Network


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

Hotspot


Dive into the research topics where Bent K. Jakobsen is active.

Publication


Featured researches published by Bent K. Jakobsen.


Blood | 2013

Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma

Gerald P. Linette; Edward A. Stadtmauer; Marcela V. Maus; Aaron P. Rapoport; Bruce L. Levine; Lyndsey Emery; Leslie A. Litzky; Adam Bagg; Beatriz M. Carreno; Patrick J. Cimino; Gwendolyn Binder-Scholl; Dominic P. Smethurst; Andrew B. Gerry; Nick Pumphrey; Alan D. Bennett; Joanna E. Brewer; Joseph Dukes; Jane Harper; Helen K. Tayton-Martin; Bent K. Jakobsen; Namir J. Hassan; Michael Kalos; Carl H. June

An obstacle to cancer immunotherapy has been that the affinity of T-cell receptors (TCRs) for antigens expressed in tumors is generally low. We initiated clinical testing of engineered T cells expressing an affinity-enhanced TCR against HLA-A*01-restricted MAGE-A3. Open-label protocols to test the TCRs for patients with myeloma and melanoma were initiated. The first two treated patients developed cardiogenic shock and died within a few days of T-cell infusion, events not predicted by preclinical studies of the high-affinity TCRs. Gross findings at autopsy revealed severe myocardial damage, and histopathological analysis revealed T-cell infiltration. No MAGE-A3 expression was detected in heart autopsy tissues. Robust proliferation of the engineered T cells in vivo was documented in both patients. A beating cardiomyocyte culture generated from induced pluripotent stem cells triggered T-cell killing, which was due to recognition of an unrelated peptide derived from the striated muscle-specific protein titin. These patients demonstrate that TCR-engineered T cells can have serious and not readily predictable off-target and organ-specific toxicities and highlight the need for improved methods to define the specificity of engineered TCRs.


Nature Biotechnology | 2005

Directed evolution of human T-cell receptors with picomolar affinities by phage display

Yi Li; Ruth Moysey; Peter Eamon Molloy; Annelise Vuidepot; Tara Mahon; Emma Baston; Steven M. Dunn; Nathaniel Liddy; Jansen P. Jacob; Bent K. Jakobsen; Jonathan M. Boulter

Peptides derived from almost all proteins, including disease-associated proteins, can be presented on the cell surface as peptide–human leukocyte antigen (pHLA) complexes. T cells specifically recognize pHLA with their clonally rearranged T-cell receptors (TCRs), whose natural affinities are limited to ∼1–100 μM. Here we describe the display of ten different human TCRs on the surface of bacteriophage, stabilized by a nonnative interchain disulfide bond. We report the directed evolution of high-affinity TCRs specific for two different pHLAs: the human T-cell lymphotropic virus type 1 (HTLV-1) tax11–19 peptide–HLA-A*0201 complex and the NY-ESO-1157–165 tumor-associated peptide antigen–HLA-A*0201 complex, with affinities of up to 2.5 nM and 26 pM, respectively, and we demonstrate their high specificity and sensitivity for targeting of cell-surface pHLAs.


Immunity | 1999

TCR Binding to Peptide-MHC Stabilizes a Flexible Recognition Interface

Benjamin E. Willcox; George F. Gao; Jessica R. Wyer; John E. Ladbury; John I. Bell; Bent K. Jakobsen; P. Anton van der Merwe

The binding of TCRs to their peptide-MHC ligands is characterized by a low affinity, slow kinetics, and a high degree of cross-reactivity. Here, we report the results of a kinetic and thermodynamic analysis of two TCRs binding to their peptide-MHC ligands, which reveal two striking features. First, significant activation energy barriers must be overcome during both association and dissociation, suggesting that conformational adjustments are required. Second, the low affinity of binding is a consequence of highly unfavorable entropic effects, indicative of a substantial reduction in disorder upon binding. This is evidence that the TCR and/or peptide-MHC have flexible binding surfaces that are stabilized upon binding. Such conformational flexibility, which may also be a feature of primary antibodies, is likely to contribute to cross-reactivity in antigen recognition.


Science Translational Medicine | 2013

Identification of a Titin-Derived HLA-A1–Presented Peptide as a Cross-Reactive Target for Engineered MAGE A3–Directed T Cells

Brian J. Cameron; Andrew B. Gerry; Joseph Dukes; Jane Harper; Vivekanandan Kannan; Frayne Bianchi; Francis Grand; Joanna E. Brewer; Minnal Gupta; Gabriela Plesa; Giovanna Bossi; Annelise Vuidepot; Alex Powlesland; Alison Legg; Katherine J. Adams; Alan D. Bennett; Nicholas J. Pumphrey; Daniel Williams; Gwendolyn Binder-Scholl; Irina Kulikovskaya; Bruce L. Levine; James L. Riley; Angel Varela-Rohena; Edward A. Stadtmauer; Aaron P. Rapoport; Gerald P. Linette; Carl H. June; Namir J. Hassan; Michael Kalos; Bent K. Jakobsen

T cells engineered to express affinity-enhanced TCRs directed to a MAGE A3 peptide cross-react with a similar, but unrelated, self-peptide. Cross-Reactive Adoptive Therapy Engineering T cells with enhanced affinity to cancer targets is a promising therapy. However, one key bottleneck in this strategy is the identification of targets that are expressed on cancer cells but not on normal healthy tissue. One way to identify these antigens is by looking at the family of cancer-testis antigens, which have restricted expression in normal tissue but are frequently up-regulated in tumors. Cameron et al. now report that a T cell engineered to target one such antigen—MAGE A3—cross-reacts with a peptide from a muscle protein, Titin. The authors developed a T cell that targeted a MAGE A3 antigen for use in adoptive immunotherapy. Although extensive preclinical investigations demonstrated no off-target antigen recognition, patients who received these T cells had serious adverse events, including fatal cardiac toxicity. The authors then used amino acid scanning to search for potential cross-reactivity of these T cells with an off-target peptide and identified a peptide derived from the muscle protein Titin. Because affinity-enhanced T cells are highly potent, this cross-reactivity was likely the cause of the off-target toxicity. This study highlights methods that may be used to prevent cross-reactivity in future trials of adoptive immunotherapy. MAGE A3, which belongs to the family of cancer-testis antigens, is an attractive target for adoptive therapy given its reactivation in various tumors and limited expression in normal tissues. We developed an affinity-enhanced T cell receptor (TCR) directed to a human leukocyte antigen (HLA)–A*01–restricted MAGE A3 antigen (EVDPIGHLY) for use in adoptive therapy. Extensive preclinical investigations revealed no off-target antigen recognition concerns; nonetheless, administration to patients of T cells expressing the affinity-enhanced MAGE A3 TCR resulted in a serious adverse event (SAE) and fatal toxicity against cardiac tissue. We present a description of the preclinical in vitro functional analysis of the MAGE A3 TCR, which failed to reveal any evidence of off-target activity, and a full analysis of the post-SAE in vitro investigations, which reveal cross-recognition of an off-target peptide. Using an amino acid scanning approach, a peptide from the muscle protein Titin (ESDPIVAQY) was identified as an alternative target for the MAGE A3 TCR and the most likely cause of in vivo toxicity. These results demonstrate that affinity-enhanced TCRs have considerable effector functions in vivo and highlight the potential safety concerns for TCR-engineered T cells. Strategies such as peptide scanning and the use of more complex cell cultures are recommended in preclinical studies to mitigate the risk of off-target toxicity in future clinical investigations.


Nature Medicine | 2015

NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma

Aaron P. Rapoport; Edward A. Stadtmauer; Gwendolyn Binder-Scholl; Olga Goloubeva; Dan T. Vogl; Simon F. Lacey; Ashraf Badros; Alfred L. Garfall; Brendan M. Weiss; Jeffrey Finklestein; Irina Kulikovskaya; Sanjoy K. Sinha; Shari Kronsberg; Minnal Gupta; Sarah Bond; Luca Melchiori; Joanna E. Brewer; Alan D. Bennett; Andrew B. Gerry; Nicholas J. Pumphrey; Daniel Williams; Helen K. Tayton-Martin; Lilliam Ribeiro; Tom Holdich; Saul Yanovich; Nancy M. Hardy; Jean Yared; Naseem Kerr; Sunita Philip; Sandra Westphal

Despite recent therapeutic advances, multiple myeloma (MM) remains largely incurable. Here we report results of a phase I/II trial to evaluate the safety and activity of autologous T cells engineered to express an affinity-enhanced T cell receptor (TCR) recognizing a naturally processed peptide shared by the cancer-testis antigens NY-ESO-1 and LAGE-1. Twenty patients with antigen-positive MM received an average 2.4 × 109 engineered T cells 2 d after autologous stem cell transplant. Infusions were well tolerated without clinically apparent cytokine-release syndrome, despite high IL-6 levels. Engineered T cells expanded, persisted, trafficked to marrow and exhibited a cytotoxic phenotype. Persistence of engineered T cells in blood was inversely associated with NY-ESO-1 levels in the marrow. Disease progression was associated with loss of T cell persistence or antigen escape, in accordance with the expected mechanism of action of the transferred T cells. Encouraging clinical responses were observed in 16 of 20 patients (80%) with advanced disease, with a median progression-free survival of 19.1 months. NY-ESO-1–LAGE-1 TCR–engineered T cells were safe, trafficked to marrow and showed extended persistence that correlated with clinical activity against antigen-positive myeloma.


Nature Medicine | 2008

Control of HIV-1 immune escape by CD8 T-cells expressing enhanced T-cell receptor

Angel Varela-Rohena; Peter Eamon Molloy; Steven M. Dunn; Yi Li; Richard G. Carroll; Anita Milicic; Tara Mahon; Deborah H. Sutton; Bruno Laugel; Ruth Moysey; Brian J. Cameron; Annelise Vuidepot; Marco E. Purbhoo; David K. Cole; Rodney E. Phillips; Carl H. June; Bent K. Jakobsen; Andrew K. Sewell; James L. Riley

HIVs considerable capacity to vary its HLA-I-restricted peptide antigens allows it to escape from host cytotoxic T lymphocytes (CTLs). Nevertheless, therapeutics able to target HLA-I-associated antigens, with specificity for the spectrum of preferred CTL escape mutants, could prove effective. Here we use phage display to isolate and enhance a T-cell antigen receptor (TCR) originating from a CTL line derived from an infected person and specific for the immunodominant HLA-A*02-restricted, HIVgag-specific peptide SLYNTVATL (SL9). High-affinity (KD < 400 pM) TCRs were produced that bound with a half-life in excess of 2.5 h, retained specificity, targeted HIV-infected cells and recognized all common escape variants of this epitope. CD8 T cells transduced with this supraphysiologic TCR produced a greater range of soluble factors and more interleukin-2 than those transduced with natural SL9-specific TCR, and they effectively controlled wild-type and mutant strains of HIV at effector-to-target ratios that could be achieved by T-cell therapy.


Immunity | 1999

T Cell Receptor and Coreceptor CD8αα Bind Peptide-MHC Independently and with Distinct Kinetics

Jessica R. Wyer; Benjamin E. Willcox; George F. Gao; Ulrich C. Gerth; Simon J. Davis; John I. Bell; P. Anton van der Merwe; Bent K. Jakobsen

The T cell surface glycoprotein CD8 enhances T cell antigen recognition by binding to MHC class I molecules. We show that human CD8 alphaalpha binds to the MHC class I molecule HLA-A2 with an extremely low affinity (Kd approximately 0.2 mM at 37 degrees C) and with kinetics that are between 2 and 3 orders of magnitude faster than reported for T cell receptor/peptide-MHC interactions. Furthermore, CD8 alphaalpha had no detectable effect on a T cell receptor (TCR) binding to the same peptide-MHC class I complex. These binding properties provide an explanation as to why the CD8/MHC class I interaction is unable to initiate cell-cell adhesion and how it can enhance TCR recognition without interfering with its specificity.


Journal of Experimental Medicine | 2005

Structural and Kinetic Basis for Heightened Immunogenicity of T Cell Vaccines

Ji-Li Chen; Guillaume Stewart-Jones; Giovanna Bossi; Nikolai Lissin; Linda Wooldridge; Ed Man-Lik Choi; Gerhard Held; P. Rod Dunbar; Robert M. Esnouf; Malkit Sami; Jonathan M. Boulter; Pierre J. Rizkallah; Christoph Renner; R. Andrew Sewell; P. Anton van der Merwe; Bent K. Jakobsen; Gillian M. Griffiths; E. Yvonne Jones; Vincenzo Cerundolo

Analogue peptides with enhanced binding affinity to major histocompatibility class (MHC) I molecules are currently being used in cancer patients to elicit stronger T cell responses. However, it remains unclear as to how alterations of anchor residues may affect T cell receptor (TCR) recognition. We correlate functional, thermodynamic, and structural parameters of TCR–peptide–MHC binding and demonstrate the effect of anchor residue modifications of the human histocompatibility leukocyte antigens (HLA)–A2 tumor epitope NY–ESO-1157–165–SLLMWITQC on TCR recognition. The crystal structure of the wild-type peptide complexed with a specific TCR shows that TCR binding centers on two prominent, sequential, peptide sidechains, methionine–tryptophan. Cysteine-to-valine substitution at peptide position 9, while optimizing peptide binding to the MHC, repositions the peptide main chain and generates subtly enhanced interactions between the analogue peptide and the TCR. Binding analyses confirm tighter binding of the analogue peptide to HLA–A2 and improved soluble TCR binding. Recognition of analogue peptide stimulates faster polarization of lytic granules to the immunological synapse, reduces dependence on CD8 binding, and induces greater numbers of cross-reactive cytotoxic T lymphocyte to SLLMWITQC. These results provide important insights into heightened immunogenicity of analogue peptides and highlight the importance of incorporating structural data into the process of rational optimization of superagonist peptides for clinical trials.


Journal of Immunology | 2008

Single and Dual Amino Acid Substitutions in TCR CDRs Can Enhance Antigen-Specific T Cell Functions

Paul F. Robbins; Yong F. Li; Mona El-Gamil; Yangbing Zhao; Jennifer A. Wargo; Zhili Zheng; Hui Xu; Richard A. Morgan; Steven A. Feldman; Laura A. Johnson; Alan D. Bennett; Steven M. Dunn; Tara Mahon; Bent K. Jakobsen; Steven A. Rosenberg

Single and dual amino acid substitution variants were generated in the TCR CDRs of three TCRs that recognize tumor-associated Ags. Substitutions that enhance the reactivity of TCR gene-modified T cells to the cognate Ag complex were identified using a rapid RNA-based transfection system. The screening of a panel of variants of the 1G4 TCR, that recognizes a peptide corresponding to amino acid residues 157–165 of the human cancer testis Ag NY-ESO-1 (SLLMWITQC) in the context of the HLA-A*02 class I allele, resulted in the identification of single and dual CDR3α and CDR2β amino acid substitutions that dramatically enhanced the specific recognition of NY-ESO-1+/HLA-A*02+ tumor cell lines by TCR gene-modified CD4+ T cells. Within this group of improved TCRs, a dual substitution in the 1G4 TCR CDR3α chain was identified that enhanced Ag-specific reactivity in gene-modified CD4+ and CD8+ T cells. Separate experiments on two distinct TCRs that recognize the MART-1 27–35 (AAGIGILTV) peptide/HLA-A*02 Ag complex characterized single amino acid substitutions in both TCRs that enhanced CD4+ T cell Ag-specific reactivity. These results indicate that simple TCR substitution variants that enhance T cell function can be identified by rapid transfection and assay techniques, providing the means for generating potent Ag complex-specific TCR genes for use in the study of T cell interactions and in T cell adoptive immunotherapy.


Molecular Cell | 1998

Structural features impose tight peptide binding specificity in the nonclassical MHC molecule HLA-E.

Christopher A. O'Callaghan; José Tormo; Benjamin E. Willcox; Veronique M. Braud; Bent K. Jakobsen; David I. Stuart; Andrew J. McMichael; John I. Bell; E. Yvonne Jones

The crystal structure of the nonclassical human class lb MHC molecule HLA-E has been determined in complex with a prototypic ligand, the nonamer peptide (VMAPRTVLL), derived from the highly conserved residues 3-11 of the human MHC class la leader sequence. The mode of peptide binding retains some of the standard features observed in MHC class la complexes, but novel features imply that HLA-E has evolved to mediate specific binding to a tightly defined set of almost identical hydrophobic peptides from the highly conserved class l leader sequences. These molecular adaptations make HLA-E a rigorous checkpoint at the cell surface reporting on the integrity of the antigen processing pathway to CD94/NKG2 receptor-bearing natural killer cells.

Collaboration


Dive into the Bent K. Jakobsen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yi Li

University of Leicester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carl H. June

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tara Mahon

Imperial College London

View shared research outputs
Top Co-Authors

Avatar

George F. Gao

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge