This new R01 proposal is a collaboration between two investigators, Wittrup and Irvine, combining protein engineering and nanoparticle synthesis expertise. Our central hypothesis is that the therapeutic index of cancer immunotherapy can be improved significantly by using novel methods to locally concentrate potent immunostimulatory molecules in tumor tissue for increased efficacy and decreased off-target toxicity. We will develop two complementary and potentially synergistic localized delivery methods for immunotherapy of cancer: pretargeting, and intratumoral nanoparticle injection. The two methods will be optimized for combined utility in syngeneic and genetically engineered mouse tumor models. We will explicate the immune therapeutic mechanisms of protocols that demonstrate efficacy. We have developed a bispecific antibody-based pretargeting protocol that provides highly tumor-specific localization of the chelator DOTA. We will site-specifically attach DOTA to the payloads IL-2, IL-12, TNF-?, ?-CTLA4 scFv, and ?-CD137 scFv. These molecules were chosen due to their demonstrated immuno-therapeutic potential in clinical trials, together with significant toxicity issues. Our protocol validated for DOTA-radiometal chelates will be adapted for specific delivery of the DOTA-labeled payloads. We have devised liposomal and stabilized micellar vehicles for surface anchoring of immunostimulatory molecules, and demonstrated their efficacy and safety from intratumoral injection into B16F10 syngeneic melanoma tumors. The same bispecific antibody used for pretargeting will be anchored on the surface of these vehicles, so that the exact same DOTA-labeled payloads can be modularly tested without re-optimization of conjugation methods. The bsAb is a scaffold that enables straightforward mimicry of immunocytokines, bispecific antibodies, and Fc conjugates by noncovalent conjugation with DOTA-labeled payloads. This will enable us to benchmark safety and efficacy of our novel approaches against these more commonly used vehicles, using the same antibody for tumor targeting and identical immunostimulatory molecules. We will test these protocols in transgenic mice expressing CEA, inoculated subcutaneously with B16F10 tumors expressing human CEA. The most successful protocols will be further tested in subcutaneous MC38-CEA tumors, and then in genetically engineered KP tumors in lung and sarcoma (floxed p53 knockout and stop-floxed activated KRAS expression via Cre recombinase delivered virally.) We will closely examine the tumor microenvironment and tumor draining lymph nodes following treatment by the most efficacious protocols, for evidence of reversal of immunosuppression by Tregs, TAMs, or MDSCs. We will also test for protective immunity and antigen spreading using syngeneic tumors lacking the antigen targeted by the bsAb (CEA).

Public Health Relevance

A team of two bioengineering labs at MIT will collaborate to develop novel methods of delivering potent immunostimulatory drugs directly to tumors while sparing healthy tissue. These methods will utilize nanoparticle synthesis and protein engineering, and will be tested in cutting-edge models of cancer in mice. The immune mechanisms affected by these therapies will be studied closely to learn generalizable lessons about the interface between cancer and the immune system.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
1R01CA174795-01
Application #
8476648
Study Section
Cancer Immunopathology and Immunotherapy Study Section (CII)
Program Officer
Welch, Anthony R
Project Start
2013-06-04
Project End
2018-04-30
Budget Start
2013-06-04
Budget End
2014-04-30
Support Year
1
Fiscal Year
2013
Total Cost
$395,388
Indirect Cost
$140,008
Name
Massachusetts Institute of Technology
Department
Internal Medicine/Medicine
Type
Schools of Arts and Sciences
DUNS #
001425594
City
Cambridge
State
MA
Country
United States
Zip Code
02139
Yang, Yu-Sang Sabrina; Moynihan, Kelly D; Bekdemir, Ahmet et al. (2018) Targeting small molecule drugs to T cells with antibody-directed cell-penetrating gold nanoparticles. Biomater Sci 7:113-124
Chen, Tiffany F; Li, Kevin K; Zhu, Eric F et al. (2018) Artificial Anti-Tumor Opsonizing Proteins with Fibronectin Scaffolds Engineered for Specificity to Each of the Murine Fc?R Types. J Mol Biol 430:1786-1798
Kelly, Ryan L; Le, Doris; Zhao, Jessie et al. (2018) Reduction of Nonspecificity Motifs in Synthetic Antibody Libraries. J Mol Biol 430:119-130
Moynihan, Kelly D; Holden, Rebecca L; Mehta, Naveen K et al. (2018) Enhancement of Peptide Vaccine Immunogenicity by Increasing Lymphatic Drainage and Boosting Serum Stability. Cancer Immunol Res 6:1025-1038
Kauke, Monique J; Traxlmayr, Michael W; Parker, Jillian A et al. (2017) An engineered protein antagonist of K-Ras/B-Raf interaction. Sci Rep 7:5831
Kwan, Byron H; Zhu, Eric F; Tzeng, Alice et al. (2017) Integrin-targeted cancer immunotherapy elicits protective adaptive immune responses. J Exp Med 214:1679-1690
Wittrup, K Dane (2017) Antitumor Antibodies Can Drive Therapeutic T Cell Responses. Trends Cancer 3:615-620
Smith, Tyrel T; Moffett, Howell F; Stephan, Sirkka B et al. (2017) Biopolymers codelivering engineered T cells and STING agonists can eliminate heterogeneous tumors. J Clin Invest 127:2176-2191
Moynihan, Kelly D; Irvine, Darrell J (2017) Roles for Innate Immunity in Combination Immunotherapies. Cancer Res 77:5215-5221
Milling, Lauren; Zhang, Yuan; Irvine, Darrell J (2017) Delivering safer immunotherapies for cancer. Adv Drug Deliv Rev 114:79-101

Showing the most recent 10 out of 27 publications