Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Timothy Brauns is active.

Publication


Featured researches published by Timothy Brauns.


The Journal of Infectious Diseases | 2004

Age-Associated Decline in Resistance to Babesia microti Is Genetically Determined

Edouard Vannier; Ingo Borggraefe; Sam R. Telford; Sanjay Menon; Timothy Brauns; Andrew Spielman; Jeffrey A. Gelfand; Henry H. Wortis

BACKGROUND Although infection by the protozoan Babesia microti is rarely symptomatic in immunocompetent young people, healthy individuals aged >50 years may experience life-threatening disease. To determine the basis for this age relationship, we developed a mouse model of babesiosis using a novel clinical isolate of B. microti. METHODS Mice were infected at 2, 6, 12, or 18 months. Parasitemia was monitored on Giemsa-stained blood smears or by flow cytometry. RESULTS In DBA/2 mice, early and persistent parasitemias increased with age at infection. BALB/c and C57BL/6 mice were resistant, regardless of age, which indicates that allelic variation determines resistance to B. microti. Unlike immunocompetent mice, SCID mice, which retain an innate immune system but lack the lymphocytes needed for adaptive immunity, developed high and persistent levels of parasitemia that were markedly reduced by transfer of naive BALB/c or DBA/2 splenocytes. BALB/c cells reduced the persistent parasitemia to a greater extent than did age-matched DBA/2 cells. Of importance, there was an age-associated loss of protection by cells of both strains. CONCLUSION The resistance to B. microti infection conferred by the adaptive immune system is genetically determined and associated with age. We postulate that there are age-related differences in the expression of alleles critical for adaptive immunity to B. microti.


PLOS ONE | 2013

Near-infrared laser adjuvant for influenza vaccine.

Satoshi Kashiwagi; Jianping Yuan; Benjamin Forbes; Mathew L. Hibert; Eugene L. Q. Lee; Laura Whicher; Calum Goudie; Yuan Yang; Tao Chen; Beth Edelblute; Brian Collette; Laurel Edington; James Trussler; Jean Nezivar; Pierre Leblanc; Roderick T. Bronson; Kosuke Tsukada; Makoto Suematsu; Jeffrey S. Dover; Timothy Brauns; Jeffrey A. Gelfand; Mark C. Poznansky

Safe and effective immunologic adjuvants are often essential for vaccines. However, the choice of adjuvant for licensed vaccines is limited, especially for those that are administered intradermally. We show that non-tissue damaging, near-infrared (NIR) laser light given in short exposures to small areas of skin, without the use of additional chemical or biological agents, significantly increases immune responses to intradermal influenza vaccination without augmenting IgE. The NIR laser-adjuvanted vaccine confers increased protection in a murine influenza lethal challenge model as compared to unadjuvanted vaccine. We show that NIR laser treatment induces the expression of specific chemokines in the skin resulting in recruitment and activation of dendritic cells and is safe to use in both mice and humans. The NIR laser adjuvant technology provides a novel, safe, low-cost, simple-to-use, potentially broadly applicable and clinically feasible approach to enhancing vaccine efficacy as an alternative to chemical and biological adjuvants.


Journal of Hematology & Oncology | 2014

A novel mycobacterial Hsp70-containing fusion protein targeting mesothelin augments antitumor immunity and prolongs survival in murine models of ovarian cancer and mesothelioma

Jianping Yuan; Satoshi Kashiwagi; Patrick Reeves; Jean Nezivar; Yuan Yang; Nadiah Hashim Arrifin; Mai Nguyen; Gilberte Jean-Mary; Xiaoyun Tong; Paramjit Uppal; Svetlana Korochkina; Ben Forbes; Tao Chen; Elda Righi; Roderick T. Bronson; Huabiao Chen; Sandra Orsulic; Timothy Brauns; Pierre Leblanc; Nathalie Scholler; Glenn Dranoff; Jeffrey A. Gelfand; Mark C. Poznansky

BackgroundAlthough dendritic cell (DC) vaccines are considered to be promising treatments for advanced cancer, their production and administration is costly and labor-intensive. We developed a novel immunotherapeutic agent that links a single-chain antibody variable fragment (scFv) targeting mesothelin (MSLN), which is overexpressed on ovarian cancer and mesothelioma cells, to Mycobacterium tuberculosis (MTB) heat shock protein 70 (Hsp70), which is a potent immune activator that stimulates monocytes and DCs, enhances DC aggregation and maturation and improves cross-priming of T cells mediated by DCs.MethodsBinding of this fusion protein with MSLN on the surface of tumor cells was measured by flow cytometry and fluorescence microscopy. The therapeutic efficacy of this fusion protein was evaluated in syngeneic and orthotopic mouse models of papillary ovarian cancer and malignant mesothelioma. Mice received 4 intraperitoneal (i.p.) treatments with experimental or control proteins post i.p. injection of tumor cells. Ascites-free and overall survival time was measured. For the investigation of anti-tumor T-cell responses, a time-matched study was performed. Splenocytes were stimulated with peptides, and IFNγ- or Granzyme B- generating CD3+CD8+ T cells were detected by flow cytometry. To examine the role of CD8+ T cells in the antitumor effect, we performed in vivo CD8+ cell depletion. We further determined if the fusion protein increases DC maturation and improves antigen presentation as well as cross-presentation by DCs.ResultsWe demonstrated in vitro that the scFvMTBHsp70 fusion protein bound to the tumor cells used in this study through the interaction of scFv with MSLN on the surface of these cells, and induced maturation of bone marrow-derived DCs. Use of this bifunctional fusion protein in both mouse models significantly enhanced survival and slowed tumor growth while augmenting tumor-specific CD8+ T-cell dependent immune responses. We also demonstrated in vitro and in vivo that the fusion protein enhanced antigen presentation and cross-presentation by targeting tumor antigens towards DCs.ConclusionsThis new cancer immunotherapy has the potential to be cost-effective and broadly applicable to tumors that overexpress mesothelin.


Cancer | 2005

A novel automated screening and interpretation process for cervical cytology using the internet transmission of low-resolution images: a feasibility study.

John H. Eichhorn; Timothy Brauns; Jeffrey A. Gelfand; Barbara A. Crothers; David C. Wilbur

Transmission over the Internet of low‐resolution images acquired by automated screening of cervical cytology specimens has the potential to provide remote interpretation and, hence, centralization of a cytology workforce.


Human Vaccines & Immunotherapeutics | 2014

Laser vaccine adjuvants. History, progress, and potential.

Satoshi Kashiwagi; Timothy Brauns; Jeffrey A. Gelfand; Mark C. Poznansky

Immunologic adjuvants are essential for current vaccines to maximize their efficacy. Unfortunately, few have been found to be sufficiently effective and safe for regulatory authorities to permit their use in vaccines for humans and none have been approved for use with intradermal vaccines. The development of new adjuvants with the potential to be both efficacious and safe constitutes a significant need in modern vaccine practice. The use of non-damaging laser light represents a markedly different approach to enhancing immune responses to a vaccine antigen, particularly with intradermal vaccination. This approach, which was initially explored in Russia and further developed in the US, appears to significantly improve responses to both prophylactic and therapeutic vaccines administered to the laser-exposed tissue, particularly the skin. Although different types of lasers have been used for this purpose and the precise molecular mechanism(s) of action remain unknown, several approaches appear to modulate dendritic cell trafficking and/or activation at the irradiation site via the release of specific signaling molecules from epithelial cells. The most recent study, performed by the authors of this review, utilized a continuous wave near-infrared laser that may open the path for the development of a safe, effective, low-cost, simple-to-use laser vaccine adjuvant that could be used in lieu of conventional adjuvants, particularly with intradermal vaccines. In this review, we summarize the initial Russian studies that have given rise to this approach and comment upon recent advances in the use of non-tissue damaging lasers as novel physical adjuvants for vaccines.


Human Vaccines & Immunotherapeutics | 2014

VaxCelerate II: Rapid development of a self-assembling vaccine for Lassa fever

Pierre Leblanc; Leonard Moise; Cybelle Luza; Kanawat Chantaralawan; Lynchy Lezeau; Jianping Yuan; Mary Field; Daniel Richer; Christine M. Boyle; William D. Martin; Jordan B. Fishman; Eric Berg; David Baker; Brandon Zeigler; Dale Mais; William R. Taylor; Russell Coleman; H. Shaw Warren; Jeffrey A. Gelfand; Anne S. De Groot; Timothy Brauns; Mark C. Poznansky

Development of effective vaccines against emerging infectious diseases (EID) can take as much or more than a decade to progress from pathogen isolation/identification to clinical approval. As a result, conventional approaches fail to produce field-ready vaccines before the EID has spread extensively. Lassa is a prototypical emerging infectious disease endemic to West Africa for which no successful vaccine is available. We established the VaxCelerate Consortium to address the need for more rapid vaccine development by creating a platform capable of generating and pre-clinically testing a new vaccine against specific pathogen targets in less than 120 d. A self-assembling vaccine is at the core of the approach. It consists of a fusion protein composed of the immunostimulatory Mycobacterium tuberculosis heat shock protein 70 (MtbHSP70) and the biotin binding protein, avidin. Mixing the resulting protein (MAV) with biotinylated pathogen-specific immunogenic peptides yields a self-assembled vaccine (SAV). To meet the time constraint imposed on this project, we used a distributed R&D model involving experts in the fields of protein engineering and production, bioinformatics, peptide synthesis/design and GMP/GLP manufacturing and testing standards. SAV immunogenicity was first tested using H1N1 influenza specific peptides and the entire VaxCelerate process was then tested in a mock live-fire exercise targeting Lassa fever virus. We demonstrated that the Lassa fever vaccine induced significantly increased class II peptide specific interferon-γ CD4+ T cell responses in HLA-DR3 transgenic mice compared to peptide or MAV alone controls. We thereby demonstrated that our SAV in combination with a distributed development model may facilitate accelerated regulatory review by using an identical design for each vaccine and by applying safety and efficacy assessment tools that are more relevant to human vaccine responses than current animal models.


Vaccine | 2017

Semiconductor diode laser device adjuvanting intradermal vaccine

Yoshifumi Kimizuka; John J. Callahan; Zilong Huang; Kaitlyn Morse; Wataru Katagiri; Ayako Shigeta; Roderick T. Bronson; Shu Takeuchi; Yusuke Shimaoka; Megan P.K. Chan; Yang Zeng; Binghao Li; Huabiao Chen; Rhea Y.Y. Tan; Conor Dwyer; Tyler Mulley; Pierre Leblanc; Calum Goudie; Jeffrey A. Gelfand; Kosuke Tsukada; Timothy Brauns; Mark C. Poznansky; David M. Bean; Satoshi Kashiwagi

A brief exposure of skin to a low-power, non-tissue damaging laser light has been demonstrated to augment immune responses to intradermal vaccination. Both preclinical and clinical studies show that this approach is simple, effective, safe and well tolerated compared to standard chemical or biological adjuvants. Until now, these laser exposures have been performed using a diode-pumped solid-state laser (DPSSL) devices, which are expensive and require labor-intensive maintenance and special training. Development of an inexpensive, easy-to-use and small device would form an important step in translating this technology toward clinical application. Here we report that we have established a handheld, near-infrared (NIR) laser device using semiconductor diodes emitting either 1061, 1258, or 1301nm light that costs less than


Cancer immunology research | 2018

AMD3100 Augments the Efficacy of Mesothelin-Targeted, Immune-Activating VIC-008 in Mesothelioma by Modulating Intratumoral Immunosuppression

Binghao Li; Yang Zeng; Patrick Reeves; Chongzhao Ran; Qiuyan Liu; Xiying Qu; Yingying Liang; Zhao Liu; Jianping Yuan; Pierre Leblanc; Zhaoming Ye; Ann E. Sluder; Jeffrey A. Gelfand; Timothy Brauns; Huabiao Chen; Mark C. Poznansky

4000, and that this device replicates the adjuvant effect of a DPSSL system in a mouse model of influenza vaccination. Our results also indicate that a broader range of NIR laser wavelengths possess the ability to enhance vaccine immune responses, allowing engineering options for the device design. This small, low-cost device establishes the feasibility of using a laser adjuvant approach for mass-vaccination programs in a clinical setting, opens the door for broader testing of this technology with a variety of vaccines and forms the foundation for development of devices ready for use in the clinic.


Journal of Immunology | 2017

Near-infrared 1064 nm laser modulates migratory dendritic cells to augment the immune response to intradermal influenza vaccine

Kaitlyn Morse; Yoshifumi Kimizuka; Megan P.K. Chan; Mai Shibata; Yusuke Shimaoka; Shu Takeuchi; Benjamin Forbes; C. Nirschl; Binghao Li; Yang Zeng; Roderick T. Bronson; Wataru Katagiri; Ayako Shigeta; Ruxandra F. Sîrbulescu; Huabiao Chen; Rhea Y.Y. Tan; Kosuke Tsukada; Timothy Brauns; Jeffrey A. Gelfand; Ann E. Sluder; Joseph J. Locascio; Mark C. Poznansky; Niroshana Anandasabapathy; Satoshi Kashiwagi

CXCR4 antagonist AMD3100 promoted conversion of Tregs to helper-like cells and reduced PD-1 expression on CD8+ T cells in mesothelioma. The combination of AMD31000 with a mesothelin-targeting immune stimulator, VIC-008, controlled tumors and prolonged survival of tumor-bearing animals. AMD3100 (plerixafor), a CXCR4 antagonist, has been demonstrated to suppress tumor growth and modulate intratumoral T-cell trafficking. However, the effect of AMD3100 on immunomodulation remains elusive. Here, we explored immunomodulation and antitumor efficacy of AMD3100 in combination with a previously developed mesothelin-targeted, immune-activating fusion protein, VIC-008, in two syngeneic, orthotopic models of malignant mesothelioma in immunocompetent mice. We showed that combination therapy significantly suppressed tumor growth and prolonged animal survival in two mouse models. Tumor control and survival benefit were associated with enhanced antitumor immunity. VIC-008 augmented mesothelin-specific CD8+ T-cell responses in the spleen and lymph nodes and facilitated intratumoral lymphocytic infiltration. However, VIC-008 treatment was associated with increased programmed cell death protein-1 (PD-1) expression on intratumoral CD8+ T cells, likely due to high CXCL12 in the tumor microenvironment. AMD3100 alone and in combination with VIC-008 modulated immunosuppression in tumors and the immune system through suppression of PD-1 expression on CD8+ T cells and conversion of regulatory T cells (Tregs) into CD4+CD25–Foxp3+IL2+CD40L+ helper-like cells. In mechanistic studies, we demonstrated that AMD3100-driven Treg reprogramming required T cell receptor (TCR) activation and was associated with loss of PTEN due to oxidative inactivation. The combination of VIC-008 augmentation of tumor-specific CD8+ T-cell responses with AMD3100 abrogation of immunosuppression conferred significant benefits for tumor control and animal survival. These data provide new mechanistic insight into AMD3100-mediated immunomodulation and highlight the enhanced antitumor effect of AMD3100 in combination with a tumor antigen–targeted therapy in mouse malignant mesothelioma, which could be clinically relevant to patients with this difficult-to-treat disease. Cancer Immunol Res; 6(5); 539–51. ©2018 AACR.


Human Vaccines & Immunotherapeutics | 2017

Q-vaxcelerate: A distributed development approach for a new Coxiella burnetii vaccine

Patrick Reeves; Susan Raju Paul; Ann E. Sluder; Timothy Brauns; Mark C. Poznansky

Brief exposure of skin to near-infrared (NIR) laser light has been shown to augment the immune response to intradermal vaccination and thus act as an immunologic adjuvant. Although evidence indicates that the NIR laser adjuvant has the capacity to activate innate subsets including dendritic cells (DCs) in skin as conventional adjuvants do, the precise immunological mechanism by which the NIR laser adjuvant acts is largely unknown. In this study we sought to identify the cellular target of the NIR laser adjuvant by using an established mouse model of intradermal influenza vaccination and examining the alteration of responses resulting from genetic ablation of specific DC populations. We found that a continuous wave (CW) NIR laser adjuvant broadly modulates migratory DC (migDC) populations, specifically increasing and activating the Lang+ and CD11b−Lang− subsets in skin, and that the Ab responses augmented by the CW NIR laser are dependent on DC subsets expressing CCR2 and Langerin. In comparison, a pulsed wave NIR laser adjuvant showed limited effects on the migDC subsets. Our vaccination study demonstrated that the efficacy of the CW NIR laser is significantly better than that of the pulsed wave laser, indicating that the CW NIR laser offers a desirable immunostimulatory microenvironment for migDCs. These results demonstrate the unique ability of the NIR laser adjuvant to selectively target specific migDC populations in skin depending on its parameters, and highlight the importance of optimization of laser parameters for desirable immune protection induced by an NIR laser–adjuvanted vaccine.

Collaboration


Dive into the Timothy Brauns's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge