longitudinal dynamics of the human b cell response …the design of next-generation vaccines. here,...

11
Longitudinal dynamics of the human B cell response to the yellow fever 17D vaccine Anna Z. Wec a , Denise Haslwanter b , Yasmina N. Abdiche c , Laila Shehata a,1 , Nuria Pedreño-Lopez d , Crystal L. Moyer e , Zachary A. Bornholdt e , Asparouh Lilov a , Juergen H. Nett a , Rohit K. Jangra b , Michael Brown a , David I. Watkins d , Clas Ahlm f , Mattias N. Forsell f , Félix A. Rey g , Giovanna Barba-Spaeth g , Kartik Chandran b , and Laura M. Walker a,2 a Adimab, LLC, Lebanon, NH 03766; b Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461; c Carterra Inc., Salt Lake City, UT 84103; d Department of Pathology, University of Miami, Miami, FL 33146; e Mapp Biopharmaceutical, Inc., San Diego, CA 92121; f Division of Infection & Immunology, Department of Clinical Microbiology, Umeå University, 90187 Umeå, Sweden; and g Structural Virology Unit, CNRS UMR 3569, Virology Department, Institut Pasteur, 75015 Paris, France Edited by Adolfo Garcia-Sastre, Icahn School of Medicine at Mount Sinai, New York, NY, and approved February 12, 2020 (received for review December 9, 2019) A comprehensive understanding of the development and evolution of human B cell responses induced by pathogen exposure will facilitate the design of next-generation vaccines. Here, we utilized a high- throughput single B cell cloning technology to longitudinally track the human B cell response to the yellow fever virus 17D (YFV-17D) vaccine. The early memory B cell (MBC) response was mediated by both classical immunoglobulin M (IgM) (IgM + CD27 + ) and switched immunoglobulin (swIg + ) MBC populations; however, classical IgM MBCs waned rapidly, whereas swIg + and atypical IgM + and IgD + MBCs were stable over time. Affinity maturation continued for 6 to 9 mo following vaccination, providing evidence for the persistence of germinal center activity long after the period of active viral replication in peripheral blood. Finally, a substantial fraction of the neutralizing antibody response was medi- ated by public clones that recognize a fusion loop-proximal antigenic site within domain II of the viral envelope glycoprotein. Overall, our findings provide a framework for understanding the dynamics and complexity of human B cell responses elicited by infection and vaccination. antiviral vaccination | antibody responses | yellow fever virus | monoclonal antibody | B cell memory M emory B cells (MBCs) formed in response to infection and vaccination provide protection against the consequences of reexposure to the same pathogen. These cells can differentiate into antibody-secreting cells to provide an immediate source of serum antibody (Ab) or they can enter germinal centers (GCs) to rediversify their B cell receptors (BCRs) in response to evolving or antigenically related pathogens (1). Over the past decade, numerous studies have shown that the MBC compartment in both humans and mice is highly heterogenous (2). In humans, both switched immunoglobulin (swIg + ) and immunoglobulin M (IgM + ) memory cells have been described, as well as atypicalMBCs that express FCRL4 or FCRL5 and/or lack expression of the canonical MBC surface marker CD27 (2, 3). Various acti- vated B cell subsets, which appear transiently after infection or vaccination, have also been reported (46). In mice, MBCs can be subcategorized based on their expression of Ig isotype as well as other surface markers, such as CD80, PD-L2, and CD73 (2, 7). Interestingly, recent studies have shown that murine IgM + MBCs formed in response to immunization with model proteins are abundant and stable over time, whereas the longevity of antigen- specific swIg + MBCs appears to be dependent on the frequency of high-avidity germline-encoded BCRs in the naïve B cell rep- ertoire (810). Although these studies have provided important insights into the hierarchy of murine IgM + and swIg + MBC populations induced by nonreplicating antigens, it remains un- clear whether, and how, these findings will translate to human MBC responses induced by infection or vaccination. Further- more, the relative contribution of different MBC subsets to the human B cell response to pathogen exposure, and if/how these subsets change over time, remains to be elucidated. Due to the high level of protection afforded by the live- attenuated yellow fever virus 17D (YFV-17D) vaccinereported to be primarily mediated by neutralizing antibodies (nAbs) targeting the YFV E protein (11)we sought to gain a com- prehensive view of the human YFV E-specific B cell response underlying this efficacy. Importantly, despite its success, our knowledge of the human B cell response to the YFV-17D vac- cine remains limited to studies of human sera and a handful of YFV E-specific human monoclonal Abs (mAbs). Recent serum depletion studies have shown that a large fraction of the YFV E- specific serum Ab response is mediated by Abs targeting domain I (DI) and/or domain II (DII) of the E protein, whereas Abs targeting domain III (DIII) are absent or present at very low titers (12). Correspondingly, the six YFV E-specific human Significance The yellow fever virus 17D (YFV-17D) vaccine is one of the safest and most effective vaccines ever developed and is widely considered to be the gold standardof antiviral vac- cination. Despite its success, remarkably little is known about the human B cell response induced by YFV-17D and the speci- ficities and functional activities of the antibodies encoded by these B cells. Here we report the molecular characterization of the complexity, dynamics, and evolution of this response at the single-cell level. Our work provides key insights into the B cell response underlying the efficacy of the YFV-17D vaccine, which may facilitate the design of vaccines for more refractory pathogens, such as malaria and HIV. Author contributions: A.Z.W., Y.N.A., A.L., and L.M.W. designed research; A.Z.W., D.H., Y.N.A., L.S., N.P.-L., A.L., and R.K.J. performed research; D.H., N.P.-L., C.L.M., Z.A.B., J.H.N., R.K.J., M.B., D.I.W., C.A., M.N.F., F.A.R., G.B.-S., and K.C. contributed new reagents/analytic tools; A.Z.W., D.H., Y.N.A., L.S., N.P.-L., A.L., M.B., K.C., and L.M.W. analyzed data; and A.Z.W. and L.M.W. wrote the paper. Competing interest statement: A.Z.W., L.S., A.L., J.H.N., M.B., and L.M.W. are employees and equity holders in Adimab LLC. C.L.M. and Z.A.B. are employees of and shareholders in Mapp Biopharmaceutical, Inc. This article is a PNAS Direct Submission. This open access article is distributed under Creative Commons Attribution-NonCommercial- NoDerivatives License 4.0 (CC BY-NC-ND). Data deposition: The sequences of neutralizing antibodies isolated in this study have been deposited in the GenBank database, https://www.ncbi.nlm.nih.gov/genbank (accession nos. MN993290MN993593). 1 Present address: Department of Immunology, University of Washington School of Med- icine, Seattle, WA 98195. 2 To whom correspondence may be addressed. Email: [email protected]. This article contains supporting information online at https://www.pnas.org/lookup/suppl/ doi:10.1073/pnas.1921388117/-/DCSupplemental. First published March 9, 2020. www.pnas.org/cgi/doi/10.1073/pnas.1921388117 PNAS | March 24, 2020 | vol. 117 | no. 12 | 66756685 IMMUNOLOGY AND INFLAMMATION Downloaded by guest on October 29, 2020

Upload: others

Post on 07-Aug-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Longitudinal dynamics of the human B cell response …the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally

Longitudinal dynamics of the human B cell response tothe yellow fever 17D vaccineAnna Z. Weca, Denise Haslwanterb, Yasmina N. Abdichec, Laila Shehataa,1, Nuria Pedreño-Lopezd, Crystal L. Moyere,Zachary A. Bornholdte, Asparouh Lilova, Juergen H. Netta, Rohit K. Jangrab, Michael Browna, David I. Watkinsd,Clas Ahlmf, Mattias N. Forsellf, Félix A. Reyg, Giovanna Barba-Spaethg, Kartik Chandranb

, and Laura M. Walkera,2

aAdimab, LLC, Lebanon, NH 03766; bDepartment of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461; cCarterra Inc.,Salt Lake City, UT 84103; dDepartment of Pathology, University of Miami, Miami, FL 33146; eMapp Biopharmaceutical, Inc., San Diego, CA 92121; fDivision ofInfection & Immunology, Department of Clinical Microbiology, Umeå University, 90187 Umeå, Sweden; and gStructural Virology Unit, CNRS UMR 3569,Virology Department, Institut Pasteur, 75015 Paris, France

Edited by Adolfo Garcia-Sastre, Icahn School of Medicine at Mount Sinai, New York, NY, and approved February 12, 2020 (received for review December9, 2019)

A comprehensive understanding of the development and evolution ofhuman B cell responses induced by pathogen exposure will facilitatethe design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally track thehuman B cell response to the yellow fever virus 17D (YFV-17D) vaccine.The early memory B cell (MBC) responsewasmediated by both classicalimmunoglobulin M (IgM) (IgM+CD27+) and switched immunoglobulin(swIg+) MBC populations; however, classical IgM MBCs waned rapidly,whereas swIg+ and atypical IgM+ and IgD+ MBCs were stable overtime. Affinitymaturation continued for 6 to 9mo following vaccination,providing evidence for the persistence of germinal center activity longafter the period of active viral replication in peripheral blood. Finally, asubstantial fraction of the neutralizing antibody response was medi-ated by public clones that recognize a fusion loop-proximal antigenicsite within domain II of the viral envelope glycoprotein. Overall, ourfindings provide a framework for understanding the dynamics andcomplexity of human B cell responses elicited by infection andvaccination.

antiviral vaccination | antibody responses | yellow fever virus | monoclonalantibody | B cell memory

Memory B cells (MBCs) formed in response to infection andvaccination provide protection against the consequences

of reexposure to the same pathogen. These cells can differentiateinto antibody-secreting cells to provide an immediate source ofserum antibody (Ab) or they can enter germinal centers (GCs) torediversify their B cell receptors (BCRs) in response to evolvingor antigenically related pathogens (1). Over the past decade,numerous studies have shown that the MBC compartment inboth humans and mice is highly heterogenous (2). In humans,both switched immunoglobulin (swIg+) and immunoglobulin M(IgM+) memory cells have been described, as well as “atypical”MBCs that express FCRL4 or FCRL5 and/or lack expression ofthe canonical MBC surface marker CD27 (2, 3). Various acti-vated B cell subsets, which appear transiently after infection orvaccination, have also been reported (4–6). In mice, MBCs canbe subcategorized based on their expression of Ig isotype as wellas other surface markers, such as CD80, PD-L2, and CD73 (2, 7).Interestingly, recent studies have shown that murine IgM+ MBCsformed in response to immunization with model proteins areabundant and stable over time, whereas the longevity of antigen-specific swIg+ MBCs appears to be dependent on the frequencyof high-avidity germline-encoded BCRs in the naïve B cell rep-ertoire (8–10). Although these studies have provided importantinsights into the hierarchy of murine IgM+ and swIg+ MBCpopulations induced by nonreplicating antigens, it remains un-clear whether, and how, these findings will translate to humanMBC responses induced by infection or vaccination. Further-more, the relative contribution of different MBC subsets to the

human B cell response to pathogen exposure, and if/how thesesubsets change over time, remains to be elucidated.Due to the high level of protection afforded by the live-

attenuated yellow fever virus 17D (YFV-17D) vaccine—reportedto be primarily mediated by neutralizing antibodies (nAbs)targeting the YFV E protein (11)—we sought to gain a com-prehensive view of the human YFV E-specific B cell responseunderlying this efficacy. Importantly, despite its success, ourknowledge of the human B cell response to the YFV-17D vac-cine remains limited to studies of human sera and a handful ofYFV E-specific human monoclonal Abs (mAbs). Recent serumdepletion studies have shown that a large fraction of the YFV E-specific serum Ab response is mediated by Abs targeting domainI (DI) and/or domain II (DII) of the E protein, whereas Abstargeting domain III (DIII) are absent or present at very lowtiters (12). Correspondingly, the six YFV E-specific human

Significance

The yellow fever virus 17D (YFV-17D) vaccine is one of thesafest and most effective vaccines ever developed and iswidely considered to be the “gold standard” of antiviral vac-cination. Despite its success, remarkably little is known aboutthe human B cell response induced by YFV-17D and the speci-ficities and functional activities of the antibodies encoded bythese B cells. Here we report the molecular characterization ofthe complexity, dynamics, and evolution of this response at thesingle-cell level. Our work provides key insights into the B cellresponse underlying the efficacy of the YFV-17D vaccine, whichmay facilitate the design of vaccines for more refractorypathogens, such as malaria and HIV.

Author contributions: A.Z.W., Y.N.A., A.L., and L.M.W. designed research; A.Z.W., D.H.,Y.N.A., L.S., N.P.-L., A.L., and R.K.J. performed research; D.H., N.P.-L., C.L.M., Z.A.B., J.H.N.,R.K.J., M.B., D.I.W., C.A., M.N.F., F.A.R., G.B.-S., and K.C. contributed new reagents/analytictools; A.Z.W., D.H., Y.N.A., L.S., N.P.-L., A.L., M.B., K.C., and L.M.W. analyzed data; andA.Z.W. and L.M.W. wrote the paper.

Competing interest statement: A.Z.W., L.S., A.L., J.H.N., M.B., and L.M.W. are employeesand equity holders in Adimab LLC. C.L.M. and Z.A.B. are employees of and shareholders inMapp Biopharmaceutical, Inc.

This article is a PNAS Direct Submission.

This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

Data deposition: The sequences of neutralizing antibodies isolated in this study havebeen deposited in the GenBank database, https://www.ncbi.nlm.nih.gov/genbank(accession nos. MN993290–MN993593).1Present address: Department of Immunology, University of Washington School of Med-icine, Seattle, WA 98195.

2To whom correspondence may be addressed. Email: [email protected].

This article contains supporting information online at https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1921388117/-/DCSupplemental.

First published March 9, 2020.

www.pnas.org/cgi/doi/10.1073/pnas.1921388117 PNAS | March 24, 2020 | vol. 117 | no. 12 | 6675–6685

IMMUNOLO

GYAND

INFLAMMATION

Dow

nloa

ded

by g

uest

on

Oct

ober

29,

202

0

Page 2: Longitudinal dynamics of the human B cell response …the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally

mAbs described to date target overlapping epitopes within DIIof the E protein (13, 14).To longitudinally track the development and evolution of the

human B cell response induced by the YFV-17D vaccine, weisolated and characterized hundreds of YFV E-specific mAbsfrom two flavivirus-naïve donors at multiple time points follow-ing YFV-17D vaccination, spanning the course of 360 d. Here wepresent an analysis of the kinetics, dynamics, and heterogeneityof this B cell response, as well as an in-depth characterization ofthe mAbs encoded by these B cells. Overall, the results provide aglobal view of the human MBC response to a highly successfulvaccine and may inform the development of vaccines againstrefractory pathogens, such as malaria and HIV.

ResultsStudy Design. To study the evolution of the human B cell re-sponse to YFV-17D, we immunized two healthy donors with theYFV-17D Stamaril vaccine and collected blood samples at days10, 14, 28, 90, 180, 270, and 360 postvaccination (SI Appendix,Fig. S1A and Table S1). Neither of the YFV-17D vaccinees had ahistory of YFV infection or vaccination, and correspondingly theprevaccination sera from both donors showed no detectablebinding reactivity to YFV-17D viral particles or recombinantYFV E or NS1 proteins (SI Appendix, Fig. S1 B–D). Further-more, neither prevaccination serum showed detectable neutralizing

activity against YFV-17D (SI Appendix, Fig. S1E). Since YFV isantigenically related to other flaviviruses, we also confirmed thatthe prevaccination sera from both donors lacked reactivity withE and NS1 proteins from commonly circulating flaviviruses(dengue virus serotypes 1 through 4 [DENV1-4], Japanese en-cephalitis virus [JEV], tick-borne encephalitis virus, West Nilevirus [WNV], and Zika virus [ZIKV]) (SI Appendix, Fig. S1 Cand D). Hence, both donors were likely flavivirus-naïve at thetime of vaccination. The IgG and IgM serum kinetics followingvaccination also supported primary exposure (SI Appendix, Fig.S1F). Consistent with prior studies (15, 16), serum neutralizingactivity against YFV-17D appeared in both donors by day 10postvaccination and persisted through the course of the study (SIAppendix, Fig. S1 E and G).

Primary YFV-17D Vaccination Induces Highly Mutated PlasmablastResponses in Flavivirus-Naïve Donors. Since previous studies haveshown that plasmablasts (PBs) appear transiently in peripheralblood ∼10 to 14 d following YFV-17D vaccination (17), wemonitored PB responses in both donors on day 10 and 14 post-vaccination. In both donors, expanded PB populations that were∼10- to 20-fold over prevaccination levels were observed at bothtime points (Fig. 1A and SI Appendix, Fig. S2).To further characterize the PB response to YFV-17D, we

single-cell-sorted ∼600 PBs from both donors and amplified the

0

10

20

30

# V H

nt.

subs

titut

ions *

**

Day 10 Day 14Donor 22 11

B

52

Day

10

107

79

131

Mut.GL

Day

14

Donor 1 Donor 2C

E

ADI-467

15

ADI-451

43

ADI-451

42

ADI-451

41

ADI-451

44

ADI-467

29

ADI-467

42

ADI-467

17

ADI-467

39

ADI-467

41

ADI-467

22

ADI-467

24

ADI-467

18

ADI-467

28

ADI-467

31

ADI-467

30

ADI-467

37

ADI-467

35

ADI-467

20

ADI-467

32

ADI-467

43

ADI-451

45

ADI-467

33

ADI-467

27

ADI-467

19

ADI-467

38

ADI-467

26

ADI-467

16

ADI-467

34

ADI-467

25

ADI-461

84

ADI-467

21

ADI-421

68

ADI-451

460

255075

100

010203040

%N

eutra

lizat

ion Day 10 PB Day 14 PB # (V

H +VL ) nt. subs.

100 nM10 nM

Donor

1

Donor

2

SHM

6 5

27 29

F Donor 1 Donor 2

Day

10

Day

14

NeutralizationIC50 [nM]<11 to 10 >10 to 100>100

Mut.GL

Donor 1 Donor 2G Neutralizing PB mAbs

A

0.1

1

10

100

0 10 14Day

Donor 2Donor 1

% P

lasm

abla

sts

amon

gC

D19

+ CD

20-/l

o B c

ells

14

10

14

10

Day

Don

or 1

Don

or 2

Non-bindingBinding

YFV-17D

D

0 50 100%PB mAbs

15 15

ADI-451

64

ADI-467

47

ADI-451

66

ADI-451

67

ADI-451

65

ADI-451

63

ADI-505

36

ADI-505

31

ADI-505

35

ADI-451

61

ADI-505

37

ADI-505

34

ADI-488

90

ADI-451

62

ADI-505

39

ADI-505

33

ADI-505

38

ADI-451

60

ADI-505

32

ADI-451

56

ADI-505

40

ADI-440

94

ADI-488

92

ADI-440

96

ADI-505

41

ADI-505

42

ADI-440

92

ADI-505

43

ADI-451

55

ADI-451

58

ADI-451

57

ADI-451

590

255075

100

010203040

%N

eutra

lizat

ion

Day 10 PB Day 14 PB # (VH +V

L ) nt. subs.

Fig. 1. Molecular characterization of YFV-17D–induced PB responses. (A) Frequency of PBs among circulating CD19+CD20—/lo B cells at days 0, 10, and 14postvaccination. (B) SHM loads of mAbs isolated from day 10 and day 14 PBs. Black bars indicate medians. (C) Proportion of mAbs isolated from day 10 andday 14 PBs that contain somatic mutations. (D) Percentage of PB-derived mAbs that showed detectable ELISA binding reactivity to whole YFV-17D particles.(E) Neutralizing activities of PB-derived mAbs against YFV-17D at 100 nM and 10 nM concentrations. Green dots indicate the number of nucleotide sub-stitutions in VH + VL for each mAb. (F) Proportion of YFV-17D binding mAbs with the indicated neutralization IC50s. MAbs that displayed <50% neutralizationat 100 nM in the preliminary screen are included in the IC50 >100 nM group. (G) Proportion of PB-derived nAbs (defined as Abs displaying >50% neutrali-zation at 100 nM) that contain somatic mutations. Statistical comparisons were made using the Mann–Whitney U test (**P < 0.01, *P < 0.05). nt, nucleotide;n.n., nonneutralizing; GL, germline; Mut., mutated; VH, variable region of the heavy chain; IC50, half-maximal inhibitory concentration.

6676 | www.pnas.org/cgi/doi/10.1073/pnas.1921388117 Wec et al.

Dow

nloa

ded

by g

uest

on

Oct

ober

29,

202

0

Page 3: Longitudinal dynamics of the human B cell response …the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally

corresponding Ab heavy- and light-chain variable (VH and VL)genes by single-cell PCR. One hundred sixty-one and 210 nativelypaired mAbs were cloned from donors 1 and 2, respectively, andexpressed as full-length IgGs in an engineered strain of Saccha-romyces cerevisiae (18). Sequence analysis showed that the PBresponses were highly diverse in both donors, with only about 15%of clones belonging to expanded clonal lineages (SI Appendix, Fig.S3). Unexpectedly, a large fraction of the PB-derived mAbs fromboth donors contained high levels of somatic hypermutation(SHM), suggesting either efficient recruitment of preexistingMBCs into the PB response or the early onset of affinity matu-ration within GC reactions or at extrafollicular sites (19) (Fig. 1B).We also expanded this analysis to include two additional flavivirus-naïve donors and observed similar results (SI Appendix, Fig. S4). Inthree out of four donors, the median level of SHM in the PB-derived mAbs was significantly higher on day 10 than on day 14(Fig. 1B and SI Appendix, Fig. S4). Correspondingly, a larger pro-portion of mAbs cloned from day 14 PBs lacked SHM, suggesting anincreased recruitment of cells from the naïve B cell compartment atthis time point (Fig. 1C). To analyze whether the somatic mutationsin the PB-derived mAbs contribute to binding activity, we generatedinferred unmutated common ancestor (UCA) mAbs from threesomatically mutated PB clones and measured their binding affinitiesto a recombinant YFV E protein. In all three cases, the UCA mAbsshowed substantially reduced binding affinities compared to themature mAbs, suggesting that somatic mutations in the PB mAbsare important for recognition of YFV E (SI Appendix, Fig. S5).We next tested the PB-derived mAbs for binding reactivity to

YFV-17D particles using a sandwich enzyme-linked immuno-sorbent assay (ELISA) (Fig. 1D and SI Appendix, Fig. S6A). Thefrequency of YFV-17D binding mAbs isolated from day 10 and14 PBs ranged from 6 to 37%, which is substantially lower thanthe percentage of antigen-specific mAbs recovered from PBsinduced by influenza vaccination or secondary DENV infection(20, 21). The low proportion of YFV-17D–reactive clones maybe due to promiscuous B cell activation (19) or low binding af-finity of the PB-derived mAbs or because the PB responses wereprimarily directed against other viral antigens not tested in thisstudy (e.g., viral core proteins). Notably, the binding and non-binding populations contained a similar proportion of poly-reactive clones, with ∼10 to 20% of PB-derived mAbs showingsome degree of polyreactivity (SI Appendix, Fig. S6B). Thus, it isunlikely that the nonbinding PB population originates from non-specifically activated, polyreactive B cell clones. Furthermore, themAbs that failed to bind YFV-17D by ELISA also showed little tono binding to a recombinant YFV E protein in a biolayer inter-ferometry (BLI) assay using an avid binding orientation, indicatingthe lack of detectable binding to YFV-17D particles by ELISA islikely not due to low affinity or assay format (SI Appendix, Fig.S6C). Despite the low frequency of YFV-specific clones, we re-covered 33 and 34 YFV-17D binding mAbs from the expanded PBpopulations in donors 1 and 2, respectively. The SHM loads of thebinding mAbs were similar to those observed in the total PBpopulation (Fig. 1B and SI Appendix, Fig. S7).The early appearance of serum neutralizing activity following

YFV-17D vaccination suggests the presence of nAbs in the PBcompartment (SI Appendix, Fig. S1 E and G). To test this di-rectly, we analyzed the neutralizing activities of the PB-derived,YFV-17D binding mAbs in a microtiter neutralization assay at100 and 10 nM concentrations. Preliminary studies with a panelof control mAbs showed that neutralization half-maximal in-hibitory concentrations (IC50s) in this assay were highly compa-rable to those observed in a classic focus reduction neutralizationtest assay (SI Appendix, Fig. S8A). The neutralizing activities ofthe PB-derived mAbs ranged from complete neutralization at10 nM to no detectable neutralization at 100 nM (Fig. 1E).Notably, a substantially higher fraction of mAbs isolated fromday 14 PBs displayed neutralizing activity compared to those

isolated from day 10 PBs, which is consistent with the increasedserum neutralizing activity on day 14 versus day 10 in both do-nors (Fig. 1F and SI Appendix, Fig. S1 E and G). Furthermore,neutralization titration experiments on the mAbs displaying atleast 50% infection inhibition at 100 nM revealed that 9 to 16%of YFV-17D binding mAbs isolated from day 14 PBs displayedmedium to high neutralizing activity (IC50s ≤10 nM) (Fig. 1F andSI Appendix, Fig. S8B). Interestingly, sequence analysis showedthat 12.5 to 33% of the PB-derived nAbs utilized VH4-4/VL1-51germline gene pairing, suggesting recognition of a common an-tigenic site (SI Appendix, Fig. S9). Finally, 53% and 20% of thenAbs isolated from donors 1 and 2, respectively, lacked somaticmutations, indicating that YFV-17D nAbs are present in thenaïve B cell repertoire (Fig. 1 E and G). We conclude that YFV-17D vaccination induces PB responses that originate from bothnaïve and MBCs, and only a minority of these B cells encode Absthat display potent neutralizing activity.

Different B Cell Populations Mediate Early and Late Memory to YFV-17D. To study the MBC response to YFV-17D, we stained purifiedB cells from each sampling time point with a panel of B cell surfacemarkers (CD19, CD20, CD27, IgM, IgD, CD21, and CD71) and afluorescently labeled recombinant YFV E protein. The YFV Eantigen used for B cell staining was recognized by two well-characterized control mAbs, 4G2 and 5A, providing evidence forproper folding (13) (SI Appendix, Fig. S10A). YFV E-specificswIg+ MBCs emerged in both donors by days 14 to 28, peakedbetween days 90 and 180, and slowly declined between days 180 and360 (Fig. 2 A and B).To further dissect the MBC response to YFV-17D, we single-

cell-sorted between 100 to 400 YFV E-reactive B cells from bothdonors at each sampling time point for mAb cloning and char-acterization. In order to capture the full heterogeneity of theMBC response, we sorted all YFV E-reactive B cells (regardlessof surface phenotype) and used index sorting to track the surfacemarkers expressed on each sorted cell (SI Appendix, Fig. S10 Band C). This analysis revealed that YFV E-specific MBC re-sponse in both donors was highly heterogenous at all time points(Fig. 2C). At the earliest sampling time point, activated naïve Bcells and IgM+CD27+ MBCs dominated the response in bothdonors, but these B cell populations waned rapidly over time(Fig. 2 C and D). By day 90, less than 15% of the YFV E-specificresponse was comprised of IgM+CD27+ MBCs, and by day 360only about 5% of YFV E-specific B cells belonged to this MBCpopulation (Fig. 2 C and D). In contrast, the swIg+ MBC pop-ulation—which was composed of both CD27+ and CD27− B cells—expanded between day 14 and day 90 and then remained stablethroughout the course of the study (Fig. 2 C and D).To determine whether the observed hierarchy of IgM+ and

swIg+ MBC populations was unique to YFV-17D vaccination, wealso stained longitudinal samples from two Puumala hantavirus(PUUV)-experienced donors with the same panel of B cell markersdescribed above and a fluorescently labeled PUUV envelope proteinsubunit (Gn) (SI Appendix, Fig. S11A). Flow cytometric analysisrevealed that PUUV Gn-specific IgM+ memory cells were presentduring early convalescence but waned rapidly in both donors (SIAppendix, Fig. S11B). In contrast, swIg+ MBCs appeared early andwere maintained at least until day 349 or 237 (the last sampling timepoints available) in donors 165 and 192, respectively (SI Appendix,Fig. S11B). Hence, the longevity of IgM+ and swIg+ MBC responsesobserved after primary YFV-17D vaccination is highly similar to thatobserved following primary infection with PUUV.Interestingly, sequence analysis revealed that over half of the

YFV E-specific mAbs isolated from phenotypically naïve(IgM+IgD+CD27−) B cells were somatically mutated, suggestingthat these B cells are GC-experienced and therefore representone or more atypical MBC subsets (SI Appendix, Fig. S12 A and B).A comparable proportion of mAbs isolated from IgM+IgD−CD27−

Wec et al. PNAS | March 24, 2020 | vol. 117 | no. 12 | 6677

IMMUNOLO

GYAND

INFLAMMATION

Dow

nloa

ded

by g

uest

on

Oct

ober

29,

202

0

Page 4: Longitudinal dynamics of the human B cell response …the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally

and IgM−IgD+CD27− B cells were also somatically mutated,suggesting that these B cells are also bona fide MBCs (SI Ap-pendix, Fig. S12 A and B). The median level of SHM in thesethree atypical MBC subsets was similar to that observed inclassical IgM MBCs (IgM+IgD+CD27+) (SI Appendix, Fig.S12B). However, in contrast to classical IgM+CD27+ MBCs, theseatypical IgM+ and/or IgD+CD27− MBC populations comprisedabout one-third of the total YFV E-specific MBC response at day180 and appeared to be stable until at least day 360 (Fig. 2 C andD). Overall, the results indicate that the early MBC response toYFV-17D vaccination is mediated by both swIg+ and classical IgMMBCs, whereas the late MBC response is dominated by swIg+ andatypical IgM+ and/or IgD+ MBCs.

Prolonged Evolution of the MBC Response to YFV-17D Vaccination. Tocharacterize the evolution of the MBC response to YFV-17Dvaccination, we tracked the SHM loads, apparent binding af-finities (KD

Apps), and neutralization potencies of the YFV E-specific mAbs at each sampling time point. In both donors, themedian level of SHM was low at day 14—with over 50% of Abslacking somatic mutations—and increased gradually over a 6- to9-mo time period (Fig. 3 A and B). By 9 mo postvaccination,SHM loads plateaued in both donors, with a median of nine andseven nucleotide substitutions in VH for donors 1 and 2, re-spectively (Fig. 3 A and B). Consistent with the mutational loadanalysis, binding studies with a recombinant YFV E proteinshowed that the KD

Apps of the MBC-derived mAbs were veryweak at early time points and progressively improved for 6 to 9 mo

0.080

0

-103

103

104

105

0.17 0.29 0.83 0.82 0.63 0.32

0.31

0-103

103

104

105

0.81

0-103

103

104

105

1.96

0-103

103

104

105

1.58

0-103

103

104

105

1.04

0-103

103

104

105

0.61

0-103

103

104

105

0.12

0-103

103

104

105

0

-103

103

104

105

YFV

E pr

otei

n | A

PC

YFV E protein | PE

Donor 1

Donor 2

Donor 1 Donor 2IgMIgD

CD27CD71CD21SHM D

ay 1

4 IgMIgDCD27CD71CD21SHM

Day

28IgM

SHM

IgDCD27CD71CD21

IgMIgDCD27CD71CD21SHM

IgMIgD

SHM

CD27CD71CD21 D

ay 9

0 IgMIgDCD27CD71CD21SHM

IgM

Day

180IgD

CD27CD71CD21SHM

IgMIgDCD27CD71CD21SHM

Day

270

IgMIgD

CD27CD71CD21SHM

IgMIgDCD27CD71CD21SHM

Day

360

IgMIgD

CD27CD71CD21SHM

IgMIgDCD27CD71CD21SHM

0

20

40

60

80

100

%YF

V E-

spec

ific

B ce

lls

0

20

40

60

80

100 IgM+IgD–CD27+

swIg+CD27–

IgM–IgD+CD27–SHM+ | atypical IgD-only memory

IgM+IgD+CD27+

IgM+IgD–CD27–SHM+ | atypical IgM-only memoryIgM+IgD+CD27–SHM+ | atypical IgM/IgD memory

IgM+IgD+CD27–CD71+SHM– orIgM+IgD+CD27–CD21loSHM–

IgM+IgD+CD27–CD21+CD71–SHM– | naïve

swIg+CD27+

14 28 90 180

270

360Day14 28 90 18

027

036

0Day

Donor 1 Donor 2

activated naive

0 14 28 90 180

270

360

0

1

2

3

%YF

V E-

spec

ific

swIg

+ B c

ell s

Donor 2Donor 1

Day

A B

C

D

Day 14 Day 28 Day 90 Day 180 Day 270 Day 360Day 0Gated on CD19+CD20+IgM—IgD— B cells

positivenegative

Fig. 2. Different B cell populations mediate early and late memory to YFV-17D. (A) Frequency of YFV E-specific swIg+ B cells at each sampling time point.Fluorescence-activated cell sorting plots shown are gated on CD19+CD20+ IgD− IgM− B cells. YFV E was labeled with two different colors to reduce back-ground binding. (B) Percentage of swIg+ B cells at each sampling time point that display YFV E reactivity. (C) Heat map showing the presence or absence ofSHM and the indicated B cell surface markers expressed on cells from which YFV E-specific mAbs were isolated. Surface marker expression was determined byflow cytometry. (D) Proportion of YFV E-specific B cells at each sampling time point that belong to the indicated B cell subsets.

6678 | www.pnas.org/cgi/doi/10.1073/pnas.1921388117 Wec et al.

Dow

nloa

ded

by g

uest

on

Oct

ober

29,

202

0

Page 5: Longitudinal dynamics of the human B cell response …the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally

14 28 90 180

270

360

0

10

20

30

Days post-vaccination

# V H

nt.

subs

itutio

ns ******

**

14 28 90 180

270

360

***** ****

A Donor 1 Donor 2 Donor 1 Donor 2B

% Y

FV E

-spe

cific

mAb

s

14 28 90 180

270

360

0

20

40

60

80

100

>15

1 to 50

14 28 90 180

270

360

6 to 1011 to 15

# VH Mutations

Days post-vaccination

14 28 90 180

270

360

10-10

10-9

10-8

10-7

10-6

Days post-vaccination

**** **

YFV

E K D

App [

M]

14 28 90 180

270

360

***** **Donor 1 Donor 2C

14 28 90 180

270

360

0

20

40

60

80

100

% Y

FV E

-spe

cific

mAb

s

Days post-vaccination

D Donor 1 Donor 2

Srong (<1 nM)Medium (>1 to 5 nM)Low (>5 to 50 nM)Weak (>50 nM)Avid-only

14 28 90 180

270

360

YFV E binding

14 28 90 180

270

360

0

20

40

60

80

100

% Y

FV E

-spe

cific

B c

ells

14 28 90 180

270

360

CD71+

CD21lo

Donor 1 Donor 2

E

Days post-vaccination

Donor 1 Donor 2

14 28 900

20

40

60

80

100

CD71— CD21loCD71+ CD21+

CD71+ CD21lo

14 28 90

Days post-vaccination

F

% Y

FV E

-spe

cific

ac

tivat

ed B

cel

ls

Days post-vaccination14 28 90 18

027

036

00.001

0.01

0.1

1

10

100

Neu

traliz

atio

n IC

50 [n

M]

14 28 90 180

270

360

n.n.Donor 1 Donor 2

14 28 90 180

270

3600

25

50

75

100

% M

BC-d

eriv

ed m

Abs

14 28 90 180

270

360

< 11 to 10>10 to 100

NeutralizationIC50 [nM]

>100 / n.n.

Donor 1 Donor 2

G HDays post-vaccination

Fig. 3. Ab affinity maturation continues for 6 to 9 mo following YFV-17D vaccination. (A) SHM loads of YFV E-specific mAbs isolated from each sampling timepoint. (B) Proportion of YFV E-specific mAbs with the indicated SHM loads. (C) KD

Apps of the YFV E-specific mAbs isolated at each sampling time point, asdetermined by BLI. MAbs that did not display detectable binding to the YFV E protein were excluded from this analysis. (D) Proportion of YFV E-specific mAbsisolated from each sampling time point with the indicated KD

Apps. Avid-only binding indicates mAbs that showed binding to YFV E in an avid orientation butnot in a monovalent orientation. (E) Neutralization IC50s of individual mAbs against YFV-17D. (F) Proportion of MBC-derived mAbs with the indicatedneutralization IC50s against YFV-17D. Shading in E and F is identical. (G) Proportion of YFV E-specific B cells that display CD21lo or CD71+ surface expression, asdetermined by index sorting analysis. (H) Proportion of YFV E-specific activated B cells that express CD71 (CD71+) and/or down-regulate CD21 (CD21lo) at days14, 28, and 90 postvaccination, as determined by index sorting analysis. Statistical comparisons were made using the Mann–Whitney U test (****P < 0.0001,**P < 0.01, *P < 0.05). Black bars indicate medians. n.n., nonneutralizing.

