Network


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

Hotspot


Dive into the research topics where Lydie Cassard is active.

Publication


Featured researches published by Lydie Cassard.


Blood | 2009

Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen

Laura A. Johnson; Richard A. Morgan; Mark E. Dudley; Lydie Cassard; James Chih-Hsin Yang; Marybeth S. Hughes; Udai S. Kammula; Richard E. Royal; Richard M. Sherry; John R. Wunderlich; Chyi Chia R Lee; Nicholas P. Restifo; Susan L. Schwarz; Alexandria P. Cogdill; Rachel J. Bishop; Hung Kim; Carmen C. Brewer; Susan F. Rudy; Carter VanWaes; Jeremy L. Davis; Aarti Mathur; Robert T. Ripley; Debbie Ann N Nathan; Carolyn M. Laurencot; Steven A. Rosenberg

Gene therapy of human cancer using genetically engineered lymphocytes is dependent on the identification of highly reactive T-cell receptors (TCRs) with antitumor activity. We immunized transgenic mice and also conducted high-throughput screening of human lymphocytes to generate TCRs highly reactive to melanoma/melanocyte antigens. Genes encoding these TCRs were engineered into retroviral vectors and used to transduce autologous peripheral lymphocytes administered to 36 patients with metastatic melanoma. Transduced patient lymphocytes were CD45RA(-) and CD45RO(+) after ex vivo expansion. After infusion, the persisting cells displayed a CD45RA(+) and CD45RO(-) phenotype. Gene-engineered cells persisted at high levels in the blood of all patients 1 month after treatment, responding patients with higher ex vivo antitumor reactivity than nonresponders. Objective cancer regressions were seen in 30% and 19% of patients who received the human or mouse TCR, respectively. However, patients exhibited destruction of normal melanocytes in the skin, eye, and ear, and sometimes required local steroid administration to treat uveitis and hearing loss. Thus, T cells expressing highly reactive TCRs mediate cancer regression in humans and target rare cognate-antigen-containing cells throughout the body, a finding with important implications for the gene therapy of cancer. This trial was registered at www.ClinicalTrials.gov as NCI-07-C-0174 and NCI-07-C-0175.


Blood | 2008

Tumor-specific Th17-polarized cells eradicate large established melanoma

Pawel Muranski; Andrea Boni; Paul A. Antony; Lydie Cassard; Kari R. Irvine; Andrew Kaiser; Chrystal M. Paulos; Douglas C. Palmer; Christopher E. Touloukian; Krzysztof Ptak; Luca Gattinoni; Claudia Wrzesinski; Christian S. Hinrichs; Keith W. Kerstann; Lionel Feigenbaum; Chi-Chao Chan; Nicholas P. Restifo

CD4+ T cells can differentiate into multiple effector subsets, but the potential roles of these subsets in anti-tumor immunity have not been fully explored. Seeking to study the impact of CD4+ T cell polarization on tumor rejection in a model mimicking human disease, we generated a new MHC class II-restricted, T-cell receptor (TCR) transgenic mouse model in which CD4+ T cells recognize a novel epitope in tyrosinase-related protein 1 (TRP-1), an antigen expressed by normal melanocytes and B16 murine melanoma. Cells could be robustly polarized into Th0, Th1, and Th17 subtypes in vitro, as evidenced by cytokine, chemokine, and adhesion molecule profiles and by surface markers, suggesting the potential for differential effector function in vivo. Contrary to the current view that Th1 cells are most important in tumor rejection, we found that Th17-polarized cells better mediated destruction of advanced B16 melanoma. Their therapeutic effect was critically dependent on interferon-gamma (IFN-gamma) production, whereas depletion of interleukin (IL)-17A and IL-23 had little impact. Taken together, these data indicate that the appropriate in vitro polarization of effector CD4+ T cells is decisive for successful tumor eradication. This principle should be considered in designing clinical trials involving adoptive transfer-based immunotherapy of human malignancies.


Nature Medicine | 2009

Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells

Luca Gattinoni; Xiao-Song Zhong; Douglas C. Palmer; Yun Ji; Christian S. Hinrichs; Zhiya Yu; Claudia Wrzesinski; Andrea Boni; Lydie Cassard; Lindsay M. Garvin; Chrystal M. Paulos; Pawel Muranski; Nicholas P. Restifo

Self-renewing cell populations such as hematopoietic stem cells and memory B and T lymphocytes might be regulated by shared signaling pathways. The Wnt–β-catenin pathway is an evolutionarily conserved pathway that promotes hematopoietic stem cell self-renewal and multipotency by limiting stem cell proliferation and differentiation, but its role in the generation and maintenance of memory T cells is unknown. We found that induction of Wnt–β-catenin signaling by inhibitors of glycogen sythase kinase-3β or the Wnt protein family member Wnt3a arrested CD8+ T cell development into effector cells. By blocking T cell differentiation, Wnt signaling promoted the generation of CD44lowCD62LhighSca-1highCD122highBcl-2high self-renewing multipotent CD8+ memory stem cells with proliferative and antitumor capacities exceeding those of central and effector memory T cell subsets. These findings reveal a key role for Wnt signaling in the maintenance of stemness in mature memory CD8+ T cells and have major implications for the design of new vaccination strategies and adoptive immunotherapies.


Journal of Clinical Investigation | 2007

Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8 + T cells via TLR4 signaling

Chrystal M. Paulos; Claudia Wrzesinski; Andrew Kaiser; Christian S. Hinrichs; Marcello Chieppa; Lydie Cassard; Douglas C. Palmer; Andrea Boni; Pawel Muranski; Zhiya Yu; Luca Gattinoni; Paul A. Antony; Steven A. Rosenberg; Nicholas P. Restifo

Lymphodepletion with total body irradiation (TBI) increases the efficacy of adoptively transferred tumor-specific CD8(+) T cells by depleting inhibitory lymphocytes and increasing homeostatic cytokine levels. We found that TBI augmented the function of adoptively transferred CD8(+) T cells in mice genetically deficient in all lymphocytes, indicating the existence of another TBI mechanism of action. Additional investigation revealed commensal gut microflora in the mesenteric lymph nodes and elevated LPS levels in the sera of irradiated mice. These findings correlated with increased dendritic cell activation and heightened levels of systemic inflammatory cytokines. Reduction of host microflora using antibiotics, neutralization of serum LPS using polymyxin B, or removal of LPS signaling components using mice genetically deficient in CD14 and TLR4 reduced the beneficial effects of TBI on tumor regression. Conversely, administration of microbial ligand-containing serum or ultrapure LPS from irradiated animals to nonirradiated antibody-lymphodepleted mice enhanced CD8(+) T cell activation and improved tumor regression. Administration of ultrapure LPS to irradiated animals further enhanced the number and function of the adoptively transferred cells, leading to long-term cure of mice with large B16F10 tumors and enhanced autoimmune vitiligo. Thus, disruption of the homeostatic balance between the host and microbes can enhance cell-based tumor immunotherapy.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Adoptively transferred effector cells derived from naïve rather than central memory CD8+ T cells mediate superior antitumor immunity

Christian S. Hinrichs; Zachary A. Borman; Lydie Cassard; Luca Gattinoni; Rosanne Spolski; Zhiya Yu; Luis Sanchez-Perez; Pawel Muranski; Steven J. Kern; Carol Logun; Douglas C. Palmer; Yun Ji; Robert N. Reger; Warren J. Leonard; Robert L. Danner; Steven A. Rosenberg; Nicholas P. Restifo

Effector cells derived from central memory CD8+ T cells were reported to engraft and survive better than those derived from effector memory populations, suggesting that they are superior for use in adoptive immunotherapy studies. However, previous studies did not evaluate the relative efficacy of effector cells derived from naïve T cells. We sought to investigate the efficacy of tumor-specific effector cells derived from naïve or central memory T-cell subsets using transgenic or retrovirally transduced T cells engineered to express a tumor-specific T-cell receptor. We found that naïve, rather than central memory T cells, gave rise to an effector population that mediated superior antitumor immunity upon adoptive transfer. Effector cells developed from naïve T cells lost the expression of CD62L more rapidly than those derived from central memory T cells, but did not acquire the expression of KLRG-1, a marker for terminal differentiation and replicative senescence. Consistent with this KLRG-1− phenotype, naïve-derived cells were capable of a greater proliferative burst and had enhanced cytokine production after adoptive transfer. These results indicate that insertion of genes that confer antitumor specificity into naïve rather than central memory CD8+ T cells may allow superior efficacy upon adoptive transfer.


Blood | 2009

Type 17 CD8+ T cells display enhanced antitumor immunity

Christian S. Hinrichs; Andrew Kaiser; Chrystal M. Paulos; Lydie Cassard; Luis Sanchez-Perez; Bianca Heemskerk; Claudia Wrzesinski; Zachary A. Borman; Pawel Muranski; Nicholas P. Restifo

Interleukin-17 (IL-17)-secreting CD8(+) T cells have been described, but they have not been thoroughly studied and they do not have a known role in cancer immunotherapy. We skewed CD8(+) T cells to secrete IL-17 through priming in Th17-polarizing conditions. IL-17-producing CD8(+) T cells demonstrated reduced expression of Eomes and diminished cytolytic differentiation in vitro. However, after adoptive transfer, these cells converted to interferon-gamma-producing effector cells and mediated regression of large, established tumors. This improved antitumor immunity was associated with increased expression of IL-7R-alpha, decreased expression of killer cell lectin-like receptor G1, and enhanced persistence of the transferred cells. This report is the first description of a cancer therapy with IL-17-secreting CD8(+) T cells. These findings have implications for the improvement of CD8(+) T cell-based adoptive immunotherapy.


Gene Therapy | 2008

Development of optimal bicistronic lentiviral vectors facilitates high-level TCR gene expression and robust tumor cell recognition

Shicheng Yang; Cyrille J. Cohen; Peter D. Peng; Yangbing Zhao; Lydie Cassard; Zhiya Yu; Zhili Zheng; Stephanie Jones; Nicholas P. Restifo; Steven A. Rosenberg; Richard A. Morgan

In human gene therapy applications, lentiviral vectors may have advantages over γ-retroviral vectors in several areas, including the ability to transduce nondividing cells, resistance to gene silencing and a potentially safer integration site profile. However, unlike γ-retroviral vectors it has been problematic to drive the expression of multiple genes efficiently and coordinately with approaches such as internal ribosome entry sites or dual promoters. Using different 2A peptides, lentiviral vectors expressing two-gene T-cell receptors directed against the melanoma differentiation antigens gp100 and MART-1 were constructed. We demonstrated that addition of amino-acid spacer sequences (GSG or SGSG) before the 2A sequence is a prerequisite for efficient synthesis of biologically active T-cell receptors and that addition of a furin cleavage site followed by a V5 peptide tag yielded optimal T-cell receptor gene expression. Furthermore, we determined that the furin cleavage site was recognized in lymphocytes and accounted for removal of residual 2A peptides at the post-translational level with an efficiency of 20–30%, which could not be increased by addition of multiple furin cleavage sites. The novel bicistronic lentiviral vector developed herein afforded robust anti-melanoma activities to engineered peripheral blood lymphocytes, including cytokine secretion, cell proliferation and lytic activity. Such optimal vectors may have immediate applications in cancer gene therapy.


Clinical Cancer Research | 2007

Toll-like Receptors in Tumor Immunotherapy

Chrystal M. Paulos; Andrew Kaiser; Claudia Wrzesinski; Christian S. Hinrichs; Lydie Cassard; Andrea Boni; Pawel Muranski; Luis Sanchez-Perez; Douglas C. Palmer; Zhiya Yu; Paul A. Antony; Luca Gattinoni; Steven A. Rosenberg; Nicholas P. Restifo

Lymphodepletion with chemotherapeutic agents or total body irradiation (TBI) before adoptive transfer of tumor-specific T cells is a critical advancement in the treatment of patients with melanoma. More than 50% of patients that are refractory to other treatments experience an objective or curative response with this approach. Emerging data indicate that the key mechanisms underlying how TBI augments the functions of adoptively transferred T cells include (a) the depletion of regulatory T cells (Treg) and myeloid-derived suppressor cells that limit the function and proliferation of adoptively transferred cells; (b) the removal of immune cells that act as “sinks” for homeostatic cytokines, whose levels increase after lymphodepletion; and (c) the activation of the innate immune system via Toll-like receptor 4 signaling, which is engaged by microbial lipopolysaccharide that translocated across the radiation-injured gut. Here, we review these mechanisms and focus on the effect of Toll-like receptor agonists in adoptive immunotherapy. We also discuss alternate regimens to chemotherapy or TBI, which might be used to safely treat patients with advanced disease and promote tumor regression.


Journal of Clinical Investigation | 2002

Modulation of tumor growth by inhibitory Fcγ receptor expressed by human melanoma cells

Lydie Cassard; Joel Cohen-Solal; Annie Galinha; Xavier Sastre-Garau; Claire Mathiot; Jérôme Galon; T. Dorval; Alain Bernheim; Wolf H. Fridman

The efficacy of anti-tumor IgG reflects the balance between opposing signals mediated by activating and inhibitory Fcγ receptors (FcγRs) expressed by effector cells. Here, we show that human malignant melanoma cells express the inhibitory low-affinity Fcγ receptor FcγRIIB1 in 40% of tested metastases. When melanoma cells were grafted in nude mice, a profound inhibition of FcγRIIB1 tumor growth that required the intracytoplasmic region of the receptor was observed. IgG immune complexes (ICs) may be required for this inhibition, since sera from nude mice bearing tumors contained IgG that decreased the proliferation of FcγRIIB1-positive cells in vitro, and tumor development of FcγRIIB1-positive melanoma lines was not inhibited in antibody-defective severe combined immunodeficiency (SCID) mice. Passive immunization of SCID mice with anti–ganglioside GD2 antibody resulted in significant inhibition of growth of FcγRIIB1-positive tumors in an intracytoplasmic-dependent manner. Altogether, these data suggest that human melanoma cells express biologically active inhibitory FcγRIIB1, which regulates their development upon direct interaction with anti-tumor antibodies. Therefore, FcγR expression on human tumors may be one component of the efficacy of antibody-mediated therapies, and FcγR-positive tumors could be the most sensitive candidates for such treatments.


Blood | 2008

Adoptive transfer of allogeneic tumor-specific T cells mediates effective regression of large tumors across major histocompatibility barriers

Andrea Boni; Pawel Muranski; Lydie Cassard; Claudia Wrzesinski; Chrystal M. Paulos; Douglas C. Palmer; Luca Gattinoni; Christian S. Hinrichs; Chi-Chao Chan; Steven A. Rosenberg; Nicholas P. Restifo

Graft-versus-tumor effects can be achieved after allogeneic bone marrow transplantation in patients with malignancies of the kidney or hematopoietic system but are often accompanied by severe graft-versus-host-disease (GVHD). We sought to maximize graft-versus-tumor while minimizing GVHD using tumor-specific allogeneic effector T cells rather than open-repertoire T cells. We transferred allogeneic CD8(+) pmel-1 or CD4(+) TRP-1 T cells specific for the melanoma-associated antigens, glycoprotein 100 (gp100) and tyrosinase-related protein-1 (TRP-1), respectively, into B16-melanoma-bearing mice. Mice receiving a preparative regimen of nonmyeloablating (5 Gy) total body irradiation experienced the rapid rejection of tumor-specific allogeneic lymphocytes with no impact on tumor growth. However, when mice were given more intense total body irradiation conditioning regimens combined with autologous bone marrow transplantation, adoptively transferred allogeneic tumor-specific T lymphocytes persisted at detectable levels for several weeks and mediated significant regression of large, vascularized tumors. We found that the risk of GVHD was low when tumor-specific T cells were transferred and significant toxicity was observed only when substantial numbers of open repertoire allogeneic naive T cells were mixed with the tumor-specific lymphocytes. Taken together, these data indicate that the use of tumor-specific allogeneic CD8(+) T cells or CD4(+) can result in significant antitumor effects in the absence of measurable GVHD.

Collaboration


Dive into the Lydie Cassard's collaboration.

Top Co-Authors

Avatar

Steven A. Rosenberg

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Christian S. Hinrichs

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Pawel Muranski

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Chrystal M. Paulos

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Claudia Wrzesinski

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Luca Gattinoni

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Nicholas P. Restifo

Government of the United States of America

View shared research outputs
Top Co-Authors

Avatar

Douglas C. Palmer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zhiya Yu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Andrea Boni

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge