Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Immunol Rev ; 320(1): 166-198, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37548063

ABSTRACT

Adoptive T-cell transfer (ACT) therapies, including of tumor infiltrating lymphocytes (TILs) and T cells gene-modified to express either a T cell receptor (TCR) or a chimeric antigen receptor (CAR), have demonstrated clinical efficacy for a proportion of patients and cancer-types. The field of ACT has been driven forward by the clinical success of CD19-CAR therapy against various advanced B-cell malignancies, including curative responses for some leukemia patients. However, relapse remains problematic, in particular for lymphoma. Moreover, for a variety of reasons, relative limited efficacy has been demonstrated for ACT of non-hematological solid tumors. Indeed, in addition to pre-infusion challenges including lymphocyte collection and manufacturing, ACT failure can be attributed to several biological processes post-transfer including, (i) inefficient tumor trafficking, infiltration, expansion and retention, (ii) chronic antigen exposure coupled with insufficient costimulation resulting in T-cell exhaustion, (iii) a range of barriers in the tumor microenvironment (TME) mediated by both tumor cells and suppressive immune infiltrate, (iv) tumor antigen heterogeneity and loss, or down-regulation of antigen presentation machinery, (v) gain of tumor intrinsic mechanisms of resistance such as to apoptosis, and (vi) various forms of toxicity and other adverse events in patients. Affinity-optimized TCRs can improve T-cell function and innovative CAR designs as well as gene-modification strategies can be used to coengineer specificity, safety, and function into T cells. Coengineering strategies can be designed not only to directly support the transferred T cells, but also to block suppressive barriers in the TME and harness endogenous innate and adaptive immunity. Here, we review a selection of the remarkable T-cell coengineering strategies, including of tools, receptors, and gene-cargo, that have been developed in recent years to augment tumor control by ACT, more and more of which are advancing to the clinic.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , T-Lymphocytes , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Immunotherapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Antigens, Neoplasm , Tumor Microenvironment
2.
Nat Biomed Eng ; 7(9): 1063-1080, 2023 09.
Article in English | MEDLINE | ID: mdl-37069267

ABSTRACT

Vectors that facilitate the engineering of T cells that can better harness endogenous immunity and overcome suppressive barriers in the tumour microenvironment would help improve the safety and efficacy of T-cell therapies for more patients. Here we report the design, production and applicability, in T-cell engineering, of a lentiviral vector leveraging an antisense configuration and comprising a promoter driving the constitutive expression of a tumour-directed receptor and a second promoter enabling the efficient activation-inducible expression of a genetic payload. The vector allows for the delivery of a variety of genes to human T cells, as we show for interleukin-2 and a microRNA-based short hairpin RNA for the knockdown of the gene coding for haematopoietic progenitor kinase 1, a negative regulator of T-cell-receptor signalling. We also show that a gene encoded under an activation-inducible promoter is specifically expressed by tumour-redirected T cells on encountering a target antigen in the tumour microenvironment. The single two-gene-encoding vector can be produced at high titres under an optimized protocol adaptable to good manufacturing practices.


Subject(s)
Lentivirus , Neoplasms , Humans , Lentivirus/genetics , T-Lymphocytes , Transgenes/genetics , Promoter Regions, Genetic/genetics , Neoplasms/genetics , Neoplasms/therapy , Tumor Microenvironment
3.
Biosens Bioelectron ; 220: 114826, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36371959

ABSTRACT

Nicotinamide riboside (NR) is a form of vitamin B3 and is one of the most studied compounds for the restoration of cellular NAD+ levels demonstrating clinical potential in many metabolic and age-related disorders. Despite its wide commercial availability as a powerful nutraceutical, our understanding of NR uptake by different cells and tissues is greatly limited by the lack of noninvasive in vivo imaging tools limiting its clinical translation. Here, we report the development and validation of a bioluminescent NR uptake probe (BiNR) for non-invasive longitudinal imaging of NR uptake both in vitro and in vivo. In addition, we optimized an assay that allows monitoring of NR flux without the need to transfect cells with the luciferase gene, enabling the use of the BiNR probe in clinical samples, as demonstrated with human T cells. Lastly, we used BiNR to investigate the role of NR uptake in cancer prevalence and metastases formation in triple negative breast cancer (TNBC) animal model. Our results demonstrate that NR supplementation results in a significant increase in cancer prevalence and metastases of TNBC to the brain. These results outline the important role of powerful nutraceuticals like NR in cancer metabolism and the need to personalize their use in certain patient populations.


Subject(s)
Biosensing Techniques , Triple Negative Breast Neoplasms , Animals , Humans , NAD , Niacinamide/metabolism , Pyridinium Compounds
4.
Front Immunol ; 13: 976628, 2022.
Article in English | MEDLINE | ID: mdl-36203587

ABSTRACT

Despite the tremendous success of adoptive T-cell therapies (ACT) in fighting certain hematologic malignancies, not all patients respond, a proportion experience relapse, and effective ACT of most solid tumors remains elusive. In order to improve responses to ACT suppressive barriers in the solid tumor microenvironment (TME) including insufficient nutrient availability must be overcome. Here we explored how enforced expression of the high-affinity glucose transporter GLUT3 impacted tumor-directed T cells. Overexpression of GLUT3 in primary murine CD8+ T cells enhanced glucose uptake and increased glycogen and fatty acid storage, and was associated with increased mitochondrial fitness, reduced ROS levels, higher abundance of the anti-apoptotic protein Mcl-1, and better resistance to stress. Importantly, GLUT3-OT1 T cells conferred superior control of B16-OVA melanoma tumors and, in this same model, significantly improved survival. Moreover, a proportion of treated mice were cured and protected from re-challenge, indicative of long-term T cell persistence and memory formation. Enforcing expression of GLUT3 is thus a promising strategy to improve metabolic fitness and sustaining CD8+ T cell effector function in the context of ACT.


Subject(s)
CD8-Positive T-Lymphocytes , Glucose Transporter Type 3/metabolism , Melanoma, Experimental , Animals , Fatty Acids , Glucose , Glucose Transporter Type 3/genetics , Glycogen , Immunologic Memory , Melanoma, Experimental/therapy , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Recurrence, Local , Reactive Oxygen Species , Tumor Microenvironment
5.
Front Immunol ; 13: 951143, 2022.
Article in English | MEDLINE | ID: mdl-35990626

ABSTRACT

Chimeric antigen receptor (CAR) T cells have emerged as a powerful immunotherapeutic tool against certain hematological malignancies but a significant proportion of patients either do not respond or they relapse, sometimes as a result of target antigen loss. Moreover, limited clinical benefit has been reported for CAR therapy against epithelial derived solid tumors. A major reason for this is the paucity of solid tumor antigens identified to date that are broadly, homogeneously and stably expressed but not found on healthy tissues. To address this, here we describe the development and evaluation of CAR T cells directed against N-glycoslylated ganglioside monosialic 3 (NGcGM3). NGcGM3 derives from the enzymatic hydroxylation of N-acetylneuraminic acid (NAc) GM3 (NAcGM3) and it is present on the surface of a range of cancers including ovarian, breast, melanoma and lymphoma. However, while NAcGM3 is found on healthy human cells, NGcGM3 is not due to the 7deletion of an exon in the gene encoding for the enzyme cytidine monophospho-N-acetylneuraminic acid hydroxylase (CMAH). Indeed, unlike for most mammals, in humans NGcGM3 is considered a neoantigen as its presence on tumors is the result of metabolic incorporation from dietary sources. Here, we have generated 3 CARs comprising different single chain variable fragments (scFvs) originating from the well-characterized monoclonal antibody (mAb) 14F7. We show reactivity of the CAR T cells against a range of patient tumor fragments and we demonstrate control of NGcGM3+ SKOV3 ovarian tumors in the absence of toxicity despite the expression of CMAH and presence of NGcGM3+ on healthy tissues in NSG mice. Taken together, our data indicate clinical potential for 14F7-based CAR T cells against a range of cancers, both in terms of efficacy and of patient safety.


Subject(s)
G(M3) Ganglioside , Ovarian Neoplasms , Animals , Female , G(M3) Ganglioside/metabolism , Humans , Immunotherapy, Adoptive , Mammals/metabolism , Mice , Neoplasm Recurrence, Local , Ovarian Neoplasms/therapy , T-Lymphocytes
6.
Leuk Lymphoma ; 63(7): 1566-1579, 2022 07.
Article in English | MEDLINE | ID: mdl-35259043

ABSTRACT

Chimeric antigen receptors (CAR)-modified T cells are an emerging therapeutic tool for chronic lymphocytic leukemia (CLL). However, in patients with CLL, well-known T-cell defects and the inhibitory properties of the tumor microenvironment (TME) hinder the efficacy of CAR T cells. We explored a novel approach combining CARs with lenalidomide, an immunomodulatory drug that tempers the immunosuppressive activity of the CLL TME. T cells from patients with CLL were engineered to express a CAR specific for CD23, a promising target antigen. Lenalidomide maintained the in vitro effector functions of CD23.CAR+ T cells effector functions in terms of antigen-specific cytotoxicity, cytokine release and proliferation. Overall, lenalidomide preserved functional CAR T-CLL cell immune synapses. In a Rag2-/-γc-/--based xenograft model of CLL, we demonstrated that, when combined with low-dose lenalidomide, CD23.CAR+ T cells efficiently migrated to leukemic sites and delayed disease progression when compared to CD23.CAR+ T cells given with rhIL-2. These observations underline the therapeutic potential of this novel CAR-based combination strategy in CLL.


Subject(s)
Immunotherapy, Adoptive , Leukemia, Lymphocytic, Chronic, B-Cell , Humans , Interleukin Receptor Common gamma Subunit , Lenalidomide/pharmacology , Lenalidomide/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , T-Lymphocytes , Tumor Microenvironment
7.
Cell ; 185(4): 585-602.e29, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35051368

ABSTRACT

The relevance of extracellular magnesium in cellular immunity remains largely unknown. Here, we show that the co-stimulatory cell-surface molecule LFA-1 requires magnesium to adopt its active conformation on CD8+ T cells, thereby augmenting calcium flux, signal transduction, metabolic reprogramming, immune synapse formation, and, as a consequence, specific cytotoxicity. Accordingly, magnesium-sufficiency sensed via LFA-1 translated to the superior performance of pathogen- and tumor-specific T cells, enhanced effectiveness of bi-specific T cell engaging antibodies, and improved CAR T cell function. Clinically, low serum magnesium levels were associated with more rapid disease progression and shorter overall survival in CAR T cell and immune checkpoint antibody-treated patients. LFA-1 thus directly incorporates information on the composition of the microenvironment as a determinant of outside-in signaling activity. These findings conceptually link co-stimulation and nutrient sensing and point to the magnesium-LFA-1 axis as a therapeutically amenable biologic system.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Magnesium/metabolism , Animals , Bacterial Infections/immunology , Caloric Restriction , Cell Line, Tumor , Cytotoxicity, Immunologic , HEK293 Cells , Humans , Immunologic Memory , Immunological Synapses/metabolism , Immunotherapy , Lymphocyte Activation/immunology , MAP Kinase Signaling System , Magnesium/administration & dosage , Male , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism
8.
Cell Mol Life Sci ; 78(1): 227-247, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32157317

ABSTRACT

Chronic inflammation that affects primarily metabolic organs, such as white adipose tissue (WAT), is considered as a major cause of human obesity-associated co-morbidities. However, the molecular mechanisms initiating this inflammation in WAT are poorly understood. By combining transcriptomics, ChIP-seq and modeling approaches, we studied the global early and late responses to a high-fat diet (HFD) in visceral (vWAT) and subcutaneous (scWAT) AT, the first being more prone to obesity-induced inflammation. HFD rapidly triggers proliferation of adipocyte precursors within vWAT. However, concomitant antiadipogenic signals limit vWAT hyperplastic expansion by interfering with the differentiation of proliferating adipocyte precursors. Conversely, in scWAT, residing beige adipocytes lose their oxidizing properties and allow storage of excessive fatty acids. This phase is followed by tissue hyperplastic growth and increased angiogenic signals, which further enable scWAT expansion without generating inflammation. Our data indicate that scWAT and vWAT differential ability to modulate adipocyte number and differentiation in response to obesogenic stimuli has a crucial impact on the different susceptibility to obesity-related inflammation of these adipose tissue depots.


Subject(s)
Adipogenesis , Adipose Tissue, White/metabolism , Cell Differentiation , Inflammation/pathology , Obesity/pathology , Adipose Tissue, White/cytology , Adipose Tissue, White/pathology , Animals , Diet, High-Fat , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation , Inflammation/etiology , Inflammation/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/complications , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/metabolism , Subcutaneous Fat/cytology , Subcutaneous Fat/metabolism , Subcutaneous Fat/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Wnt Proteins/metabolism
10.
Nat Biotechnol ; 38(4): 426-432, 2020 04.
Article in English | MEDLINE | ID: mdl-32015549

ABSTRACT

Approaches to increase the activity of chimeric antigen receptor (CAR)-T cells against solid tumors may also increase the risk of toxicity and other side effects. To improve the safety of CAR-T-cell therapy, we computationally designed a chemically disruptable heterodimer (CDH) based on the binding of two human proteins. The CDH self-assembles, can be disrupted by a small-molecule drug and has a high-affinity protein interface with minimal amino acid deviation from wild-type human proteins. We incorporated the CDH into a synthetic heterodimeric CAR, called STOP-CAR, that has an antigen-recognition chain and a CD3ζ- and CD28-containing endodomain signaling chain. We tested STOP-CAR-T cells specific for two antigens in vitro and in vivo and found similar antitumor activity compared to second-generation (2G) CAR-T cells. Timed administration of the small-molecule drug dynamically inactivated the activity of STOP-CAR-T cells. Our work highlights the potential for structure-based design to add controllable elements to synthetic cellular therapies.


Subject(s)
Receptors, Antigen, T-Cell/chemistry , Receptors, Chimeric Antigen/chemistry , Small Molecule Libraries/pharmacology , T-Lymphocytes/drug effects , Cell Engineering , Cells, Cultured , Humans , Immunotherapy, Adoptive , Jurkat Cells , Lymphocyte Activation/drug effects , PC-3 Cells , Protein Binding , Protein Engineering , Protein Multimerization , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/antagonists & inhibitors , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Signal Transduction , Small Molecule Libraries/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
11.
Am J Physiol Renal Physiol ; 314(6): F1154-F1165, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29488390

ABSTRACT

Different complex mechanisms control the morphology of podocyte foot processes and their interactions with the underlying basement membrane. Injuries to this system often cause glomerular dysfunction and albuminuria. The present study aimed at identifying early markers of glomerular damage in diabetic nephropathy. For this purpose, we performed a microarray analysis on kidneys of 3-wk-old peroxisome proliferator-activated receptor-γ (PPARγ)-null and AZIP/F1 mice, which are two models of diabetic nephropathy due to lipodystrophy. This was followed by functional annotation of the enriched clusters of genes. One of the significant changes in the early stages of glomerular damage was the increase of hemicentin 1 (HMCN1). Its expression and distribution were then studied by real-time PCR and immunofluorescence in various models of glomerular damage and on podocyte cell cultures. HMCN1 progressively increased in the glomeruli of diabetic mice, according to disease severity, as well as in puromycin aminonucleoside (PA)-treated rats. Studies on murine and human podocytes showed an increased HMCN1 deposition upon different pathological stimuli, such as hyperglycemia, transforming growth factor-ß (TGF-ß), and PA. In vitro silencing studies showed that HMCN1 mediated the rearrangements of podocyte cytoskeleton induced by TGF-ß. Finally, we demonstrated an increased expression of HMCN1 in the kidneys of patients with proteinuric nephropathies. In summary, our studies identified HMCN1 as a new molecule involved in the dynamic changes of podocyte foot processes. Its increased expression associated with podocyte dysfunction points to HMCN1 as a possible marker for the early glomerular damage occurring in different proteinuric nephropathies.


Subject(s)
Calcium-Binding Proteins/metabolism , Diabetic Nephropathies/metabolism , Extracellular Matrix Proteins/metabolism , Immunoglobulins/metabolism , Nephrosis/metabolism , Podocytes/metabolism , Animals , Calcium-Binding Proteins/genetics , Cells, Cultured , Cytoskeleton/metabolism , Cytoskeleton/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Female , Glucose/pharmacology , Humans , Immunoglobulins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Nephrosis/genetics , Nephrosis/pathology , PPAR gamma/genetics , PPAR gamma/metabolism , Podocytes/drug effects , Podocytes/pathology , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/pathology , Rats, Sprague-Dawley , Signal Transduction , Transforming Growth Factor beta/pharmacology , Up-Regulation
12.
J Invest Dermatol ; 138(3): 500-510, 2018 03.
Article in English | MEDLINE | ID: mdl-28964716

ABSTRACT

PPARγ regulates multiple aspects of skin physiology, including sebocyte differentiation, keratinocyte proliferation, epithelial stem cell survival, adipocyte biology, and inflammatory skin responses. However, the effects of its global deletion, namely of nonredundant key functions of PPARγ signaling in mammalian skin, are yet unknown because of embryonic lethality. Here, we describe the skin and hair phenotype of a whole-body PPARγ-null mouse (PpargΔ/Δ), obtained by preserving PPARγ expression in the placenta. PpargΔ/Δ mice exhibited total lipoatrophy and complete absence of sebaceous glands. Right after birth, hair follicle (HF) morphogenesis was transiently delayed, along with reduced expression of HF differentiation markers and of transcriptional regulators necessary for HF development. Later, adult PpargΔ/Δ mice developed scarring alopecia and severe perifollicular inflammation. Skin analyses in other models of lipodystrophy, AZIPtg/+ and Adipoq-Cretg/+Ppargfl/fl mice, coupled with skin graft experiments, showed that the early defects observed in hair morphogenesis were caused by the absence of adipose tissue. In contrast, the late alteration of HF cycle and appearance of inflammation were observed only in PpargΔ/Δ mice and likely were due to the lack sebaceous glands. Our findings underscore the increasing appreciation for the importance of adipose tissue-mediated signals in HF development and function.


Subject(s)
Hair Follicle/growth & development , Lipodystrophy/pathology , Morphogenesis , PPAR gamma/physiology , Animals , Cell Differentiation , Disease Models, Animal , Homeostasis , Mice , Mice, Knockout , PPAR gamma/genetics
13.
Oncotarget ; 7(32): 51581-51597, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27323395

ABSTRACT

Chimeric antigen receptor (CAR)-modified T-cell adoptive immunotherapy is a remarkable therapeutic option proven effective in the treatment of hematological malignancies. In order to optimize cell manufacturing, we sought to develop a novel clinical-grade protocol to obtain CAR-modified cytokine-induced killer cells (CIKs) using the Sleeping Beauty (SB) transposon system. Administration of irradiated PBMCs overcame cell death of stimulating cells induced by non-viral transfection, enabling robust gene transfer together with efficient T-cell expansion. Upon single stimulation, we reached an average of 60% expression of CD123- and CD19- specific 3rd generation CARs (CD28/OX40/TCRzeta). Furthermore, modified cells displayed persistence of cell subsets with memory phenotype, specific and effective lytic activity against leukemic cell lines and primary blasts, cytokine secretion, and proliferation. Adoptive transfer of CD123.CAR or CD19.CAR lymphocytes led to a significant anti-tumor response against acute myelogenous leukemia (AML) and acute lymphoblastic leukemia (ALL) disseminated diseases in NSG mice. Notably, we found no evidence of integration enrichment near cancer genes and transposase expression at the end of the differentiation. Taken all together, our findings describe a novel donor-derived non-viral CAR approach that may widen the repertoire of available methods for T cell-based immunotherapy.


Subject(s)
Genetic Therapy/methods , Immunotherapy, Adoptive/methods , Leukemia/pathology , Leukemia/therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/transplantation , Transposases/genetics , Acute Disease , Adolescent , Animals , Antigens, CD19/genetics , Antigens, CD19/metabolism , Cell Line, Tumor , Child , Child, Preschool , Combined Modality Therapy , Female , Humans , Infant , Interleukin-3 Receptor alpha Subunit/genetics , Interleukin-3 Receptor alpha Subunit/metabolism , Leukemia/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/metabolism , Transposases/metabolism , Xenograft Model Antitumor Assays
14.
Nucl Recept Signal ; 13: e001, 2015.
Article in English | MEDLINE | ID: mdl-25945080

ABSTRACT

The peroxisome proliferator-activated receptors (PPARs) are a group of nuclear receptors that function as transcription factors regulating the expression of genes involved in cellular differentiation, development, metabolism and also tumorigenesis. Three PPAR isotypes (α, ß/δ and γ) have been identified, among which PPARß/δ is the most difficult to functionally examine due to its tissue-specific diversity in cell fate determination, energy metabolism and housekeeping activities. PPARß/δ acts both in a ligand-dependent and -independent manner. The specific type of regulation, activation or repression, is determined by many factors, among which the type of ligand, the presence/absence of PPARß/δ-interacting corepressor or coactivator complexes and PPARß/δ protein post-translational modifications play major roles. Recently, new global approaches to the study of nuclear receptors have made it possible to evaluate their molecular activity in a more systemic fashion, rather than deeply digging into a single pathway/function. This systemic approach is ideally suited for studying PPARß/δ, due to its ubiquitous expression in various organs and its overlapping and tissue-specific transcriptomic signatures. The aim of the present review is to present in detail the diversity of PPARß/δ function, focusing on the different information gained at the systemic level, and describing the global and unbiased approaches that combine a systems view with molecular understanding.


Subject(s)
PPAR delta/metabolism , PPAR-beta/metabolism , Systems Biology/methods , Animals , Base Sequence , Cell Physiological Phenomena , Genomics , Humans , PPAR delta/chemistry , PPAR delta/genetics , PPAR-beta/chemistry , PPAR-beta/genetics
15.
Br J Haematol ; 161(3): 389-401, 2013 May.
Article in English | MEDLINE | ID: mdl-23432359

ABSTRACT

Current therapeutic regimens for acute myeloid leukaemia (AML) are still associated with high rates of relapse. Immunotherapy with T-cells genetically modified to express chimeric antigen receptors (CARs) represents an innovative approach. Here we investigated the targeting of the interleukin three receptor alpha (IL3RA; CD123) molecule, which is overexpressed on AML bulk population, CD34(+) leukaemia progenitors, and leukaemia stem cells (LSC) compared to normal haematopoietic stem/progenitor cells (HSPCs), and whose overexpression is associated with poor prognosis. Cytokine-induced killer (CIK) cells were transduced with SFG-retroviral-vector encoding an anti-CD123 CAR. Transduced cells were able to strongly kill CD123(+) cell lines, as well as primary AML blasts. Interestingly, secondary colony experiments demonstrated that anti-CD123.CAR preserved in vitro HSPCs, in contrast to a previously generated anti-CD33.CAR, while keeping an identical cytotoxicity profile towards AML. Furthermore, limited killing of normal monocytes and CD123-low-expressing endothelial cells was noted, thus indicating a low toxicity profile of the anti-CD123.CAR. Taken together, our results indicate that CD123-specific CARs strongly enhance anti-AML CIK functions, while sparing HSPCs and normal low-expressing antigen cells, paving the way to develop novel immunotherapy approaches for AML treatment.


Subject(s)
Cytokine-Induced Killer Cells/immunology , Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Acute/pathology , Receptors, Cell Surface/physiology , Cell Line, Tumor/metabolism , Coculture Techniques , Cytokines/metabolism , Cytotoxicity Tests, Immunologic , Endothelial Cells , Female , HEK293 Cells , Hematopoietic Stem Cells , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-3 Receptor alpha Subunit/antagonists & inhibitors , Leukemia, Monocytic, Acute/pathology , Male , Monocytes , Recombinant Fusion Proteins/physiology , Transduction, Genetic , Tumor Stem Cell Assay
16.
Blood ; 117(18): 4736-45, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21406718

ABSTRACT

Chronic lymphocytic leukemia (CLL) is characterized by an accumulation of mature CD19(+)CD5(+)CD20(dim) B lymphocytes that typically express the B-cell activation marker CD23. In the present study, we cloned and expressed in T lymphocytes a novel chimeric antigen receptor (CAR) targeting the CD23 antigen (CD23.CAR). CD23.CAR(+) T cells showed specific cytotoxic activity against CD23(+) tumor cell lines (average lysis 42%) and primary CD23(+) CLL cells (average lysis 58%). This effect was obtained without significant toxicity against normal B lymphocytes, in contrast to CARs targeting CD19 or CD20 antigens, which are also expressed physiologically by normal B lymphocytes. Moreover, CLL-derived CD23.CAR(+) T cells released inflammatory cytokines (1445-fold more TNF-ß, 20-fold more TNF-α, and 4-fold more IFN-γ). IL-2 was also produced (average release 2681 pg/mL) and sustained the antigen-dependent proliferation of CD23.CAR(+) T cells. Redirected T cells were also effective in vivo in a CLL Rag2(-/-)γ(c)(-/-) xenograft mouse model. Compared with mice treated with control T cells, the infusion of CD23.CAR(+) T cells resulted in a significant delay in the growth of the MEC-1 CLL cell line. These data suggest that CD23.CAR(+) T cells represent a selective immunotherapy for the elimination of CD23(+) leukemic cells in patients with CLL.


Subject(s)
Immunotherapy, Adoptive/methods , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Receptors, IgE/antagonists & inhibitors , Animals , B-Lymphocytes/immunology , Cell Line, Tumor , Coculture Techniques , Cytokines/biosynthesis , Cytotoxicity, Immunologic , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression , Humans , Interleukin-2/biosynthesis , Lymphocyte Activation , Mice , Mice, Knockout , Receptors, IgE/genetics , Receptors, IgE/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
17.
Mol Ther ; 17(5): 880-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19259067

ABSTRACT

Adoptive transfer of antigen-specific cytotoxic T lymphocytes (CTLs) can induce objective clinical responses in patients with malignant diseases. The option of providing a proliferative and survival advantage to adoptively transferred CTLs remains a challenge to improve their efficacy. Host lymphodepletion and administration of recombinant interleukin-2 (IL-2) are currently used to improve CTL survival and expansion after adoptive transfer, but these approaches are frequently associated with significant side effects and may increase proliferation of T regulatory cells. IL-7 is a crucial homeostatic cytokine that has been safely administered as a recombinant protein. However, while IL-7 induces robust expansion of naive and memory T lymphocytes, the lack of expression of the IL-7 receptor alpha chain (IL-7Ralpha) by CTLs precludes their response to this cytokine. We found that CTLs can be genetically modified to re-express IL-7Ralpha, and that this manipulation restores the response of these cells to IL-7 without apparent modification of their antigen specificity or dependency, and without changing their response to other common gamma (gammac) chain cytokines. This approach may allow selective expansion of CTLs without the unwanted effects associated with IL-2.


Subject(s)
Interleukin-7/immunology , Receptors, Interleukin-7/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-2/immunology , Mice , Mice, SCID , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-7/genetics
18.
Blood ; 110(8): 2793-802, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17638856

ABSTRACT

The antitumor effect of adoptively transferred tumor-specific cytotoxic T lymphocytes (CTLs) is impaired by the limited capacity of these cells to expand within the tumor microenvironment. Administration of interleukin 2 (IL-2) has been used to overcome this limitation, but the systemic toxicity and the expansion of unwanted cells, including regulatory T cells, limit the clinical value of this strategy. To discover whether transgenic expression of lymphokines by the CTLs themselves might overcome these limitations, we evaluated the effects of transgenic expression of IL-2 and IL-15 in our model of Epstein Barr Virus-specific CTLs (EBV-CTLs). We found that transgenic expression of IL-2 or IL-15 increased the expansion of EBV-CTLs both in vitro and in vivo in a severe combined immunodeficiency disease (SCID) mouse model and enhanced antitumor activity. Although the proliferation of these cytokine genes transduced CTLs remained strictly antigen dependent, clinical application of this approach likely requires the inclusion of a suicide gene to deal with the potential development of T-cell mutants with autonomous growth. We found that the incorporation of an inducible caspase-9 suicide gene allowed efficient elimination of transgenic CTLs after exposure to a chemical inducer of dimerization, thereby increasing the safety and feasibility of the approach.


Subject(s)
Cytokines/biosynthesis , Cytokines/genetics , Genes, Transgenic, Suicide/physiology , Immunotherapy, Adoptive/methods , T-Lymphocytes, Cytotoxic/physiology , Adoptive Transfer , Animals , Blotting, Western , Cell Survival , Genetic Vectors , Herpesvirus 4, Human , Humans , Immunophenotyping , Interleukin-15/biosynthesis , Interleukin-15/genetics , Interleukin-2/biosynthesis , Interleukin-2/genetics , Lymphocyte Activation/physiology , Mice , Mice, SCID , Transduction, Genetic , Transgenes
19.
Haematologica ; 92(3): 381-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17339188

ABSTRACT

Chimeric T-cell receptors (ChTCR), are a fascinating technological step in the field of immunotherapy for orienting the activity of immune cells towards specific molecular targets expressed on the cell surface of various tumors, including hematologic malignancies. The main characteristics of ChTCR are their ability to redirect T-cell specificity and their killing/effector activity toward a selected target in a non MHC-restricted manner, exploiting the antigen binding properties of monoclonal antibodies. ChTCR are, in fact, artificial T-cell receptors constituted by an antigen-recognizing antibody molecule linked to a T-cell triggering domain. Various hematologic malignancies represent optimal targets for the exploitation of ChTCR, because of the bright expression of specific antigens on the surface of tumor cells. Thus, CD19 and CD20 have been targeted for B-cell lymphoid tumors (acute lymphoblastic leukemia-ALL, lymphomas and chronic lymphocytic leukemia-CLL), CD33 for myeloid leukemia, and CD30 for lymphomas. Even though technical and safety progresses are still needed to improve the profile of gene transfer and protein expression of ChTCR, phase 1 trials will be carried out in the near future to demonstrate the feasibility of their clinical translation and, it is be hoped, give preliminary indications about their anti-tumor efficacy.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy/methods , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , Adult , Animals , Antigens, CD/immunology , Antigens, CD19/immunology , Antigens, CD20/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Child , Clinical Trials as Topic , Drug Delivery Systems , Forecasting , Hematologic Neoplasms/therapy , Humans , Ki-1 Antigen/immunology , Killer Cells, Natural/immunology , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , Sialic Acid Binding Ig-like Lectin 3 , T-Cell Antigen Receptor Specificity , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...