Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 29
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Vaccines (Basel) ; 12(7)2024 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-39066446

RÉSUMÉ

The human epidermal growth factor receptor 2 (HER2) is a transmembrane tyrosine kinase receptor and tumor-associated antigen abnormally expressed in various types of cancer, including breast, ovarian, and gastric cancer. HER2 overexpression is highly correlated with increased tumor aggressiveness, poorer prognosis, and shorter overall survival. Consequently, multiple HER2-targeted therapies have been developed and approved; however, only a subset of patients benefit from these treatments, and relapses are common. More potent and durable HER2-targeted therapies are desperately needed for patients with HER2-positive cancers. In this study, we developed a lipid nanoparticle (LNP)-based therapy formulated with mRNA encoding a novel HER2-CD3-Fc bispecific antibody (bsAb) for HER2-positive cancers. The LNPs efficiently transfected various types of cells, such as HEK293S, SKOV-3, and A1847, leading to robust and sustained secretion of the HER2-CD3-Fc bsAb with high binding affinity to both HER2 and CD3. The bsAb induced potent T-cell-directed cytotoxicity, along with secretion of IFN-λ, TNF-α, and granzyme B, against various types of HER2-positive tumor cells in vitro, including A549, NCI-H460, SKOV-3, A1847, SKBR3, and MDA-MB-231. The bsAb-mediated antitumor effect is highly specific and strictly dependent on its binding to HER2, as evidenced by the gained resistance of A549 and A1847 her2 knockout cells and the acquired sensitivity of mouse 4T1 cells overexpressing the human HER2 extracellular domain (ECD) or epitope-containing subdomain IV to the bsAb-induced T cell cytotoxicity. The bsAb also relies on its binding to CD3 for T-cell recruitment, as ablation of CD3 binding abolished the bsAb's ability to elicit antitumor activity. Importantly, intratumoral injection of the HER2-CD3-Fc mRNA-LNPs triggers a strong antitumor response and completely blocks HER2-positive tumor growth in a mouse xenograft model of human ovarian cancer. These results indicate that the novel HER2-CD3-Fc mRNA-LNP-based therapy has the potential to effectively treat HER2-positive cancer.

2.
Int J Mol Sci ; 24(17)2023 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-37686170

RÉSUMÉ

Natural killer (NK) cells are cytotoxic lymphocytes that are critical for the innate immune system. Engineering NK cells with chimeric antigen receptors (CARs) allows CAR-NK cells to target tumor antigens more effectively. In this report, we present novel CAR mRNA-LNP (lipid nanoparticle) technology to effectively transfect NK cells expanded from primary PBMCs and to generate functional CAR-NK cells. CD19-CAR mRNA and BCMA-CAR mRNA were embedded into LNPs that resulted in 78% and 95% CAR expression in NK cells, respectively. BCMA-CAR-NK cells after transfection with CAR mRNA-LNPs killed multiple myeloma RPMI8226 and MM1S cells and secreted IFN-gamma and Granzyme B in a dose-dependent manner in vitro. In addition, CD19-CAR-NK cells generated with CAR mRNA-LNPs killed Daudi and Nalm-6 cells and secreted IFN-gamma and Granzyme B in a dose-dependent manner. Both BCMA-CAR-NK and CD19-CAR-NK cells showed significantly higher cytotoxicity, IFN-gamma, and Granzyme B secretion compared with normal NK cells. Moreover, CD19-CAR-NK cells significantly blocked Nalm-6 tumor growth in vivo. Thus, non-viral delivery of CAR mRNA-LNPs can be used to generate functional CAR-NK cells with high anti-tumor activity.


Sujet(s)
Myélome multiple , Récepteurs chimériques pour l'antigène , Humains , Récepteurs chimériques pour l'antigène/génétique , Granzymes/génétique , Antigène de maturation des cellules B , Cellules tueuses naturelles , Myélome multiple/génétique , Myélome multiple/thérapie , Protéines adaptatrices de la transduction du signal , Antigènes CD19
3.
Cancers (Basel) ; 15(10)2023 May 22.
Article de Anglais | MEDLINE | ID: mdl-37345198

RÉSUMÉ

The epithelial cell adhesion molecule (EpCAM) is often overexpressed in many types of tumors, including colorectal cancer. We sequenced and humanized an EpCAM mouse antibody and used it to develop bispecific EpCAM-CD3 antibodies. Three different designs were used to generate bispecific antibodies such as EpCAM-CD3 CrossMab knob-in-hole, EpCAM ScFv-CD3 ScFv (BITE), and EpCAM ScFv-CD3 ScFv-human Fc designs. These antibody designs showed strong and specific binding to the EpCAM-positive Lovo cell line and T cells, specifically killed EpCAM-positive Lovo cells and not EpCAM-negative Colo741 cells in the presence of T cells, and increased T cells' IFN-gamma secretion in a dose-dependent manner. In addition, transfection of HEK-293 cells with EpCAM ScFv-CD3 ScFv human Fc mRNA-LNPs resulted in antibody secretion that killed Lovo cells and did not kill EpCAM-negative Colo741 cells. The antibody increased IFN-gamma secretion against Lovo target cells and did not increase it against Colo741 target cells. EpCAM-CD3 hFc mRNA-LNP transfection of several cancer cell lines (A1847, C30, OVCAR-5) also demonstrated functional bispecific antibody secretion. In addition, intratumoral delivery of the EpCAM-CD3 human Fc mRNA-LNPs into OVCAR-5 tumor xenografts combined with intravenous injection of T cells significantly blocked xenograft tumor growth. Thus, EpCAM-CD3 hFc mRNA-LNP delivery to tumor cells shows strong potential for future clinical studies.

4.
Biomedicines ; 9(10)2021 Oct 09.
Article de Anglais | MEDLINE | ID: mdl-34680541

RÉSUMÉ

Multiple myeloma (MM) is a hematological cancer caused by abnormal proliferation of plasma cells in the bone marrow, and novel types of treatment are needed for this deadly disease. In this study, we aimed to develop novel CS1 CAR-T cells and bispecific CS1-BCMA CAR-T cells to specifically target multiple myeloma. We generated a new CS1 (CD319, SLAM-7) antibody, clone (7A8D5), which specifically recognized the CS1 antigen, and we applied it for the generation of CS1-CAR. CS1-CAR-T cells caused specific killing of CHO-CS1 target cells with secretion of IFN-gamma and targeted multiple myeloma cells. In addition, bispecific CS1-BCMA-41BB-CD3 CAR-T cells effectively killed CHO-CS1 and CHO-BCMA target cells, killed CS1/BCMA-positive multiple myeloma cells, and secreted IFN-gamma. Moreover, CS1-CAR-T cells and bispecific CS1-BCMA CAR-T cells effectively blocked MM1S multiple myeloma tumor growth in vivo. These data for the first time demonstrate that novel CS1 and bispecific CS1-BCMA-CAR-T cells are effective in targeting MM cells and provide a basis for future clinical trials.

5.
Cancers (Basel) ; 13(5)2021 Feb 26.
Article de Anglais | MEDLINE | ID: mdl-33652767

RÉSUMÉ

CD19 and CD37 proteins are highly expressed in B-cell lymphoma and have been successfully targeted with different monotherapies, including chimeric antigen receptor (CAR)-T cell therapy. The goal of this study was to target lymphoma with novel CD37, humanized CD37, and bi-specific humanized CD37-CD19 CAR-T cells. A novel mouse monoclonal anti-human CD37 antibody (clone 2B8D12F2D4) was generated with high binding affinity for CD37 antigen (KD = 1.6 nM). The CD37 antibody specifically recognized cell surface CD37 protein in lymphoma cells and not in multiple myeloma or other types of cancer. The mouse and humanized CD37-CAR-T cells specifically killed Raji and CHO-CD37 cells and secreted IFN-gamma. In addition, we generated bi-specific humanized hCD37-CD19 CAR-T cells that specifically killed Raji cells, CHO-CD37, and Hela-CD19 cells and did not kill control CHO or Hela cells. Moreover, the hCD37-CD19 CAR-T cells secreted IFN-gamma against CD37-positive and CD19-positive target CHO-CD37, Hela-CD19 cells, respectively, but not against CD19 and CD37-negative parental cell line. The bi-specific hCD37-CD19 significantly inhibited Raji xenograft tumor growth and prolonged mouse survival in NOD scid gamma mouse (NSG) mouse model. This study demonstrates that novel humanized CD37 and humanized CD37-CD19 CAR-T cells specifically targeted either CD37 positive or CD37 and CD19-positive cells and provides a basis for future clinical studies.

6.
Front Biosci (Landmark Ed) ; 25(9): 1765-1786, 2020 06 01.
Article de Anglais | MEDLINE | ID: mdl-32472757

RÉSUMÉ

Placental alkaline phosphatase, PLAP encoded by ALPP gene in humans is mainly expressed in placenta and testis, and not expressed in any other normal tissues. PLAP is overexpressed in colorectal cancers which makes it an attractive target for CAR (chimeric antigen receptor)-T cell therapy. PLAP mRNA expression was detected in 21.5% (25 out of 116) of colorectal cancer cell lines and this expression was confirmed by FACS at the protein level. In addition, IHC staining on primary colorectal cancer tumors demonstrated PLAP expression in >20% of colorectal cancer tumors. We generated mouse and humanized PLAP ScFv-CAR-T cells and demonstrated high specificity against PLAP-positive colon cancer cells using RTCA (real-time cytotoxicity assay) and IFN-gamma secretion. In addition, humanized-CAR-T cells significantly decreased Lovo xenograft tumor growth in vivo. The combination of hPLAP-CAR-T cells with PD-1, PD-L1 or LAG-3 checkpoint inhibitors significantly increased the activity of hPLAP-CAR-T cells. This study demonstrates ability of novel PLAP-CAR-T cells to kill colorectal cancers and that the extent of killing can be increased by combination with checkpoint inhibitors.


Sujet(s)
Phosphatase alcaline/immunologie , Tumeurs du côlon/immunologie , Isoenzymes/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Anticorps à chaîne unique/immunologie , Lymphocytes T/immunologie , Phosphatase alcaline/antagonistes et inhibiteurs , Phosphatase alcaline/métabolisme , Animaux , Cellules Caco-2 , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/immunologie , Tumeurs du côlon/métabolisme , Tumeurs du côlon/thérapie , Protéines liées au GPI/antagonistes et inhibiteurs , Protéines liées au GPI/immunologie , Protéines liées au GPI/métabolisme , Cellules HCT116 , Cellules HEK293 , Cellules HT29 , Humains , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Immunothérapie adoptive/méthodes , Interféron gamma/immunologie , Interféron gamma/métabolisme , Isoenzymes/antagonistes et inhibiteurs , Isoenzymes/métabolisme , Mâle , Souris de lignée NOD , Souris knockout , Souris SCID , Récepteurs chimériques pour l'antigène/métabolisme , Anticorps à chaîne unique/métabolisme , Lymphocytes T/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
7.
Front Biosci (Landmark Ed) ; 25(2): 270-282, 2020 01 01.
Article de Anglais | MEDLINE | ID: mdl-31585889

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell immunotherapy has demonstrated clinical success in treatment of B-cell hematologic cancers. In this study, we compared human Transferrin epitope tagged CAR-T cells with non-tagged CAR-T cells for cytotoxicity, IFN-gamma secretion and tumor clearance in NSG mice. CD19-TF-CAR-T cells had similar cytotoxicity in vitro to CD19-CAR-T cells against cells expressing CD19 antigen: exogenously CD19+ Hela cells and endogenously CD19+ Raji cells. In addition, CD22-TF CAR-T cells were similarly cytotoxic against CD22+ CHO cells and CD22+  Raji cells. Both CD19-TF or CD22-TF-CAR-T cells secreted less IFN-gamma as compared to non-tagged CAR-T cells. In a Raji xenograft NSG mouse model, CD19-TF-CAR-T cells were as effective as CD19-CAR-T cells in reducing tumor growth and extending mouse survival. The results show that CD19-TF-CAR-T cells can be monitored using TF antibody in vitro and ex vivo, and that these cells effectively killed Raji cells in vitro and in vivo with reduced secretion of IFN-gamma. Thus, these TF-tagged CAR-T cells might have improved safety and provide a basis for future clinical studies.


Sujet(s)
Antigènes CD19/immunologie , Épitopes/immunologie , Immunothérapie adoptive/méthodes , Lymphomes/thérapie , Récepteurs chimériques pour l'antigène/immunologie , Transferrine/immunologie , Animaux , Antigènes CD19/génétique , Antigènes CD19/métabolisme , Cellules CHO , Lignée cellulaire tumorale , Cricetinae , Cricetulus , Cytotoxicité immunologique/immunologie , Épitopes/génétique , Épitopes/métabolisme , Cellules HeLa , Humains , Lymphomes/immunologie , Lymphomes/métabolisme , Souris de lignée NOD , Souris knockout , Souris SCID , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Transferrine/génétique , Transferrine/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
8.
Cancers (Basel) ; 12(1)2019 Dec 23.
Article de Anglais | MEDLINE | ID: mdl-31878090

RÉSUMÉ

CAR-T (chimeric antigen receptor T cells) immunotherapy is effective in many hematological cancers; however, efficacy in solid tumors is disappointing. Doublecortin-like kinase 1 (DCLK1) labels tumor stem cells (TSCs) in genetic mouse models of colorectal cancer (CRC). Here, we describe a novel CAR-T targeting DCLK1 (CBT-511; with our proprietary DCLK1 single-chain antibody variable fragment) as a treatment strategy to eradicate CRC TSCs. The cell surface expression of DCLK1 and cytotoxicity of CBT-511 were assessed in CRC cells (HT29, HCT116, and LoVo). LoVo-derived tumor xenografts in NOD Scid gamma (NSGTM)mice were treated with CBT-511 or mock CAR-T cells. Adherent CRC cells express surface DCLK1 (two-dimensional, 2D). A 4.5-fold increase in surface DCLK1 was observed when HT29 cells were grown as spheroids (three-dimensional, 3D). CBT-511 induced cytotoxicity (2D; p < 0.0001), and increased Interferon gamma (IFN-γ) release in CRC cells (2D) compared to mock CAR-T (p < 0.0001). Moreover, an even greater increase in IFN-γ release was observed when cells were grown in 3D. CBT-511 reduced tumor growth by approximately 50 percent compared to mock CAR-T. These data suggest that CRC cells with increased clonogenic capacity express increased surface DCLK1. A DCLK1-targeted CAR-T can induce cytotoxicity in vitro and inhibit xenograft growth in vivo.

9.
J Vis Exp ; (153)2019 11 12.
Article de Anglais | MEDLINE | ID: mdl-31789308

RÉSUMÉ

Chimeric antigen receptor (CAR) T-cell therapy for cancer has achieved significant clinical benefit for resistant and refractory hematological malignancies such as childhood acute lymphocytic leukemia. Efforts are currently underway to extend this promising therapy to solid tumors in addition to other hematological cancers. Here, we describe the development and production of potent CAR T cells targeting antigens with unique or preferential expression on solid and liquid tumor cells. The in vitro potency of these CAR T cells is then evaluated in real-time using the highly sensitive impedance-based xCELLigence assay. Specifically, the impact of different costimulatory signaling domains, such as glucocorticoid-induced tumor necrosis factor receptor (TNFR)-related protein (GITR), on the in vitro potency of CAR T cells is examined. This report includes protocols for: generating CAR T cells for preclinical studies using lentiviral gene transduction, expanding CAR T cells, validating CAR expression, and running and analyzing xCELLigence potency assays.


Sujet(s)
Apoptose , Lymphomes/anatomopathologie , Tumeurs du pancréas/anatomopathologie , Récepteurs chimériques pour l'antigène/métabolisme , Lymphocytes T/anatomopathologie , Humains , Lymphomes/immunologie , Lymphomes/métabolisme , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Cellules cancéreuses en culture
10.
Cancers (Basel) ; 11(5)2019 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-31052261

RÉSUMÉ

Hypoxia is a major characteristic of the solid tumor microenvironment. To understand how chimeric antigen receptor-T cells (CAR-T cells) function in hypoxic conditions, we characterized CD19-specific and BCMA-specific human CAR-T cells generated in atmospheric (18% oxygen) and hypoxic (1% oxygen) culture for expansion, differentiation status, and CD4:CD8 ratio. CAR-T cells expanded to a much lower extent in 1% oxygen than in 18% oxygen. Hypoxic CAR-T cells also had a less differentiated phenotype and a higher CD4:CD8 ratio than atmospheric CAR-T cells. CAR-T cells were then added to antigen-positive and antigen-negative tumor cell lines at the same or lower oxygen level and characterized for cytotoxicity, cytokine and granzyme B secretion, and PD-1 upregulation. Atmospheric and hypoxic CAR-T cells exhibited comparable cytolytic activity and PD-1 upregulation; however, cytokine production and granzyme B release were greatly decreased in 1% oxygen, even when the CAR-T cells were generated in atmospheric culture. Together, these data show that at solid tumor oxygen levels, CAR-T cells are impaired in expansion, differentiation and cytokine production. These effects may contribute to the inability of CAR-T cells to eradicate solid tumors seen in many patients.

11.
Methods Mol Biol ; 1884: 349-360, 2019.
Article de Anglais | MEDLINE | ID: mdl-30465215

RÉSUMÉ

T cells engineered with chimeric antigen receptors (CARs) are emerging as powerful cancer immunotherapies. Remarkable efficacies have been demonstrated in treating B-cell malignancies with CAR-T cells, leading to the FDA's first approval of gene therapy. Currently, numerous clinical trials for hematological malignancies and solid tumors are underway worldwide. Production of CAR-T cells with proper qualities is essential for CAR-T success in vivo. Here we detail optimized protocols for the generation of CAR-T cells for preclinical studies using lentiviral gene transfer, expansion of CAR-T cells in culture, detection of CAR expression, and evaluation of CAR-T cellular cytotoxicity in vitro.


Sujet(s)
Ingénierie cellulaire/méthodes , Immunothérapie adoptive/méthodes , Tumeurs/thérapie , Récepteurs chimériques pour l'antigène/immunologie , Lymphocytes T/immunologie , Ingénierie cellulaire/instrumentation , Tests de cytotoxicité immunologique/instrumentation , Tests de cytotoxicité immunologique/méthodes , Cytométrie en flux/instrumentation , Cytométrie en flux/méthodes , Vecteurs génétiques/génétique , Cellules HEK293 , Humains , Lentivirus/génétique , Tumeurs/immunologie , Récepteurs chimériques pour l'antigène/génétique , Lymphocytes T/métabolisme , Transduction génétique/instrumentation , Transduction génétique/méthodes
12.
Cancers (Basel) ; 10(9)2018 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-30208593

RÉSUMÉ

The cell-surface protein B cell maturation antigen (BCMA, CD269) has emerged as a promising target for CAR-T cell therapy for multiple myeloma. In order to create a novel BCMA CAR, we generated a new BCMA monoclonal antibody, clone 4C8A. This antibody exhibited strong and selective binding to human BCMA. BCMA CAR-T cells containing the 4C8A scFv were readily detected with recombinant BCMA protein by flow cytometry. The cells were cytolytic for RPMI8226, H929, and MM1S multiple myeloma cells and secreted high levels of IFN-γ in vitro. BCMA-dependent cytotoxicity and IFN-γ secretion were also observed in response to CHO (Chinese Hamster Ovary)-BCMA cells but not to parental CHO cells. In a mouse subcutaneous tumor model, BCMA CAR-T cells significantly blocked RPMI8226 tumor formation. When BCMA CAR-T cells were given to mice with established RPMI8226 tumors, the tumors experienced significant shrinkage due to CAR-T cell activity and tumor cell apoptosis. The same effect was observed with 3 humanized BCMA-CAR-T cells in vivo. These data indicate that novel CAR-T cells utilizing the BCMA 4C8A scFv are effective against multiple myeloma and warrant future clinical development.

13.
Front Biosci (Landmark Ed) ; 23(12): 2245-2254, 2018 06 01.
Article de Anglais | MEDLINE | ID: mdl-29772559

RÉSUMÉ

T cells expressing Chimeric antigen receptors or CAR-T cells are used as a novel treatment against hematological and solid cancers. In this report, we designed CAR with glucocorticoid-induced TNFR-related protein (GITR) co-stimulatory domain to study its ability to co-activate CAR-T cells. EGFR-GITR-CD3 CAR-T cells were cytotoxic against EGFR-positive: pancreatic and ovarian cancer cells but not against EGFR-negative cancer cells. The cytotoxic activity of EGFR-GITR-CD3 CAR-T cells was comparable or better than EGFR-28-CD3 or EGFR-41BB-CD3 CAR-T cells. We designed also EGFR-CD3-GITR-CAR and EGFR-ΔGITR-CD3 with deleted 184-192 amino-acids of co-stimulatory GITR domain, and showed that EGFR-GITR-CD3 had significantly higher cytotoxic activity against EGFR-positive cells. The EGFR-GITR-CD3 cells secreted significantly higher levels of IFN-gamma than EGFR-CD3-GITR and EGFR-ΔGITR-CD3 cells. In addition, Mesothelin-GITR-CD3 CAR-T cells also killed mesothelin-positive ovarian cancer cell lines, and pancreatic cancer cells. Moreover, CD19-GITR-CD3 CAR-T cells had significant cytotoxic activity against CD19-positive cancer cells in vitro and in Raji xenograft tumors in vivo. Thus, our results clearly show that GITR co-stimulatory domain can be used as a novel co-stimulatory domain in CAR-T cells.


Sujet(s)
Cytotoxicité immunologique/immunologie , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Tumeurs/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Lymphocytes T/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes , Animaux , Antigènes CD19/génétique , Antigènes CD19/immunologie , Antigènes CD19/métabolisme , Antigènes CD3/génétique , Antigènes CD3/immunologie , Antigènes CD3/métabolisme , Lignée cellulaire tumorale , Protéine associée au récepteur du TNF induit par les corticoïdes/génétique , Protéine associée au récepteur du TNF induit par les corticoïdes/métabolisme , Cellules HEK293 , Humains , Cellules MCF-7 , Mâle , Mésothéline , Souris de lignée NOD , Souris knockout , Souris SCID , Tumeurs/anatomopathologie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Lymphocytes T/métabolisme
14.
Cancers (Basel) ; 9(10)2017 Oct 21.
Article de Anglais | MEDLINE | ID: mdl-29065481

RÉSUMÉ

CD47 is a glycoprotein of the immunoglobulin superfamily that is often overexpressed in different types of hematological and solid cancer tumors and plays important role in blocking phagocytosis, increased tumor survival, metastasis and angiogenesis. In the present report, we designed CAR (chimeric antigen receptor)-T cells that bind CD47 antigen. We used ScFv (single chain variable fragment) from mouse CD47 antibody to generate CD47-CAR-T cells for targeting different cancer cell lines. CD47-CAR-T cells effectively killed ovarian, pancreatic and other cancer cells and produced high level of cytokines that correlated with expression of CD47 antigen. In addition, CD47-CAR-T cells significantly blocked BxPC3 pancreatic xenograft tumor growth after intratumoral injection into NSG mice. Moreover, we humanized mouse CD47 ScFv and showed that it effectively bound CD47 antigen. The humanized CD47-CAR-T cells also specifically killed ovarian, pancreatic, and cervical cancer cell lines and produced IL-2 that correlated with expression of CD47. Thus, CD47-CAR-T cells can be used as a novel cellular therapeutic agent for treating different types of cancer.

15.
Front Biosci (Landmark Ed) ; 22(10): 1644-1654, 2017 06 01.
Article de Anglais | MEDLINE | ID: mdl-28410137

RÉSUMÉ

Autologous T cells expressing chimeric antigen receptors (CARs) specific for CD19 have demonstrated remarkable efficacy as therapeutics for B cell malignancies. In the present study, we generated FLAG-tagged CD19-specific CAR-T cells (CD19-FLAG) and compared them to their non-tagged counterparts for their effects on solid and hematological cancer cells in vitro and in vivo. For solid tumors, we used HeLa cervical carcinoma cells engineered to overexpress CD19 (HeLa-CD19), and for hematological cancer we used Raji Burkitt's lymphoma cells, which endogenously express CD19. Like non-tagged CD19 CAR-T cells, CD19-FLAG CAR-T cells expanded in culture >100-fold and exhibited potent cytolytic activity against both HeLa-CD19 and Raji cells in vitro. CD19-FLAG CAR-T cells also secreted significantly more IFN-gamma and IL-2 than the control T cells. In vivo, CD19-FLAG CAR-T cells significantly blocked the growth of HeLa-CD19 solid tumors, increased tumor cleaved caspase-3 levels, and expanded systemically. CD19-FLAG CAR-T cells also significantly reduced Raji tumor burden and extended mouse survival. These results demonstrate the strong efficacy of FLAG-tagged CD19 CAR-T cells in solid and hematological cancer models.


Sujet(s)
Antigènes CD19/immunologie , Tumeurs/immunologie , Oligopeptides/immunologie , Lymphocytes T/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes , Animaux , Antigènes CD19/génétique , Antigènes CD19/métabolisme , Lignée cellulaire tumorale , Cellules cultivées , Cytokines/immunologie , Cytokines/métabolisme , Cytotoxicité immunologique/immunologie , Cellules HeLa , Tumeurs hématologiques/génétique , Tumeurs hématologiques/immunologie , Tumeurs hématologiques/métabolisme , Humains , Sous-unité gamma commune aux récepteurs des interleukines/déficit , Sous-unité gamma commune aux récepteurs des interleukines/génétique , Mâle , Souris de lignée NOD , Souris knockout , Souris SCID , Tumeurs/génétique , Tumeurs/métabolisme , Oligopeptides/génétique , Oligopeptides/métabolisme , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/immunologie , Protéines de fusion recombinantes/métabolisme , Lymphocytes T/métabolisme , Résultat thérapeutique
16.
Anticancer Agents Med Chem ; 17(10): 1344-1350, 2017.
Article de Anglais | MEDLINE | ID: mdl-28071584

RÉSUMÉ

Cellular immunotherapies such as CAR-T cell therapy and TCR-T cell therapy are relatively new, highly promising approaches for the treatment of cancer. In CAR-T cell therapy, a patient's T cells are engineered to express chimeric antigen receptors targeting tumor-associated cell surface antigens. In TCR-T cell therapy, the patient's T cells are engineered to express receptors targeting intracellular antigens. This report will summarize presentations from the recent CAR-TCR summit in Boston on September 13-16, 2016. These presentations were given by leaders in the field and many were divided into three streams: Discovery and Genetic T Cell Engineering; Translation and Clinical Development; and Manufacturing, Supply Chain and Commercialization. The report summarizes major pharmaceutical companies developing these novel therapies and provides challenges and perspectives for future therapeutic developments.


Sujet(s)
Immunothérapie , Tumeurs/thérapie , Récepteurs aux antigènes/immunologie , Boston , Humains
17.
Mediators Inflamm ; 2015: 628340, 2015.
Article de Anglais | MEDLINE | ID: mdl-26457007

RÉSUMÉ

While it has long been established that the chemokine receptor CCR9 and its ligand CCL25 are essential for the movement of leukocytes into the small intestine and the development of small-intestinal inflammation, the role of this chemokine-receptor pair in colonic inflammation is not clear. Toward this end, we compared colonic CCL25 protein levels in healthy individuals to those in patients with ulcerative colitis. In addition, we determined the effect of CCR9 pharmacological inhibition in the mdr1a(-/-) mouse model of ulcerative colitis. Colon samples from patients with ulcerative colitis had significantly higher levels of CCL25 protein compared to healthy controls, a finding mirrored in the mdr1a(-/-) mice. In the mdr1a(-/-) mice, CCR9 antagonists significantly decreased the extent of wasting and colonic remodeling and reduced the levels of inflammatory cytokines in the colon. These findings indicate that the CCR9:CCL25 pair plays a causative role in ulcerative colitis and suggest that CCR9 antagonists will provide a therapeutic benefit in patients with colonic inflammation.


Sujet(s)
Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/métabolisme , Récepteurs CCR/antagonistes et inhibiteurs , Récepteurs CCR/métabolisme , Sous-famille B de transporteurs à cassette liant l'ATP/génétique , Sous-famille B de transporteurs à cassette liant l'ATP/métabolisme , Animaux , Chimiokines CC/génétique , Chimiokines CC/métabolisme , Rectocolite hémorragique/génétique , Femelle , Humains , Techniques in vitro , Souris , Souris knockout , Sulfonamides/usage thérapeutique
18.
Immunology ; 141(1): 111-22, 2014 Jan.
Article de Anglais | MEDLINE | ID: mdl-24116850

RÉSUMÉ

The concentration of CXCL12/SDF-1 in the bloodstream is tightly regulated, given its central role in leucocyte and stem/progenitor cell egress from bone marrow and recruitment to sites of inflammation or injury. The mechanism responsible for this regulation is unknown. Here we show that both genetic deletion and pharmacological inhibition of CXCR7, a high-affinity CXCL12 receptor, caused pronounced increases in plasma CXCL12 levels. The rise in plasma CXCL12 levels was associated with an impairment in the ability of leucocytes to migrate to a local source of CXCL12. Using a set of complementary and highly sensitive techniques, we found that CXCR7 protein is expressed at low levels in multiple organs in both humans and mice. In humans, CXCR7 was detected primarily on venule endothelium and arteriole smooth muscle cells. CXCR7 expression on venule endothelium was also documented in immunodeficient mice and CXCR7(+/lacZ) mice. The vascular expression of CXCR7 therefore gives it immediate access to circulating CXCL12. These studies suggest that endothelial CXCR7 regulates circulating CXCL12 levels and that CXCR7 inhibitors might be used to block CXCL12-mediated cell migration for therapeutic purposes.


Sujet(s)
Chimiokine CXCL12/immunologie , Endothélium vasculaire/immunologie , Régulation de l'expression des gènes/immunologie , Cellules endothéliales de la veine ombilicale humaine/immunologie , Récepteurs CXCR/immunologie , Animaux , Mouvement cellulaire/immunologie , Chimiokine CXCL12/sang , Endothélium vasculaire/cytologie , Endothélium vasculaire/métabolisme , Cellules endothéliales de la veine ombilicale humaine/cytologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Humains , Leucocytes/cytologie , Leucocytes/immunologie , Leucocytes/métabolisme , Souris , Spécificité d'organe/immunologie , Récepteurs CXCR/biosynthèse
19.
Metabolism ; 62(11): 1623-32, 2013 Nov.
Article de Anglais | MEDLINE | ID: mdl-23953944

RÉSUMÉ

OBJECTIVE: CCR2 inhibition has produced promising experimental and clinical anti-hyperglycemic effects. These results support the thesis that insulin resistance and Type 2 diabetes (T2D) are associated with chronic unresolved inflammation. The aim of this study was to provide a broad analysis of the various physiological changes occurring in mouse models of T2D in connection with pharmacological CCR2 inhibition. MATERIALS/METHODS: A mouse-active chemical analogue of the clinical candidate CCX140-B was tested in diet-induced obese (DIO) mice and db/db mice. Measurements included: adipose tissue inflammatory macrophage counts; peripheral blood glucose levels at steady-state and after glucose and insulin challenges; peripheral blood insulin and adiponectin levels; 24-h urine output and urinary glucose levels; pancreatic islet number and size; hepatic triglyceride and glycogen content; and hepatic glucose-6-phosphatase levels. RESULTS: In DIO mice, the CCR2 antagonist completely blocked the recruitment of inflammatory macrophages to visceral adipose tissue. The mice exhibited reduced hyperglycemia and insulinemia, improved insulin sensitivity, increased circulating adiponectin levels, decreased pancreatic islet size and increased islet number. It also reduced urine output, glucose excretion, hepatic glycogen and triglyceride content and glucose 6-phosphatase levels. Similar effects were observed in the db/db diabetic mice. CONCLUSIONS: These data indicate that pharmacological inhibition of CCR2 in models of T2D can reduce inflammation in adipose tissue, alter hepatic metabolism and ameliorate multiple diabetic parameters. These mechanisms may contribute to the promising anti-diabetic effects seen in humans with at least one CCR2 antagonist.


Sujet(s)
Tissu adipeux/anatomopathologie , Diabète expérimental/traitement médicamenteux , Diabète expérimental/métabolisme , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Hypoglycémiants/pharmacologie , Insulinorésistance , Macrophages , Obésité/métabolisme , Récepteurs CCR2/antagonistes et inhibiteurs , Adiponectine/sang , Animaux , Marqueurs biologiques/sang , Glycémie/métabolisme , Diabète expérimental/sang , Diabète expérimental/étiologie , Diabète expérimental/anatomopathologie , Diabète de type 2/sang , Diabète de type 2/étiologie , Diabète de type 2/anatomopathologie , Alimentation riche en graisse , Relation dose-effet des médicaments , Glucosephosphatase/métabolisme , Glycogène/métabolisme , Glycosurie/diagnostic , Hypoglycémiants/usage thérapeutique , Inflammation/métabolisme , Insuline/administration et posologie , Insuline/sang , Cellules à insuline/anatomopathologie , Foie/métabolisme , Mâle , Souris , Souris de lignée C57BL , Obésité/sang , Obésité/complications , Obésité/étiologie , Récepteurs CCR2/métabolisme , Triglycéride/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE