Network


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

Hotspot


Dive into the research topics where Mitchell Thorn is active.

Publication


Featured researches published by Mitchell Thorn.


Clinical Cancer Research | 2015

Phase I Hepatic Immunotherapy for Metastases study of intra-arterial chimeric antigen receptor modified T cell therapy for CEA+ liver metastases

Steven C. Katz; Rachel A. Burga; Elise McCormack; Li Juan Wang; Wesley Mooring; Gary R. Point; Pranay D. Khare; Mitchell Thorn; Qiangzhong Ma; Brian F. Stainken; Earle O. Assanah; Robin Davies; N. Joseph Espat; Richard P. Junghans

Purpose: Chimeric antigen receptor–modified T cells (CAR-T) have demonstrated encouraging results in early-phase clinical trials. Successful adaptation of CAR-T technology for CEA-expressing adenocarcinoma liver metastases, a major cause of death in patients with gastrointestinal cancers, has yet to be achieved. We sought to test intrahepatic delivery of anti-CEA CAR-T through percutaneous hepatic artery infusions (HAIs). Experimental Design: We conducted a phase I trial to test HAI of CAR-T in patients with CEA+ liver metastases. Six patients completed the protocol, and 3 received anti-CEA CAR-T HAIs alone in dose-escalation fashion (108, 109, and 1010 cells). We treated an additional 3 patients with the maximum planned CAR-T HAI dose (1010 cells × 3) along with systemic IL2 support. Results: Four patients had more than 10 liver metastases, and patients received a mean of 2.5 lines of conventional systemic therapy before enrollment. No patient suffered a grade 3 or 4 adverse event related to the CAR-T HAIs. One patient remains alive with stable disease at 23 months following CAR-T HAI, and 5 patients died of progressive disease. Among the patients in the cohort that received systemic IL2 support, CEA levels decreased 37% (range, 19%–48%) from baseline. Biopsies demonstrated an increase in liver metastasis necrosis or fibrosis in 4 of 6 patients. Elevated serum IFNγ levels correlated with IL2 administration and CEA decreases. Conclusions: We have demonstrated the safety of anti-CEA CAR-T HAIs with encouraging signals of clinical activity in a heavily pretreated population with large tumor burdens. Further clinical testing of CAR-T HAIs for liver metastases is warranted. Clin Cancer Res; 21(14); 3149–59. ©2015 AACR.


Cancer Immunology, Immunotherapy | 2015

Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-T

Rachel A. Burga; Mitchell Thorn; Gary R. Point; Prajna Guha; Cang T. Nguyen; Lauren A. Licata; Ronald P. DeMatteo; Alfred Ayala; N. Joseph Espat; Richard P. Junghans; Steven C. Katz

Chimeric antigen receptor-modified T cell (CAR-T) technology, a promising immunotherapeutic tool, has not been applied specifically to treat liver metastases (LM). While CAR-T delivery to LM can be optimized by regional intrahepatic infusion, we propose that liver CD11b+Gr-1+ myeloid-derived suppressor cells (L-MDSC) will inhibit the efficacy of CAR-T in the intrahepatic space. We studied anti-CEA CAR-T in a murine model of CEA+ LM and identified mechanisms through which L-MDSC expand and inhibit CAR-T function. We established CEA+ LM in mice and studied purified L-MDSC and responses to treatment with intrahepatic anti-CEA CAR-T infusions. L-MDSC expanded threefold in response to LM, and their expansion was dependent on GM-CSF, which was produced by tumor cells. L-MDSC utilized PD-L1 to suppress anti-tumor responses through engagement of PD-1 on CAR-T. GM-CSF, in cooperation with STAT3, promoted L-MDSC PD-L1 expression. CAR-T efficacy was rescued when mice received CAR-T in combination with MDSC depletion, GM-CSF neutralization to prevent MDSC expansion, or PD-L1 blockade. As L-MDSC suppressed anti-CEA CAR-T, infusion of anti-CEA CAR-T in tandem with agents targeting L-MDSC is a rational strategy for future clinical trials.


Cancer Gene Therapy | 2016

Regional CAR-T cell infusions for peritoneal carcinomatosis are superior to systemic delivery.

Steven C. Katz; Gary R. Point; M Cunetta; Mitchell Thorn; Prajna Guha; N J Espat; C Boutros; N Hanna; Richard P. Junghans

Metastatic spread of colorectal cancer (CRC) to the peritoneal cavity is common and difficult to treat, with many patients dying from malignant bowel obstruction. Chimeric antigen receptor T cell (CAR-T) immunotherapy has shown great promise, and we previously reported murine and phase I clinical studies on regional intrahepatic CAR-T infusion for CRC liver metastases. We are now studying intraperitoneal (IP) delivery of CAR-Ts for peritoneal carcinomatosis. Regional IP infusion of CAR-T resulted in superior protection against carcinoembryonic antigen (CEA+) peritoneal tumors, when compared with systemically infused CAR-Ts. IP CAR-Ts also provided prolonged protection against IP tumor re-challenges and demonstrated an increase in effector memory phenotype over time. IP CAR-Ts provided protection against tumor growth at distant subcutaneous (SC) sites in association with increases in serum IFNγ levels. Given the challenges posed by immunoinhibitory pathways in solid tumors, we combined IP CAR-T treatment with suppressor cell targeting. High frequencies of myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg) were found within the IP tumors, with MDSC expressing high levels of immunosuppressive PD-L1. Combinatorial IP CAR-T treatment with depleting antibodies against MDSC and Treg further improved efficacy against peritoneal metastases. Our data support further development of combinatorial IP CAR-T immunotherapy for peritoneal malignancies.


Cancer Gene Therapy | 2016

Tumor-associated GM-CSF overexpression induces immunoinhibitory molecules via STAT3 in myeloid-suppressor cells infiltrating liver metastases

Mitchell Thorn; Prajna Guha; M Cunetta; N J Espat; George Miller; Richard P. Junghans; Steven C. Katz

Assumptions that liver immune cells and immunosuppressive pathways are similar to their counterparts in other spaces have led to gaps in our understanding of intrahepatic neoplasm aggressiveness. Myeloid-derived suppressor cells (MDSCs) are potent inhibitors of antitumor immunity and pose a major obstacle to solid tumor treatment. Liver MDSCs (L-MDSCs) associated with liver metastases (LM) are particularly problematic by contributing to intrahepatic immunosuppression that promotes tumor progression. L-MDSCs have been reported to expand in response to granulocyte-macrophages colony-stimulating factor (GM-CSF) and suppress antitumor immunity in LM. To extend these findings, we examined mechanisms of intrahepatic immunosuppression exploited by L-MDSCs. We found that the majority of L-MDSCs co-expressed GM-CSF receptor (GM-CSF-R), indoleamine 2,3-dioxygenase (IDO) and programmed death ligand 1 (PD-L1), while demonstrating high levels of signal transducer and activator of transcription factor 3 (STAT3) activation. GM-CSF-secreting tumor cells induced STAT3 phosphorylation in L-MDSCs in addition to expression of IDO and PD-L1. GM-CSF or GM-CSF-R blockade markedly reduced L-MDSC IDO and PD-L1 expression, implicating tumor-derived GM-CSF in supporting L-MDSC-immunoinhibitory molecule expression. Small-molecule inhibitors of Janus-activated kinase 2 (JAK2) and STAT3 also dramatically diminished IDO and PD-L1 expression in L-MDSCs. We determined that STAT3 exerts transcriptional control over L-MDSC IDO and PD-L1 expression by binding to the IDO1 and PD-L1 promoters. Our data suggest that the GM-CSF/JAK2/STAT3 axis in L-MDSCs drives immunosuppression in a model of LM and blockade of this pathway may enable rescue of intrahepatic antitumor immunity.


Journal of Leukocyte Biology | 2013

Biliary obstruction results in PD-1-dependent liver T cell dysfunction and acute inflammation mediated by Th17 cells and neutrophils

Lauren A. Licata; Cang T. Nguyen; Rachel A. Burga; Vincent Falanga; N. Joseph Espat; Alfred Ayala; Mitchell Thorn; Richard P. Junghans; Steven C. Katz

Biliary obstruction is a common clinical problem that is associated with intrahepatic inflammation and impaired immunity. PD‐1 is well known to mediate T cell dysfunction but has been reported to promote and attenuate acute inflammation in various injury models. With the use of a well‐established murine model of BDL, we studied the effects of intrahepatic PD‐1 expression on LTC function, inflammation, and cholestasis. Following BDL, PD‐1 expression increased significantly among LTCs. Increased PD‐1 expression following BDL was associated with decreased LTC proliferation and less IFN‐γ production. Elimination of PD‐1 expression resulted in significantly improved proliferative capacity among LTC following BDL, in addition to a more immunostimulatory cytokine profile. Not only was LTC function rescued in PD‐1−/− mice, but also, the degrees of biliary cell injury, cholestasis, and inflammation were diminished significantly compared with WT animals following BDL. PD‐1‐mediated acute inflammation following BDL was associated with expansions of intrahepatic neutrophil and Th17 cell populations, with the latter dependent on IL‐6. PD‐1 blockade represents an attractive strategy for reversing intrahepatic immunosuppression while limiting inflammatory liver damage.


Cancer Gene Therapy | 2014

Neutrophil:lymphocyte ratios and serum cytokine changes after hepatic artery chimeric antigen receptor-modified T-cell infusions for liver metastases

A Saied; Lauren A. Licata; Rachel A. Burga; Mitchell Thorn; Elise McCormack; Brian F. Stainken; E O Assanah; P D Khare; Robin Davies; N J Espat; Richard P. Junghans; Steven C. Katz

Our phase I Hepatic Immunotherapy for Metastases (HITM) trial tested the safety of chimeric antigen receptor-modified T-cell (CAR-T) hepatic artery infusions (HAI) for unresectable carcinoembryonic antigen (CEA)+ liver metastases (LM). High neutrophil:lymphocyte ratios (NLR) predict poor outcome in cancer patients and we hypothesized that NLR changes would correlate with early responses to CAR-T HAI. Six patients completed the protocol. Three patients received CAR-T HAI in dose escalation (1 × 108, 1 × 109 and 1 × 1010 cells) and the remainder received three doses (1 × 1010 cells) with interleukin (IL)2 support. Serum cytokines and NLR were measured at multiple time points. The mean NLR for all patients was 13.9 (range 4.8–38.1). NLR increased in four patients following treatment with a mean fold change of 1.9. Serum IL6 levels and NLR fold changes demonstrated a trend towards a positive correlation (r=0.77, P=0.10). Patients with poor CEA responses were significantly more likely to have higher NLR level increases (P=0.048). Increased NLR levels were associated with poor responses following CAR-T HAI. NLR variations and associated cytokine changes may be useful surrogates of response to CAR-T HAI.


Journal of Translational Medicine | 2013

Anti-KIT designer T cells for the treatment of gastrointestinal stromal tumor

Steven C. Katz; Rachel A. Burga; Seema Naheed; Lauren A. Licata; Mitchell Thorn; Doreen Osgood; Cang T. Nguyen; N. Joseph Espat; Jonathan A. Fletcher; Richard P. Junghans

