Darrell J Irvine - US grants
Affiliations: | Department of Biological Engineering | Massachusetts Institute of Technology, Cambridge, MA, United States |
We are testing a new system for linking grants to scientists.
The funding information displayed below comes from the NIH Research Portfolio Online Reporting Tools and the NSF Award Database.The grant data on this page is limited to grants awarded in the United States and is thus partial. It can nonetheless be used to understand how funding patterns influence mentorship networks and vice-versa, which has deep implications on how research is done.
You can help! If you notice any innacuracies, please sign in and mark grants as correct or incorrect matches.
High-probability grants
According to our matching algorithm, Darrell J Irvine is the likely recipient of the following grants.Years | Recipients | Code | Title / Keywords | Matching score |
---|---|---|---|---|
2004 — 2005 | Irvine, Darrell J | R21Activity Code Description: To encourage the development of new research activities in categorical program areas. (Support generally is restricted in level of support and in time.) |
Vaccine-Driven Modulation of Dendritic Cell Trafficking @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): In both immunization- and natural infection-driven immune responses, dendritic cells (DCs) play a critical role in initiating T cell activation, as they are the only cells known to have the capacity to prime naIve T cells in vivo. It is poorly understood how the trafficking of dendritic cells is controlled by the immunization site microenvironment, and whether or not this process can be optimized for subunit or DNA vaccines. Given the uncertainty in our understanding and the intrinsic complexity of the DC life cycle during an immune response, we would like to dissect this process through controlled release of factors that allow selective manipulation of DC trafficking. We hypothesize that vaccines which control the dynamics of dendritic cells entering, patrolling and leaving the vaccination site will have a significant impact on both the number and state of DCs reaching the lymph nodes for subsequent T cell activation, and thus on resulting immune responses. To test this hypothesis, we would like to determine the immunological consequence of modulating DC migration into and out of the immunization site quantitatively. To do this, we have devised a set of tools that enables us to modulate each step in DC trafficking precisely in vivo. Our model immunization system is comprised of two components that can be co-injected as a vaccine: microspheres that release factors which modulate DC trafficking with controlled rates, and hydrogel nanoparticles that deliver antigen and immobilized maturation factors in concert. Using this system, we will analyze the number and state of DCs at the injection site and the draining lymph node and the number and state of cognate T cells in the draining lymph node as a function of time. We will examine the effect of 1) modulating DC and DC precursor attraction to the immunization site and 2) modulating DC emigration from the immunization site to draining lymph nodes. DC trafficking will be quantitatively manipulated by immunizing with microspheres that release DC chemoattractants or modulating factors such as prostaglandins with defined release kinetics and total doses. Such studies are expected to advance our understanding of dendritic cell function, catalyze the development of more advanced materials for control of immune responses, allow the construction of a mathematical model to describe dendritic cell dynamics, and enable more rational design of vaccines. |
1 |
2004 — 2010 | Irvine, Darrell | N/AActivity Code Description: No activity code was retrieved: click on the grant title for more information |
Career: Colloidal Micelles as Multifunctional Vaccines @ Massachusetts Institute of Technology 0348259 |
0.915 |
2005 — 2006 | Irvine, Darrell J | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Imaging Cytokine Transfer At the Immunological Synapse @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): Signaling between cells can occur via receptor-receptor interactions for closely-opposed membranes or by transfer of soluble signaling molecules such as cytokines, such intercellular communications is especially key to the initiation of adaptive immune responses via the activation of T cells by antigen presenting cells (APCs). Recent studies have revealed the formation of an organized structure known as the immunological synapse between T cells and APCs during T cell activation, characterized by clustering of receptor pairs at the interface. Soluble cytokines are also known to be critical mediators of events during T cell activation, yet little is known about the transfer of these molecules from T cell to APC and vice versa during antigen recognition. To characterize cytokine transfer during T cell activation we will undertake the following specific aims: 1. We will develop a new technology aimed at detecting secreted cytokines near the surface of cells and within the immunological synapse as a means for characterizing in situ the temporal and spatial distribution of cytokines during T cell recognition via 3D time-lapse fluorescence imaging. 2. We will apply this approach to determining the spatial distribution of the key cytokine interleukin-2 (IL-2) near the surface of T cells and APCs during during T cell recognition as a function of time. 3. We will extend this technology to simultaneously characterize the spatial and temporal distribution of 3 cytokines believed to be exchanged between T cells and APCs during activation, IL-2, interleukin-12, and interferon-gamma (IFN-gamma). The proposed approach builds on existing enzyme-linked immunoabsorbent (ELISA) methods, but uses new technology for determination of local soluble molecule concentrations near the cell surface, using 3D fluorescence microscopy for direct spatially-resolved analysis. Results from these studies will 1) provide a new technology for characterizing soluble molecule transfer between cells, applicable to many biological systems, and 2) provide the first quantitative description of cytokine transfer between cells during T cell activation. Application of this new technology to this and other systems will allow the spatial and temporal complexity of cytokine signaling between cells to be directly characterized and thus allow rational therapies to be developed that target cytokine signaling. |
1 |
2007 — 2008 | Irvine, Darrell J | R21Activity Code Description: To encourage the development of new research activities in categorical program areas. (Support generally is restricted in level of support and in time.) |
Chemokine and Antigen-Carrying Nanoparticle Co-Delivery For An Oral Hiv Vaccine @ Massachusetts Institute of Technology [unreadable] DESCRIPTION (provided by applicant): In order to provide prophylactic protection against HIV, a successful vaccine will need to elicit mucosal immunity, in order to fight the virus at the mucosal sites that are major points of entry. Among the potential routes for vaccines to promote mucosal immunity, oral delivery is of great interest as a simple, needle-free mode of administration, but is made challenging by the need to protect vaccines during transit through the gastrointestinal tract and achieve sufficient uptake of the vaccine by the mucosal tissue. We have recently developed a polymeric vaccine system comprised of chemokines and antigen-loaded nanoparticles co-delivered within alginate hydrogel microsphere carriers. Alginate microspheres are candidate materials for oral drug delivery due to their stability at low pH, and alginate particles have previously been reported to promote mucosal immunity via oral vaccination. Thus, we propose that the alginate microsphere carriers we have developed can (1) protect antigen/co-delivered chemokine (or cytokine) during transit through the gastrointestinal (Gl) tract and (2) promote uptake by the gut-associated lymphoid tissues (GALT). Microparticles are known to be transcytosed by M cells overlying Peyer's patches, and extracellular deposition of alginate carriers in the GALT would allow for local generation of dendritic cell (DC)-attracting chemokine gradients that would draw DCs to these microdepots of antigen-loaded nanoparticles. We propose here to perform initial tests of this concept for the generation of oral vaccines that could prime mucosal immunity to HIV. The specific aims are: Aim 1: We will characterize the ability of alginate microsphere carriers prepared by different means to protect antigen delivery nanoparticles and co-delivered chemokines from simulated gastric fluid in vitro and to be taken up by gut mucosa in vivo, and characterize GALT uptake of alginate carriers as a function of particle size. Aim 2: Using optimal carrier sizes and compositions defined from the studies in Aim 1, we will measure immune responses triggered by nanoparticles delivering gp120 antigen and CpG alone or with co-delivery of the chemokine CCL20, and analyze antigen dissemination and dendritic cell trafficking in response to these vaccine particles. The studies described will enable the rationale design of microsphere carriers that can promote strong mucosal and systemic immune responses to recombinant HIV antigens, as a step toward a prophylactic HIV vaccine. [unreadable] [unreadable] [unreadable] |
1 |
2007 — 2009 | Irvine, Darrell J | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Dissecting Lymphocyte Chemotaxis Using Synthetic Chemokine-Releasing Microspheres @ Massachusetts Institute of Technology [unreadable] DESCRIPTION (provided by applicant): Lymphocyte trafficking is governed in part by a host of chemotactic signals encountered in lymphoid organs and peripheral tissues. How the quantitative characteristics of chemokine gradients derived from 'point' sources (the physiologic setting of attractants secreted from discrete cells) control the 3D navigation of lymphocytes through tissues and their localization in secondary lymphoid organs (SLO) remains poorly defined. We propose to use a simplified in vitro model system to quantitatively examine how lymphocytes interpret chemokine gradients released from local sources. Synthetic hydrogel microspheres that encapsulate and slowly release chemokines at predetermined rates will be used to engineer chemokine gradients in collagen gel surrogate ECMs. By fluorescently labeling human T and B lymphocytes and encapsulating a fluorescently-labeled 'tracer1 quantity of chemokine in microspheres, we will simultaneously track by videomicroscopy lymphocyte migration and the evolution of chemokine gradients over time. We will use this system to dissect aspects of T cell and B cell chemotaxis relevant to homeostasis and antigen priming in the SLO: (1) We will determine how lymphocyte migration is modulated by isolated chemokine point sources as a function of chemokine gradient characteristics. (2) To understand how lymphocytes respond to clusters of spatially discrete chemokine sources, we will use chemokine-releasing microspheres dispersed at varying density within a defined volume of a collagen matrix to determine how lymphocyte navigation through 3D volumes is influenced by the presence of multiple point sources of chemokine. (3) Using physically adjacent depots of CXCL13- and CCL21-releasing microspheres, we will determine how T and B lymphocyte self-organization is controlled by chemokine gradient characteristics, and characterize how lymphocyte activation alters T and B cell trafficking at a CXCL13/CCL21 gradient interface as a function of time and gradient characteristics. These studies will reveal the quantitative requirements for soluble chemokine gradients to induce lymphocyte chemotaxis and control lymphocyte positioning, in a system modeling the physiologic setting of discrete, 'point'-like chemokine sources. These results will have implications for understanding how trafficking in the immune system is regulated and will shed light on how chemokines might be targeted to inhibit or enhance ectopic lymphoid tissue formation for immunotherapy. [unreadable] [unreadable] [unreadable] |
1 |
2008 | Irvine, Darrell J | P01Activity Code Description: For the support of a broadly based, multidisciplinary, often long-term research program which has a specific major objective or a basic theme. A program project generally involves the organized efforts of relatively large groups, members of which are conducting research projects designed to elucidate the various aspects or components of this objective. Each research project is usually under the leadership of an established investigator. The grant can provide support for certain basic resources used by these groups in the program, including clinical components, the sharing of which facilitates the total research effort. A program project is directed toward a range of problems having a central research focus, in contrast to the usually narrower thrust of the traditional research project. Each project supported through this mechanism should contribute or be directly related to the common theme of the total research effort. These scientifically meritorious projects should demonstrate an essential element of unity and interdependence, i.e., a system of research activities and projects directed toward a well-defined research program goal. |
Lipid-Enveloped Nanoparticle Vaccines For Hiv @ Dana-Farber Cancer Institute AIDS Virus; ATGN; Acquired Immune Deficiency Syndrome Virus; Acquired Immunodeficiency Syndrome Virus; Adjuvant; Adsorption; Affect; Affinity; Antibodies; Antibody Formation; Antibody Production; Antibody Response; Antigenic Determinants; Antigens; Auricle Helix; B blood cells; B-Cell Activation; B-Cells; B-Lymphocytes; Binding; Binding (Molecular Function); Binding Determinants; Bursa-Dependent Lymphocytes; Bursa-Equivalent Lymphocyte; CD4 Positive T Lymphocytes; CD4 T cells; CD4 lymphocyte; CD4+ T cell; CD4+ T-Lymphocyte; CD4-Positive Lymphocytes; Caliber; Cell Membrane Lipids; Cells; Cells, CD4; Chemicals; Chimera Protein; Chimeric Proteins; Class Switching; Collaborations; Condition; Core Particle; Data; Dendritic Cells; Dendritic cell activation; Development; Diameter; Dioxanedione Polymer with Dimethyldioxanedione Polymer; Dose; Drugs, Nonproprietary; Encapsulated; Envelope Glycoprotein gp120, HIV; Epitopes; Epitopes, T-Lymphocyte; Figs; Figs - dietary; Fusion Protein; Generations; Generic Drugs; Glycolic-Lactic Acid Polyester; HIV; HIV Envelope Protein gp120; HIV vaccine; HIV/AIDS Vaccines; HTLV-III; HTLV-III gp120; Helix; Helix (Snails); Helix of ear; Helper Cells; Helper T-Cells; Helper-Inducer T-Lymphocyte; Human Immunodeficiency Viruses; Human T-Cell Leukemia Virus Type III; Human T-Cell Lymphotropic Virus Type III; Human T-Lymphotropic Virus Type III; Immune response; Immune system; Immunization; Immunoglobulin Class Switching; Immunologic Stimulation; Immunological Stimulation; Immunostimulation; Inducer Cells; Isotype Switching; LAV-HTLV-III; Lead; Ligand Binding; Ligands; Lipids; Lymph; Lymph node proper; Lymphadenopathy-Associated Virus; Mammals, Mice; Mediating; Membrane; Membrane Lipids; Mice; Molecular Configuration; Molecular Conformation; Molecular Interaction; Molecular Stereochemistry; Murine; Mus; Nature; Nucleosome Core; Nucleosome Core Particle; Oligo; Oligonucleotides; PADRE 45; PADRE peptide; PL-PG copolymer; PLG polymer; PLGA; Pattern; Pb element; Peptides; Phenotype; Poly(Glycolide Lactide)Copolymer; Poly(Lactide-Co-Glycolide); Poly(Lactide-Co-Glycoside); Polyglactin; Proteins; Relative; Relative (related person); Reticuloendothelial System, Lymph; Reticuloendothelial System, Lymph Node; Sensitization, Immunologic; Sensitization, Immunological; Specificity; Surface; Switchings, Class; Switchings, Immunoglobulin Class; Switchings, Isotype; T-Cell Activation; T-Cell Epitopes; T-Cells, Helper-Inducer; T-Lymphocyte Epitopes; T-Lymphocytes, Helper; T-Lymphocytes, Inducer; T4 Cells; T4 Lymphocytes; TLR protein; TLR3; TLR3 gene; TLR4; TLR4 gene; TLR7; TLR7 gene; TOLL; Testing; Toll-like receptors; Vaccines; Veiled Cells; Virion; Virus Particle; Virus-HIV; Work; World Health; antibody biosynthesis; base; biodegradable polymer; bioresorbable polymer; body system, allergic/immunologic; conformation; conformational state; degradable polymer; density; ear helix; env Protein gp120, HIV; gene product; generic; gp120; gp120 ENV Glycoprotein; gp120(HIV); hToll; heavy metal Pb; heavy metal lead; helper T cell; host response; human immunodeficiency virus vaccine; immunogen; immunoglobulin biosynthesis; immunoresponse; improved; in vivo; lymph gland; lymph nodes; lymphatic fluid; membrane structure; nano particle; nanoparticle; neutralizing antibody; novel; organ system, allergic/immunologic; pan HLA DR-binding epitope; particle; poly(glycolide-co-lactide); polylactic acid-polyglycolic acid copolymer; polylactic-co-glycolic acid copolymer; prophylactic; response; size; trafficking; uptake |
0.924 |
2009 — 2013 | Irvine, Darrell J | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Nanoparticle-Mediated Support of Cancer Immunotherapy @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): Cell-based immunotherapies are in active development for treatment of cancer, and adoptive cell therapy (ACT) of cancer with ex vivo activated/expanded T-cells is one of the more promising treatments being currently tested in patients. However, strategies to enhance the persistence and effector functions of ACT T- cells are sought, to increase the frequency of objective responses. One way to enhance the function of ACT T- cells is via genetic engineering of the lymphocytes themselves, introducing chimeric receptors or costimulatory molecules. Here we propose a complementary strategy, based on the linkage of nanoparticle drug carriers (functionalized biodegradable polymer nanoparticles) to ACT T-cells, enabling T-cells to carry and release drug cargos in their local environment. This novel approach to tumor immunotherapy offers several potential advantages over systemic adjuvant drug therapy: (1) T-cells will be uniformly exposed to drugs designed to act directly on the ACT cells themselves, irrespective of their particular tissue location and focusing the drug action on the target cells, (2) the biodistribution of drug treatment will follow the homing of T-cells and may allow a reduction in dose and limitation of systemic toxicity, and (3) sustained action of pro-immunity or anti-tumor drugs will be achieved via sustained release from cell-bound nanoparticle (NP) drug carriers. Our specific aims are: (1) We will determine the maximal NP load and basal persistence time of particle drug carriers attached to ACT T-cells transferred into tumor-bearing hosts. (2) We will test the hypothesis that interleukin cytokine support of ACT T-cells is augmented by T-cell-mediated delivery, compared to systemic injection of cytokine. (3) We will determine whether T-cell-mediated delivery of TLR ligands to tumors can substantially enhance ACT, and compare cell-mediated delivery to systemic TLR ligand injection. (4) We will determine whether combining delivery of antigen and TLR ligands as an in situ vaccine boost for ACT T-cells can provide substantial synergy in promoting anti-tumor responses. These studies will identify nanoparticle compositions that 'arm'T-cells for more effective anti-tumor function, by modulating either the function of T-cells themselves or cells in their local microenvironments, both tumor sites and secondary lymphoid organs. Public Health Relevance: Adoptive cell therapy is a promising strategy for cancer treatment, but combination therapy will likely be necessary to achieve eradication of advanced metastatic cancer. Here we propose an approach based on the attachment of biodegradable drug-loaded nanoparticles to T-lymphocytes, using T-cells as living drug delivery agents to carry therapeutics into lymphoid organs and tumor tissues. If successful, this method for engineering ACT T-cells would be simple to implement in the clinic and complementary to other strategies for augmenting ACT (such as T cell genetic engineering), and could potentially be extended to other therapeutic immune cell populations. |
1 |
2011 — 2015 | Irvine, Darrell J | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Engineered Lipid Vesicles as Potent Vaccine Vectors For Hiv @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): Successful vaccination against HIV will likely require the generation of coordinated humoral and cellular immunity. Among current strategies, protein vaccines fail to stimulate cellular responses, while plasmid DNA or live recombinant vectors capable of promoting cellular and humoral responses together face other issues, such as potency in humans (DNA), or challenges of vector-specific immunity and/or safety (live vectors). We propose here a new class of lipid-based vesicles for protein vaccine delivery, composed of multilamellar lipid vesicles stabilized by the introduction of crosslinks connecting the bilayers of the structure. These novel materials are synthesized and encapsulate protein antigens in a mild all-aqueous process, favoring the preservation of epitopes on complex antigens. In preliminary studies, we show that these novel vaccine carriers can be repeatedly administered for homologous boosting: Following a prime and 2 booster immunizations with a model antigen, these particles massively expand antigen-specific CD8+ T-cells, to the best of our knowledge substantially stronger than any previously reported protein vaccine. These T-cells were functional in terms of their capacity to produce IFN-?, and formed a substantial memory population a month after the second boost. In other preliminary studies, we found that this same system drives potent humoral responses against a candidate malaria antigen. Based on these promising initial results, we propose here to test the promise of this new vaccine delivery approach in the context of HIV vaccination, aiming to determine whether potent, durable T-cell and humoral responses can be elicited against HIV gag and env antigens. In addition, we will explore whether particular aspects of this vaccine system (e.g., the deposition of particles at the injection site) can be exploited to direct the localization of memory cells to key mucosal tissue sites that could promote protection against HIV infection. Our specific aims are: (1) How do the humoral and cellular responses to gag and env antigens evolve as a function of antigen dose and boosting with ICMV particle vaccination? (2) Are alternative clinically-relevant TLR agonist/danger signals capable of eliciting stronger immune responses than MPLA when delivered by ICMV vectors? (3) How do particles retained at the injection site influence memory/effector cell trafficking following ICMV particle immunization, compared to traditional protein immunization? (4) Can microneedle delivery be used to allow repeated needle-free particle boosting to be performed, without the need for refrigerated vaccines? Identification of ICMV formulations that can raise immune responses against HIV immunogens comparable to those shown in our preliminary data for OVA or malaria antigens will set the stage for subsequent testing in macaque protection models (either using ICMV vaccination alone or in tandem with other strong HIV vaccine candidates. |
1 |
2013 — 2017 | Irvine, Darrell J | P01Activity Code Description: For the support of a broadly based, multidisciplinary, often long-term research program which has a specific major objective or a basic theme. A program project generally involves the organized efforts of relatively large groups, members of which are conducting research projects designed to elucidate the various aspects or components of this objective. Each research project is usually under the leadership of an established investigator. The grant can provide support for certain basic resources used by these groups in the program, including clinical components, the sharing of which facilitates the total research effort. A program project is directed toward a range of problems having a central research focus, in contrast to the usually narrower thrust of the traditional research project. Each project supported through this mechanism should contribute or be directly related to the common theme of the total research effort. These scientifically meritorious projects should demonstrate an essential element of unity and interdependence, i.e., a system of research activities and projects directed toward a well-defined research program goal. |
Vaccine Adjuvant/ Delivery Systems Core @ Massachusetts General Hospital New adjuvant/delivery strategies that could enable protein and peptide vaccines to elicit both potent cellular and humoral immunity would be highly complementary to existing approaches in HIV vaccine development. In this Core, we will supply and further develop two classes of potent novel adjuvants recently developed in the Irvine lab to support the subunit vaccine studies of projects 1 and 2 in studying human immune responses to HIV immunogens in BLT humanized mice: stabilized lipid nanocapsules (SLNCs) and membrane-targeted micelles (MTMs). SLNCs are a lipid nanocapsule system for delivery of protein or peptide antigens together with molecular danger signals such as TLR agonists (TLRa), which retain entrapped antigen/TLRa much more effectively than traditional liposomes, leading to enhanced antigen/adjuvant delivery to dendritic cells and sustained accumulation of antigen in draining lymph nodes following vaccination. The second adjuvant system, membrane-targeted micelles (MTM), is based on the conjugation of molecular adjuvants or peptide antigen cargos to a micelle-forming lipid tail. These amphiphilic molecules self-assemble to form nanoparticles -15 nm in diameter in water, but also dynamically dis-assemble to anchor their lipid tails in the membrane on contact with cells. MTMs dramatically target TLRa or peptides to lymph nodes, forming intranodal vaccine depots following s.c. injection, and driving strongly enhanced immune responses to co-administered protein antigens. We will provide these potent adjuvants on demand for BLT humanized mouse immunization studies for projects 1 and 2. In addition, we will further develop these adjuvants to maximize the effectiveness in BLT mice while designing for negligible toxicity/reactogenicity, to ensure their relevance for human vaccine development. Results from these studies will further the BLT humanized mouse model as a tool for vaccine development, by identifying effective adjuvants and routes of administration for vaccine delivery, and provide new adjuvants applicable to diverse subunit vaccines beyond HIV. |
0.94 |
2013 — 2017 | Irvine, Darrell J | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Targeting Immunosuppression Blockade to T Cells For Cancer Immunotherapy @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): Cell-based immunotherapies are in active development for treatment of cancer, and adoptive cell therapy (ACT) with ex vivo activated/expanded T-cells is one of the most promising treatments currently being tested in patients. In ACT, autologous tumor-reactive T-cells are activated/expanded ex vivo and then reinfused to combat metastatic tumors. However, strategies to protect T-cells from the highly immuno-suppressive environment generated by tumors are needed to increase the frequency and durability of objective responses. We recently demonstrated that the efficacy of ACT can be dramatically enhanced by conjugation of cytokine- loaded nanoparticles (NPs) to the surfaces of T-cells ex vivo prior to transfer into tumor-bearing recipients (Stephan et al. Nat. Med. 2010). T-cell-bound particles provided pseudo-autocrine drug delivery to the transferred cells that greatly increased the effective potency of adjuvant cytokines while simultaneously eliminating systemic exposure to the drug. However, a limitation of this approach is the one-time nature of the intervention: ACT T-cells can only be loaded once with a cargo of adjuvant drug prior to transfer, and the duration of stimulation is inherently limited by expansion of the cell population in vivo. What is needed is a strategy to arm T-cells with nanoparticle drug carriers in vivo, directly in th blood or tissues, so that a single population of anti-tumor lymphocytes transferred into a patient could be repeatedly stimulated with supporting adjuvant drugs that are targeted to this cell population. We propose a set of approaches to achieve this goal, based on the injection of nanoparticles that carry immunosuppression-blocking adjuvant drugs, which are designed to bind to tumor cells and/or ACT T-cells, and which could be re-administered at will to continuously provide supporting signals to lymphocytes during ACT therapy. As a proof of concept, we will focus on delivery of a small-molecule inhibitor of the key T-cell phosphatase Shp1, which is a downstream target of a broad spectrum of negative regulatory signals in the tumor microenvironment including IL-10, TGF-¿, CTLA-4, and PD-1. We propose to test 3 major strategies, which are not mutually exclusive: (i) non-specific in situ targeting of NPs to tumor cells and lymphocytes via thiol-reactive particles; (ii) antibody-targeted NP delivery of Shp inhibitors specifically to ACT T-cells; and (iii) combined costimulation and targeting of ACT T-cells, by targeting Shp inhibitor-carrying NPs to ACT T-cells via stimulatory ligands that both target NPs to the T-cells and provide an activating/costimulatory signal to the recipient cells. As a final goal we will test whether the most potent of these approaches can synergize with vaccination to allow endogenous T-cell responses to have anti-tumor efficacy, thereby eliminating the need for adoptive transfer of T-cells for potent tumor rejection. |
1 |
2014 — 2018 | Irvine, Darrell J Jones, R. Brad |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
T-Cell-Mediated Targeting of Therapeutics to Hiv Reservoirs @ Massachusetts Institute of Technology DESCRIPTION (provided by applicant): In developed nations, the prognosis for people living with HIV has improved with the advent of anti-retroviral therapy (ART). However, ART is neither able to cure infection nor to fully restore health due, in part, to ongoing HIV replication in tisses sanctuaries. Studies in both the SIV-infected rhesus macaque and in HIV-infected humans have implicated lymphoid tissue - lymph nodes in particular - as important sites of ongoing replication. The factors governing bio-distribution of drugs are complex and multifactorial and we perceive the effective delivery of free (non-encapsulated) drug to all sites of potential HIV replication to represent a formidable challenge. The delivery of nanoparticle (NP) encapsulated drugs is an alternative, but here the efficient clearance of NP drug carriers by the reticuloendothelial system can be expected to interfere with delivery. We propose in this R01 study to adapt a technology that we have previously developed to covalently attach NPs to T-cells, thus converting them into living carriers of ARV drug cargo that we term T-Pharmacytes (TPH). A unique aspect of our approach is that NPs remain at the cell surface for weeks, and do not interfere with T-cell trafficking or effector functions. In our first Aim we will conjugate NPs to bulk CD4+ or CD8+ T-cells and take advantage of the natural homing of these cells to the lymphoid tissues to deliver ARVs to these HIV sanctuary sites in the NOD/SCID BLT mouse model. In a potentially very powerful approach, our second Aim will be to directly link NPs to well-characterized HIV-specific CTL clones that have been isolated from HIV- infected subjects, and test these in a huCD4+ NSG mouse model. In this latter approach, we will load NPs with both ARVs and an IL-15 superagonist (IL-15SA). The latter will both support the in vivo survival of adoptively transferred cells, and will act as a latency-reversing drug (LRD). We envision a chain of events whereby: (1) HIV-specific TPH will traffic into HIV sanctuary sites (2) TPH will encounter cognate antigen resulting in the lysis of productively infected cells and the local accumulation of TPH (3) IL-15SA and ARVs delivery will be concentrated at these sites resulting in i) induction of HIV expression from local latently-infected cells and ii) suppression of any new rounds of infection (4) HIV-specific TPH (enhanced in function and survival by IL-15SA) will recognize and eliminate exposed latently-infected cells. Our third Aim seeks to build upon preliminary studies to directly conjugate bulk or HIV-specific T-cells to NPs in vivo thus negating the need for ex vivo manipulations of cells. This approach will be developed and tested by a multidisciplinary team bringing expertise in nanotechnology/cell therapy, HIV T-cell immunology, and humanized mouse models of HIV infection. Ultimately our results will provide insights into the relative importance of lymphoid tissue viral reservoirs to overall persistence and may lead to therapeutic approaches comprising either the ongoing suppression of, or the eradication of, HIV reservoirs from ARV-treated individuals. |
1 |
2016 — 2019 | Irvine, Darrell J | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Lymph Node-Targeted Molecular Vaccines @ Massachusetts Institute of Technology ? DESCRIPTION (provided by applicant): Therapeutic strategies aiming to promote immune responses against tumors are of great interest for their potential to destroy metastatic cancer despite its genetic heterogeneity and ability to evade traditional chemotherapy or targeted drugs. Therapeutic vaccines are particularly compelling, because of their low toxicity and broad applicability to diverse human cancers. Recently, the first signs of therapeutic efficacy in cancer vaccines have begun to be reported, and the first cancer vaccine to be approved by the FDA (the cellular vaccine Provenge) was licensed in 2010. Vaccines based on polypeptide antigens, such as long peptides (peptides of 20-40 amino acids that can be processed and presented by diverse human HLAs) are much simpler to manufacture and have also recently begun to show signs of efficacy in patients. However, such vaccines, typically formulated as soluble polypeptides mixed with various adjuvant compounds, leave much room for improvement in terms of their potency in promoting T-cell responses. In an effort to enhance polypeptide vaccines, we sought a readily translatable strategy to enhance vaccine targeting to lymph nodes, where immune responses are initiated. Taking cues from another area of clinical cancer management, we noted that identification of sentinel lymph nodes (LNs) draining sites of primary tumor resection is often performed by the injection of dyes that avidly bind to the ubiquitous serum protein albumin. Albumin-binding dyes are efficiently carried through lymphatics and accumulate in the lymph node, allowing visual identification of the draining nodes following surgery. Mimicking this process, we designed molecular vaccines composed of peptide antigens or immunostimulatory oligonucleotides (single-stranded CpG oligos) conjugated to lipophilic tails with an intervening polymer or oligonucleotide spacer. Strikingly, when these lipid-polar block vaccine amphiphiles were synthesized with (i) lipophilic tails exhibiting high affinity for albumin and (ii) long polar spacers/cargos, they exhibited dramaticall enhanced (>10-fold) accumulation in LNs following parenteral injection relative to soluble peptide/CpG. This enhanced LN targeting of both antigens and molecular adjuvants elicited dramatically enhanced CD8+ T-cell responses, comparable to viral vectors. Based on this promising preliminary data, we propose to establish the mechanisms by which these albumin-hitchhiking amphiphile vaccines function and to test the extensibility of this approach to other adjuvant and immunomodulatory factors. In addition, we have discovered that amph-vaccines efficiently transit across the airway mucosa, and we will test the capacity of pulmonary amphiphiles to serve as simultaneous vaccines priming new T-cell responses in draining LNs and direct modulators of the lung tumor microenvironment in models of lung metastasis and primary GEM lung adenocarcinomas. |
1 |
2016 — 2020 | Irvine, Darrell J | P01Activity Code Description: For the support of a broadly based, multidisciplinary, often long-term research program which has a specific major objective or a basic theme. A program project generally involves the organized efforts of relatively large groups, members of which are conducting research projects designed to elucidate the various aspects or components of this objective. Each research project is usually under the leadership of an established investigator. The grant can provide support for certain basic resources used by these groups in the program, including clinical components, the sharing of which facilitates the total research effort. A program project is directed toward a range of problems having a central research focus, in contrast to the usually narrower thrust of the traditional research project. Each project supported through this mechanism should contribute or be directly related to the common theme of the total research effort. These scientifically meritorious projects should demonstrate an essential element of unity and interdependence, i.e., a system of research activities and projects directed toward a well-defined research program goal. |
Lymph Node-Targeted Molecular Vaccines For Mucosal Hiv Immunity @ Texas Biomedical Research Institute PROJECT SUMMARY/ABSTRACT ? PROJECT 3 Protective immunity against HIV may be greatly facilitated by the generation of strong immune defenses localized to mucosal portals of entry, where multiple mechanisms of cellular and humoral immunity can participate in virus neutralization. Projects 1 and 2 of this HIVRAD program focus on generating a mechanistic understanding of striking protection observed in macaques through a combination of IgG and IgA antibodies at mucosal surfaces; Project 3 aims to develop immunization methods capable of inducing such immune responses in concert with cellular immunity at mucosal surfaces. We have recently developed a novel strategy to target both polypeptide antigens and molecular adjuvants to lymph nodes, by conjugating these vaccine components to amphiphilic albumin-binding lipid tails, which efficiently causes albumin-mediated lymph node uptake (Liu et al. Nature 2014). This simple modification to create amphiphile (amph)-vaccines increases the T-cell response to molecular vaccines by 30-fold and humoral responses by 20-fold, and greatly increases the therapeutic efficacy of cancer vaccines in mice. This strategy for lymph node targeted adjuvant delivery simultaneously enhances the safety profile of potent molecular adjuvants, by blocking systemic dissemination. It has also recently been demonstrated that mucosal immunization can be dramatically enhanced by exploiting neonatal Fc receptor (FcRn)-mediated transcytosis of Fc-antigen fusion proteins across mucosal barriers. Because albumin is also recycled through FcRn binding, we hypothesize that amph-vaccines will be able to bind to endogenous albumin in interstitial fluid and achieve similar efficient transport across mucosal barriers to permit needle-free mucosal vaccination through the nasal mucosa to promote humoral immunity at distal reproductive tract mucosal sites. Our specific aims are: (1) Synthesize a set of candidate amphiphile-adjuvants for lymph node targeting of mucosa-draining lymph nodes; (2) Determine limits and best strategies for lymph node targeting of antigens; (3) Define pharmacokinetics and early innate immune/toxicity responses of lymph node-targeted vaccines for mucosal vaccination in non-human primates; and (4) Test the capacity of LN- targeted vaccines to enhance the immunogenicity of HIV subunit vaccines, promote mucosal cellular and humoral immunity, and promote protection against mucosal SHIV challenge in non-human primates. Results from this project will establish the translation of ?albumin hitchhiking? as a strategy to potentiate cellular and humoral to HIV at mucosal surfaces in non-human primates, and test the capacity of this promising new approach to maximize protection against heterologous SHIV challenge through induction of polyfunctional immunity at mucosal portals of entry. |
0.93 |
2018 — 2020 | Irvine, Darrell J Weiss, Ron (co-PI) [⬀] |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Synthetic Biology-Regulated Rna Vaccines @ Massachusetts Institute of Technology PI: Irvine, Darrell J. Project Summary/Abstract: Strategies to promote the magnitude and quality of T cell and antibody responses following immunization have broad relevance for the development of new prophylactic and therapeutic vaccines for the treatment of cancer and infectious diseases. Recent studies, including work from our own laboratories, have demonstrated that the kinetic pattern of antigen and adjuvant exposure to lymphoid tissues has a substantial impact on the immune response to vaccination. However, active control over the temporal pattern of antigen/inflammatory cue delivery to lymph nodes is lacking in all current vaccine approaches. Here we propose an approach applying methods from synthetic biology to create nucleic acid-based vaccines where vaccine antigen/adjuvant expression dynamics can be controlled by (i) exogenous regulation by orally-available FDA-approved small molecule drugs or (ii) intrinsically programmed in genetic circuits carried by the RNA. Based on the promising features of RNA-based vaccines, in preliminary studies we established a lipid nanoparticle-delivered self- replicating alphavirus replicon RNA as the platform for these regulated vaccines. We will systematically study the impact of vaccine antigen and adjuvant kinetics on the immune response to vaccination, create pre- programmed vaccine kinetic patterns, and test the capacity of regulated replicons to enable single-shot vaccines with prime and boost controlled by an orally-available small molecule drug. Our specific aims are (1) To optimize small molecule-regulated expression of antigen and molecular adjuvants from RNA replicons, (2) To use the regulated replicon platform to define optimal kinetics of antigen and adjuvant expression during vaccination, (3) To design RNA-based replicon genetic circuits with pre-programmed temporal patterns, and (4) To determine factors limiting replicon expression lifetimes in vivo, and engineer strategies to prolong expression toward the goal of small molecule-regulated prime-boost regimens. These studies will lead to fundamental discoveries in basic immunology, provide a framework for rationally designing immunization regimens, and create technologies to practically implement them. |
1 |
2019 | Irvine, Darrell J | R13Activity Code Description: To support recipient sponsored and directed international, national or regional meetings, conferences and workshops. |
2019 Cancer Nanotechnology Gordon Research Conference and Gordon Research Seminar @ Gordon Research Conferences PI: Irvine, Darrell J. Project Title: A booster vaccine for CAR T cells PA/INSTITUTE/STUDY SECTION ASSIGNMENTS: PA: PAR-18-648 Please assign this application to the following: Institutes/Centers: National Cancer Institute ? NCI Project Summary/Abstract The 2019 Cancer Nanotechnology Gordon Research Conference (CN-GRC) provides a unique and highly acclaimed forum where scientists at every stage of their career can share unpublished, innovative cancer research made possible by the use of nanotechnology. Held biennially since 2011, the conference has had a strong and steadily growing attendance. To further support the emerging generation of scientists in this field, the second Cancer Nanotechnology Gordon Research Seminar (CN-GRS) will be carried out in conjunction with the GRC, highlighting talks by graduate students and postdocs. This conference is an important venue bringing together chemists, physicists, engineers, biologists, clinicians, industry leaders and regulatory experts focused on the highly interdisciplinary science of cancer nanotechnology. Exciting new areas to be highlighted in the 2019 meeting include the role of nanotechnology in immune-oncology and manipulation of the microbiome to treat cancer. The conference will bring together a lively group of exceptional scientists to peaceful and scenic Mount Snow, VT in order to: 1) share cutting-edge research in cancer nanotechnology by experts representing the whole spectrum of life sciences; 2) foster discussion, exchange of ideas, and promote collaborations and 3) enhance the training and development of young scientists and promote diversity. A high priority of the CN-GRC and CN- GRS is to increase the diversity of participants. Towards this goal, funds are requested for travel and registration support of female and under-represented speakers. |
0.901 |
2019 — 2021 | Irvine, Darrell J Jones, R. Brad |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
@ Weill Medical Coll of Cornell Univ Project Abstract Although modern therapies have dramatically improved the outlooks for people living with HIV they are unable to cure infection, leaving these individuals burdened by a lifelong commitment to antiretroviral (ARV) medication. For any given individual, maintaining lifelong adherence to medication can present substantial challenges. Moreover, these expensive medications are not accessible for many individuals, in particular those in resource poor settings. It would therefore be of tremendous value to develop novel therapies that can drive HIV into remission, by which we mean into a state where levels of virus remain low or undetectable even when one stops taking ARV medication. At present, no such therapeutic intervention exists. Recent studies have shown that a type of molecule called BCL-2/BCL-XL antagonists is able to promote the death of HIV-infected cells, which could potentially lead to remission. A concern of these BCL-2/BCL-XL antagonists, however, is that they are associated with side-effects that are likely to be considered unacceptable. Relatedly, these molecules are not highly specific to HIV-infected cells and can also cause the death of some uninfected 'bystander' cells. We have developed a technology that allows for the selective targeting of drug-loaded gold nanoparticles to certain cell populations in vivo. In the current proposal we aim to use this technology to more selectively target BCL-2/BCL-XL antagonists to infected cell populations. In Aim 2, this targeting will be relatively broad ? for example, targeting all memory CD4+ T-cells. In Aim 3, we will test approaches to specifically target delivery to only HIV infected cells. For both of these approaches 'latency reversing agents (LRAs)' may also be needed to induce some expression of HIV and promote the death of infected cells. In Aim 2, these LRAs will be provided along with BCL-2/BCL-XL antagonist be co-loading gold nanoparticles. In Aim 3, LRAs will be provided first in order to induce HIV expression, allowing subsequent specific targeting of BCL-2/BCL-XL antagonists to HIV-infected cells. Our proposal will take both of these complementary approaches from in vitro experiments through to an in vivo preclinical model. Our ultimate objective is to observe efficacy of the novel therapeutics developed by this project in these preclinical models. If observed, this would enable future clinical trials of these new therapies in people living with HIV, and potentially leading to viral remission without the need for ongoing ARV therapy. |
0.901 |
2019 — 2021 | Hammond, Paula T [⬀] Irvine, Darrell J |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
@ Massachusetts Institute of Technology An immunosuppressive or immune excluded tumor microenvironment (TME) plays a key role in limiting the response of many tumor types to immunotherapy. One attractive strategy to accomplish increased lymphocyte infiltration in tumors is the use of cytokines, which can directly impact multiple immune pathways and reprogram the TME to enable a robust immune response against cancer cells. Unfortunately, despite this obvious potential, many cytokines have been limited clinically due to toxicity concerns. Rational drug delivery strategies that can rescue the therapeutic potential of cytokines could act as an important step in our ability to carefully manipulate the anti-tumor immune response in the TME and open the door for more effective immunotherapies. Nanoparticles (NPs) are a promising vehicle for the rescue of toxic cytokines. While many studies have used NPs to improve efficacy and toxicity, there remains a substantial knowledge gap surrounding the role of NP biophysical properties on enhanced delivery. There is much that is not yet understood about how nanoparticles traffic and how these differences can affect therapeutic outcomes. We are uniquely positioned to investigate the role of NP biophysical properties on cytokine delivery given our extensive experience in both NP design for targeted tumor cell delivery and in polyelectrolyte layer-by-layer (LbL) assembly. LbL-NP systems can be designed to modulate the release of multiple drugs from the core and from surrounding layers, often with time dependent staged release; whereas, manipulating the outer layer to possess certain surface chemistries and targeting moieties can significantly impact trafficking of particles on both the anatomical and cellular level. Using this system will allow for a systematic investigation of the role of these unique NP properties on effective cytokine delivery. The goal of this work is to understand and control the delivery of cytokines against solid tumors using LbL-NPs as a tool, with a focus on the impact of trafficking, localization and release kinetics of the particle and payload. Our work will focus on interleukin-12 (IL-12), one of the most potent and toxic proinflammatory cytokines for which we have recently demonstrated improved efficacy and lowered systemic toxicity by using LbL-NPs that bind to the surface membrane of ovarian cancer cells. Our studies will take place within the context of advanced serous ovarian cancer (OC), which has shown limited response to existing immunotherapies, and non-small cell lung cancers (NCSLC), which is highly responsive, but only for a defined subset of patients. IL-12 loaded NPs with external layers possessing a range of surface chemistries and targeting moieties will be examined for cellular and subcellular uptake and immune cell stimulation. Cytokine release kinetics will be examined and optimized, and nanoparticle systems will be examined in vivo for delivery of cytokines alone and in combination with anti- PD1 treatments in orthotopic syngeneic animal models. 1 |
1 |
2020 — 2021 | Irvine, Darrell J | P30Activity Code Description: To support shared resources and facilities for categorical research by a number of investigators from different disciplines who provide a multidisciplinary approach to a joint research effort or from the same discipline who focus on a common research problem. The core grant is integrated with the center's component projects or program projects, though funded independently from them. This support, by providing more accessible resources, is expected to assure a greater productivity than from the separate projects and program projects. |
@ Massachusetts Institute of Technology Microscopy Core: Project Summary/Abstract Central to the mission of the Koch Institute is to understand the molecular and cellular changes associated with cancer development and progression and to apply that knowledge to developing new treatments and diagnostic approaches. Advanced state-of-the-art imaging tools are essential for these studies. The Koch Institute Microscopy Core provides a broad range of light microscopy-based imaging systems and related image analysis platforms that enable both ex vivo and intravital imaging of nanoparticles, cells, tissues and tumors. In the current funding period, the capabilities of this Core have been expanded and enhanced. This includes the acquisition of new instrumentation and upgrades to many imaging systems, with support from Institutional funding. Importantly, we have recruited Dr. Jeffrey Kuhn to the Core Leader position. Dr. Kuhn is an interdisciplinary expert in cell biology, single molecule biochemistry, and imaging instrument and software development. Core use by Koch Institute Investigators has been strong during this funding cycle: 77% of Center Members use the Core, account for 94% of Core service use, and include investigators from all three Research Programs. In the upcoming funding period, the Microscopy Core will continue to offer a wide range of state-of-the-art services to support Center Member research programs. The Microscopy Core is committed to maintaining instrumentation and capabilities at the leading edge, and will continue to closely monitor technological improvements and evaluate emerging capabilities in the context of Center Member needs and interests. A primary goal is to conduct a hands-on and complete assessment of Core capabilities and Center Member needs with respect to both instrumentation and data analysis capabilities. Replacement of older instruments and acquisition of new capabilities and upgrades will be guided by Center Member research needs and will be funded by Institutional sources. Specific planned initiatives include: enhancing expertise in single molecule localization microscopy; expanding hands-on training opportunities; developing resources to support advanced image processing, visualization, quantitation and machine learning; and collaborating with other Koch Institute Cores to develop a monthly workshop series to support Center Members? access to imaging technology platforms and data analysis. This Shared Resource is essential to the success of the Koch Institute mission and provides exceptional value to the CCSG. The requested CCSG budget for Year 49 is increased by 27.2% over the Core CCSG budget for the current period (Year 48), reflecting increased support for expert personnel. The CCSG budgets of other Koch Institute Cores have been reduced to offset the proposed increase in this, and other, Cores. |
1 |
2020 — 2021 | Irvine, Darrell J | R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Enhancing Car-T Cell Activity Against Solid Tumors by Vaccine Boosting Through the Chimeric Receptor @ Massachusetts Institute of Technology Project Summary/Abstract 30 lines or less: Adoptive cell therapy (ACT) with chimeric antigen receptor (CAR) T cells has shown dramatic clinical responses in hematologic cancers, with a high proportion of durable complete remissions elicited in leukemias and lymphomas. This success has led to a strong commercial investment establishing adoptive cell therapy as a viable clinical therapy and the first licensure of CAR-T therapy by the FDA in 2017. However, achieving the full promise of CAR-T ACT, especially in solid tumors, will require further advances in this form of cellular therapy. A key challenge is maintaining a sufficient pool of functional CAR-T cells in vivo. In addition, even in hematologic tumors treated effectively with CAR T cells, chemotherapeutic lymphodepletion regimens with high toxicity are often required to ensure the engraftment and initial expansion of the donor cells. We recently discovered an efficient strategy for molecularly targeting peptide vaccines and vaccine adjuvants to lymph nodes, through the use of albumin-binding phospholipid-polymer linkers conjugated to antigens/molecular adjuvants. Albumin constitutively traffics from blood to lymph, and serves as an effective chaperone to concentrate these ?amphiphile-vaccine? (amph-vax) components in lymph nodes that would otherwise be rapidly dispersed in the bloodstream following parenteral injection. However, these lipid-polymer conjugates also exhibit the property that they insert in cell membranes on arrival in lymph nodes. We propose here to exploit these dual lymph node targeting and membrane-decorating properties of amph-vax molecules to create a booster vaccine for CAR T cells, which can be used to repeatedly expand and rejuvenate CAR-T directly in vivo- in native lymph nodes and/or tumors. To evaluate this approach in the presence of a complete host immune system, we will test this concept both with human T cells and with an immunocompetent syngeneic mouse model of melanoma recently developed by our lab. Our specific aims are to (1) Characterize the biology of synthetic antigen presentation of amphiphile-ligands from the surface of antigen presenting cells to CAR T cells in vivo, (2) to demonstrate an amph-vax design generalizable to any CAR, (3) to evaluate the capacity of a CAR-T vaccine to expand T cells with enhanced functionality and persistence in vivo, and (4) to test the utility of intratumoral amph-vax delivery to enhance CAR-T and endogenous T cell priming in tandem. These studies will establish a robust technology platform to transform multiple aspects of adoptive cell therapy and address key limitations in existing ACT therapeutic strategies. |
1 |
2021 | Irvine, Darrell J Wittrup, Karl D |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Localized Immunotherapy Using Alum-Binding Therapeutics @ Massachusetts Institute of Technology Project Summary/Abstract Combination treatments aiming to stimulate synergistic immune pathways employing cytokines or immunomodulatory antibodies are generally more effective than monotherapies in preclinical models of cancer immunotherapy. However when given systemically, these combination treatments suffer from high toxicity from on-target off-tumor stimulation as well as low local concentrations at the tumor site due to poor tumor penetrance and high clearance rates. Local intratumoral therapy is a viable approach to bypass some of the challenges associated with systemic delivery, but requires optimization to promote retention of the therapeutic agent at the injection site and minimize leakage into the circulation. We have recently developed an approach to enhance vaccine efficacy by engineering the binding of immunogens to the commonly used adjuvant aluminum hydroxide (alum) via a site-specific phosphoserine (pSer) peptide tag. The pSer moieties undergo a ligand-exchange reaction with free hydroxyl groups on the surface of alum leading to stable anchoring of proteins on alum particles. We propose here to apply this alum-anchoring platform in the context of cancer to retain potent immune agonists within the tumor site, promoting a robust systemic immune response with minimal toxicity. Our preliminary results show that this simple approach can be used to load stimulatory cytokines onto alum for retention at the tumor site up to a month, stimulating a strong anti-tumor response from a single shot treatment. We plan to develop and optimize this translational strategy through the following specific aims: (1) use in-cell phosphorylation to produce phosphoserine-tagged cytokines and other candidate immune agonists for optimal alum binding, (2) determine optimal treatment regimens for these intratumoral alum-bound therapeutic agents in vivo in multiple tumor models, (3) define the mechanism of action through which this therapy elicits a response, (4) evaluate the systemic immune response and assess strategies to enhance abscopal effects by promoting the transfer of immunostimulatory payloads to motile lymphocytes for trafficking to distal untreated tumors. These studies will establish a robust technology platform capable of safely delivering treatments currently viewed as too toxic, by addressing key limitations in existing localized therapeutic strategies |
1 |
2021 | Bathe, Mark [⬀] Irvine, Darrell J |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Investigation of Synthetic Dna-Based Viral Particles For Spatially Controlled Antigen Presentation @ Massachusetts Institute of Technology PROJECT SUMMARY Strategies to enhance antigenicity, antibody affinity maturation, and memory induction in response to subunit vaccines are of broad relevance for the design of effective vaccines against infectious diseases, and may be especially important for difficult-to-neutralize pathogens such as HIV. One approach to enhance the efficacy of subunit vaccines is to formulate antigens in a multivalent, nanoparticulate form, which promotes several aspects of humoral immunity, and most notably enhances crosslinking of B cell receptors (BCRs). This approach has been exploited both in licensed vaccines (e.g., the HPV and HBV vaccines), and in a great variety of vaccines in preclinical and clinical development. However, to date it remains unclear what are the ideal characteristics of nanoparticle antigen display. In this project, we use the unique technology of scaffolded DNA origami to engineer nanoparticles on the 10?100 nanometer scale that offer the ability to investigate the impact of scaffold size, antigen copy number up to more than 100, antigen-BCR affinity, as well as the nanoscale spatial organization and dimensionality of antigen presentation on BCR activation. Specifically, we test the relative importance of these parameters on B-cell activation, which are of central importance to the development of a successful subunit vaccines, using the germline targeting engineered outer domain of HIV-1 gp120, termed eOD-GT8, and its variants with different affinities, as a testbed. In vitro evaluation of early B-cell signaling and pathway activation will be characterized, and contrasted with the benchmark strongly activating 60-mer control organized on a protein scaffold. Single-cell fluorescence imaging is used to investigate the detailed mechanism of BCR-binding and B-cell activation based on the optimal immunogen presentation found. These constructs are then used to test the impact of these optimal HIV DNA-NP constructs on T-cell and B-cell response in vivo using mouse models. Taken together, our results will offer the elucidation of the optimal immunogen presentation parameters for effective immune cell response in the development of more effective subunit vaccines, with major translational potential for HIV and other infectious diseases. |
1 |
2021 | Irvine, Darrell J Weiss, Ron (co-PI) [⬀] |
R01Activity Code Description: To support a discrete, specified, circumscribed project to be performed by the named investigator(s) in an area representing his or her specific interest and competencies. |
Spongebot: Genetically Engineered Cells to Suppress Sars-Cov-2 and Future Viruses @ Massachusetts Institute of Technology SpongeBot represents a new class of genetically modified cells to address the COVID-19 pandemic, leveraging a novel antiviral platform developed under the proposer team?s existing NIH NIBIB R01 project. This antiviral platform facilitates rapid, targeted SpongeBot development and deployment against SARS-CoV-2, its viral mutations, as well as entirely new viruses - providing a barrier to future viral pandemics. SARS-CoV-2 is highly communicable and individuals can transmit the disease even prior to becoming symptomatic, sharply increasing the rate of disease spread. During the first two weeks following infection, the innate immune system attempts to slow down the rapidly multiplying pathogen to provide time for the adaptive immune system to develop more specific and effective mechanisms to destroy the virus. However, in individuals with decreased or compromised immune responses, the excessive viral load can lead to elevated inflammation, severe tissue damage, and ultimately death. SpongeBot, our bioengineered cell-based therapy solution, provides vital support to the body?s immune system, through its genetically designed ability to sequester and destroy SARS-CoV-2 viral particles at sites of injury, in addition to attenuation of the immune system?s hyperinflammatory response to the virus. Administering SpongeBot cells to an infected individual reduces and keeps viral load below dangerous thresholds, prevents harmful hyperinflammation, and provides the adaptive immune system the time required to mount an effective defense against the virus. SpongeBot can be administered prophylactically to at-risk populations (e.g., healthcare workers, the elderly, or immunocompromised individuals), or therapeutically at any stage during the course of viral infection. Importantly, SpongeBot therapy is extremely safe; the base technology has a long proven clinical safety track record. Unlike the lengthy development times necessary for vaccines or antiviral medications, a targeted SpongeBot therapy against a predicted virus can be placed in clinical trials immediately. SpongeBot development for a novel virus would be ready for deployment in only about 12 weeks. |
1 |
2021 | Crotty, Shane P Irvine, Darrell J |
R61Activity Code Description: As part of a bi-phasic approach to funding exploratory and/or developmental research, the R61 provides support for the first phase of the award. This activity code is used in lieu of the R21 activity code when larger budgets and/or project periods are required to establish feasibility for the project. |
Immune Engineering of Optimized Sequential Immunization Strategies For Hiv Vaccines @ Massachusetts Institute of Technology Project Summary Preclinical studies and early stage human trials evaluating passively transferred broadly neutralizing antibodies (bnAbs) suggest that a vaccine capable of eliciting bnAbs would provide effective protection from HIV infection. However, the difficulty of inducing bnAbs through vaccination has led to a focus in the field on vaccine strategies based on sequential immunizations meant to guide the developing B cell response. These sequential immunization strategies range from germline targeting to lineage-guided design to immunofocusing, and combinations thereof. While logical from a structural biology perspective, important immunological questions remain unanswered for such vaccines: In such a strategy, at what interval should sequential immunogens be administered? How does competition from antigen-specific but non-neutralizing B cell precursors impact vaccine ?shepherding?? Is this sequential immunization process hindered by limited T cell help? In addition, vaccines comprised of 4 or more injections will be a challenge to implement globally. How do we make such a vaccine practical? In this phase R61/R33 application, we propose systematic studies in small animal models to evaluate fundamental vaccine immunology issues facing such strategies. We proposed several novel approaches to examine, and potentially solve, these issues: In aim 1 we will characterize the immunology of staggered sequential immunizations, in aim 2 we develop an approach to delete competitor B cells during vaccination, in aim 3 we develop vaccines employing augmented T cell help, and in aim 4, we propose technologies to enable sequential immunogen exposure following a single injection. We will test these concepts in the context of vaccines aiming to elicit bnAbs against the CD4 binding site (VRC01-class responses) and the Env fusion peptide, using physiologically relevant conditions. The most impactful of these immunization strategies will be downselected for testing in non-human primates (NHP) during the R33 phase, the animal model for HIV immunization closest to humans. Our work is guided by recent advances by the Irvine, Crotty, and Silvestri labs in understand GC kinetics, bnAb B cell competition,vaccine delivery systems, novel adjuvants, and roles of T cell help in rare B cell recruitment. |
1 |