Network


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

Hotspot


Dive into the research topics where Kelly D. Moynihan is active.

Publication


Featured researches published by Kelly D. Moynihan.


Nature | 2014

Structure-based programming of lymph-node targeting in molecular vaccines

Haipeng Liu; Kelly D. Moynihan; Yiran Zheng; Gregory L. Szeto; Adrienne V. Li; Bonnie Huang; Debra S. Van Egeren; Clara Park; Darrell J. Irvine

In cancer patients, visual identification of sentinel lymph nodes (LNs) is achieved by the injection of dyes that bind avidly to endogenous albumin, targeting these compounds to LNs, where they are efficiently filtered by resident phagocytes. Here we translate this ‘albumin hitchhiking’ approach to molecular vaccines, through the synthesis of amphiphiles (amph-vaccines) comprising an antigen or adjuvant cargo linked to a lipophilic albumin-binding tail by a solubility-promoting polar polymer chain. Administration of structurally optimized CpG-DNA/peptide amph-vaccines in mice resulted in marked increases in LN accumulation and decreased systemic dissemination relative to their parent compounds, leading to 30-fold increases in T-cell priming and enhanced anti-tumour efficacy while greatly reducing systemic toxicity. Amph-vaccines provide a simple, broadly applicable strategy to simultaneously increase the potency and safety of subunit vaccines.


Nature Medicine | 2016

Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses

Kelly D. Moynihan; Cary Francis Opel; Gregory L. Szeto; Alice Tzeng; Eric F. Zhu; Jesse M. Engreitz; Robert T. Williams; Kavya Rakhra; Michael H Zhang; Adrienne Rothschilds; Sudha Kumari; Ryan L. Kelly; Byron Hua Kwan; Wuhbet Abraham; Kevin Hu; Naveen K. Mehta; Monique J. Kauke; Heikyung Suh; Jennifer R. Cochran; Douglas A. Lauffenburger; K. Dane Wittrup; Darrell J. Irvine

Checkpoint blockade with antibodies specific for cytotoxic T lymphocyte–associated protein (CTLA)-4 or programmed cell death 1 (PDCD1; also known as PD-1) elicits durable tumor regression in metastatic cancer, but these dramatic responses are confined to a minority of patients. This suboptimal outcome is probably due in part to the complex network of immunosuppressive pathways present in advanced tumors, which are unlikely to be overcome by intervention at a single signaling checkpoint. Here we describe a combination immunotherapy that recruits a variety of innate and adaptive immune cells to eliminate large tumor burdens in syngeneic tumor models and a genetically engineered mouse model of melanoma; to our knowledge tumors of this size have not previously been curable by treatments relying on endogenous immunity. Maximal antitumor efficacy required four components: a tumor-antigen-targeting antibody, a recombinant interleukin-2 with an extended half-life, anti-PD-1 and a powerful T cell vaccine. Depletion experiments revealed that CD8+ T cells, cross-presenting dendritic cells and several other innate immune cell subsets were required for tumor regression. Effective treatment induced infiltration of immune cells and production of inflammatory cytokines in the tumor, enhanced antibody-mediated tumor antigen uptake and promoted antigen spreading. These results demonstrate the capacity of an elicited endogenous immune response to destroy large, established tumors and elucidate essential characteristics of combination immunotherapies that are capable of curing a majority of tumors in experimental settings typically viewed as intractable.


Cancer immunology research | 2015

Engineering New Approaches to Cancer Vaccines

Naveen K. Mehta; Kelly D. Moynihan; Darrell J. Irvine

Recently, a number of promising approaches have been developed using synthetic chemistry, materials science, and bioengineering-based strategies to address challenges in the design of more effective cancer vaccines. At the stage of initial priming, potency can be improved by maximizing vaccine delivery to lymph nodes. Because lymphatic uptake from peripheral tissues is strongly size dependent, antigens and adjuvants packaged into optimally sized nanoparticles access the lymph node with much greater efficiency than unformulated vaccines. Once primed, T cells must home to the tumor site. Because T cells acquire the necessary surface receptors in the local lymph node draining the tissue of interest, vaccines must be engineered that reach organs, such as the lung and gut, which are common sites of tumor lesions but inaccessible by traditional vaccination routes. Particulate vaccine carriers can improve antigen exposure in these organs, resulting in greater lymphocyte priming. Immunomodulatory agents can also be injected directly into the tumor site to stimulate a systemic response capable of clearing even distal lesions; materials have been designed that entrap or slowly release immunomodulators at the tumor site, reducing systemic exposure and improving therapeutic efficacy. Finally, lessons learned from the design of biomaterial-based scaffolds in regenerative medicine have led to the development of implantable vaccines that recruit and activate antigen-presenting cells to drive antitumor immunity. Overall, these engineering strategies represent an expanding toolkit to create safe and effective cancer vaccines. Cancer Immunol Res; 3(8); 836–43. ©2015 AACR.


Nature Communications | 2016

Targeting dendritic cells to accelerate T-cell activation overcomes a bottleneck in tuberculosis vaccine efficacy

Kristin L. Griffiths; Mushtaq Ahmed; Shibali Das; Radha Gopal; William Horne; Terry D. Connell; Kelly D. Moynihan; Jay K. Kolls; Darrell J. Irvine; Maxim N. Artyomov; Javier Rangel-Moreno; Shabaana A. Khader

The development of a tuberculosis (TB) vaccine that induces sterilizing immunity to Mycobacterium tuberculosis infection has been elusive. Absence of sterilizing immunity induced by TB vaccines may be due to delayed activation of mucosal dendritic cells (DCs), and subsequent delay in antigen presentation and activation of vaccine-induced CD4+ T-cell responses. Here we show that pulmonary delivery of activated M. tuberculosis antigen-primed DCs into vaccinated mice, at the time of M. tuberculosis exposure, can overcome the delay in accumulation of vaccine-induced CD4+ T-cell responses. In addition, activating endogenous host CD103+ DCs and the CD40–CD40L pathway can similarly induce rapid accumulation of vaccine-induced lung CD4+ T-cell responses and limit early M. tuberculosis growth. Thus, our study provides proof of concept that targeting mucosal DCs can accelerate vaccine-induced T-cell responses on M. tuberculosis infection, and provide insights to overcome bottlenecks in TB vaccine efficacy.


Nature Communications | 2017

High-throughput quantitation of inorganic nanoparticle biodistribution at the single-cell level using mass cytometry

Yu-Sang Sabrina Yang; Prabhani U. Atukorale; Kelly D. Moynihan; Ahmet Bekdemir; Kavya Rakhra; Li Tang; Francesco Stellacci; Darrell J. Irvine

Inorganic nanoparticles (NPs) are studied as drug carriers, radiosensitizers and imaging agents, and characterizing nanoparticle biodistribution is essential for evaluating their efficacy and safety. Tracking NPs at the single-cell level with current technologies is complicated by the lack of reliable methods to stably label particles over extended durations in vivo. Here we demonstrate that mass cytometry by time-of-flight provides a label-free approach for inorganic nanoparticle quantitation in cells. Furthermore, mass cytometry can enumerate AuNPs with a lower detection limit of ∼10 AuNPs (3 nm core size) in a single cell with tandem multiparameter cellular phenotyping. Using the cellular distribution insights, we selected an amphiphilic surface ligand-coated AuNP that targeted myeloid dendritic cells in lymph nodes as a peptide antigen carrier, substantially increasing the efficacy of a model vaccine in a B16-OVA melanoma mouse model. This technology provides a powerful new level of insight into nanoparticle fate in vivo.


Journal of Controlled Release | 2017

Radiation-enhanced delivery of systemically administered amphiphilic-CpG oligodeoxynucleotide

Oliver K. Appelbe; Kelly D. Moynihan; Amy Catherine Flor; Nick Rymut; Darrell J. Irvine; Stephen J. Kron

ABSTRACT Along with vaccines and checkpoint blockade, immune adjuvants may have an important role in tumor immunotherapy. Oligodeoxynucleotides containing unmethylated cytidyl guanosyl dinucleotide motifs (CpG ODN) are TLR9 ligands with attractive immunostimulatory properties, but intratumoral administration has been required to induce an effective anti‐tumor immune response. Following on recent studies with radiation‐targeted delivery of nanoparticles, we examined enhanced tumor‐specific delivery of amphiphile‐CpG, an albumin‐binding analog of CpG ODN, following systemic administration 3 days after tumor irradiation. The combination of radiation and CpG displayed superior tumor control over either treatment alone. Intravital imaging of fluorescently labeled amphiphilic‐CpG revealed increased accumulation in irradiated tumors along with decreased off‐target accumulation in visceral organs. Within 48 h after amphiphile‐CpG administration, immune activation could be detected by increased Granzyme B and Interferon gamma activity in the tumor as well as in circulating monocytes and activated CD8+ T cells. Using radiotherapy to enhance the targeting of CpG to tumors may help advance this once promising therapy to clinical relevance. Graphical abstract Figure. No caption available.


Journal of Immunology | 2018

Combined HDAC and BET Inhibition Enhances Melanoma Vaccine Immunogenicity and Efficacy

Alexander Badamchi-Zadeh; Kelly D. Moynihan; Rafael A. Larocca; Malika Aid; Nicholas M. Provine; M. Justin Iampietro; Ekaterina Kinnear; Pablo Penaloza-MacMaster; Peter Abbink; Eryn Blass; John S. Tregoning; Darrell J. Irvine; Dan H. Barouch

The combined inhibition of histone deacetylases (HDAC) and the proteins of the bromodomain and extraterminal (BET) family have recently shown therapeutic efficacy against melanoma, pancreatic ductal adenocarcinoma, testicular, and lymphoma cancers in murine studies. However, in such studies, the role of the immune system in therapeutically controlling these cancers has not been explored. We sought to investigate the effect of the HDAC inhibitor romidepsin (RMD) and the BET inhibitor IBET151, both singly and in combination, on vaccine-elicited immune responses. C57BL/6 mice were immunized with differing vaccine systems (adenoviral, protein) in prime-boost regimens under treatment with RMD, IBET151, or RMD+IBET151. The combined administration of RMD+IBET151 during vaccination resulted in a significant increase in the frequency and number of Ag-specific CD8+ T cells. RMD+IBET151 treatment significantly increased the frequency of vaccine-elicited IFN-γ+ splenic CD8+ T cells and conferred superior therapeutic and prophylactic protection against B16-OVA melanoma. RNA sequencing analyses revealed strong transcriptional similarity between RMD+IBET151 and untreated Ag-specific CD8+ T cells except in apoptosis and IL-6 signaling–related genes that were differentially expressed. Serum IL-6 was significantly increased in vivo following RMD+IBET151 treatment, with recombinant IL-6 administration replicating the effect of RMD+IBET151 treatment on vaccine-elicited CD8+ T cell responses. IL-6 sufficiency for protection was not assessed. Combined HDAC and BET inhibition resulted in greater vaccine-elicited CD8+ T cell responses and enhanced therapeutic and prophylactic protection against B16-OVA melanoma. Increased IL-6 production and the differential expression of pro- and anti-apoptotic genes following RMD+IBET151 treatment are likely contributors to the enhanced cancer vaccine responses.


Cancer immunology research | 2017

Abstract A42: Combination immunotherapy of an autochthonous murine lung cancer model expressing human CEA as a tumor-associated self-antigen

Kavya Rakhra; Eric F. Zhu; Wuhbet Abraham; Kelly D. Moynihan; Naveen K. Mehta; Karl Dane Wittrup; Darrell J. Irvine

While cancer immunotherapies like checkpoint inhibitors have resulted in unprecedented clinical success, they only benefit a subset of patients. To improve therapeutic outcomes for greater numbers of patients, one strategy is to rationally combine different immunotherapy modalities. We recently demonstrated that attaching albumin-binding lipophilic tails to peptide antigens or molecular adjuvants (creating amphiphile vaccines) results in enhanced T-cell responses. Additionally, we observed significant tumor regression upon combining tumor-antigen targeting antibodies with extended half-life IL-2 (exPK-IL-2) in mouse models of melanoma and prostate cancer. In the present work, we combined both approaches to treat a spontaneous model of lung adenocarcinoma expressing carcinoembryonic antigen (CEA), an oncofetal protein expressed in some human lung cancers. KrasLSL-G12D/+;;p53fl/fl mice were crossed with transgenic mice expressing human-CEA to generate a CEA-tolerant background. Lung tumors were induced by infection with a lentivirus expressing Cre recombinase and human CEA. Due to the extended latency of tumor initiation in this model, we also generated a CEA-expressing cell line from these mice to test the efficacy of different combination immunotherapy regimens. We discovered that weekly treatments combining a CEA-targeting amphiphile-vaccine, exPK-IL-2, and an anti-CEA antibody with checkpoint inhibitors (anti-PD-1 and -CTLA4) resulted in sustained tumor regression in 50% of mice bearing established tumors. We are currently testing this combination immunotherapy on autochthonous lung tumors. Our results suggest that breaking tolerance to a tumor-associated self-antigen (CEA) and combining immunotherapies to recruit both innate and adaptive immune effectors can have a potent therapeutic effect in intractable tumors like lung cancer. Citation Format: Kavya Rakhra, Eric F. Zhu, Wuhbet Abraham, Kelly D. Moynihan, Naveen Mehta, Karl D. Wittrup, Darrell J. Irvine. Combination immunotherapy of an autochthonous murine lung cancer model expressing human CEA as a tumor-associated self-antigen. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2016 Oct 20-23; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2017;5(3 Suppl):Abstract nr A42.


Cancer immunology research | 2017

Abstract A25: High-resolution visualization of immune response across the intact melanoma tumor

Sudha Kumari; Kelly D. Moynihan; Darrell J. Irvine

Tumor immunotherapy relies on improved anti-tumor activity of the immune cells. Efficacious immunotherapy often correlates with increased CD8+ T cell infiltration within the tumor, subsequently resulting into reduction in tumor growth and morphological remodeling. However, pan- melanoma visualization of immune cell infiltration and associated macro-scale alterations in tumor morphology and vasculature remains extremely challenging. This is largely owing to the dense pigmentation, thick extracellular matrix deposition and friable texture of the tumor, rendering it optically inaccessible using the routine histology and organ clarification techniques. Here, we describe a rapid and inexpensive optical clarification technique that enables examination of immune infiltration as well as vascular changes, both at tissue level as well as single cell-resolution within an intact tumor. The aforementioned technique, termed Tumor-optimized three-dimensional imaging of solvent cleared organs (T-DISCO), combines in vivo fluorescent labeling of the endogenous immune cells with organic solvents-based tissue clarification and eventual image acquisition using a standard confocal microscope. This technique when applied to the B16F10 melanoma model undergoing a multi-component immunotherapy, revealed changes within the tumor microenvironment, including vascular normalization and intra-tumoral trafficking of the T cells. Thus, T-DISCO represents a powerful tool to examine and investigate immune cell behavior deep within the intact tumor. Citation Format: Sudha Kumari, Kelly Moynihan, Darrell J. Irvine. High-resolution visualization of immune response across the intact melanoma tumor. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2016 Oct 20-23; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2017;5(3 Suppl):Abstract nr A25.


Cancer immunology research | 2017

Abstract A52: Eradication of large established tumors with combination immunotherapy engaging innate and adaptive immunity

Kelly D. Moynihan; Cary Francis Opel; Gregory Szeto; Alice Tzeng; Zhu Eric; Jesse M. Engreitz; Williams Robert; Kavya Rakhra; Michael Zhang; Adrienne Rothschilds; Sudha Kumari; Ryan L. Kelly; Byron Hua Kwan; Wuhbet Abraham; Kevin Hu; Naveen K. Mehta; Monique J. Kauke; Heikyung Suh; Douglas A. Lauffenburger; K. Dane Wittrup; Darrell J. Irvine

Checkpoint blockade against CTLA-4 or PD-1 has demonstrated that an endogenous immune response can be stimulated to elicit durable regressions in advanced cancer, but these dramatic responses are currently confined to a minority of patients. This outcome is probably due in part to the complex network of immunosuppressive pathways present in advanced tumors, which are unlikely to be overcome by intervention at a single signaling checkpoint, requiring a counter-directed network of pro-immunity signals. Here we demonstrate a combination immunotherapy that recruits a diverse set of innate and adaptive immune effectors, enabling robust elimination of tumor burdens that to our knowledge have not previously been curable by treatments relying on endogenous immunity. Maximal anti-tumor efficacy required four components: a tumor antigen targeting antibody, an extended half-life IL-2, anti-PD-1, and a powerful T-cell vaccine. This combination elicited durable cures in a majority of animals, formed immunological memory in multiple transplanted tumor models, and induced sustained tumor regression in an autochthonous BRrafV600E/Pten-/- melanoma model. Multiple innate immune cell subsets, CD8+ T-cells, and cross-presenting dendritic cells were critical to successful therapy. Treatment induced high levels of intratumoral inflammatory cytokines and immune cell infiltration, enhanced antibody-mediated tumor antigen uptake, and promoted antigen spreading. These results demonstrate the capacity of an elicited endogenous immune response to destroy large, established tumors and elucidate essential characteristics of combination immunotherapies capable of curing a majority of tumors in experimental settings typically viewed as intractable. Citation Format: Kelly Dare Moynihan, Cary Opel, Gregory Szeto, Alice Tzeng, Zhu Eric, Jesse Engreitz, Williams Robert, Kavya Rakhra, Michael Zhang, Adrienne Rothschilds, Sudha Kumari, Ryan L. Kelly, Byron Kwan, Wuhbet Abraham, Kevin Hu, Naveen Mehta, Monique Kauke, Heikyung Suh, Douglas A. Lauffenburger, K. Dane Wittrup, Darrell J. Irvine. Eradication of large established tumors with combination immunotherapy engaging innate and adaptive immunity. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2016 Oct 20-23; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2017;5(3 Suppl):Abstract nr A52.

Collaboration


Dive into the Kelly D. Moynihan's collaboration.

Top Co-Authors

Avatar

Darrell J. Irvine

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Cary Francis Opel

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Naveen K. Mehta

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Eric F. Zhu

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

K. Dane Wittrup

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wuhbet Abraham

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Gregory L. Szeto

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Monique J. Kauke

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Alice Tzeng

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge