Paul Chaplin’s research while affiliated with Bavarian Nordic and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (116)


Abstract LB223: Intratumoral administration of modified vaccinia Ankara expressing a tumor associated antigen and the costimulatory molecules IL-12 and 4-1BBL induces potent systemic long-term immune responses against murine tumors
  • Article

April 2023

·

30 Reads

Cancer Research

Cigdem Atay Langbein

·

·

Mahmad Ismail Housni

·

[...]

·

Maria Hinterberger

In the last decade, immunotherapy has revolutionized cancer treatment. However, the lack of potent therapy-induced immune responses against solid tumors due to the immune-suppressive tumor microenvironment (TME) is still a major drawback. One approach to reprogram TME is virotherapy, which can target tumors via destruction of infected tumor cells as well as activating immune cell populations in the TME. A versatile and safe tool for virotherapy is modified vaccinia virus Ankara (MVA), which has already been approved by the FDA as a non-replicating vaccine against smallpox and monkeypox and is also in clinical trials against other infectious diseases and cancer. In the current study, we deciphered the anti-tumorigenic effect of a novel MVA vector encoding a tumor associated antigen (TAA) and the costimulatory molecules IL-12 and 4-1BBL, called MVA-TAA-4-1BBL-IL12. We could show that repetitive intratumoral (IT) injection of MVA-TAA-4-1BBL-IL12 resulted in strong tumor growth control, shrinkage and/or complete elimination of tumors and induction of tumor-specific CD8 T cell response. Furthermore, MVA-TAA-4-1BBL-IL12 treatment promoted a systemic anti-tumor immune response not only against the treated tumor but also untreated, distant tumors. Importantly, local virotherapy also resulted in the generation of tumor-specific memory response, which protected mice against local recurrence after rechallenge. As a conclusion, our findings show that MVA-TAA-4-1BBL-IL12 IT treatment induces an effective anti-tumorigenic immune response in different solid tumor models, which gives rise to long-term protection against cancer recurrence. Citation Format: Cigdem Atay Langbein, Raphael Giessel, Mahmad Ismail Housni, Kerstin Lämmermann, Barbara Bathke, André Riedl, Jürgen Hausmann, Markus Kalla, Paul Chaplin, Hubertus Hochrein, José Medina-Echeverz, Maria Hinterberger. Intratumoral administration of modified vaccinia Ankara expressing a tumor associated antigen and the costimulatory molecules IL-12 and 4-1BBL induces potent systemic long-term immune responses against murine tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr LB223.


Characterization of transgenes contained in TAEK-VAC-HerBy. (a) Schematic map of TAEK-VAC-HerBy showing the transgenes in the Intergenic Region (IGR) 88/89 of the MVA-BN genome. Expression of transgenes is controlled by different poxvirus promoters; (b) Transgene expression detected by flow cytometry. Briefly, HeLa cells were infected overnight either with mock or with TVH at a MOI of 4. Expression of vaccinia, HER2, Brachyury and CD40L (blue histogram) against mock control (red histogram) is shown; (c) Analysis of subcellular localization of Brachyury-ΔNLS and Brachyury-NLS by immunofluorescence staining. A31 cells were either mock infected or infected at a MOI of 2 with MVA-BN, MVA-Brachyury or TVH. At 17 h post infection, cells were fixed, permeabilized, and stained with anti-Brachyury antibody followed by PE-conjugated secondary antibody. Upper panel represents Brachyury staining, followed by clear field (middle panel) and overlay (lower panel). To better show subcellular localization of Brachyury-ΔNLS or Brachyury-NLS, inlays display higher magnification of representative cells indicated by arrow; (d) Differential binding of HER2 antibodies to mutated HER2 present in TVH. Briefly, CT26.WT cells were infected overnight with TVH at a MOI of 5. Then, infected CT26.WT cells as well as control CT26.HER2 cells were stained with different HER2 antibody clones, namely HER2 24D2, Herceptin® or Perjeta®. Data show HER2 expression detected by Herceptin® and Perjeta® normalized to HER2 24D2-mediated detection. Data shown in B-D are representative of several independent experiments.
TVH activates innate immune cells in vitro. (a, b) Activation and cytokine production of dendritic cells upon TVH infection. Monocyte-derived DCs (mo-DC) were generated after enrichment of CD14⁺ monocytes from human PBMCs and cultured for 7 days in the presence of GM-CSF and IL-4 as explained in Materials and Methods. Mo-DCs were infected overnight either with MVA-BN or TVH at different MOIs. LPS and mock infections were used as positive and negative controls, respectively. Supernatants were then collected, and cells were stained for flow cytometry analysis. (a) Mean Fluorescence Intensity (MFI) of MHC class II (MHC-II), CD86 and CD70 gated on CD1a⁺ DCs; (b) TNF-α, IL-12p70, IL-6 and IL-18 quantification in supernatants. A representative donor out of four tested is shown; (c-e) Cryopreserved PBMCs from the same donor were infected with TVH at a MOI of 0.5 or stimulated with ConA as a positive control. After overnight incubation, supernatants were collected, and cells were either stained for flow cytometry analysis or NK cells were magnetically sorted. Sorted NK cells were incubated with Cell Proliferation Dye (CPD) labelled K562 target cells in a 1:1 ratio (effector:target). (c) Cytokine secretion profile showing IL-6, IL-12p70, IFN-γ and TNF-α; (d) Frequency of CD69 positive cells on CD16⁺CD56low and CD16⁻CD56high NK cells detected by flow cytometry, indicating TVH-induced activity and cytotoxicity of NK cells. (e) Percentage of dead CPD K562 target cells after being incubated with sorted NK cells. Data are expressed as Mean ± SEM and are representative of two independent experiments. One-way ANOVA was performed on figures c-e. ns, non-significant; *, p < 0.05; **p < 0.01; ***p < 0.005; ****p < 0.001.
Brachyury and HER2 expressed in TVH are loaded onto HLA class I molecules. (a, b) TVH infection of THP-1-derived macrophages. THP-1 cells are cultured in the presence of 200 ng/ml PMA for 72 h. Then, cells are washed and rested in complete medium for additional 48 h until the start of the experiment. (a) Phenotype of THP-1-derived macrophages compared to undifferentiated THP-1 cells. Expression of hCD16, hCD14, hCD80, hCD86 and HLA-ABC is detected by flow cytometry. (b) Expression of HER2, Brachyury and CD40L on THP-1-derived macrophages. THP-1-derived macrophages were infected with TVH at Inf.U 4/cell. After overnight incubation, expression of HER2, Brachyury and CD40L was analysed by flow cytometry. (c-d) Analysis of peptides loaded onto HLA-ABC by mass spectrometry, and brief results of ProImmune ProPresent® MAPPS Antigen Presentation assay. Infected THP-1-derived macrophages are lysed, and HLA-peptide molecules recovered in an immune affinity step. Peptides are eluted from the HLA molecules and analysed by LC–MS/MS. Identified sequences are subjected to rigorous analysis to identify true positive peptides with high confidence. (c) Summary table of peptides detected by the ProPresent® Assay in TVH-infected macrophages. The likelihood of peptides to be real identities is described by their Expect Value. Peptides with an Expect Value ≤ 0.05 are indicative of identity; (d) Summary table for the features of TAA-derived peptides MPNPEGRYTF and GLDPNAMYSFL, showing predicted HLA-binding calculated using SYFPEITHI, whether they are listed in the IEDB database, and their binding rank to HLA-B*15:01 and HLA-A*02:01 defined by Net-MHC4.0.
Intravenous injection of TVH into cynomolgus monkeys induces innate immune cell driven cytokine release and activation. TVH administration scheme was defined in Table 1. Briefly, 3 homologous intravenous injections of either TBS, 1 × 10⁹ Inf.U TVH or 6.75 × 10⁹ Inf.U TVH were performed on days 1, 22 and 43. Blood was taken prior to each immunization, 4 h and 24 h after each immunization for cytokine and chemokine analysis. For analysis of innate immune cell subset activation, PBMCs were taken on day 0 and day 2 after the first immunization. (a) Quantification of IL-6, IL-18, IFN-γ, CCL2 and CCL4 in sera at 0 (predose), 4 and 24 h after each immunization by Luminex is indicated as arbitrary units (AU). (b) Frequency of NK cells; (c) Mean Fluorescence Intensity (MFI) of CD69 on NK cells; (d) Percentage of Ki67⁺ NK cells; (e) MFI of CD80 on pDCs; (f) on cDC2-DCs, (g) CD14⁺ monocytes, (h) CD16⁺ monocytes, and (i) CD14⁺ CD16⁺ monocytes are shown for the times only after prime immunization. Data in figures b-i shown for individual animals and as Mean (depicted by columns) ± SEM. Two-way ANOVA was performe to evaluate statistically significant differences. ns, non-significant; *p < 0.05; **p < 0.01; ***p < 0.005; ****p < 0.001.
Induction of HER2- and MVA-specific IgG upon systemic immunization of cynomolgus monkeys using TVH. TVH administration scheme was defined in Table 1. Briefly, 3 homologous intravenous injections of either TBS, 1 × 10⁹ Inf.U TVH or 6.75 × 10⁹ Inf.U TVH were performed on days 1, 22 and 43. Blood was collected prior to the first immunization (predose: Day -12 and on days 22, 43, 46 and 71; day 71 only in Recovery Group; n = 4). Sera were analyzed using (a) MVA-specific IgG ELISA and (b) HER2-specific IgG ELISA. In this graph, IgG titers for individual animals are shown as well as GMTs (Geometric Mean Titers) depicted by horizonal bars +/-SEM. Two-way ANOVA was performed on figures a-b. ns, non-significant; *p < 0.05; **p < 0.01; ***p < 0.005; ****p < 0.001.
Preclinical development of a first-in-class vaccine encoding HER2, Brachyury and CD40L for antibody enhanced tumor eradication
  • Article
  • Full-text available

March 2023

·

129 Reads

·

2 Citations

The induction of antiviral innate immunity by systemic immunization with live virus can be employed to positively impact the response to therapeutic vaccination. We previously demonstrated that systemic immunization with a non-replicating MVA encoding CD40 ligand (CD40L) enhances innate immune cell activation and function, and triggers potent antitumor CD8⁺ T cell responses in different murine tumor models. Antitumor efficacy was increased when combined with tumor targeting antibodies. Here we report the development of TAEK-VAC-HerBy (TVH), a first-in-class human tumor antibody enhanced killing (TAEK) vaccine based on the non-replicating MVA-BN viral vector. It encodes the membrane bound form of human CD40L, HER2 and the transcription factor Brachyury. TVH is designed for therapeutic use in HER2- or Brachyury-expressing cancer patients in combination with tumor targeting antibodies. To preclude possible oncogenic activities in infected cells and to prevent binding of vaccine-encoded HER2 by monoclonal antibodies trastuzumab and pertuzumab, genetic modifications of HER2 were introduced in the vaccine. Brachyury was genetically modified to prevent nuclear localization of the protein thereby inhibiting its transcriptional activity. CD40L encoded in TVH enhanced human leukocyte activation and cytokine secretion in vitro. Lastly, TVH intravenous administration to non-human primates was proven immunogenic and safe in a repeat-dose toxicity study. Nonclinical data presented here highlight TVH as a first-in-class immunotherapeutic vaccine platform currently under clinical investigation.

Download

LB1579. A single and a two-dose regimen of MVA-BN elicit similar memory responses to a boost vaccination two years after the initial regimen, indicating potential flexibility for a delayed second dose of MVA-BN in the ongoing monkeypox emergency

December 2022

·

24 Reads

·

1 Citation

Open Forum Infectious Diseases

Background The ongoing monkeypox outbreak, a WHO-declared PHEIC, requires fast and broad rollout of MVA-BN vaccinations among risk groups. An initial single-dose strategy, followed by a delayed second dose beyond the approved 4-week interval, could help managing tight vaccine resources. Methods Bavarian Nordic has run a study in vaccinia-naïve participants, randomized to receive either 1 MVA-BN vaccination (N = 181) or 2 MVA-BN vaccinations 4 weeks apart (N = 183). Of note, this study also evaluated participants with prior smallpox vaccination, who received 1 MVA-BN boost (N = 200). Subsets of approximately 75 participants of the vaccinia-naïve subjects having received either 1 or 2 doses of the vaccine were administered an MVA-BN boost 2 years later. Neutralizing antibodies (nAb) were measured using Vaccinia PRNT. Results The 1xMVA-BN and 2xMVA-BN groups responded as expected to the primary regimen, with modest increases in nAb GMTs at Week 2 (5.1 and 4.8, respectively) that further increased at Week 4 (7.2 and 7.5). Two weeks after the second primary vaccination (at Week 6) (2xMVA-BN) nAb GMTs peaked (45.6) before stabilizing at Week 8 (34.0). In both the 1x and 2xMVA-BN groups, the nAb levels were near baseline 2 years after primary vaccination. After the MVA-BN boost at 2 years, both groups exhibited rapid anamnestic responses with nAb GMTs that peaked 2 weeks post-boost (80.7 and 125.3), at higher levels than those observed at any timepoint following primary vaccination, remaining elevated at 6 months post-boost (25.6 and 49.3). These anamnestic responses support the presence of durable immunological memory in both groups. No safety concerns were identified and the most common adverse event following a 2-year MVA-BN boost was injection site erythema in 82.2% of participants. Conclusion A single dose of MVA-BN elicits a durable memory immune response for at least 2 years. Similarly, high nAb responses following a boost dose among subjects in the initial 1xMVA-BN or 2xMVA-BN groups indicate that the approved 4-week interval of the two-dose regimen could optionally be extended up to at least 2 years. NCT00316524 and NCT00686582 Disclosures Heinz Weidenthaler, MD, Bavarian Nordic GmbH: Employee and received warrants Florian Lienert, PhD, Bavarian Nordic GmbH: employee Employee and received warrants Daniela Reichhardt, PhD, Bavarian Nordic A/S: employee Employee and received warrants Darja Schmidt, Dr, Bavarian Nordic GmbH: Employee and received warrants Günter Silbernagl, MSc, Bavarian Nordic: Employed|Bavarian Nordic GmbH: employee Employee and received warrants Paul Chaplin, PhD, Bavarian Nordic A/S: employee (CEO)|Bavarian Nordic A/S: Stocks/Bonds.


Figure 2. GMTs Over Time in the Initial and Follow-up Studies
Demographics of Study Participants at Initial and Follow-up Study Baselines
Overview of Related Unsolicited Adverse Events Experienced by > 1 Participant in the Follow-up Study
Single and 2-dose vaccinations with modified vaccinia Ankara-Bavarian Nordic® induce durable B cell memory responses comparable to replicating smallpox vaccines

November 2022

·

24 Reads

·

28 Citations

The Journal of Infectious Diseases

Background Though Modified Vaccinia Ankara - Bavarian Nordic (MVA-BN®). vaccination is approved for smallpox and monkeypox prevention, immunological persistence and booster effects remain undescribed. Methods Participants naïve to smallpox vaccination were randomized to 1 dose MVA-BN (1×MVA, N = 181), 2 doses MVA-BN (2×MVA, N = 183), or placebo (N = 181). Participants with previous smallpox vaccination received 1 MVA-BN booster (HSPX+, N = 200). Subsets of the formerly naïve groups (∼75 each) received an MVA-BN booster 2 years later. Results Neutralizing antibody (nAb) geometric mean titers (GMTs) increased from 1.1 (baseline, both naïve groups) to 7.2 and 7.5 (Week 4, 1×MVA and 2×MVA, respectively), and further to 45.6 (Week 6, 2×MVA after second vaccination). In HSPX+, nAb GMT rapidly increased from 21.6 (baseline) to 175.1 (Week 2). At 2 years, GMTs for 1×MVA, 2×MVA, and HSPX+ were 1.1, 1.3, and 10.3, respectively. After boosting in the previously naïve groups, nAb GMTs increased rapidly in 2 weeks to 80.7 (1×MVA) and 125.3 (2×MVA), higher than after primary vaccination and comparable to boosted HSPX+ subjects. Six months after boosting, GMTs were 25.6 (1×MVA) and 49.3 (2×MVA). No safety concerns were identified. Conclusion Anamnestic responses without sustained high nAb titers support presence of durable immunological memory following MVA-BN immunization.


Effect of T-cell depletion on efficacy. BALB/c mice (n = 5) either were vaccinated (IN) twice 3 weeks apart with 1 × 10⁸ TCID50 MVA-RSV or received TBS. RSV challenge (IN, 10⁶ pfu) was performed 2 weeks after the last vaccination. One day prior and 1 day after RSV challenge, mice were either treated (IP) with an anti-CD4 (CD4 depl.) or anti-CD8 (CD8 depl.) or an isotype-matched control antibody (isotype control). For long-term depletion of CD4 T cells, injection with the anti-CD4 antibody was performed 2 days before prime vaccination and thereafter twice a week. (A) RSV-specific IgG in serum was measured on Day 33 (12 days post second vaccination) by ELISA. Titers were comparable between vaccinated mice treated with the isotype control antibody and anti-CD4 or anti-CD8 antibody (p = 0.9965 for CD4-depleted mice and p = 0.5426 for CD8-depleted mice; Tukey test). (B) RSV-specific mucosal IgA and IgG in BAL fluids were measured on Day 39 (4 days post challenge) by ELISA. Mice treated with anti-CD4 or anti-CD8 antibody had comparable RSV-specific mucosal IgA (p = 0.9717 for CD4-depleted mice and p = 0.1087 for CD8-depleted mice, Tukey test) as well as mucosal IgG (p = 0.9991 for CD4-depleted mice and p = 0.8205 for CD8-depleted mice, Tukey test) to vaccinated mice treated with the isotype control antibody. No antibodies could be measured in the CD4 long-term depleted group. For both panels the geometric mean titers (GMT) with 95% confidence interval (CI) are shown. Representative data of one out of two mouse studies are shown. ns: not statistically significant. Four days post challenge mice were sacrificed and the viral load in lung was measured by (C) plaque assay. Mean pfu per half lung ± SEM is shown. (D) Viral load detected by RT-qPCR. Mean L-gene copies ± SEM is shown; the black line indicates the lower limit of detection (LLOD = 15 gcs). Plaque assay data showed significant differences between TBS-treated control mice and all other groups (***p < 0.0005, **p < 0.005 Dunn’s method). RT-qPCR data showed significant differences between TBS-treated control mice and vaccinated control mice, CD4 depl. mice, and CD8 depl. mice (****p < 0.00005, ***p < 0.0005 Dunn’s method). Pooled data of two separately performed mouse studies are shown (n = 9 or 10).
Efficacy in CD8 T cell (β2M -/-)- or antibody (T11µMT)-deficient C57BL/6 mice. C57BL/6 WT mice (n = 5), T11µMT (n = 5), or β2M -/- (n = 4) were vaccinated (IN) twice 3 weeks apart with 1 × 10⁸ TCID50 MVA-RSV. WT mice treated with TBS served as controls. RSV challenge (IN, 10⁶ pfu) was performed 2 weeks after last vaccination. Four days post challenge mice were sacrificed, and the viral load in lung was measured by (A) plaque assay. Mean pfu per half lung ± SEM is shown. (B) Viral load detected by RT-qPCR. Mean L-gene copies ± SEM is shown; the black line indicates the lower limit of detection (LLOD = 15 gcs). Plaque assay data (pfu) showed significant differences between TBS-treated control mice and vaccinated control mice (*p < 0.05, Dunn’s method). RT-qPCR data (L-gene copies) showed significant differences between TBS-treated control mice and all other groups (***p < 0.0005, **p < 0.005, Tukey test). ns: not statistically significant.
Efficacy of passively transferred RSV-specific antibodies. Either BALB/c mice (n = 5) were vaccinated (IN) twice 3 weeks apart with 1 × 10⁸ TCID50 MVA-RSV or non-vaccinated BALB/c mice received 1 ml of serum (IP) from MVA-RSV-vaccinated (positive serum) or mock-vaccinated control mice (mock serum) 1 day before challenge. (A) RSV-specific IgG in serum was measured on Day 39 (4 days post serum transfer) by ELISA. Geometric mean titers (GMT) with 95% confidence interval (CI) are shown. Titers were comparable between MVA-RSV-vaccinated mice and mice receiving positive serum (p = 0.4127, Dunn`s method). (B) RSV-specific mucosal IgA and IgG in BAL fluids were measured on Day 39 (4 days post serum transfer) by ELISA. GMT with 95% CI is shown. RSV-specific mucosal IgG titers were comparable between vaccinated mice and mice receiving RSV-specific serum antibodies (p = 0.2313, Dunn’s method). For Figures 3B and 1B , identical results for RSV-specific mucosal IgA and IgG for MVA-RSV-vaccinated control animals were shown as this experiment was run in parallel to reduce the number of animals required. (C) Viral load in lung was measured 4 days post RSV challenge (IN, 10⁶ pfu) by plaque assay. Mean pfu per half lung ± SEM is shown. Viral load (pfu) determined by plaque assay was significantly (****p < 0.00005;Mann–Whitney U-test) reduced by the passive transfer of RSV-specific antibodies compared to the viral load of mice receiving the mock serum. (D) Viral load detected by RT-qPCR. Mean L-gene copies ± SEM is shown; the black line indicates the lower limit of detection (LLOD = 15 gcs). There was no statistically significant difference (p = 0.066, Mann–Whitney U-test) in RSV-specific L-gene copies between mice receiving RSV-specific antibodies and mice receiving the mock serum. Pooled data of two separately performed mouse studies are shown (n = 9 or 10). ns: not statistically significant.
Role of IgA for RSV protection. C57BL/6 WT (n = 5 to 10) or IgA-deficient (IgA-/-, n = 5 to 10) mice were vaccinated (IN) twice 3 weeks apart with 1 × 10⁸ TCID50 MVA-RSV. C57BL/6 WT mice treated with TBS served as controls. (A) RSV-specific IgG in serum was measured on Day 34 (13 days post last vaccination) by ELISA. Geometric mean titers (GMT) with 95% confidence interval (CI) are shown. Titers were similar between vaccinated WT mice and IgA -/- mice (p = 0.054, Mann–Whitney U-test). (B) RSV-specific mucosal IgA and IgG in BAL fluids were measured on Day 39 (4 days post challenge) by ELISA. GMT with 95% CI is shown. Mucosal IgG titers were comparable between vaccinated WT mice and IgA -/- mice (p = 0.4206, Mann–Whitney U-test). Mucosal IgA was only detected in vaccinated WT mice but not in IgA -/- mice, as expected. (C) Viral load in lung was measured 4 days post RSV challenge (IN, 10⁶ pfu) by plaque assay. Mean pfu per half lung ± SEM is shown. The lungs of vaccinated IgA -/- mice were completely free from infectious virus analogous to the vaccinated WT control group (*p < 0.05; Dunn’s method). (D) Viral load detected by RT-qPCR. Mean L-gene copies ± SEM is shown; the black line indicates the lower limit of detection (LLOD = 15 gcs). L-gene copies were significantly reduced in IgA -/- mice compared to TBS-treated WT mice (**p < 0.005, ***p < 0.0005, Tukey test). ns: not statistically significant. For Figures 4C, D and 2A, B , identical results for plaque assay (pfu) and RT-qPCR (L-gene copies) for TBS-treated and vaccine-treated control animals were shown as this experiment was run in parallel to reduce the number of animals required.
A Recombinant MVA-Based RSV Vaccine Induces T-Cell and Antibody Responses That Cooperate in the Protection Against RSV Infection

June 2022

·

86 Reads

·

16 Citations

Respiratory syncytial virus (RSV) causes a respiratory disease with a potentially fatal outcome especially in infants and elderly individuals. Several vaccines failed in pivotal clinical trials, and to date, no vaccine against RSV has been licensed. We have developed an RSV vaccine based on the recombinant Modified Vaccinia Virus Ankara-BN® (MVA-RSV), containing five RSV-specific antigens that induced antibody and T-cell responses, which is currently tested in clinical trials. Here, the immunological mechanisms of protection were evaluated to determine viral loads in lungs upon vaccination of mice with MVA-RSV followed by intranasal RSV challenge. Depletion of CD4 or CD8 T cells, serum transfer, and the use of genetically engineered mice lacking the ability to generate either RSV-specific antibodies (T11µMT), the IgA isotype (IgA knockout), or CD8 T cells (β2M knockout) revealed that complete protection from RSV challenge is dependent on CD4 and CD8 T cells as well as antibodies, including IgA. Thus, MVA-RSV vaccination optimally protects against RSV infection by employing multiple arms of the adaptive immune system.


Experimental design and immunogenicity of ABNCoV2. (A) Schematic study design with non-human primates (NHP) (N = 6 per group) vaccinated intramuscularly with 100 or 15 μg ABNCoV2 in weeks 0 and 14 and challenged with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in week 20. Four (4) additional non-vaccinated NHP were added as controls. At regular intervals as indicated, animals were bled. (B) RBD-specific IgG was measured by ELISA, and (C) SARS-CoV-2 neutralizing antibodies were assessed by PRNT. For comparison, 10 human plasma samples positive for SARS-CoV-2 antibodies (human convalescents, gray symbols) were analyzed as well. Filled circles represent individual values and columns depict the geometric mean (ELISA, B) or geometric mean titers (PRNT, C) of the group +/geometric standard deviation. The horizontal dotted line in (B) represents the mean + 2 times the standard deviation of 50 untreated naive NHP to indicate background responses.
Chemokine changes following SARS-CoV-2 challenge. NHP (N = 6 per group) were vaccinated intramuscularly with 100 μg ABNCoV2 (red symbols) or 15 μg ABNCoV2 (blue symbols) in weeks 0 and 14 and challenged by the combined intranasal/intratracheal route with SARS-CoV-2 in week 20. As controls, four non-vaccinated rhesus macaques (black symbols) were challenged at the same time. CXCL10 (top graphs) and CXCL11 (bottom graphs) were measured in serum (A) and bronchoalveolar lavage (BAL) (B) on the days post-challenge as indicated. Each curve represents an individual animal. The symbol shapes identify individual animals as stated and the same symbols are used for the same animals throughout all post-challenge data.
Viral load in bronchoalveolar lavage following SARS-CoV-2 challenge. Rhesus macaques were vaccinated and challenged as described in Figure 2 . sgRNA was measured in bronchoalveolar lavage on days 2, 4, and 6 post-challenge. (A) sgRNA copies per ml BAL are shown for each animal in the 100-μg ABNCoV2 group by red symbols, in the 15-μg ABNCoV2 group by blue symbols, and in the non-vaccinated control group by black symbols. The symbol shapes identify individual animals; the same symbols are used for the same animals throughout all post-challenge data. Horizontal bars depict geometric mean values. (B) Total amount of sgRNA in BAL per animal (symbols) and median values (horizontal bars) are calculated as area under the curve (AUC). Significant differences by Mann–Whitney test were seen between each vaccinated group compared with the control group (p = 0.0095).
Immune responses following SARS-CoV-2 challenge. Rhesus macaques were vaccinated and challenged as described in Figure 2 . SARS-CoV-2 neutralizing antibodies (A) were measured at the time of challenge (week 20) and 2 weeks post-challenge (week 22) by PRNT, and RBD-specific IFN-γ T-cell responses (B) were measured 2 weeks post-challenge by ELISpot. (A) Symbols represent individual PRNT50 titers; horizontal bars depict the GMT of the group. For comparison, PRNT50 titers and GMT of 10 human plasma samples positive for SARS-CoV-2 antibodies (human convalescents) are shown. (B) Symbols represent individual spot-forming units (SFU) per one million PBMCs; horizontal bars depict mean SFU per one million PBMCs. Different symbol shapes represent individual animals, and the same symbol shapes are used for the same animals throughout all post-challenge data.
Neutralization of SARS-CoV-2 variants of concern. (A) NHP were vaccinated as described in Figure 1 . Two weeks following the second administration, vaccine-induced antibodies were tested for their capacity to neutralize Wuhan-like SARS-CoV-2 (FR-4286) as well as the alpha (B.1.1.7), beta (B.1.351), and delta variants (B1.617.2) by PRNT. Filled circles represent PRNT50 titers of individual NHP, and columns depict the GMT of the group +/geometric standard deviation. Kruskal–Wallis test showed no statistically significant differences between the neutralization of Wuhan-like FR-4286 and the variants. (B) Ten human plasma samples positive for SARS-CoV-2 antibodies (human convalescents) were analyzed for comparison.
A Capsid Virus-Like Particle-Based SARS-CoV-2 Vaccine Induces High Levels of Antibodies and Protects Rhesus Macaques

April 2022

·

429 Reads

·

25 Citations

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a worldwide pandemic. Here, we present non-human primate immunogenicity and protective efficacy data generated with the capsid virus-like particle (cVLP)-based vaccine ABNCoV2 that has previously demonstrated immunogenicity in mice. In rhesus macaques, a single vaccination with either 15 or 100 μg ABNCoV2 induced binding and neutralizing antibodies in a dose-dependent manner, at levels comparable to those measured in human convalescents. A second vaccine administration led to a >50-fold increase in neutralizing antibodies, with 2-log higher mean levels in the 100-μg ABNCoV2 group compared with convalescent samples. Upon SARS-CoV-2 challenge, a significant reduction in viral load was observed for both vaccine groups relative to the challenge control group, with no evidence of enhanced disease. Remarkably, neutralizing antibody titers against an original SARS-CoV-2 isolate and against variants of concern were comparable, indicating a potential for broad protection afforded by ABNCoV2, which is currently in clinical testing.


Human antibody responses following vaccinia immunization using protein microarrays and correlation with cell-mediated and antibody dependent cellular cytotoxicity responses

March 2021

·

58 Reads

·

15 Citations

The Journal of Infectious Diseases

Background There are limited data regarding immunological correlates of protection for the modified vaccinia Ankara (MVA) smallpox vaccine. Methods Five hundred twenty-three vaccinia-naïve subjects were randomized to receive two vaccine doses, either lyophilized MVA subcutaneously (SC), liquid MVA SC or liquid MVA intradermally (ID) 28 days apart. For a subset of subjects, antibody-dependent cellular cytotoxicity (ADCC), IFN- γ release enzyme-linked immunospot (ELISPOT), and protein microarray antibody-binding assays were conducted. Protein microarray responses were assessed for correlations with PRNT, enzyme-linked immunosorbent assay (ELISA), ADCC and ELISPOT results. Results MVA elicited significant microarray antibody responses to 15 of 224 antigens, mostly virion membrane proteins, at day 28 or 42, particularly, WR113/D8L and WR101H3L. In the Liquid-SC group, responses to nine antigens, including WR113/D8L and WR101/H3L, correlated with PRNT results. Three correlated for the Liquid-ID group. No significant correlations were observed with ELISPOT responses. In the Liquid-ID group, WR052/F13L, a membrane glycoprotein, correlated with ADCC responses. Conclusions MVA elicited antibodies to fifteen vaccinia strain antigens representing virion membrane. Antibody responses to two proteins strongly increased and significantly correlated with increases in PRNT. Responses to these proteins are potential correlates of protection and may serve as immunogens for future vaccine development.


Intratumoral virotherapy with 4-1BBL armed modified vaccinia Ankara eradicates solid tumors and promotes protective immune memory

February 2021

·

221 Reads

·

16 Citations

Background Human cancers are extraordinarily heterogeneous in terms of tumor antigen expression, immune infiltration and composition. A common feature, however, is the host′s inability to mount potent immune responses that prevent tumor growth effectively. Often, naturally primed CD8 ⁺ T cells against solid tumors lack adequate stimulation and efficient tumor tissue penetration due to an immune hostile tumor microenvironment. Methods To address these shortcomings, we cloned tumor-associated antigens (TAA) and the immune-stimulatory ligand 4-1BBL into the genome of modified vaccinia Ankara (MVA) for intratumoral virotherapy. Results Local treatment with MVA-TAA-4-1BBL resulted in control of established tumors. Intratumoral injection of MVA localized mainly to the tumor with minimal leakage to the tumor-draining lymph node. In situ infection by MVA-TAA-4-1BBL triggered profound changes in the tumor microenvironment, including the induction of multiple proinflammatory molecules and immunogenic cell death. These changes led to the reactivation and expansion of antigen-experienced, tumor-specific cytotoxic CD8 ⁺ T cells that were essential for the therapeutic antitumor effect. Strikingly, we report the induction of a systemic antitumor immune response including tumor antigen spread by local MVA-TAA-4-1BBL treatment which controlled tumor growth at distant, untreated lesions and protected against local and systemic tumor rechallenge. In all cases, 4-1BBL adjuvanted MVA was superior to MVA. Conclusion Intratumoral 4-1BBL-armed MVA immunotherapy induced a profound reactivation and expansion of potent tumor-specific CD8 ⁺ T cells as well as favorable proinflammatory changes in the tumor microenvironment, leading to elimination of tumors and protective immunological memory.



P04.02 A novel cancer immunotherapy combines rMVA-CD40L with tumor targeting antibodies

October 2020

·

40 Reads

·

1 Citation

Background Virus-based vaccines and appropriate costimulation potently enhance antigen-specific T cell immunity against cancer. In the present study, we exploit both innate and adaptive immune responses triggered by a novel recombinant modified vaccinia virus Ankara (rMVA) encoding a Tumor-Associated Antigen (TAA) and the costimulatory CD40L against solid tumors in combination regimes to overcome tumor-induced resistance to immunotherapy. Material and Methods Subcutaneous murine tumors were induced in C57BL/6 or Balb/c mice using syngeneic tumor cell lines. When tumors were established (60–80 mm ³ ) mice were intravenously injected with rMVA-CD40L. Tumor growth monitoring and immune cell analysis was performed. Results Therapeutic treatment with rMVA-CD40L resulted in the control of established tumors in several independent tumor models. This antitumor effect was based on the generation of non-exhausted, systemic tumor-specific cytotoxic CD8 ⁺ T cells that was essential for therapeutic efficacy. Strikingly, rMVA-CD40L also induced strong NK cell activation and enhanced cytotoxicity. Moreover, the combination of rMVA-CD40L and tumor targeting antibodies resulted in increased therapeutic antitumor efficacy. This therapeutic combination relied on Fcγ receptor-expressing immune cells as well as on NK cells. Conclusion We describe a novel and translationally relevant therapeutic synergy between viral vaccination and CD40L costimulation. We show strengthened antitumor immune responses when both rMVA-CD40L-induced innate and adaptive immune mechanisms are exploited by combining immunotherapeutic regimes, such as TAA targeting antibodies. This finding could have a direct positive impact in therapeutic regimens where TAA targeting antibodies could be employed. Disclosure Information M. Hinterberger: A. Employment (full or part-time); Significant; Bavarian Nordic. J. Medina-Echeverz: A. Employment (full or part-time); Significant; Bavarian Nordic. M. Testori: A. Employment (full or part-time); Significant; Bavarian Nordic. M. Geiger: A. Employment (full or part-time); Significant; Bavarian Nordic. R. Giessel: A. Employment (full or part-time); Significant; Bavarian Nordic. B. Bathke: A. Employment (full or part-time); Significant; Bavarian Nordic. R. Kassub: A. Employment (full or part-time); Significant; Bavarian Nordic. F. Gräbnitz: A. Employment (full or part-time); Significant; Bavarian Nordic. G. Fiore: A. Employment (full or part-time); Significant; Bavarian Nordic. S. Wennier: A. Employment (full or part-time); Significant; Bavarian Nordic. P. Chaplin: A. Employment (full or part-time); Significant; Bavarian Nordic. M. Suter: A. Employment (full or part-time); Significant; Bavarian Nordic. H. Hochrein: A. Employment (full or part-time); Significant; Bavarian Nordic. H. Lauterbach: A. Employment (full or part-time); Significant; Bavarian Nordic.


Citations (60)


... HER-Vaxx (IMU-131), a peptide-based vaccine, demonstrated an improved OS of 14.0 months when compared with chemotherapy with OS of 8.3 months (0.08) in a phase II trial [83]. Another vaccine, BVAC-B, showed limited clinical activity, whereas TAEK-VAC-HerBy, a novel human tumor antibody enhanced killing (TAEK) vaccine (VAC) encoding the membrane bound form of human CD40L, HER2 (Her) and the human transcription factor Brachyury (By), is currently in a phase I trial (NCT04246671) [84,85]. Chimeric antigen receptor T-cell (CAR-T) therapy targeting HER2 is under investigation, primarily for solid tumors (NCT04650451 and NCT04511871). ...

Reference:

Human Epidermal Growth Factor Receptor 2 Positive Advanced Gastric or Esophagogastric Adenocarcinoma: Reflecting on the Past to Gain a New Insights
Preclinical development of a first-in-class vaccine encoding HER2, Brachyury and CD40L for antibody enhanced tumor eradication

... The latter includes the demonstration of non-inferior immunogenicity compared to the replicating-vaccinia vaccine ACAM2000 together with a more favorable safety profile for MVA-BN compared to ACAM2000 as demonstrated in a pivotal phase 3 randomized clinical trial [42]. While one dose of MVA-BN was previously shown to induce low titers of vaccinia-specific and MPVX-specific neutralizing antibody titers [43][44][45], our SLR found VE estimates against mpox infections for one dose of MVA-BN suggestive of high effectiveness. This challenges the view that neutralizing antibody levels correlate with protection against Orthopoxvirus infection. ...

Single and 2-dose vaccinations with modified vaccinia Ankara-Bavarian Nordic® induce durable B cell memory responses comparable to replicating smallpox vaccines

The Journal of Infectious Diseases

... The lower measured viral load in IM-INand IN-IN-vaccinated mouse lungs may have been as a result of enhanced mucosal antibody responses, or may reflect a more multifactorial mechanism of RSV protection. CD8 + and CD4 + T cells, and specifically T RM , have also been described as important for protection against RSV, and in other studies, control over RSV infection has been demonstrated by T cells in the absence of RSV-specific antibodies [69][70][71][72] . ...

A Recombinant MVA-Based RSV Vaccine Induces T-Cell and Antibody Responses That Cooperate in the Protection Against RSV Infection

... Currently, various virus-like particle (VLP) vaccines have utilised S protein or its fragments as the primary antigenic component. [4][5][6][7] Significant progress has been made in the application of VLPs in vaccine development over the past 30 years. 8,9 In addition to having a spatial structure and composition similar to natural virus particles, VLPs vaccines can also enable effective uptake and processing by antigen-presenting cells due to their highly ordered and repetitive antigen structure, thereby inducing higher levels of humoral and cellular immunity. ...

A Capsid Virus-Like Particle-Based SARS-CoV-2 Vaccine Induces High Levels of Antibodies and Protects Rhesus Macaques

... To further characterize the humoral immune response before and after the third immunization and determine the EBV glycoprotein-specific IgG isotypes elicited by the MVA vaccines, MVA-gp350-immunized mice mainly exhibited glycoproteinspecific IgG1. We observed no increase in IgG1 or IgG2a levels after the third MVA-EBV5-2 dose (Day 84) in either sex, which together with the results of our kinetic response analysis in Figures 2B, 3B, suggested that a third MVA-EBV5-2 immunization did not further boost IgG responses; thus, we reduced the number of MVA-EBV5-2 doses to two in subsequent immunogenicity experiments, which is representative of regimens used in the clinic for other MVA-based vaccines (56,60,79). ...

Human antibody responses following vaccinia immunization using protein microarrays and correlation with cell-mediated and antibody dependent cellular cytotoxicity responses

The Journal of Infectious Diseases

... T cell stimulators such as tumour necrosis factor ligand superfamily member 9 (TNFSF9) 104,107 , and single-chain antibodies against immune checkpoints, such as cytotoxic T lymphocyte-associated protein 4 (CTLA-4) 104 or PD1 (ref. 108), as well as various immunostimulatory cytokines 92 . ...

Intratumoral virotherapy with 4-1BBL armed modified vaccinia Ankara eradicates solid tumors and promotes protective immune memory

... MVA has a proven clinical track record and a high safety profile (9). Therefore, MVA can also be used in immunocompromised animals (10) and humans (11). MVA-BN is an approved prophylactic vaccine against smallpox and monkeypox in the European Union (12) and the USA (13). ...

MVA-BN as monkeypox vaccine for healthy and immunocompromised

International Journal of Infectious Diseases

... Such mechanisms, in conjunction with Bcl-xL expression, more fit mitochondria, and IL-2 autocrine production, provide cis CD137costimulated T cells with clear competitive survival and functional advantages. This advantage was prominently observed upon competition to infiltrate transplanted tumors and in response to vaccination with MVA vectors 34,43 . The intriguing effects of Eomes and T-bet expression are probably behind these observations, but such transcriptional biology is very complex and gives rise to different outcomes depending on balanced coexpression of these two transcription factors 44 . ...

P04.02 A novel cancer immunotherapy combines rMVA-CD40L with tumor targeting antibodies
  • Citing Article
  • October 2020

... Notably, both the ChAd/mRNA and mRNA/mRNA regimens, but not the MVA-based vaccine candidates, induced a transcriptional response of the complement cascade, which is also suggested to be involved in the pathogenesis of VITT (73). This is in line with the beneficial safety profile of recombinant and non-recombinant MVA-based vaccines reported in numerous studies (8,74). ...

Broad Antibody and Cellular Immune Response from a Phase 2 Clinical Trial with a Novel Multivalent Poxvirus Based RSV Vaccine
  • Citing Article
  • July 2020

The Journal of Infectious Diseases

... It is possible that persons with viremia despite ART prescription were also those less engaged in care and had fewer opportunities for vaccination; conversely, providers often defer vaccines for those with low CD4 count for concerns of safety or lack of efficacy [22,23]. The gap in vaccinating persons with CD4 <200 raises questions about whether some PWH or their providers had safety concerns, even though MVA-BN is a replication-deficient vaccine and is safe to administer in advanced immunosuppression [24]. ...

A randomized phase II trial to compare safety and immunogenicity of the MVA-BN smallpox vaccine at various doses in adults with a history of AIDS
  • Citing Article
  • February 2020

Vaccine