Network


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

Hotspot


Dive into the research topics where Leah Mitchell is active.

Publication


Featured researches published by Leah Mitchell.


Science Translational Medicine | 2016

Phase 1 trial of vocimagene amiretrorepvec and 5-fluorocytosine for recurrent high-grade glioma

Timothy F. Cloughesy; Joseph Landolfi; Daniel Hogan; Stephen Bloomfield; Bob S. Carter; Clark C. Chen; J. Bradley Elder; Steven N. Kalkanis; Santosh Kesari; Albert Lai; Ian Y. Lee; Linda M. Liau; Tom Mikkelsen; Phioanh L. Nghiemphu; David Piccioni; Tobias Walbert; Alice Chu; Asha Das; Oscar Diago; Dawn Gammon; Harry E. Gruber; Michelle M. Hanna; Douglas J. Jolly; Noriyuki Kasahara; David R. McCarthy; Leah Mitchell; Derek Ostertag; Joan M. Robbins; Maria Rodriguez-Aguirre; Michael A. Vogelbaum

Toca 511 and Toca FC show promising results in treating recurrent high-grade glioma, and a specific molecular signature correlates with treatment-related survival. Tag-team attack on glioma Toca FC (extended-release 5-fluorocytosine) and Toca 511 (vocimagene amiretrorepvec) are an investigational therapeutic combination for glioma, consisting of two parts: a prodrug that is inactive on its own and a modified virus that infects the tumor and delivers an enzyme, which then activates the drug and allows it to kill the glioma cells. Cloughesy et al. tested this therapy in 45 human patients with recurrent or progressive high-grade glioma and discovered that the treatment was well tolerated and improved survival compared to an external control group. In addition, the authors identified a gene signature that correlated with response to the treatment, which may help identify the patients most likely to benefit from this approach. Toca 511 (vocimagene amiretrorepvec) is an investigational nonlytic, retroviral replicating vector (RRV) that delivers a yeast cytosine deaminase, which converts subsequently administered courses of the investigational prodrug Toca FC (extended-release 5-fluorocytosine) into the antimetabolite 5-fluorouracil. Forty-five subjects with recurrent or progressive high-grade glioma were treated. The end points of this phase 1, open-label, ascending dose, multicenter trial included safety, efficacy, and molecular profiling; survival was compared to a matching subgroup from an external control. Overall survival for recurrent high-grade glioma was 13.6 months (95% confidence interval, 10.8 to 20.0) and was statistically improved relative to an external control (hazard ratio, 0.45; P = 0.003). Tumor samples from subjects surviving more than 52 weeks after Toca 511 delivery disproportionately displayed a survival-related mRNA expression signature, identifying a potential molecular signature that may correlate with treatment-related survival rather than being prognostic. Toca 511 and Toca FC show excellent tolerability, with RRV persisting in the tumor and RRV control systemically. The favorable assessment of Toca 511 and Toca FC supports confirmation in a randomized phase 2/3 trial (NCT02414165).


Neuro-oncology | 2017

Toca 511 gene transfer and treatment with the prodrug, 5-fluorocytosine, promotes durable antitumor immunity in a mouse glioma model

Leah Mitchell; Fernando Lopez Espinoza; Daniel Mendoza; Yuki Kato; Akihito Inagaki; Kei Hiraoka; Noriyuki Kasahara; Harry E. Gruber; Douglas J. Jolly; Joan M. Robbins

Abstract Background. Toca 511 (vocimagene amiretrorepvec) is a retroviral replicating vector encoding an optimized yeast cytosine deaminase (CD). Tumor-selective expression of CD converts the prodrug, 5-fluorocytosine (5-FC), into the active chemotherapeutic, 5-fluorouracil (5-FU). This therapeutic approach is being tested in a randomized phase II/III trial in recurrent glioblastoma and anaplastic astrocytoma (NCT0241416). The aim of this study was to identify the immune cell subsets contributing to antitumor immune responses following treatment with 5-FC in Toca 511–expressing gliomas in a syngeneic mouse model. Methods. Flow cytometry was utilized to monitor and characterize the immune cell infiltrate in subcutaneous Tu-2449 gliomas in B6C3F1 mice treated with Toca 511 and 5-FC. Results. Tumor-bearing animals treated with Toca 511 and 5-FC display alterations in immune cell populations within the tumor that result in antitumor immune protection. Attenuated immune subsets were exclusive to immunosuppressive cells of myeloid origin. Depletion of immunosuppressive cells temporally preceded a second event which included expansion of T cells which were polarized away from Th2 and Th17 in the CD4+ T cell compartment with concomitant expansion of interferon gamma–expressing CD8+ T cells. Immune alterations correlated with clearance of Tu-2449 subcutaneous tumors and T cell–dependent protection from future tumor challenge. Conclusions. Treatment with Toca 511 and 5-FC has a concentrated effect at the site of the tumor which causes direct tumor cell death and alterations in immune cell infiltrate, resulting in a tumor microenvironment that is more permissive to establishment of a T cell mediated antitumor immune response.


Neuro-oncology | 2017

Retroviral replicating vector-mediated gene therapy achieves long-term control of tumor recurrence and leads to durable anticancer immunity

Kei Hiraoka; Akihito Inagaki; Yuki Kato; Tiffany T. Huang; Leah Mitchell; Shuichi Kamijima; Masamichi Takahashi; Hiroshi Matsumoto; Katrin Hacke; Carol A. Kruse; Derek Ostertag; Joan M. Robbins; Harry E. Gruber; Douglas J. Jolly; Noriyuki Kasahara

Abstract Background. Prodrug-activator gene therapy with Toca 511, a tumor-selective retroviral replicating vector (RRV) encoding yeast cytosine deaminase, is being evaluated in recurrent high-grade glioma patients. Nonlytic retroviral infection leads to permanent integration of RRV into the cancer cell genome, converting infected cancer cell and progeny into stable vector producer cells, enabling ongoing transduction and viral persistence within tumors. Cytosine deaminase in infected tumor cells converts the antifungal prodrug 5-fluorocytosine into the anticancer drug 5-fluorouracil, mediating local tumor destruction without significant systemic adverse effects. Methods. Here we investigated mechanisms underlying the therapeutic efficacy of this approach in orthotopic brain tumor models, employing both human glioma xenografts in immunodeficient hosts and syngeneic murine gliomas in immunocompetent hosts. Results. In both models, a single injection of replicating vector followed by prodrug administration achieved long-term survival benefit. In the immunodeficient model, tumors recurred repeatedly, but bioluminescence imaging of tumors enabled tailored scheduling of multicycle prodrug administration, continued control of disease burden, and long-term survival. In the immunocompetent model, complete loss of tumor signal was observed after only 1–2 cycles of prodrug, followed by long-term survival without recurrence for >300 days despite discontinuation of prodrug. Long-term survivors rejected challenge with uninfected glioma cells, indicating immunological responses against native tumor antigens, and immune cell depletion showed a critical role for CD4+ T cells. Conclusion. These results support dual mechanisms of action contributing to the efficacy of RRV-mediated prodrug-activator gene therapy: long-term tumor control by prodrug conversion-mediated cytoreduction, and induction of antitumor immunity.


Molecular therapy. Nucleic acids | 2017

Retroviral Replicating Vector Delivery of miR-PDL1 Inhibits Immune Checkpoint PDL1 and Enhances Immune Responses In Vitro

Amy H. Lin; Christopher G. Twitty; Ryan Burnett; Andrew Hofacre; Leah Mitchell; Fernando Lopez Espinoza; Harry E. Gruber; Douglas J. Jolly

Tumor cells express a number of immunosuppressive molecules that can suppress anti-tumor immune responses. Efficient delivery of small interfering RNAs to treat a wide range of diseases including cancers remains a challenge. Retroviral replicating vectors (RRV) can be used to stably and selectively introduce genetic material into cancer cells. Here, we designed RRV to express shRNA (RRV-shPDL1) or microRNA30-derived shRNA (RRV-miRPDL1) using Pol II or Pol III promoters to downregulate PDL1 in human cancer cells. We also designed RRV expressing cytosine deaminase (yCD2) and miRPDL1 for potential combinatorial therapy. Among various configurations tested, we showed that RRV-miRPDL1 vectors with Pol II or Pol III promoter replicated efficiently and exhibited sustained downregulation of PDL1 protein expression by more than 75% in human cancer cell lines with high expression of PDL1. Immunologic effects of RRV-miRPDL1 were assessed by a trans-suppression lymphocyte assay. In vitro data showed downregulation of PDL1+ tumor cells restored activation of CD8+ T cells and bio-equivalency compared to anti-PDL1 antibody treatment. These results suggest RRV-miRPDL1 may be an alternative therapeutic approach to enhance anti-tumor immunity by overcoming PDL1-induced immune suppression from within cancer cells and this approach may also be applicable to other cancer targets.


Molecular Therapy - Oncolytics | 2018

A Retroviral Replicating Vector Encoding Cytosine Deaminase and 5-FC Induces Immune Memory in Metastatic Colorectal Cancer Models

Kader Yagiz; Maria Rodriguez-Aguirre; Fernando Lopez Espinoza; Tiffany T. Montellano; Daniel Mendoza; Leah Mitchell; Carlos E. Ibanez; Noriyuki Kasahara; Harry E. Gruber; Douglas J. Jolly; Joan M. Robbins

Treatment of tumors with Toca 511, a gamma retroviral replicating vector encoding cytosine deaminase, followed by 5-fluorocytosine (5-FC) kills tumors by local production of 5-fluorouracil (5-FU). In brain tumor models, this treatment induces systemic anti-tumor immune responses and long-term immune-mediated survival. Phase 1 Toca 511 and Toca FC (extended-release 5-FC) clinical trials in patients with recurrent high-grade glioma show durable complete responses and promising survival data compared to historic controls. The work described herein served to expand on our earlier findings in two models of metastatic colorectal carcinoma (mCRC). Intravenous (i.v.) delivery of Toca 511 resulted in substantial tumor-selective uptake of vector into metastatic lesions. Subsequent treatment with 5-FC resulted in tumor shrinkage, improved survival, and immune memory against future rechallenge with the same CT26 CRC cell line. Similar results were seen in a brain metastasis model of mCRC. Of note, 5-FC treatment resulted in a significant decrease in myeloid-derived suppressor cells (MDSCs) in mCRC tumors in both the liver and brain. These results support the development of Toca 511 and Toca FC as a novel immunotherapeutic approach for patients with mCRC. A phase 1 study of i.v. Toca 511 and Toca FC in solid tumors, including mCRC, is currently underway (NCT02576665).


Cancer Research | 2018

Abstract 5630: Immune profile of tumor microenvironment helps predict response in patients treated with an investigational immunotherapeutic consisting of a retroviral replicating vector (Toca 511) and an extended-release formulation of 5-fluorocytosine (Toca FC)

Derek Ostertag; William Accomando; Leah Mitchell; Maria Rodriguez-Aguirre; Daniel Hogan; Oscar Diago; Dawn Gammon; Ali Haghighi; Harry E. Gruber; Asha Das; Douglas J. Jolly

Toca 511 (vocimagene amiretrorepvec) is an investigational, conditionally lytic, retroviral replicating vector (RRV). RRVs selectively infect cancer cells due to defects in innate and adaptive immune responses found in cancers that support virus replication, and cell division requirements for virus integration into the genome. Toca 511 spreads through cancer cells, stably delivering an optimized cytosine deaminase (CD) gene that converts the prodrug Toca FC (investigational, extended-release 5-fluorocytosine) into 5-fluorouracil (5-FU), a canonical chemotherapeutic. In preclinical tumor models, as infected cancer cells are killed, diffusible 5-FU also kills nearby susceptible cells, including uninfected cancer cells, and myeloid derived suppressor cells (MDSC) that contribute to immune-suppression in the tumor microenvironment. This action by Toca 511 and Toca FC has been shown in animal models to generate a durable anti-tumor immune response that can be transferred to naive, untreated animals. The Toca 511 and Toca FC immunotherapeutic are proposed to remodel the tumor microenvironment to break tumor tolerance resulting in induction of antitumor activity by the patient9s immune system and durable complete responses. In a phase 1 clinical study for recurrent high grade glioma (NCT01470794), Toca 511 was injected into resection cavity walls at time of resection followed by multiple courses of oral Toca FC. We observed multi-year durable and objective responses; including 5 ongoing complete responses in a group of 23 patients in the higher dose single agent treatment cohorts given approximately the same Toca 511 doses and having the same entry criteria as an ongoing Phase 3 study in recurrent high grade glioma (NCT02414165). Patient tumors at time of resection were analyzed by exome sequencing, RNA sequencing, IHC, and TCR sequencing, before Toca 511 treatment. In this study, we report higher levels of tumor infiltrating T cells by TCR sequencing, before the start of treatment, were significantly associated with responding patients compared to patients whose disease progressed. The significance of this data is supported by the preclinical mechanism of action reported previously. Additionally, we plan to report on T cell, B cell, and myeloid populations in the tumor as measured by IHC and RNA sequencing and their relationship to clinical response. Data reported here will provide mechanistic context to the immunotherapeutic mode of action proposed to account for durable responses seen in treatment of brain tumors with Toca 511 and Toca FC. Citation Format: Derek Ostertag, William Accomando, Leah Mitchell, Maria Rodriguez-Aguirre, Daniel Hogan, Oscar Diago, Dawn Gammon, Ali Haghighi, Harry Gruber, Asha Das, Douglas Jolly. Immune profile of tumor microenvironment helps predict response in patients treated with an investigational immunotherapeutic consisting of a retroviral replicating vector (Toca 511) and an extended-release formulation of 5-fluorocytosine (Toca FC) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5630.


Cancer Research | 2016

Abstract 3212: Combining Toca 511 and 5-fluorocytosine with αPD-1 or αCTLA-4 antibody significantly reduces tumor burden compared to either checkpoint inhibitor alone or in combination in a subcutaneous mouse glioma model

Leah Mitchell; Fernando Lopez Espinoza; Kader Yagiz; Daniel Mendoza; Maria Rodriguez-Aguirre; Sean Mitchell; Douglas J. Jolly; Joan M. Robbins

Toca 511 (vocimagene amiretrorepvec), a retroviral replicating vector encoding an optimized yeast cytosine deaminase (CD) protein, selectively replicates and spreads in malignant cells. In infected cells, CD enzyme is expressed and converts 5-FC (5-fluorocytosine, an oral anti-fungal drug) to 5-FU, leading to both direct tumor cytotoxicity and extended immunotherapeutic effects. As of 9/25/2015, 116 patients with recurrent high grade glioma have been treated with Toca 511 & Toca FC in 3 ongoing Phase 1 clinical trials (NCT01156584, NCT01470794, NCT01985256), in which potential benefits were found, including extended overall survival and a favorable safety profile. The purpose of this study was to determine if combinations of Toca 511+5-FC with αPD-1, αCTLA-4, or both, are therapeutically useful in a syngeneic mouse model, and for which combinations we could see meaningful variations in tumor infiltrating immune cells. 2×10⁁6 Tu-2449SC glioma cells (2% pretransduced with Toca 511 to provide a uniform starting inoculum) were implanted on the right flanks of B6C3F1 mice and treatment initiated when tumors reached ≈100mm3. 5-FC treated mice received 500 mg/kg/day IP (5 days per week). αPD-1 was administered at 300μg/mouse IP followed by 3 maintenance doses every 3 days of 200μg/mouse. αCTLA-4 (100μg/mouse, IP) was administered every 3 days (total of 6 treatments). Isotype antibodies were administered to control groups. The study was terminated after 3 cycles of 5-FC. Tumors and spleens were collected and analyzed by flow cytometry for 26 unique immune cell populations. Tumor burden was significantly reduced with the addition of Toca 511+5-FC to either antibody treatment alone or in combination compared to antibody treatment groups. Treatment with Toca 511 and 5-FC alone was highly effective at controlling tumor growth, therefore, additive treatment with αPD-1, αCTLA-4, or αPD-1 + αCTLA-4 did not significantly further reduce tumor burden. The combination of αPD-1 and αCTLA-4, but not either antibody alone, significantly reduced tumor burden. Treatment with 5-FC significantly reduced tumor associated macrophages as well as myeloid derived suppressor cells and increased CD4+ T cell populations. Of the T cell populations that were increased in the tumors, a greater percentage of T cells were producing IFNγ. Combining Toca 511+5-FC with αPD-1, αCTLA-4 or the combination of antibodies significantly reduced tumor growth compared to antibody treatments alone. Tumor associated immune cell population analysis also revealed that Toca 511+5-FC altered the immune cell populations in the tumor to generate a more permissive environment for immune activation and anti-tumor immune response. Toca 511 & Toca FC may be broadly applicable in combination with checkpoint inhibitors to help activate the immune system. Citation Format: Leah Mitchell, Fernando Lopez Espinoza, Kader Yagiz, Daniel Mendoza, Maria Rodriguez-Aguirre, Sean Mitchell, Douglas J. Jolly, Joan M. Robbins. Combining Toca 511 and 5-fluorocytosine with αPD-1 or αCTLA-4 antibody significantly reduces tumor burden compared to either checkpoint inhibitor alone or in combination in a subcutaneous mouse glioma model. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3212.


Immune Monitoring / Clinical Correlates | 2018

Abstract B010: Antitumor cellular immune response elicited by Toca 511 and Toca FC therapy in preclinical and clinical studies

Tiffany T. Montellano; Derek Ostertag; Daniel Hogan; Oscar Diago; Dawn Gammon; Ali Haghighi; William Accomando; Leah Mitchell; Asha Das; Harry E. Gruber; Douglas J. Jolly


Neuro-oncology | 2017

ATIM-21. INTRAVENOUS DELIVERY OF TOCA 511 IN PATIENTS WITH HIGH GRADE GLIOMA RESULTS IN QUANTIFIABLE EXPRESSION OF CYTOSINE DEAMINASE IN TUMOR TISSUE

Tobias Walbert; Daniela A. Bota; Michael A. Vogelbaum; Steven N. Kalkanis; Linda M. Liau; Tom Mikkelsen; Harry E. Gruber; Jolene Shorr; Maria Rodriguez-Aquirre; Derek Ostertag; Leah Mitchell; Douglas J. Jolly; Timothy F. Cloughesy


Neuro-oncology | 2017

TMIC-42. TOCA 511 AND 5-FC INDUCES T CELL-MEDIATED ANTITUMOR IMMUNITY IN A MOUSE GLIOMA MODEL WHICH IS ENHANCED BY THE ADDITION OF A THERAPEUTIC ANTIBODY AGAINST CTLA-4 AND CORRELATIVE WITH A REDUCTION IN MEMORY T REGULATORY CELLS.

Douglas J. Jolly; Leah Mitchell; Kader Yagiz; Fernando Lopez Espinoza; Daniel Mendoza; Anthony Munday; Harry E. Gruber

Collaboration


Dive into the Leah Mitchell's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tom Mikkelsen

Henry Ford Health System

View shared research outputs
Top Co-Authors

Avatar

Linda M. Liau

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge