Network


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

Hotspot


Dive into the research topics where Thomas M. Schmitt is active.

Publication


Featured researches published by Thomas M. Schmitt.


Blood | 2010

The B-cell tumor–associated antigen ROR1 can be targeted with T cells modified to express a ROR1-specific chimeric antigen receptor

Michael Hudecek; Thomas M. Schmitt; Sivasubramanian Baskar; Maria Teresa Lupo-Stanghellini; Tetsuya Nishida; Tori N. Yamamoto; Marie Bleakley; Cameron J. Turtle; Wen-Chung Chang; Harvey A. Greisman; Brent L. Wood; David G. Maloney; Michael C. Jensen; Christoph Rader; Stanley R. Riddell

Monoclonal antibodies and T cells modified to express chimeric antigen receptors specific for B-cell lineage surface molecules such as CD20 exert antitumor activity in B-cell malignancies, but deplete normal B cells. The receptor tyrosine kinase-like orphan receptor 1 (ROR1) was identified as a highly expressed gene in B-cell chronic lymphocytic leukemia (B-CLL), but not normal B cells, suggesting it may serve as a tumor-specific target for therapy. We analyzed ROR1-expression in normal nonhematopoietic and hematopoietic cells including B-cell precursors, and in hematopoietic malignancies. ROR1 has characteristics of an oncofetal gene and is expressed in undifferentiated embryonic stem cells, B-CLL and mantle cell lymphoma, but not in major adult tissues apart from low levels in adipose tissue and at an early stage of B-cell development. We constructed a ROR1-specific chimeric antigen receptor that when expressed in T cells from healthy donors or CLL patients conferred specific recognition of primary B-CLL and mantle cell lymphoma, including rare drug effluxing chemotherapy resistant tumor cells that have been implicated in maintaining the malignancy, but not mature normal B cells. T-cell therapies targeting ROR1 may be effective in B-CLL and other ROR1-positive tumors. However, the expression of ROR1 on some normal tissues suggests the potential for toxi-city to subsets of normal cells.


Science Translational Medicine | 2013

Transferred WT1-reactive CD8+ T cells can mediate antileukemic activity and persist in post-transplant patients

Aude G. Chapuis; Gunnar B. Ragnarsson; Hieu Nguyen; Colette Chaney; Jeffrey S. Pufnock; Thomas M. Schmitt; Natalie Duerkopp; Ilana Roberts; Galina Pogosov; William Ho; Sebastian Ochsenreither; Matthias Wölfl; Merav Bar; Jerald P. Radich; Cassian Yee; Philip D. Greenberg

Donor-derived WT1-specific CD8+ T cells transferred after hematopoietic cell transplantation mediate antileukemic activity and can establish persistent responses without toxicity to normal tissues in high-risk leukemia patients. Targeting Leukemic Cells for Destruction After a patient is diagnosed with leukemia, the first line of treatment is generally chemotherapy. If it doesn’t work, the patient can get a bone marrow transplant, which can sometimes cure otherwise untreatable leukemia. However, for patients who have already relapsed after a transplant, or whose cancer has particularly unfavorable characteristics, few options remain. Such patients’ prognosis is generally very poor, with a high risk of relapse and death from leukemia. Immune cells derived from the donor bone marrow can help fight the cancer by attacking malignant cells inside the graft recipient. At the same time, however, they often attack the recipient’s healthy cells as well and cause graft-versus-host disease (GVHD), which can itself be lethal. Now, Chapuis and coauthors report a way to harness the power of the donors’ immune cells against some leukemias, without triggering GVHD in the bone marrow recipients. In this pilot trial, the authors enrolled 11 patients who had leukemia with poor prognostic characteristics and who had each undergone a bone marrow transplant. The patients then received T cells that were derived from their respective donors and selected for their ability to recognize Wilms tumor antigen 1 (WT1). Very small amounts of WT1 are present in some healthy cells, but its expression in malignant cells is much greater and corresponds to the aggressiveness of the cancer. In the first seven patients, the authors found that T cells that recognized WT1 could suppress the leukemia temporarily, but the new cells disappeared within 14 days, and the cancer rebounded. In the last four patients, the authors tried a modified protocol, pretreating the cells with interleukin-21 (IL-21) before infusion into the recipients. T cells pretreated with IL-21 developed characteristics of memory cells, including a greatly extended life span. At this time, all four patients who received IL-21–treated T cells are still alive, with no recurrence of their cancer up to 30 months after the T cell infusion. Notably, none of the patients who received WT1-specific T cells in this study have developed GVHD, supporting the idea that WT1 targeting is specific to the tumor cells and safe for patient use. The study by Chapuis et al. is a small pilot trial, and its results must be replicated with larger groups of patients before this protocol can become standard practice. Nevertheless, the combination of targeting WT1 in leukemia and pretreating leukemia-targeting T cells with IL-21 to extend their life span looks very promising and offers a potential safe and effective treatment for patients who have few other options. Relapse remains a leading cause of death after allogeneic hematopoietic cell transplantation (HCT) for patients with high-risk leukemias. The potentially beneficial donor T cell–mediated graft-versus-leukemia (GVL) effect is often mitigated by concurrent graft-versus-host disease (GVHD). Providing T cells that can selectively target Wilms tumor antigen 1 (WT1), a transcription factor overexpressed in leukemias that contributes to the malignant phenotype, represents an opportunity to promote antileukemic activity without inducing GVHD. HLA-A*0201–restricted WT1-specific donor-derived CD8+ cytotoxic T cell (CTL) clones were administered after HCT to 11 relapsed or high-risk leukemia patients without evidence of on-target toxicity. The last four treated patients received CTL clones generated with exposure to interleukin-21 (IL-21) to prolong in vivo CTL survival, because IL-21 can limit terminal differentiation of antigen-specific T cells generated in vitro. Transferred cells exhibited direct evidence of antileukemic activity in two patients: a transient response in one patient with advanced progressive disease and the induction of a prolonged remission in a patient with minimal residual disease (MRD). Additionally, three treated patients at high risk for relapse after HCT survive without leukemia relapse, GVHD, or additional antileukemic treatment. CTLs generated in the presence of IL-21, which were transferred in these latter three patients and the patient with MRD, all remained detectable long term and maintained or acquired in vivo phenotypic and functional characteristics associated with long-lived memory CD8+ T cells. This study supports expanding efforts to immunologically target WT1 and provides insights into the requirements necessary to establish potent persistent T cell responses.


Human Gene Therapy | 2009

T Cell Receptor Gene Therapy for Cancer

Thomas M. Schmitt; Gunnar B. Ragnarsson; Philip D. Greenberg

T cell-based adoptive immunotherapy has been shown to be a promising treatment for various types of cancer. However, adoptive T cell therapy currently requires the custom isolation and characterization of tumor-specific T cells from each patient-a process that can be not only difficult and time-consuming but also often fails to yield high-avidity T cells, which together have limited the broad application of this approach as a clinical treatment. Employing T cell receptor (TCR) gene therapy as a component of adoptive T cell therapy strategies can overcome many of these obstacles, allowing autologous T cells with a defined specificity to be generated in a much shorter time period. Initial studies using this approach have been hampered by a number of technical difficulties resulting in low TCR expression and acquisition of potentially problematic specificities due to mispairing of introduced TCR chains with endogenous TCR chains. The last several years have seen substantial progress in our understanding of the multiple facets of TCR gene therapy that will have to be properly orchestrated for this strategy to succeed. Here we outline the challenges of TCR gene therapy and the advances that have been made toward realizing the promise of this approach.


Cancer Cell | 2015

T Cells Engineered against a Native Antigen Can Surmount Immunologic and Physical Barriers to Treat Pancreatic Ductal Adenocarcinoma

Ingunn M. Stromnes; Thomas M. Schmitt; Ayaka Hulbert; J. Scott Brockenbrough; Hieu Nguyen; Carlos Cuevas; Ashley M. Dotson; Xiaoxia Tan; Jennifer L. Hotes; Philip D. Greenberg; Sunil R. Hingorani

Pancreatic ductal adenocarcinomas (PDAs) erect physical barriers to chemotherapy and induce multiple mechanisms of immune suppression, creating a sanctuary for unimpeded growth. We tested the ability of T cells engineered to express an affinity-enhanced T cell receptor (TCR) against a native antigen to overcome these barriers in a genetically engineered model of autochthonous PDA. Engineered T cells preferentially accumulate in PDA and induce tumor cell death and stromal remodeling. However, tumor-infiltrating T cells become progressively dysfunctional, a limitation successfully overcome by serial T cell infusions that resulted in a near-doubling of survival without overt toxicities. Similarly engineered human T cells lyse PDA cells in vitro, further supporting clinical advancement of this TCR-based strategy for the treatment of PDA.


Immunological Reviews | 2014

Re‐adapting T cells for cancer therapy: from mouse models to clinical trials

Ingunn M. Stromnes; Thomas M. Schmitt; Aude G. Chapuis; Sunil R. Hingorani; Philip D. Greenberg

Adoptive T‐cell therapy involves the isolation, expansion, and reinfusion of T lymphocytes with a defined specificity and function as a means to eradicate cancer. Our research has focused on specifying the requirements for tumor eradication with antigen‐specific T cells and T cells transduced to express a defined T‐cell receptor (TCR) in mouse models and then translating these strategies to clinical trials. Our design of T‐cell‐based therapy for cancer has reflected efforts to identify the obstacles that limit sustained effector T‐cell activity in mice and humans, design approaches to enhance T‐cell persistence, develop methods to increase TCR affinity/T‐cell functional avidity, and pursue strategies to overcome tolerance and immunosuppression. With the advent of genetic engineering, a highly functional population of T cells can now be rapidly generated and tailored for the targeted malignancy. Preclinical studies in faithful and informative mouse models, in concert with knowledge gained from analyses of successes and limitations in clinical trials, are shaping how we continue to develop, refine, and broaden the applicability of this approach for cancer therapy.


Blood | 2013

Enhanced-affinity murine T-cell receptors for tumor/self-antigens can be safe in gene therapy despite surpassing the threshold for thymic selection

Thomas M. Schmitt; David H. Aggen; Ingunn M. Stromnes; Michelle L. Dossett; Sarah A. Richman; David M. Kranz; Philip D. Greenberg

Many of the most promising tumor antigens for T-cell-based cancer immunotherapies are unmodified self-antigens. Unfortunately, the avidity of T cells specific for these antigens is limited by central tolerance during T-cell development in the thymus, resulting in decreased anti-tumor efficacy of these T cells. One approach to overcoming this obstacle is to mutate T-cell receptor (TCR) genes from naturally occurring T cells to enhance the affinity for the target antigen. These enhanced-affinity TCRs can then be developed for use in TCR gene therapy. Although TCRs with significantly enhanced affinity have been generated using this approach, it is not clear whether these TCRs, which bypass the affinity limits imposed by negative selection, remain unresponsive to the low levels of self-antigen generally expressed by some normal tissues. Here we show that 2 variants of a high-affinity WT1-specific TCR with enhanced affinity for WT1 are safe and do not mediate autoimmune tissue infiltration or damage when transduced into peripheral CD8 T cells and transferred in vivo. However, if expressed in developing T cells and subjected to thymic selection, the same enhanced-affinity TCRs signal tolerance mechanisms in the thymus, resulting in T cells with attenuated antigen sensitivity in the periphery.


Blood | 2012

Cyclin-A1 represents a new immunogenic targetable antigen expressed in acute myeloid leukemia stem cells with characteristics of a cancer-testis antigen

Sebastian Ochsenreither; Ravindra Majeti; Thomas M. Schmitt; Derek L. Stirewalt; Ulrich Keilholz; Keith R. Loeb; Brent L. Wood; Yongiae E. Choi; Marie Bleakley; Edus H. Warren; Michael Hudecek; Yoshiki Akatsuka; Irving L. Weissman; Philip D. Greenberg

Targeted T-cell therapy is a potentially less toxic strategy than allogeneic stem cell transplantation for providing a cytotoxic antileukemic response to eliminate leukemic stem cells (LSCs) in acute myeloid leukemia (AML). However, this strategy requires identification of leukemia-associated antigens that are immunogenic and exhibit selective high expression in AML LSCs. Using microarray expression analysis of LSCs, hematopoietic cell subpopulations, and peripheral tissues to screen for candidate antigens, cyclin-A1 was identified as a candidate gene. Cyclin-A1 promotes cell proliferation and survival, has been shown to be leukemogenic in mice, is detected in LSCs of more than 50% of AML patients, and is minimally expressed in normal tissues with exception of testis. Using dendritic cells pulsed with a cyclin-A1 peptide library, we generated T cells against several cyclin-A1 oligopeptides. Two HLA A*0201-restricted epitopes were further characterized, and specific CD8 T-cell clones recognized both peptide-pulsed target cells and the HLA A*0201-positive AML line THP-1, which expresses cyclin-A1. Furthermore, cyclin-A1-specific CD8 T cells lysed primary AML cells. Thus, cyclin-A1 is the first prototypic leukemia-testis-antigen to be expressed in AML LSCs. The pro-oncogenic activity, high expression levels, and multitude of immunogenic epitopes make it a viable target for pursuing T cell-based therapy approaches.


Gene Therapy | 2012

Single-chain VαVβ T-cell receptors function without mispairing with endogenous TCR chains.

David H. Aggen; Adam S. Chervin; Thomas M. Schmitt; Boris Engels; Jennifer D. Stone; Sarah A. Richman; Brian M. Baker; Philip D. Greenberg; Hans Schreiber; David M. Kranz

Transduction of exogenous T-cell receptor (TCR) genes into patients’ activated peripheral blood T cells is a potent strategy to generate large numbers of specific T cells for adoptive therapy of cancer and viral diseases. However, the remarkable clinical promise of this powerful approach is still being overshadowed by a serious potential consequence: mispairing of the exogenous TCR chains with endogenous TCR chains. These ‘mixed’ heterodimers can generate new specificities that result in graft-versus-host reactions. Engineering TCR constant regions of the exogenous chains with a cysteine promotes proper pairing and reduces the mispairing, but, as we show here, does not eliminate the formation of mixed heterodimers. By contrast, deletion of the constant regions, through use of a stabilized Vα/Vβ single-chain TCR (scTv), avoided mispairing completely. By linking a high-affinity scTv to intracellular signaling domains, such as Lck and CD28, the scTv was capable of activating functional T-cell responses in the absence of either the CD3 subunits or the co-receptors, and circumvented mispairing with endogenous TCRs. Such transduced T cells can respond to the targeted antigen independent of CD3 subunits via the introduced scTv, without the transduced T cells acquiring any new undefined and potentially dangerous specificities.


Molecular Therapy | 2009

Adoptive Immunotherapy of Disseminated Leukemia With TCR-transduced, CD8+ T Cells Expressing a Known Endogenous TCR

Michelle L. Dossett; Ryan M. Teague; Thomas M. Schmitt; Xiaoxia Tan; Laurence J. N. Cooper; Cristina Pinzon; Philip D. Greenberg

Adoptive T-cell immunotherapy has shown promise in the treatment of human malignancies, but the challenge of isolating T cells with high avidity for tumor antigens in each patient has limited application of this approach. The transfer into T cells of T-cell receptor (TCR) genes encoding high-affinity TCRs recognizing defined tumor-associated antigens can potentially circumvent this obstacle. Using a well-characterized murine model of adoptive T-cell immunotherapy for widely disseminated leukemia, we demonstrate that TCR gene-modified T cells can cure mice of disseminated tumor. One goal of such adoptive therapy is to establish a persistent memory response to prevent recurrence; however, long-term function of transferred TCR-transduced T cells is limited due to reduced expression of the introduced TCR in vivo in quiescent resting T cells. However, by introducing the TCR into a cell with a known endogenous specificity, activation of these T cells by stimulation through the endogenous TCR can be used to increase expression of the introduced TCR, potentially providing a strategy to increase the total number of tumor-reactive T cells in the host and restore more potent antitumor activity.


Journal of Immunology | 2012

Abrogation of Src Homology Region 2 Domain-Containing Phosphatase 1 in Tumor-Specific T Cells Improves Efficacy of Adoptive Immunotherapy by Enhancing the Effector Function and Accumulation of Short-Lived Effector T Cells In Vivo

Ingunn M. Stromnes; Carla Fowler; Chanel C. Casamina; Christina M. Georgopolos; Megan S. McAfee; Thomas M. Schmitt; Xiaoxia Tan; Tae Don Kim; Inpyo Choi; Joseph N. Blattman; Philip D. Greenberg

T cell expression of inhibitory proteins can be a critical component for the regulation of immunopathology owing to self-reactivity or potentially exuberant responses to pathogens, but it may also limit T cell responses to some malignancies, particularly if the tumor Ag being targeted is a self-protein. We found that the abrogation of Src homology region 2 domain-containing phosphatase-1 (SHP-1) in tumor-reactive CD8+ T cells improves the therapeutic outcome of adoptive immunotherapy in a mouse model of disseminated leukemia, with benefit observed in therapy employing transfer of CD8+ T cells alone or in the context of also providing supplemental IL-2. SHP-1−/− and SHP-1+/+ effector T cells were expanded in vitro for immunotherapy. Following transfer in vivo, the SHP-1−/− effector T cells exhibited enhanced short-term accumulation, followed by greater contraction, and they ultimately formed similar numbers of long-lived, functional memory cells. The increased therapeutic effectiveness of SHP-1−/− effector cells was also observed in recipients that expressed the tumor Ag as a self-antigen in the liver, without evidence of inducing autoimmune toxicity. SHP-1−/− effector CD8+ T cells expressed higher levels of eomesodermin, which correlated with enhanced lysis of tumor cells. Furthermore, reduction of SHP-1 expression in tumor-reactive effector T cells by retroviral transduction with vectors that express SHP-1–specific small interfering RNA, a translatable strategy, also exhibited enhanced antitumor activity in vivo. These studies suggest that abrogating SHP-1 in effector T cells may improve the efficacy of tumor elimination by T cell therapy without affecting the ability of the effector cells to persist and provide a long-term response.

Collaboration


Dive into the Thomas M. Schmitt's collaboration.

Top Co-Authors

Avatar

Philip D. Greenberg

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Aude G. Chapuis

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Ingunn M. Stromnes

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Hieu Nguyen

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Daniel Hunter

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Felecia Wagener

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Sunil R. Hingorani

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Gunnar B. Ragnarsson

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Xiaoxia Tan

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Cassian Yee

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge