Catalogue Search | MBRL
Search Results Heading
Explore the vast range of titles available.
MBRLSearchResults
-
DisciplineDiscipline
-
Is Peer ReviewedIs Peer Reviewed
-
Item TypeItem Type
-
SubjectSubject
-
YearFrom:-To:
-
More FiltersMore FiltersSourceLanguage
Done
Filters
Reset
41
result(s) for
"Sheppard, Neil C"
Sort by:
Pretreatment with IL-15 and IL-18 rescues natural killer cells from granzyme B-mediated apoptosis after cryopreservation
2024
Human natural killer (NK) cell-based therapies are under assessment for treating various cancers, but cryopreservation reduces both the recovery and function of NK cells, thereby limiting their therapeutic feasibility. Using cryopreservation protocols optimized for T cells, here we find that ~75% of NK cells die within 24 h post-thaw, with the remaining cells displaying reduced cytotoxicity. Using CRISPR-Cas9 gene editing and confocal microscopy, we find that cryopreserved NK cells largely die via apoptosis initiated by leakage of granzyme B from cytotoxic vesicles. Pretreatment of NK cells with a combination of Interleukins-15 (IL-15) and IL-18 prior to cryopreservation improves NK cell recovery to ~90-100% and enables equal tumour control in a xenograft model of disseminated Raji cell lymphoma compared to non-cryopreserved NK cells. The mechanism of IL-15 and IL-18-induced protection incorporates two mechanisms: a transient reduction in intracellular granzyme B levels via degranulation, and the induction of antiapoptotic genes.
Natural killer (NK) cells are assessed for various therapies, but sub-optimal cryopreservation dampens their clinical feasibility. Here the authors show that pretreating human NK cells with IL-15/IL-18 prior to cryopreservation improves NK cell post-thaw viability and functions, potentially via anti-apoptosis gene induction and granzyme B degranulation.
Journal Article
ZFP36 disruption is insufficient to enhance the function of mesothelin-targeting human CAR-T cells
2024
Loss of inflammatory effector function, such as cytokine production and proliferation, is a fundamental driver of failure in T cell therapies against solid tumors. Here, we used CRISPR/Cas9 to genetically disrupt ZFP36, an RNA binding protein that regulates the stability of mRNAs involved in T cell inflammatory function, such as the cytokines IL2 and IFNγ, in human T cells engineered with a clinical-stage mesothelin-targeting CAR to determine whether its disruption could enhance antitumor responses. ZFP36 disruption slightly increased antigen-independent activation and cytokine responses but did not enhance overall performance in vitro or in vivo in a xenograft tumor model with NSG mice. While ZFP36 disruption does not reduce the function of CAR-T cells, these results suggest that singular disruption of ZFP36 is not sufficient to improve their function and may benefit from a multiplexed approach.
Journal Article
Genetic engineering of T cells for immunotherapy
by
Riley, James L
,
Ellis, Gavin I
,
Sheppard, Neil C
in
Acute lymphoblastic leukemia
,
Antigens
,
Autoimmune diseases
2021
Genetically engineered T cell immunotherapies have provided remarkable clinical success to treat B cell acute lymphoblastic leukaemia by harnessing a patient’s own T cells to kill cancer, and these approaches have the potential to provide therapeutic benefit for numerous other cancers, infectious diseases and autoimmunity. By introduction of either a transgenic T cell receptor or a chimeric antigen receptor, T cells can be programmed to target cancer cells. However, initial studies have made it clear that the field will need to implement more complex levels of genetic regulation of engineered T cells to ensure both safety and efficacy. Here, we review the principles by which our knowledge of genetics and genome engineering will drive the next generation of adoptive T cell therapies.This Review discusses strategies for the genetic engineering of adoptive T cell immunotherapies with a focus on approaches harnessing transgenic T cell receptors or chimeric antigen receptors to treat cancer. The authors also discuss the more complex levels of genetic regulation that will be needed to ensure both safety and efficacy.
Journal Article
Nanomaterials for T-cell cancer immunotherapy
2021
T-cell-based immunotherapies hold promise for the treatment of many types of cancer, with three approved products for B-cell malignancies and a large pipeline of treatments in clinical trials. However, there are several challenges to their broad implementation. These include insufficient expansion of adoptively transferred T cells, inefficient trafficking of T cells into solid tumours, decreased T-cell activity due to a hostile tumour microenvironment and the loss of target antigen expression. Together, these factors restrict the number of therapeutically active T cells engaging with tumours. Nanomaterials are uniquely suited to overcome these challenges, as they can be rationally designed to enhance T-cell expansion, navigate complex physical barriers and modulate tumour microenvironments. Here, we present an overview of nanomaterials that have been used to overcome clinical barriers to T-cell-based immunotherapies and provide our outlook of this emerging field at the interface of cancer immunotherapy and nanomaterial design.This Review discusses the progress and the remaining challenges for the clinical application of nanomaterial-based T-cell immunotherapies.
Journal Article
Swiss army knife T cell: one T cell many tumor targets
2024
Tumor cells often have mutated (neoantigens) or abnormally expressed proteins (tumor-associated antigens, TAAs).1,2 Antigen presentation is a process by which every nucleated cell in the body presents small samples of its proteins to the immune system in a context of peptide-loaded Human Leukocyte Antigen (HLA)3 (Fig. 1a). Dolton and colleagues studied T cells from MM909.24, a patient with stage IV melanoma who had successful TIL immunotherapy treatment and remains cancer free for a decade and counting.7 50% of expanded TIL cells from this patient were able to destroy the patient’s melanoma cell line in vitro. Using this database, 3 of the top 10 predicted peptides were recognized by the MEL8 TCR, and these came from 3 distinct TAAs: Melan A, Bone marrow stromal cell antigen 2 (BST2), and insulin-like growth factor 2 mRNA-binding protein 2 (IMP2) (Fig. 1d).
Journal Article
Human IL-6 fosters long-term engraftment of patient-derived disease-driving myeloma cells in immunodeficient mice
by
Susanibar-Adaniya, Sandra P.
,
Sheppard, Neil C.
,
Shultz, Leonard D.
in
Amyloid
,
Amyloidosis
,
Anemia
2024
Multiple myeloma is a largely incurable and life-threatening malignancy of antibody-secreting plasma cells. An effective and widely available animal model that recapitulates human myeloma and related plasma cell disorders is lacking. We show that busulfan-conditioned human IL-6-transgenic (hIL-6-transgenic) NSG (NSG+hIL6) mice reliably support the engraftment of malignant and premalignant human plasma cells, including from patients diagnosed with monoclonal gammopathy of undetermined significance, pre- and postrelapse myeloma, plasma cell leukemia, and amyloid light chain amyloidosis. Consistent with human disease, NSG+hIL6 mice engrafted with patient-derived myeloma cells developed serum M spikes, and a majority developed anemia, hypercalcemia, and/or bone lesions. Single-cell RNA sequencing showed nonmalignant and malignant cell engraftment, the latter expressing a wide array of mRNAs associated with myeloma cell survival and proliferation. Myeloma-engrafted mice given CAR T cells targeting plasma cells or bortezomib experienced reduced tumor burden. Our results establish NSG+hIL6 mice as an effective patient-derived xenograft model for study and preclinical drug development of multiple myeloma and related plasma cell disorders.
Journal Article
Responsive biomaterials: optimizing control of cancer immunotherapy
2024
Immunotherapy has emerged as an eminent and effective modality in the treatment of cancer. However, current cancer immunotherapies lack spatial and temporal control, resulting in systemic side effects and suboptimal patient outcomes. Responsive biomaterials have proven to be powerful tools for controlling cancer immunotherapies by providing precise control over the delivery and kinetics of immunotherapeutic cargoes. Here, we discuss biological barriers to cancer immunotherapy and how biomaterial-based strategies that respond to different stimuli — both internal and external — can be used to increase the therapeutic efficacy while reducing the toxicity of cancer immunotherapies. We examine the use of biomaterials that respond to physiological stimuli (pH, enzymes and redox potential) and exogenous energetic stimuli (light, magnetism and ultrasound) and expand upon the use of these strategies in propagating three key approaches in cancer immunotherapy: cancer vaccines, T cell-based therapy and therapies involving sustained delivery.
Immunotherapy represents an important advance in cancer treatment, yet faces challenges owing to lack of precise control, leading to systemic effects and suboptimal results for patients. This Review explores how responsive biomaterials can enhance cancer immunotherapies by responding to various internal and external stimuli to regulate the delivery and behaviour of therapeutic agents, thereby improving efficacy and reducing toxicity in treatment methods such as cancer vaccines, T cell-based therapies and sustained delivery systems.
Journal Article
Vaccination against Endogenous Retrotransposable Element Consensus Sequences Does Not Protect Rhesus Macaques from SIVsmE660 Infection and Replication
by
Piaskowski, Shari M.
,
Rakasz, Eva G.
,
Nixon, Douglas F.
in
Acquired immune deficiency syndrome
,
AIDS
,
AIDS vaccines
2014
The enormous sequence diversity of HIV remains a major roadblock to the development of a prophylactic vaccine and new approaches to induce protective immunity are needed. Endogenous retrotransposable elements (ERE) such as endogenous retrovirus K (ERV)-K and long interspersed nuclear element-1 (LINE-1) are activated during HIV-1-infection and could represent stable, surrogate targets to eliminate HIV-1-infected cells. Here, we explored the hypothesis that vaccination against ERE would protect macaques from acquisition and replication of simian immunodeficiency virus (SIV). Following vaccination with antigens derived from LINE-1 and ERV-K consensus sequences, animals mounted immune responses that failed to delay acquisition of SIVsmE660. We observed no differences in acute or set point viral loads between ERE-vaccinated and control animals suggesting that ERE-specific responses were not protective. Indeed, ERE-specific T cells failed to expand anamnestically in vivo following infection with SIVsmE660 and did not recognize SIV-infected targets in vitro, in agreement with no significant induction of targeted ERE mRNA by SIV in macaque CD4+ T cells. Instead, lower infection rates and viral loads correlated significantly to protective TRIM5α alleles. Cumulatively, these data demonstrate that vaccination against the selected ERE consensus sequences in macaques did not lead to immune-mediated recognition and killing of SIV-infected cells, as has been shown for HIV-infected human cells using patient-derived HERV-K-specific T cells. Thus, further research is required to identify the specific nonhuman primate EREs and retroviruses that recapitulate the activity of HIV-1 in human cells. These results also highlight the complexity in translating observations of the interplay between HIV-1 and human EREs to animal models.
Journal Article
Polyethyleneimine is a potent mucosal adjuvant for viral glycoprotein antigens
by
Hillson, William R
,
Sattentau, Quentin J
,
Cole, Suzanne
in
631/45/612/1231
,
631/45/612/1256
,
631/61/24/590/2291
2012
No mucosal adjuvant formulation is approved for clinical use, even though boosting immunity at sites of pathogen entry should increase the efficacy of nonreplicating vaccines. Wegmann
et al
. report that polyethyleneimine (PEI) acts as a potent mucosal adjuvant for protein antigens from influenza and herpes simplex virus, protecting mice against otherwise lethal infections.
Protection against mucosally transmitted infections probably requires immunity at the site of pathogen entry
1
, yet there are no mucosal adjuvant formulations licensed for human use. Polyethyleneimine (PEI) represents a family of organic polycations used as nucleic acid transfection reagents
in vitro
and DNA vaccine delivery vehicles
in vivo
2
,
3
. Here we show that diverse PEI forms have potent mucosal adjuvant activity for viral subunit glycoprotein antigens. A single intranasal administration of influenza hemagglutinin or herpes simplex virus type-2 (HSV-2) glycoprotein D with PEI elicited robust antibody-mediated protection from an otherwise lethal infection, and was superior to existing experimental mucosal adjuvants. PEI formed nanoscale complexes with antigen, which were taken up by antigen-presenting cells
in vitro
and
in vivo
, promoted dendritic cell trafficking to draining lymph nodes and induced non-proinflammatory cytokine responses. PEI adjuvanticity required release of host double-stranded DNA that triggered Irf3-dependent signaling. PEI therefore merits further investigation as a mucosal adjuvant for human use.
Journal Article
Small-molecule-mediated control of the anti-tumour activity and off-tumour toxicity of a supramolecular bispecific T cell engager
by
Sheppard, Neil C.
,
El-Mayta, Rakan
,
Billingsley, Margaret M.
in
631/61/350/354
,
631/61/54/152
,
Amantadine
2024
The broader clinical use of bispecific T cell engagers for inducing anti-tumour toxicity is hindered by their on-target off-tumour toxicity and the associated neurotoxicity and cytokine-release syndrome. Here we show that the off-tumour toxicity of a supramolecular bispecific T cell engager binding to the T cell co-receptor CD3 and to the human epidermal growth factor receptor 2 on breast tumour cells can be halted by disengaging the T cells from the tumour cells via the infusion of the small-molecule drug amantadine, which disassembles the supramolecular aggregate. In mice bearing human epidermal growth factor receptor 2-expressing tumours and with a human immune system, high intravenous doses of such a ‘switchable T cell nanoengager’ elicited strong tumour-specific adaptive immune responses that prevented tumour relapse, while the infusion of amantadine restricted off-tumour toxicity, cytokine-release syndrome and neurotoxicity. Supramolecular chemistry may be further leveraged to control the anti-tumour activity and off-tumour toxicity of bispecific antibodies.
The off-tumour toxicity of a supramolecular bispecific T cell engager can be halted by disengaging T cells from the tumour cells via the disassembly of the supramolecular aggregate through the infusion of the small-molecule drug amantadine.
Journal Article