Wec et al. PNAS | March 24, 2020 | vol. 117 | no. 12 | 6679

IMMUNOLO

GYAND

INFLAMMATION

Dow

nloa

ded

by g

uest

on

Oct

ober

29,

202

0

Page 6: Longitudinal dynamics of the human B cell response …the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally

following vaccination (Fig. 3 C and D). In parallel with the in-crease in affinity, we observed the emergence of highly potentnAbs (IC50s <1 nM) beginning at day 90 (Fig. 3 E and F). Thesepotent nAbs were derived from multiple MBC subsets, includingatypical IgM+ and/or IgD+ MBCs (SI Appendix, Fig. S13). In-terestingly, the average binding avidities and neutralization po-tencies of the mAbs isolated from day 14 MBCs were lower thanthose isolated from day 14 PBs, suggesting preferential re-cruitment of high-affinity nAbs into the PB compartment (Figs.1F and 3F and SI Appendix, Fig. S14).Finally, we assessed ongoing B cell activation by analyzing ex-

pression of the B cell activation/proliferation marker CD71 onYFV E-specific MBCs (4). This marker was expressed on 75 to85% YFV E-specific B cells at day 14 and remained elevated for 6to 9 mo in both donors (Fig. 3G). Since down-regulation of CD21

has also been shown to be associated with activation of MBCsfollowing infection or vaccination (4–6), we also monitored CD21expression on YFV E-specific B cells at each sampling time point.In both donors, YFV E-specific CD21lo cells were highly abundanton days 14 and 28 postvaccination, comprising about 40 to 80% ofthe YFV E-specific response, and then declined rapidly by day 90(Fig. 3G). At day 14, about 25% of YFV E-specific CD21lo cellsexpressed IgM BCRs and lacked both CD27 expression and so-matic mutations (SI Appendix, Fig. S15A), suggesting a naïve B cellorigin. In contrast, at late time points (day 90 and onward) almostall of the YFV E-specific CD21lo cells contained somatic muta-tions, supporting a GC origin (SI Appendix, Fig. S15B). Notably,at day 14 postvaccination, there was a high degree of overlapbetween the CD71+ and CD21lo populations, with 50 to 80% ofYFV E-specific activated B cells (defined as CD71+ and/or

D

0

5

10

15

20

25

30

# V H

nt.

subs

titut

ions

Day 14 28 90 180 270 360 14 28 90 180 270 360

Donor 1 Donor 2

VH3-72 All other germlines

E

VL2-14VL3-25

VL3-1VL2-11VL2-8

VK1-9VK3-15

VL3-10

VL3-9VK1-27

87

VK1-33

VL2-23

Donor 1

Donor 2

VK3-11

VK1-39VK1-5

VK1D-12VL1-47VL1-51VL9-49

110

5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32

05

10152025

CDR H3 length (amino acids)

% m

Abs

5 6 7 8 9 10 1 1 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32

05

10152025

CDR H3 length (amino acids)

All other germlinesVH3-72

Unselected repertoires

Donor 1 Donor 2

A B

C

10-10

10-9

10-8

10-7

10-6

14 28 90 180 270 360

YFV

E K D

App [

M]

14 28 90 180 270 360

Donor 1 Donor 2

VH3-72 All other germlines Avid affinity

Day

** * * * * ** ***

Unselected

D14 MBCD28 MBCD90 MBC

D180 MBCD270 MBCD360 MBC

VH1-18

VH1-2VH1-3

VH1-46

VH1-69

VH1-8

VH2-26

VH2-5

VH2-70

VH3-11

VH3-13

VH3-15

VH3-20

VH3-21

VH3-23

VH3-30

VH3-30-3

VH3-33

VH3-43D

VH3-48

VH3-49

VH3-64

VH3-66

VH3-7

VH3-72

VH3-74

VH3-9

VH4-30-2

VH4-30-4

VH4-31

VH4-34

VH4-38-2

VH4-39

VH4-4

VH4-59

VH4-61

VH5-51

VH6-1

VH7-4-1

D14 MBCD30 MBCD90 MBC

D180 MBCD270 MBCD360 MBC

%Repertoire

Donor 1

Donor 2

051015202530354045

Fig. 4. YFV E-specific mAbs show preferential usage of the VH3-72 germline gene. (A) VH germline gene usage of YFV E-specific mAbs isolated from eachsampling time point. VH germline gene frequencies of unselected human MBCs (Unselected) were obtained from multiple high-throughput sequencingstudies and shown for comparison (46). (B) VL germline gene usage of mAbs utilizing the VH3-72 germline gene. The numbers in the center of the pies denotethe total number of VH3-72 mAbs. (C) CDR H3 length distribution of YFV E-specific mAbs utilizing the VH3-72 germline gene, mAbs utilizing all other VH

germline genes, or unselected Abs from MBCs (46). (D) KDApps of mAbs utilizing the VH3-72 germline gene or all other VH germline genes, as determined by

BLI. Avid KDApps are plotted for mAbs isolated from day 14 MBCs because only a small subset of these mAbs showed detectable binding to YFV E in a

monovalent orientation. (E) SHM loads of YFV E-specific mAbs utilizing the VH3-72 germline gene or all other VH germline genes. Black bars indicate medians.Statistical comparisons were made using the Mann–Whitney U test (***P < 0.001, **P < 0.01, *P < 0.05).

6680 | www.pnas.org/cgi/doi/10.1073/pnas.1921388117 Wec et al.

Dow

nloa

ded

by g

uest

on

Oct

ober

29,

202

0

Page 7: Longitudinal dynamics of the human B cell response …the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally

CD21lo) displaying a CD71+CD21lo phenotype (Fig. 3H). How-ever, by days 28 to 90, the CD71+CD21lo population wanedto <20% of the activated B cell response in both donors (Fig. 3H).At these time points, the majority of YFV E-specific activated Bcells displayed either a CD71+CD21+ or CD71−CD21lo phenotypeand were heterogenous with respect to isotype and CD27 ex-pression, demonstrating that these B cells belong to multipledistinct MBC populations (Figs. 3H and 2C). Overall, the resultsindicate that the YFV E-specific MBC response continues toevolve for 6 to 9 mo following primary vaccination, suggesting thatGC activity persists for many months following the period of activeviral replication in peripheral blood (22).

The Majority of MBC-Derived mAbs Target Epitopes within orProximal to the Fusion Loop on DII of the YFV E Protein. We nextsought to investigate Ab immunodominance hierarchy in the Bcell response to YFV-17D. As a first step, we analyzed thegermline gene usage of the isolated mAbs at each sampling timepoint. Strikingly, in both donors, mAbs utilizing the VH3-72germline gene dominated the response at all time points (Fig.4A). A large fraction of these mAbs also utilized one of fivedominant light-chain germline genes and displayed shorter-than-average heavy-chain complementary determining region 3 (CDRH3)lengths, suggesting a shared mode of antigen recognition (Fig. 4

B and C). Interestingly, the KDApps of the mAbs utilizing VH3-72

were significantly lower than those observed for mAbs utilizingother VH germlines, despite containing similar levels of SHM(Fig. 4 D and E).To define the number of distinct antigenic sites targeted by the

MBC-derived mAbs, we next performed pairwise competition ex-periments using the newly isolated mAbs and two well-characterizedcontrol mAbs, 4G2 and 5A, which recognize proximal but non-overlapping epitopes within DII of the YFV E monomer (Fig. 5Aand SI Appendix, Fig. S16); 4G2 is a pan-flavivirus mAb that targetsthe fusion loop (FL) (23), whereas 5A is a YFV E-specific mAb thatbinds to a FL-proximal epitope overlapping the proposed prM as-sociation region (Fig. 5A) (13). Competition experiments were per-formed using high-throughput surface plasmon resonance on aCarterra LSA instrument (24). In addition, we tested the mAbs forreactivity with a recombinant YFV-17D DIII protein by BLI.The majority of mAbs recognized one of eight distinct anti-

genic sites, which were defined based on reactivity with DIII andcompetition with 4G2, 5A, and three of the newly isolated mAbs(ADI-49147, ADI-44112, and ADI-45107) (Fig. 5B and DatasetS1). Consistent with previous studies of human sera (12), only asmall subset of mAbs (6 of 772) recognized epitopes within DIII(Fig. 5B and Dataset S1). In contrast, over half of the mAbs fromboth donors competed with 4G2 and/or 5A, suggesting that the

5A scFvFusion peptide 4G2 binding site

DII

DIII

DI

AVK1-27VK1-33

VK1-39

VK1-5

VK1-9

VK3-11

VL2-11

VL2-14

VL2-23VL2-8VL3-1

VL3-25

Donor 1

5A4G2 / 5A4G2ADI-44112Other

1

30

20

10

40

50

60

1

30

20

10

40

50

60

70

80

VK1-33

VK1-39

VK1-5

VK3-11

VL1-47VL2-11

VL2-14

VL2-8

VL3-1

VL3-10

VL3-25

Donor 2

B Donor 1 Donor 2

Day post-vaccination14 28 9018

027

036

00

25

50

75

100

%m

Abs

14 28 90 180

270

360

5A / ADI-451075A4G2 / 5A4G2ADI-44112

DIII;ADI-49147 competitor

DIII; otherOther

ADI-45107

D

VH3-72Other

4G2 competitors

250

C

E

YFV

E K D

App [

M]

10-10

10-9

10-8

10-7

10-6

VH3-72

other

**

Fig. 5. Abs targeting epitopes within or proximal to the FL dominate the MBC response to YFV-17D vaccination. (A) Crystal structure of a soluble YFV E dimerin complex with scFv 5A (13) (Protein Data Bank ID code 6IW2), with the monomers comprising the antiparallel YFV E dimer represented as ribbon or space-filling model. Domains I (red), II (yellow), and III (blue) within the individual monomer unit are colored. The location of the fusion peptide, the site of 4G2binding, is labeled in cyan (23). (B) Proportion of mAbs within each competition group at each sampling time point. (C) MAbs utilizing the VH3-72 germlinegene are shaded according to their competition group; light-chain germline genes are indicated on the right. (D) Proportion of 4G2-competitor mAbs thatutilize the VH3-72 germline gene. (E) KD

Apps of 4G2-competitor mAbs that either use the VH3-72 germline gene or all other germline genes. Statisticalcomparisons were made using the Mann–Whitney U test (**P < 0.01). scFv, single-chain variable fragment.

Wec et al. PNAS | March 24, 2020 | vol. 117 | no. 12 | 6681

IMMUNOLO

GYAND

INFLAMMATION

Dow

nloa

ded

by g

uest

on

Oct

ober

29,

202

0

Page 8: Longitudinal dynamics of the human B cell response …the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally

majority of the YFV E-specific response is mediated by Abs thattarget epitopes within or proximal to the FL on DII (Fig. 5B).Interestingly, nearly all of the mAbs that utilized the VH3-72germline gene competed with 4G2, suggesting recognition ofthe FL (Fig. 5C). Correspondingly, analysis of the sequencefeatures of the mAbs clustered by competition group revealedthat over half of the mAbs that competed with 4G2 utilized theVH3-72 germline gene (Fig. 5D). Consistent with our previousobservation (Fig. 4E), the 4G2 competitor mAbs utilizing VH3-72 showed significantly higher affinities compared to those uti-lizing other VH germline genes (Fig. 5E). Finally, although theproportion of mAbs targeting each antigenic site did not changedramatically over time, we observed a suppression of 4G2/5Acompetitor mAbs at later time points in donor 1 (days 270 and360) (Fig. 5B). Furthermore, in both donors, mAbs that com-peted with both 5A and ADI-45107 did not emerge until days 28to 90 (Fig. 5B). Altogether, the results suggest that the vastmajority of the YFV E-specific response is directed againstepitopes within or proximal to the FL on domain II and thatthere are only minor shifts in Ab immunodominance hierarchyduring the maturation of the B cell response to YFV-17D.

The Majority of Highly Potent nAbs Recognize FL-Proximal Epitopes.We next investigated the relationship between antigenic site andneutralization potency. Strikingly, over 90% of the mAbs thatcompeted either with 5A only or both 5A and ADI-45107showed neutralizing activity (Fig. 6A). Moreover, the majority(78%) of highly potent nAbs (IC50 <1 nM) in the panel belongedto these two competition groups (Fig. 6 B and C). Analysis of thesequence features of 5A-only or 5A/ADI-45107 competitor nAbsrevealed that nearly 40% utilized VH4-4/VL1-51 germline genepairing and did not show evidence of a convergent CDRH3 se-quence, suggesting a common mode of germline-encoded anti-gen recognition (Fig. 6D and SI Appendix, Table S2). In line withprior studies of other flaviviruses (25), most of the DIII-directedmAbs also showed highly potent neutralizing activity (Fig. 6 Aand B). In contrast to the 5A competitors and DIII-directedmAbs, only a minority of the mAbs belonging to other compe-tition groups showed neutralizing activity (Fig. 6 A and B). Forexample, only 12% and 20% of mAbs that competed with 4G2only or both 4G2 and 5A, respectively, displayed neutralizationIC50s <100 nM (Fig. 6 A and B). Overall, the results demonstratethat, in these two donors, the nAb response to YFV-17D isprimarily mediated by Abs that recognize FL-proximal epitopeswithin DII of the YFV E protein.

A Subset of mAbs Display Binding Cross-Reactivity with OtherFlavivirus E Proteins. Since human flavivirus infections areknown to induce flavivirus cross-reactive Ab responses (26–28),we next evaluated whether any of the isolated mAbs displayedbinding reactivity to recombinant DENV-2, DENV-4, WNV, orZIKV E proteins. In both donors, about 6% of YFV E-reactivemAbs showed cross-reactivity to at least one heterologous fla-vivirus E protein (Fig. 7A). The majority of these cross-reactivemAbs targeted the highly conserved FL and bound to all fiveflavivirus E proteins with high apparent affinities (KD

Apps <10 nM)(Fig. 7 B and C). Correspondingly, the small subset of mAbsthat bound to epitopes outside of the FL generally displayedmore limited cross-reactivity profiles and lower affinities (Fig.7C). Importantly, only 6 out of 50 cross-binding mAbs showedneutralizing activity against YFV-17D, and none of these mAbsshowed significant cross-neutralizing activity against ZIKV,WNV, or JEV (Fig. 7 and SI Appendix, Fig. S17). We concludethat YFV-17D vaccination induces a subset of Abs that displaybroad flavivirus binding activity, the majority of which target thehighly conserved FL and show little to no cross-neutralizing activity.

DiscussionA deep understanding of the kinetics, dynamics, and specificitiesof human MBC responses elicited by successful vaccines mayprovide insights into the design of vaccines for refractory path-ogens. To dissect the antigen-specific MBC response induced bythe YFV-17D vaccine—widely considered to be the “gold-standard” of antiviral vaccination—we cloned and character-ized hundreds of YFV E-specific mAbs from two healthy donorsat multiple time points following primary YFV-17D vaccination.In most individuals, YFV serum neutralizing activity appears

∼10 to 14 d following primary YFV-17D vaccination (29, 30),which corresponds to the peak PB response and suggests thepresence of nAbs in this compartment. Indeed, we found thatabout one-third of the YFV E-specific mAbs isolated from day14 PBs showed neutralizing activity. The PB-derived nAbs orig-inated from both mutated and unmutated cells, suggesting thatboth activated naïve and MBCs contribute to the nAb responseto YFV-17D. Interestingly, the average binding avidities andneutralization potencies of the mAbs isolated from day 14 PBswere higher than those isolated from day 14 MBCs, which isconsistent with previous studies in mouse models showing thathigher-affinity cells are preferentially recruited into the PBcompartment (31, 32). Hence, the immune system appears toplace a priority on the rapid production of high-affinity serum

4G2

5A

A B

C

D

0

25

50

75

100

%m

Abs

4G2

4G2 /

5A 5A

5A / A

DI-451

07

ADI-451

07

DIII bin

der

ADI-441

12Othe

r0.001

0.01

0.1

1

10

100

1000

Neu

traliz

atio

n IC

50 [n

M]

4G2

4G2 /

5A 5A

5A / A

DI-451

07

ADI-451

07

DIII bin

der

ADI-441

12Othe

r

5A and ADI-45107

45 DIII; ADI-49147 competitor

4G2 and 5A4G2Other

5A

ADI-45107

mAbs with IC50<1 nM

< 11 to 10>10 to 100 >100 / n.n.

Neutralization IC50 [nM]

VK1-12

VK1-39VK1-5

VK2-28

VK3-11

VK3-15

VK3-20VK4-1

VL1-51

VL2-11

VL2-14

VL2-8

VL3-21

VL3-25

VH3-11VH3-23VH3-30VH3-33

VH3-7VH3-72

VH3-9VH4-4

VH4-59VH4-61

0

10

20

30

% nAbs 38

Fig. 6. The majority of highly potent nAbs recognize FL-proximal epitopes.(A) Proportion of mAbs in each competition group with the indicated neu-tralization IC50s against YFV-17D. (B) Neutralization IC50s of individual mAbsagainst YFV-17D across the indicated competition groups. Black bars indicatemedians. IC50 values for 4G2 and 5A mAbs are indicated by horizontal lines.(C) Antigenic sites targeted by highly potent nAbs (IC50 <1 nM). The numberin the center of the pie indicates the total number of highly potent nAbs. (D)VH and VL germline gene usage of 5A-only and 5A/ADI-45107 competitornAbs.

6682 | www.pnas.org/cgi/doi/10.1073/pnas.1921388117 Wec et al.

Dow

nloa

ded

by g

uest

on

Oct

ober

29,

202

0

Page 9: Longitudinal dynamics of the human B cell response …the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally

nAbs, which likely provides the basis for the early protectionafforded by the YFV-17D vaccine.Previous studies in mouse models have demonstrated that

antigen-specific IgM+ MBCs are induced early after immuniza-tion and remain numerically stable over time (8–10). In contrast,the longevity of swIg+ MBCs has been suggested to be dependenton the frequency of high-affinity germline-encoded precursors inthe naïve B cell repertoire, with the presence of high-affinitynaïve precursors associated with unstable swIg+ MBCs (8, 9).Interestingly, despite the presence of germline-encoded BCRswith high avidity for YFV E in both donors, we observed thatclassical IgM+ MBCs waned rapidly over time, whereas swIg+

MBCs remained stable until at least day 360 postvaccination.Although the results from these two vaccinated donors may notbe representative of the entire YFV-17D–vaccinated pop-ulation, we also observed transient IgM+ MBCs and long-livedswIg+ MBCs in the context of PUUV infection and others havereported long-term maintenance of swIg+ MBCs followingsmallpox vaccination (33), suggesting that primary viral infec-tions commonly induce a stable swIg+ MBC population. Thediscrepancy between our results and those observed in mousemodels may reflect differences between the hierarchy ofIgM+ versus swIg+ MBC response in mice and humans and/ordifferences in the response to replicating versus nonreplicatingantigens.We observed increases in SHM, Ab affinity, and neutralization

potency for 6 to 9 mo following YFV-17D vaccination, suggest-ing that GC activity continues for many months following theperiod of detectable viral replication in peripheral blood. Thiscould either be due to low level viral replication in secondarylymphoid organs or prolonged capture of inert viral antigens on

the surface of follicular dendritic cells (FDCs). Although wecannot distinguish between these two possibilities, previousstudies have shown that viral antigens, in the form of complement-coated immune complexes, can be sequestered by FDCs for ex-tended periods of time for presentation to GC B cells (34–37).Importantly, recent studies have also shown that MBCs induced byEbola virus infection and influenza virus vaccination continue toundergo affinity maturation for 6 to 12 mo (6, 38, 39), suggestingthat prolonged maturation of the MBC response may be morecommon than previously appreciated. Given that Ab responsesinduced by live viral infections have been shown to have half-lives of 50 y or more, whereas responses to nonreplicatingprotein antigens wane relatively quickly and require boostervaccinations (40), it will be of interest to investigate whetherprotracted affinity maturation is also observed for vaccines that donot induce long-lived Ab responses (e.g., the tetanus toxoid anddiphtheria vaccines).In both donors the nAb response induced by YFV-17D vac-

cination was primarily directed against FL-proximal epitopesoverlapping the 5A binding site. Interestingly, a substantialfraction of these “5A-class” nAbs displayed convergent VH/VLgermline gene pairing, suggestive of a so-called public Ab re-sponse (41). The relatively high abundance of these two germlinegenes (VH4-4 and VL1-51) in the naïve B cell repertoire perhapsprovides a molecular explanation for how the YFV-17D vaccineis able to induce protective neutralizing Ab responses in such anextraordinarily high proportion of individuals (>95%) (42, 43).Furthermore, a subset of these nAbs displayed exceptionallypotent neutralizing activity, with IC50s that were about 10 timeslower than previously described YFV mAbs (13, 14). Given therecent YFV outbreaks in Brazil and the Democratic Republic of

ADI-450

97

ADI-441

55

ADI-422

35

ADI-491

56

ADI-421

19

ADI-422

03

ADI-441

44

ADI-427

75

ADI-422

18

ADI-422

00

ADI-441

33

ADI-422

39

ADI-490

25

ADI-422

19

ADI-421

80

ADI-441

48

ADI-502

14

ADI-491

97

ADI-422

20

ADI-491

58

ADI-428

02

ADI-489

67

ADI-421

83

ADI-441

54

ADI-421

82

ADI-441

27

ADI-491

72

ADI-441

20

ADI-491

37

ADI-422

22

ADI-502

23

ADI-502

21

ADI-491

80

ADI-491

78

ADI-502

26

ADI-491

59

ADI-427

98

ADI-502

28

ADI-441

23

ADI-450

86

ADI-489

50

ADI-465

93

ADI-489

05

ADI-488

98

ADI-441

11

ADI-451

11

ADI-422

42

ADI-421

95

ADI-421

76

ADI-491

04

ZIKVDENV-2DENV-4

WNVYFV

4G2 / 5A 4G2 ADI-441

12

Other5A DIII

YFVZIKVWNVJEV

KDApp [nM]

IC50 [nM]

N.B.

<11 to 10>10 to 100>100 to 500

>100 / n.n.

<11 to 10>10 to 100

Competition group# of E proteins

recognized

339 45154321 (YFV only)

50

4G2

DIII; ADI-49147ADI-44112

5A4G2/5A

Other

A B

C

Donor 1 Donor 2

Fig. 7. A subset of mAbs show broad flavivirus cross-reactivity. (A) Proportion of mAbs that react with one or more of the flavivirus E proteins tested (YFV,DENV-1, DENV-2, ZIKV, and WNV). Numbers in the center of the pies indicate the number of mAbs analyzed. (B) Proportion of cross-reactive mAbs thatrecognize the indicated antigenic sites. (C, Top) Heat map shows the binding cross-reactivity profiles of 50 mAbs that showed binding reactivity to at least oneflavivirus E protein in addition to YFV E. (C, Bottom) Heat map shows the neutralizing activities of the cross-binding mAbs against YFV-17D, ZIKV, WNV, andJEV. Competition group assignments for the individual mAbs are indicated at the top of the heat map. N.B., nonbinding; n.n., nonneutralizing.

Wec et al. PNAS | March 24, 2020 | vol. 117 | no. 12 | 6683

IMMUNOLO

GYAND

INFLAMMATION

Dow

nloa

ded

by g

uest

on

Oct

ober

29,

202

0

Page 10: Longitudinal dynamics of the human B cell response …the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally

Congo, coupled with YFV-17D vaccine supply shortages and thelack of effective treatments for YFV disease, these mAbs rep-resent promising candidates for prophylaxis and/or therapy.Interestingly, about 6% of Abs induced by YFV-17D vacci-

nation showed binding reactivity to heterologous flavivirus Eproteins. As expected based on prior studies of flavivirus in-fection (26–28), we found that the majority of these mAbs tar-geted the highly conserved FL and lacked measurable cross-neutralizing activity. Although nonneutralizing Abs are unlikelyto contribute to protection or Ab-dependent enhancement ofsecondary flavivirus infections (44), the corresponding cross-reactive B cells may be preferentially activated and expandedupon subsequent flavivirus infection. Thus, it will be of interestto study if and how preexisting YFV immunity impacts the B cellresponse to heterologous flavivirus exposure and vice versa.Altogether, the results of this study provide a comprehensive

view of the dynamics and complexity of the human MBC re-sponse induced by the highly successful YFV-17D vaccine.Similar studies performed on other vaccines and natural infec-tions may shed light on features of the MBC response associatedwith the induction and maintenance of long-term protective Abresponses.

Materials and MethodsHuman Subjects. Informed consent to participate in this study was obtainedbefore vaccination. Study subjects aged between 25 and 32 y were vacci-nated with the YFV-17D Stamaril vaccine. Heparinized blood (50 to 100 cc)was obtained from subjects before vaccination and on days 10, 14, 28, 90,180, 270, and 360 following vaccination. Samples were processed in theImmune Monitoring and Flow Cytometry Core laboratory at the GeiselSchool of Medicine at Dartmouth College to obtain plasma and to isolateperipheral blood-derived B cells. Isolated cells and plasma were stored fro-zen in aliquots at −80 °C. This study complies with all relevant ethicalregulations for work with human participants and was approved by the

Committee for the Protection of Human Subjects, Dartmouth-HitchcockMedical Center, and Dartmouth College.

Blood samples from the two PUUV-experienced donors were obtainedon days 20, 35, 82, 130, 161, 237, and 349 postdiagnosis. Both donors werehospitalized due to PUUV-associated disease. Samples were processed at theUniversity Hospital of Northern Sweden to obtain plasma and to isolateperipheral blood-derived B cells. The two subjects were 43 (donor 165) and 75(donor 192) y of age at the time of hospitalization. Ethical approval wasobtained by the regional Ethical Review Board at Umea ̊ University, Umea ̊,Sweden. Signed informed consent was obtained from both donors.

Single B Cell Sorting and Variable Gene Amplification.MBCs were stained usinganti-human CD19, CD20, CD3, CD8, CD14, CD16, IgD, IgM, CD27, CD21, CD71,and a mixture of dual-labeled (APC and PE) YFV E tetramers. B cells thatshowed reactivity to the YFV E tetramers were single-cell-sorted for mAbcloning, as described previously (45).

Microtiter Neutralization Assays. Monoclonal antibodies were serially dilutedin cell culture medium and incubated at room temperature with the testviruses (YFV-17D or ZIKV) or test reporter virus particles (RVPs) (WNV or JEV)for 1 h then added to cells. Infection levels were measured 2 d later bystaining infected cells with the pan-flavivirus mouse mAb 4G2 on a Cytation-5 automated fluorescence microscope (BioTek).

Data Availability. All data are included in the manuscript and SI Appendix. Allreagent and data requests should be directed to L.M.W. The sequences ofneutralizing antibodies isolated in this study have been deposited in theGenBank database, https://www.ncbi.nlm.nih.gov/genbank (accession nos.MN993290–MN993593).

ACKNOWLEDGMENTS. We thank Dr. Ted Pierson for his generous gift of theflavivirus RVP system and Alan Bergeron from the Dartmouth Immune Mon-itoring and Flow Cytometry Core laboratory for carrying out human periph-eral blood mononuclear cell processing and banking. This work wassupported by the NIH grant NIAID R01 AI134824 (to K.C.) and by the trans-national Agence Nationale de la Recherche/Deutsche Forschungsgemein-schaft grant FlavImmunity/ANR-17-CE15-0031-01 (to G.B.-S.).

1. L. Mesin, J. Ersching, G. D. Victora, Germinal center B cell dynamics. Immunity 45,471–482 (2016).

2. F. Weisel, M. Shlomchik, Memory B cells of mice and humans. Annu. Rev. Immunol. 35,255–284 (2017).

3. J. J. Taylor, M. K. Jenkins, K. A. Pape, Heterogeneity in the differentiation andfunction of memory B cells. Trends Immunol. 33, 590–597 (2012).

4. A. H. Ellebedy et al., Defining antigen-specific plasmablast and memory B cell subsetsin human blood after viral infection or vaccination. Nat. Immunol. 17, 1226–1234(2016).

5. D. Lau et al., Low CD21 expression defines a population of recent germinal centergraduates primed for plasma cell differentiation. Sci. Immunol. 2, eaai8153 (2017).

6. S. F. Andrews et al., Activation dynamics and immunoglobulin evolution of pre-existing and newly generated human memory B cell responses to influenza hemag-glutinin. Immunity 51, 398–410.e5 (2019).

7. G. V. Zuccarino-Catania et al., CD80 and PD-L2 define functionally distinct memory Bcell subsets that are independent of antibody isotype. Nat. Immunol. 15, 631–637(2014).

8. K. A. Pape et al., Naive B cells with high-avidity germline-encoded antigen receptorsproduce persistent IgM(+) and transient IgG(+) memory B cells. Immunity 48, 1135–1143 e4 (2018).

9. K. A. Pape, J. J. Taylor, R. W. Maul, P. J. Gearhart, M. K. Jenkins, Different B cellpopulations mediate early and late memory during an endogenous immune re-sponse. Science 331, 1203–1207 (2011).

10. I. Dogan et al., Multiple layers of B cell memory with different effector functions. Nat.Immunol. 10, 1292–1299 (2009).

11. R. A. Mason, N. M. Tauraso, R. O. Spertzel, R. K. Ginn, Yellow fever vaccine: Direct challengeof monkeys given graded doses of 17D vaccine. Appl. Microbiol. 25, 539–544 (1973).

12. O. Vratskikh et al., Dissection of antibody specificities induced by yellow fever vac-cination. PLoS Pathog. 9, e1003458 (2013).

13. X. Lu et al., Double lock of a human neutralizing and protective monoclonal antibodytargeting the yellow fever virus envelope. Cell Rep. 26, 438–446 e5 (2019).

14. S. Daffis et al., Antibody responses against wild-type yellow fever virus and the 17Dvaccine strain: Characterization with human monoclonal antibody fragments andneutralization escape variants. Virology 337, 262–272 (2005).

15. R. W. Wieten et al., A single 17D yellow fever vaccination provides lifelong immunity;characterization of yellow-fever-specific neutralizing antibody and T-cell responsesafter vaccination. PLoS One 11, e0149871 (2016).

16. N. P. Lindsey et al., Persistence of yellow fever virus-specific neutralizing antibodiesafter vaccination among US travellers. J. Travel Med. 25, tay108 (2018).

17. S. Kohler et al., The early cellular signatures of protective immunity induced by liveviral vaccination. Eur. J. Immunol. 42, 2363–2373 (2012).

18. M. S. Gilman et al., Rapid profiling of RSV antibody repertoires from the memory B

cells of naturally infected adult donors. Sci. Immunol. 1, eaaj1879 (2016).19. R. Di Niro et al., Salmonella infection drives promiscuous B cell activation followed by

extrafollicular affinity maturation. Immunity 43, 120–131 (2015).20. W. Dejnirattisai et al., A new class of highly potent, broadly neutralizing antibodies

isolated from viremic patients infected with dengue virus. Nat. Immunol. 16, 170–177

(2015).21. J. Wrammert et al., Rapid cloning of high-affinity human monoclonal antibodies

against influenza virus. Nature 453, 667–671 (2008).22. B. Reinhardt, R. Jaspert, M. Niedrig, C. Kostner, J. L’age-Stehr, Development of vire-

mia and humoral and cellular parameters of immune activation after vaccination with

yellow fever virus strain 17D: A model of human flavivirus infection. J. Med. Virol. 56,

159–167 (1998).23. W. D. Crill, G. J. Chang, Localization and characterization of flavivirus envelope gly-

coprotein cross-reactive epitopes. J. Virol. 78, 13975–13986 (2004).24. A. Sivasubramanian et al., Broad epitope coverage of a human in vitro antibody li-

brary. MAbs 9, 29–42 (2017).25. E. N. Gallichotte et al., Role of Zika virus envelope protein domain III as a target of

human neutralizing antibodies. MBio 10, e01485-19 (2019).26. C. H. Calisher et al., Antigenic relationships between flaviviruses as determined by

cross-neutralization tests with polyclonal antisera. J. Gen. Virol. 70, 37–43 (1989).27. K. L. Mansfield et al., Flavivirus-induced antibody cross-reactivity. J. Gen. Virol. 92,

2821–2829 (2011).28. L. Priyamvada et al., Human antibody responses after dengue virus infection are

highly cross-reactive to Zika virus. Proc. Natl. Acad. Sci. U.S.A. 113, 7852–7857

(2016).29. T. P. Monath et al., Comparative safety and immunogenicity of two yellow fever 17D

vaccines (ARILVAX and YF-VAX) in a phase III multicenter, double-blind clinical trial.

Am. J. Trop. Med. Hyg. 66, 533–541 (2002).30. J. Lang et al., Comparison of the immunogenicity and safety of two 17D yellow fever

vaccines. Am. J. Trop. Med. Hyg. 60, 1045–1050 (1999).31. J. J. Taylor, K. A. Pape, H. R. Steach, M. K. Jenkins, Humoral immunity. Apoptosis and

antigen affinity limit effector cell differentiation of a single naïve B cell. Science 347,

784–787 (2015).32. D. Paus et al., Antigen recognition strength regulates the choice between extra-

follicular plasma cell and germinal center B cell differentiation. J. Exp. Med. 203,

1081–1091 (2006).33. S. Crotty et al., Cutting edge: Long-term B cell memory in humans after smallpox

vaccination. J. Immunol. 171, 4969–4973 (2003).

6684 | www.pnas.org/cgi/doi/10.1073/pnas.1921388117 Wec et al.

Dow

nloa

ded

by g

uest

on

Oct

ober

29,

202

0

Page 11: Longitudinal dynamics of the human B cell response …the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally

34. B. A. Heesters et al., Endocytosis and recycling of immune complexes by folliculardendritic cells enhances B cell antigen binding and activation. Immunity 38, 1164–1175 (2013).

35. B. A. Smith et al., Persistence of infectious HIV on follicular dendritic cells. J. Immunol.166, 690–696 (2001).

36. B. A. Heesters, R. C. Myers, M. C. Carroll, Follicular dendritic cells: Dynamic antigenlibraries. Nat. Rev. Immunol. 14, 495–504 (2014).

37. J. G. Tew, T. E. Mandel, Prolonged antigen half-life in the lymphoid follicles ofspecifically immunized mice. Immunology 37, 69–76 (1979).

38. C. W. Davis et al., Longitudinal analysis of the human B cell response to Ebola virusinfection. Cell 177, 1566–1582 e17 (2019).

39. K. Matsuda et al, Prolonged evolution of the memory B cell response inducedby a replicating adenovirus-influenza H5 vaccine. Sci. Immunol. 4, eaau2710(2019).

40. I. J. Amanna, N. E. Carlson, M. K. Slifka, Duration of humoral immunity to commonviral and vaccine antigens. N. Engl. J. Med. 357, 1903–1915 (2007).

41. A. Lanzavecchia, Dissecting human antibody responses: Useful, basic and surprising

findings. EMBO Mol. Med. 10, e8879 (2018).42. B. Briney, A. Inderbitzin, C. Joyce, D. R. Burton, Commonality despite excep-

tional diversity in the baseline human antibody repertoire. Nature 566, 393–397

(2019).43. N. D. Collins, A. D. Barrett, Live attenuated yellow fever 17D vaccine: A legacy vaccine

still controlling outbreaks in modern day. Curr. Infect. Dis. Rep. 19, 14 (2017).44. M. J. Luppe et al., Yellow fever (YF) vaccination does not increase dengue severity:

A retrospective study based on 11,448 dengue notifications in a YF and dengue

endemic region. Travel Med. Infect. Dis. 30, 25–31 (2019).45. T. Tiller et al., Efficient generation of monoclonal antibodies from single human B

cells by single cell RT-PCR and expression vector cloning. J. Immunol. Methods 329,

112–124 (2008).46. A. Kovaltsuk et al., Observed antibody space: A resource for data mining next-

generation sequencing of antibody repertoires. J. Immunol. 201, 2502–2509 (2018).

Wec et al. PNAS | March 24, 2020 | vol. 117 | no. 12 | 6685

IMMUNOLO

GYAND

INFLAMMATION

Dow

nloa

ded

by g

uest

on

Oct

ober

29,

202

0