• No results found

Molecular determinants of FVIII immunogenicity in hemophilia A - Chapter 3+: Reply to: Mannose-sensitive receptors mediate the uptake of factor VIII therapeutics by human dendritic cells

N/A
N/A
Protected

Academic year: 2021

Share "Molecular determinants of FVIII immunogenicity in hemophilia A - Chapter 3+: Reply to: Mannose-sensitive receptors mediate the uptake of factor VIII therapeutics by human dendritic cells"

Copied!
8
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

Molecular determinants of FVIII immunogenicity in hemophilia A

Wróblewska, A.

Publication date

2013

Link to publication

Citation for published version (APA):

Wróblewska, A. (2013). Molecular determinants of FVIII immunogenicity in hemophilia A.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)

3+

Reply to: Mannose-sensitive

receptors mediate the uptake of

factor VIII therapeutics by human

dendritic cells

Eszter Herczenik, Simon D. van Haren, Aleksandra Wroblewska, Paul Kaijen, Maartje van den Biggelaar, Alexander B. Meijer, Luisa Martinez-Pomares, Anja ten Brinke and

Jan Voorberg

E.H., S.H. & A.W. contributed equally to this work

(3)

72

Repl

y t

o letter

3+

Correspondence: Mannose-sensitive receptors mediate the

uptake of factor VIII therapeutics by human dendritic cells

Yohann Repessé, Suryasarathi Dasgupta, Ana-Maria Navarrete, Sandrine Delignat, Srinivas V. Kaveri, Sébastien Lacroix-Desmazes

To the Editor: Administration of factor VIII (FVIII) to manage bleedings in patients

with hemophilia A induces inhibitory anti-FVIII antibodies in a substantial number of patients. The initial step in the development of the naive anti-FVIII immune response is the uptake of the exogenously administered FVIII by dendritic cells (DCs), the professional subset of antigen-presenting cells. The interesting study by Herczenik et al 1 suggests the implication of a variety of receptors that mediate

the internalization of FVIII by DCs.

Here, we wish to emphasize the importance of mannose-bearing residues on different commercially available FVIII therapeutics in endocytosis through mannose-sensitive receptors by human DCs in vitro. We first found that kinetics (data not shown) and dose dependency of FVIII endocytosis by immature DCs were similar whether FVIII was directly labeled 2 or was detected in permeabilized

cells by a labeled antibody 1 (Figure 1A). We then compared the endocytosis of

different FVIII therapeutics by DCs: 2 recombinant full-length FVIII (Kogenate and Advate) produced in baby hamster kidney or Chine hamster ovary cells, respectively, a recombinant B domain–deleted FVIII (Refacto) produced in Chine hamster ovary cells, and a plasma-derived FVIII (Factane). Kogenate and Advate were endocytosed with identical kinetics and to a greater extent than Refacto and Factane (Figure 1B). The reduced endocytosis of FVIII in the case of the plasma-derived product in our experimental setup is probably due to the presence of von Willebrand factor, as reported earlier. 3 We then confirmed that

the endocytosis of the 4 FVIII products results in FVIII presentation to CD4+ T

lymphocytes. DCs were incubated with 5 IU/ml of FVIII (a concentration close to that reached in vivo following replacement therapy) in the presence of D9E9, a human FVIII C1 domain–specific T-cell line. 4 All tested FVIII therapeutics induced

the secretion of IFN-g by D9E9 (Figure 1C), demonstrating similar endocytic and intracellular processing of the different products. Furthermore, the incubation of the 4 FVIII therapeutics with DCs in the presence of mannan was accompanied by a reduction in the amount of endocytosed FVIII (reduction of 56% ± 15%, 48% ± 24%, 39% ± 19%, and 76% ± 1% for Kogenate, Advate, Refacto, and Factane, respectively; Figure 1D). The reduction of endocytosis of both Kogenate and Refacto by mannan had been shown to reduce D9E9 activation. 2 Our results

indicate that the contribution of mannose-ending sugars to the endocytosis of FVIII by DCs does not differ significantly depending on the plasmatic or recombinant origin of FVIII, or on the presence or absence of the B domain in the recombinant molecule. Interestingly, mannose-sensitive receptors expressed by human monocyte–derived immature DCs 5 are also expressed by splenic and

circulating myeloid DCs (data not shown). In the work by Herczenik et al. 1 the

(4)

Repl

y t

o letter

3+

immunogenicity of FVIII in vivo. It is not clear whether this effect was due to the quenching of phosphatidylserine–binding moieties of FVIII and due to an ensuing reduction of FVIII aggregation, 6 due to steric hindrance following the binding of

the IgG to FVIII, or due to the blocking of a specific, yet unidentified, receptor for FVIII. Together with our observations, the data suggest that the generation of a novel FVIII therapeutic compromised in its capability to interact with DCs upon the removal of selected N-glycosylation sites may be a novel strategy to produce a hemostatically proficient drug with potentially reduced immunogenicity in patients with hemophilia A.

Figure 1. Endocytosis of different commercially available FVIII products by human DCs implicates mannose-senstive receptors. A. Dose-dependent labeling of DCs following incubation with FVIII. Human monocyte–

derived immature DCs were incubated in X-VIVO 15 with FVIII or FITC-labeled FVIII (Kogenate) for 120 minutes at 4 and 37°C, respectively. 2 In the case of nonlabeled FVIII, cells were permeabilized by using 0.1% saponin and FVIII was detected by using the monoclonal anti-FVIII IgG, mAb 77IP52H7 (10 μg/mL), coupled to fluorescein isothiocyanate (FITC). The percentage of labeling at 4°C was subtracted from that measured at 37°C to quantify FVIII endocytosis. B. Dose-dependent endocytosis of therapeutic FVIII concentrates by DCs. C. Activation of the FVIII-specific human CD4+ T-cell clone D9E9. DCs were generated from healthy blood donors with the MHC II haplotype DRB1*1501/DRB5*01, which matches D9E9. DCs were incubated with D9E9 in the medium alone or with 5 IU/ml of different FVIII preparations, for 20 hours at 37°C. The activation of D9E9 was assessed by measuring IFN-g in culture supernatants. The amount of IFN-g produced in the presence of Kogenate was considered as 100%. D. Inhibition of FVIII endocytosis by mannan. DCs were incubated alone or in the presence of mannan (1 mg/ml) for 30 minutes at 37°C before the addition of FVIII for 2 hours at 37°C. Results represent the percentage of inhibition of FVIII endocytosis in the presence of mannan using the condition without mannan as the reference. Error bars indicate SD for 2 to 4 independent experiments.

(5)

74

Repl

y t

o letter

3+

We thank Joelle Treton (INSERM U662, Hospital Saint-Louis, Paris, France) for providing us with blood from healthy MHC-matched donors. mAb 77IP52H7 and Factane were gifts from LFB. D9E9 and BO2C11 were gifts from J. M. Saint-Remy and M. Jacquemin (CMVB, KUL, Belgium). Kogenate, Helixate, Advate and Refacto were kind gifts from Bayer Healthcare, CSL-Behring, Baxter Bioscience, and Pfizer-Wyeth, respectively.

References

1. Herczenik E, van Haren SD, Wroblewska A, Kaijen P, van den Biggelaar M, Meijer AB, et al. Uptake of blood coagulation factor VIII by dendritic cells is mediated via its C1 domain. J Allergy Clin Immunol 2012;129:501-9, 509.e1-5.

2. Dasgupta S, Navarrete AM, Bayry J, Delignat S, Wootla B, Andre S, et al. A role for exposed mannosylations in presentation of human therapeutic self-proteins to CD4+ T lymphocytes. Proc

Natl Acad Sci USA 2007;104:8965-70.

3. Dasgupta S, Repesse Y, Bayry J, Navarrete AM, Wootla B, Delignat S, et al. VWF protects FVIII from endocytosis by dendritic cells and subsequent presentation to immune effectors. Blood 2007;109:610-2.

4. Jacquemin M, Vantomme V, Buhot C, Lavend’homme R, Burny W, Demotte N, et al. CD4+ T-cell

clones specific for wild-type factor VIII: a molecular mechanism responsible for a higher incidence of inhibitor formation in mild/moderate hemophilia A. Blood 2003;101:1351-8.

5. Navarrete AM, Delignat S, Teillaud JL, Kaveri SV, Lacroix-Desmazes S, Bayry J. CD4+CD25+

regulatory T cell-mediated changes in the expression of endocytic receptors and endocytosis process of human dendritic cells. Vaccine 2011;29:2649-52.

6. Miclea RD, Purohit VS, Balu-Iyer SV. O-Phospho-L-serine, multi-functional excipient for B domain deleted recombinant factor VIII. AAPS J 2007;9:E251-9.

(6)

Repl y t o letter

3+

A B

Figure 1. Proliferation of a FVIII C2 domain– specific CD4+ T-cell clone is mannan insensitive. A. Monocytes were isolated from

a matched HLA DRB1*0101/DRB1*1301 and cultured with 1000 U/ml of GM-CSF and 800 U/ ml of IL-4 for 4-7 days as described.1 DCs were first pretreated for 15 minutes with 1 mg/ml of mannan and incubated with 100 nM of FVIII for 1 hour to allow endocytosis. Then the cells were washed twice, resuspended at a concentration of 5×103/100 μl, and combined with 5×104/100 μl of carboxyfluorescein succinimidyl ester–

labeled CD4+ T-cell clone 32A-18, which was

derived from a DRB1*0101-positive hemophilia A patient. 2,3 After 6 days of culture, the mean fluorescent intensity of the resuspended CD4+ T cells was measured and the data were analyzed with FlowJo 7.6.4 software (Tree Inc, Ashland, Ore). B. The glycosylation site N2118 in the C1 domain remains distant from the KM33 epitope that includes residues K2092 and F2093. The carbohydrate structure depicted is a core-fucosylated high-mannose structure that was modeled by using the glycam Webserver (http://glycam.ccrc.uga.edu/ccrc/). The image was generated by using the PyMOL Molecular Graphics System, Version 1.3, Schrödinger, LLC (New York, NY). FITC, Fluorescein isothiocyanate.

To the Editor

Eszter Herczenik, Simon D. van Haren, Aleksandra Wroblewska, Paul Kaijen, Maartje van den Biggelaar, Alexander B. Meijer, Anja ten Brinke, Jan Voorberg

We first would like to thank Yohann Repessé and his coworkers 1 for their valuable

comments regarding our recent article on the uptake of blood coagulation factor VIII (FVIII) by dendritic cells (DCs). 2 In this report, we provided evidence that

mannose-sensitive receptors do not play a prominent role in the uptake of FVIII by DCs. In contrast to the recent work of Repessé and coworkers 3 only a

very small reduction in FVIII endocytosis was observed on preincubation with mannan. 2 Moreover, siRNA mediated knockdown of the macrophage mannose

receptor (CD206) did not influence the endocytosis of FVIII. Conversely, Repessé and coworkers demonstrate that the endocytosis of various therapeutic FVIII preparates such as Kogenate, Advate, Refacto and Factane by DCs is reduced in the presence of mannan. 1 To further address the role of mannose-ending glycans

in the contribution to FVIII endocytosis by DCs, we performed experiments by using patient-derived C2 domain–specific T-cell clone 32A-18 4,5 and HLA-matched

DCs. Prior to the addition of 100 nM of FVIII for 1 hour, we pretreated the HLA-matched DCs for 15 minutes with mannan. Subsequently, the DCs were combined

(7)

76

Repl

y t

o letter

3+

with carboxyfluorescein succinimidyl ester–labeled FVIII-specific T cells. After 6 days, we observed no decrease in fluorescent intensity in the samples in which T cells were added to untreated DCs or DCs treated only with mannan (Figure 1A, upper left and right panels). When DCs were preincubated with FVIII, the fluorescence of the CD4+ T cells decreased (Figure 1A, bottom left panel),

which indicates that the majority of the CD4+ T cells proliferated in response

to FVIII (Figure 1A). Similarly, we detected comparable CD4+ T-cell responses in

the sample that contained DCs that received mannan prior to the addition of FVIII (Figure 1A, bottom right panel). These observations suggest that mannan did not prevent FVIII endocytosis and subsequent peptide presentation to FVIII-specific CD4+ T cells by DCs. We have shown earlier that mAb KM33 prevents the

endocytosis of FVIII by DCs and associates with a reduced immunogenicity of FVIII in vivo. 2 We have also demonstrated that KM33 does not block the in vitro

binding of FVIII to the mannose receptor. 2 These observations strongly suggest

that the mannose receptor does not play a major role in FVIII endocytosis by DCs. We have previously indicated that residues K2092 and F2093 contribute to the binding of KM33 to FVIII (Figure 1B). 6 Mannose-ending glycans are present

at N239 and N2118 of FVIII. 7 Analysis of the 3-dimensional structure of FVIII

reveals that N2118 is not located in proximity to residues K2092 and F2093 (Figure 1B). We therefore propose that mannan-sensitive receptors do not play a major role in the endocytosis of FVIII by DCs. We are currently addressing which mannan-insensitive mechanisms contribute to the uptake of FVIII by DCs. We thank Dr. K. P. Pratt and E. A. James for providing the CD4+ T-cell clone used

(8)

Repl

y t

o letter

3+

References

1. Repessé Y, Dasgupta S, Navarrete A, Delignat S, Kaveri SV, Lacroix-Desmazes S. Mannose-sensitive receptors mediate the uptake of factor VIII therapeutics by human dendritic cells. J Allergy Clin Immunol 2012;129:1172-3.

2. Herczenik E, van Haren SD, Wroblewska A, Kaijen P, van den Biggelaar M, Meijer AB, et al. Uptake of blood coagulation factor VIII by dendritic cells is mediated via its C1 domain. J Allergy Clin Immunol 2012;129:501-9, 509.e1-5.

3. Dasgupta S, Navarrete AM, Bayry J, Delignat S, Wootla B, André S, et al. A role for exposed mannosylations in presentation of human therapeutic self-proteins to CD4+ T lymphocytes. Proc

Natl Acad Sci USA 2007;104:8965-70.

4. James EA, Kwok WW, Ettinger RA, Thompson AR, Pratt KP. T-cell responses over time in a mild hemophilia A inhibitor subject: epitope identification and transient immunogenicity of the corresponding self-peptide. J Thromb Haemost 2007;5:2399-407.

5. Ettinger RA, James EA, Kwok WW, Thompson AR, Pratt KP. Lineages of human T-cell clones, including T helper 17/T helper 1 cells, isolated at different stages of anti-factor VIII immune responses. Blood 2009;114:1423-8.

6. Meems H, Meijer AB, Cullinan DB, Mertens K, Gilbert GE. Factor VIII C1 domain residues Lys 2092 and Phe 2093 contribute to membrane binding and cofactor activity. Blood 2009;114: 3938-46.

7. Medzihradszky KF, Besman MJ, Burlingame AL. Structural characterization of site-specific N-glycosylation of recombinant human factor VIII by reversed-phase high performance liquid chromatography-electrospray ionization mass spectrometry. Anal Chem 1997;69:3986-94.

Referenties

GERELATEERDE DOCUMENTEN

De algemene conclusie op basis van de verschillende deelonderzoeken luidt dat samenwerking een complexe, rommelige en chaotische aangelegenheid is in een context waar steeds van

The focus is on the interaction between the people in organisations with different interests, views and practices, coming from various domains of society and faced with a

Vandaaruit kwam ze terecht bij landelijke kennisinstituten waaronder de Nederlandse Federatie voor Jeugd en Jongeren werk, het Nederlands Instituut voor Natuurvrienden werk (NIVON)

De BeweegKuur evolueert van een gesloten tot een open innovatie met een lokale point of view, hetgeen cruciaal is voor de effectiviteit van de interventie (vrij naar aanleiding van

We have shown how the variability in IRAS 13224–3809 behaves in a different manner to what is typically observed in AGNs, and even for accreting sources in general. Non-

Testing the deep-crustal heating model using quiescent neutron-star very-faint X-ray transients and the possibility of partially accreted crusts in accreting neutron stars..

The column density evolution of Fe , Fe , Si  and C  (both ground-state and excited levels) is fit with an excitation-only model, which includes the following

Marnix’ zoon bevond zich tezelfder tijd eveneens in Leiden, maar volgt Vulcanius naar Antwerpen; vanaf mei 1581 studeert hij weer in Leiden, opnieuw onder toezicht van