Network


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

Hotspot


Dive into the research topics where Cary Francis Opel is active.

Publication


Featured researches published by Cary Francis Opel.


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.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Antigen specificity can be irrelevant to immunocytokine efficacy and biodistribution

Alice Tzeng; Byron Hua Kwan; Cary Francis Opel; Tejas Navaratna; K. Dane Wittrup

Significance Cytokines (potent immunostimulatory proteins) exert powerful antitumor effects but often cause severe whole-body inflammation when used as cancer therapies. Contrary to the current paradigm that fusion to antitumor antibodies can constrain cytokine activity to tumors, we have found that, for some immunocytokines incorporating the cytokine IL-2, the cytokine moiety overrides antibody-mediated targeting, localizing the fusion protein to IL-2 receptor-expressing cells rather than tumor cells. Although the IL-2 immunocytokines did not selectively home to tumors, they persisted longer in circulation than free IL-2, such that a nontoxic immunocytokine dose could synergize with tumor-specific antibody to cure mice with aggressive solid tumors. Cytokine therapy can activate potent, sustained antitumor responses, but collateral toxicity often limits dosages. Although antibody–cytokine fusions (immunocytokines) have been designed with the intent to localize cytokine activity, systemic dose-limiting side effects are not fully ameliorated by attempted tumor targeting. Using the s.c. B16F10 melanoma model, we found that a nontoxic dose of IL-2 immunocytokine synergized with tumor-specific antibody to significantly enhance therapeutic outcomes compared with immunocytokine monotherapy, concomitant with increased tumor saturation and intratumoral cytokine responses. Examination of cell subset biodistribution showed that the immunocytokine associated mainly with IL-2R–expressing innate immune cells, with more bound immunocytokine present in systemic organs than the tumor microenvironment. More surprisingly, immunocytokine antigen specificity and Fcγ receptor interactions did not seem necessary for therapeutic efficacy or biodistribution patterns because immunocytokines with irrelevant specificity and/or inactive mutant Fc domains behaved similarly to tumor-specific immunocytokine. IL-2–IL-2R interactions, rather than antibody–antigen targeting, dictated immunocytokine localization; however, the lack of tumor targeting did not preclude successful antibody combination therapy. Mathematical modeling revealed immunocytokine size as another driver of antigen targeting efficiency. This work presents a safe, straightforward strategy for augmenting immunocytokine efficacy by supplementary antibody dosing and explores underappreciated factors that can subvert efforts to purposefully alter cytokine biodistribution.


Journal of Clinical Investigation | 2017

Biopolymers codelivering engineered T cells and STING agonists can eliminate heterogeneous tumors

Tyrel T. Smith; Howell F. Moffett; Sirkka B. Stephan; Cary Francis Opel; Amy G. Dumigan; Xiuyun Jiang; Venu G. Pillarisetty; Smitha P. S. Pillai; K. Dane Wittrup; Matthias T. Stephan

Therapies using T cells that are programmed to express chimeric antigen receptors (CAR T cells) consistently produce positive results in patients with hematologic malignancies. However, CAR T cell treatments are less effective in solid tumors for several reasons. First, lymphocytes do not efficiently target CAR T cells; second, solid tumors create an immunosuppressive microenvironment that inactivates T cell responses; and third, solid cancers are typified by phenotypic diversity and thus include cells that do not express proteins targeted by the engineered receptors, enabling the formation of escape variants that elude CAR T cell targeting. Here, we have tested implantable biopolymer devices that deliver CAR T cells directly to the surfaces of solid tumors, thereby exposing them to high concentrations of immune cells for a substantial time period. In immunocompetent orthotopic mouse models of pancreatic cancer and melanoma, we found that CAR T cells can migrate from biopolymer scaffolds and eradicate tumors more effectively than does systemic delivery of the same cells. We have also demonstrated that codelivery of stimulator of IFN genes (STING) agonists stimulates immune responses to eliminate tumor cells that are not recognized by the adoptively transferred lymphocytes. Thus, these devices may improve the effectiveness of CAR T cell therapy in solid tumors and help protect against the emergence of escape variants.


ACS Nano | 2013

Emergent Properties of Nanosensor Arrays: Applications for Monitoring IgG Affinity Distributions, Weakly Affined Hypermannosylation, and Colony Selection for Biomanufacturing

Nigel F. Reuel; Brittany R. Grassbaugh; Sebastian Kruss; J. Zachary Mundy; Cary Francis Opel; Adebola O. Ogunniyi; Kamal Egodage; Ramon Wahl; Bernhard Helk; Jingqing Zhang; Z. Ilke Kalcioglu; Kevin Tvrdy; Darin O. Bellisario; Bin Mu; Steven Blake; Krystyn J. Van Vliet; J. Christopher Love; Karl Dane Wittrup; Michael S. Strano

It is widely recognized that an array of addressable sensors can be multiplexed for the label-free detection of a library of analytes. However, such arrays have useful properties that emerge from the ensemble, even when monofunctionalized. As examples, we show that an array of nanosensors can estimate the mean and variance of the observed dissociation constant (KD), using three different examples of binding IgG with Protein A as the recognition site, including polyclonal human IgG (KD μ = 19 μM, σ(2) = 1000 mM(2)), murine IgG (KD μ = 4.3 nM, σ(2) = 3 μM(2)), and human IgG from CHO cells (KD μ = 2.5 nM, σ(2) = 0.01 μM(2)). Second, we show that an array of nanosensors can uniquely monitor weakly affined analyte interactions via the increased number of observed interactions. One application involves monitoring the metabolically induced hypermannosylation of human IgG from CHO using PSA-lectin conjugated sensor arrays where temporal glycosylation patterns are measured and compared. Finally, the array of sensors can also spatially map the local production of an analyte from cellular biosynthesis. As an example, we rank productivity of IgG-producing HEK colonies cultured directly on the array of nanosensors itself.


Scientific Reports | 2015

A graphene-based physiometer array for the analysis of single biological cells.

Geraldine L C Paulus; Justin T. Nelson; Katherine Y. Lee; Qing Hua Wang; Nigel F. Reuel; Brittany R. Grassbaugh; Sebastian Kruss; Markita P. Landry; Jeon Woong Kang; Emma Vander Ende; Jingqing Zhang; Bin Mu; Ramachandra R. Dasari; Cary Francis Opel; K. Dane Wittrup; Michael S. Strano

A significant advantage of a graphene biosensor is that it inherently represents a continuum of independent and aligned sensor-units. We demonstrate a nanoscale version of a micro-physiometer – a device that measures cellular metabolic activity from the local acidification rate. Graphene functions as a matrix of independent pH sensors enabling subcellular detection of proton excretion. Raman spectroscopy shows that aqueous protons p-dope graphene – in agreement with established doping trajectories, and that graphene displays two distinct pKa values (2.9 and 14.2), corresponding to dopants physi- and chemisorbing to graphene respectively. The graphene physiometer allows micron spatial resolution and can differentiate immunoglobulin (IgG)-producing human embryonic kidney (HEK) cells from non-IgG-producing control cells. Population-based analyses allow mapping of phenotypic diversity, variances in metabolic activity, and cellular adhesion. Finally we show this platform can be extended to the detection of other analytes, e.g. dopamine. This work motivates the application of graphene as a unique biosensor for (sub)cellular interrogation.


Journal for ImmunoTherapy of Cancer | 2013

Combined treatment using adoptive cell therapy, extended pharmacokinetic IL-2, and tumor-specific antibodies leads to cures of established B16F10 tumors and extended in vivo T cell survival

Cary Francis Opel; K. Dane Wittrup

IL2 is frequently given alongside adoptive cell therapy in order to enhance T cell function and survival, however, the protein has a poor pharmacokinetic profile and severe negative side effects. Fc-IL2 is a monovalent Fc fusion with IL2. The addition of the Fc domain to the cytokine significantly improved the persistence of the molecule in vivo, leading to enhanced activation of many types of immune cells, including T cells. The treatment described here is the combination of ACT, Fc-IL2, and/or antibodies targeting tumor associated antigens. The application of these three agents in preclinical experiments showed increased persistence of transferred cells, extended survival of treated animals, regression of tumor size (see Figure ​Figure1),1), and in some cases complete cures of established tumors with immune memory, as demonstrated by the rejection of secondary tumor challenge. Although the combination of all three agents was the most effective, the survival benefit of ACT and Fc-IL2 without antibody was significant enough to justify its use regardless of the availability of appropriate antibodies. The experimental model consisted of C57BL/6 mice subcutaneously injected with the B16F10 cell line. Following tumor establishment for 6 days, patient mice were lymphodepleted by total body irradiation and treated with a single injection of pmel-1 T cells, which express a T cell receptor (TCR) specific for B16F10 cells. In addition, over the next 24 days, 5 injections of Fc-IL2 and an antibody specific for B16F10 cells, TA99, were performed. Luciferase expressing pmel-1 cells were also injected in order to track the duration and intensity of the T cell response to the B16F10 tumors. These results show that effective adjuvants, such as Fc-IL2, have the potential to improve the clinical outcomes of adoptive cell therapy. Figure 1 Growth curves of B16F10 tumors treated with combination immunotherapy.


Journal of Molecular Biology | 2018

Artificial Anti-Tumor Opsonizing Proteins with Fibronectin Scaffolds Engineered for Specificity to Each of the Murine FcγR Types

Tiffany F. Chen; Kevin Li; Eric F. Zhu; Cary Francis Opel; Monique J. Kauke; Heeyoon Kim; Eta Atolia; K. Dane Wittrup

We have engineered a panel of novel Fn3 scaffold-based proteins that bind with high specificity and affinity to each of the individual mouse Fcγ receptors (mFcγR). These binders were expressed as fusions to anti-tumor antigen single-chain antibodies and mouse serum albumin, creating opsonizing agents that invoke only a single mFcγR response rather than the broader activity of natural Fc isotypes, as well as all previously reported Fc mutants. This panel isolated the capability of each of the four mFcγRs to contribute to macrophage phagocytosis of opsonized tumor cells and in vivo tumor growth control with these monospecific opsonizing fusion proteins. All activating receptors (mFcγRI, mFcγRIII, and mFcγRIV) were capable of driving specific tumor cell phagocytosis to an equivalent extent, while mFcγRII, the inhibitory receptor, did not drive phagocytosis. Monospecific opsonizing fusion proteins that bound mFcγRI alone controlled tumor growth to an extent similar to the most active IgG2a murine isotype. As expected, binding to the inhibitory mFcγRII did not delay tumor growth, but unexpectedly, mFcγRIII also failed to control tumor growth. mFcγRIV exhibited detectable but lesser tumor-growth control leading to less overall survival compared to mFcγRI. Interestingly, in vivo macrophage depletion demonstrates their importance in tumor control with mFcγRIV engagement, but not with mFcγRI. This panel of monospecific mFcγR-binding proteins provides a toolkit for isolating the functional effects of each mFcγR in the context of an intact immune system.


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.


Cancer immunology research | 2016

Abstract B120: Characterization and combination immunotherapy treatment of an inducible autochthonous murine lung cancer model expressing human carcinoembryonic antigen (CEA) as a tumor-associated self-antigen

Eric F. Zhu; Kavya Rakhra; Naveen K. Mehta; Kelly D. Moynihan; Cary Francis Opel; Darrell J. Irvine; K. Dane Wittrup

Previous work from our lab has demonstrated that a combination of anti-tumor antibody and an IL-2 fusion protein that exhibits extended serum half-life elicits an immune response that can effectively control a wide variety of tumor models. We have since then combined this therapeutic regimen with a vaccine exhibiting efficient lymph node trafficking that can generate an impressive population of tumor-antigen specific CD8+ T-cells but by itself does not provide good anti-tumor efficacy. The efficacy of this combination immunotherapy is further boosted by immune checkpoint blockade, leading to a robust four-component therapy: 1) anti-tumor antigen antibody; 2) IL-2 fusion protein; 3) anti-tumor antigen vaccine; 4) anti-PD-1 antibody. Although this four-pronged approach is demonstrably effective in the syngeneic subcutaneous melanoma model B16F10, we wish to test its efficacy in a more physiological model. To this end, we have turned to a model developed by the Jacks Lab, known as the KP model: an inducible lung tumor model where lentivirus-driven integration and expression of Cre is able to activate oncogenic Kras and completely remove p53 function. Because our therapeutic regimen requires a targetable tumor-associated antigen with respect to both the antibody and vaccine, we chose to induce expression of human carcinoembryonic antigen (CEA) in these tumors, as CEA has a well-studied structure and biology, and frequently expresses aberrantly in many forms of human adenocarcinomas. Additionally, our lab has previously engineered an antibody targeting CEA possessing picomolar affinity. Finally, to remove any endogenous immunological response against human CEA as a foreign antigen in our mouse system, we have crossed the KP model with a mouse model transgenic for human CEA, which in the literature has been described to have identical spatiotemporal expression of CEA as found in humans and should allow for central tolerance of this antigen. In the course of this work, we have successfully introduced human CEA into our lentivirus constructs and shown tumorigenesis by these constructs in the KP model coincides with expression of tumor-associated CEA, as detected by qPCR. On the therapeutic side, we have tailored the vaccine to successfully drive an anti-CEA CD8+ T-cell response. Performing preliminary therapeutic experiments in a transplant model of the KP tumor with our four-component therapy, we saw tumor control compared to untreated tumors. Upon interrogating the CD8+ T-cell response against CEA, we found 1-15% of CD8+ T-cells in the blood respond to CEA stimulation by intracellular cytokine staining. With regards to the lung tumor model, in the course of establishing the system we have also observed that the growth kinetics of tumors expressing CEA lags behind those tumors without CEA, even in the transgenic background. Preliminary immunophenotyping work by flow cytometry suggests that tumors with CEA seem to have a reduced myeloid-derived suppressor cell (MDSC) population and a higher CD8a+ dendritic cell (DC) population compared to tumors without CEA, suggesting that the former may have a less immunosuppressive tumor microenvironment that is better able to prime an anti-tumor CD8+ T-cell response. We will be planning to conduct therapeutic trials in the more physiological lung tumor KP model in the near future, as well as investigate the differences in the immune response with tumors expressing or lacking CEA. Citation Format: Eric F. Zhu, Kavya Rakhra, Naveen Mehta, Kelly D. Moynihan, Cary F. Opel, Darrell J. Irvine, K. Dane Wittrup. Characterization and combination immunotherapy treatment of an inducible autochthonous murine lung cancer model expressing human carcinoembryonic antigen (CEA) as a tumor-associated self-antigen. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr B120.


Cancer Cell | 2015

Synergistic Innate and Adaptive Immune Response to Combination Immunotherapy with Anti-Tumor Antigen Antibodies and Extended Serum Half-Life IL-2

Eric F. Zhu; Shuning A. Gai; Cary Francis Opel; Byron Hua Kwan; Rishi Surana; Martin C. Mihm; Monique J. Kauke; Kelly D. Moynihan; Alessandro Angelini; Robert T. Williams; Matthias T. Stephan; Jacob S. Kim; Michael B. Yaffe; Darrell J. Irvine; Louis M. Weiner; Glenn Dranoff; K. Dane Wittrup

Collaboration


Dive into the Cary Francis Opel's collaboration.

Top Co-Authors

Avatar

Darrell J. Irvine

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

Eric F. Zhu

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Karl Dane Wittrup

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Kelly D. Moynihan

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
Top Co-Authors

Avatar

Byron Hua Kwan

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
Researchain Logo
Decentralizing Knowledge