Network


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

Hotspot


Dive into the research topics where Eric F. Zhu is active.

Publication


Featured researches published by Eric F. Zhu.


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.


Journal of Experimental Medicine | 2017

Integrin-targeted cancer immunotherapy elicits protective adaptive immune responses

Byron Hua Kwan; Eric F. Zhu; Alice Tzeng; Harun R. Sugito; Ahmed A. Eltahir; Botong Ma; Mary K. Delaney; Patrick A. Murphy; Monique J. Kauke; Alessandro Angelini; Noor Momin; Naveen K. Mehta; Alecia M. Maragh; Richard O. Hynes; Glenn Dranoff; Jennifer R. Cochran; K. Dane Wittrup

Certain RGD-binding integrins are required for cell adhesion, migration, and proliferation and are overexpressed in most tumors, making them attractive therapeutic targets. However, multiple integrin antagonist drug candidates have failed to show efficacy in cancer clinical trials. In this work, we instead exploit these integrins as a target for antibody Fc effector functions in the context of cancer immunotherapy. By combining administration of an engineered mouse serum albumin/IL-2 fusion with an Fc fusion to an integrin-binding peptide (2.5F-Fc), significant survival improvements are achieved in three syngeneic mouse tumor models, including complete responses with protective immunity. Functional integrin antagonism does not contribute significantly to efficacy; rather, this therapy recruits both an innate and adaptive immune response, as deficiencies in either arm result in reduced tumor control. Administration of this integrin-targeted immunotherapy together with an anti–PD-1 antibody further improves responses and predominantly results in cures. Overall, this well-tolerated therapy achieves tumor specificity by redirecting inflammation to a functional target fundamental to tumorigenic processes but expressed at significantly lower levels in healthy tissues, and it shows promise for translation.


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 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 | 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 immunology research | 2013

Koch Institute Symposium on Cancer Immunology and Immunotherapy

Adam Drake; Nikhil S. Joshi; Gregory L. Szeto; Eric F. Zhu; Herman N. Eisen; Darrell J. Irvine

The 12th annual summer symposium of The Koch Institute for Integrative Cancer Research at MIT was held in Cambridge, Massachusetts, on June 14, 2013. The symposium, entitled “Cancer Immunology and Immunotherapy,” focused on recent advances in preclinical research in basic immunology and biomedical engineering and their clinical application in cancer therapies. The day-long gathering also provided a forum for discussion and potential collaborations between engineers and clinical investigators. The major topics presented included (i) enhancement of adoptive cell therapy by engineering to improve the ability and functionality of T cells against tumor cells; (ii) current therapies using protein and antibody therapeutics to modulate endogenous antitumor immunity; and (iii) new technologies to identify molecular targets and assess therapeutic efficacy, and devices to control and target drug delivery more effectively and efficiently. Cancer Immunol Res; 1(4); 217–22. ©2013 AACR.


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


Chemistry of Materials | 2017

Redox Interfaces for Electrochemically Controlled Protein–Surface Interactions: Bioseparations and Heterogeneous Enzyme Catalysis

Xiao Su; Jonas Hübner; Monique J. Kauke; Luiza Dalbosco; Jonathan Thomas; Christopher C. Gonzalez; Eric F. Zhu; Matthias Franzreb; Timothy F. Jamison; T. Alan Hatton


Cell Reports | 2016

Temporally Programmed CD8α+ DC Activation Enhances Combination Cancer Immunotherapy

Alice Tzeng; Monique J. Kauke; Eric F. Zhu; Kelly D. Moynihan; Cary Francis Opel; Nicole J. Yang; Naveen K. Mehta; Ryan L. Kelly; Gregory L. Szeto; Willem W. Overwijk; Darrell J. Irvine; K. Dane Wittrup


Archive | 2014

SYNERGISTIC TUMOR TREATMENT WITH EXTENDED-PK IL-2 AND ADOPTIVE CELL THERAPY

Karl Dane Wittrup; Cary Francis Opel; Eric F. Zhu; Shuning Gai

Collaboration


Dive into the Eric F. Zhu'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

Monique J. Kauke

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

K. 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

Alice Tzeng

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

Karl Dane Wittrup

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge