affinity, as such variants would have the least reduction in ELISA signal after extended washing in the presence of competitor protein. Procedure in detail is described in the Supplementary data. Subcloning CTLA-4 variants into the IgG2-Fc fusion expression vector The IgG2-Fc fusion expression vector is a derivative of pCDNA3.1(+) (Life Technologies, #V79020) engineered to express a fusion protein comprising the human CTLA-4 protein from positions M1 to D161, including the signal sequence (Harper et al., 1991) and the human IgG2-Fc domain (from positions E216 to K447 using the EU numbering system). Unique AgeI and ClaI sites were introduced through silent mutations at I31–P32–V33 and I115–D116 positions in the CTLA-4-ECD-encoding region to facilitate cloning of PCR-amplified variants into the vector. Production and purification of variant CTLA4-Ig fusion proteins COS-7 cells (ATCC, #CRL-165) were transiently transfected with plasmid DNA encoding variant CTLA4-Ig fusion proteins using FuGENE 6 transfection reagent (Roche, #11 815 075 001), as per the manufacturer’s recommended conditions. Following transfection, cells were cultured with DMEM medium (Invitrogen, #11964-142) containing 10% fetal bovine serum (FBS) (Hyclone, #SV30014.03) and 1 × PSG (penicillin–streptomycin–glycine, Invitrogen, #10378-016) at 37°C and 5% CO2 for 4 days. Variant CTLA4-Ig fusion proteins were purified by Protein A affinity chromatography. SDS/PAGE analysis was used to assess identity and purity of variant CTLA4-Ig protein preparations. Size exclusion chromatography (SEC) was used to measure the content of aggregates in variant CTLA4-Ig protein preparations. Typical protein preparations exhibited >90% monomeric peak area, where the monomeric species is a disulfide-linked homodimer comprised of two CTLA4-Ig polypeptides. Reagent and control proteins Abatacept, the first-generation CTLA4-Ig comprising a fusion protein of wild-type CTLA-4 with modified Fc domain of human IgG1 (Linsley et al., 1991), was purchased from Bristol-Myers Squibb (Princeton, NJ). Belatacept, a next-generation CTLA4-Ig protein Directed evolution of CD86-selective CTLA4-Ig differing from abatacept by two mutations (A29Y and L104E) in the CTLA-4 ECD (Larsen et al., 2005), was produced at Perseid Therapeutics (Redwood, CA). As surrogate control proteins for binding assay, wild-type CTLA-4 or modified CTLA-4 with the two amino acids mutations fused with Fc domain of human IgG2 was produced at Perseid Therapeutics. CD80-mIg or CD86-mIg protein was produced at Perseid Therapeutics. 161 Astellas Pharma, Inc. (Tsukuba, Japan) that are awarded accreditation status by the AAALAC International and performed in accordance with the ethics criteria contained in the bylaws of the committee. All the other experimental procedures performed on animals were approved by the IACUC of Perseid Therapeutics and conducted in accordance with the current edition of the Guide for the Care and Use of Laboratory Animals as standardly used by PHS/OLAW-assured and AAALAC-accredited institutions. Biacore measurements and analysis Rabbit anti-mouse IgG antibody (GE Healthcare, #BR-1005-14) was immobilized on CM-5 sensor chips (GE Healthcare, #BR-1000-14) according to the manufacturer’s protocol. Human or murine CD80-murine Ig (CD80-Ig) or CD86-murine Ig (CD86-Ig) proteins were bound to antibody-coated sensor chips. CTLA4-Ig proteins were diluted in HBS-EP buffer (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.005% v/v surfactant P20, GE Healthcare, #BR1001-88) and flowed over ligand-coated sensor chips for 2 min, followed by 5- or 20-min incubation with HBS-EP buffer containing no protein at the same flow rate, according to the purpose of analysis. For more accurate comparison of mutant CTLA4-Ig fusion proteins having very slow dissociation rates from ligands, kinetic assays were conducted using 20 min dissociation times. No discernible dissociation of captured ligands was observed during the dissociation periods (data not shown). Eight dilutions of CTLA4-Ig proteins ranging from 500 to 0.2 nM were analyzed against a blank reference (HBS-EP buffer alone). Ligand was removed from the chip surface between cycles by incubation with 10 mM glycine, pH 1.7. Data were analyzed by BIAevaluation software (v4.1, available from GE Healthcare) using the ‘Fit kinetics, Simultaneous ka/kd’ function. Data were fit using the 1:1 Langmuir model to determine the association rate constant (ka) and the dissociation rate constant (kd) and to calculate the equilibrium dissociation constant, KD. Sequence optimization Protein sequences of CTLA-4 ECD variants were aligned using VectorNTI (Invitrogen) and GeneDoc (http://www.nrbsc.org/gfx/ genedoc) software and ordered by relative CD86 binding affinity. Individual mutations were weighted according to their frequency among variants with high CD86 binding affinity relative to their frequency in the unselected library. Mutations were further weighted by examining potential changes to the protein structure using available NMR and X-ray crystal structures (Metzler et al., 1997; Schwartz et al., 2001; Stamper et al., 2001) viewed using Cn3Dv4.1 (NCBI) and Swiss-PdbViewer (ExPASy). Weighting information was used to determine candidate amino acid positions for reversion to wild-type sequence. Sequence-optimized clones were constructed by de novo assembly of overlapping oligonucleotides (60-mers with 30 nucleotide overlap) encoding the full-length variant CTLA-4-ECD and cloned into the IgG2-Fc fusion expression vector as described herein. Homology modeling The homology model was constructed from the Protein Data Bank coordinate set 1I8L of CTLA-4 bound to CD86 (Schwartz et al., 2001) using Accelrys Discovery Studio Visualizer 2 to determine optimal fitting of variant amino acid side chain rotamers within their local environments. Animals The experimental procedures on in vitro mice studies were approved by the Institutional Animal Care and Use Committee (IACUC) of T-cell proliferation assays Human blood samples were obtained healthy volunteers (Palo Alto Medical Foundation, CA) under IRB consent. For human T-cell recall antigen-specific proliferation assay, peripheral blood mononuclear cells (PBMC) were freshly isolated from the donors prescreened for reactivity to Mycobacterium tuberculosis purified protein derivative (PPD) using a Ficoll (Sigma-Aldrich, Histopaque, #10771) gradient separation as per the manufacturer’s recommended protocol. PBMC were suspended in RPMI 1640 medium containing 10% FBS and 1% penicillin/streptomycin (Gibco, #15140), and 1 × 105cells/well were added to cell culture plates with PPD recall antigen (5 µg/ml, Mycos, #P-1000-001). Baseline control wells contained PBMC only without PPD antigen. For human mixed lymphocyte reactions (MLR) assay, human PBMC were prepared as above. From one donor (responder), 1 × 105 PBMC were mixed with irradiated (26 Gy) 1 × 105 PBMC from a second donor (stimulator) in 96-well round bottom culture plates. Baseline control wells contained responder PBMC only. For murine MLR assay, splenocytes of C57BL/6 (Charles River Labs, Japan) and Balb/c (Charles River Labs, Japan) were isolated by standard technique. Splenocytes of C57BL/6 were prepared in RPMI 1640 medium supplemented with 2 mM L-glutamine, 100 U/ml penicillin, 100 µg/ml streptomycin (Sigma-Aldrich, #P0781), 55 µM 2-mercaptoethanol (Gibco, #21985-023) and 10% FBS, and 1.5 × 105 of splenocytes from C57BL/6 (responder) were mixed with irradiated (20 Gy) 1.5 × 105 of splenocytes obtained from BALB/c (stimulator) in 96-well round bottom culture plates. Baseline control wells contained responder splenocytes only. For all T-cell response assays, test compounds were serially diluted in the same medium and added to the wells. Culture plates were incubated for 2–5 days at 37°C and 5% CO2 with 1 µCi/well 3H thymidine (GE Healthcare, Inc. or Moravek Biochemicals, Inc.) added to each well for an additional 6–18 h. The cells were harvested with a cell harvester (Perkin Elmer, Waltham, MA) onto glass filter membranes (Perkin Elmer). Cell proliferation was measured by incorporation of radiolabel counted by a scintillation counter (Perkin Elmer) and reported as counts per minute (CPM). Assay data were analyzed by Prism 5 (GraphPad Software, Inc., La Jolla, CA) with non-linear regression curve fitting using the sigmoidal dose–response (variable slope) equation and least squares fit method with the top and bottom parameters set to share the values from all the data sets. Geometric mean IC50 for each test compound was generated from at least three independent experiments. In vivo mouse antibody response with KLH immunization Female BALB/c mice at the age of 8–12 weeks (Charles River Labs, Hollister, CA) were immunized on day 0 by subcutaneous (SC) injection with 200 µg of Keyhole Limpet Hemocyanin (KLH) (Sigma-Aldrich, #H7017) at the base of the tail. To induce secondary antibody response, mice were immunized with SC injection of 200 µg of KLH at the base of 162 S.Oshima et al. the tail after resting for at least 8 weeks from primary KLH immunization. Mice were treated with test compounds via SC at the doses indicated in parallel with the primary or secondary KLH immunization. Mice were bled to detect anti-KLH antibody at 21 days after primary immunization or at 14 days after secondary immunization, respectively. Anti-KLH IgG in serum was measured by ELISA (Life Diagnostics, #4010-1). Results Directed evolution strategy and library screening The iterative directed evolution process employed consisted of multiple CTLA-4 variant libraries over three rounds of screening, in which separate libraries were created to explore different types, frequency and location of amino acid diversity within the CTLA-4 protein structure, while keeping library sizes within the limits of phage display screening throughput (∼1 × 109) (Supplementary Tables SI and SII). Naturally occurring diversity was obtained from analysis of orthologous CTLA-4 protein sequences from different mammalian species (Supplementary Fig. S1), while random diversity was created by incorporation of NNS degenerate codons (Supplementary Table SI). Library 1 (Supplementary Table SI) interrogated orthologous diversity at CTLA-4 residues that were predicted to be at or near the protein–protein interface based on structural studies (Schwartz et al., 2001). As many of those residues mapped to regions of CTLA-4 homologous to the complementarity-determining regions (CDR1 and CDR3) of the variable-immunoglobulin domain, libraries from Series 2 (Supplementary Table SI) interrogated randomized diversity in each of the variable-CDR-like loops of CTLA-4 (CRD1, CDR2, CDR3 and C’‘D loop, Supplementary Table SI). Series 3 libraries were built to interrogate combinations of the most favored diversity from previous libraries (1 and 2) with orthologous diversity in the ‘framework’-like regions of CTLA-4 variable-immunoglobulin-like domain. Libraries were screened by monovalent phage display panning (Bass et al., 1990) against human CD86-Ig or CD80-Ig fusion proteins. Enriched phage was confirmed for ligand binding activity by phage ELISA and for identity by DNA sequencing. Individual variants were expressed as CTLA4-IgG2-Fc fusion proteins in transiently transfected COS cells, and the binding kinetics of variant CTLA4-Ig fusion proteins to immobilized CD80-Ig or CD86-Ig were evaluated using a surface plasmon resonance (Biacore) assay with a 5 min dissociation phase. Amino acid changes and improvements in ligand binding affinity relative to wild-type CTLA4-Ig (abatacept) for variants selected from each round of library screening are shown in Fig. 1, Supplementary Figures S2 and S3. Different profiles were observed for variants selected from separate libraries, in which variants selected from Library 3 have a wide range of different ligand binding selectivity (Fig. 1), with some variants preferentially improved for CD86 binding (such as 201685). Sequence optimization The CTLA4-Ig variants obtained from library screening have ∼20 amino acid changes per clone relative to the wild-type CTLA-4 sequence. In order to minimize potential immunogenicity risks associated with sequence divergence from wild type, a sequence optimization process was adopted with the aim of reducing mutation load without compromising function. A large set of variants spanning a large binding activity range (subset shown in Supplementary Fig. S2) was analyzed using sequence-based (Fox et al., 2007) and structural analyses (data not shown) to establish correlations between mutant residues and Fig. 1 Ligand binding activity of CTLA4-Ig variant proteins selected from phage screening of Library 3. Binding kinetics of CTLA-Ig variant proteins to immobilized CD80-Ig and CD86-Ig proteins were measured using a surface plasmon resonance (Biacore) assay with a 5 min dissociation phase. Binding affinity is expressed as fold improvement in KD (equilibrium dissociation constant) relative to wild-type CTLA4-Ig (abatacept) for binding to CD86 (black bars) or CD80 (gray bars). Data are representative of two independent measurements. binding activity. Potentially neutral mutations that correlated poorly with binding activity were candidates for reversion during sequence optimization, while those that correlated strongly with binding activity were preserved in the initial step. Clones 201685, 201731 and 201682 were selected as parent frameworks for this approach, as they have high CD86 binding affinity and considerable sequence diversity represented among their 19–21 mutations (Supplementary Fig. S4). Potentially neutral mutations in the parent clones were reverted to wild type using site-directed mutagenesis. A total of four descendent clones (D1–D4) were expressed as soluble CTLA4-IgG2 fusion proteins and characterized for CD86 binding affinity by Biacore (Supplementary Fig. S4 and Table SIII). Of the four descendants, Clone D3 retained the highest level of CD86 binding affinity relative to its parent (0.86-fold reduction), indicating that the seven reversions were likely neutral in the context of the Clone 201731 framework. This sequence optimization process reduced the mutation load of Clone 201731 from 19 to 12 amino acid changes without significant loss of CD86 ligand binding activity. In order to further optimize ligand binding activity relative to mutation load, variants of Clone D3 (the D3 series) were generated with different mutation loads by reversion and re-assortment of the 12 amino acid changes (Supplementary Fig. S5). The D3 series variants were expressed as soluble CTLA4-IgG2 fusion proteins and characterized for CD86/CD80 binding affinity by Biacore (Supplementary Fig. S6). ASP2408 was identified as a variant with the highest CD86 binding selectivity with the lowest mutation load, while ASP2409 was identified as a variant with the highest CD86 binding activity with reduced mutation load relative to Clone D3 (Fig. 2a and b). Directed evolution of CD86-selective CTLA4-Ig 163 Fig. 2 Binding affinity on CD86, selectivity on ligands and mutation load of D3-related variants (the D3 series). (a) Relationship of fold improvement in the CD86 binding affinity of variants relative to abatacept vs mutation load. (b) Relationship of the CD86 binding selectivity of variants vs mutation load. Table I. Ligand binding activity of ASP2408, ASP2409 and control proteins to immobilized human CD80-Ig and CD86-Ig CTLA4-Ig CD86_human CD80_human ka (1/Ms) Abatacept Belatacept ASP2408 ASP2409 kd (1/s) KD (M) −2 6 1.08 × 10 9.52 × 10−4 2.17 × 10−4 8.23 × 10−5 1.62 × 10 2.25 × 106 2.69 × 106 2.74 × 106 Fold −9 6.66 × 10 4.23 × 10−10 8.05 × 10−11 3.00 × 10−11 1 16 83 220 Selectivity ka (1/Ms) kd (1/s) 0.054 0.083 3.0 2.1 KD (M) −4 6 1.32 × 10 1.79 × 106 1.33 × 106 1.27 × 106 4.75 × 10 6.26 × 10−5 3.20 × 10−4 8.12 × 10−5 −10 3.61 × 10 3.50 × 10−11 2.41 × 10−10 6.42 × 10−11 Fold Selectivity 1 10 1.5 5.6 18 12 0.33 0.47 Binding kinetics were measured using Biacore assay with a 5 or a 20 min dissociation phase. ka represents the association rate constant, kd represents the dissociation rate constant and KD represents the equilibrium dissociation constant. Values are expressed as the mean of 2 or more independent measurements. Fold indicates the fold difference in KD relative to abatacept. Selectivity is expressed as the ratio of KD for each ligand. Binding kinetics for belatacept, ASP2408 and ASP2409 were obtained using a 20 min dissociation time, while those for abatacept were obtained using a 5 min dissociation time. Table II. Ligand binding activity of ASP2408, ASP2409 and control proteins to immobilized murine CD80-Ig and CD86-Ig CTLA4-Ig CD86_murine ka (1/Ms) Abatacept-IgG2 Belatacept-IgG2 ASP2408 ASP2409 5 8.23 × 10 6.16 × 105 1.41 × 106 1.13 × 106 CD80_murine kd (1/s) KD (M) −4 5.63 × 10 2.19 × 10−3 6.65 × 10−5 1.46 × 10−4 Fold −10 6.84 × 10 3.55 × 10−9 4.70 × 10−11 1.29 × 10−10 1 0.19 15 5.3 Selectivity 0.17 N.D. 1.1 2.5 ka (1/Ms) 5 9.81 × 10 N.D. 1.44 × 106 9.50 × 105 kd (1/s) KD (M) −4 1.16 × 10 7.92 × 10−4 7.77 × 10−5 3.02 × 10−4 −10 1.19 × 10 N.D. 5.40 × 10−11 3.18 × 10−10 Fold Selectivity 1 N.D. 2.2 0.37 5.7 N.D. 0.87 0.41 Abatacept-IgG2 and belatacept-IgG2, which consist of the same CTLA-4 sequence as abatacept and belatacept, respectively, fused with Fc domain of human IgG2, were assessed as a surrogate protein. Binding kinetics were measured using Biacore assay with a 5 or a 20 min dissociation phase, and the values are indicated as in Table I. N.D., not determined. Ligand binding activity of ASP2408 and ASP2409 Binding activity for human ligands of ASP2408 and ASP2409 is shown in Table I. ASP2408 and ASP2409 have binding affinity to human CD86 with KD of 80.5 and 30.0 pM, respectively, which represent approximately 83-fold and 220-fold improvements in binding affinity relative to abatacept, respectively. The CD80 binding affinity of ASP2408 is similar to that of abatacept (KD of 241 pM for ASP2408 vs 361 pM for abatacept), whereas ASP2409 has 5.6-fold higher affinity (KD of 64.2 pM for ASP2409 vs 361 pM for abatacept). ASP2408 and ASP2409 are selective for CD86 binding relative to abatacept, as the affinity of ASP2408 for CD86 is 3.0-fold higher than that for CD80 (80.5 vs 241 pM) and the affinity of ASP2409 for CD86 is 2.1-fold higher than that for CD80 (30.0 vs 64.2 pM). This selectivity profile is opposite to that observed for abatacept and belatacept, which are selective for CD80 binding (18-fold and 12-fold, respectively). We also examined the binding affinity of ASP2408 and ASP2409 with murine CD80 and CD86 (Table II). ASP2408 and ASP2409 also showed improved binding affinity for murine CD86 relative to a surrogate protein of abatacept. In contrast to the binding activity for human CD86, ASP2408 showed more potent affinity to murine CD86 relative to ASP2409 (47.0 vs 129 pM, respectively). The potential for off-target binding of ASP2408 and ASP2409 was assessed by immunohistochemical staining of a wide range of normal tissues from human, cynomolgus monkey, rat and mouse, with no unexpected staining observed (data not shown). Modeling of ASP2408 and ASP2409 amino acid changes to the CTLA-4 protein structure There are 6 and 10 amino acid changes present in the ECD of ASP2408 and ASP2409, respectively, relative to wild type (Fig. 3a). The location 164 S.Oshima et al. Fig. 3 Homology model of ASP2408 and ASP2409 in complex with CD86. (a) Sequence alignment of ASP2408, ASP2409 and wild-type CTLA4-IgG2. Numbering is relative to the start of the mature CTLA-4 ECD (Peach et al., 1994), identity to the wild-type CTLA4-IgG2 is represented by dots and differences are indicated by single letter amino acid code. (b and c) Views of the homology model of ASP2408 and ASP2409 in complex with CD86 to illustrate the location of mutations relative to the ligand binding interface. The solvent accessible surface of CD86 has been color according to its electrostatic potential; blue and red areas having positive and negative potentials at their surface, respectively. The alpha carbon (Ca) trace of ASP2408 and ASP2409 is shown in light blue. The Ca trace of the highly conserved ligand binding motif (M97YPPPY102) is highlighted in green. The carbon atoms of mutations are color coded gold for hydrophobic residue (V32I, A50M, S64P, S70F), blue for positively charged residues (M54K), red for negatively charged residues (A24E, G55E, N56D, L104E) and purple for hydrophilic non-charged residues (T30N, I65S). (d) An expanded view of CTLA4/CD86 binding interface region, with the CTLA-4 residues known to directly participate in CD86 binding (E31 R33, T51, M97YPPPYY103) are shown in green (Schwartz et al., 2001). The carbon atoms of hydrophobic V32I, A50M, S70F and negative-charged L104E side chains are shown in gold, purple, yellow and red, respectively, and proximal CTLA-4 hydrophobic side chains of residues Y52, L58, F60, V75, V94 and L96 are shown in light blue. of each mutation within the protein structure was visualized by creating homology models of ASP2408 and ASP2409 in complex with CD86 (Fig. 3b and c). The amino acid changes in ASP2408 and ASP2409 are positioned throughout the CTLA-4 domain, with some changes such as S64P well removed from the CD86 ligand binding surface. The remaining changes are positioned in proximity to the ligand binding interface, where they may participate more directly in enhancing ligand binding. Several of the proximal changes are hydrophobic in nature (A50M, S70F in both, V32I in ASP2409). An expanded view of these hydrophobic changes reveals that they are clustered in a hydrophobic region adjacent to the ligand binding loop M97YPPPY102, with their side chains orientated away from the ligand binding interface. In addition, L104E, which is a key mutation in belatacept (Larsen et al., 2005), is also present in ASP2409 and likely contributes directly to ligand binding. In vitro immunosuppressive activity of ASP2408 and ASP2409 The immunosuppressive activity of ASP2408 and ASP2409 was measured in vitro using several primary human and murine T-cell proliferation assays (Table III). Co-stimulatory signal is provided by endogenous APC within the PBMC population. In the M. tuberculosis PPD stimulation assay, modulation of memory T cells was assessed using PBMC harvested from donors prescreened for reactivity to PPD antigen and cultured in the presence of PPD to recall antigen-experienced T cells. CTLA4-Ig variants inhibited memory T-cell proliferation in a dose-dependent manner (Supplementary Fig. S7a). In human and murine MLR assays, modulation of allo-reactive T-cell was evaluated, where co-stimulatory signal was provided by endogenous APC within the allogeneic PBMC or splenocyte population. Allogeneic T-cell proliferation was also inhibited in a dose-dependent manner by the CTLA4-Ig variants (Supplementary Fig. S7b and c). Suppressive potency for each CTLA4-Ig variant was calculated as the geometric mean IC50 and is summarized in Table III. In all assay formats tested, the improved immunosuppressive potency of ASP2408, ASP2409 and belatacept relative to abatacept correlates with their improved CD86 binding affinity. Of note, ASP2408 was more potent than ASP2409 in inhibiting murine T-cell proliferation, consistent with the higher binding affinity of ASP2408 vs ASP2409 to the murine CD86 ligand. Directed evolution of CD86-selective CTLA4-Ig 165 Table III. IC50 values of CTLA4-Ig variants in each in vitro T-cell stimulation assay CTLA4-Ig PPD_human IC50, M (95% CI) Abatacept Belatacept ASP2408 ASP2409 −9 2.2 × 10 (7.2 × 10−10–6.6 × 10−9) 1.8 × 10−10 (9.3 × 10−11–3.3 × 10−10) 3.6 × 10−11 (2.1 × 10−11–6.0 × 10−11) 1.7 × 10−11 (6.3 × 10−12–4.6 × 10−11) MLR_human Fold 1 12 60 130 IC50, M (95% CI) −9 7.3 × 10 (3.3 × 10−9–1.6 × 10−8) 4.2 × 10−10 (1.3 × 10−10–1.3 × 10−9) 4.2 × 10−11 (1.4 × 10−11–1.3 × 10−10) 2.2 × 10−11 (6.9 × 10−12–7.1 × 10−11) MLR_murine Fold 1 18 170 330 IC50, M (95% CI) −9 Fold 8.6 × 10 (1.7 × 10−9–4.3 × 10−8) >1.3 × 10−7 1 4.5 × 10−10 (1.2 × 10−10–1.7 × 10−9) 2.5 × 10−9 (4.6 × 10−10–1.3 × 10−8) 19 N.D. 3.5 IC50 values were determined from concentration–response curves and are represented as the geometric mean with the 95% confidence interval (CI) from at least three experiments. N.D., not determined. Fig. 4 Immunosuppressive effects of ASP2408 and ASP2409 on primary and secondary anti-KLH antibody responses in mice. Female BALB/c mice were immunized with 200 µg KLH, and anti-KLH IgG antibody responses were measured as described in the Materials and Methods section. Test compounds were administered subcutaneously at the doses indicated on the same day of primary or secondary KLH immunization (n = 3–8/group). Anti-KLH antibody responses were measured at 21 days after primary immunization with KLH and treatment with (a) ASP2408 or (b) ASP2409. Anti-KLH antibody responses were measured at 14 days after secondary KLH immunization and treatment with (c) ASP2408 and (d) ASP2409. The hashed line indicates the arithmetic mean value of the group. The error bars show the standard error of the mean. Statistical significance was analyzed by using Dunnett’s multiple comparison test. *P < 0.05 when compared with primary or secondary KLH immunized animals untreated with test compounds. In vivo immunosuppressive activity of ASP2408 and ASP2409 T-cell-dependent B cell differentiation requires co-stimulatory signal, as demonstrated by inhibition of antigen-specific antibodies when co-stimulation is blocked using CTLA4-Ig (Linsley et al., 1992). To test the in vivo immunosuppressive effects of ASP2408 and ASP2409, we investigated murine primary and secondary antibody responses induced by KLH immunization. As shown in Fig. 4, ASP2408 and ASP2409 with a single SC dose at the time of the last KLH immunization inhibited the level of anti-KLH antibody generation in primary and secondary antibody responses in a dose-dependent fashion. In primary anti-KLH antibody response, significant suppression was observed by ASP2408 at the 0.4 mg/kg dose (Fig. 4a), while a 2 mg/kg dose of ASP2409 (Fig. 4b) was required to achieve a significant effect. Complete suppression of primary anti-KLH antibody response was observed by ASP2408 treatment at 1 mg/kg (Fig. 4a), and ASP2409 was not able to achieve complete suppression at dose levels up to 2 mg/kg (Fig. 4b). In secondary anti-KLH antibody response, 166 higher dose levels of ASP2408 or ASP2409 were required to observe suppressive effects relative to the primary response. For ASP2408, although a significant suppressive effect was observed at the 0.5 mg/kg dose, complete suppression was not observed at dose levels up to 10 mg/kg (Fig. 4c). For ASP2409, a significant suppressive effect was observed at the 5 mg/kg dose and complete suppression was not observed at dose levels up to 20 mg/kg (Fig. 4d). These findings indicate that in vivo immunosuppressive effect of ASP2408 was more potent relative to ASP2409 in mice, which was consistent with in vitro immunosuppressive effect using murine splenocyte and with affinity to the murine CD80 and CD86 ligands (shown in Tables II and III and Supplementary Fig. S7). Discussion We employed a directed evolution approach known as MolecularBreeding™ or DNA shuffling (Stemmer, 1994; Minshull and Stemmer, 1999) to improve the ligand binding properties of a class of protein therapeutics approved for rheumatoid arthritis and kidney transplantation indications. DNA shuffling methods harness the power of iterative genetic recombination and have proved versatility in dissecting protein structure–function relationships (Schürmann et al., 2013) and improving diverse protein properties such as cytokine activity (Chang et al., 1999), enzymatic activity (Fox et al., 2007), thermal stability (Ruller et al., 2008), ligand binding affinity (Luginbühl et al., 2006), expression level and solubility (Keenan et al., 2005). Here we used synthetic DNA shuffling (Ness et al., 2002) to improve CD86 binding affinity and generate the first examples of CD86-selective CTLA4-Ig proteins that have entered into clinical trials. Multiple CTLA-4 variant libraries containing over 3 billion unique clones were screened iteratively over three rounds in order to effectively sample the large sequence space (Crameri et al., 1998) represented within natural CTLA-4 orthologs (∼6 × 1021 unique clones, Supplementary Fig. S1). In the initial rounds of screening (Libraries 1 and 2, Supplementary Table SI), natural or random diversity was explored in the region of the well-characterized CTLA-4 ligand binding interface (Peach et al., 1994; Metzler et al., 1997; Schwartz et al., 2001; Stamper et al., 2001). Comparing results from these different approaches (Supplementary Fig. S3a and b), libraries created from natural orthologous diversity yielded CTLA4-Ig variants with higher CD86 binding affinity and selectivity. This result is consistent with the observation that re-assortment of naturally occurring diversity can provide accelerated directed evolution relative to random mutagenesis (Crameri, et al., 1998). Although neither library generated variants with substantially improved activity relative to belatacept, beneficial mutations were selected that could be recombined in further library iterations. In later rounds of screening (Library 3, Supplementary Table SI), beneficial mutations from previous rounds were combined with natural orthologous diversity located throughout the CTLA-4 protein structure. Results from Library 3 screening underscore the power of the family shuffling approach, as dramatic improvements in CD86 binding affinity and selectivity were observed (Fig. 1). The majority of variants selected from Library 3 have CD86 affinity improved substantially beyond that of belatacept, with several clones displaying >5-fold higher affinity relative to belatacept and up to 50-fold higher affinity relative to abatacept. Improvement in CD86 binding affinity generally correlated with improved CD86 binding selectivity, with several clones showing strong (5- to 6-fold) CD86 binding selectivity profiles. A sequence optimization process was employed to reduce the mutation load of top variants selected from Library 3. Of the four reversion constructs created, only Clone D3 retained sufficient ligand S.Oshima et al. binding activity (Supplementary Table SIII), emphasizing the inherent uncertainty in predicting relative contributions of multiple amino acid changes within a 3D protein structure. Based on the amino acid changes in Clone D3, further sequence optimization was performed. Among these variants of Clone D3 (the D3 series), ASP2408 was identified as a variant with the highest CD86 binding selectivity with the lowest mutation load, while ASP2409 was identified as a variant with the highest CD86 binding activity with reduced mutation load relative to Clone D3 (Fig. 2a and b). Several amino acid mutations are common for ASP2408 and ASP2409, such as hydrophobic changes (S64P, S70F), positive charge changes (M54K) and neutral changes (T30N, A50M). Unique mutations are also present in ASP2408 (G55E: negative charge change) and in ASP2409 (A24E, N56D and L104E: negative charge changes; V32I and I65S: neutral changes), respectively (Fig. 3a). Homology modeling reveals several mutations V32I, A50M, S70F form a core hydrophobic patch below the ligand binding loop M97YPPPY102 (Fig. 3d). These mutations appear to interact with adjacent hydrophobic residues to increase the overall hydrophobicity of the region, which could act to stabilize a more productive binding conformation for the CD86 ligand relative to CD80. Further experiments would be required to test this hypothesis. Further characterization of ASP2408 and ASP2409 binding profile using Biacore conditions optimized for measuring slow dissociation rates reveals dramatic improvements in human and murine CD86 binding relative to abatacept (Table I and II). Importantly, the ligand binding selectivity profile for ASP2408 and ASP2409 (3.0-fold and 2.1-fold selective for CD86, respectively) has been completely reversed relative to abatacept and belatacept, which are 18-fold and 12-fold selective for CD80, respectively. Selectively improved CD86 binding affinity correlates with improved immunosuppressive potency as measured by in vitro T cell proliferation assays (Table III). Further, we found that the immune modulatory effect of CTLA4-Igs in mouse KLH immunization models was clearly consistent with improved binding affinity for CD86 (Fig. 4). These findings support and extend previous observations where immunosuppressive potency of CTLA4Ig variants correlates more with binding affinity to CD86 than CD80 (Vincenti, 2008; Latek et al., 2009; Xu et al., 2012). Structure-based rational design and high-throughput mutagenesis methods have been previously used to engineer CTLA4-Igs for improved CD80/CD86 ligand binding (Larsen et al., 2005; Xu et al., 2012; Bernett et al., 2013). Although these studies have provided fundamental advances in the understanding of the molecular interactions between CTLA-4 and its ligands, none have generated CTLA4-Igs with such pronounced and selective improvements in CD86 ligand binding affinity as represented by ASP2408 and ASP2409. The magnitude of improvement in CD86 binding from previous studies is in the range of 16-fold (Larsen et al., 2005), 17-fold (Xu et al., 2012) and 23-fold (Bernett et al., 2013) relative to abatacept, while ASP2408 and ASP2409 have substantial further improvements of 83-fold and 220-fold, respectively. Of note, ASP2408 and ASP2409 represent the first examples of CTLA4-Igs with binding selectivity for CD86, having 3.0-fold and 2.1-fold higher affinity for CD86 vs CD80, respectively. CTLA4-Igs from previous engineering efforts with the highest improvements in CD86 binding are still selective for CD80, having 12-fold (Larsen et al., 2005), 6-fold (Xu et al., 2012) and 2-fold (Bernett et al., 2013) higher CD80 binding affinity relative to CD86. It is anticipated that such attributes will enable ASP2408 and ASP2409 to have improved dosing convenience and potentially improved efficacy and safety profiles relative to previous generations of modified CTLA4-Igs. Improvements in CD86 binding affinity and selectivity are hypothesized to confer clinically meaningful advantages relative to Directed evolution of CD86-selective CTLA4-Ig current CTLA4-Ig therapies by several interrelated mechanisms. Because improved CD86 affinity correlates with improved immunosuppressive potency, CTLA4-Igs with improved CD86 binding affinity would be able to provide therapeutic levels of CD86 occupancy at substantially lower serum drug concentrations. Thus, increased convenience of drug administration could be realized by allowing lower dose levels and less frequent dosing intervals. This concept is reinforced by the observation that ASP2408 or ASP2409 requires between 60- and 360-fold lower protein concentration relative to abatacept to achieve similar inhibition of primary human T cell proliferation in vitro (Table III). In addition, increased affinity to CD86 could enable increased therapeutic efficacy by virtue of providing more complete saturation of the dominant co-stimulatory ligand CD86 throughout the dosing cycle. In a related set of hypotheses, CD86-selective CTLA4-Igs are anticipated to occupy substantially less CD80 ligand at therapeutic dose levels. Increased CD80 availability could provide increased efficacy by enhancing regulatory functions involved in the maintenance of immune tolerance. Because Tregs require CD28 signaling for development and survival (Tang et al., 2003), and CD80 can be a productive ligand for CD28 (Jansson et al., 2005), it follows that increased CD80 availability could enhance Treg cell generation via increased CD80–CD28 interactions. Treg function could also be improved by increased CD80 availability, as CTLA-4 is critical for Treg function (Walker and Sansom, 2011) and CD80 is the preferred ligand for CTLA-4 (Jansson et al., 2005). Additionally, increased CD80 availability could allow increased CD80-mediated immune functions known to be associated with antiviral responses (Lumsden et al., 2000), anti-B cell lymphoma (Suvas et al., 2002), antitumor responses in CNS (Ando et al., 2002) and maintenance of bone marrow plasma cells (Rozanski et al., 2011). Increased preservation of such immune responses may reduce immunosuppressive complications of standard CTLA4-Ig therapy such as infection and malignancy (Wojciechowski and Vincenti, 2012; Keating, 2013). In conclusion, we employed DNA shuffling directed evolution methods and created CTLA4-Ig variants, ASP2408 and ASP2409, with specific improvements in CD86 binding affinity. We provide the first description of CD86-selective CTLA4-Igs with potential for improved clinical effectiveness. Author contribution S.O. and E.K. equally contributed to make this manuscript. E.K. was project lead. S.O. led in vitro (murine cells) studies. M.P. led library design, screening and sequence optimization. M.N. led murine KLH studies. S.C. led Biacore analysis, R.F. led in vitro human PPD and MLR assays, M.R. performed Biacore measurements, and K.W. performed human PPD and MLR assays. C.W. performed murine KLH studies. Y.C. led protein production and characterization. M.W. created homology models of CTLA-4/CD86 interactions. F.A., R.B. and Q.Z. performed construct design, protein production and library design/screening. M.N., B.L., S.V., B.D., S.B. and Y.H. provided scientific leadership. Supplementary data Supplementary data are available at PEDS online. Acknowledgements We thank Anithakumari Dulapalli, Tasnim Kothambawala, Judy Gao, Edward Pascua and William Ngo for protein production and characterization. This paper and all the work associated with this manuscript are dedicated to the memory of our wonderful colleague, B.R.L., who recently passed away. 167 References Ando,H., Saio,M., Ohe,N., et al. (2002) Int J Oncol, 20, 807–812. Bass,S., Greene,R. and Wells,J.A. (1990) Proteins, 8, 309–314. Bernett,M.J., Chu,S.Y., Leung,I., et al. (2013) MAbs, 5, 384–396. Bhatia,S., Edidin,M., Almo,S.C. and Nathenson,S.G. (2005) Proc Natl Acad Sci USA, 102, 15569–15574. Bour-Jordan,H., Esensten,J.H., Martinez-Llordella,M., Penaranda,C., Stumpf, M. and Bluestone,J.A. (2011) Immunol Rev, 241, 180–205. Carman,J.A., Davis,P.M., Yang,W.P., Zhu,J., Chang,H., He,A., Truong,A., Suchard,S.J. and Nadler,S.G. (2009) J Clin Immunol, 29, 479–489. Chang,C.C., Chen,T.T., Cox,B.W., Dawes,G.N., Stemmer,W.P., Punnonen,J. and Patten,P.A. (1999) Nat Biotechnol, 17, 793–797. Crameri,A., Raillard,S.A., Bermudez,E. and Stemmer,W.P. (1998) Nature, 391, 288–291. Fox,R.J., Davis,S.C., Mundorff,E.C., et al. (2007) Nat Biotechnol, 25, 338–344. Guzman,L.M., Belin,D., Carson,M.J. and Beckwith,J. (1995) J Bacteriol, 177, 4121–4130. Harper,K., Balzano,C., Rouvier,E., Mattéi,M.G., Luciani,M.F. and Golstein,P. (1991) J Immunol, 147, 1037–1044. Jansson,A., Barnes,E., Klenerman,P., Harlén,M., Sørensen,P., Davis,S.J. and Nilsson,P. (2005) J Immunol, 175, 1575–1585. Keating,G.M. (2013) Drugs, 73, 1095–1119. Keenan,R.J., Siehl,D.L., Gorton,R. and Castle,L.A. (2005) Proc Natl Acad Sci USA, 102, 8887–8892. Larsen,C.P., Pearson,T.C., Adams,A.B., et al. (2005) Am J Transplant, 5, 443–453. Latek,R., Fleener,C., Lamian,V., Kulbokas,E., Davis,P.M., Suchard,S.J., Curran, M., Vincenti,F. and Townsend,R. (2009) Transplantation, 87, 926–933. Linsley,P.S., Brady,W., Urnes,M., Grosmaire,L.S., Damle,N.K. and Ledbetter,J. A. (1991) J Exp Med, 174, 561–569. Linsley,P.S., Wallace,P.M., Johnson,J., Gibson,M.G., Greene,J.L., Ledbetter,J. A., Singh,C. and Tepper,M.A. (1992) Science, 257, 792–795. Luginbühl,B., Kanyo,Z., Jones,R.M., Fletterick,R.J., Prusiner,S.B., Cohen,F.E., Williamson,R.A., Burton,D.R. and Plückthun,A. (2006) J Mol Biol, 363, 75–97. Lumsden,J.M., Roberts,J.M., Harris,N.L., Peach,R.J. and Ronchese,F. (2000) J Immunol, 164, 79–85. Metzler,W.J., Bajorath,J., Fenderson,W., et al. (1997) Nat Struct Biol, 4, 527–531. Minshull,J. and Stemmer,W.P. (1999) Curr Opin Chem Biol, 3, 284–290. Ness,J.E., Kim,S., Gottman,A., Pak,R., Krebber,A., Borchert,T.V., Govindarajan, S., Mundorff,E.C. and Minshull,J. (2002) Nat Biotechnol, 20, 1251–1255. Peach,R.J., Bajorath,J., Brady,W., Leytze,G., Greene,J., Naemura,J. and Linsley, P.S. (1994) J Exp Med, 180, 2049–2058. Pentcheva-Hoang,T., Egen,J.G., Wojnoonski,K. and Allison,J.P. (2004) Immunity, 21, 401–413. Picken,R.N., Mazaitis,A.J., Maas,W.K., Rey,M. and Heyneker,H. (1983) Infect Immun, 42, 269–275. Rozanski,C.H., Arens,R., Carlson,L.M., Nair,J., Boise,L.H., Chanan-Khan,A. A., Schoenberger,S.P. and Lee,K.P. (2011) J Exp Med, 208, 1435–1446. Ruller,R., Deliberto,L., Ferreira,T.L. and Ward,R.J. (2008) Proteins, 70, 1280–1293. Salomon,B. and Bluestone,J.A. (2001) Annu Rev Immunol, 19, 225–252. Schürmann,N., Trabuco,L.G., Bender,C., Russell,R.B. and Grimm,D. (2013) Nat Struct Mol Biol, 20, 818–826. Schwartz,J.C., Zhang,X., Fedorov,A.A., Nathenson,S.G. and Almo,S.C. (2001) Nature, 410, 604–608. Stamper,C.C., Zhang,Y., Tobin,J.F., et al. (2001) Nature, 410, 608–611. Stemmer,W.P. (1994) Proc Natl Acad Sci USA, 91, 10747–10751. Suvas,S., Singh,V., Sahdev,S., Vohra,H. and Agrewala,J.N. (2002) J Biol Chem, 277, 7766–7775. Tang,Q., Henriksen,K.J., Boden,E.K., Tooley,A.J., Ye,J., Subudhi,S.K., Zheng, X.X., Strom,T.B. and Bluestone,J.A. (2003) J Immunol, 171, 3348–3352. Trinh,V.A. and Hwu,W.J. (2012) Expert Opin Biol Ther, 12, 773–782. Vincenti,F. (2008) J Allergy Clin Immunol, 121, 299–306; quiz 307–298. Walker,L.S. and Sansom,D.M. (2011) Nat Rev Immunol, 11, 852–863. Wojciechowski,D. and Vincenti,F. (2012) Curr Opin Organ Transplant, 17, 640–647. Xu,Z., Juan,V., Ivanov,A., Ma,Z., Polakoff,D., Powers,D.B., Dubridge,R.B., Wilson,K. and Akamatsu,Y. (2012) J Immunol, 189, 4470–4477.
© Copyright 2025 Paperzz