Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 10(5)2022 05.
Article in English | MEDLINE | ID: mdl-35577500

ABSTRACT

Despite promising clinical results in a small subset of malignancies, therapies based on engineered chimeric antigen receptor and T-cell receptor T cells are associated with serious adverse events, including cytokine release syndrome and neurotoxicity. These toxicities are sometimes so severe that they significantly hinder the implementation of this therapeutic strategy. For a long time, existing preclinical models failed to predict severe toxicities seen in human clinical trials after engineered T-cell infusion. However, in recent years, there has been a concerted effort to develop models, including humanized mouse models, which can better recapitulate toxicities observed in patients. The Accelerating Development and Improving Access to CAR and TCR-engineered T cell therapy (T2EVOLVE) consortium is a public-private partnership directed at accelerating the preclinical development and increasing access to engineered T-cell therapy for patients with cancer. A key ambition in T2EVOLVE is to design new models and tools with higher predictive value for clinical safety and efficacy, in order to improve and accelerate the selection of lead T-cell products for clinical translation. Herein, we review existing preclinical models that are used to test the safety of engineered T cells. We will also highlight limitations of these models and propose potential measures to improve them.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Receptors, Chimeric Antigen , Animals , Cytokine Release Syndrome , Humans , Immunotherapy, Adoptive/adverse effects , Mice , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/therapeutic use , T-Lymphocytes
2.
Nat Commun ; 10(1): 2087, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31064990

ABSTRACT

T cells expressing CD19-targeting chimeric antigen receptors (CARs) reveal high efficacy in the treatment of B cell malignancies. Here, we report that T cell receptor fusion constructs (TRuCs) comprising an antibody-based binding domain fused to T cell receptor (TCR) subunits can effectively reprogram an intact TCR complex to recognize tumor surface antigens. Unlike CARs, TRuCs become a functional component of the TCR complex. TRuC-T cells kill tumor cells as potently as second-generation CAR-T cells, but at significant lower cytokine release and despite the absence of an extra co-stimulatory domain. TRuC-T cells demonstrate potent anti-tumor activity in both liquid and solid tumor xenograft models. In several models, TRuC-T cells are more efficacious than respective CAR-T cells. TRuC-T cells are shown to engage the signaling capacity of the entire TCR complex in an HLA-independent manner.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Receptors, Artificial/immunology , Single-Chain Antibodies/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD19/immunology , Antigens, Neoplasm/immunology , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred NOD , Neoplasms/immunology , Primary Cell Culture , Protein Domains , Receptors, Antigen, T-Cell/genetics , Receptors, Artificial/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/genetics , Treatment Outcome , Xenograft Model Antitumor Assays
3.
PLoS One ; 8(3): e59594, 2013.
Article in English | MEDLINE | ID: mdl-23555045

ABSTRACT

The inability to obtain sufficient numbers of transduced cells remains a limitation in gene therapy. One strategy to address this limitation is in vivo pharmacologic selection of transduced cells. We have previously shown that knockdown of HPRT using lentiviral delivered shRNA facilitates efficient selection of transduced murine hematopoietic progenitor cells (HPC) using 6-thioguanine (6TG). Herein, we now extend these studies to human HPC. We tested multiple shRNA constructs in human derived cell lines and identified the optimal shRNA sequence for knockdown of HPRT and 6TG resistance. We then tested this vector in human umbilical cord blood derived HPC in vitro and in NOD/SCID recipients. Knockdown of HPRT effectively provided resistance to 6TG in vitro. 6TG treatment of mice resulted in increased percentages of transduced human CD45(+) cells in the peripheral blood and in the spleen in particular, in both myeloid and lymphoid compartments. 6TG treatment of secondary recipients resulted in higher percentages of transduced human cells in the bone marrow, confirming selection from the progeny of long-term repopulating HPCs. However, the extent of selection of cells in the bone marrow at the doses of 6TG tested and the toxicity of higher doses, suggest that this strategy may be limited to selection of more committed progenitor cells. Together, these data suggest that human HPC can be programmed to be resistant to purine analogs, but that HPRT knockdown/6TG-based selection may not be robust enough for in vivo selection.


Subject(s)
Gene Knockdown Techniques , Hematopoietic Stem Cells/metabolism , Hypoxanthine Phosphoribosyltransferase/deficiency , Hypoxanthine Phosphoribosyltransferase/genetics , Transduction, Genetic/methods , Animals , Antigens, CD34/metabolism , Cell Line , Drug Resistance/drug effects , Drug Resistance/genetics , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/enzymology , Humans , Lymphocytes/cytology , Mice , Myeloid Cells/cytology , Thioguanine/pharmacology , Umbilical Cord/cytology
4.
J Cell Physiol ; 226(5): 1292-307, 2011 May.
Article in English | MEDLINE | ID: mdl-20945395

ABSTRACT

Diabetes mellitus (DM) is a primary risk factor for cardiovascular diseases and heart failure. Activation of the retinoic acid receptor (RAR) and retinoid X receptor (RXR) has an anti-diabetic effect; but, a role in diabetic cardiomyopathy remains unclear. Using neonatal and adult cardiomyocytes, we determined the role of RAR and RXR in hyperglycemia-induced apoptosis and expression of renin-angiotensin system (RAS) components. Decreased nuclear expression of RARα and RXRα, activation of apoptotic signaling and cell apoptosis was observed in high glucose (HG) treated neonatal and adult cardiomyocytes and diabetic hearts in Zucker diabetic fatty (ZDF) rats. HG-induced apoptosis and reactive oxygen species (ROS) generation was prevented by both RAR and RXR agonists. Silencing expression of RARα and RXRα, by small interference RNA, promoted apoptosis under normal conditions and significantly enhanced HG-induced apoptosis, indicating that RARα and RXRα are required in regulating cell apoptotic signaling. Blocking angiotensin type 1 receptor (AT(1) R); but, not AT(2) R, attenuated HG-induced apoptosis and ROS generation. Moreover, HG induced gene expression of angiotensinogen, renin, AT(1) R, and angiotensin II (Ang II) synthesis were inhibited by RARα agonists and promoted by silencing RARα. Activation of RXRα, downregulated the expression of AT(1) R; and RXRα silencing accelerated HG induced expression of angiotensinogen and Ang II synthesis, whereas there was no significant effect on renin gene expression. These results indicate that reduction in the expression of RARα and RXRα has an important role in hyperglycemia mediated apoptosis and expression of RAS components. Activation of RAR/RXR signaling protects cardiomyocytes from hyperglycemia, by reducing oxidative stress and inhibition of the RAS.


Subject(s)
Apoptosis/drug effects , Diabetes Mellitus/drug therapy , Hyperglycemia/drug therapy , Hypoglycemic Agents/pharmacology , Myocytes, Cardiac/drug effects , Receptors, Retinoic Acid/agonists , Renin-Angiotensin System/drug effects , Retinoid X Receptor alpha/agonists , Signal Transduction/drug effects , Tretinoin/pharmacology , Angiotensin II/metabolism , Angiotensin Receptor Antagonists/pharmacology , Animals , Animals, Newborn , Blood Glucose/metabolism , Cells, Cultured , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation , Hyperglycemia/genetics , Hyperglycemia/metabolism , Hyperglycemia/pathology , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , RNA Interference , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Rats, Zucker , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Renin-Angiotensin System/genetics , Retinoic Acid Receptor alpha , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Signal Transduction/genetics , Time Factors
5.
Neoplasia ; 12(3): 224-34, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20234816

ABSTRACT

Work from our laboratory and others has demonstrated that activation of the nuclear receptor peroxisome proliferator-activated receptor-gamma (PPARgamma) inhibits transformed growth of non-small cell lung cancer (NSCLC) cell lines in vitro and in vivo. We have demonstrated that activation of PPARgamma promotes epithelial differentiation of NSCLC by increasing expression of E-cadherin, as well as inhibiting expression of COX-2 and nuclear factor-kappaB. The Snail family of transcription factors, which includes Snail (Snail1), Slug (Snail2), and ZEB1, is an important regulator of epithelial-mesenchymal transition, as well as cell survival. The goal of this study was to determine whether the biological responses to rosiglitazone, a member of the thiazolidinedione family of PPARgamma activators, are mediated through the regulation of Snail family members. Our results indicate that, in two independent NSCLC cell lines, rosiglitazone specifically decreased expression of Snail, with no significant effect on either Slug or ZEB1. Suppression of Snail using short hairpin RNA silencing mimicked the effects of PPARgamma activation, in inhibiting anchorage-independent growth, promoting acinar formation in three-dimensional culture, and inhibiting invasiveness. This was associated with the increased expression of E-cadherin and decreased expression of COX-2 and matrix metaloproteinases. Conversely, overexpression of Snail blocked the biological responses to rosiglitazone, increasing anchorage-independent growth, invasiveness, and promoting epithelial-mesenchymal transition. The suppression of Snail expression by rosiglitazone seemed to be independent of GSK-3 signaling but was rather mediated through suppression of extracellular signal-regulated kinase activity. These findings suggest that selective regulation of Snail may be critical in mediating the antitumorigenic effects of PPARgamma activators.


Subject(s)
Carcinoma, Non-Small-Cell Lung/prevention & control , Cell Proliferation , Lung Neoplasms/prevention & control , PPAR gamma/pharmacology , Transcription Factors/antagonists & inhibitors , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/prevention & control , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Hypoglycemic Agents/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Matrix Metalloproteinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Signal Transduction , Snail Family Transcription Factors , Thiazolidinediones/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Cancer Res ; 69(5): 1733-8, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19208832

ABSTRACT

Cancer progression and metastasis involves interactions between tumor cells and the tumor microenvironment (TME). We reported that mice deficient for cytosolic phospholipase A(2) (cPLA(2)-KO) are protected against the development of lung tumors. The goal of this study was to examine the role of cPLA(2) in the TME. Mouse lung cancer cells (CMT167 and Lewis lung carcinoma cells) injected directly into lungs of syngeneic mice formed a primary tumor, and then metastasized to other lobes of the lung and to the mediastinal lymph nodes. Identical cells injected into cPLA(2)-KO mice showed a dramatic decrease in the numbers of secondary metastatic tumors. This was associated with decreased macrophage staining surrounding the tumor. Wild-type mice transplanted with cPLA(2)-KO bone marrow had a marked survival advantage after inoculation with tumor cells compared with mice receiving wild-type (WT) bone marrow. In vitro, coculturing CMT167 cells with bone marrow-derived macrophages from WT mice increased production of interleukin 6 (IL-6) by cancer cells. This increase was blocked in cocultures using cPLA(2)-KO macrophages. Correspondingly, IL-6 staining was decreased in tumors grown in cPLA(2)-KO mice. These data suggest that stromal cPLA(2) plays a critical role in tumor progression by altering tumor-macrophage interactions and cytokine production.


Subject(s)
Bone Marrow Cells/enzymology , Lung Neoplasms/prevention & control , Macrophages/enzymology , Phospholipases A2, Cytosolic/physiology , Animals , Cell Line, Tumor , Dinoprostone/physiology , Disease Progression , Female , Humans , Interleukin-6/biosynthesis , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis
7.
J Cell Physiol ; 215(1): 172-81, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17941088

ABSTRACT

Cardiomyocyte apoptosis has an important role in the transition from compensatory cardiac remodeling to heart failure. All-trans retinoic acid (RA), a bioactive vitamin A derivative, prevents stretch- and angiotensin II (Ang II)-induced cardiac hypertrophy. However, the anti-apoptotic potential of RA in the heart remains unexplored. Here, we demonstrate that stretch- and Ang II-induced apoptosis is prevented by RA in neonatal cardiomyocytes. RA improved mitochondrial function by inhibiting the stretch- and Ang II-induced reduction in mitochondrial membrane potential, cytochrome c release and by increasing the Bcl2/Bax ratio. RA inhibited stretch- and Ang II-induced intracellular reactive oxygen species (ROS) generation and upregulated the SOD2 level. Hydrogen peroxide-induced increases in the number of TUNEL-positive cells and percentage of Annexin V positive cells, were dose-dependently inhibited by RA. The thiol antioxidant, N-acetyl cysteine (NAC), completely inhibited stretch- and Ang II-induced apoptosis. Using diazoxide (mitochondrial ATP-sensitive K(+) channel opener) and SDS (NADPH oxidase activator), we confirmed that RA suppressed both mitochondrial- and NADPH oxidase-derived ROS. We also observed that both RAR and RXR were involved in preventing Ang II- and stretch-induced ROS production and apoptosis, by using selective retinoid receptor agonists and antagonists. Our data provide the first evidence that RA prevents Ang II and stretch induced apoptosis, by inhibiting ROS generation and increasing the anti-oxidant defense system, suggesting that RA-mediated signaling may provide a new therapeutic target for the prevention of the cardiac remodeling process.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin II/pharmacology , Apoptosis/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Reactive Oxygen Species/metabolism , Tretinoin/pharmacology , Animals , Cells, Cultured , Cytoprotection/drug effects , Hydrogen Peroxide/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , NADPH Oxidases/antagonists & inhibitors , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Retinoid X Receptors/metabolism , Stress, Mechanical
8.
Am J Physiol Heart Circ Physiol ; 294(2): H633-44, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18156191

ABSTRACT

This study was designed to determine the effect of all-trans retinoic acid (RA) on the development of cardiac remodeling in a pressure overload rat model. Male Sprague-Dawley rats were subjected to sham operation and the aortic constriction procedure. A subgroup of sham control and aortic constricted rats were treated with RA for 5 mo after surgery. Pressure-overloaded rats showed significantly increased interstitial and perivascular fibrosis, heart weight-to-body weight ratio, and gene expression of atrial natriuretic peptide and brain natriuretic peptide. Echocardiographic analysis showed that pressure overload induced systolic and diastolic dysfunction, as evidenced by decreased fractional shortening, ejection fraction, stroke volume, and increased E-to-E(a) ratio and isovolumic relaxation time. RA treatment prevented the above changes in cardiac structure and function and hypertrophic gene expression in pressure-overloaded rats. RA restored the ratio of Bcl-2 to Bax, inhibited cleavage of caspase-3 and -9, and prevented the decreases in the levels of SOD-1 and SOD-2. Pressure overload-induced phosphorylation of ERK1/2, JNK, and p38 was inhibited by RA, via upregulation of mitogen-activated protein kinase phosphatase (MKP)-1 and MKP-2. The pressure overload-induced production of angiotensin II was inhibited by RA via upregulation of expression of angiotensin-converting enzyme (ACE)2 and through inhibition of the expression of cardiac and renal renin, angiotensinogen, ACE, and angiotensin type 1 receptor. Similar results were observed in cultured neonatal cardiomyocytes in response to static stretch. These results demonstrate that RA has a significant inhibitory effect on pressure overload-induced cardiac remodeling, through inhibition of the expression of renin-angiotensin system components.


Subject(s)
Aorta, Thoracic/physiology , Heart/drug effects , Renin-Angiotensin System/drug effects , Tretinoin/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Atrial Natriuretic Factor/pharmacology , Blood Pressure/drug effects , Blotting, Western , Cells, Cultured , Fibrosis/prevention & control , Heart Rate/drug effects , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/prevention & control , Ligation , Male , Mitogen-Activated Protein Kinases/physiology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Natriuretic Peptide, Brain/pharmacology , Physical Stimulation , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tretinoin/therapeutic use , Ultrasonography
9.
Mol Cell Biochem ; 283(1-2): 115-22, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16444593

ABSTRACT

In view of the known involvement of oxidative stress and calcineurin (Ca(2+)-calmodulin dependent protein phosphatase) in beta-Adrenergic stimulated events, we examined the influence of eugenol (an antioxidant generally regarded as safe by the Food and Agricultural Organization of the United Nations) on isoproterenol-induced apoptosis in neonatal cardiomyocytes. In comparison to unstimulated controls, cardiomyocytes stimulated with 50 microM isoproterenol for 48 h demonstrated (a) increased intracellular Ca(2+) levels (b) oxidative stress involving enhanced reactive oxygen species, decreased GSH/GSSG ratio, enhanced lipid peroxidation, increased activities of superoxide dismutase and glutathione peroxidase (c) apoptosis, evidenced by increased number of annexin V/TUNEL positive cells, enhanced membrane fluidity, decreased mitochondrial membrane potential, increased activities of caspase 3 and 9 along with (d) increased calcineurin activity. Pre-incubation of cardiomyocytes with 100 microM eugenol for 1 h, followed by isoproterenol treatment for 48 h, led to reversal of enhanced intracellular Ca(2+) levels, oxidative stress, calcineurin activation and apoptosis caused by isoproterenol. In addition, similar treatment of cardiomyocytes with 10 nM FK506, a calcineurin inhibitor, could also attenuate isoproterenol-induced apoptosis. These results indicate the beneficial effects of eugenol in preventing cardiomyocyte apoptosis.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Calcineurin/metabolism , Eugenol/pharmacology , Myocytes, Cardiac/pathology , Oxidative Stress/physiology , Animals , Animals, Newborn , Calcium/metabolism , Cardiotonic Agents/pharmacology , Caspases/metabolism , Cells, Cultured , Glutathione Peroxidase/metabolism , Isoproterenol/pharmacology , Lipid Peroxidation/drug effects , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/drug effects , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
10.
Indian J Clin Biochem ; 21(2): 107-13, 2006 Sep.
Article in English | MEDLINE | ID: mdl-23105625

ABSTRACT

Recent reports on the involvement of calcineurin in cardiac hypertrophy and its susceptibility to free radicals, prompted us to examine possible beneficial effects of dietary antioxidants in this regard. In continuation of initialin vitro studies revealing eugenol to be a potent calcineurin inhibitor, we investigated its ability to reverse isoproterenol-induced cardiac hypertrophy in rats. Intraperitoneal administration of isoproterenol (1 mg/kg body wt/day for 10 days) induced cardiac hypertrophy with increased heart weight and enhanced apoptosis of myocytes concomitant with accumulation of reactive oxygen species, decreased glutathione contents, increased activities of calcineurin and protein kinase C in ventricular tissue. Administering eugenol for 3 days (1 mg/kg body wt/twice a day), followed by combined administration of isoproterenol and eugenol resulted in significant reversal of cardiac hypertrophy and restoration of above changes. These results suggest that eugenol, a natural antioxidant of dietary origin, may offer potential benefits in the management of cardiac hypertrophy.

11.
Int J Cardiol ; 105(3): 327-31, 2005 Dec 07.
Article in English | MEDLINE | ID: mdl-16274778

ABSTRACT

BACKGROUND: Among various intracellular signaling cascades associated with cardiac hypertrophy, the involvement of calcineurin (CaN; Ca2+-calmodulin dependent protein phosphatase) is gaining credence because of its enhanced activity in ventricular myocardium and the ability of CaN inhibitors to prevent pressure-overload hypertrophy. Since our recent findings attribute clinical significance to serum CaN, the present investigation was conducted to evaluate its significance in cardiac hypertrophy. METHODS: The study group comprised of patients diagnosed for hypertensive hypertrophy, hypertrophic cardiomyopathy, chronic coronary artery disease with compensatory left ventricular hypertrophy, dilated cardiomyopathy and acute myocardial infarction. Serum contents of CaN and calmodulin were determined and activities of CaN as well as of acid and alkaline phosphatases were assayed and correlated with 2D echocardiography findings. The results were compared with those obtained from age-matched healthy volunteers. RESULTS: Serum CaN activity, but not of acid or alkaline phosphatases, was significantly enhanced by 2-fold in hypertensive hypertrophy, 3-fold in hypertrophic cardiomyopathy and 3.75-fold in chronic coronary artery disease associated with left ventricular hypertrophy, unaccompanied by changes in serum contents of calmodulin and CaN. No such increases were observed in acute myocardial infarction and dilated cardiomyopathy. CONCLUSIONS: Positive correlations observed between serum CaN activity and enhanced left ventricular mass in cardiac hypertrophy suggest that assaying serum CaN activity may be useful in the diagnosis and management of left ventricular hypertrophy.


Subject(s)
Calcineurin/blood , Hypertrophy, Left Ventricular/blood , Adult , Aged , Female , Humans , Hypertrophy, Left Ventricular/pathology , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...