Figure - available from: Cancers
This content is subject to copyright.
Strategies for T cell selection for adoptive cell therapy (ACT) of cancer. Resected tumour and peripheral blood samples are two main sources of T cells for ACT of cancer. Tumour samples require processing to obtain tumour infiltrating lymphocytes (TILs). Heterogenous TIL population can be expanded according to the rapid expansion protocol (REP). Alternatively, TILs expressing programmed death receptor 1 (PD-1) or 4-1BB can be selected, to increase frequency of tumour-specific T cells, which are subsequently expanded. Another approach includes cancer cell sequencing followed by in silico peptide prediction and tandem minigenes (TMGs) or synthetic peptide generation. Subsequently, T cells isolated from cancer patient, after optional enrichment for PD-1⁺/4-1BB⁺ cells, are tested for reactivity to predicted neopeptides with one of the following approaches: binding to MHC tetramers loaded with synthetic peptides (pMHC tetramers); and expression of activation marker (e.g., 4-1BB) in the presence of APCs transfected with TMGs or pulsed with synthetic peptides. Then, antigen-specific T cells are sorted and their TCRs are sequenced in aim to introduce tumour-specific TCRs into polyclonal T cells. Subsequently, TCR-engineered T cells are expanded for therapeutically relevant numbers (the figure was created with BioRender).

Strategies for T cell selection for adoptive cell therapy (ACT) of cancer. Resected tumour and peripheral blood samples are two main sources of T cells for ACT of cancer. Tumour samples require processing to obtain tumour infiltrating lymphocytes (TILs). Heterogenous TIL population can be expanded according to the rapid expansion protocol (REP). Alternatively, TILs expressing programmed death receptor 1 (PD-1) or 4-1BB can be selected, to increase frequency of tumour-specific T cells, which are subsequently expanded. Another approach includes cancer cell sequencing followed by in silico peptide prediction and tandem minigenes (TMGs) or synthetic peptide generation. Subsequently, T cells isolated from cancer patient, after optional enrichment for PD-1⁺/4-1BB⁺ cells, are tested for reactivity to predicted neopeptides with one of the following approaches: binding to MHC tetramers loaded with synthetic peptides (pMHC tetramers); and expression of activation marker (e.g., 4-1BB) in the presence of APCs transfected with TMGs or pulsed with synthetic peptides. Then, antigen-specific T cells are sorted and their TCRs are sequenced in aim to introduce tumour-specific TCRs into polyclonal T cells. Subsequently, TCR-engineered T cells are expanded for therapeutically relevant numbers (the figure was created with BioRender).

Source publication
Article
Full-text available
In recent years, much research has been focused on the field of adoptive cell therapies (ACT) that use native or genetically modified T cells as therapeutic tools. Immunotherapy with T cells expressing chimeric antigen receptors (CARs) demonstrated great success in the treatment of haematologic malignancies, whereas adoptive transfer of autologous...

Citations

... Adoptive T cell therapy using ex vivo expanded cytotoxic T lymphocytes (CTL) against tumor-associated antigens provides an important immune defense against cancer and has achieved durable remissions in selected malignancies. 1,2 Although there are clear benefits using CAR-T or adoptive T cell therapy, progress in the field has been impeded by exhaustion and senescence of these tumor targeting effector cells, which limits proliferation and functional activities against tumor cells. [3][4][5][6][7][8][9] Among proposed methodologies to revitalize T cells, epigenetic reprogramming into iPSC precursor cells, followed by T cell lineage redifferentiation has shown encouraging results. ...
Article
Full-text available
A major hurdle in adoptive T cell therapy is cell exhaustion and failure to maintain anti-tumor responses. Here, we introduce an induced pluripotent stem cell (iPSC) strategy for reprogramming and revitalization of precursor exhausted BCMA-specific T cells to effectively target multiple myeloma (MM). Heteroclitic BCMA72-80 LMFLLRKI)-specific CD8+ memory cytotoxic T lymphocytes (CTL) were epigenetically reprogrammed to a pluripotent state, developed into hematopoietic progenitor cells (HPC: CD34+ CD43+/CD14- CD235a-), differentiated into the T cell lineage and evaluated for their poly-functional activities against MM. The final T cell products demonstrated; 1) mature CD8αβ+ memory phenotype, 2) high expression of activation/costimulatory molecules (CD38, CD28, 41BB), 3) no expression of immune checkpoint and senescence markers (CTLA4, PD1, LAG3, TIM3; CD57), and 4) robust proliferation and poly-functional immune responses to MM. The BCMC-specific iPSC-T cells possessed a single T cell receptor clonotype with cognate BCMA peptide recognition and specificity for targeting MM. RNAseq analyses revealed distinct genome-wide shifts and a distinctive transcriptional profile in selected iPSC clones, which can develop CD8αβ+ memory T cells. This includes a repertoire of gene regulators promoting T cell lineage-development, memory CTL activation, and immune response regulation [LCK, IL7R; 4-1BB, TRAIL, GZMB, FOXF1, ITGA1]. This study highlights the potential application of iPSC technology to an adaptive T cell therapy protocol and identifies specific transcriptional patterns that could serve as a biomarker for selection of suitable iPSC clones for successful development of antigen-specific CD8αβ+ memory T cells to improve MM patient outcome.
... Therefore, the induction of target antigen expression favors the efficacy of CART cell therapy after RT [89,90]. The immune cells present different sensitivity to irreversible damage induced by RT. ...
Article
Full-text available
Tumor behavior is determined by its interaction with the tumor microenvironment (TME). Chimeric antigen receptor (CART) cell therapy represents a new form of cellular immunotherapy (IT). Immune cells present a different sensitivity to radiation therapy (RT). RT can affect tumor cells both modifying the TME and inducing DNA damage, with different effects depending on the low and high doses delivered, and can favor the expression of CART cells. CART cells are patients’ T cells genetically engineered to recognize surface structure and to eradicate cancer cells. High-dose radiation therapy (HDRT, >10–20 Gy/fractions) converts immunologically “cold” tumors into “hot” ones by inducing necrosis and massive inflammation and death. LDRT (low-dose radiation therapy, >5–10 Gy/fractions) increases the expansion of CART cells and leads to non-immunogenetic death. An innovative approach, defined as the LATTICE technique, combines a high dose in higher FDG- uptake areas and a low dose to the tumor periphery. The association of RT and immune checkpoint inhibitors increases tumor immunogenicity and immune response both in irradiated and non-irradiated sites. The aim of this narrative review is to clarify the knowledge, to date, on CART cell therapy and its possible association with radiation therapy in solid tumors.
... One such advanced alternative therapeutic modality is CAR-T cells, which provide specific target engagement and generate a robust antitumor response. [269][270][271] Despite their effectiveness, CAR-T cells have considerable safety concerns due to uncontrolled activation, growth, and termination. To address this, CAR-T cell systems that employ small-molecule safety switches have been developed with reduced toxicities. ...
Article
Living systems use proximity to regulate biochemical processes. Inspired by this phenomenon, bifunctional modalities that induce proximity have been developed to redirect cellular processes. An emerging example of this class is molecules that induce ubiquitin-dependent proteasomal degradation of a protein of interest, and their initial development sparked a flurry of discovery for other bifunctional modalities. Recent advances in this area include modalities that can change protein phosphorylation, glycosylation, and acetylation states, modulate gene expression, and recruit components of the immune system. In this review, we highlight bifunctional modalities that perform functions other than degradation and have great potential to revolutionize disease treatment, while also serving as important tools in basic research to explore new aspects of biology.
... RT induces DNA damage and thus promotes the occurrence of new mutations within the tumor. These de novo mutations often result in the expression of cancer antigens and neoantigens, hence favoring CAR T cell therapy initiation after RT when CAR target antigen expression is induced [12][13][14][15]. ...
Article
Full-text available
CAR T cell-based therapies have revolutionized the treatment of hematological malignancies such as leukemia and lymphoma within the last years. In contrast to the success in hematological cancers, the treatment of solid tumors with CAR T cells is still a major challenge in the field and attempts to overcome these hurdles have not been successful yet. Radiation therapy is used for management of various malignancies for decades and its therapeutic role ranges from local therapy to a priming agent in cancer immunotherapy. Combinations of radiation with immune checkpoint inhibitors have already proven successful in clinical trials. Therefore, a combination of radiation therapy may have the potential to overcome the current limitations of CAR T cell therapy in solid tumor entities. So far, only limited research was conducted in the area of CAR T cells and radiation. In this review we will discuss the potential and risks of such a combination in the treatment of cancer patients.
... Among the different ACT methods developed, chimeric antigen receptor (CAR)-T cell gene therapy has attracted the most attention for killing cancer cells. CAR-modified T cells may identify different types of antigens regardless of how they are presented on MHC molecules (298). To date, there are currently 24 clinical trials examining the efficacy of CAR-T cell treatment for GBM according to Clinicaltrials.gov ...
Article
Glioblastoma, also referred to as glioblastoma multiforme (GBM), is grade IV astrocytoma characterized by being fast-growing and the most aggressive brain tumor. In adults, it is the most prevalent type of malignant brain tumor. Despite the advancements in both diagnosis tools and therapeutic treatments, GBM is still associated with poor survival rate without any statistically significant improvement in the past three decades. Patient's genome signature is one of the key factors causing the development of this tumor, in addition to previous radiation exposure and other environmental factors. Researchers have identified genomic and subsequent molecular alterations affecting core pathways that trigger the malignant phenotype of this tumor. Targeting intrinsically altered molecules and pathways is seen as a novel avenue in GBM treatment. The present review shed light on signaling pathways and intrinsically altered molecules implicated in GBM development. It discussed the main challenges impeding successful GBM treatment, such as the blood brain barrier and tumor microenvironment (TME), the plasticity and heterogeneity of both GBM and TME and the glioblastoma stem cells. The present review also presented current advancements in GBM molecular targeted therapy in clinical trials. Profound and comprehensive understanding of molecular participants opens doors for innovative, more targeted and personalized GBM therapeutic modalities.
... Additionally, the viability and persistence of infused cells are important factors to increase therapeutic potential for adoptive transfer. There is an emerging need for strategies to improve the viability of ex vivo expanded cells (42,43). In Treg tra cking, we found that bvPLA2 treated Tregs were more frequently detected in most tissues, including the liver, after transferred 7 days. ...
Preprint
Full-text available
Background Parkinson’s disease is a long-term neurodegenerative disease characterized by dopaminergic neuronal loss and the aggregation of alpha-synuclein in the brain. Cell therapy using regulatory T cells has therapeutic potential on Parkinson’s progression in a 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced mouse model; however, several challenges were associated with its applications. Polyclonal regulatory T cells can move to sites other than the disease loci and cause undesirable suppression. Moreover, the efficiency of regulatory T cells is reduced during the expansion process, accompanied by phenotypic changes. Here, we propose a strategy for regulatory T cell expansion using alpha-synuclein and bee venom phospholipase A2. Methods We presented alpha-synuclein to T cells via dendritic cells following bee venom phospholipase A2 treatment and analyzed their phenotype and mobility. These regulatory T cells were transferred to 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced Parkinson’s mouse model. First, we measured motor function using pole test. Next, we examined the expression of tyrosine hydroxylase, alpha-synuclein, ionized calcium binding adaptor molecule 1, and nitric oxide synthase 2 in the substantia nigra by immunohistostaining. The mRNA expression related to neuroinflammation was measured using real-time reverse transcription polymerase chain reaction. Results Our method increased the mobility of regulatory T cells towards the site of abundant alpha-synuclein in vitro and in vivo. Alpha-synuclein-specific bee venom phospholipase A2-treated regulatory T cells showed noteworthy neuroprotective effects against motor function deficits, dopaminergic neuronal loss, and alpha-synuclein accumulation in the Parkinson's mouse model. Furthermore, adoptive transfer of alpha-synuclein-specific bee venom phospholipase A2-treated regulatory T cells exerted immunosuppressive effects on activated microglia, especially pro-inflammatory microglia, in Parkinson’s mice. Conclusions Our findings suggest that the combination of alpha-synuclein and bee venom phospholipase A2 may provide a significant improvement in neuroprotective activities of regulatory T cells and suggest the effective clinical application of T cell therapy in Parkinson’s.
... The vast majority of potentially actionable microbial and cancer antigens are major histocompatibility complex (MHC)-bound peptides. 1,2 Although it is possible to isolate T cell receptors (TCR) targeting some of these antigens and manufacture TCR-transgenic T cells ex vivo followed by their injection into affected individuals, such adoptive immunotherapy approaches face numerous technical hurdles and are currently available only for a minority of relevant cancer antigens. 3 Several effective strategies rely on ex vivo expansion of native antigen-specific T cells, enabling targeting of a vast array of antigens. ...
Article
Full-text available
Antigen-specific T cells expansion ex vivo followed by adoptive transfer enables the targeting of a multitude of microbial and cancer antigens. However, clinical scale T-cell expansion from rare precursors requires repeated stimulations which may lead to T-cell dysfunction and limited therapeutic potential. We used a clinically-compliant protocol to expand Epstein-Barr virus (EBV) and Wilms Tumor 1 (WT1) antigen-specific CD8⁺ T cells, and leveraged T-cell exhaustion-associated inhibitory receptor blockade to improve T-cell expansion. Several inhibitory receptors were expressed early by ex vivo expanded antigen-specific CD8⁺ T cells, including PD-1 and TIM-3, with co-expression matching evidence of T-cell dysfunction as the cultures progressed. The introduction of anti-PD-L1 and anti-TIM-3 blockade in combination (but not individually) to the culture led to markedly improved antigen-specific T-cell expansion without inducing T-cell dysfunction. Further, single-cell RNA sequencing (RNA-Seq) and T-cell receptor (TCR) repertoire profiling revealed that double blockade does not impart specific transcriptional programs in T cells or alterations in TCR repertoires. However, combined blockade may impact gene expression in a minority of clonotypes in a donor-specific fashion. We conclude that antigen-specific CD8⁺ T-cell manufacturing can be improved by using TIM-3 and PD-L1/PD-1 axis blockade in combination. This approach is readily applicable to several adoptive-immunotherapy strategies.
... T cells equipped with a TCR can recognize peptides presented on MHC molecules and target cancer cells. Nevertheless, and to overcome the limitation of antigen presentation, CAR-modified T cells recognize various types of antigens regardless of their presentation on MHC molecules (135). The aforementioned categories have different mechanisms of action to eliminate cancer cells. ...
Article
Cancer still ranks as one of the top causes of morbidity and mortality despite recent improvements in standard chemotherapy, radiotherapy, and surgery. This underlines some of the difficulties in creating successful therapeutic strategies, but it also highlights the shortcomings of conventional methods. In order to enhance the standard treatment of cancer patients, biology‑driven therapies are emerging towards more specific and effective clinical options. In the present review, both conventional and novel methods for cancer treatment were addressed, with a particular focus on Glioblastoma multiforme (GBM) therapies. GBM is one of the most challenging cancers for conventional treatments, and survival rates of patients remain very low. In the present review, focus was addressed on employed chemo‑ and radiotherapies along with developing novel targeted and immunotherapies assessed in clinical trials on patients with GBM or yet to be evaluated clinically. It was aimed to evaluate efficiency of treatments in suppressing GBMs, roadblocks and challenges. A brief discussion of a few promising delivery methods for targeted drug and gene therapy for cancer was also provided. Increment advancements in this field emphasizes the significance of combining different treatment strategies for improved survival and quality of patients' lives.
... No follow up of this study or additional clinical documentation regarding CAR T cell therapy has been reported since then [41]. Potential barriers include the insufficient T cell penetration into solid masses due to physical obstacles, the lack of targetable antigens solely expressed on tumor cells and the limited CAR T cell persistence after infusion and homing of potent immunosuppressive cells that tender T cells dysfunction in the TME [42]. An alternative and less complex approach of ACT is the CAR-NK cell therapy [43,44]. ...
Article
Full-text available
Sarcomas are uncommon malignancies of mesenchymal origin that can arise throughout the human lifespan, at any part of the body. Surgery remains the optimal treatment modality whilst response to conventional treatments, such as chemotherapy and radiation, is minimal. Immunotherapy has emerged as a novel approach to treat different cancer types but efficacy in soft tissue sarcoma and bone sarcoma is limited to distinct subtypes. Growing evidence shows that cancer-stroma cell interactions and their microenvironment play a key role in the effectiveness of immunotherapy. However, the pathophysiological and immunological properties of the sarcoma tumor microenvironment in relation to immunotherapy advances, has not been broadly reviewed. Here, we provide an up-to-date overview of the different immunotherapy modalities as potential treatments for sarcoma, identify barriers posed by the sarcoma microenvironment to immunotherapy, highlight their relevance for impeding effectiveness, and suggest mechanisms to overcome these barriers.
... T cells can recognize tumor-associated antigens without using the major histocompatibility complex if they have a chimeric antigen receptor made artificially [57]. In the therapy of resistant B cell malignancies, chimeric antigen receptor-T cells that targeted the pan-B-cell marker CD19 showed an excellent reaction [126]. ...
Article
Full-text available
Background Findings of new targeted treatments with adequate safety evaluations are essential for better cancer cures and mortality rates. Immunotherapy holds promise for patients with relapsed disease, with the ability to elicit long-term remissions. Emerging promising clinical results in B-cell malignancy using gene-altered T-lymphocytes uttering chimeric antigen receptors have sparked a lot of interest. This treatment could open the path for a major difference in the way we treat tumors that are resistant or recurring. Main body Genetically altered T cells used to produce tumor-specific chimeric antigen receptors are resurrected fields of adoptive cell therapy by demonstrating remarkable success in the treatment of malignant tumors. Because of the molecular complexity of chimeric antigen receptors-T cells, a variety of engineering approaches to improve safety and effectiveness are necessary to realize larger therapeutic uses. In this study, we investigate new strategies for enhancing chimeric antigen receptors-T cell therapy by altering chimeric antigen receptors proteins, T lymphocytes, and their relations with another solid tumor microenvironment (TME) aspects. Furthermore, examine the potential region of chimeric antigen receptors-T cells therapy to become a most effective treatment modality, taking into account the basic and clinical and practical aspect. Short conclusions Chimeric antigen receptors-T cells have shown promise in the therapy of hematological cancers. Recent advancements in protein and cell editing, as well as genome-editing technologies, have paved the way for multilayered T cell therapy techniques that can address numerous important demands. At around the same time, there is crosstalk between various intended aspects within the chimeric antigen receptors-T cell diverse biological complexity and possibilities. These breakthroughs substantially improve the ability to comprehend these complex interactions in future solid tumor chimeric antigen receptor-T cell treatment and open up new treatment options for patients that are currently incurable.