Network


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

Hotspot


Dive into the research topics where Brandon K. Sack is active.

Publication


Featured researches published by Brandon K. Sack.


Nature Medicine | 2016

Protection against malaria at 1 year and immune correlates following PfSPZ vaccination

Andrew S. Ishizuka; Kirsten E. Lyke; Adam DeZure; Andrea A. Berry; Thomas L. Richie; Floreliz Mendoza; Mary E. Enama; Ingelise J. Gordon; Lee-Jah Chang; Uzma N Sarwar; Kathryn L. Zephir; LaSonji A. Holman; Eric R. James; Peter F. Billingsley; Anusha Gunasekera; Sumana Chakravarty; Anita Manoj; Minglin Li; Adam Ruben; Tao Li; Abraham G. Eappen; Richard E. Stafford; Natasha K C; Tooba Murshedkar; Hope DeCederfelt; Sarah Plummer; Cynthia S. Hendel; Laura Novik; Pamela Costner; Jamie G. Saunders

An attenuated Plasmodium falciparum (Pf) sporozoite (SPZ) vaccine, PfSPZ Vaccine, is highly protective against controlled human malaria infection (CHMI) 3 weeks after immunization, but the durability of protection is unknown. We assessed how vaccine dosage, regimen, and route of administration affected durable protection in malaria-naive adults. After four intravenous immunizations with 2.7 × 105 PfSPZ, 6/11 (55%) vaccinated subjects remained without parasitemia following CHMI 21 weeks after immunization. Five non-parasitemic subjects from this dosage group underwent repeat CHMI at 59 weeks, and none developed parasitemia. Although Pf-specific serum antibody levels correlated with protection up to 21–25 weeks after immunization, antibody levels waned substantially by 59 weeks. Pf-specific T cell responses also declined in blood by 59 weeks. To determine whether T cell responses in blood reflected responses in liver, we vaccinated nonhuman primates with PfSPZ Vaccine. Pf-specific interferon-γ-producing CD8 T cells were present at ∼100-fold higher frequencies in liver than in blood. Our findings suggest that PfSPZ Vaccine conferred durable protection to malaria through long-lived tissue-resident T cells and that administration of higher doses may further enhance protection.


Cell Reports | 2014

Interferon-Mediated Innate Immune Responses against Malaria Parasite Liver Stages

Jessica L. Miller; Brandon K. Sack; Michael Baldwin; Ashley M. Vaughan; Stefan H. I. Kappe

Mosquito-transmitted malaria parasites infect hepatocytes and asymptomatically replicate as liver stages. Using RNA sequencing, we show that a rodent malaria liver-stage infection stimulates a robust innate immune response including type I interferon (IFN) and IFNγ pathways. Liver-stage infection is suppressed by these infection-engendered innate responses. This suppression was abrogated in mice deficient in IFNγ, the type I IFN α/β receptor (IFNAR), and interferon regulatory factor 3. Natural killer and CD49b(+)CD3(+) natural killer T (NKT) cells increased in the liver after a primary infection, and CD1d-restricted NKT cells, which secrete IFNγ, were critical in reducing liver-stage burden of a secondary infection. Lack of IFNAR signaling abrogated the increase in NKT cell numbers in the liver, showing a link between type I IFN signaling, cell recruitment, and subsequent parasite elimination. Our findings demonstrate innate immune sensing of malaria parasite liver-stage infection and that the ensuing innate responses can eliminate the parasite.


Infection and Immunity | 2014

Model for In Vivo Assessment of Humoral Protection against Malaria Sporozoite Challenge by Passive Transfer of Monoclonal Antibodies and Immune Serum

Brandon K. Sack; Jessica L. Miller; Ashley M. Vaughan; Alyse N. Douglass; Alexis Kaushansky; Sebastian A. Mikolajczak; Alida Coppi; Gloria González-Aseguinolaza; Moriya Tsuji; Fidel Zavala; Photini Sinnis; Stefan H. I. Kappe

ABSTRACT Evidence from clinical trials of malaria vaccine candidates suggests that both cell-mediated and humoral immunity to pre-erythrocytic parasite stages can provide protection against infection. Novel pre-erythrocytic antibody (Ab) targets could be key to improving vaccine formulations, which are currently based on targeting antigens such as the circumsporozoite protein (CSP). However, methods to assess the effects of sporozoite-specific Abs on pre-erythrocytic infection in vivo remain underdeveloped. Here, we combined passive transfer of monoclonal Abs (MAbs) or immune serum with a luciferase-expressing Plasmodium yoelii sporozoite challenge to assess Ab-mediated inhibition of liver infection in mice. Passive transfer of a P. yoelii CSP MAb showed inhibition of liver infection when mice were challenged with sporozoites either intravenously or by infectious mosquito bite. However, inhibition was most potent for the mosquito bite challenge, leading to a more significant reduction of liver-stage burden and even a lack of progression to blood-stage parasitemia. This suggests that Abs provide effective protection against a natural infection. Inhibition of liver infection was also achieved by passive transfer of immune serum from whole-parasite-immunized mice. Furthermore, we demonstrated that passive transfer of a MAb against P. falciparum CSP inhibited liver-stage infection in a humanized mouse/P. falciparum challenge model. Together, these models constitute unique and sensitive in vivo methods to assess serum-transferable protection against Plasmodium sporozoite challenge.


PLOS ONE | 2013

Quantitative Bioluminescent Imaging of Pre-Erythrocytic Malaria Parasite Infection Using Luciferase-Expressing Plasmodium yoelii

Jessica L. Miller; Sara A. Murray; Ashley M. Vaughan; Anke Harupa; Brandon K. Sack; Michael Baldwin; Ian Nicholas Crispe; Stefan H. I. Kappe

The liver stages of Plasmodium parasites are important targets for the development of anti-malarial vaccine candidates and chemoprophylaxis approaches that aim to prevent clinical infection. Analyzing the impact of interventions on liver stages in the murine malaria model system Plasmodium yoelii has been cumbersome and requires terminal procedures. In vivo imaging of bioluminescent parasites has previously been shown to be an effective and non-invasive alternative to monitoring liver stage burden in the Plasmodium berghei model. Here we report the generation and characterization of a transgenic P. yoelii parasite expressing the reporter protein luciferase throughout the parasite life cycle. In vivo bioluminescent imaging of these parasites allows for quantitative analysis of P. yoelii liver stage burden and parasite development, which is comparable to quantitative RT-PCR analysis of liver infection. Using this system, we show that both BALB/cJ and C57BL/6 mice show comparable susceptibility to P. yoelii infection with sporozoites and that bioluminescent imaging can be used to monitor protective efficacy of attenuated parasite immunizations. Thus, this rapid, simple and noninvasive method for monitoring P. yoelii infection in the liver provides an efficient system to screen and evaluate the effects of anti-malarial interventions in vivo and in real-time.


Molecular therapy. Methods & clinical development | 2014

Development of gene transfer for induction of antigen-specific tolerance

Brandon K. Sack; Roland W. Herzog; Cox Terhorst; David M. Markusic

Gene replacement therapies, like organ and cell transplantation, are likely to introduce neoantigens that elicit rejection via humoral and/or effector T-cell immune responses. Nonetheless, thanks to an ever-growing body of preclinical studies; it is now well accepted that gene transfer protocols can be specifically designed and optimized for induction of antigen-specific immune tolerance. One approach is to specifically express a gene in a tissue with a tolerogenic microenvironment such as the liver or thymus. Another strategy is to transfer a particular gene into hematopoietic stem cells or immunological precursor cells thus educating the immune system to recognize the therapeutic protein as “self.” In addition, expression of the therapeutic protein in protolerogenic antigen-presenting cells such as immature dendritic cells and B cells has proven to be promising. All three approaches have successfully prevented unwanted immune responses in preclinical studies aimed at the treatment of inherited protein deficiencies, e.g., lysosomal storage disorders and hemophilia, and of type 1 diabetes and multiple sclerosis. In this review, we focus on current gene transfer protocols that induce tolerance, including gene delivery vehicles and target tissues, and discuss successes and obstacles in different disease models.


Science Translational Medicine | 2017

Complete attenuation of genetically engineered Plasmodium falciparum sporozoites in human subjects

James G. Kublin; Sebastian A. Mikolajczak; Brandon K. Sack; Matt E. Fishbaugher; Annette M. Seilie; Lisa Shelton; Tracie VonGoedert; Melike Firat; Sara Magee; Emma Fritzen; Will Betz; Heather S. Kain; Dorender A. Dankwa; Ryan W. J. Steel; Ashley M. Vaughan; D. Noah Sather; Sean C. Murphy; Stefan H. I. Kappe

A genetically attenuated Plasmodium falciparum strain called Pf GAP3KO is completely attenuated, safe, and immunogenic in adults. A triple punch knocks out the malaria parasite Vaccination with weakened infectious forms of the malaria parasite is the most promising approach to protect against malaria infection. However, creating genetically defined and weakened parasite strains that are safe for vaccination remains challenging. In a new study, Kublin et al. show that genetic engineering of the malaria parasite by the precise removal of three genes creates a parasite strain that infects humans and is well tolerated but cannot cause malaria. These genetically attenuated parasites thus appear safe for vaccination and stimulate the human immune system to generate responses that have the potential to block infection. Immunization of humans with whole sporozoites confers complete, sterilizing immunity against malaria infection. However, achieving consistent safety while maintaining immunogenicity of whole parasite vaccines remains a formidable challenge. We generated a genetically attenuated Plasmodium falciparum (Pf) malaria parasite by deleting three genes expressed in the pre-erythrocytic stage (Pf p52−/p36−/sap1−). We then tested the safety and immunogenicity of the genetically engineered (Pf GAP3KO) sporozoites in human volunteers. Pf GAP3KO sporozoites were delivered to 10 volunteers using infected mosquito bites with a single exposure consisting of 150 to 200 bites per subject. All subjects remained blood stage–negative and developed inhibitory antibodies to sporozoites. GAP3KO rodent malaria parasites engendered complete, protracted immunity against infectious sporozoite challenge in mice. The results warrant further clinical testing of Pf GAP3KO and its potential development into a vaccine strain.


Infection and Immunity | 2014

Immunization of Mice with Live-Attenuated Late Liver Stage-Arresting Plasmodium yoelii Parasites Generates Protective Antibody Responses to Preerythrocytic Stages of Malaria

Gladys J. Keitany; Brandon K. Sack; Hannah M. Smithers; Lin Chen; Ihn Kyung Jang; Leslie Sebastian; Megha Gupta; D. Noah Sather; Marissa Vignali; Ashley M. Vaughan; Stefan H. I. Kappe; Ruobing Wang

ABSTRACT Understanding protective immunity to malaria is essential for the design of an effective vaccine to prevent the large number of infections and deaths caused by this parasitic disease. To date, whole-parasite immunization with attenuated parasites is the most effective method to confer sterile protection against malaria infection in clinical trials. Mouse model studies have highlighted the essential role that CD8+ T cells play in protection against preerythrocytic stages of malaria; however, there is mounting evidence that antibodies are also important in these stages. Here, we show that experimental immunization of mice with Plasmodium yoelii fabb/f − (Pyfabb/f −), a genetically attenuated rodent malaria parasite that arrests late in the liver stage, induced functional antibodies that inhibited hepatocyte invasion in vitro and reduced liver-stage burden in vivo. These antibodies were sufficient to induce sterile protection from challenge by P. yoelii sporozoites in the absence of T cells in 50% of mice when sporozoites were administered by mosquito bite but not when they were administered by intravenous injection. Moreover, among mice challenged by mosquito bite, a higher proportion of BALB/c mice than C57BL/6 mice developed sterile protection (62.5% and 37.5%, respectively). Analysis of the antibody isotypes induced by immunization with Pyfabb/f − showed that, overall, BALB/c mice developed an IgG1-biased response, whereas C57BL/6 mice developed an IgG2b/c-biased response. Our data demonstrate for the first time that antibodies induced by experimental immunization of mice with a genetically attenuated rodent parasite play a protective role during the preerythrocytic stages of malaria. Furthermore, they highlight the importance of considering both the route of challenge and the genetic background of the mouse strains used when interpreting vaccine efficacy studies in animal models of malaria infection.


Vaccine | 2015

Novel approaches to whole sporozoite vaccination against malaria.

Else M. Bijker; Steffen Borrmann; Stefan H. I. Kappe; Benjamin Mordmüller; Brandon K. Sack; Shahid M. Khan

The parasitic disease malaria threatens more than 3 billion people worldwide, resulting in more than 200 million clinical cases and almost 600,000 deaths annually. Vaccines remain crucial for prevention and ultimately eradication of infectious diseases and, for malaria, whole sporozoite based immunization has been shown to be the most effective in experimental settings. In addition to immunization with radiation-attenuated sporozoites, chemoprophylaxis and sporozoites (CPS) is a highly efficient strategy to induce sterile protection in humans. Genetically attenuated parasites (GAP) have demonstrated significant protection in rodent studies, and are now being advanced into clinical testing. This review describes the existing pre-clinical and clinical data on CPS and GAP, discusses recent developments and examines how to transform these immunization approaches into vaccine candidates for clinical development.


PLOS Pathogens | 2015

Mechanisms of Stage-Transcending Protection Following Immunization of Mice with Late Liver Stage-Arresting Genetically Attenuated Malaria Parasites

Brandon K. Sack; Gladys J. Keitany; Ashley M. Vaughan; Jessica L. Miller; Ruobing Wang; Stefan H. I. Kappe

Malaria, caused by Plasmodium parasite infection, continues to be one of the leading causes of worldwide morbidity and mortality. Development of an effective vaccine has been encumbered by the complex life cycle of the parasite that has distinct pre-erythrocytic and erythrocytic stages of infection in the mammalian host. Historically, malaria vaccine development efforts have targeted each stage in isolation. An ideal vaccine, however, would target multiple life cycle stages with multiple arms of the immune system and be capable of eliminating initial infection in the liver, the subsequent blood stage infection, and would prevent further parasite transmission. We have previously shown that immunization of mice with Plasmodium yoelii genetically attenuated parasites (GAP) that arrest late in liver stage development elicits stage-transcending protection against both a sporozoite challenge and a direct blood stage challenge. Here, we show that this immunization strategy engenders both T- and B-cell responses that are essential for stage-transcending protection, but the relative importance of each is determined by the host genetic background. Furthermore, potent anti-blood stage antibodies elicited after GAP immunization rely heavily on FC-mediated functions including complement fixation and FC receptor binding. These protective antibodies recognize the merozoite surface but do not appear to recognize the immunodominant merozoite surface protein-1. The antigen(s) targeted by stage-transcending immunity are present in both the late liver stages and blood stage parasites. The data clearly show that GAP-engendered protective immune responses can target shared antigens of pre-erythrocytic and erythrocytic parasite life cycle stages. As such, this model constitutes a powerful tool to identify novel, protective and stage-transcending T and B cell targets for incorporation into a multi-stage subunit vaccine.


Immunity | 2017

Natural Parasite Exposure Induces Protective Human Anti-Malarial Antibodies.

Gianna Triller; Stephen W. Scally; Giulia Costa; Maria Pissarev; Cornelia Kreschel; Alexandre Bosch; Eric Marois; Brandon K. Sack; Rajagopal Murugan; Ahmed M. Salman; Chris J. Janse; Shahid M. Khan; Stefan H. I. Kappe; Ayola A. Adegnika; Benjamin Mordmüller; Elena A. Levashina; Jean-Philippe Julien; Hedda Wardemann

Summary Antibodies against the NANP repeat of circumsporozoite protein (CSP), the major surface antigen of Plasmodium falciparum (Pf) sporozoites, can protect from malaria in animal models but protective humoral immunity is difficult to induce in humans. Here we cloned and characterized rare affinity‐matured human NANP‐reactive memory B cell antibodies elicited by natural Pf exposure that potently inhibited parasite transmission and development in vivo. We unveiled the molecular details of antibody binding to two distinct protective epitopes within the NANP repeat. NANP repeat recognition was largely mediated by germline encoded and immunoglobulin (Ig) heavy‐chain complementarity determining region 3 (HCDR3) residues, whereas affinity maturation contributed predominantly to stabilizing the antigen‐binding site conformation. Combined, our findings illustrate the power of exploring human anti‐CSP antibody responses to develop tools for malaria control in the mammalian and the mosquito vector and provide a molecular basis for the structure‐based design of next‐generation CSP malaria vaccines. Graphical Abstract Figure. No caption available. HighlightsLong‐term natural Pf exposure induces weak human CSP‐memory B cell responsesAnti‐CSP memory B cell antibodies protect from Pf transmission and developmentPf‐inhibitory antibodies can recognize two distinct CSP NANP conformationsNANP repeat recognition is largely mediated by germline‐encoded residues &NA; CSP is the target of protective antibodies against the malaria parasite Plasmodium falciparum (Pf). Here, Triller and Scally et al. identified potent Pf‐inhibitory human anti‐CSP memory B cell antibodies induced by natural exposure and unveiled the molecular details of antigen binding to two protective CSP repeat epitopes.

Collaboration


Dive into the Brandon K. Sack's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adam DeZure

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew S. Ishizuka

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Cynthia S. Hendel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Floreliz Mendoza

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Hope DeCederfelt

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ingelise J. Gordon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jamie G. Saunders

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kathryn L. Zephir

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge