91 results on '"Louahed J"'
Search Results
2. Expression of tumor-associated antigens in breast cancer subtypes
- Author
-
Curigliano, G, Bagnardi, V, Ghioni, M, Louahed, J, Brichard, V, Lehmann, F, Marra, A, Trapani, D, Criscitiello, C, Viale, G, Curigliano G., Bagnardi V., Ghioni M., Louahed J., Brichard V., Lehmann F. F., Marra A., Trapani D., Criscitiello C., Viale G., Curigliano, G, Bagnardi, V, Ghioni, M, Louahed, J, Brichard, V, Lehmann, F, Marra, A, Trapani, D, Criscitiello, C, Viale, G, Curigliano G., Bagnardi V., Ghioni M., Louahed J., Brichard V., Lehmann F. F., Marra A., Trapani D., Criscitiello C., and Viale G.
- Abstract
Objectives: Tumor-associated antigens (TAAs) are frequently overexpressed in several cancer types. The aim of this study was to investigate the expression of TAAs in breast cancer. Material and methods: A total of 250 selected invasive breast cancers including 50 estrogen receptor (ER)-positive (Luminal B like), 50 triple-negative (TN), 50 ER-positive lobular type, 50 ER- and progesterone receptor (PgR)-positive (Luminal A like) and 50 cerbB2-positive breast cancers, were assessed for New York esophageal squamous cell carcinoma-1 (NY-ESO-1), Wilms tumor antigen (WT-1) and PReferentially expressed Antigen of MElanoma (PRAME) antigen expression by immunohistochemistry (IHC). Results: A significantly higher expression of cancer testis (CT)-antigens NY-ESO-1 and WT-1 antigen was detected in TN breast cancers compared with ER-positive tumors. NY-ESO-1 overexpression (score 2 + and 3+) assessed by monoclonal and polyclonal antibodies was detected in 9 (18%) TN cancers as compared to 2 (4%) ER-positive tumors (p = 0.002). WT1 over-expression (score 2 + and 3+) was confirmed in 27 (54%) TN tumor samples as compared to 6 (12%) ER-positive (p < 0.0001). PRAME over-expression (score 2 + and 3+) was detected in 8 (16%) HER2 positive tumor samples as compared to no TN and ER-positive cancers (p = 0.0021). Conclusions: NY-ESO-1 and WT1 antigens are overexpressed in TN breast cancers. Because of the limited therapeutic options for this patient subgroup, CT antigen-based vaccines might prove to be useful for patients with this phenotype of breast cancer.
- Published
- 2020
3. Antibody production by injection of living cells expressing non self antigens as cell surface type II transmembrane fusion protein
- Author
-
Nizet, Yannick, Gillet, Laurent, Schroeder, Hélène, Lecuivre, Corinne, Louahed, J., Renauld, J.-C., Gianello, Pierre, and Vanderplasschen, Alain
- Published
- 2011
- Full Text
- View/download PDF
4. A Th1/IFNG gene signature is prognostic in the adjuvant setting of resectable high-risk melanoma but not in non-small cell lung cancer
- Author
-
Dizier, B. Callegaro, A. Debois, M. Dreno, B. Hersey, P. Gogas, H.J. Kirkwood, J.M. Vansteenkiste, J.F. Sequist, L.V. Atanackovic, D. Goeman, J. van Houwelingen, H. Salceda, S. Wang, F. Therasse, P. Debruyne, C. Spiessens, B. Brichard, V.G. Louahed, J. Ulloa-Montoya, F.
- Abstract
Purpose: Immune components of the tumor microenvironment (TME) have been associated with disease outcome. We prospectively evaluated the association of an immune-related gene signature (GS) with clinical outcome in melanoma and non-small cell lung cancer (NSCLC) tumor samples from two phase III studies. Experimental Design: The GS was prospectively validated using an adaptive signature design to optimize it for the sample type and technology used in phase III studies. One-third of the samples were used as “training set”; the remaining two thirds, constituting the “test set,” were used for the prospective validation of the GS. Results: In the melanoma training set, the expression level of eight Th1/IFNg-related genes in tumor-positive lymph node tissue predicted the duration of disease-free survival (DFS) and overall survival (OS) in the placebo arm. This GS was prospectively and independently validated as prognostic in the test set. Building a multivariate Cox model in the test set placebo patients from clinical covariates and the GS score, an increased number of melanoma-involved lymph nodes and the GS were associated with DFS and OS. This GS was not associated with DFS in NSCLC, although expression of the Th1/IFNg -related genes was associated with the presence of lymphocytes in tumor samples in both indications. Conclusions: These findings provide evidence that expression of Th1/IFNg genes in the TME, as measured by this GS, is associated with clinical outcome in melanoma. This suggests that, using this GS, patients with stage IIIB/C melanoma can be classified into different risk groups. © 2019 American Association for Cancer Research.
- Published
- 2020
5. Interleukin-9
- Author
-
Renauld, J.‐C, primary, Houssiau, F., additional, Louahed, J., additional, Vink, A., additional, Snick, J. Van, additional, and Uyttenhove, C., additional
- Published
- 1993
- Full Text
- View/download PDF
6. Interleukin-9 Regulates NF-κB Activity Through BCL3 Gene Induction
- Author
-
Richard, M., Louahed, J., Demoulin, J. -B, and Jean-Christophe Renauld
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
BCL3 encodes a protein with close homology to IκB proteins and interacts with p50 NF-κB homodimers. However, the regulation and transcriptional activity of BCL3 remain ill-defined. We observed here that interleukin-9 (IL-9) and IL-4, but not IL-2 or IL-3, transcriptionally upregulated BCL3 expression in T cells and mast cells. BCL3 induction by IL-9 was detected as soon as 4 hours after stimulation and appeared to be dependent on the Jak/STAT pathway. IL-9 stimulation was associated with an increase in p50 homodimers DNA binding activity, which was mimicked by stableBCL3 expression. This contrasts with tumor necrosis factor (TNF)-dependent NF-κB activation, which occurs earlier, involves p65/p50 dimers, and is dependent on IκB degradation. Moreover, IL-9 stimulation or BCL3 transient transfection similarly inhibited NF-κB–mediated transcription in response to TNF. Taken together, our observations show a new regulatory pathway for the NF-κB transcription factors through STAT-dependent upregulation ofBCL3 gene expression.
- Published
- 1999
- Full Text
- View/download PDF
7. Differential activity of dexamethasone on IL-2-, IL-4-, or IL-9-induced proliferation of murine factor-dependent T cell lines
- Author
-
Louahed, J., Renauld, J. -C, Jean-Baptiste Demoulin, Baughman, G., Bourgeois, S., Sugamura, K., and Snick, J.
- Subjects
Immunology ,Immunology and Allergy - Abstract
Mouse helper T cell lines were developed that proliferate permanently without Ag and APCs in response to either IL-2, IL-4, or IL-9, three cytokines whose receptors interact with the IL-2R gamma-chain for signal transduction. Depending on the growth factor, a marked difference was observed regarding the ability of dexamethasone (DEX) to inhibit cell proliferation. In three different cell lines, proliferation induced by IL-2 was completely arrested, while that supported by IL-9 was hardly affected. With IL-4, proliferation was also maintained but less markedly than with IL-9. Although DEX was able to induce apoptosis in these cells, the inhibition of IL-2-induced proliferation was not the result of apoptosis, as this process was equally antagonized by all three factors. Moreover, addition of IL-4 or IL-9 to cultures previously incubated with IL-2 and DEX for several days restored cell proliferation. Finally, autonomous cell variants derived from the factor-dependent cell lines were still protected by IL-4 and IL-9 against growth inhibition by DEX. Together, these results indicate that growth stimulation in the presence of glucocorticoids and inhibition of apoptosis involve distinct aspects of cytokine activities.
- Published
- 1996
- Full Text
- View/download PDF
8. IL-9 protects against bleomycin-induced lung injury: involvement of prostaglandins
- Author
-
Arras, M., Louahed, J., Heilier, J. -F, Delos, M., Brombacher, F., Jean-Christophe Renauld, Lison, D., Huaux, F., UCL - MD/MNOP - Département de morphologie normale et pathologique, UCL - MD/ESP - Ecole de santé publique, UCL - MD/MIGE - Département de microbiologie, d'immunologie et de génétique, UCL - (SLuc) Centre de toxicologie clinique, UCL - (SLuc) Service de biochimie médicale, and UCL - (MGD) Service d'anatomie pathologique
- Subjects
Lung Diseases ,Time Factors ,L-Lactate Dehydrogenase ,Indomethacin ,Interleukin-9 ,Mice, Inbred Strains ,Mice, Transgenic ,Lung Injury ,Original Research Paper ,Bleomycin ,Mice ,Water Supply ,Prostaglandins ,Animals ,Bronchoalveolar Lavage Fluid ,Lung - Abstract
IL-9 is a Th2 cytokine that exerts pleiotropic activities, and might be involved in the regulation of lung inflammatory processes. To characterize the activity of IL-9 on lung injury, we compared the pulmonary responses to bleomycin (blm) in IL-9 transgenic (Tg5) and wild-type (FVB) mice. Following intratracheal instillation of lethal doses of blm, the mortality rate was markedly reduced in Tg5 mice compared to their wild-type counterparts (ie, 25% mortality for Tg5 versus 85% for FVB mice, 21 days after instillation of 0.05U blm/mouse). Histological and biochemical analyses showed that blm induced less lung injury and less epithelial damage in Tg5 as compared to FVB animals. This protection of Tg5 mice was accompanied by an expansion of eosinophils and B cells in the lungs. In addition, TGF-beta and prostaglandin-E2 (PGE2) levels in broncho-alveolar lavage fluid were also increased in transgenic mice. The contribution of B cells and eosinophils to the protective mechanism did not appear essential since eosinophil-deficient (IL-5 KO) and B-deficient (muMT) mice overexpressing IL-9 were also resistant to high doses of blm. We could rule out that TGF-beta was a key factor in the protective effect of IL-9 by blocking this mediator with neutralizing antibodies. Indomethacin treatment, which inhibited PGE2 production in both strains, suppressed the protection in Tg5 mice, supporting the idea that IL-9 controls blm-induced lung injury through a prostaglandin-dependent mechanism.
- Published
- 2005
9. P25. Access to diagnostics: a bottleneck for immunotherapeutics development - case example of MAGE-A3 cancer immunotherapeutic
- Author
-
Lykopoulos, K, primary, Collins, P, additional, Hall, M, additional, Ehness, R, additional, Barth, J, additional, and Louahed, J, additional
- Published
- 2014
- Full Text
- View/download PDF
10. Antibody production by injection of living cells expressing non self antigens as cell surface type II transmembrane fusion protein
- Author
-
UCL - SSS/DDUV/MEXP - Médecine expérimentale, UCL - SSS/IREC/CHEX - Pôle de chirgurgie expérimentale et transplantation, UCL - (SLuc) Service de chirurgie et transplantation abdominale, Nizet, Yannick, Gillet, Laurent, Schroeder, Hélène, Lecuivre, Corinne, Louahed, J., Renauld, Jean-Christophe, Gianello, Pierre, Vanderplasschen, Alain, UCL - SSS/DDUV/MEXP - Médecine expérimentale, UCL - SSS/IREC/CHEX - Pôle de chirgurgie expérimentale et transplantation, UCL - (SLuc) Service de chirurgie et transplantation abdominale, Nizet, Yannick, Gillet, Laurent, Schroeder, Hélène, Lecuivre, Corinne, Louahed, J., Renauld, Jean-Christophe, Gianello, Pierre, and Vanderplasschen, Alain
- Abstract
Antigen expression and purification are laborious, time consuming and frequently difficult steps in the process of antibody production. In the present study, we developed a method avoiding these two steps. This method relies on the injection of histocompatible living cells stably expressing the antigen as a cell surface type II transmembrane fusion protein. A vector, nicknamed pCD1-CD134L, was constructed to express the antigen fused at the carboxyterminal end of the human CD134 ligand (CD134L) type II transmembrane protein on the surface of eucaryotic cells. This vector was shown to induce cell surface expression of epitopes from human c-Myc (soluble protein), uterogloblin-related protein 1 (secreted protein) and CD94 (type II transmembrane protein). Using this vector, we developed a method to produce antibodies without antigen production. The flowchart of this method is as follows: (i) cloning of the antigen in the pCD1-CD134L vector; (ii) production of a histocompatible cell line stably expressing the CD134L-antigen fusion protein; (iii) testing for cell surface expression of the fusion protein by targeting the CD134L carrier; and (iv) prime-boost immunisation with living cells expressing the fusion protein. This method was successfully used for production of polyclonal antibodies raised against Ixodes ricinus calreticulin (secreted protein) in mice and for production of monoclonal antibodies raised against an epitope of Vaccinia virus A56 (type I transmembrane protein) protein in rat. The present study is the first to demonstrate the use of a type II transmembrane protein as a carrier for cell surface display of antigens.
- Published
- 2011
11. P26 Expression of MAGE-A3 and its prognostic value in hepatocellular carcinoma in Asian patients
- Author
-
Lee, P., primary, Tanwandee, T., additional, Chotirosniramit, A., additional, D’Agostino, D., additional, Louahed, J., additional, Myo, A., additional, and Therasse, P., additional
- Published
- 2011
- Full Text
- View/download PDF
12. PP55 A MAGE-A3 specific quantitative RT-PCR assay used for patient accrual in MAGRIT, a Phase III ASCI (Antigen-Specific Cancer Immunotherapeutic) trial in adjuvant NSCLC
- Author
-
Coche, T., primary, Louahed, J., additional, Gruselle, O., additional, Wu, H., additional, Hoffmann, H., additional, Esteban-Gonzales, E., additional, and Therasse, P., additional
- Published
- 2009
- Full Text
- View/download PDF
13. Association of gene expression signature and clinical efficacy of MAGE-A3 antigen-specific cancer immunotherapeutic (ASCI) as adjuvant therapy in resected stage IB/II non-small cell lung cancer (NSCLC)
- Author
-
Vansteenkiste, J. F., primary, Zielinski, M., additional, Dahabreh, I. J., additional, Linder, A., additional, Lehmann, F., additional, Gruselle, O., additional, Therasse, P., additional, Louahed, J., additional, and Brichard, V. G., additional
- Published
- 2008
- Full Text
- View/download PDF
14. Expression of defined genes identified by pretreatment tumor profiling: Association with clinical responses to the GSK MAGE- A3 immunotherapeutic in metastatic melanoma patients (EORTC 16032–18031)
- Author
-
Louahed, J., primary, Gruselle, O., additional, Gaulis, S., additional, Coche, T., additional, Eggermont, A. M., additional, Kruit, W., additional, Dreno, B., additional, Sileni, V. Chiarion, additional, Lehmann, F., additional, and Brichard, V. G., additional
- Published
- 2008
- Full Text
- View/download PDF
15. 116 POSTER A recombinant HER2 protein evaluated for cancer immunotherapy: induction of specific antibodies and T-cells
- Author
-
Limentani, S.A., primary, Campone, M., additional, Dorval, T., additional, Curigliano, G., additional, White, S., additional, De Boer, R., additional, Canon, J., additional, Bachelot, T., additional, Cormont, F., additional, and Louahed, J., additional
- Published
- 2006
- Full Text
- View/download PDF
16. Evaluation of a recombinant HER2 vaccine: Induction of specific antibodies, T-cells and preliminary activity in metastatic breast cancer patients
- Author
-
Limentani, S. A., primary, Campone, M., additional, Dorval, T., additional, Tan-Chiu, E., additional, Curigliano, G., additional, De Boer, R., additional, Canon, J., additional, Bachelot, T., additional, Louahed, J., additional, and Brichard, V. G., additional
- Published
- 2006
- Full Text
- View/download PDF
17. Expression of mast cell specific proteases and high affinity IgE receptor by IL-9 activated T cell clones
- Author
-
UCL - MD/MIGE - Département de microbiologie, d'immunologie et de génétique, Louahed, J., Kermouni, A., Van Snick, Jacques, Renauld, Jean-Christophe, UCL - MD/MIGE - Département de microbiologie, d'immunologie et de génétique, Louahed, J., Kermouni, A., Van Snick, Jacques, and Renauld, Jean-Christophe
- Published
- 1994
18. IL-9 induces expression of granzymes and high-affinity IgE receptor in murine T helper clones.
- Author
-
Louahed, J, primary, Kermouni, A, additional, Van Snick, J, additional, and Renauld, J C, additional
- Published
- 1995
- Full Text
- View/download PDF
19. Interleukin-9 is a major anti-apoptotic factor for thymic lymphomas
- Author
-
Renauld, JC, primary, Vink, A, additional, Louahed, J, additional, and Van Snick, J, additional
- Published
- 1995
- Full Text
- View/download PDF
20. Rat (and mouse) monoclonal antibodies
- Author
-
Delaunay, T., primary, Louahed, J., additional, and H., Bazin, additional
- Published
- 1990
- Full Text
- View/download PDF
21. Role of insulin receptor substrate-2 in interleukin-9-dependent proliferation
- Author
-
Demoulin, J. B., Grasso, L., Atkins, J. M., Stevens, M., Louahed, J., Levitt, R. C., Nicolaides, N. C., and Renauld, J. C.
- Published
- 2000
- Full Text
- View/download PDF
22. IL-9 induces expression of granzymes and high-affinity IgE receptor in murine T helper clones
- Author
-
Louahed, J., Kermouni, A., Snick, J., and Jean-Christophe Renauld
- Subjects
Immunology ,Immunology and Allergy - Abstract
Interleukin 9 (IL-9) is a TH2 cytokine that has been shown to promote the antigen-independent growth of some mouse T helper clones. To characterize the specificity of IL-9-mediated T cell activation, we used a murine T cell clone that could grow with either IL-9 or IL-2. After differential hybridization of a cDNA library, we isolated three genes that were expressed preferentially in the presence of IL-9. Two of them correspond respectively to granzyme A and granzyme B, two proteases expressed by activated T cells. By Northern blot hybridization and functional assays, we found that IL-9 induced the expression of granzyme B in several T cell clones as well as in mast cell lines. In addition, other proteases such as the mouse mast cell proteases were also found to be expressed by IL-9-activated T cell clones. The third IL-9-induced cDNA corresponds to the alpha-chain of the high-affinity receptor for IgE. Several T cell clones expressed this IgE receptor mRNA and were able to bind IgE with high affinity. Taken together, our results indicate that IL-9 induces a mast cell-like phenotype in T cell clones.
23. A calcium-activated chloride channel blocker inhibits goblet cell metaplasia and mucus overproduction
- Author
-
Zhou Y, Shapiro M, Dong Q, Louahed J, Weiss C, Wan S, Chen Q, Dragwa C, Savio D, Huang M, Fuller C, Tomer Y, Nicholas Nicolaides, McLane M, and Rc, Levitt
24. Expression of tumor-associated antigens in breast cancer subtypes
- Author
-
Giuseppe Viale, Frederic Lehmann, Vincenzo Bagnardi, Giuseppe Curigliano, Carmen Criscitiello, Dario Trapani, Jamila Louahed, Vincent Brichard, Mariacristina Ghioni, Antonio Marra, Curigliano, G, Bagnardi, V, Ghioni, M, Louahed, J, Brichard, V, Lehmann, F, Marra, A, Trapani, D, Criscitiello, C, and Viale, G
- Subjects
Adult ,Tumor-associated antigens ,Cancer testis antigens ,Receptor, ErbB-2 ,Estrogen receptor ,Breast Neoplasms ,Triple Negative Breast Neoplasms ,lcsh:RC254-282 ,Cancer testis antigen ,03 medical and health sciences ,Breast cancer ,0302 clinical medicine ,Antigen ,Antigens, Neoplasm ,PRAME ,Biomarkers, Tumor ,Tumor-associated antigen ,Humans ,NY-ESO-1 ,Medicine ,030212 general & internal medicine ,WT1 Proteins ,Aged ,business.industry ,Membrane Proteins ,Cancer ,General Medicine ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Immunohistochemistry ,WT1 ,Receptors, Estrogen ,030220 oncology & carcinogenesis ,Cancer research ,Cancer/testis antigens ,Female ,Original Article ,Surgery ,Immunotherapy ,Receptors, Progesterone ,business - Abstract
Objectives Tumor-associated antigens (TAAs) are frequently overexpressed in several cancer types. The aim of this study was to investigate the expression of TAAs in breast cancer. Material and methods A total of 250 selected invasive breast cancers including 50 estrogen receptor (ER)-positive (Luminal B like), 50 triple-negative (TN), 50 ER-positive lobular type, 50 ER- and progesterone receptor (PgR)-positive (Luminal A like) and 50 cerbB2-positive breast cancers, were assessed for New York esophageal squamous cell carcinoma-1 (NY-ESO-1), Wilms tumor antigen (WT-1) and PReferentially expressed Antigen of MElanoma (PRAME) antigen expression by immunohistochemistry (IHC). Results A significantly higher expression of cancer testis (CT)-antigens NY-ESO-1 and WT-1 antigen was detected in TN breast cancers compared with ER-positive tumors. NY-ESO-1 overexpression (score 2 + and 3+) assessed by monoclonal and polyclonal antibodies was detected in 9 (18%) TN cancers as compared to 2 (4%) ER-positive tumors (p = 0.002). WT1 over-expression (score 2 + and 3+) was confirmed in 27 (54%) TN tumor samples as compared to 6 (12%) ER-positive (p, Highlights • Tumor-associated antigens are frequently overexpressed in several cancer types, being also associated with poorer patients’ survival outcomes. • Our study confirmed that NY-ESO-1 and WT1 antigens are higher expressed in triple-negative than in other breast cancer subtypes. • Given the limited therapeutic options for triple-negative breast cancer patients, the assessment of WT1 and NY-ESO-1 antigens expression in breast cancer tissue at surgery may allow to identify patients potentially candidate to adjuvant peptide vaccines, alone or in combination with other systemic therapies.
- Published
- 2020
- Full Text
- View/download PDF
25. IL-9 exerts biological function on antigen-experienced murine T cells and exacerbates colitis induced by adoptive transfer.
- Author
-
de Heusch M, Steenwinckel V, Cochez PM, Louahed J, Warnier G, Lemaire MM, Renauld JC, and Dumoutier L
- Subjects
- Animals, Colitis etiology, Colitis genetics, Colitis pathology, Disease Models, Animal, Hyaluronan Receptors genetics, Hyaluronan Receptors immunology, Interleukin-2 genetics, Interleukin-2 immunology, Interleukin-9 genetics, Mice, Mice, Knockout, Mice, SCID, Receptors, Interleukin-9 genetics, Receptors, Interleukin-9 immunology, Th17 Cells pathology, Th17 Cells transplantation, Th2 Cells pathology, Th2 Cells transplantation, Adoptive Transfer adverse effects, Colitis immunology, Interleukin-9 immunology, Th17 Cells immunology, Th2 Cells immunology
- Abstract
IL-9 is involved in various T cell-dependent inflammatory models including colitis, encepahlitis, and asthma. However, the regulation and specificity of IL-9 responsiveness by T cells during immune responses remains poorly understood. Here, we addressed this question using two different models: experimental colitis induced by transfer of naive CD4
+ CD45RBhigh T cells into immunodeficient mice, and OVA-specific T cell activation. In the colitis model, constitutive IL-9 expression exacerbated inflammation upon transfer of CD4+ CD45RBhigh T cells from WT but not from Il9r-/- mice, indicating that IL-9 acts directly on T cells. Suprisingly, such naïve CD4+ CD45RBhigh T cells failed to express the Il9r or respond to IL-9 in vitro, in contrast with CD4+ CD45RBlow T cells. By using OVA-specific T cells, we observed that T cells acquired the capacity to respond to IL-9 along with CD44 upregulation, after long-lasting (5 to 12 days) in vivo antigenic stimulation. Il9r expression was associated with Th2 and Th17 phenotypes. Interestingly, in contrast to the IL-2 response, antigen restimulation downregulated IL-9 responsiveness. Taken together, our results demonstrate that IL-9 does not act on naïve T cells but that IL-9 responsiveness is acquired by CD4+ T cells after in vivo activation and acquisition of memory markers such as CD44., (© 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)- Published
- 2020
- Full Text
- View/download PDF
26. A Th1/IFNγ Gene Signature Is Prognostic in the Adjuvant Setting of Resectable High-Risk Melanoma but Not in Non-Small Cell Lung Cancer.
- Author
-
Dizier B, Callegaro A, Debois M, Dreno B, Hersey P, Gogas HJ, Kirkwood JM, Vansteenkiste JF, Sequist LV, Atanackovic D, Goeman J, van Houwelingen H, Salceda S, Wang F, Therasse P, Debruyne C, Spiessens B, Brichard VG, Louahed J, and Ulloa-Montoya F
- Subjects
- Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Humans, Interferon-gamma metabolism, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms pathology, Melanoma drug therapy, Melanoma genetics, Prognosis, Prospective Studies, Survival Rate, Th1 Cells metabolism, Transcriptome, Biomarkers, Tumor genetics, Carcinoma, Non-Small-Cell Lung pathology, Gene Expression Regulation, Neoplastic, Interferon-gamma immunology, Melanoma pathology, Th1 Cells immunology, Tumor Microenvironment immunology
- Abstract
Purpose: Immune components of the tumor microenvironment (TME) have been associated with disease outcome. We prospectively evaluated the association of an immune-related gene signature (GS) with clinical outcome in melanoma and non-small cell lung cancer (NSCLC) tumor samples from two phase III studies., Experimental Design: The GS was prospectively validated using an adaptive signature design to optimize it for the sample type and technology used in phase III studies. One-third of the samples were used as "training set"; the remaining two thirds, constituting the "test set," were used for the prospective validation of the GS., Results: In the melanoma training set, the expression level of eight Th1/IFNγ-related genes in tumor-positive lymph node tissue predicted the duration of disease-free survival (DFS) and overall survival (OS) in the placebo arm. This GS was prospectively and independently validated as prognostic in the test set. Building a multivariate Cox model in the test set placebo patients from clinical covariates and the GS score, an increased number of melanoma-involved lymph nodes and the GS were associated with DFS and OS. This GS was not associated with DFS in NSCLC, although expression of the Th1/IFNγ-related genes was associated with the presence of lymphocytes in tumor samples in both indications., Conclusions: These findings provide evidence that expression of Th1/IFNγ genes in the TME, as measured by this GS, is associated with clinical outcome in melanoma. This suggests that, using this GS, patients with stage IIIB/C melanoma can be classified into different risk groups., (©2019 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
27. Expression of tumor-associated antigens in breast cancer subtypes.
- Author
-
Curigliano G, Bagnardi V, Ghioni M, Louahed J, Brichard V, Lehmann FF, Marra A, Trapani D, Criscitiello C, and Viale G
- Subjects
- Adult, Aged, Biomarkers, Tumor analysis, Female, Humans, Immunohistochemistry, Middle Aged, Receptors, Estrogen metabolism, Receptors, Progesterone metabolism, Triple Negative Breast Neoplasms metabolism, Antigens, Neoplasm analysis, Breast Neoplasms immunology, Membrane Proteins analysis, Receptor, ErbB-2 analysis, WT1 Proteins analysis
- Abstract
Objectives: Tumor-associated antigens (TAAs) are frequently overexpressed in several cancer types. The aim of this study was to investigate the expression of TAAs in breast cancer., Material and Methods: A total of 250 selected invasive breast cancers including 50 estrogen receptor (ER)-positive (Luminal B like), 50 triple-negative (TN), 50 ER-positive lobular type, 50 ER- and progesterone receptor (PgR)-positive (Luminal A like) and 50 cerbB2-positive breast cancers, were assessed for New York esophageal squamous cell carcinoma-1 (NY-ESO-1), Wilms tumor antigen (WT-1) and PReferentially expressed Antigen of MElanoma (PRAME) antigen expression by immunohistochemistry (IHC)., Results: A significantly higher expression of cancer testis (CT)-antigens NY-ESO-1 and WT-1 antigen was detected in TN breast cancers compared with ER-positive tumors. NY-ESO-1 overexpression (score 2 + and 3+) assessed by monoclonal and polyclonal antibodies was detected in 9 (18%) TN cancers as compared to 2 (4%) ER-positive tumors (p = 0.002). WT1 over-expression (score 2 + and 3+) was confirmed in 27 (54%) TN tumor samples as compared to 6 (12%) ER-positive (p < 0.0001). PRAME over-expression (score 2 + and 3+) was detected in 8 (16%) HER2 positive tumor samples as compared to no TN and ER-positive cancers (p = 0.0021)., Conclusions: NY-ESO-1 and WT1 antigens are overexpressed in TN breast cancers. Because of the limited therapeutic options for this patient subgroup, CT antigen-based vaccines might prove to be useful for patients with this phenotype of breast cancer., (Copyright © 2019 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
28. Association of homogeneous inflamed gene signature with a better outcome in patients with metastatic melanoma treated with MAGE-A3 immunotherapeutic.
- Author
-
Baurain JF, Robert C, Mortier L, Neyns B, Grange F, Lebbe C, Ulloa-Montoya F, De Sousa Alves PM, Gillet M, Louahed J, Jarnjak S, and Lehmann FF
- Abstract
Purpose: This study assessed clinical activity, safety and immunogenicity of MAGE-A3 immunotherapeutic in patients with MAGE-A3-positive metastatic melanoma., Patients and Methods: In this open-label, multicentre, uncontrolled, Phase II study (ClinicalTrials.gov NCT00896480), patients received ≤24 doses of MAGE-A3 immunotherapeutic (4-cycle schedule). At screening, two skin lesions were biopsied for MAGE-A3 expression analysis and presence/absence of a previously identified gene signature (GS) associated with favourable clinical outcome. Clinical activity was assessed in terms of clinical response, time-to-treatment failure (TTF) and progression-free survival (PFS). Adverse events (AEs) and serious AEs (SAEs) were recorded. MAGE-A3-specific immune responses were assessed. Clinical activity and immunogenicity were analysed overall and separately in patients with 2/2 (GS+/+), 1/2 (GS+/-) or 0/2 (GS-/-) biopsies presenting GS., Results: Of 49 screened patients, 32 had MAGE-A3-positive tumours; 24 (8 GS+/+, 8 GS+/-, 8 GS-/-) were treated. Two complete (GS+/+ patients) and two partial responses (one GS+/+, one GS+/-) were reported; of note, one of the two complete responses was unlikely to be related to the study treatment. Median TTF and PFS were 14.8 and 7.2 months for GS+/+, 2.3 and 2.8 months for GS+/- and 2.4 and 2.9 months for GS-/- patients. Three grade 3 AEs and two SAEs unrelated to treatment were reported. All patients were seropositive for MAGE-A3 antibodies on vaccination with no differences between the different GS profiles. MAGE-A3-specific CD4+ and CD8+ T cell immunogenicity was detected; 12/16 (75.0%) of patients presented CD4+ T cell responses., Conclusion: Treatment with MAGE-A3 immunotherapeutic showed signs of clinical activity in GS+/+ patients. Treatment was well tolerated and immunogenic. No differences in immune responses according to GS status were observed., Trial Registration Number: NCT00896480 (Results)., Competing Interests: Competing interests: MG, JL, FU-M and SJ are employees of the GSK group of companies. FFL and PMDSA were employees of the GSK group of companies during the conduct of the study. FFL, JL, PMDSA, FU-M and SJ hold shares in the GSK group of companies as part of their employee remuneration.
- Published
- 2018
- Full Text
- View/download PDF
29. MAGE-A3 immunotherapeutic as adjuvant therapy for patients with resected, MAGE-A3-positive, stage III melanoma (DERMA): a double-blind, randomised, placebo-controlled, phase 3 trial.
- Author
-
Dreno B, Thompson JF, Smithers BM, Santinami M, Jouary T, Gutzmer R, Levchenko E, Rutkowski P, Grob JJ, Korovin S, Drucis K, Grange F, Machet L, Hersey P, Krajsova I, Testori A, Conry R, Guillot B, Kruit WHJ, Demidov L, Thompson JA, Bondarenko I, Jaroszek J, Puig S, Cinat G, Hauschild A, Goeman JJ, van Houwelingen HC, Ulloa-Montoya F, Callegaro A, Dizier B, Spiessens B, Debois M, Brichard VG, Louahed J, Therasse P, Debruyne C, and Kirkwood JM
- Subjects
- Adult, Aged, Antigens, Neoplasm genetics, Chemotherapy, Adjuvant, Disease-Free Survival, Double-Blind Method, Female, Humans, Injections, Intramuscular, Internationality, Male, Melanoma mortality, Melanoma pathology, Melanoma surgery, Middle Aged, Neoplasm Invasiveness pathology, Neoplasm Proteins genetics, Neoplasm Staging, Prognosis, Risk Assessment, Skin Neoplasms mortality, Skin Neoplasms pathology, Skin Neoplasms surgery, Survival Analysis, Treatment Outcome, Melanoma, Cutaneous Malignant, Antigens, Neoplasm drug effects, Immunoconjugates therapeutic use, Immunotherapy methods, Melanoma drug therapy, Neoplasm Proteins drug effects, Skin Neoplasms drug therapy
- Abstract
Background: Despite newly approved treatments, metastatic melanoma remains a life-threatening condition. We aimed to evaluate the efficacy of the MAGE-A3 immunotherapeutic in patients with stage IIIB or IIIC melanoma in the adjuvant setting., Methods: DERMA was a phase 3, double-blind, randomised, placebo-controlled trial done in 31 countries and 263 centres. Eligible patients were 18 years or older and had histologically proven, completely resected, stage IIIB or IIIC, MAGE-A3-positive cutaneous melanoma with macroscopic lymph node involvement and an Eastern Cooperative Oncology Group performance score of 0 or 1. Randomisation and treatment allocation at the investigator sites were done centrally via the internet. We randomly assigned patients (2:1) to receive up to 13 intramuscular injections of recombinant MAGE-A3 with AS15 immunostimulant (MAGE-A3 immunotherapeutic; 300 μg MAGE-A3 antigen plus 420 μg CpG 7909 reconstituted in AS01B to a total volume of 0·5 mL), or placebo, over a 27-month period: five doses at 3-weekly intervals, followed by eight doses at 12-weekly intervals. The co-primary outcomes were disease-free survival in the overall population and in patients with a potentially predictive gene signature (GS-positive) identified previously and validated here via an adaptive signature design. The final analyses included all patients who had received at least one dose of study treatment; analyses for efficacy were in the as-randomised population and for safety were in the as-treated population. This trial is registered with ClinicalTrials.gov, number NCT00796445., Findings: Between Dec 1, 2008, and Sept 19, 2011, 3914 patients were screened, 1391 randomly assigned, and 1345 started treatment (n=895 for MAGE-A3 and n=450 for placebo). At final analysis (data cutoff May 23, 2013), median follow-up was 28·0 months [IQR 23·3-35·5] in the MAGE-A3 group and 28·1 months [23·7-36·9] in the placebo group. Median disease-free survival was 11·0 months (95% CI 10·0-11·9) in the MAGE-A3 group and 11·2 months (8·6-14·1) in the placebo group (hazard ratio [HR] 1·01, 0·88-1·17, p=0·86). In the GS-positive population, median disease-free survival was 9·9 months (95% CI 5·7-17·6) in the MAGE-A3 group and 11·6 months (5·6-22·3) in the placebo group (HR 1·11, 0·83-1·49, p=0·48). Within the first 31 days of treatment, adverse events of grade 3 or worse were reported by 126 (14%) of 894 patients in the MAGE-A3 group and 56 (12%) of 450 patients in the placebo group, treatment-related adverse events of grade 3 or worse by 36 (4%) patients given MAGE-A3 vs six (1%) patients given placebo, and at least one serious adverse event by 14% of patients in both groups (129 patients given MAGE-A3 and 64 patients given placebo). The most common adverse events of grade 3 or worse were neoplasms (33 [4%] patients in the MAGE-A3 group vs 17 [4%] patients in the placebo group), general disorders and administration site conditions (25 [3%] for MAGE-A3 vs four [<1%] for placebo) and infections and infestations (17 [2%] for MAGE-A3 vs seven [2%] for placebo). No deaths were related to treatment., Interpretation: An antigen-specific immunotherapeutic alone was not efficacious in this clinical setting. Based on these findings, development of the MAGE-A3 immunotherapeutic for use in melanoma has been stopped., Funding: GlaxoSmithKline Biologicals SA., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
30. Safety and immunogenicity of MAGE-A3 cancer immunotherapeutic with dacarbazine in patients with MAGE-A3-positive metastatic cutaneous melanoma: an open phase I/II study with a first assessment of a predictive gene signature.
- Author
-
Grob JJ, Mortier L, D'Hondt L, Grange F, Baurain JF, Dréno B, Lebbe C, Robert C, Dompmartin A, Neyns B, Gillet M, Louahed J, Jarnjak S, and Lehmann FF
- Abstract
Background: We assessed safety, immunogenicity and clinical activity of recombinant MAGE-A3 antigen combined with AS15 immunostimulant (MAGE-A3 immunotherapeutic) in association with dacarbazine in patients with metastatic melanoma., Methods: In this open-label, phase I/II, uncontrolled multicentre trial conducted in Belgium and France, patients with MAGE-A3-positive melanoma received up to 24 doses of MAGE-A3 immunotherapeutic (four cycles) coadministered with eight doses of dacarbazine. Adverse events (AE) were recorded until 31 days postvaccination, and serious AEs (SAE), until 30 days following the last dose. MAGE-A3-specific antibodies were measured by ELISA. Clinical activity of MAGE-A3 immunotherapeutic was assessed in patients positive/negative for previously identified gene signature (GS) associated with clinical outcome., Results: Forty-eight patients were enrolled and treated (32 GS+, 15 GS-, 1 unknown GS status); two patients completed the study. All patients reported AEs, the most common were 'general disorders and administration site conditions' (94%). Treatment-related AEs were reported by 85% of patients; the most common was pain at injection site (38%). Sixteen SAEs were reported by 21% of patients; two were considered as treatment related (neutropenia and thrombocytopenia; grade 4). Postdose 4, all patients were seropositive for MAGE-A3-specific antibodies, with a geometric mean titre of 2778.7 ELISA units (EU)/mL (95% CI 1638.3 to 4712.8). One complete and three partial responses were reported (only in GS+ patients). Median overall survival was 11.4 months for GS+ and 5.3 months for GS- patients., Conclusion: Although this trial shows poor results compared with the new results with checkpoint inhibitors, it gives an interesting insight in rapidly developing fields like combinations of immunotherapy and chemotherapy, new generation vaccines and the use of gene profile as a predictive marker., Trial Registration Number: NCT00849875., Competing Interests: Competing interests: MG, JL and SJ are employees of the GSK group of companies. FFL was an employee of the GSK group of companies during the conduct of the study. FFL, JL and SJ own stock/stock options in the GSK group of companies. BD received grant/personal fees from GSK group of companies during the conduct of the study and from BMS, Roche and Novartis outside the submitted work. LM declares that his institution received support from GSK group of companies and Novartis to carry out clinical studies. LM reports personal fees from Roche, BMS, AMGEN, Novartis and GSK group of companies outside submitted work. JJG reports personal fees from GSK group of companies for his participation to an advisory board during the conduct of the study. CR received personal fees for participating to advisory boards for GSK, Novartis, Amgen, Roche, BMS and MSD. CL declares personal fees from Novartis, Roche, BMS, MSD and AMGEN for her participation to advisory boards.
- Published
- 2017
- Full Text
- View/download PDF
31. Gene expression of MAGE-A3 and PRAME tumor antigens and EGFR mutational status in Taiwanese non-small cell lung cancer patients.
- Author
-
Pan SH, Su KY, Spiessens B, Kusuma N, Delahaye NF, Gruselle O, Myo A, de Creus A, Louahed J, Chang GC, Yu SL, and Yang PC
- Subjects
- Adult, Aged, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Female, Humans, Lung Neoplasms metabolism, Lung Neoplasms pathology, Male, Middle Aged, Mutation, Polymorphism, Genetic, Retrospective Studies, Taiwan, Antigens, Neoplasm metabolism, Biomarkers, Tumor metabolism, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms genetics, Neoplasm Proteins metabolism
- Abstract
Aim: To determine the frequency of expression of the tumor-associated antigens (TAAs) melanoma-associated antigen A3 (MAGE-A3) and preferentially expressed antigen of melanoma (PRAME) and the rate of EGFR mutations in a Taiwanese non-small cell lung cancer (NSCLC) population including only adenocarcinomas and squamous cell carcinomas. Furthermore, to investigate associations between TAA expression and EGFR mutations and to evaluate these TAAs as prognostic markers for overall survival. The occurrence of single nucleotide polymorphisms in MAGEA3 and PRAME was also assessed., Methods: Archival fresh-frozen tumor tissue specimens were tested by quantitative reverse transcription polymerase chain reaction assays to detect MAGE-A3 and PRAME expression. EGFR mutations were detected by mass spectroscopy and single nucleotide polymorphisms by gene sequencing., Results: Of the 156 adenocarcinomas examined, 3.3% expressed MAGE-A3, 32.2% expressed PRAME and 62.8% had EGFR mutations. Of the 128 squamous cell carcinomas, 29.8% expressed MAGE-A3, 59.2% expressed PRAME and 20.5% harbored EGFR mutations. TAA expression was similar across subgroups determined by patient or tumor characteristics. There was no association between TAA expression and EGFR mutation status and TAA expression was found not to be a prognostic marker for survival. Single nucleotide polymorphisms were identified, one of which with a possible impact on MAGE-A3 expression., Conclusions: In this NSCLC population, expression of MAGE-A3 and PRAME was more frequent in squamous cell carcinomas than in adenocarcinomas tumors. EGFR mutations were not associated with TAA expression for either histology and were three times more frequent in adenocarcinomas than in squamous cell carcinomas tumors., (© 2016 The Authors. Asia-Pacific Journal of Clinical Oncology Published by John Wiley & Sons Australia, Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
32. Safety and Immunogenicity of the PRAME Cancer Immunotherapeutic in Patients with Resected Non-Small Cell Lung Cancer: A Phase I Dose Escalation Study.
- Author
-
Pujol JL, De Pas T, Rittmeyer A, Vallières E, Kubisa B, Levchenko E, Wiesemann S, Masters GA, Shen R, Tjulandin SA, Hofmann HS, Vanhoutte N, Salaun B, Debois M, Jarnjak S, De Sousa Alves PM, Louahed J, Brichard VG, and Lehmann FF
- Subjects
- Carcinoma, Non-Small-Cell Lung pathology, Female, Humans, Lung Neoplasms pathology, Male, Carcinoma, Non-Small-Cell Lung drug therapy, Chemotherapy, Adjuvant methods, Immunotherapy methods, Lung Neoplasms drug therapy
- Abstract
Introduction: Adjuvant platinum-based chemotherapy is standard treatment for surgically resected stage II to IIIA NSCLC, but the relapse rate is high. The preferentially expressed antigen of melanoma (PRAME) tumor antigen is expressed in two-thirds of NSCLC and offers an attractive target for antigen-specific immunization. A phase I dose escalation study assessed the safety and immunogenicity of a PRAME immunotherapeutic consisting of recombinant PRAME plus proprietary immunostimulant AS15 in patients with surgically resected NSCLC (NCT01159964)., Methods: Patients with PRAME-positive resected stage IB to IIIA NSCLC were enrolled in three consecutive cohorts to receive up to 13 injections of PRAME immunotherapeutic (recombinant PRAME protein dose of 20 μg, 100 μg, or 500 μg, with a fixed dose of AS15). Adverse events, predefined dose-limiting toxicity, and the anti-PRAME humoral response (measured by enzyme-linked immunosorbent assay) were coprimary end points. Anti-PRAME cellular responses were assessed., Results: A total of 60 patients were treated (18 received 20 μg of PRAME, 18 received 100 μg of PRAME, and 24 received 500 μg of PRAME). No dose-limiting toxicity was reported. Adverse events considered by the investigator to be causally related to treatment were grade 1 or 2, and most were injection site reactions or fever. All patients had detectable anti-PRAME antibodies after four immunizations. The percentages of patients with PRAME-specific CD4-positive T cells were higher at the dose of 500 μg compared with lower doses. No predefined CD8-positive T-cell responses were detected., Conclusion: The PRAME immunotherapeutic had an acceptable safety profile. All patients had anti-PRAME humoral responses that were not dose related, and 80% of those treated at the highest dose showed a cellular immune response. The dose of 500 μg was selected. However, further development was stopped after negative results with a similar immunotherapeutic in patients with NSCLC., (Copyright © 2016 International Association for the Study of Lung Cancer. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
33. The prevalence of expression of MAGE-A3 and PRAME tumor antigens in East and South East Asian non-small cell lung cancer patients.
- Author
-
Thongprasert S, Yang PC, Lee JS, Soo R, Gruselle O, Myo A, Louahed J, Lehmann FF, Brichard VG, and Coche T
- Subjects
- Adenocarcinoma metabolism, Adenocarcinoma pathology, Adult, Aged, Aged, 80 and over, Asia, Southeastern epidemiology, Biomarkers, Tumor analysis, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell pathology, Asia, Eastern epidemiology, Female, Gene Expression Regulation, Neoplastic, Humans, Immunotherapy methods, Lung Neoplasms pathology, Male, Middle Aged, Neoplasm Staging, Retrospective Studies, Smoking epidemiology, Antigens, Neoplasm metabolism, Carcinoma, Non-Small-Cell Lung metabolism, Lung Neoplasms metabolism, Neoplasm Proteins metabolism, Prevalence
- Abstract
Introduction: Treatment of non-small cell lung cancer (NSCLC) is an important and often unmet medical need regardless of the disease stage at the time of first diagnosis. Antigen-specific immunotherapy may be a feasible therapeutic option if tumor associated antigens (TAAs) that can be targeted by the patient's immune system are identified. The study objective (NCT01837511) was to investigate the expression rates of MAGE-A3 and PRAME in tumors from East Asian NSCLC patients, and the associations between TAA expression and clinico-pathologic patient characteristics., Methods: Archived formalin-fixed paraffin-embedded tumor tissue specimens were tested for MAGE-A3 and PRAME expression by quantitative reverse transcription polymerase chain reaction. Exploratory analyses of the impact of patient and tumor characteristics on antigen expression were performed by multivariate logistic regression analyses., Results: A total of 377 specimens were tested and a valid expression result was obtained for 86.5% and 92.6% for MAGE-A3 and PRAME, respectively. Of the specimens with valid test results, 26.4% expressed MAGE-A3, 49.9% PRAME, 20.0% both and 57.5% expressed at least one TAA. The same pattern of associations between antigen expression and patient and tumor characteristics was found for both TAAs: higher rates of antigen-positive tumors were found in squamous cell carcinomas compared to adenocarcinomas, and for smokers compared to non-smokers., Conclusions: Expression of MAGE-A3 and PRAME suggests an association with tumor histology and the patient's smoking status. The rates of TAA-positive tumors found in these East and South East Asian NSCLC patients indicate that both antigens may serve as targets for antigen-specific immunotherapies., (Copyright © 2016 The Authors. Published by Elsevier Ireland Ltd.. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
34. Efficacy of the MAGE-A3 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-positive non-small-cell lung cancer (MAGRIT): a randomised, double-blind, placebo-controlled, phase 3 trial.
- Author
-
Vansteenkiste JF, Cho BC, Vanakesa T, De Pas T, Zielinski M, Kim MS, Jassem J, Yoshimura M, Dahabreh J, Nakayama H, Havel L, Kondo H, Mitsudomi T, Zarogoulidis K, Gladkov OA, Udud K, Tada H, Hoffman H, Bugge A, Taylor P, Gonzalez EE, Liao ML, He J, Pujol JL, Louahed J, Debois M, Brichard V, Debruyne C, Therasse P, and Altorki N
- Subjects
- Aged, Antigens, Neoplasm metabolism, Biomarkers, Tumor metabolism, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Chemotherapy, Adjuvant, Double-Blind Method, Female, Follow-Up Studies, Humans, Lung Neoplasms immunology, Lung Neoplasms metabolism, Lung Neoplasms pathology, Male, Middle Aged, Neoplasm Proteins metabolism, Neoplasm Recurrence, Local immunology, Neoplasm Recurrence, Local metabolism, Neoplasm Recurrence, Local pathology, Neoplasm Staging, Prognosis, Survival Rate, Antigens, Neoplasm immunology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Immunoconjugates therapeutic use, Immunotherapy, Lung Neoplasms drug therapy, Neoplasm Proteins immunology, Neoplasm Recurrence, Local drug therapy
- Abstract
Background: Fewer than half of the patients with completely resected non-small-cell lung cancer (NSCLC) are cured. Since the introduction of adjuvant chemotherapy in 2004, no substantial progress has been made in adjuvant treatment. We aimed to assess the efficacy of the MAGE-A3 cancer immunotherapeutic in surgically resected NSCLC., Methods: In this randomised, double-blind, placebo-controlled trial, we recruited patients aged at least 18 years with completely resected stage IB, II, and IIIA MAGE-A3-positive NSCLC who did or did not receive adjuvant chemotherapy from 443 centres in 34 countries (Europe, the Americas, and Asia Pacific). Patients were randomly assigned (2:1) to receive 13 intramuscular injections of recMAGE-A3 with AS15 immunostimulant (MAGE-A3 immunotherapeutic) or placebo during 27 months. Randomisation and treatment allocation at the investigator site was done centrally via internet with stratification for chemotherapy versus no chemotherapy. Participants, investigators, and those assessing outcomes were masked to group assignment. A minimisation algorithm accounted for the number of chemotherapy cycles received, disease stage, lymph node sampling procedure, performance status score, and lifetime smoking status. The primary endpoint was broken up into three co-primary objectives: disease-free survival in the overall population, the no-chemotherapy population, and patients with a potentially predictive gene signature. The final analyses included the total treated population (all patients who had received at least one treatment dose). This trial is registered with ClinicalTrials.gov, number NCT00480025., Findings: Between Oct 18, 2007, and July 17, 2012, we screened 13 849 patients for MAGE-A3 expression; 12 820 had a valid sample and of these, 4210 (33%) had a MAGE-A3-positive tumour. 2312 of these patients met all eligibility criteria and were randomly assigned to treatment: 1515 received MAGE-A3 and 757 received placebo and 40 were randomly assigned but never started treatment. 784 patients in the MAGE-A3 group also received chemotherapy, as did 392 in the placebo group. Median follow-up was 38·1 months (IQR 27·9-48·4) in the MAGE-A3 group and 39·5 months (27·9-50·4) in the placebo group. In the overall population, median disease-free survival was 60·5 months (95% CI 57·2-not reached) for the MAGE-A3 immunotherapeutic group and 57·9 months (55·7-not reached) for the placebo group (hazard ratio [HR] 1·02, 95% CI 0·89-1·18; p=0·74). Of the patients who did not receive chemotherapy, median disease-free survival was 58·0 months (95% CI 56·6-not reached) in those in the MAGE-A3 group and 56·9 months (44·4-not reached) in the placebo group (HR 0·97, 95% CI 0·80-1·18; p=0·76). Because of the absence of treatment effect, we could not identify a gene signature predictive of clinical benefit to MAGE-A3 immunotherapeutic. The frequency of grade 3 or worse adverse events was similar between treatment groups (246 [16%] of 1515 patients in the MAGE-A3 group and 122 [16%] of 757 in the placebo group). The most frequently reported grade 3 or higher adverse events were infections and infestations (37 [2%] in the MAGE-A3 group and 19 [3%] in the placebo group), vascular disorders (30 [2%] vs 17 [3%]), and neoplasm (benign, malignant, and unspecified (29 [2%] vs 16 [2%])., Interpretation: Adjuvant treatment with the MAGE-A3 immunotherapeutic did not increase disease-free survival compared with placebo in patients with MAGE-A3-positive surgically resected NSCLC. Based on our results, further development of the MAGE-A3 immunotherapeutic for use in NSCLC has been stopped., Funding: GlaxoSmithKline Biologicals SA., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
35. A non-randomized dose-escalation Phase I trial of a protein-based immunotherapeutic for the treatment of breast cancer patients with HER2-overexpressing tumors.
- Author
-
Limentani SA, Campone M, Dorval T, Curigliano G, de Boer R, Vogel C, White S, Bachelot T, Canon JL, Disis M, Awada A, Berlière M, Amant F, Levine E, Burny W, Callegaro A, de Sousa Alves PM, Louahed J, Brichard V, and Lehmann FF
- Subjects
- Adult, Aged, Breast Neoplasms metabolism, Dose-Response Relationship, Drug, Drug Administration Schedule, Drug Dosage Calculations, Female, Gene Expression Regulation, Neoplastic, Humans, Immunologic Factors adverse effects, Immunotherapy, Middle Aged, Recombinant Proteins immunology, Survival Analysis, Treatment Outcome, Breast Neoplasms therapy, Immunologic Factors administration & dosage, Receptor, ErbB-2 immunology, Receptor, ErbB-2 metabolism, Recombinant Proteins administration & dosage, Up-Regulation
- Abstract
This Phase I dose-escalation study (NCT00058526) assessed the safety and immunogenicity of an anti-cancer immunotherapeutic (recombinant HER2 protein (dHER2) combined with the immunostimulant AS15) in patients with early-stage HER2-overexpressing breast cancer (BC). Sixty-one trastuzumab-naive patients with stage II-III HER2-positive BC received the dHER2 immunotherapeutic after surgical resection and adjuvant therapy. They were allocated into four cohorts receiving different doses of dHER2 (20, 100, 500 µg) combined with a fixed AS15 dose. Safety and immunogenicity (dHER2-specific antibody responses) were assessed. After completing the immunization schedule (three or six doses over 14 weeks) and a six-month follow-up, the patients were followed for 5 years for late toxicity, long-term immunogenicity, and clinical status. The immunizations were well tolerated, and increasing doses of dHER2 had no impact on the frequency or severity of adverse events. Few late toxicities were reported, and after 5 years 45/54 patients (83.3 %) were still alive, while 28/45 (62 %) with known disease status were disease free. Regarding the immunogenicity of the compound, a positive association was found between the dHER2 dose, the immunization schedule, and the prevalence of dHER2-specific humoral responses. Among the patients receiving the most intense immunization schedule with the highest dHER2 dose, 6/8 maintained their dHER2-specific antibody response 5 years after immunization. The dHER2 immunotherapeutic had an acceptable safety profile in early HER2-positive BC patients. dHER2-specific antibody responses were induced, with the rate of responders increasing with the dHER2 dose and the number and frequency of immunizations.
- Published
- 2016
- Full Text
- View/download PDF
36. A phase I/II trial of the safety and clinical activity of a HER2-protein based immunotherapeutic for treating women with HER2-positive metastatic breast cancer.
- Author
-
Curigliano G, Romieu G, Campone M, Dorval T, Duck L, Canon JL, Roemer-Becuwe C, Roselli M, Neciosup S, Burny W, Callegaro A, de Sousa Alves PM, Louahed J, Brichard V, and Lehmann FF
- Subjects
- Adjuvants, Immunologic adverse effects, Antineoplastic Agents therapeutic use, Breast Neoplasms metabolism, Disease-Free Survival, Drug Administration Schedule, Female, Humans, Immunotherapy, Middle Aged, Receptor, ErbB-2 genetics, Recombinant Proteins adverse effects, Trastuzumab therapeutic use, Treatment Outcome, Adjuvants, Immunologic administration & dosage, Antineoplastic Agents administration & dosage, Breast Neoplasms therapy, Receptor, ErbB-2 metabolism, Recombinant Proteins administration & dosage, Trastuzumab administration & dosage
- Abstract
The objectives of this phase I/II study (NCT00140738) were to evaluate the safety and clinical activity of a cancer immunotherapeutic agent (recombinant HER2 protein (dHER2) and the immunostimulant AS15) in patients with HER2-overexpressing metastatic breast cancer (MBC). Forty HER2-positive MBC patients received up to 18 doses (12q2w, 6q3w) of dHER2 immunotherapeutic, as first- or second-line therapy following response to trastuzumab-based treatment as maintenance. Toxicity was graded by the Common Terminology Criteria for Adverse Events (CTCAE) and clinical activity was evaluated by target lesion assessment according to the Response Evaluation Criteria in Solid Tumors (RECIST). Immunogenicity was assessed. The dHER2 immunotherapeutic was well tolerated: grade 1/2 adverse events (AEs) were most common. No cardiac events were observed and one patient experienced an asymptomatic decrease of left ventricular ejection fraction below the normal range (47 %). Both humoral and cellular immunogenicity to the dHER2 antigen was observed. No patient discontinued the immunizations because of AEs but 35/40 withdrew prematurely, 34 because of disease progression (24/34 before or at the tumor assessment after dose 6). One patient achieved a complete response lasting 11 months and one patient had a partial response lasting 3.5 months. Ten patients experienced stable disease ≥26 weeks with 4/10 still in stable disease at the last tumor assessment after 47 weeks. Immunization of MBC patients with the dHER2 immunotherapeutic was associated with minimal toxicity and no cardiac events. Clinical activity was observed with two objective responses and prolonged stable disease for 10/40 patients.
- Published
- 2016
- Full Text
- View/download PDF
37. A Comprehensive Preclinical Model Evaluating the Recombinant PRAME Antigen Combined With the AS15 Immunostimulant to Fight Against PRAME-expressing Tumors.
- Author
-
Gérard C, Baudson N, Ory T, Segal L, and Louahed J
- Subjects
- Adjuvants, Immunologic toxicity, Animals, Antibodies blood, Antigens, Neoplasm toxicity, Cell Line, Tumor, Drug Therapy, Combination, Female, Immunotherapy, Macaca fascicularis, Male, Mice, Mice, Transgenic, Neoplasms immunology, Recombinant Proteins therapeutic use, Recombinant Proteins toxicity, T-Lymphocytes immunology, Adjuvants, Immunologic therapeutic use, Antigens, Neoplasm therapeutic use, Neoplasms drug therapy
- Abstract
The PRAME tumor antigen is a potential target for immunotherapy. We assessed the immunogenicity, the antitumor activity, and the safety and the tolerability of a recombinant PRAME protein (recPRAME) combined with the AS15 immunostimulant (recPRAME+ AS15) in preclinical studies in mice and Cynomolgus monkeys. Four groups of 12 CB6F1 mice received 4 injections of phosphate-buffered saline (PBS), recPRAME, AS15, or recPRAME+AS15. Immunized mice were injected with tumor cells expressing PRAME (CT26-PRAME) 2 weeks or 2 months after the last injection. The mean tumor surface was measured twice a week. Two groups of 10 monkeys received 7 injections of saline or recPRAME+ AS15. T-cell responses were measured by flow cytometry using intracellular cytokine staining (ICS). In CB6F1 mice, repeated injections of recPRAME+ AS15 induced high PRAME-specific antibody titers and mostly CD4+ T cells producing cytokines. This immune response was long-lasting in these animals and was associated with protection against a challenge with PRAME-expressing tumor cells (CT26-PRAME) applied either 2 weeks or 2 months after the last injection; these data indicate the induction of an immune memory. In HLA-A02.01/HLA-DR1 transgenic mice, recPRAME+ AS15 induced both CD4+ and CD8+ T-cell responses, indicating that this antigen can be processed by the human leukocyte antigen and is potentially immunogenic in humans. In addition, a repeated-dose toxicity study in monkeys showed that 7 biweekly injections of recPRAME+ AS15 were well tolerated, and induced PRAME-specific antibodies and T cells. In conclusion, these preclinical data indicate that repeated injections of the PRAME cancer immunotherapeutic are immunogenic and have an acceptable safety profile.
- Published
- 2015
- Full Text
- View/download PDF
38. Safety and Immunogenicity of MAGE-A3 Cancer Immunotherapeutic with or without Adjuvant Chemotherapy in Patients with Resected Stage IB to III MAGE-A3-Positive Non-Small-Cell Lung Cancer.
- Author
-
Pujol JL, Vansteenkiste JF, De Pas TM, Atanackovic D, Reck M, Thomeer M, Douillard JY, Fasola G, Potter V, Taylor P, Bosquée L, Scheubel R, Jarnjak S, Debois M, de Sousa Alves P, Louahed J, Brichard VG, and Lehmann FF
- Subjects
- Adult, Aged, Aged, 80 and over, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung pathology, Chemotherapy, Adjuvant, Cisplatin administration & dosage, Cohort Studies, Female, Humans, Immunotherapy methods, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Male, Middle Aged, Neoplasm Staging, Recombinant Proteins therapeutic use, Vinblastine administration & dosage, Vinblastine analogs & derivatives, Vinorelbine, Antigens, Neoplasm therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Non-Small-Cell Lung therapy, Lung Neoplasms therapy, Neoplasm Proteins therapeutic use
- Abstract
Introduction: To assess the safety and immunogenicity of MAGE-A3 immunotherapeutic in patients with stage IB-III MAGE-A3-positive non-small-cell lung cancer (NSCLC) who were or were not undergoing standard cisplatin/vinorelbine chemotherapy., Methods: This open, prospective, multicenter, parallel-group phase I study (NCT00455572) enrolled patients with resected (cohorts 1-3) or unresectable (cohort 4) MAGE-A3-positive NSCLC. MAGE-A3 immunotherapeutic (300 μg recombinant MAGE-A3 formulated with AS15) was administered (eight doses, 3 weeks apart) concurrent with (cohort 1), after (cohort 2), or without (cohort 3) standard-adjuvant chemotherapy, or after standard radiotherapy and/or chemotherapy (cohort 4)., Results: Sixty-seven patients received greater than or equal to 1 dose of MAGE-A3 immunotherapeutic. Grade 3/4 adverse events (AEs) were reported for 16 out of 19 (84%), 2 out of 18 (11%), 5 out of 18 (28%), and 1 out of 12 (8%) patients in cohorts 1, 2, 3, and 4, respectively. Many grade 3/4 AEs in cohort 1 (e.g., neutropenia) were typical of chemotherapy. Six patients, including three in cohort 1, reported study treatment-related grade 3/4 AEs (injection-site reactions or musculoskeletal/back pain, which resolved within 5 days). One patient (in cohort 4) died, but this and the other serious adverse events were not study treatment related. MAGE-A3-specific antibody responses to immunotherapy were induced in all patients evaluated in all cohorts. MAGE-A3-specific CD4 T-cell responses to immunotherapy were detected in 4 out of 11 (36%), 4 out of 15 (27%), 2 out of 8 (25%), and 5 out of 6 (83%) evaluated patients in cohorts 1, 2, 3, and 4, respectively; and CD8 T-cell responses were only detected in four patients., Conclusion: In resected and unresectable NSCLC patients and irrespective of whether standard chemotherapy was concurrent or not, MAGE-A3 immunotherapeutic is well tolerated and induces MAGE-A3-specific immune responses.
- Published
- 2015
- Full Text
- View/download PDF
39. Development of a Quantitative Real-Time RT-PCR Assay for the Detection of MAGE-A3-Positive Tumors.
- Author
-
Gruselle O, Coche T, and Louahed J
- Subjects
- Base Sequence, Carcinoma, Non-Small-Cell Lung genetics, Humans, Lung Neoplasms genetics, Molecular Sequence Data, Sequence Homology, Nucleic Acid, Antigens, Neoplasm genetics, Biomarkers, Tumor genetics, Carcinoma, Non-Small-Cell Lung diagnosis, Lung Neoplasms diagnosis, Neoplasm Proteins genetics, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction methods, Reverse Transcriptase Polymerase Chain Reaction methods
- Abstract
Melanoma antigen A3 (MAGE-A3) is a member of the MAGE family of tumor antigens and a relevant candidate for use in cancer immunotherapy. However, not all tumors express MAGE-A3, and closely related members of the MAGE family can be co-expressed with MAGE-A3 in the same tumor. Therefore, in the frame of MAGE-A3 clinical trials, it appeared necessary to evaluate tumors for MAGE-A3 expression with a highly specific quantitative assay to select patients who are eligible for anti-MAGE-A3 immunotherapy treatment. Herein, we describe the development and validation of a quantitative real-time RT-PCR (RT-qPCR) assay for the determination of MAGEA3 gene expression in tumor tissues. In the early phases of development, the designed primers and probe were not able to distinguish between MAGE-A3 and MAGE-A6. To ensure the specificity for MAGE-A3 over MAGE-A6, our strategy was to use a 5'-nuclease probe (or hydrolysis probe). The final assay was shown to be specific and linear within the analytical range, with an acceptable CV for repeatability and intermediate precision. When compared with a reference semiquantitative RT-PCR assay, the two methods were in good agreement, with only 4.23% of the samples giving discordant results. In conclusion, we have developed a MAGE-A3-specific RT-qPCR assay, compatible with a high-throughput setting for the estimation of MAGEA3 gene expression in present and future clinical trials., (Copyright © 2015 American Society for Investigative Pathology and the Association for Molecular Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
40. Tumor mouse model confirms MAGE-A3 cancer immunotherapeutic as an efficient inducer of long-lasting anti-tumoral responses.
- Author
-
Gérard C, Baudson N, Ory T, and Louahed J
- Subjects
- Animals, Antigens, Neoplasm metabolism, Cancer Vaccines immunology, Cell Line, Tumor, CpG Islands, Cytokines metabolism, Humans, Mice, Mice, Inbred C57BL, Recombinant Proteins pharmacology, Antigens, Neoplasm genetics, Antineoplastic Agents pharmacology, Immunotherapy methods, Neoplasm Proteins genetics, Neoplasms immunology, Neoplasms therapy
- Abstract
Purpose: MAGE-A3 is a potential target for immunotherapy due to its tumor-specific nature and expression in several tumor types. Clinical data on MAGE-A3 immunotherapy have raised many questions that can only be addressed by using animal models. In the present study, different aspects of the murine anti-tumor immune responses induced by a recombinant MAGE-A3 protein (recMAGE-A3) in combination with different immunostimulants (AS01, AS02, CpG7909 or AS15) were investigated., Experimental Design and Results: Based on cytokine profile analyses and protection against challenge with MAGE-A3-expressing tumor, the combination recMAGE-A3+AS15 was selected for further experimental work, in particular to study the mechanisms of anti-tumor responses. By using MHC class I-, MHC class II-, perforin-, B-cell- and IFN-γ- knock-out mice and CD4+ T cell-, CD8+ T cell- and NK cell- depleted mice, we demonstrated that CD4+ T cells and NK cells are the main anti-tumor effectors, and that IFN-γ is a major effector molecule. This mouse tumor model also established the need to repeat recMAGE-A3+AS15 injections to sustain efficient anti-tumor responses. Furthermore, our results indicated that the efficacy of tumor rejection by the elicited anti-MAGE-A3 responses depends on the proportion of tumor cells expressing MAGE-A3., Conclusions: The recMAGE-A3+AS15 cancer immunotherapy efficiently induced an antigen-specific, functional and long-lasting immune response able to recognize and eliminate MAGE-A3-expressing tumor cells up to several months after the last immunization in mice. The data highlighted the importance of the immunostimulant to induce a Th1-type immune response, as well as the key role played by IFN-γ, CD4+ T cells and NK cells in the anti-tumoral effect.
- Published
- 2014
- Full Text
- View/download PDF
41. MEK inhibition, alone or in combination with BRAF inhibition, affects multiple functions of isolated normal human lymphocytes and dendritic cells.
- Author
-
Vella LJ, Pasam A, Dimopoulos N, Andrews M, Knights A, Puaux AL, Louahed J, Chen W, Woods K, and Cebon JS
- Subjects
- Antigens, Neoplasm immunology, Cell Differentiation, Cell Proliferation drug effects, Cross-Priming immunology, Cytokines biosynthesis, Dendritic Cells cytology, Epitopes, T-Lymphocyte immunology, Humans, Imidazoles pharmacology, Lipopolysaccharides immunology, Lymphocyte Activation drug effects, Lymphocyte Subsets drug effects, Lymphocyte Subsets immunology, Lymphocyte Subsets metabolism, Oximes pharmacology, Pyridones pharmacology, Pyrimidinones pharmacology, Dendritic Cells drug effects, Dendritic Cells metabolism, Lymphocytes drug effects, Lymphocytes metabolism, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins B-raf antagonists & inhibitors
- Abstract
Combination therapy with BRAF and MEK inhibition is currently in clinical development for the treatment of BRAF-mutated malignant melanoma. BRAF inhibitors are associated with enhanced antigen-specific T-lymphocyte recognition in vivo. Consequently, BRAF inhibition has been proposed as proimmunogenic and there has been considerable enthusiasm for combining BRAF inhibition with immunotherapy. MEK inhibitors inhibit ERK phosphorylation regardless of BRAF mutational status and have been reported to impair T-lymphocyte and modulate dendritic cell function. In this study, we investigate the effects on isolated T lymphocytes and monocyte-derived dendritic cells (moDC) of a MEK (trametinib) and BRAF (dabrafenib) inhibitor combination currently being evaluated in a randomized controlled clinical trial. The effects of dabrafenib and trametinib, alone and in combination, were studied on isolated normal T lymphocytes and moDCs. Lymphocyte viability, together with functional assays including proliferation, cytokine production, and antigen-specific expansion, were assessed. MoDC phenotype in response to lipopolysaccharide stimulation was evaluated by flow cytometry, as were effects on antigen cross-presentation. Dabrafenib did not have an impact on T lymphocytes or moDCs, whereas trametinib alone or in combination with dabrafenib suppressed T-lymphocyte proliferation, cytokine production, and antigen-specific expansion. However, no significant decrease in CD4(+) or CD8(+) T-lymphocyte viability was observed following kinase inhibition. MoDC cross-presentation was suppressed in association with enhanced maturation following combined inhibition of MEK and BRAF. The results of this study demonstrate that MEK inhibition, alone or in combination with BRAF inhibition, can modulate immune cell function, and further studies in vivo will be required to evaluate the potential clinical impact of these findings.
- Published
- 2014
- Full Text
- View/download PDF
42. Predictive gene signature in MAGE-A3 antigen-specific cancer immunotherapy.
- Author
-
Ulloa-Montoya F, Louahed J, Dizier B, Gruselle O, Spiessens B, Lehmann FF, Suciu S, Kruit WH, Eggermont AM, Vansteenkiste J, and Brichard VG
- Subjects
- Adult, Aged, Aged, 80 and over, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung immunology, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Lung Neoplasms genetics, Lung Neoplasms immunology, Male, Melanoma genetics, Melanoma immunology, Middle Aged, Molecular Targeted Therapy methods, Odds Ratio, Predictive Value of Tests, Protein Array Analysis, Recombinant Proteins therapeutic use, Reverse Transcriptase Polymerase Chain Reaction, Skin Neoplasms genetics, Skin Neoplasms immunology, Treatment Outcome, Adjuvants, Immunologic therapeutic use, Antigens, Neoplasm immunology, Biomarkers, Tumor immunology, Cancer Vaccines therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Immunotherapy methods, Lung Neoplasms drug therapy, Melanoma drug therapy, Neoplasm Proteins immunology, Skin Neoplasms drug therapy, Transcriptome
- Abstract
Purpose: To detect a pretreatment gene expression signature (GS) predictive of response to MAGE-A3 immunotherapeutic in patients with metastatic melanoma and to investigate its applicability in a different cancer setting (adjuvant therapy of resected early-stage non-small-cell lung cancer [NSCLC])., Patients and Methods: Patients were participants in two phase II studies of the recombinant MAGE-A3 antigen combined with an immunostimulant (AS15 or AS02B). mRNA from melanoma biopsies was analyzed by microarray analysis and quantitative polymerase chain reaction. These results were used to identify and cross-validate the GS, which was then applied to the NSCLC data., Results: In the patients with melanoma, 84 genes were identified whose expression was potentially associated with clinical benefit. This effect was strongest when the immunostimulant AS15 was included in the immunotherapy (hazard ratio [HR] for overall survival, 0.37; 95% CI, 0.13 to 1.05; P = .06) and was less strong with the other immunostimulant AS02B (HR, 0.84; 95% CI, 0.36 to 1.97; P = .70). The same GS was then used to predict the outcome for patients with resected NSCLC treated with MAGE-A3 plus AS02B; actively treated GS-positive patients showed a favorable disease-free interval compared with placebo-treated GS-positive patients (HR, 0.42; 95% CI, 0.17 to 1.03; P = .06), whereas among GS-negative patients, no such difference was found (HR, 1.17; 95% CI, 0.59 to 2.31; P = .65). The genes identified were mainly immune related, involving interferon gamma pathways and specific chemokines, suggesting that their pretreatment expression influences the tumor's immune microenvironment and the patient's clinical response., Conclusion: An 84-gene GS associated with clinical response for MAGE-A3 immunotherapeutic was identified in metastatic melanoma and confirmed in resected NSCLC.
- Published
- 2013
- Full Text
- View/download PDF
43. Selection of immunostimulant AS15 for active immunization with MAGE-A3 protein: results of a randomized phase II study of the European Organisation for Research and Treatment of Cancer Melanoma Group in Metastatic Melanoma.
- Author
-
Kruit WH, Suciu S, Dreno B, Mortier L, Robert C, Chiarion-Sileni V, Maio M, Testori A, Dorval T, Grob JJ, Becker JC, Spatz A, Eggermont AM, Louahed J, Lehmann FF, Brichard VG, and Keilholz U
- Subjects
- Adjuvants, Immunologic administration & dosage, Adult, Aged, Aged, 80 and over, Cancer Vaccines administration & dosage, Cancer Vaccines immunology, Female, Humans, Injections, Intramuscular, Kaplan-Meier Estimate, Male, Middle Aged, Recombinant Proteins therapeutic use, Treatment Outcome, Adjuvants, Immunologic therapeutic use, Antigens, Neoplasm immunology, Antigens, Neoplasm therapeutic use, Cancer Vaccines therapeutic use, Melanoma drug therapy, Melanoma immunology, Neoplasm Proteins immunology, Neoplasm Proteins therapeutic use, Skin Neoplasms drug therapy, Skin Neoplasms immunology, Vaccination methods
- Abstract
Purpose: Active immunization against the tumor-specific MAGE-A3 antigen is followed by a few but impressive and durable clinical responses. This randomized phase II trial evaluated two different immunostimulants combined with the MAGE-A3 protein to investigate whether a more robust and persistent immune response could be associated with increased clinical benefit., Patients and Methods: Patients with MAGE-A3-positive stage III or IV M1a melanoma were randomly assigned to receive the MAGE-A3 protein combined either with AS02B or with AS15 immunostimulant. Clinical end points were toxicity and rates of objective clinical responses, progression-free survival (PFS), and overall survival (OS)., Results: Seventy-five patients were treated, with 36 eligible patients per arm. Both treatments were well tolerated. In the AS15 arm, four objective responses were observed (three complete responses and one partial response) versus one partial response in the AS02B arm. In the AS15 and AS02B arms, the PFS rates after 6 months were 25% and 14%, respectively; and the median OS times were 33 months and 19.9 months, respectively, with a median observation period of 48 months. Antibodies against MAGE-A3, found in all patients, showed three-fold higher titers in the AS15 arm. The anti-MAGE-A3 cellular response was also more pronounced in the AS15 arm., Conclusion: In the MAGE-A3+AS15 arm, clinical activity was higher and the immune response more robust. Therefore, the AS15 immunostimulant was selected for combination with the MAGE-A3 protein in phase III trials.
- Published
- 2013
- Full Text
- View/download PDF
44. Cancer regression and neurological toxicity cases after anti-MAGE-A3 TCR gene therapy.
- Author
-
Brichard VG, Louahed J, and Clay TM
- Subjects
- Female, Humans, Male, Antigens, Neoplasm, Genetic Therapy methods, Immunotherapy, Adoptive, Melanoma therapy, Neoplasm Proteins, Receptors, Antigen, T-Cell
- Published
- 2013
- Full Text
- View/download PDF
45. Gene signature in melanoma associated with clinical activity: a potential clue to unlock cancer immunotherapy.
- Author
-
Gajewski TF, Louahed J, and Brichard VG
- Subjects
- Animals, Cancer Vaccines immunology, Gene Expression Profiling, Humans, Immunity genetics, Melanoma immunology, Vaccines, Subunit immunology, Vaccines, Subunit therapeutic use, Cancer Vaccines therapeutic use, Immunotherapy methods, Melanoma genetics, Melanoma therapy
- Abstract
Immunotherapeutic approaches for melanoma and other cancers can impart profound clinical benefit but only for a subset of patients. Interpatient heterogeneity could, in principle, be due to somatic differences in the tumor between individuals or alternatively be accounted for distinct germline polymorphisms in immunoregulatory genes of the host. Analysis of these possibilities has been initiated by investigating gene expression profiling of the tumor microenvironment in the context of clinical trials of cancer vaccines. Distinct gene expression profiles have been identified on pretreatment biopsies that are associated with a positive or negative clinical outcome. These observations suggest that such profiling might be useful as a predictive biomarker for clinical benefit from vaccines and other immunotherapy approaches, and analysis of specific gene products has begun to suggest new therapeutic interventions to overcome mechanisms of tumor resistance.
- Published
- 2010
- Full Text
- View/download PDF
46. IL-9 promotes IL-13-dependent paneth cell hyperplasia and up-regulation of innate immunity mediators in intestinal mucosa.
- Author
-
Steenwinckel V, Louahed J, Lemaire MM, Sommereyns C, Warnier G, McKenzie A, Brombacher F, Van Snick J, and Renauld JC
- Subjects
- Animals, Biomarkers, Hyperplasia genetics, Hyperplasia immunology, Hyperplasia metabolism, Hyperplasia pathology, Interleukin-13 deficiency, Interleukin-13 genetics, Interleukin-13 metabolism, Interleukin-9 genetics, Interleukin-9 metabolism, Kinetics, Mice, Mice, Knockout, Phospholipases A2 metabolism, Ribonuclease, Pancreatic metabolism, Immunity, Innate immunology, Interleukin-13 immunology, Interleukin-9 immunology, Intestinal Mucosa immunology, Paneth Cells immunology, Up-Regulation immunology
- Abstract
IL-9 contributes to lung inflammatory processes such as asthma, by promoting mast cell differentiation, B cell activation, eosinophilia, and mucus production by lung epithelial cells. The observation that IL-9 overexpressing mice show increased mast cell numbers in the intestinal mucosa suggests that this cytokine might also play a role in intestinal inflammation. In colons from IL-9 transgenic mice, the expression of Muc2, a major intestinal mucin gene, was up-regulated, together with that of CLCA3 chloride channel and resistin like alpha, which are goblet cell-associated genes. Additional IL-9 up-regulated genes were identified and included innate immunity genes such as angiogenin 4 and the PLA2g2a phospholipase A(2), which are typical Paneth cell markers. Histochemical staining of Paneth cells by phloxine/tartrazine showed that IL-9 induces Paneth cell hyperplasia in Lieberkühn glands of the small intestine, and in the colonic mucosa, where this cell type is normally absent. Expression of Paneth cell markers, including angiogenin 4, PLA2g2a, and cryptdins, was induced in the colon of wild-type mice after two to four daily administrations of IL-9. By crossing IL-9 transgenic mice with IL-13(-/-) mice, or by injecting IL-9 into IL-4R(-/-) mice, we showed that IL-13 was required for the up-regulation of these Paneth cell-specific genes by IL-9. Taken together, our data indicate that Paneth cell hyperplasia and expression of their various antimicrobial products contribute to the immune response driven by TH2 cytokines, such as IL-9 and IL-13 in the intestinal mucosa.
- Published
- 2009
- Full Text
- View/download PDF
47. The delivery site of a monovalent influenza vaccine within the respiratory tract impacts on the immune response.
- Author
-
Minne A, Louahed J, Mehauden S, Baras B, Renauld JC, and Vanbever R
- Subjects
- Administration, Inhalation, Administration, Intranasal, Administration, Oral, Animals, Antibodies, Viral biosynthesis, Female, Immunity, Cellular, Immunization methods, Immunoglobulin A, Secretory biosynthesis, Immunoglobulin G biosynthesis, Immunoglobulin G blood, Influenza A Virus, H3N2 Subtype immunology, Influenza Vaccines immunology, Injections, Intramuscular, Mice, Mice, Inbred BALB C, Mice, Inbred Strains, Th1 Cells immunology, Influenza Vaccines administration & dosage, Respiratory System immunology
- Abstract
Pulmonary vaccination is a promising immunization route. However, there still remains a crucial need to characterize the different parameters affecting the efficacy of inhaled vaccination. This study aimed at assessing the impact of antigen distribution within the respiratory tract on the immune response to a monovalent A/Panama/2007/99 H3N2 influenza split virus vaccine administered to BALB/c mice. Varying the administration technique allowed the targeting of the vaccine to different sites of the mouse respiratory tract, i.e. the nasal cavity, the upper or central airways, or the deep lung. This targeting was verified by using ovalbumin as a tracer compound. The immune responses generated following influenza vaccine administration to the different respiratory tract sites were compared to each other and to those elicited by intramuscular and peroral intragastric immunization. Delivery of the vaccine to the different respiratory regions generated systemic, local and cellular virus-specific immune responses, which increased with the depth of vaccine deposition, culminating in deep-lung vaccination. The latter induced virus-specific serum immunoglobulin G and neutralizing antibody titres as elevated as intramuscular vaccination, whereas the production of mucosal secretory immunoglobulin A was significantly superior in deep-lung-vaccinated animals. The analysis of cytokines secreted by mononuclear cells during an in vitro recall response indicated that deep-lung vaccination induced a local shift of the cellular immune response towards a T helper type 1 phenotype as compared to intramuscular vaccination. In conclusion, antigen distribution within the respiratory tract has a major effect on the immune response, with the deep lung as the best target for inhaled influenza vaccination.
- Published
- 2007
- Full Text
- View/download PDF
48. IL-13 mediates in vivo IL-9 activities on lung epithelial cells but not on hematopoietic cells.
- Author
-
Steenwinckel V, Louahed J, Orabona C, Huaux F, Warnier G, McKenzie A, Lison D, Levitt R, and Renauld JC
- Subjects
- Animals, Asthma genetics, Asthma metabolism, Asthma pathology, Chemokine CCL11, Chemokines, CC biosynthesis, Chemokines, CC immunology, Epithelial Cells metabolism, Epithelial Cells pathology, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells pathology, Interleukin-13 deficiency, Interleukin-9 deficiency, Leukocytes immunology, Leukocytes metabolism, Leukocytes pathology, Lung immunology, Lung metabolism, Lung pathology, Mice, Mice, Knockout, Mucus immunology, Mucus metabolism, Pulmonary Eosinophilia genetics, Pulmonary Eosinophilia metabolism, Pulmonary Eosinophilia pathology, Receptors, Interleukin-9 deficiency, Receptors, Interleukin-9 immunology, Up-Regulation immunology, Asthma immunology, Epithelial Cells immunology, Hematopoietic Stem Cells immunology, Interleukin-13 immunology, Interleukin-9 immunology, Pulmonary Eosinophilia immunology
- Abstract
Increased IL-9 expression, either systemically or under the control of lung-specific promoter, induces an asthma-like phenotype, including mucus overproduction, mastocytosis, lung eosinophilia, and airway hyperresponsiveness. These activities correlate with increased production of other Th2 cytokines such as IL-4, IL-5, and IL-13 in IL-9 Tg mice. To determine the exact role of IL-13 in this phenotype, mice overexpressing IL-9 were crossed with IL-13-deficient mice. In these animals, IL-9 could still induce mastocytosis and B lymphocyte infiltration of the lungs. Although IL-9-induced eosinophilia in the peritoneal cavity was not diminished in the absence of IL-13, IL-13 was required for IL-9 to increase eotaxin expression and lung eosinophilia. Mucus production and up-regulation of lung epithelial genes upon IL-9 overexpression were completely abolished in the absence of IL-13. Using hemopoietic cell transfer experiments with recipients that overexpressed IL-9 but were deficient in the IL-9 receptor (IL-9R), we could demonstrate that the effect of IL-9 on lung epithelial cells is indirect and could be fully restored by transfer of hemopoietic cells expressing IL-9R. Mucus production by lung epithelial cells was only up-regulated when hemopoietic cells simultaneously expressed functional IL-9R and IL-13 genes, indicating that IL-13 is not a cofactor but a direct mediator of the effect of IL-9 on lung epithelial cells. Taken together, these data indicate that IL-9 can promote asthma through IL-13-independent pathways via expansion of mast cells, eosinophils, and B cells, and through induction of IL-13 production by hemopoietic cells for mucus production and recruitment of eosinophils by lung epithelial cells.
- Published
- 2007
- Full Text
- View/download PDF
49. B lymphocytes are critical for lung fibrosis control and prostaglandin E2 regulation in IL-9 transgenic mice.
- Author
-
Arras M, Louahed J, Simoen V, Barbarin V, Misson P, van den Brûle S, Delos M, Knoops L, Renauld JC, Lison D, and Huaux F
- Subjects
- Animals, Bronchoalveolar Lavage Fluid chemistry, Collagen metabolism, Cyclooxygenase 1 metabolism, Cyclooxygenase 2 metabolism, Dinoprostone biosynthesis, Female, Interleukin-9 genetics, Interleukin-9 immunology, Intramolecular Oxidoreductases metabolism, Lung cytology, Lung drug effects, Lung pathology, Macrophages, Alveolar metabolism, Macrophages, Peritoneal enzymology, Macrophages, Peritoneal metabolism, Mice, Mice, Transgenic, Prostaglandin-E Synthases, Silicon Dioxide pharmacology, Subcellular Fractions, B-Lymphocytes immunology, Dinoprostone metabolism, Gene Expression Regulation, Interleukin-9 metabolism, Pulmonary Fibrosis immunology
- Abstract
We previously showed that overexpression of IL-9 controls lung fibrosis induced by silica particles in mice (Arras and colleagues; Am J Respir Cell Mol Biol 2001;24:368-375). This protection was associated with an expansion of lung B lymphocytes. To explore the contribution of these cells in the protective effect of IL-9, we crossed IL-9 transgenic (IL-9+) and B-deficient (B-) mice. The antifibrotic effect of IL-9 was abolished in mice deficient in B lymphocytes (B-IL-9+) and restored by reconstituting these mice with B lymphocytes. The expression of the antifibrotic mediator prostaglandin (PG)E2 was markedly increased in the lung of IL-9+ mice at baseline, and similarly high levels were found in both wild-type and transgenic strains upon silica treatment. This PGE2 expression was completely abolished in B- mice, both at baseline and upon silica administration. In vitro, alveolar and peritoneal macrophages from IL-9+ mice had an increased capacity to produce PGE2 in response to LPS or silica. This capacity was markedly reduced in macrophages obtained from B- mice and restored by co-incubating macrophages with B lymphocytes from IL-9+ mice. The increased PGE2 response of IL-9+ macrophages was dependent on cyclooxygenase 2 expression, based on transcript analysis and inhibition by NS398. We conclude that B lymphocytes are essential for the protection against lung fibrosis and macrophage overexpression of PGE2 in IL-9 transgenic animals.
- Published
- 2006
- Full Text
- View/download PDF
50. IL-9 promotes but is not necessary for systemic anaphylaxis.
- Author
-
Knoops L, Louahed J, Van Snick J, and Renauld JC
- Subjects
- Anaphylaxis enzymology, Animals, Chymases, Disease Models, Animal, Female, Immunization, Interleukin-9 genetics, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Ovalbumin immunology, Passive Cutaneous Anaphylaxis immunology, Receptors, Interleukin deficiency, Receptors, Interleukin genetics, Receptors, Interleukin-9, Serine Endopeptidases blood, Anaphylaxis etiology, Anaphylaxis immunology, Interleukin-9 immunology
- Abstract
Anaphylaxis represents an extreme form of allergic reaction, consisting of a sensitization phase during which allergen-specific IgE are produced and an acute effector phase triggered by allergen-induced degranulation of mast cells. We studied the role of IL-9, a Th2 cytokine implicated in asthma, in different models of murine anaphylaxis. Using a passive model of systemic anaphylaxis, in which anti-DNP IgE Abs were administered before challenge with DNP-BSA, we found that IL-9-transgenic mice or wild-type mice treated with IL-9 for 5 days were highly sensitive to fatal anaphylaxis. This effect was reproduced in both anaphylaxis-susceptible and -resistant backgrounds (FVB/N or [FVB/N x BALB/c] F(1) mice, respectively) and correlated with increased serum concentrations of mouse mast cell protease-1 level, a protein released upon mast cells degranulation. By contrast, IL-9 did not increase the susceptibility to passive cutaneous anaphylaxis. IL-9 expression also increased the susceptibility to fatal anaphylaxis when mice were sensitized by immunization against OVA before challenge with the same Ag. In this model, serum from sensitized, IL-9-transgenic mice was more potent in transferring susceptibility to OVA challenge into naive mice, indicating that IL-9 also promotes the sensitization stage. Finally, using IL-9R-deficient mice, we found that despite its anaphylaxis-promoting activity, IL-9 is dispensable for development of both passive and active anaphylaxis, at least in the C57BL/6 mouse background. Taken together, the data reported in this study indicate that IL-9 promotes systemic anaphylaxis reactions, acting at both the sensitization and effector stages, but is not absolutely required for this process.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.