16 results on '"Travis L. Jensen"'
Search Results
2. The antibody landscapes following AS03 and MF59 adjuvanted H5N1 vaccination
- Author
-
Johannes B. Goll, Aarti Jain, Travis L. Jensen, Rafael Assis, Rie Nakajima, Algis Jasinskas, Lynda Coughlan, Sami R. Cherikh, Casey E. Gelber, S. Khan, D. Huw Davies, Philip Meade, Daniel Stadlbauer, Shirin Strohmeier, Florian Krammer, Wilbur H. Chen, and Philip L. Felgner
- Subjects
Pharmacology ,and promotion of well-being ,Prevention ,Immunology ,Prevention of disease and conditions ,Influenza ,Vaccine Related ,Infectious Diseases ,Emerging Infectious Diseases ,3.4 Vaccines ,Clinical Research ,Biodefense ,Pneumonia & Influenza ,Pharmacology (medical) ,Immunization ,Infection - Abstract
Current seasonal and pre-pandemic influenza vaccines induce short-lived predominantly strain-specific and limited heterosubtypic responses. To better understand how vaccine adjuvants AS03 and MF59 may provide improved antibody responses to vaccination, we interrogated serum from subjects who received 2 doses of inactivated monovalent influenza A/Indonesia/05/2005 vaccine with or without AS03 or MF59 using hemagglutinin (HA) microarrays (NCT01317758 and NCT01317745). The arrays were designed to reflect both full-length and globular head HA derived from 17 influenza A subtypes (H1 to H16 and H18) and influenza B strains. We observed significantly increased strain-specific and broad homo- and heterosubtypic antibody responses with both AS03 and MF59 adjuvanted vaccination with AS03 achieving a higher titer and breadth of IgG responses relative to MF59. The adjuvanted vaccine was also associated with the elicitation of stalk-directed antibody. We established good correlation of the array antibody responses to H5 antigens with standard HA inhibition and microneutralization titers.
- Published
- 2022
3. AS03-Adjuvanted H5N1 Avian Influenza Vaccine Modulates Early Innate Immune Signatures in Human Peripheral Blood Mononuclear Cells
- Author
-
C. Buddy Creech, Travis L. Jensen, Casey E. Gelber, Andrew J. Link, Kristen L. Hoek, Kathryn M. Edwards, Leigh M Howard, Shawn Levy, Sebastian Joyce, Nripesh Prasad, and Johannes B. Goll
- Subjects
Adult ,Squalene ,0301 basic medicine ,H5N1 vaccine ,medicine.medical_treatment ,alpha-Tocopherol ,Polysorbates ,Adaptive Immunity ,Biology ,Peripheral blood mononuclear cell ,Major Articles and Brief Reports ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Adjuvants, Immunologic ,Double-Blind Method ,Interferon ,Influenza, Human ,Leukocytes ,medicine ,Humans ,Immunology and Allergy ,030212 general & internal medicine ,Innate immune system ,Influenza A Virus, H5N1 Subtype ,Antigen processing ,Gene Expression Profiling ,Acquired immune system ,Immunity, Innate ,Drug Combinations ,030104 developmental biology ,Infectious Diseases ,Influenza Vaccines ,Immunology ,Adjuvant ,Signal Transduction ,medicine.drug - Abstract
Background Adjuvant System 03 (AS03) markedly enhances responses to influenza A/H5N1 vaccines, but the mechanisms of this enhancement are incompletely understood. Methods Using ribonucleic acid sequencing on peripheral blood mononuclear cells (PBMCs) from AS03-adjuvanted and unadjuvanted inactivated H5N1 vaccine recipients, we identified differentially expressed genes, enriched pathways, and genes that correlated with serologic responses. We compared bulk PBMC findings with our previously published assessments of flow-sorted immune cell types. Results AS03-adjuvanted vaccine induced the strongest differential signals on day 1 postvaccination, activating multiple innate immune pathways including interferon and JAK-STAT signaling, Fcγ receptor (FcγR)-mediated phagocytosis, and antigen processing and presentation. Changes in signal transduction and immunoglobulin genes predicted peak hemagglutinin inhibition (HAI) titers. Compared with individual immune cell types, activated PBMC genes and pathways were most similar to innate immune cells. However, several pathways were unique to PBMCs, and several pathways identified in individual cell types were absent in PBMCs. Conclusions Transcriptomic analysis of PBMCs after AS03-adjuvanted H5N1 vaccination revealed early activation of innate immune signaling, including a 5- to 8-fold upregulation of FcγR1A/1B/1C genes. Several early gene responses were correlated with HAI titer, indicating links with the adaptive immune response. Although PBMCs and cell-specific results shared key innate immune signals, unique signals were identified by both approaches.
- Published
- 2018
4. Human Antibody Responses Following Vaccinia Immunization Using Protein Microarrays and Correlation With Cell-Mediated Immunity and Antibody-Dependent Cellular Cytotoxicity Responses
- Author
-
C. Buddy Creech, Paul Chaplin, Mark J. Mulligan, Wilbur H. Chen, Sharon E. Frey, Thomas M. Kaufman, Lisa A. Jackson, Anna Wald, Samer S. El-Kamary, Jack T. Stapleton, Nadine Rouphael, Travis L. Jensen, Shital M. Patel, Heather Hill, Christine Johnston, Patricia L. Winokur, Hana M. El Sahly, D. Huw Davies, Kathryn M. Edwards, Tammy P Blevins, Zuhair K. Ballas, Wendy L. Rasmussen, Robert B. Belshe, Johannes B. Goll, Srilatha Edupuganti, Robert L. Atmar, Magdalena Tary-Lehmann, and Wendy A. Keitel
- Subjects
0301 basic medicine ,Modified vaccinia Ankara ,viruses ,Protein Array Analysis ,Vaccinia virus ,Vaccines, Attenuated ,complex mixtures ,03 medical and health sciences ,chemistry.chemical_compound ,Major Articles and Brief Reports ,0302 clinical medicine ,Antigen ,Vaccines, DNA ,Vaccinia ,Immunology and Allergy ,Humans ,030212 general & internal medicine ,Antigens, Viral ,Antibody-dependent cell-mediated cytotoxicity ,Immunity, Cellular ,biology ,ELISPOT ,Virion membrane ,Antibody-Dependent Cell Cytotoxicity ,hemic and immune systems ,Viral Vaccines ,Titer ,030104 developmental biology ,Infectious Diseases ,chemistry ,Immunology ,Antibody Formation ,biology.protein ,Immunization ,Antibody ,Smallpox Vaccine - Abstract
Background There are limited data regarding immunological correlates of protection for the modified vaccinia Ankara (MVA) smallpox vaccine. Methods A total of 523 vaccinia-naive subjects were randomized to receive 2 vaccine doses, as lyophilized MVA given subcutaneously, liquid MVA given subcutaneously (liquid-SC group), or liquid MVA given intradermally (liquid-ID group) 28 days apart. For a subset of subjects, antibody-dependent cellular cytotoxicity (ADCC), interferon-γ release enzyme-linked immunospot (ELISPOT), and protein microarray antibody-binding assays were conducted. Protein microarray responses were assessed for correlations with plaque reduction neutralization titer (PRNT), enzyme-linked immunosorbent assay, 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 9 antigens, including WR113/D8L and WR101/H3L, correlated with PRNT results. Three were correlated in 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 15 vaccinia strain antigens representing virion membrane. Antibody responses to 2 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. Clinical Trials Registration NCT00914732.
- Published
- 2020
5. Proteomic Analysis of Human Immune Responses to Live-Attenuated Tularemia Vaccine
- Author
-
Duc M. Duong, Yie-Hwa Chang, Nicholas T. Seyfried, Nadine Rouphael, Mark J. Mulligan, Travis L. Jensen, Luming Yin, Patrick Sanz, Daniel F. Hoft, Johannes B. Goll, Casey E. Gelber, David C. Wood, Evan J. Anderson, Muktha S Natrajan, and Robert A Johnson
- Subjects
0301 basic medicine ,Immunology ,Antigen presentation ,lcsh:Medicine ,Peripheral blood mononuclear cell ,complex mixtures ,Article ,Tularemia ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,proteomics ,human immune response ,Drug Discovery ,MHC class I ,medicine ,antigen processing ,Pharmacology (medical) ,LC-MS/MS ,tularemia vaccine ,Francisella tularensis ,Pharmacology ,biology ,Antigen processing ,lcsh:R ,phagocytosis ,biomarkers ,respiratory system ,medicine.disease ,biology.organism_classification ,bacterial infections and mycoses ,Tularemia vaccine ,030104 developmental biology ,Infectious Diseases ,inflammation ,030220 oncology & carcinogenesis ,biology.protein ,2D-DIGE - Abstract
Francisella tularensis (F. tularensis) is an intracellular pathogen that causes a potentially debilitating febrile illness known as tularemia. F. tularensis can be spread by aerosol transmission and cause fatal pneumonic tularemia. If untreated, mortality rates can be as high as 30%. To study the host responses to a live-attenuated tularemia vaccine, peripheral blood mononuclear cell (PBMC) samples were assayed from 10 subjects collected pre- and post-vaccination, using both the 2D-DIGE/MALDI-MS/MS and LC-MS/MS approaches. Protein expression related to antigen processing and presentation, inflammation (PPAR&gamma, nuclear receptor), phagocytosis, and gram-negative bacterial infection was enriched at Day 7 and/or Day 14. Protein candidates that could be used to predict human immune responses were identified by evaluating the correlation between proteome changes and humoral and cellular immune responses. Consistent with the proteomics data, parallel transcriptomics data showed that MHC class I and class II-related signals important for protein processing and antigen presentation were up-regulated, further confirming the proteomic results. These findings provide new biological insights that can be built upon in future clinical studies, using live attenuated strains as immunogens, including their potential use as surrogates of protection.
- Published
- 2020
- Full Text
- View/download PDF
6. Transcriptomic and Metabolic Responses to a Live-Attenuated Francisella tularensis Vaccine
- Author
-
Mark J. Mulligan, Nadine Rouphael, Steven E. Bosinger, James L. Edwards, William F. Hooper, Muktha S Natrajan, Katherine L. Sanders, Casey E. Gelber, Patrick Sanz, Daniel F. Hoft, Yating Wang, Shuzhao Li, Johannes B. Goll, Evan J. Anderson, Robert A Johnson, and Travis L. Jensen
- Subjects
0301 basic medicine ,suppression of immune response ,DVC-LVS ,T cell ,Immunology ,Antigen presentation ,genetic processes ,TNF ,lcsh:Medicine ,LC–MS ,Article ,NF-κB ,Francisella tularenis vaccine ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,TLR ,human immune response ,Drug Discovery ,MHC class I ,medicine ,Pharmacology (medical) ,natural sciences ,RNA-Seq ,NOD-like receptor ,Francisella tularensis ,tularemia vaccine ,B cell ,Pharmacology ,Innate immune system ,biology ,lcsh:R ,biology.organism_classification ,metabolomics ,innate immune signaling ,interferon α/β signaling ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,bacteria ,Antibody - Abstract
The immune response to live-attenuated Francisella tularensis vaccine and its host evasion mechanisms are incompletely understood. Using RNA-Seq and LC&ndash, MS on samples collected pre-vaccination and at days 1, 2, 7, and 14 post-vaccination, we identified differentially expressed genes in PBMCs, metabolites in serum, enriched pathways, and metabolites that correlated with T cell and B cell responses, or gene expression modules. While an early activation of interferon &alpha, /&beta, signaling was observed, several innate immune signaling pathways including TLR, TNF, NF-&kappa, B, and NOD-like receptor signaling and key inflammatory cytokines such as Il-1&alpha, Il-1&beta, and TNF typically activated following infection were suppressed. The NF-&kappa, B pathway was the most impacted and the likely route of attack. Plasma cells, immunoglobulin, and B cell signatures were evident by day 7. MHC I antigen presentation was more actively up-regulated first followed by MHC II which coincided with the emergence of humoral immune signatures. Metabolomics analysis showed that glycolysis and TCA cycle-related metabolites were perturbed including a decline in pyruvate. Correlation networks that provide hypotheses on the interplay between changes in innate immune, T cell, and B cell gene expression signatures and metabolites are provided. Results demonstrate the utility of transcriptomics and metabolomics for better understanding molecular mechanisms of vaccine response and potential host&ndash, pathogen interactions.
- Published
- 2020
7. Plasmablast, Memory B Cell, CD4+ T Cell, and Circulating Follicular Helper T Cell Responses to a Non-Replicating Modified Vaccinia Ankara Vaccine
- Author
-
Heather Hill, Yongxian Xu, Carol M. Kao, Lalita Priyamvada, Lilin Lai, Jens Wrammert, Sarah Kabbani, Travis L. Jensen, Evan J. Anderson, Nadine Rouphael, Inci Yildirim, Mark J. Mulligan, Johannes B. Goll, and Lisa A. Jackson
- Subjects
0301 basic medicine ,Modified vaccinia Ankara ,animal structures ,viruses ,T cell ,Immunology ,lcsh:Medicine ,mva ,complex mixtures ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Plaque reduction neutralization test ,Drug Discovery ,Medicine ,Pharmacology (medical) ,030212 general & internal medicine ,Memory B cell ,vaccinia ,Pharmacology ,biology ,business.industry ,lcsh:R ,Vaccination ,Titer ,smallpox ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,follicular helper t cells (tfh), plasmablasts ,chemistry ,antibody secreting cells ,biology.protein ,Vaccinia ,Antibody ,business - Abstract
Background: Vaccinia is known to induce antibody and cellular responses. Plasmablast, circulating follicular helper T (cTFH) cells, cytokine-expressing CD4 T cells, and memory B cells were compared between subcutaneous (SC) and needle-free jet injection (JI) recipients of non-replicating modified vaccinia Ankara (MVA) vaccine. Methods: Vaccinia-naï, ve adults received MVA SC or by JI on Days 1 and 29. Vaccinia-specific antibodies were quantified by plaque reduction neutralization test (PRNT) and enzyme-linked immunosorbent assay. Plasmablast, cTFH, and cytokine-expressing CD4 T cells were assessed on Days 1, 8, 15, 29, 36, 43 (cTFH and CD4+ only) and 57. Memory B cells were measured on Days 1 and 57. Results: Of the 36 enrolled subjects, only 22 received both vaccinations and had evaluable specimens after the second vaccine. Plasmablasts peaked one week after each vaccine. Day 15 plasmablasts correlated with peak PRNT titers. cTFH peaked on Days 8 and 36 and correlated with Day 36 plasmablasts. CD4+ peaked at Day 29 and one-third produced &ge, 2 cytokines. Day 57 memory B cells ranged from 0.1% to 0.17% of IgG-secreting B cells. Conclusions: This study provides insights into the cellular responses to non-replicating MVA, currently used as a vector for a variety of novel vaccines.
- Published
- 2020
8. Systems Vaccinology for a Live Attenuated Tularemia Vaccine Reveals Unique Transcriptional Signatures That Predict Humoral and Cellular Immune Responses
- Author
-
William F. Hooper, Patrick Sanz, Marcelo B. Sztein, Johannes B. Goll, Robert A Johnson, Heather Hill, Dmitri Kazmin, Chris C. Ibegbu, Muktha S Natrajan, Evan J. Anderson, Mark J. Mulligan, Nadine Rouphael, Travis L. Jensen, Bali Pulendran, David S. Weiss, and Lilin Lai
- Subjects
0301 basic medicine ,T cell ,Immunology ,lcsh:Medicine ,Biology ,Article ,Tularemia ,03 medical and health sciences ,transcriptomics ,0302 clinical medicine ,Immune system ,Drug Discovery ,medicine ,Live attenuated influenza vaccine ,Pharmacology (medical) ,030212 general & internal medicine ,Pharmacology ,Innate immune system ,Viral Vaccine ,lcsh:R ,medicine.disease ,Tularemia vaccine ,tularemia ,comparative vaccines ,Vaccination ,live attenuated vaccines ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,immune signaling - Abstract
Background: Tularemia is a potential biological weapon due to its high infectivity and ease of dissemination. This study aimed to characterize the innate and adaptive responses induced by two different lots of a live attenuated tularemia vaccine and compare them to other well-characterized viral vaccine immune responses. Methods: Microarray analyses were performed on human peripheral blood mononuclear cells (PBMCs) to determine changes in transcriptional activity that correlated with changes detected by cellular phenotyping, cytokine signaling, and serological assays. Transcriptional profiles after tularemia vaccination were compared with yellow fever [YF-17D], inactivated [TIV], and live attenuated [LAIV] influenza. Results: Tularemia vaccine lots produced strong innate immune responses by Day 2 after vaccination, with an increase in monocytes, NK cells, and cytokine signaling. T cell responses peaked at Day 14. Changes in gene expression, including upregulation of STAT1, GBP1, and IFIT2, predicted tularemia-specific antibody responses. Changes in CCL20 expression positively correlated with peak CD8+ T cell responses, but negatively correlated with peak CD4+ T cell activation. Tularemia vaccines elicited gene expression signatures similar to other replicating vaccines, inducing early upregulation of interferon-inducible genes. Conclusions: A systems vaccinology approach identified that tularemia vaccines induce a strong innate immune response early after vaccination, similar to the response seen after well-studied viral vaccines, and produce unique transcriptional signatures that are strongly correlated to the induction of T cell and antibody responses.
- Published
- 2019
- Full Text
- View/download PDF
9. Targeted Sequencing of 7 Genes Can Help Reduce Pathologic Misclassification of MDS
- Author
-
Steven D. Gore, Eric Padron, Amy E. DeZern, Donnie Hebert, James M. Foran, Steven H. Kroft, Myron A. Waclawiw, Alexandra M. Harrington, Jane Jijun Liu, Wael Saber, Steffanie H. Wilson, Jason Walker, Tareq Al Baghdadi, Daniel T. Starczynowski, Johannes B. Goll, Edward J. Gorak, Ling Zhang, Mikkael A. Sekeres, Jaroslaw P. Maciejewski, H. Joachim Deeg, Rafael Bejar, Nancy L. DiFronzo, R. Coleman Lindsley, Rami S. Komrokji, Robert S. Fulton, Harrison Reed, Benjamin L. Ebert, Travis L. Jensen, Gregory A. Abel, and Matthew J. Walter
- Subjects
business.industry ,Immunology ,Medicine ,Cell Biology ,Hematology ,Computational biology ,business ,Biochemistry ,Gene - Abstract
Introduction: The NHLBI National MDS Study (NCT02775383) is a prospective cohort study conducted at 92 community hospitals and 29 academic centers. It enrolls patients undergoing work up for suspected MDS to understand the genetic, epigenetic, and biological factors associated with the initiation and progression of the disease. Previously untreated, cytopenic participants undergo both local and centralized pathology review and are assigned a diagnosis, including MDS, MDS/MPN, AML with blasts < 30%, and "Other". Emerging data suggests that Next Generation Sequencing (NGS), along with cytogenetics and clinical variables, may improve MDS diagnostic precision. Given that our study relies on central review (with additional tertiary pathology review used to adjudicate disagreements), we examined whether targeted gene sequencing data could be used to increase the agreement between local and central pathologic diagnosis of MDS vs. Other. Methods: Peripheral blood and bone marrow (BM) biopsy specimens from cytopenic patients, along with clinical history, CBC, and other results including karyotyping, FISH and pathology reports from local pathologists were reviewed by central pathologists. The updated 2016 WHO classifications were used to diagnose MDS. Targeted exon sequencing of 96 genes was performed using BM specimens. A subset of 648 individuals that were classified as MDS (n=212) or Other (n=436, including 90 CCUS and 89 individuals with other cancers) by pathology assessments were selected. A mean coverage of 1,317X was achieved and variants had a minimum variant allele frequency (VAF) of 2% (except FLT3). Variants for 596 subjects were manually reviewed to retain likely disease-causing variants to build a binary classifier (MDS vs. Other) using the maximum VAF per gene as input (Figure 1). Subjects diagnosed with MDS or Other by both central and local pathology were used for training, validation, and testing, and were considered "gold standard" (GS) cases (n=546). These subjects were split into 4 random groups with equal proportions of MDS cases. 75% of the GS cases were used to train and validate lasso-regularized logistic regression models using 3-fold cross validation. ROC curve analysis was carried out using the remaining 25% of GS cases (Test Set 1) on the best model to identify an optimal probability cut off point for classifying subjects as MDS. Model performance was then tested on 50 subjects for which the central and local pathology diagnosis disagreed (Test Set 2), as well as on 52 additional subjects irrespective of agreement (Test Set 3). Results : The best performing logistic regression model retained 7 genes as most informative in a discriminating diagnosis of MDS from Other based on their VAFs, in order of impact: TP53, SF3B1, U2AF1, ASXL1, TET2,STAG2, and SRSF2. We used this model to assign probabilities for each of the subjects in Test Set 1 and to estimate the performance using ROC analysis (Figure 1), resulting in a high area under the curve (AUC) of 0.89. We chose a probability cut-off of ≥0.17, being associated with a high percentage of correct classification of MDS with a sensitivity and specificity of 0.90 and 0.81, respectively. Among the cohort of 50 subjects with a discordant local and central pathology diagnosis (Test Set 2), the classifier accurately reassigned 37 subjects (accuracy = 74%) from the local to the central pathology. The blinded tertiary pathology reviewer agreed with central in all Test Set 2 cases. This included 24/34 MDS cases that had been labeled as Other by local pathology (positive predictive value [PPV]=0.89). 3/16 final pathology-classified Other cases were mis-classified as MDS by the local pathologist (negative predictive value [NPV] = 0.57). Next, we assessed the ability of the model to predict MDS vs. Other for 52 additional independent subjects using the third pathologist's diagnosis to break any ties (Test Set 3). The classifier correctly predicted 15/21 MDS cases (PPV=0.83) and misclassified 6/31 Others as MDS (NPV=0.82). The overall accuracy was 83%. Conclusions: We identified that VAFs for 7 genes can correctly re-classify subjects as either MDS or Other in 74% of cases that were misclassified between local and central pathology review. Further assessment on an independent cohort showed an accuracy of 83% of the model. Taken together, these data suggest that complementing pathology reviews with targeted sequencing of 7 genes could improve MDS diagnosis. Disclosures Lindsley: MedImmune: Research Funding; Jazz Pharmaceuticals: Consultancy, Research Funding; Bluebird Bio: Consultancy; Takeda Pharmaceuticals: Consultancy. Bejar:Aptose Biosciences: Current Employment; AbbVie/Genentech: Honoraria; Astex/Otsuka: Honoraria; Takeda: Honoraria, Research Funding; Celgene/BMS: Honoraria, Research Funding; Daiichi-Sankyo: Honoraria; Forty-Seven/Gilead: Honoraria; Genoptix/NeoGenomics: Honoraria. DeZern:MEI: Consultancy; Astex: Research Funding; Abbvie: Consultancy; Celgene: Consultancy, Honoraria. Foran:H3Biosciences: Research Funding; Aptose: Research Funding; Kura Oncology: Research Funding; Trillium: Research Funding; Takeda: Research Funding; Revolution Medicine: Consultancy; Xencor: Research Funding; Agios: Honoraria, Research Funding; Aprea: Research Funding; Actinium: Research Funding; Servier: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Abbvie: Research Funding; BMS: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Boehringer Ingelheim: Research Funding. Gore:Abbvie: Consultancy, Honoraria, Research Funding. Komrokji:Acceleron: Honoraria; Incyte: Honoraria; Abbvie: Honoraria; Agios: Speakers Bureau; BMS: Honoraria, Speakers Bureau; Jazz: Honoraria, Speakers Bureau; Geron: Honoraria; Novartis: Honoraria. Maciejewski:Alexion, BMS: Speakers Bureau; Novartis, Roche: Consultancy, Honoraria. Padron:Novartis: Honoraria; BMS: Research Funding; Incyte: Research Funding; Kura: Research Funding. Starczynowski:Captor Therapeutics: Consultancy; Tolero Therapeutics: Research Funding; Kurome Therapeutics: Consultancy, Current equity holder in private company, Research Funding. Sekeres:BMS: Consultancy; Takeda/Millenium: Consultancy; Pfizer: Consultancy.
- Published
- 2020
10. Alterations in the Human Plasma Lipidome in Response to Tularemia Vaccination
- Author
-
Eric A. Ortlund, Jacob D. Franke, David A. Ford, Kristal M. Maner-Smith, Jennifer K. Colucci, Manoj Khadka, Travis L. Jensen, Daniel F. Hoft, Mark J. Mulligan, Johannes B. Goll, Carolyn J. Albert, Steve E. Bosinger, Patrick Sanz, Robert S. P. Johnson, Evan J. Anderson, Nadine Rouphael, Casey E. Gelber, and Muktha S Natrajan
- Subjects
0301 basic medicine ,oxylipins ,030231 tropical medicine ,Immunology ,lcsh:Medicine ,Inflammation ,Biology ,Article ,Tularemia ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,vaccine response ,Drug Discovery ,Lipidomics ,medicine ,Pharmacology (medical) ,Francisella tularensis ,030304 developmental biology ,Pharmacology ,0303 health sciences ,5-hydroxyeicosatetraenoic (5HETE) ,lcsh:R ,030302 biochemistry & molecular biology ,Lipid signaling ,Lipidome ,medicine.disease ,biology.organism_classification ,Tularemia vaccine ,tularemia ,3. Good health ,030104 developmental biology ,Infectious Diseases ,chemistry ,inflammation ,Cholesteryl ester ,lipidomics ,lipids (amino acids, peptides, and proteins) ,medicine.symptom - Abstract
Tularemia is a highly infectious and contagious disease caused by the bacterium Francisella tularensis. To better understand human response to a live-attenuated tularemia vaccine and the biological pathways altered post-vaccination, healthy adults were vaccinated, and plasma was collected pre- and post-vaccination for longitudinal lipidomics studies. Using tandem mass spectrometry, we fully characterized individual lipid species within predominant lipid classes to identify changes in the plasma lipidome during the vaccine response. Separately, we targeted oxylipins, a subset of lipid mediators involved in inflammatory pathways. We identified 14 differentially abundant lipid species from eight lipid classes. These included 5-hydroxyeicosatetraenoic acid (5-HETE) which is indicative of lipoxygenase activity and, subsequently, inflammation. Results suggest that 5-HETE was metabolized to a dihydroxyeicosatrienoic acid (DHET) by day 7 post-vaccination, shedding light on the kinetics of the 5-HETE-mediated inflammatory response. In addition to 5-HETE and DHET, we observed pronounced changes in 34:1 phosphatidylinositol, anandamide, oleamide, ceramides, 16:1 cholesteryl ester, and other glycerophospholipids, several of these changes in abundance were correlated with serum cytokines and T cell activation. These data provide new insights into alterations in plasma lipidome post-tularemia vaccination, potentially identifying key mediators and pathways involved in vaccine response and efficacy.
- Published
- 2020
11. Molecular Evolution of Herpes Simplex Virus 2 Complete Genomes: Comparison between Primary and Recurrent Infections
- Author
-
Lynda A. Morrison, Travis L. Jensen, Robert B. Belshe, Johannes B. Goll, Maria Korom, Miguel A. Minaya, and Sree H. Datla
- Subjects
0301 basic medicine ,Nonsynonymous substitution ,viruses ,Herpesvirus 2, Human ,Immunology ,Genome, Viral ,Biology ,medicine.disease_cause ,Microbiology ,Genome ,DNA sequencing ,Evolution, Molecular ,03 medical and health sciences ,symbols.namesake ,South Africa ,Genetic drift ,INDEL Mutation ,Molecular evolution ,Recurrence ,Virology ,medicine ,Humans ,Primary isolate ,Phylogeny ,Sanger sequencing ,Clinical Trials as Topic ,Herpes Genitalis ,Polymorphism, Genetic ,Genetic Drift ,High-Throughput Nucleotide Sequencing ,Sequence Analysis, DNA ,Virus Shedding ,030104 developmental biology ,Herpes simplex virus ,Scotland ,Genetic Diversity and Evolution ,Insect Science ,DNA, Viral ,North America ,symbols ,Female ,Virus Activation - Abstract
Herpes simplex virus 1 (HSV-1) and HSV-2 are large, double-stranded DNA viruses that cause lifelong persistent infections characterized by periods of quiescence and recurrent disease. How HSV evolves within an infected individual experiencing multiple episodes of recurrent disease over time is not known. We determined the genome sequences of viruses isolated from two subjects in the Herpevac Trial for Women who experienced primary HSV-2 genital disease and compared them with sequences of viruses isolated from the subsequent fifth or sixth episode of recurrent disease in the same individuals. Each of the HSV-2 genome sequences was initially obtained using next-generation sequencing and completed with Sanger sequencing. Polymorphisms over the entire genomes were mapped, and amino acid variants resulting from nonsynonymous changes were analyzed based on the secondary and tertiary structures of a previously crystallized protein. A phylogenetic reconstruction was used to assess relationships among the four HSV-2 samples, other North American sequences, and reference sequences. Little genetic drift was detected in viruses shed by the same subjects following repeated reactivation events, suggesting strong selective pressure on the viral genome to maintain sequence fidelity during reactivations from its latent state within an individual host. Our results also demonstrate that some primary HSV-2 isolates from North America more closely resemble the HG52 laboratory strain from Scotland than the low-passage-number clinical isolate SD90e from South Africa or laboratory strain 333. Thus, one of the sequences reported here would be a logical choice as a reference strain for inclusion in future studies of North American HSV-2 isolates. IMPORTANCE The extent to which the HSV-2 genome evolves during multiple episodes of reactivation from its latent state within an infected individual is not known. We used next-generation sequencing techniques to determine whole-genome sequences of four viral samples from two subjects in the Herpevac Trial. The sequence of each subject's well-documented primary isolate was compared with the sequence of the isolate from their fifth or sixth episode of recurrent disease. Only 19 genetic polymorphisms unique to the primary or recurrent isolate were identified, 10 in subject A and 9 in subject B. These observations indicate remarkable genetic conservation between primary and recurrent episodes of HSV-2 infection and imply that strong selection pressures exist to maintain the fidelity of the viral genome during repeated reactivations from its latent state. The genome conservation observed also has implications for the potential success of a therapeutic vaccine.
- Published
- 2017
12. 994. AS03-Adjuvanted H5N1 Avian Influenza Vaccine Modulates Early Innate Immune Signatures in Peripheral Blood Mononuclear Cells
- Author
-
Travis L. Jensen, Leigh M Howard, Heather Hill, C. Buddy Creech, Kathryn M. Edwards, Casey E. Gelber, and Johannes B. Goll
- Subjects
Abstracts ,Infectious Diseases ,Innate immune system ,Oncology ,B. Poster Abstracts ,business.industry ,Immunology ,medicine ,AS03 ,medicine.disease_cause ,business ,Peripheral blood mononuclear cell ,Influenza A virus subtype H5N1 - Abstract
Background Influenza A/H5N1 vaccines have been poorly immunogenic. Addition of Adjuvant System 03 (AS03) markedly enhances immune responses, but the mechanisms of this enhancement are unclear. Methods We compared gene expression in peripheral blood mononuclear cells (PBMCs) between recipients of AS03-adjuvanted and unadjuvanted inactivated split-virus H5N1 vaccine on days 1, 3, 7, and 28 postvaccination. We used a systems vaccinology approach to assess functional classifications of differentially expressed (DE) genes between the two vaccine groups, identify DE genes that correlate with serologic responses, and compare these findings with previous cell-specific assessments. Results AS03-adjuvanted vaccine induced the strongest differential gene expression signals on day 1 after vaccination (Figure 1). Multiple innate immune signaling pathways were activated, including the interferon, JAK-STAT, and TNF pathways, and FC gamma receptor (Fc_R) mediated phagocytosis. Immune pathways specific for antigen processing and presentation and influenza A responses were also enriched. Early differential expression of several signal transduction (day 1) and immunoglobulin (day 7) genes were predictive of peak HAI titer (Figure 2). Compared with cell-specific responses, DE gene, and immunologic pathways of PBMCs were most similar to innate immune cell subsets. However, several pathways were unique to PBMCs, and several cell-type-specific pathways, particularly from neutrophils, were absent in PBMCs (Figure 3). Conclusion Transcriptomic analysis of PBMCs after AS03-adjuvanted H5N1 vaccination revealed early differential regulation of multiple innate immune signaling pathways and enrichment of pathways involved in antigen presentation and influenza immune responses. Early expression of several genes was associated with peak HAI responses, suggesting a potential role for application of these signatures in earlier determination of vaccine responses. While PBMC and immune cell-specific results shared key innate immune signals, unique signals were identified by either approach. Disclosures L. Howard, Pfizer: Grant Investigator, Grant recipient. C. B. Creech, Pfizer: Grant Investigator, Research grant. Novartis: Grant Investigator, Research grant. K. Edwards, Novartis: Grant Investigator, Research grant. Novartis: Scientific Advisor, Consulting fee.
- Published
- 2018
13. Transcriptome Analysis in Human Breast Milk and Blood after Inactivated or Attenuated Influenza Immunization
- Author
-
Carey R. Petrie, Casey E. Gelber, Travis L. Jensen, Phillip J. Dexheimer, Johannes B. Goll, Elizabeth P. Schlaudecker, David I. Bernstein, and Mark C. Steinhoff
- Subjects
0301 basic medicine ,biology ,business.industry ,Lymphocyte ,Influenzavirus B ,Alpha interferon ,Poster Abstract ,Peripheral blood mononuclear cell ,Virology ,Immunoglobulin G ,Transcriptome ,Abstracts ,03 medical and health sciences ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Oncology ,Antigen ,Immunology ,medicine ,biology.protein ,business ,Breast feeding - Abstract
Background The goal of this study was to identify transcriptomic signatures (RNA-Seq) in human peripheral blood mononuclear cells (PBMCs) and breast milk lymphocyte cells (BMLCs) in response to trivalent inactivated influenza vaccine (TIV) or live attenuated influenza vaccine (LAIV). Methods We performed a randomized, double-blind study in breastfeeding women who received either LAIV and intramuscular placebo, or TIV and intranasal placebo. A subset of subjects with available samples (LAIV, n = 10 and TIV, n = 6) was used for this study. Human milk was collected on days 0, 2, 8, and 28, and blood samples were collected on days 0 and 28. PBMC and BMLC RNA was extracted for RNA-Seq and differentially expressed (DE) gene analysis. Results We identified a total of 382 DE BMLC genes in the LAIV group, most of which were up-regulated at day 28. DE genes were preferentially involved in innate immune signaling pathways including cytokine-cytokine receptor interaction, TNF signaling, and NF-kappa B signaling. For TIV, 3 DE genes were identified of which 2 (IL1A and IL1B) overlapped with LAIV. Response time trends for co-expressed gene clusters by vaccine group showed that LAIV generally induced an early (day 2) up-regulation of innate immune signaling pathway genes, while TIV induced peak innate immune signaling gene responses ahead of LAIV (day 8 vs. day 28). A group of known interferon-alpha/β-inducible genes (IFIT3, OAS3, IFI44L, MX1, OAS2, IFIT1, IFI6) showed higher responses at day 2 for TIV but stronger peak levels by day 28 for the LAIV group (Fig 1). While no such innate immune signaling responses were observed in PBMCs at day 28, we identified an up-regulation of IgG gene (IGHG1 and IGHG3) expression in the TIV group (Fig 2). Conclusion We observed increased innate immune signaling responses in BMLC but not in PBMC at day 28 for the LAIV group. We hypothesize that breastfeeding extends the innate response to LAIV via mucosal immunity. Gene cluster time trends indicated an earlier innate immune signaling response for TIV. The day 28 increase in IGHG3 gene expression levels in TIV group PBMCs was correlated with corresponding increases in serum ELISA IgG titers for the influenza B antigen (Fig 3). Additional studies are required to investigate the differences in innate response signaling seen for BMLC and PBMC in this study. Disclosures All authors: No reported disclosures.
- Published
- 2017
14. Proteomics show antigen presentation processes in human immune cells after AS03-H5N1 vaccination
- Author
-
Kristen L. Hoek, Sebastian Joyce, Heather Hill, Laura E. Gordy, Parimal Samir, Xinnan Niu, Travis L. Jensen, Johannes B. Goll, C. Buddy Creech, Kathryn M. Edwards, Allison C. Galassie, Leigh M Howard, Andrew J. Link, and Tara M. Allos
- Subjects
Proteomics ,0301 basic medicine ,Proteome ,Neutrophils ,T-Lymphocytes ,medicine.medical_treatment ,Antigen presentation ,Human leukocyte antigen ,Biochemistry ,Monocytes ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Adjuvants, Immunologic ,Antigen ,Influenza, Human ,MHC class I ,medicine ,Humans ,Protein Interaction Maps ,030212 general & internal medicine ,Molecular Biology ,Cells, Cultured ,Antigen Presentation ,B-Lymphocytes ,Influenza A Virus, H5N1 Subtype ,biology ,Antigen processing ,MHC class I antigen ,Killer Cells, Natural ,030104 developmental biology ,Influenza Vaccines ,Immunology ,biology.protein ,Adjuvant - Abstract
Adjuvants enhance immunity elicited by vaccines through mechanisms that are poorly understood. Using a systems biology approach, we investigated temporal protein expression changes in five primary human immune cell populations: neutrophils, monocytes, natural killer cells, T cells, and B cells after administration of either an Adjuvant System 03 adjuvanted or unadjuvanted split-virus H5N1 influenza vaccine. Monocytes demonstrated the strongest differential signal between vaccine groups. On day 3 post-vaccination, several antigen presentation-related pathways, including MHC class I-mediated antigen processing and presentation, were enriched in monocytes and neutrophils and expression of HLA class I proteins was increased in the Adjuvant System 03 group. We identified several protein families whose proteomic responses predicted seroprotective antibody responses (>1:40 hemagglutination inhibition titer), including inflammation and oxidative stress proteins at day 1 as well as immunoproteasome subunit (PSME1 and PSME2) and HLA class I proteins at day 3 in monocytes. While comparison between temporal proteomic and transcriptomic results showed little overlap overall, enrichment of the MHC class I antigen processing and presentation pathway in monocytes and neutrophils was confirmed by both approaches.
- Published
- 2017
15. Front Cover: Proteomics show antigen presentation processes in human immune cells after AS03-H5N1 vaccination
- Author
-
Xinnan Niu, Kathryn M. Edwards, Heather Hill, Travis L. Jensen, Leigh M Howard, Johannes B. Goll, Tara M. Allos, Sebastian Joyce, Kristen L. Hoek, Allison C. Galassie, Laura E. Gordy, Andrew J. Link, Parimal Samir, and C. Buddy Creech
- Subjects
Antigen presentation ,Biology ,Proteomics ,medicine.disease_cause ,Biochemistry ,Virology ,Influenza A virus subtype H5N1 ,Vaccination ,Immune system ,Front cover ,Immunology ,medicine ,AS03 ,Molecular Biology - Published
- 2017
16. Cell-Based Systems Biology Analysis of Human AS03-Adjuvanted H5N1 Avian Influenza Vaccine Responses: A Phase I Randomized Controlled Trial
- Author
-
Kristen L. Hoek, Shawn Levy, Xinnan Niu, Sebastian Joyce, Andrew J. Link, Travis L. Jensen, Allison C. Galassie, Johannes B. Goll, C. Buddy Creech, Nripesh Prasad, Heather Hill, Tara M. Allos, Kathryn M. Edwards, Laura E. Gordy, Parimal Samir, and Leigh M Howard
- Subjects
Male ,0301 basic medicine ,Neutrophils ,medicine.medical_treatment ,Gene Expression ,lcsh:Medicine ,NK cells ,Antibodies, Viral ,medicine.disease_cause ,Monocytes ,White Blood Cells ,0302 clinical medicine ,Animal Cells ,Medicine and Health Sciences ,Influenza A virus ,Public and Occupational Health ,lcsh:Science ,Immune Response ,Antigen Presentation ,Vaccines ,Multidisciplinary ,Systems Biology ,Immunogenicity ,Vaccination ,Antibody titer ,Middle Aged ,Vaccination and Immunization ,3. Good health ,Killer Cells, Natural ,Influenza Vaccines ,Female ,Cellular Types ,Adjuvant ,Research Article ,Adult ,Adolescent ,H5N1 vaccine ,Immune Cells ,Immunology ,Biology ,Young Adult ,03 medical and health sciences ,Adjuvants, Immunologic ,Double-Blind Method ,Antigen ,Influenza, Human ,Genetics ,medicine ,Humans ,AS03 ,Blood Cells ,Influenza A Virus, H5N1 Subtype ,Interleukin-6 ,lcsh:R ,Biology and Life Sciences ,Dendritic Cells ,Cell Biology ,Hemagglutination Inhibition Tests ,Virology ,Chemokine CXCL10 ,030104 developmental biology ,Antibody Formation ,lcsh:Q ,Preventive Medicine ,030215 immunology - Abstract
BACKGROUNDVaccine development for influenza A/H5N1 is an important public health priority, but H5N1 vaccines are less immunogenic than seasonal influenza vaccines. Adjuvant System 03 (AS03) markedly enhances immune responses to H5N1 vaccine antigens, but the underlying molecular mechanisms are incompletely understood.OBJECTIVE AND METHODSWe compared the safety (primary endpoint), immunogenicity (secondary), gene expression (tertiary) and cytokine responses (exploratory) between AS03-adjuvanted and unadjuvanted inactivated split-virus H5N1 influenza vaccines. In a double-blinded clinical trial, we randomized twenty adults aged 18-49 to receive two doses of either AS03-adjuvanted (n = 10) or unadjuvanted (n = 10) H5N1 vaccine 28 days apart. We used a systems biology approach to characterize and correlate changes in serum cytokines, antibody titers, and gene expression levels in six immune cell types at 1, 3, 7, and 28 days after the first vaccination.RESULTSBoth vaccines were well-tolerated. Nine of 10 subjects in the adjuvanted group and 0/10 in the unadjuvanted group exhibited seroprotection (hemagglutination inhibition antibody titer > 1:40) at day 56. Within 24 hours of AS03-adjuvanted vaccination, increased serum levels of IL-6 and IP-10 were noted. Interferon signaling and antigen processing and presentation-related gene responses were induced in dendritic cells, monocytes, and neutrophils. Upregulation of MHC class II antigen presentation-related genes was seen in neutrophils. Three days after AS03-adjuvanted vaccine, upregulation of genes involved in cell cycle and division was detected in NK cells and correlated with serum levels of IP-10. Early upregulation of interferon signaling-related genes was also found to predict seroprotection 56 days after first vaccination.CONCLUSIONSUsing this cell-based systems approach, novel mechanisms of action for AS03-adjuvanted pandemic influenza vaccination were observed.TRIAL REGISTRATIONClinicalTrials.gov NCT01573312.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.