BackgroundImatinib mesylate is an effective treatment for metastatic gastrointestinal stromal tumor (GIST). However, most patients eventually develop resistance and there are few other treatment options. Immunotherapy using genetically modified or designer T cells (dTc) has gained increased attention for several malignancies in recent years. The aims of this study were to develop and test novel anti-KIT dTc engineered to target GIST cells.MethodsHuman anti-KIT dTc were created by retroviral transduction with novel chimeric immune receptors (CIR). The gene for stem cell factor (SCF), the natural ligand for KIT, was cloned into 1st generation (SCF-CD3ζ, 1st gen) and 2nd generation (SCF-CD28-CD3ζ, 2nd gen) CIR constructs. In vitro dTc proliferation and tumoricidal capacity in the presence of KIT+ tumor cells were measured. In vivo assessment of dTc anti-tumor efficacy was performed by treating immunodeficient mice harboring subcutaneous GIST xenografts with dTc tail vein infusions.ResultsWe successfully produced the 1st and 2nd gen anti-KIT CIR and transduced murine and human T cells. Average transduction efficiencies for human 1st and 2nd gen dTc were 50% and 42%. When co-cultured with KIT+ tumor cells, both 1st and 2nd gen dTc proliferated and produced IFNγ. Human anti-KIT dTc were efficient at lysing GIST in vitro compared to untransduced T cells. In mice with established GIST xenografts, treatment with either 1st or 2nd gen human anti-KIT dTc led to significant reductions in tumor growth rates.ConclusionsWe have constructed a novel anti-KIT CIR for production of dTc that possess specific activity against KIT+ GIST in vitro and in vivo. Further studies are warranted to evaluate the therapeutic potential and safety of anti-KIT dTc.


Journal of Leukocyte Biology | 2014

Liver metastases induce reversible hepatic B cell dysfunction mediated by Gr-1+CD11b+ myeloid cells

Mitchell Thorn; Gary Point; Rachel A. Burga; Cang T. Nguyen; N. Joseph Espat; Steven C. Katz

LM escape immune surveillance, in part, as a result of the expansion of CD11b+MC, which alter the intrahepatic microenvironment to promote tumor tolerance. HBC make up a significant proportion of liver lymphocytes and appear to delay tumor progression; however, their significance in the setting of LM is poorly defined. Therefore, we characterized HBC and HBC/CD11b+MC interactions using a murine model of LM. Tumor‐bearing livers showed a trend toward elevated absolute numbers of CD19+ HBC. A significant increase in the frequency of IgMloIgDhi mature HBC was observed in mice with LM compared with normal mice. HBC derived from tumor‐bearing mice demonstrated increased proliferation in response to TLR and BCR stimulation ex vivo compared with HBC from normal livers. HBC from tumor‐bearing livers exhibited significant down‐regulation of CD80 and were impaired in inducing CD4+ T cell proliferation ex vivo. We implicated hepatic CD11b+MC as mediators of CD80 down‐modulation on HBC ex vivo via a CD11b‐dependent mechanism that required cell‐to‐cell contact and STAT3 activity. Therefore, CD11b+MC may compromise the ability of HBC to promote T cell activation in the setting of LM as a result of diminished expression of CD80. Cross‐talk between CD11b+MC and HBC may be an important component of LM‐induced immunosuppression.


Archive | 2013

Vertebrate Immune System Evolution and Comparative Primate Immunity

Jessica F. Brinkworth; Mitchell Thorn

The primate immune system is the outcome of hundreds of millions of years of complex evolution. Its divergence from the immune systems of other species and variation within the order affect primate disease susceptibility, infectious disease emergence, and human health. There is, however, limited information on differences in baseline immune function within the primate order. As such, a comparative immunological review of vertebrate immunity can offer insight into the evolutionary divergence of primate immune system function. The goal of this chapter is to provide an evolutionary background for the development and diversification of the primate immune system. Here, we put primate immunity in the context of the emergence of the major structures and cellular components and the vertebrate immune system over evolutionary time with special attention paid, when information is available, to examples of how baseline immunity has diverged between primate species.


Journal of Clinical Oncology | 2013

Targeting myeloid-derived suppressor cells and the PD-1/PD-L1 axis to enhance immunotherapy with anti-CEA designer T cells for the treatment of colorectal liver metastases.

Rachel A. Burga; Mitchell Thorn; Cang T. Nguyen; Lauren A. Licata; N. Joseph Espat; Richard P. Junghans; Steven C. Katz

Collaboration


Dive into the Mitchell Thorn's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard P. Junghans

Roger Williams Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rachel A. Burga

Roger Williams Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cang T. Nguyen

Roger Williams Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gary Point

Roger Williams Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lauren A. Licata

Roger Williams Medical Center

View shared research outputs
Top Co-Authors

Avatar

Prajna Guha

Roger Williams Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marissa Cunetta

Roger Williams Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge