Network


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

Hotspot


Dive into the research topics where Aaron T. Mayer is active.

Publication


Featured researches published by Aaron T. Mayer.


ACS Nano | 2012

Enhanced Delivery of Chemotherapy to Tumors Using a Multicomponent Nanochain with Radio-Frequency-Tunable Drug Release

Lisa Bauer; Randall Toy; Emily Tran; Jenna Pansky; Elizabeth Doolittle; Erik Schmidt; Elliott Hayden; Aaron T. Mayer; Ruth A. Keri; Mark A. Griswold; Efstathios Karathanasis

While nanoparticles maximize the amount of chemotherapeutic drug in tumors relative to normal tissues, nanoparticle-based drugs are not accessible to the majority of cancer cells because nanoparticles display patchy, near-perivascular accumulation in tumors. To overcome the limitations of current drugs in their molecular or nanoparticle form, we developed a nanoparticle based on multicomponent nanochains to deliver drug to the majority of cancer cells throughout a tumor while reducing off-target delivery. The nanoparticle is composed of three magnetic nanospheres and one doxorubicin-loaded liposome assembled in a 100 nm long chain. These nanoparticles display prolonged blood circulation and significant intratumoral deposition in tumor models in rodents. Furthermore, the magnetic particles of the chains serve as a mechanical transducer to transfer radio frequency energy to the drug-loaded liposome. The defects on the liposomal walls trigger the release of free drug capable of spreading throughout the entire tumor, which results in a widespread anticancer effect.


Bioconjugate Chemistry | 2015

Novel Radiotracer for ImmunoPET Imaging of PD-1 Checkpoint Expression on Tumor Infiltrating Lymphocytes.

Arutselvan Natarajan; Aaron T. Mayer; Lingyun Xu; Robert Reeves; Jacob Gano; Sanjiv S. Gambhir

Immune checkpoint signaling through the programmed death 1 (PD-1) axis to its ligand (PD-L1) significantly dampens anti-tumor immune responses. Cancer patients treated with checkpoint inhibitors that block this suppressive signaling have exhibited objective response rates of 20-40% for advanced solid tumors, lymphomas, and malignant melanomas. This represents a tremendous advance in cancer treatment. Unfortunately, all patients do not respond to immune checkpoint blockade. Recent findings suggest that patients with tumor infiltrating lymphocytes (TILs) expressing PD-1 may be most likely to respond to αPD-1/PD-L1 checkpoint inhibitors. There is a compelling need for diagnostic and prognostic imaging tools to assess the PD-1 status of TILs in vivo. Here we have developed a novel immunoPET tracer to image PD-1 expressing TILs in a transgenic mouse model bearing melanoma. A (64)Cu labeled anti-mouse antibody (IgG) PD-1 immuno positron emission tomography (PET) tracer was developed to detect PD-1 expressing murine TILs. Quality control of the tracer showed >95% purity by HPLC and >70% immunoreactivity in an in vitro cell binding assay. ImmunoPET scans were performed over 1-48 h on Foxp3+.LuciDTR4 mice bearing B16-F10 melanoma tumors. Mice receiving anti-PD-1 tracer (200 ± 10 μCi/10-12 μg/200 μL) revealed high tracer uptake in lymphoid organs and tumors. BLI images of FoxP3(+) CD4(+) Tregs known to express PD-1 confirmed lymphocyte infiltration of tumors at the time of PET imaging. Biodistribution measurements performed at 48 h revealed a high (11×) tumor to muscle uptake ratio of the PET tracer (p < 0.05). PD-1 tumors exhibited 7.4 ± 0.7%ID/g tracer uptake and showed a 2× fold signal decrease when binding was blocked by unlabeled antibody. To the best of our knowledge this data is the first report to image PD-1 expression in living subjects with PET. This radiotracer has the potential to assess the prognostic value of PD-1 in preclinical models of immunotherapy and may ultimately aid in predicting response to therapies targeting immune checkpoints.


Journal of Controlled Release | 2014

Treatment of cancer micrometastasis using a multicomponent chain-like nanoparticle

Randall Toy; Aaron Abramowski; Pete Vicente; Samantha Tucci; Lisa Bauer; Aaron T. Mayer; Morgan Tam; Elizabeth Doolittle; Jenna Pansky; Emily Tran; Dishen Lin; William P. Schiemann; Ketan B. Ghaghada; Mark A. Griswold; Efstathios Karathanasis

While potent cytotoxic agents are available to oncologists, the clinical utility of these agents is limited due to their non-specific distribution in the body and toxicity to normal tissues leading to use of suboptimal doses for eradication of metastatic disease. Furthermore, treatment of micrometastases is impeded by several biobarriers, including their small size and high dispersion to organs, making them nearly inaccessible to drugs. To circumvent these limitations in treating metastatic disease, we developed a multicomponent, flexible chain-like nanoparticle (termed nanochain) that possesses a unique ability to gain access to and be deposited at micrometastatic sites. Moreover, coupling nanochain particles to radiofrequency (RF)-triggered cargo delivery facilitated widespread delivery of drug into hard-to-reach cancer cells. Collectively, these features synergistically facilitate effective treatment and ultimately eradication of micrometastatic disease using a low dose of a cytotoxic drug.


Science Translational Medicine | 2018

Eradication of spontaneous malignancy by local immunotherapy

Idit Sagiv-Barfi; Debra K. Czerwinski; Shoshana Levy; Israt S. Alam; Aaron T. Mayer; Sanjiv S. Gambhir; Ronald Levy

In situ vaccination with low doses of TLR ligands and anti-OX40 antibodies can cure widespread cancers in preclinical models. Deliver locally, act globally Mobilizing endogenous T cells to fight tumors is the goal of many immunotherapies. Sagiv-Barfi et al. investigated a combination therapy in multiple types of mouse cancer models that could provide sustainable antitumor immunity. Specifically, they combined intratumoral delivery of a TLR9 ligand with OX40 activation to ramp up T cell responses. This dual immunotherapy led to shrinkage of distant tumors and long-term survival of the animals, even in a stringent spontaneous tumor model. Both of these stimuli are in clinical trials as single agents and could likely be combined at great benefit for cancer patients. It has recently become apparent that the immune system can cure cancer. In some of these strategies, the antigen targets are preidentified and therapies are custom-made against these targets. In others, antibodies are used to remove the brakes of the immune system, allowing preexisting T cells to attack cancer cells. We have used another noncustomized approach called in situ vaccination. Immunoenhancing agents are injected locally into one site of tumor, thereby triggering a T cell immune response locally that then attacks cancer throughout the body. We have used a screening strategy in which the same syngeneic tumor is implanted at two separate sites in the body. One tumor is then injected with the test agents, and the resulting immune response is detected by the regression of the distant, untreated tumor. Using this assay, the combination of unmethylated CG–enriched oligodeoxynucleotide (CpG)—a Toll-like receptor 9 (TLR9) ligand—and anti-OX40 antibody provided the most impressive results. TLRs are components of the innate immune system that recognize molecular patterns on pathogens. Low doses of CpG injected into a tumor induce the expression of OX40 on CD4+ T cells in the microenvironment in mouse or human tumors. An agonistic anti-OX40 antibody can then trigger a T cell immune response, which is specific to the antigens of the injected tumor. Remarkably, this combination of a TLR ligand and an anti-OX40 antibody can cure multiple types of cancer and prevent spontaneous genetically driven cancers.


The Journal of Nuclear Medicine | 2017

Practical ImmunoPET radiotracer design considerations for human immune checkpoint imaging

Aaron T. Mayer; Arutselvan Natarajan; Sydney R. Gordon; Roy L. Maute; Melissa N. McCracken; Aaron M. Ring; Irving L. Weissman; Sanjiv S. Gambhir

Immune checkpoint blockade has emerged as a promising cancer treatment paradigm. Unfortunately, there are still a large number of patients and malignancies that do not respond to therapy. A major barrier to validating biomarkers for the prediction and monitoring of responders to clinical checkpoint blockade has been the lack of imaging tools to accurately assess dynamic immune checkpoint expression. Here, we sought to optimize noninvasive immuno-PET imaging of human programmed death-ligand 1 (PD-L1) expression, in a preclinical model, using a small high-affinity engineered protein scaffold (HAC-PD1). Six HAC-PD1 radiotracer variants were developed and used in preclinical imaging and biodistribution studies to assess their ability to detect human PD-L1 expression in vivo. Radiotracer design modifications included chelate, glycosylation, and radiometal. HACA-PD1 was adopted as the naming convention for aglycosylated tracer variants. NOD scid γ-(NSG) mice were inoculated with subcutaneous tumors engineered to either be constitutively positive (CT26 hPD-L1) or be negative (ΔmPD-L1 CT26) for human PD-L1 expression. When the tumors had grown to an average size of 1 cm in diameter, mice were injected with 0.75–2.25 MBq (∼10 μg) of an engineered radiotracer variant and imaged. At 1 h after injection, organs were harvested for biodistribution. Of the practical immuno-PET tracer modifications considered, glycosylation was the most prominent design factor affecting tracer uptake, specificity, and clearance. In imaging studies, aglycosylated 64Cu-NOTA-HACA-PD1 most accurately visualized human PD-L1 expression in vivo. We reasoned that because of the scaffold’s small size (14 kDa), its pharmacokinetics may be suitable for labeling with the short-lived and widely clinically available radiometal 68Ga. At 1 h after injection, 68Ga-NOTA-HACA-PD1 and 68Ga-DOTA-HACA-PD1 exhibited promising target-to-background ratios in ex vivo biodistribution studies (12.3 and 15.2 tumor-to-muscle ratios, respectively). Notably, all HAC-PD1 radiotracer variants enabled much earlier detection of human PD-L1 expression (1 h after injection) than previously reported radiolabeled antibodies (>24 h after injection). This work provides a template for assessing immuno-PET tracer design parameters and supports the translation of small engineered protein radiotracers for imaging human immune checkpoints.


Biomaterials | 2017

Vaccine nanocarriers: Coupling intracellular pathways and cellular biodistribution to control CD4 vs CD8 T cell responses

Marcela Rincon-Restrepo; Aaron T. Mayer; Sylvie Hauert; Daniel K. Bonner; Edward A. Phelps; Jeffrey A. Hubbell; Melody A. Swartz; Sachiko Hirosue

Nanoparticle delivery systems are known to enhance the immune response to soluble antigens (Ags) and are thus a promising tool for the development of new vaccines. Our laboratory has engineered two different nanoparticulate systems in which Ag is either encapsulated within the core of polymersomes (PSs) or decorated onto the surface of nanoparticles (NPs). Previous studies showed that PSs are better at enhancing CD4 T cells and antibody titers, while NPs preferentially augment cytotoxic CD8 T cells. Herein, we demonstrate that the differential activation of T cell immunity reflects differences in the modes of intracellular trafficking and distinct biodistribution of the Ag in lymphoid organs, which are both driven by the properties of each nanocarrier. Furthermore, we found that Ags within PSs promoted better CD4 T cell activation and induced a higher frequency of CD4 T follicular helper (Tfh) cells. These differences correlated with changes in the frequency of germinal center B cells and plasma cell formation, which reflects the previously observed antibody titers. Our results show that PSs are a promising vector for the delivery of Ags for B cell vaccine development. This study demonstrates that nanocarrier design has a large impact on the quality of the induced adaptive immune response.


Journal of Clinical Investigation | 2018

Imaging activated T cells predicts response to cancer vaccines

Israt S. Alam; Aaron T. Mayer; Idit Sagiv-Barfi; Kezheng Wang; Ophir Vermesh; Debra K. Czerwinski; Emily M. Johnson; Michelle L. James; Ronald Levy; Sanjiv S. Gambhir

In situ cancer vaccines are under active clinical investigation, given their reported ability to eradicate both local and disseminated malignancies. Intratumoral vaccine administration is thought to activate a T cell–mediated immune response, which begins in the treated tumor and cascades systemically. In this study, we describe a PET tracer (64Cu-DOTA-AbOX40) that enabled noninvasive and longitudinal imaging of OX40, a cell-surface marker of T cell activation. We report the spatiotemporal dynamics of T cell activation following in situ vaccination with CpG oligodeoxynucleotide in a dual tumor–bearing mouse model. We demonstrate that OX40 imaging was able to predict tumor responses on day 9 after treatment on the basis of tumor tracer uptake on day 2, with greater accuracy than both anatomical and blood-based measurements. These studies provide key insights into global T cell activation following local CpG treatment and indicate that 64Cu-DOTA-AbOX40 is a promising candidate for monitoring clinical cancer immunotherapy strategies.


The Journal of Nuclear Medicine | 2017

Imaging B cells in a mouse model of multiple sclerosis using 64Cu-Rituximab-PET

Michelle L. James; Aileen Hoehne; Aaron T. Mayer; Kendra Lechtenberg; Monica Moreno; Gayatri Gowrishankar; Ohad Ilovich; Arutselvan Natarajan; Emily M. Johnson; Joujou Nguyen; Lisa Quach; May Han; Marion S. Buckwalter; Sudeep Chandra; Sanjiv S. Gambhir

B lymphocytes are a key pathologic feature of multiple sclerosis (MS) and are becoming an important therapeutic target for this condition. Currently, there is no approved technique to noninvasively visualize B cells in the central nervous system (CNS) to monitor MS disease progression and response to therapies. Here, we evaluated 64Cu-rituximab, a radiolabeled antibody specifically targeting the human B cell marker CD20, for its ability to image B cells in a mouse model of MS using PET. Methods: To model CNS infiltration by B cells, experimental autoimmune encephalomyelitis (EAE) was induced in transgenic mice that express human CD20 on B cells. EAE mice were given subcutaneous injections of myelin oligodendrocyte glycoprotein fragment1–125 emulsified in complete Freund adjuvant. Control mice received complete Freund adjuvant alone. PET imaging of EAE and control mice was performed 1, 4, and 19 h after 64Cu-rituximab administration. Mice were perfused and sacrificed after the final PET scan, and radioactivity in dissected tissues was measured with a γ-counter. CNS tissues from these mice were immunostained to quantify B cells or were further analyzed via digital autoradiography. Results: Lumbar spinal cord PET signal was significantly higher in EAE mice than in controls at all evaluated time points (e.g., 1 h after injection: 5.44 ± 0.37 vs. 3.33 ± 0.20 percentage injected dose [%ID]/g, P < 0.05). 64Cu-rituximab PET signal in brain regions ranged between 1.74 ± 0.11 and 2.93 ± 0.15 %ID/g for EAE mice, compared with 1.25 ± 0.08 and 2.24 ± 0.11 %ID/g for controls (P < 0.05 for all regions except striatum and thalamus at 1 h after injection). Similarly, ex vivo biodistribution results revealed notably higher 64Cu-rituximab uptake in the brain and spinal cord of huCD20tg EAE, and B220 immunostaining verified that increased 64Cu-rituximab uptake in CNS tissues corresponded with elevated B cells. Conclusion: B cells can be detected in the CNS of EAE mice using 64Cu-rituximab PET. Results from these studies warrant further investigation of 64Cu-rituximab in EAE models and consideration of use in MS patients to evaluate its potential for detecting and monitoring B cells in the progression and treatment of this disease. These results represent an initial step toward generating a platform to evaluate B cell–targeted therapeutics en route to the clinic.


The Journal of Nuclear Medicine | 2018

The Immunoimaging Toolbox

Aaron T. Mayer; Sanjiv S. Gambhir

The recent clinical success of cancer immunotherapy has renewed interest in the development of tools to image the immune system. In general, immunotherapies attempt to enable the body’s own immune cells to seek out and destroy malignant disease. Molecular imaging of the cells and molecules that regulate immunity could provide unique insight into the mechanisms of action, and failure, of immunotherapies. In this article, we will provide a comprehensive overview of the current state-of-the-art immunoimaging toolbox with a focus on imaging strategies and their applications toward immunotherapy.


The Journal of Nuclear Medicine | 2018

Response: Optimizing Strategies for Immune Checkpoint Imaging with Immuno-PET in Preclinical Study

Aaron T. Mayer; Sanjiv S. Gambhir

TO THE EDITOR: Recently, we have read with interest the paper by Mayer et al. published in The Journal of Nuclear Medicine (1). The authors assessed the effects of 6 immuno-PET radiotracers on human programmed cell death ligand 1 (PD-L1) immune checkpoint imaging and discussed important design considerations that may affect biodistribution of radiotracers. Those radiotracers were specifically against human PD-L1 but did not cross-react with murine PD-L1. As we inferred, clinical immuno-PET tracers can bind not only to PD-L1 expressed by tumors, but also to PD-L1 expressed by normal cells. It is known that PD-L1 is expressed wildly on T cells, B cells, monocytes, and endothelial cells in both humans and mice (2). Therefore, radiotracer can be taken up by PD-L1–positive cells in organs, including lymphoid organs, lung, and liver, resulting in unexpected background signal and confounding determination of PD-L1 level in tumors. To optimize the immuno-PET imaging effect, especially in terms of background signal, we suggest using antimurine radiotracers and murine tumor cell lines for syngeneic tumor engraftments, because these will better fit the putative clinical status, rather than performing in vivo study in human tumor xenografts. We are also concerned about the inherent characteristic of PDL1 after immuno-PET imaging. It is known that radiotracers can induce cell internalization; thus, the targeted receptor could be involved and relocated from membrane to cytoplasm (3,4). During immuno-PET imaging, PD-L1 is internalized but the metabolic mechanism is unclear, partially including degradation and repopulation back to the tumor cell surface. Moreover, whether the affinity between PD-L1 and tracer would change after being detected by immuno-PET for the first evaluation and monitoring assessment during treatment remains unknown. To identify the potential affinity change, we suggest conducting another immunoPET scan or surface plasmon resonance after the radiotracer is entirely eliminated. Additionally, it is possible that the expression level of PD-L1 may not be a favorable biomarker for predicting anti–PD-L1 response. By analyzing the outcome of patients with different PD-L1 level, Robert et al. reported no difference in overall survival between the high-expression PD-L1 group and low or negative group after immunotherapy with anti–PD-L1 antibody (5). Therefore, high uptake of radiotracer at a tumor site may not predict a good response whereas low uptake may not indicate a poor response. To better predict anti–PD-L1 response, a combination of PD-L1 status and other cancer genetic biomarkers should be further considered (6). Generally, immuno-PET imaging represents a novel imaging procedure and is helpful for selecting optimal patients and monitoring the expression status of specific molecules during anti–PD-L1 treatment. It could become the go-to complement to immunotherapy in the near future. REFERENCES

Collaboration


Dive into the Aaron T. Mayer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Efstathios Karathanasis

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elizabeth Doolittle

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Emily Tran

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Jenna Pansky

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Randall Toy

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Ruth A. Keri

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Elliott Hayden

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Lisa Bauer

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge