Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Monika Lusky is active.

Publication


Featured researches published by Monika Lusky.


Immunology Letters | 1997

Gene therapy with recombinant adenovirus vectors: evaluation of the host immune response

M. Christ; Monika Lusky; Fabienne Stoeckel; Dominique Dreyer; Annick Dieterlé; Anne-Isabelle Michou; Andrea Pavirani; Majid Mehtali

E1, E3-deleted, replication-deficient recombinant adenoviruses are widely studied as vectors for their capacity to transfer therapeutic genes in vivo. They can infect a wide variety of dividing and quiescent cells from different organs and possess a large packaging capacity. One of the major limitations in the use of these vectors for gene therapy is the transient expression of the transgene in vivo and the poor transduction efficiency when re-administered. Despite the deletion of the viral E1 region, low level of early and late viral genes are expressed in vivo. Thus, viral antigens plus those derived from transgene expression in transduced cells contribute to cellular immune responses leading to the destruction of these cells. Production of anti-adenovirus antibodies, the cellular immune response as well as the early non-specific clearance of the vectors, constitute barriers to successful gene therapy. New vectors have been derived with additional deletions in the E2a or the E4 regions. Such second generation vectors were evaluated in vivo. These studies have revealed the complexity of the immune mechanisms elicited by these vectors and the importance of several parameters in these evaluations (i.e. mouse strains, nature of the transgene, route of administration...). In order to inhibit the production of neutralizing antibodies to adenovirus that prevent from further readministration of the vectors, immunosuppressive strategies were undertaken. Treatment regimens with immunosuppressive drugs (cyclophosphamide, FK506) or with monoclonal antibodies that block either the T cell receptor or costimulation pathways allow prolonged transgene expression and/or readministration of adenoviral vectors. In addition, transduction efficiencies may be increased by transiently inhibiting non-specific immune mechanisms that lead to the dramatic early clearance of the vectors. Taken together, these strategies may improve further gene therapy protocols by decreasing the host immune response to adenoviral vectors.


Human Gene Therapy | 2000

Modulation of the Inflammatory Properties and Hepatotoxicity of Recombinant Adenovirus Vectors by the Viral E4 Gene Products

M. Christ; Bernard Louis; Fabienne Stoeckel; Annick Dieterlé; Linda Grave; Dominique Dreyer; Jacqueline Kintz; Dalila Ali Hadji; Monika Lusky; Majid Mehtali

Liver toxicity and inflammation were assessed in C57BL/6, CBA, and BALB/c mice injected intravenously with a series of recombinant adenoviruses deleted simultaneously in E1/E3, in E1/E3/E2A, or in E1/E3/E4. All vectors were either devoid of transgenes or carried in E1 the human CFTR cDNA under the control of the CMV promoter. Injection of the E1/E3-deleted vector induced a significant liver dystrophy and inflammatory responses that were accompanied by an increased serum transaminase concentration. The vector toxicity remained elevated on additional deletion of the E2A gene and was further enhanced when hCFTR was expressed. In contrast, additional deletion of E4 led to a reduction in hepatotoxicity, suggesting an active role of E4 gene products in liver injury. However, deletion of E4 also led to a loss of transgene expression. To identify the individual E4 product(s) involved in liver toxicity and in the regulation of transgene expression, a series of isogenic E1/E3-deleted vectors, with or without the hCFTR transgene, and containing various combinations of functional E4 open reading frames (ORFs), were evaluated in vitro and in vivo. We demonstrate that liver injury was markedly reduced with vectors containing either ORF3 alone or ORF3,4 while vectors containing ORF4, ORF6,7 or ORF3,6,7 still displayed elevated hepatotoxicity and inflammatory responses. Moreover, transgene expression was restored when ORF3,4 or ORF3,6,7 was retained in the vector. These results highlight the importance of the E4 gene products in the design of improved in vivo gene transfer vectors.


Clinical Cancer Research | 2010

Targeted chemotherapy for head and neck cancer with a chimeric oncolytic adenovirus coding for bifunctional suicide protein FCU1.

João D. Dias; Ilkka Liikanen; Kilian Guse; Johann Foloppe; Marta Sloniecka; Iulia Diaconu; Ville Rantanen; Minna Eriksson; Tanja Hakkarainen; Monika Lusky; Philippe Erbs; Sophie Escutenaire; Anna Kanerva; Sari Pesonen; Vincenzo Cerullo; Akseli Hemminki

Purpose: Transfer of prodrug activation systems into tumors by using replication-deficient viruses has been suggested to be an effective method for achieving high local and low systemic anticancer drug concentrations. However, most current suicide gene therapy strategies are still hindered by poor efficiency of in vivo gene transfer, inefficient tumor penetration, limited bystander cell killing effect, and need of large prodrug doses. We hypothesized that local amplification provided by a replication competent platform would help overcome these limitations. Experimental Design: We generated a transductionally and transcriptionally targeted oncolytic adenovirus Ad5/3-Δ24FCU1 expressing the fusion suicide gene FCU1. FCU1 encodes a bifunctional fusion protein that efficiently catalyzes the direct conversion of 5-FC, a relatively nontoxic antifungal agent, into the toxic metabolites 5-fluorouracil and 5-fluorouridine monophosphate, bypassing the natural resistance of certain human tumor cells to 5-fluorouracil. Results: We examined the efficacy of Ad5/3-Δ24FCU1 and the replication-defective control Ad5/3-FCU1 with and without 5-FC. FCU1 expression was confirmed by Western blot, whereas enzymatic conversion levels in vitro and in vivo were determined by high-performance liquid chromatography separation. Significant antitumor effect was observed in vitro and in vivo in a murine model of head and neck squamous cell carcinoma. Although we observed a decrease in viral DNA copy number in vitro and in tumors treated with Ad5/3-Δ24FCU1 and 5-FC, suggesting an effect on virus replication, the highest antitumor effect was observed for this combination. Conclusions: It seems feasible and efficacious to combine adenovirus replication to the FCU1 prodrug activation system. Clin Cancer Res; 16(9); 2540–9. ©2010 AACR.


Molecular Therapy | 2010

Phase II Clinical Trial of Intratumoral Application of TG1042 (Adenovirus-interferon-γ) in Patients With Advanced Cutaneous T-cell Lymphomas and Multilesional Cutaneous B-cell Lymphomas

Reinhard Dummer; Stefan B. Eichmüller; Sylke Gellrich; Chalid Assaf; Brigitte Dreno; Meinhard Schiller; O. Dereure; Marion Baudard; Martine Bagot; Amir Khammari; Pascal Bleuzen; Vincent Bataille; Anna Derbij; Nicole Wiedemann; Tim Waterboer; Monika Lusky; Bruce Acres; Mirjana Urosevic-Maiwald

Cutaneous lymphomas (CLs) are a heterogeneous group of lymphoproliferative disorders that are manageable by immunotherapy. Twenty-one patients were enrolled in a prospective open-label, dose-escalation multicenter study evaluating the effects of repeated TG1042 [adenovirus-interferon (IFN)-gamma] intralesional injections in patients with primary CLs, of which 18 were of T-cell and 3 of B-cell type. Repeated intralesional therapy using TG1042 consistently results in local tumor regressions in about half of treated patients and one-third of patients also in regressions in noninjected distant lesions, likely reflecting the systemic immune activation after intralesional therapy. Treatment was well tolerated with few adverse events including injection site reactions, chills, lymphopenia, and fever. Immune monitoring in the peripheral blood demonstrated systemic immune activation and the induction of antibodies against tumor antigens in some patients without clear association with clinical responses. CLs, in particular B-cell lymphomas with high objective response rates, seem to be excellent targets for this type of immunotherapy.Cutaneous lymphomas (CLs) are a heterogeneous group of lymphoproliferative disorders that are manageable by immunotherapy. Twenty-one patients were enrolled in a prospective open-label, dose-escalation multicenter study evaluating the effects of repeated TG1042 [adenovirus-interferon (IFN)-γ] intralesional injections in patients with primary CLs, of which 18 were of T-cell and 3 of B-cell type. Repeated intralesional therapy using TG1042 consistently results in local tumor regressions in about half of treated patients and one-third of patients also in regressions in noninjected distant lesions, likely reflecting the systemic immune activation after intralesional therapy. Treatment was well tolerated with few adverse events including injection site reactions, chills, lymphopenia, and fever. Immune monitoring in the peripheral blood demonstrated systemic immune activation and the induction of antibodies against tumor antigens in some patients without clear association with clinical responses. CLs, in particular B-cell lymphomas with high objective response rates, seem to be excellent targets for this type of immunotherapy.


Human Gene Therapy | 2001

Radioisotopic Imaging Allows Optimization of Adenovirus Lung Deposition for Cystic Fibrosis Gene Therapy

Sté Phanie Lerondel; Alain Le Pape; Claude Sene; Laurence Faure; Serge Bernard; P. Diot; Elena Nicolis; Majid Mehtali; Monika Lusky; Giulio Cabrini; Andrea Pavirani

Cystic fibrosis is a common, heriditary disease resulting from mutations of the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Airway transfer of the CFTR gene is a potential strategy to treat or prevent the lung pathology that is the main cause of morbidity and mortality. Among the vectors used for gene therapy, adenoviruses have shown their ability to transfer the CFTR gene to respiratory epithelial cells, using either instillation or nebulization. Our objective was to characterize the lung deposition of aerosolized adenovirus by quantitative radioisotopic imaging, the only noninvasive technique allowing in vivo quantitation of inhaled drugs. We first labeled an adenovirus expressing human CFTR with the gamma-emitting radioisotope, technetium 99m (99mTc), and determined the best labeling conditions to allow preservation of virus bioactivity. We then administered the radioaerosol to baboons, determined lung regional deposition of 99mTc-labeled adenovirus, and compared the expression of CFTR transcripts 3 and 21 days after inhalation. The expression of vector-encoded mRNA ranged from 4 to 22% with respect to the endogenous CFTR mRNA depending on the lung segments. Moreover, we have developed a model using 99mTc-DTPA (diethylenetriamine pentaacetic acid), which can be used, as an alternative to adenovirus, to determine the profile of lung deposition of the vector. This study demonstrates that scintigraphy is a useful technique to achieve optimization of gene administration to the airways.


Journal of Gene Medicine | 2000

Differential influence of the E4 adenoviral genes on viral and cellular promoters.

Linda Grave; Dominique Dreyer; Annick Dieterlé; Pierre Leroy; Anne-Isabelle Michou; Cecile Doderer; Andrea Pavirani; Monika Lusky; Majid Mehtali

Strong and stable transgene expression is fundamental to the success of recombinant adenovirus vectors in human gene therapy. However, control of transgene expression is a complex process, involving both viral and cellular factors. In this study, the influence of the E4 adenoviral region on the activity of various promoters was investigated in vitro and in vivo.


Current Gene Therapy | 2002

Transductional Targeting with Recombinant Adenovirus Vectors

Valerie Legrand; Philippe Leissner; Arend Jan Winter; Majid Mehtali; Monika Lusky

Replication-deficient adenoviruses are considered as gene delivery vectors for the genetic treatment of a variety of diseases. The ability of such vectors to mediate efficient expression of therapeutic genes in a broad spectrum of dividing and non-dividing cell types constitutes an advantage over alternative gene transfer vectors. However, this broad tissue tropism may also turn disadvantageous when genes encoding potentially harmful proteins (e.g. cytokines, toxic proteins) are expressed in surrounding normal tissues. Therefore, specific restrictions of the viral tropism would represent a significant technological advance towards safer and more efficient gene delivery vectors, in particular for cancer gene therapy applications. In this review, we summarize various strategies used to selectively modify the natural tropism of recombinant adenoviruses. The advantages, limitations and potential impact on gene therapy operations of such modified vectors are discussed.


Expert Review of Vaccines | 2010

Oncolytic vaccinia virus: a silver bullet?

Monika Lusky; Philippe Erbs; Johann Foloppe; R Bruce Acres

Currently, the most successful curative intent treatment for many cancers is surgery, yet many tumors remain inoperable. Radiotherapy can be effective and is often used in combination with surgery; for example, it is an effective treatment option for prostate cancer. However, metastatic disease is amenable to neither of these treatments and has the poorest outlook [1]. Chemotherapy is not often a curative treatment but can prolong a patient’s life, sometimes with an acceptable impact on quality of life. Targeted therapies, such as monoclonal antibodies, are now being used to target tumors specifically, for example, to reduce growth factor-dependent growth stimulation or to inhibit tumor vascularization. Despite their good specificity, the toxicity profile of monclonal antibodies has proven to be as problematic as chemo therapy. Harnessing the power of the immune response is the most recently applied weapon in the fight against cancer and it has great potential. Many cancer immunotherapeutic vaccines are now in clinical trials [2] and are beginning to show encouraging results. A landmark in cancer immunotherapy, Dendreon’s prostate cancer vaccine, sipuleucel-T (Provenge; Seattle, WA, USA) was the first antigen-specific thera peutic cancer vaccine to receive approval, in 2010, from the US FDA [101]. Early in the 20th Century, various infectious agents were tested for the treatment of cancer. The best known are Coley’s toxins, derived from a mixture of bacterial agents, which achieved some remarkable results in the treatment of sarcoma [3]. At approximately the same time, crude preparations of viruses were also being tested for the treatment of cancer. Some successes were reported and, similarly to Coley’s toxins, these were associated with fever, inflammation and innate immune responses. In the 1950s, Southam and colleagues attempted to treat cancers of various origins with better characterized viral strains, including vaccinia, mumps, West Nile, Ilheus and Bunyamwera viruses [4]. The history of oncolytic viruses is nicely described in a review by Kelly and Russell [5]. Purified viruses, in particular Newcastle Disease virus [6] and vaccinia virus (VV) [7], have been tested for their ability to ‘antigenize’ a tumor by injecting patients with viral oncolysates of tumor cells infected with a virus. Some clinical responses as a result of this treatment have been reported. Various viral constructs have also been used to target the expression of genes in tumors following various modes of administration. These include VV and other pox viruses (myxoma, racoonpox), herpes simplex virus, vesicular stomatitis virus (a rhabdovirus), Newcastle disease virus, adenovirus and a-virus. Many reports describe antitumor activity by the oncolytic mechanism of action of these viruses, exploiting the inherent capacity of oncolytic viruses to Monika Lusky


OncoTargets and Therapy | 2017

Immunogenicity of oncolytic vaccinia viruses JX-GFP and TG6002 in a human melanoma in vitro model: studying immunogenic cell death, dendritic cell maturation and interaction with cytotoxic T lymphocytes

Bernd Heinrich; Johannes Klein; Maike Delic; Katrin Goepfert; Vanessa Engel; Linda Geberzahn; Monika Lusky; Philippe Erbs; Xavier Préville; Markus Moehler

Oncolytic virotherapy is an emerging immunotherapeutic modality for cancer treatment. Oncolytic viruses with genetic modifications can further enhance the oncolytic effects on tumor cells and stimulate antitumor immunity. The oncolytic vaccinia viruses JX-594-GFP+/hGM-CSF (JX-GFP) and TG6002 are genetically modified by secreting granulocyte-macrophage colony-stimulating factor (GM-CSF) or transforming 5-fluorocytosine (5-FC) into 5-fluorouracil (5-FU). We compared their properties to kill tumor cells and induce an immunogenic type of cell death in a human melanoma cell model using SK29-MEL melanoma cells. Their influence on human immune cells, specifically regarding the activation of dendritic cells (DCs) and the interaction with the autologous cytotoxic T lymphocyte (CTL) clone, was investigated. Melanoma cells were infected with either JX-GFP or TG6002 alone or in combination with 5-FC and 5-FU. The influence of viral infection on cell viability followed a time- and multiplicity of infection dependent manner. Combination of virus treatment with 5-FU resulted in stronger reduction of cell viability. TG6002 in combination with 5-FC did not significantly strengthen the reduction of cell viability in this setting. Expression of calreticulin and high mobility group 1 protein (HMGB1), markers of immunogenic cell death (ICD), could be detected after viral infection. Accordingly, DC maturation was noted after viral oncolysis. DCs presented stronger expression of activation and maturation markers. The autologous CTL clone IVSB expressed the activation marker CD69, but viral treatment failed to enhance cytotoxicity marker. In summary, vaccinia viruses JX-GFP and TG6002 lyse melanoma cells and induce additional immunostimulatory effects to promote antitumor immune response. Further investigation in vivo is needed to consolidate the data.


PLOS ONE | 2014

TG1042 (Adenovirus-interferon-γ) in primary cutaneous B-cell lymphomas: a phase II clinical trial.

B. Dréno; Mirjana Urosevic-Maiwald; Youn H. Kim; Joan Guitart; Madeleine Duvic; O. Dereure; Amir Khammari; Anne Chantal Knol; Anna Derbij; Monika Lusky; Isabelle Didillon; Anne Marie Santoni; Bruce Acres; Vincent Bataille; Marie Pierre Chenard; Pascal Bleuzen; J.-M. Limacher; Reinhard Dummer

Rational While a variety of registered therapies exist for Cutaneous T Cell Lymphoma, no such therapy is available for Cutaneous B Cell Therapy. In this context we performed a phase II, open label, multicenter, non-comparative study to evaluate the efficacy and safety of repeated intra-lesional administrations of TG1042 (adenovirus-interferon-γ) in patients with relapsing primary cutaneous B-cell lymphomas (CBCL). Method Thirteen patients have been enrolled and received intralesional injections of TG1042 containing 5×1010 viral particles into up to six lesions simultaneously. Injections were performed on days 1, 8 and 15 of each of four consecutive 28 day cycles. Results Eleven (85%) out of 13 enrolled patients showed an objective response after injections of TG1042. Seven patients (54%) exhibited complete and four (31%) displayed partial response. The median time to disease progression in the study population was 23.5 months (range 6.25 to 26+). Most commonly observed adverse events were minor to moderate flu-like symptoms, fatigue and injection site reactions. Conclusions Our study showed that treatment with TG1042 was associated with a clinical benefit in the majority of the patients with relapsing CBCL, including tumor regression, a clinically meaningful duration of response and a good treatment tolerance. Trial Registration www.clinicaltrials.gov NCT00394693

Collaboration


Dive into the Monika Lusky's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Majid Mehtali

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge