Network


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

Hotspot


Dive into the research topics where Timothy T. Spear is active.

Publication


Featured researches published by Timothy T. Spear.


Cancer Immunology, Immunotherapy | 2016

TCR gene-modified T cells can efficiently treat established hepatitis C-associated hepatocellular carcinoma tumors

Timothy T. Spear; Glenda G. Callender; Jeffrey J. Roszkowski; Kelly Moxley; Patricia Simms; Kendra C. Foley; David C. Murray; Gina Scurti; Mingli Li; Justin T. Thomas; Alexander Langerman; Elizabeth Garrett-Mayer; Yi Zhang; Michael I. Nishimura

Abstract The success in recent clinical trials using T cell receptor (TCR)-genetically engineered T cells to treat melanoma has encouraged the use of this approach toward other malignancies and viral infections. Although hepatitis C virus (HCV) infection is being treated with a new set of successful direct anti-viral agents, potential for virologic breakthrough or relapse by immune escape variants remains. Additionally, many HCV+ patients have HCV-associated disease, including hepatocellular carcinoma (HCC), which does not respond to these novel drugs. Further exploration of other approaches to address HCV infection and its associated disease are highly warranted. Here, we demonstrate the therapeutic potential of PBL-derived T cells genetically engineered with a high-affinity, HLA-A2-restricted, HCV NS3:1406-1415-reactive TCR. HCV1406 TCR-transduced T cells can recognize naturally processed antigen and elicit CD8-independent recognition of both peptide-loaded targets and HCV+ human HCC cell lines. Furthermore, these cells can mediate regression of established HCV+ HCC in vivo. Our results suggest that HCV TCR-engineered antigen-reactive T cells may be a plausible immunotherapy option to treat HCV-associated malignancies, such as HCC.


Cancer Immunology, Immunotherapy | 2016

Strategies to genetically engineer T cells for cancer immunotherapy

Timothy T. Spear; Kaoru Nagato; Michael I. Nishimura

Immunotherapy is one of the most promising and innovative approaches to treat cancer, viral infections, and other immune-modulated diseases. Adoptive immunotherapy using gene-modified T cells is an exciting and rapidly evolving field. Exploiting knowledge of basic T cell biology and immune cell receptor function has fostered innovative approaches to modify immune cell function. Highly translatable clinical technologies have been developed to redirect T cell specificity by introducing designed receptors. The ability to engineer T cells to manifest desired phenotypes and functions is now a thrilling reality. In this review, we focus on outlining different varieties of genetically engineered T cells, their respective advantages and disadvantages as tools for immunotherapy, and their promise and drawbacks in the clinic.


Proceedings of the National Academy of Sciences of the United States of America | 2016

How structural adaptability exists alongside HLA-A2 bias in the human αβ TCR repertoire

Sydney J. Blevins; Brian G. Pierce; Nishant K. Singh; Timothy P. Riley; Yuan Wang; Timothy T. Spear; Michael I. Nishimura; Zhiping Weng; Brian M. Baker

Significance T-cell receptor (TCR) recognition of antigenic peptides presented by major histocompatibility complex (MHC) proteins defines specificity in cellular immunity. Evidence suggests that TCRs are intrinsically biased toward MHC proteins, yet how this bias coexists alongside the considerable structural variability that is necessary for TCRs to engage different ligands has been a longstanding puzzle. By examining structural and sequence data, we found evidence that human αβ TCRs have an inherent compatibility with structural and chemical properties of MHC proteins. This compatibility leads TCRs to an intrinsic MHC bias but does not compel the formation of particular modes of binding, providing a solution to how TCRs can be MHC-biased but still structurally adaptable. How T-cell receptors (TCRs) can be intrinsically biased toward MHC proteins while simultaneously display the structural adaptability required to engage diverse ligands remains a controversial puzzle. We addressed this by examining αβ TCR sequences and structures for evidence of physicochemical compatibility with MHC proteins. We found that human TCRs are enriched in the capacity to engage a polymorphic, positively charged “hot-spot” region that is almost exclusive to the α1-helix of the common human class I MHC protein, HLA-A*0201 (HLA-A2). TCR binding necessitates hot-spot burial, yielding high energetic penalties that must be offset via complementary electrostatic interactions. Enrichment of negative charges in TCR binding loops, particularly the germ-line loops encoded by the TCR Vα and Vβ genes, provides this capacity and is correlated with restricted positioning of TCRs over HLA-A2. Notably, this enrichment is absent from antibody genes. The data suggest a built-in TCR compatibility with HLA-A2 that biases receptors toward, but does not compel, particular binding modes. Our findings provide an instructional example for how structurally pliant MHC biases can be encoded within TCRs.


Journal of Leukocyte Biology | 2016

Hepatitis C virus‐cross‐reactive TCR gene‐modified T cells: a model for immunotherapy against diseases with genomic instability

Timothy T. Spear; Timothy P. Riley; Gretchen E. Lyons; Glenda G. Callender; Jeffrey J. Roszkowski; Yuan Wang; Patricia Simms; Gina Scurti; Kendra C. Foley; David C. Murray; Lance M. Hellman; Rachel H. McMahan; Makio Iwashima; Elizabeth Garrett-Mayer; Hugo R. Rosen; Brian M. Baker; Michael I. Nishimura

A major obstacle hindering the development of effective immunity against viral infections, their associated disease, and certain cancers is their inherent genomic instability. Accumulation of mutations can alter processing and presentation of antigens recognized by antibodies and T cells that can lead to immune escape variants. Use of an agent that can intrinsically combat rapidly mutating viral or cancer‐associated antigens would be quite advantageous in developing effective immunity against such disease. We propose that T cells harboring cross‐reactive TCRs could serve as a therapeutic agent in these instances. With the use of hepatitis C virus, known for its genomic instability as a model for mutated antigen recognition, we demonstrate cross‐reactivity against immunogenic and mutagenic nonstructural protein 3:1406‐1415 and nonstructural protein 3:1073‐1081 epitopes in PBL‐derived, TCR‐gene‐modified T cells. These single TCR‐engineered T cells can CD8‐independently recognize naturally occurring and epidemiologically relevant mutant variants. TCR‐peptide MHC modeling data allow us to rationalize how TCR structural properties accommodate recognition of certain mutated epitopes and how these substitutions impact the requirement of CD8 affinity enhancement for recognition. A better understanding of such TCRs’ promiscuous behavior may allow for exploitation of these properties to develop novel, adoptive T cell‐based therapies for viral infections and cancers exhibiting similar genomic instability.


Proceedings of the National Academy of Sciences of the United States of America | 2017

How an alloreactive T-cell receptor achieves peptide and MHC specificity

Yuan Wang; Nishant K. Singh; Timothy T. Spear; Lance M. Hellman; Kurt H. Piepenbrink; Rachel H. McMahan; Hugo R. Rosen; Craig W. Vander Kooi; Michael I. Nishimura; Brian M. Baker

Significance T-cell alloreactivity drives transplant rejection. Alloreactive recognition is believed to proceed with limited specificity, accounting for the high numbers of alloreactive T cells in humans. Paradoxically, however, many T cells recognize alloantigens with high specificity, and receptors from such T cells are being explored for use in cancer immunotherapy. Here, we explain how a T-cell receptor (TCR) achieves high specificity toward a peptide antigen presented by allo-major histocompatibility complex (MHC). Counter to prevailing theories of alloreactivity, we find that TCR recognition is driven by a cooperative interplay between features unique to both the allo-MHC and the peptide, such that binding is both MHC- and peptide-centric. Our results have broad implications for the determinants of immune recognition and efforts in immunotherapy. T-cell receptor (TCR) allorecognition is often presumed to be relatively nonspecific, attributable to either a TCR focus on exposed major histocompatibility complex (MHC) polymorphisms or the degenerate recognition of allopeptides. However, paradoxically, alloreactivity can proceed with high peptide and MHC specificity. Although the underlying mechanisms remain unclear, the existence of highly specific alloreactive TCRs has led to their use as immunotherapeutics that can circumvent central tolerance and limit graft-versus-host disease. Here, we show how an alloreactive TCR achieves peptide and MHC specificity. The HCV1406 TCR was cloned from T cells that expanded when a hepatitis C virus (HCV)-infected HLA-A2− individual received an HLA-A2+ liver allograft. HCV1406 was subsequently shown to recognize the HCV nonstructural protein 3 (NS3):1406–1415 epitope with high specificity when presented by HLA-A2. We show that NS3/HLA-A2 recognition by the HCV1406 TCR is critically dependent on features unique to both the allo-MHC and the NS3 epitope. We also find cooperativity between structural mimicry and a crucial peptide “hot spot” and demonstrate its role, along with the MHC, in directing the specificity of allorecognition. Our results help explain the paradox of specificity in alloreactive TCRs and have implications for their use in immunotherapy and related efforts to manipulate TCR recognition, as well as alloreactivity in general.


Journal of Immunological Methods | 2016

Differential scanning fluorimetry based assessments of the thermal and kinetic stability of peptide-MHC complexes.

Lance M. Hellman; Liusong Yin; Yuan Wang; Sydney J. Blevins; Timothy P. Riley; Orrin S. Belden; Timothy T. Spear; Michael I. Nishimura; Lawrence J. Stern; Brian M. Baker

Measurements of thermal stability by circular dichroism (CD) spectroscopy have been widely used to assess the binding of peptides to MHC proteins, particularly within the structural immunology community. Although thermal stability assays offer advantages over other approaches such as IC50 measurements, CD-based stability measurements are hindered by large sample requirements and low throughput. Here we demonstrate that an alternative approach based on differential scanning fluorimetry (DSF) yields results comparable to those based on CD for both class I and class II complexes. As they require much less sample, DSF-based measurements reduce demands on protein production strategies and are amenable for high throughput studies. DSF can thus not only replace CD as a means to assess peptide/MHC thermal stability, but can complement other peptide-MHC binding assays used in screening, epitope discovery, and vaccine design. Due to the physical process probed, DSF can also uncover complexities not observed with other techniques. Lastly, we show that DSF can also be used to assess peptide/MHC kinetic stability, allowing for a single experimental setup to probe both binding equilibria and kinetics.


Journal of Leukocyte Biology | 2017

Comparative exploration of multidimensional flow cytometry software: a model approach evaluating T cell polyfunctional behavior

Timothy T. Spear; Michael I. Nishimura; Patricia Simms

Advancement in flow cytometry reagents and instrumentation has allowed for simultaneous analysis of large numbers of lineage/functional immune cell markers. Highly complex datasets generated by polychromatic flow cytometry require proper analytical software to answer investigators’ questions. A problem among many investigators and flow cytometry Shared Resource Laboratories (SRLs), including our own, is a lack of access to a flow cytometry‐knowledgeable bioinformatics team, making it difficult to learn and choose appropriate analysis tool(s). Here, we comparatively assess various multidimensional flow cytometry software packages for their ability to answer a specific biologic question and provide graphical representation output suitable for publication, as well as their ease of use and cost. We assessed polyfunctional potential of TCR‐transduced T cells, serving as a model evaluation, using multidimensional flow cytometry to analyze 6 intracellular cytokines and degranulation on a per‐cell basis. Analysis of 7 parameters resulted in 128 possible combinations of positivity/negativity, far too complex for basic flow cytometry software to analyze fully. Various software packages were used, analysis methods used in each described, and representative output displayed. Of the tools investigated, automated classification of cellular expression by nonlinear stochastic embedding (ACCENSE) and coupled analysis in Pestle/simplified presentation of incredibly complex evaluations (SPICE) provided the most user‐friendly manipulations and readable output, evaluating effects of altered antigen‐specific stimulation on T cell polyfunctionality. This detailed approach may serve as a model for other investigators/SRLs in selecting the most appropriate software to analyze complex flow cytometry datasets. Further development and awareness of available tools will help guide proper data analysis to answer difficult biologic questions arising from incredibly complex datasets.


Cancer Immunology, Immunotherapy | 2017

Critical biological parameters modulate affinity as a determinant of function in T-cell receptor gene-modified T-cells

Timothy T. Spear; Yuan Wang; Kendra C. Foley; David C. Murray; Gina Scurti; Patricia Simms; Elizabeth Garrett-Mayer; Lance M. Hellman; Brian M. Baker; Michael I. Nishimura

T-cell receptor (TCR)-pMHC affinity has been generally accepted to be the most important factor dictating antigen recognition in gene-modified T-cells. As such, there is great interest in optimizing TCR-based immunotherapies by enhancing TCR affinity to augment the therapeutic benefit of TCR gene-modified T-cells in cancer patients. However, recent clinical trials using affinity-enhanced TCRs in adoptive cell transfer (ACT) have observed unintended and serious adverse events, including death, attributed to unpredicted off-tumor or off-target cross-reactivity. It is critical to re-evaluate the importance of other biophysical, structural, or cellular factors that drive the reactivity of TCR gene-modified T-cells. Using a model for altered antigen recognition, we determined how TCR–pMHC affinity influenced the reactivity of hepatitis C virus (HCV) TCR gene-modified T-cells against a panel of naturally occurring HCV peptides and HCV-expressing tumor targets. The impact of other factors, such as TCR–pMHC stabilization and signaling contributions by the CD8 co-receptor, as well as antigen and TCR density were also evaluated. We found that changes in TCR–pMHC affinity did not always predict or dictate IFNγ release or degranulation by TCR gene-modified T-cells, suggesting that less emphasis might need to be placed on TCR–pMHC affinity as a means of predicting or augmenting the therapeutic potential of TCR gene-modified T-cells used in ACT. A more complete understanding of antigen recognition by gene-modified T-cells and a more rational approach to improve the design and implementation of novel TCR-based immunotherapies is necessary to enhance efficacy and maximize safety in patients.


Molecular Therapy - Oncolytics | 2017

HCV T Cell Receptor Chain Modifications to Enhance Expression, Pairing, and Antigen Recognition in T Cells for Adoptive Transfer

Kendra C. Foley; Timothy T. Spear; David C. Murray; Kaoru Nagato; Elizabeth Garrett-Mayer; Michael I. Nishimura

T cell receptor (TCR)-gene-modified T cells for adoptive cell transfer can mediate objective clinical responses in melanoma and other malignancies. When introducing a second TCR, mispairing between the endogenous and introduced α and β TCR chains limits expression of the introduced TCR, which can result in impaired efficacy or off-target reactivity and autoimmunity. One approach to promote proper TCR chain pairing involves modifications of the introduced TCR genes: introducing a disulfide bridge, substituting murine for human constant regions, codon optimization, TCR chain leucine zipper fusions, and a single-chain TCR. We have introduced these modifications into our hepatitis C virus (HCV) reactive TCR and utilize a marker gene, CD34t, which allows us to directly compare transduction efficiency with TCR expression and T cell function. Our results reveal that of the TCRs tested, T cells expressing the murine Cβ2 TCR or leucine zipper TCR have the highest levels of expression and the highest percentage of lytic and interferon-γ (IFN-γ)-producing T cells. Our studies give us a better understanding of how TCR modifications impact TCR expression and T cell function that may allow for optimization of TCR-modified T cells for adoptive cell transfer to treat patients with malignancies.


Journal of Leukocyte Biology | 2018

TCR modifications that enhance chain pairing in gene‐modified T cells can augment cross‐reactivity and alleviate CD8 dependence

Timothy T. Spear; Kendra C. Foley; Elizabeth Garrett-Mayer; Michael I. Nishimura

T cell receptor (TCR) gene‐modified T cells are a promising immunotherapy but require refinement to improve clinical responses and limit off‐target toxicities. A variety of TCR and gene‐delivery vector modifications have been developed to enhance introduced TCR expression and limit introduced/endogenous TCR chain mispairing, improving target antigen recognition and minimizing mispairing‐induced cross‐reactivity. Using our well‐characterized HCV1406 TCR, we previously compared the impact of various chain pairing enhancing modifications on TCR expression and cognate antigen recognition. HCV1406 TCR is also natively cross‐reactive against naturally occurring altered peptide ligands (APLs), which was shown to be dependent on high TCR surface density. In this report, we observed in a Jurkat model that absent TCR chain pairing competition alleviated CD8‐dependent APL recognition and induced novel cross‐reactivity of HCV1406 TCR. We then compared chain pairing enhancing modifications’ effects on TCR cross‐reactivity in Jurkat and T cells, showing C‐terminal leucine zippers and constant region murinization alleviated CD8 dependence and induced novel APL recognition. While modifications enhancing TCR chain pairing intend to avoid cross‐reactivity by limiting mispairing with the endogenous TCR, these data suggest they may also enhance natural cross‐reactivity and reduce dependence on CD8. These observations have significant implications on the design/implementation of TCR gene‐modified T cells.

Collaboration


Dive into the Timothy T. Spear's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elizabeth Garrett-Mayer

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Brian M. Baker

University of Notre Dame

View shared research outputs
Top Co-Authors

Avatar

Yuan Wang

University of Notre Dame

View shared research outputs
Top Co-Authors

Avatar

Kendra C. Foley

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar

Patricia Simms

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David C. Murray

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar

Gina Scurti

Loyola University Chicago

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge