Network


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

Hotspot


Dive into the research topics where Viia Valge-Archer is active.

Publication


Featured researches published by Viia Valge-Archer.


Journal of Immunology | 2003

Program Death-1 Engagement Upon TCR Activation Has Distinct Effects on Costimulation and Cytokine-Driven Proliferation: Attenuation of ICOS, IL-4, and IL-21, But Not CD28, IL-7, and IL-15 Responses

Frann Bennett; Deborah Luxenberg; Vincent Ling; I-Ming Wang; Kim Marquette; David Lowe; Nighat Khan; Geertruida M. Veldman; Kenneth A. Jacobs; Viia Valge-Archer; Mary Collins; Beatriz M. Carreno

The program death 1 (PD-1) receptor and its ligands, PD-1 ligand (PD-L)1 and PD-L2, define a novel regulatory pathway with potential inhibitory effects on T, B, and monocyte responses. In the present study, we show that human CD4+ T cells express PD-1, PD-L1, and PD-L2 upon activation, and Abs to the receptor can be agonists or antagonists of the pathway. Under optimal conditions of stimulation, ICOS but not CD28 costimulation can be prevented by PD-1 engagement. IL-2 levels induced by costimulation are critical in determining the outcome of the PD-1 engagement. Thus, low to marginal IL-2 levels produced upon ICOS costimulation account for the greater sensitivity of this pathway to PD-1-mediated inhibition. Interestingly, exogenous IL-2, IL-7, and IL-15 but not IL-4 and IL-21 can rescue PD-1 inhibition, suggesting that among these cytokines only those that activate STAT5 can rescue PD-1 inhibition. As STAT5 has been implicated in the maintenance of IL-2Rα expression, these results suggest that IL-7 and IL-15 restore proliferation under conditions of PD-1 engagement by enhancing high-affinity IL-2R expression and hence, IL-2 responsiveness.


Science Translational Medicine | 2016

The tumor microenvironment and Immunoscore are critical determinants of dissemination to distant metastasis

Bernhard Mlecnik; Gabriela Bindea; Amos Kirilovsky; Helen K. Angell; Anna C. Obenauf; Marie Tosolini; Sarah E. Church; Pauline Maby; Angela Vasaturo; Mihaela Angelova; Tessa Fredriksen; Stéphanie Mauger; Maximilian J. Waldner; Anne Berger; Michael R. Speicher; Franck Pagès; Viia Valge-Archer; Jérôme Galon

Local lymphatic vessel density and immune cytotoxicity prevent metastatic invasion. Managing metastasis Because of the poor prognosis of metastatic cancer, it is critical to determine exactly how different factors contribute to cancer spread. Mlecnik et al. examined the impact of tumor-intrinsic, microenvironmental, and immunological factors on tumor metastasis in colorectal cancer patients. They found that decreased presence of lymphatic vessels and reduced immune cytotoxicity were more strongly associated with the metastatic process than tumor-intrinsic factors such as chromosomal instability or cancer-associated mutations. These data support testing the Immunoscore as a biomarker to predict metastasis and guide therapy. Although distant metastases account for most of the deaths in cancer patients, fundamental questions regarding mechanisms that promote or inhibit metastasis remain unanswered. We show the impact of mutations, genomic instability, lymphatic and blood vascularization, and the immune contexture of the tumor microenvironment on synchronous metastases in large cohorts of colorectal cancer patients. We observed large genetic heterogeneity among primary tumors, but no major differences in chromosomal instability or key cancer-associated mutations. Similar patterns of cancer-related gene expression levels were observed between patients. No cancer-associated genes or pathways were associated with M stage. Instead, mutations of FBXW7 were associated with the absence of metastasis and correlated with increased expression of T cell proliferation and antigen presentation functions. Analyzing the tumor microenvironment, we observed two hallmarks of the metastatic process: decreased presence of lymphatic vessels and reduced immune cytotoxicity. These events could be the initiating factors driving both synchronous and metachronous metastases. Our data demonstrate the protective impact of the Immunoscore, a cytotoxic immune signature, and increased marginal lymphatic vessels, against the generation of distant metastases, regardless of genomic instability.


Science Translational Medicine | 2014

Functional Network Pipeline Reveals Genetic Determinants Associated with in Situ Lymphocyte Proliferation and Survival of Cancer Patients

Bernhard Mlecnik; Gabriela Bindea; Helen K. Angell; Maria Stella Sasso; Anna C. Obenauf; Tessa Fredriksen; Lucie Lafontaine; Amélie M. Bilocq; Amos Kirilovsky; Marie Tosolini; Maximilian J. Waldner; Anne Berger; Wolf H. Fridman; Arash Rafii; Viia Valge-Archer; Franck Pagès; Michael R. Speicher; Jérôme Galon

Genomic alterations result in changes of immune cell densities within the tumor. There Goes the Neighborhood Just as a new homeowner will remodel when moving into a house, a tumor also alters the local microenvironment. One of the least tumor-friendly things in the tumor microenvironment is the immune response. Mlecnik et al. now examine the network of changes to cytokines that contribute to shaping the intratumoral immune response in colorectal tumors. The authors found changes in local expression of 13 cytokines. Deletion of one cytokine, interleukin 15, was associated with tumor recurrence and reduced patient survival, potentially by affecting the local proliferation of B and T cells. Their results show that chromosome instability contributes to the regulation of cytokines and downstream tumor immune response. The tumor microenvironment is host to a complex network of cytokines that contribute to shaping the intratumoral immune reaction. Chromosomal gains and losses, coupled with expression analysis, of 59 cytokines and receptors and their functional networks were investigated in colorectal cancers. Changes in local expression for 13 cytokines were shown. Metastatic patients exhibited an increased frequency of deletions of cytokines from chromosome 4. Interleukin 15 (IL15) deletion corresponded with decreased IL15 expression, a higher risk of tumor recurrence, and reduced patient survival. Decreased IL15 expression affected the local proliferation of B and T lymphocytes. Patients with proliferating B and T cells at the invasive margin and within the tumor center had significantly prolonged disease-free survival. These results delineate chromosomal instability as a mechanism of modulating local cytokine expression in human tumors and underline the major role of IL15. Our data provide further mechanisms resulting in changes of specific immune cell densities within the tumor, and the importance of local active lymphocyte proliferation for patient survival.


mAbs | 2010

In vitro potency, pharmacokinetic profiles, and pharmacological activity of optimized anti-IL-21R antibodies in a mouse model of lupus.

Yulia Vugmeyster; Heath Guay; Pamela Szklut; Ming D. Qian; Macy Jin; Angela Widom; Vikki Spaulding; Frann Bennett; Leslie Lowe; Tatyana Andreyeva; David Lowe; Steven Lane; George Thom; Viia Valge-Archer; Davinder Gill; Deborah Young; Laird Bloom

Using phage display, we generated a panel of optimized neutralizing antibodies against the human and mouse receptors for interleukin 21 (IL-21), a cytokine that is implicated in the pathogenesis of many types of autoimmune disease. Two antibodies, Ab-01 and Ab-02, which differed by only four amino acids in VL CDR3, showed potent inhibition of human and mouse IL-21R in cell-based assays and were evaluated for their pharmacological and pharmacodynamic properties. Ab-01, but not Ab-02, significantly reduced a biomarker of disease (anti-dsDNA antibodies) and IgG deposits in the kidney in the MRL-Faslpr mouse model of lupus, suggesting that anti-IL-21R antibodies may prove useful in the treatment of lupus. Ab-01 also had a consistently higher exposure (AUC0-∞) than Ab-02 following a single dose in rodents or cynomolgus monkeys (2-3-fold or 4-7-fold, respectively). Our data demonstrate that small differences in CDR3 sequences of optimized antibodies can lead to profound differences in in vitro and in vivo properties, including differences in pharmacological activity and pharmacokinetic profiles. The lack of persistent activity of Ab-02 in the MRL-Faslpr mouse lupus model may have been a consequence of faster elimination, reduced potency in blocking the effects of mouse IL-21R, and more potent/earlier onset of the anti-product response relative to Ab-01.


Cancer immunology research | 2017

Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery

Suzanne Mosely; John E. Prime; Richard Sainson; Jens-Oliver Koopmann; Dennis Wang; Danielle Greenawalt; Miika Ahdesmaki; Rebecca Leyland; Stefanie Mullins; Luciano Pacelli; Danielle Marcus; Judith Anderton; Amanda Watkins; Jane Coates Ulrichsen; Philip Brohawn; Brandon W. Higgs; Matthew McCourt; Hazel Jones; James Harper; Michelle Morrow; Viia Valge-Archer; Ross Stewart; Simon J. Dovedi; Robert W. Wilkinson

Murine syngeneic tumor models are used to study responses to antitumor immunotherapies. To rationalize model selection, the underlying genetic and immunologic biology of the models was analyzed, allowing parallels to be drawn between models and human disease phenotypes. Murine syngeneic tumor models are critical to novel immuno-based therapy development, but the molecular and immunologic features of these models are still not clearly defined. The translational relevance of differences between the models is not fully understood, impeding appropriate preclinical model selection for target validation, and ultimately hindering drug development. Across a panel of commonly used murine syngeneic tumor models, we showed variable responsiveness to immunotherapies. We used array comparative genomic hybridization, whole-exome sequencing, exon microarray analysis, and flow cytometry to extensively characterize these models, which revealed striking differences that may underlie these contrasting response profiles. We identified strong differential gene expression in immune-related pathways and changes in immune cell–specific genes that suggested differences in tumor immune infiltrates between models. Further investigation using flow cytometry showed differences in both the composition and magnitude of the tumor immune infiltrates, identifying models that harbor “inflamed” and “non-inflamed” tumor immune infiltrate phenotypes. We also found that immunosuppressive cell types predominated in syngeneic mouse tumor models that did not respond to immune-checkpoint blockade, whereas cytotoxic effector immune cells were enriched in responsive models. A cytotoxic cell–rich tumor immune infiltrate has been correlated with increased efficacy of immunotherapies in the clinic, and these differences could underlie the varying response profiles to immunotherapy between the syngeneic models. This characterization highlighted the importance of extensive profiling and will enable investigators to select appropriate models to interrogate the activity of immunotherapies as well as combinations with targeted therapies in vivo. Cancer Immunol Res; 5(1); 29–41. ©2016 AACR.


OncoImmunology | 2015

A high density of tertiary lymphoid structure B cells in lung tumors is associated with increased CD4+ T cell receptor repertoire clonality

Wei Zhu; Claire Germain; Zheng Liu; Yinong Sebastian; Priyanka Devi; Samantha Knockaert; Philip Brohawn; Kim Lehmann; Diane Damotte; Pierre Validire; Yihong Yao; Viia Valge-Archer; Scott A. Hammond; Marie-Caroline Dieu-Nosjean; Brandon W. Higgs

T and B cell receptor (TCR and BCR, respectively) Vβ or immunoglobulin heavy chain complementarity-determining region 3 sequencing allows monitoring of repertoire changes through recognition, clonal expansion, affinity maturation, and T or B cell activation in response to antigen. TCR and BCR repertoire analysis can advance understanding of antitumor immune responses in the tumor microenvironment. TCR and BCR repertoires of sorted CD4+, CD8+ or CD19+ cells in tumor, non-tumoral distant tissue (NT), and peripheral compartments (blood/draining lymph node [P]) from 47 non-small cell lung cancer (NSCLC) patients (agemedian = 68 y) were sequenced. The clonotype spectra were assessed among different tissues and correlated with clinical and immunological parameters. In all tissues, CD4+ and CD8+ TCR repertoires had greater clonality relative to CD19+ BCR. CD4+ T cells exhibited greater clonality in NT compared to tumor (p = 0.002) and P (p < 0.001), concentrated among older patients (age > 68). Younger patients exhibited greater CD4+ T cell diversity in P compared to older patients (p = 0.05), and greater CD4+ T cell clonality in tumor relative to P (p < 0.001), with fewer shared clonotypes between tumor and P than older patients (p = 0.04). More interestingly, greater CD4+ and CD8+ T cell clonality in tumor and P, respectively (both p = 0.05), correlated with high density of tumor-associated tertiary lymphoid structure (TLS) B cells, a biomarker of higher overall survival in NSCLC. Results indicate distinct adaptive immune responses in NSCLC, where peripheral T cell diversity is modulated by age, and tumor T cell clonal expansion is favored by the presence of TLSs in the tumor microenvironment.


Journal of the National Cancer Institute | 2018

Comprehensive Intrametastatic Immune Quantification and Major Impact of Immunoscore on Survival.

Bernhard Mlecnik; Marc Van den Eynde; Gabriela Bindea; Sarah E. Church; Angela Vasaturo; Tessa Fredriksen; Lucie Lafontaine; Nacilla Haicheur; Florence Marliot; Daphné Debetancourt; Géraldine Pairet; Anne Jouret-Mourin; Jean-François Gigot; Catherine Hubert; Etienne Danse; Cristina Dragean; Javier Carrasco; Yves Humblet; Viia Valge-Archer; Anne Berger; Franck Pagès; Jean-Pascal Machiels; Jérôme Galon

Background This study assesses how the metastatic immune landscape is impacting the response to treatment and the outcome of colorectal cancer (CRC) patients. Methods Complete curative resection of metastases (n = 441) was performed for two patient cohorts (n = 153). Immune densities were quantified in the center and invasive margin of all metastases. Immunoscore and T and B cell (TB) score were analyzed in relation to radiological and pathological responses and patients disease-free (DFS) and overall survival (OS) using multivariable Cox proportional hazards models. All statistical tests were two-sided. Results The spatial distribution of immune cells within metastases was nonuniform. Patients, as well as metastases of the same patient, had variable immune infiltrates and response to therapy. A beneficial response was statistically significantly associated with increased immune densities. Among all metastases, Immunoscore (I) and TB score evaluated in the least immune-infiltrated metastases were the strongest predictors for DFS and OS (five-year follow-up, Immunoscore: I 3-4: DFS rate = 27.9%, 95% CI = 15.2 to 51.3; vs I 0-1-2: DFS rate = 12.3%, 95% CI = 4.9 to 30.6; HR = 0.45, 95% CI = 0.28 to 0.70, P = .02; I 3-4: OS rate = 64.6%, 95% CI = 46.6 to 89.6; vs I 0-1-2: OS rate = 32.5%, 95% CI = 17.2 to 61.4; HR = 0.32, 95% CI = 0.15 to 0.66, P = .001, C-index = 65.9%; five-year follow-up, TB score: TB 3-4: DFS rate = 25.7%, 95% CI = 14.2 to 46.6; vs TB 0-1-2: DFS rate = 5.0%, 95% CI = 0.8 to 32.4; HR = 0.36, 95% CI = 0.22 to 0.57, P < .001; TB 3-4: OS rate = 63.7%, 95% CI = 46.4 to 87.5; vs TB 0-1-2: OS rate: 21.4%, 95% CI = 9.2 to 49.8; HR = 0.25, 95% CI = 0.12 to 0.51, P < .001, C-index = 67.8%). High TB score and Immunoscore patients had a median survival of 70.5 months, while low patients survived only 25.1 to 38.3 months. Nonresponding patients with high-immune infiltrates had prolonged DFS (HR = 0.28, 95% CI = 0.15 to 0.52, P = .001) and OS (HR = 0.25, 95% CI = 0.1 to 0.62, P = .001). The immune parameters remained the only statistically significant prognostic factor associated with DFS and OS in multivariable analysis (P < .001), while response to treatment was not. Conclusions Response to treatment and prolonged survival of metastatic CRC patients were statistically significantly associated with high-immune densities quantified into the least immune-infiltrated metastasis.


Oncotarget | 2016

Phenotypic screening reveals TNFR2 as a promising target for cancer immunotherapy

Geoffrey S. Williams; Bina Mistry; Sandrine Guillard; Jane Coates Ulrichsen; Alan Sandercock; Jun Wang; Andrea González-Muñoz; Julie Parmentier; Chelsea Black; Jo Soden; Jim Freeth; Jelena Jovanović; Rebecca Leyland; Rafia S. Al-Lamki; Andrew J. Leishman; Steven J. Rust; Ross Stewart; Lutz Jermutus; John R. Bradley; Vahe Bedian; Viia Valge-Archer; Ralph Minter; Robert W. Wilkinson

Antibodies that target cell-surface molecules on T cells can enhance anti-tumor immune responses, resulting in sustained immune-mediated control of cancer. We set out to find new cancer immunotherapy targets by phenotypic screening on human regulatory T (Treg) cells and report the discovery of novel activators of tumor necrosis factor receptor 2 (TNFR2) and a potential role for this target in immunotherapy. A diverse phage display library was screened to find antibody mimetics with preferential binding to Treg cells, the most Treg-selective of which were all, without exception, found to bind specifically to TNFR2. A subset of these TNFR2 binders were found to agonise the receptor, inducing iκ-B degradation and NF-κB pathway signalling in vitro. TNFR2 was found to be expressed by tumor-infiltrating Treg cells, and to a lesser extent Teff cells, from three lung cancer patients, and a similar pattern was also observed in mice implanted with CT26 syngeneic tumors. In such animals, TNFR2-specific agonists inhibited tumor growth, enhanced tumor infiltration by CD8+ T cells and increased CD8+ T cell IFN-γ synthesis. Together, these data indicate a novel mechanism for TNF-α-independent TNFR2 agonism in cancer immunotherapy, and demonstrate the utility of target-agnostic screening in highlighting important targets during drug discovery.


British Journal of Cancer | 2017

MHC class I chain-related protein A and B (MICA and MICB) are predominantly expressed intracellularly in tumour and normal tissue

Hormas Ghadially; Lee Brown; Chris Lloyd; Leeanne Lewis; Arthur Lewis; Janette Dillon; Richard C.A. Sainson; Jelena Jovanović; Natalie Tigue; David Bannister; Lisa Bamber; Viia Valge-Archer; R. Wilkinson

Background:Major histocompatibility complex (MHC) class I chain-related protein A (MICA) and MHC class I chain-related protein B (MICB) are polymorphic proteins that are induced upon stress, damage or transformation of cells which act as a ‘kill me’ signal through the natural-killer group 2, member D receptor expressed on cytotoxic lymphocytes. MICA/B are not thought to be constitutively expressed by healthy normal cells but expression has been reported for most tumour types. However, it is not clear how much of this protein is expressed on the cell surface.Methods:Using a novel, well-characterised antibody and both standard and confocal microscopy, we systematically profiled MICA/B expression in multiple human tumour and normal tissue.Results:High expression of MICA/B was detected in the majority of tumour tissues from multiple indications. Importantly, MICA/B proteins were predominantly localised intracellularly with only occasional evidence of cell membrane localisation. MICA/B expression was also demonstrated in most normal tissue epithelia and predominantly localised intracellularly. Crucially, we did not observe qualitative differences in cell surface expression between tumour and MICA/B expressing normal epithelia.Conclusions:This demonstrates for the first time that MICA/B is more broadly expressed in normal tissue and that expression is mainly intracellular with only a small fraction appearing on the cell surface of some epithelia and tumour cells.


Cancer Research | 2016

Abstract 1451: Analysis of expression MHC class I chain-related gene A and B (MICA/B) in normal and tumor tissue

Hormas Ghadially; Lee Brown; Arthur Lewis; Meggan Czapiga; Viia Valge-Archer; Robert W. Wilkinson

MHC class I chain-related gene A and B (MICA and MICB) are highly polymorphic proteins that are induced upon stress, damage or transformation of cells which act as a “kill me” signal through the NKG2D receptor expressed on Natural Killer, CD8+ and γδ T cells. Experimentally, the MIC/NKG2D axis has been shown to be important for the recognition of tumour cells by cytotoxic cells of the immune system and many tumours have evolved strategies to evade the detection by NKG2D expressing cells, e.g. by shedding MIC from the cell surface. Expression of MIC has been reported for most tumour types and in normal gastrointestinal tract epithelium but the published data is often difficult to interpret. Additionally, it is not clear how much of the protein is expressed on the cell surface as MIC cell surface expression is known to be regulated tightly on multiple levels. A validated MICA/B IHC assay was developed using an in-house tool antibody to profile multiple frozen human normal and tumour tissue microarrays (TMA’s) by both standard and confocal microscopy techniques. Using a stringently characterised novel antibody that detects MICA as well as MICB this study describes the expression patterns in a wide range of tumours and normal tissues. With this method we generated data with unprecedented resolution, which enabled us to analyse the expression of MICA and MICB not only on the cellular but also on the sub-cellular level. Citation Format: Hormas Ghadially, Lee Brown, Arthur Lewis, Meggan Czapiga, Viia Valge-Archer, Robert W. Wilkinson. Analysis of expression MHC class I chain-related gene A and B (MICA/B) in normal and tumor tissue. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1451.

Collaboration


Dive into the Viia Valge-Archer'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
Top Co-Authors

Avatar

Beatriz M. Carreno

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge