Person: Anderson, Ana
Loading...
Email Address
AA Acceptance Date
Birth Date
Research Projects
Organizational Units
Job Title
Last Name
Anderson
First Name
Ana
Name
Anderson, Ana
8 results
Search Results
Now showing 1 - 8 of 8
Publication TIM3+FOXP3+ regulatory T cells are tissue-specific promoters of T-cell dysfunction in cancer(Landes Bioscience, 2013) Sakuishi, Kaori; Ngiow, Shin Foong; Sullivan, Jenna M.; Teng, Michele W. L.; Kuchroo, Vijay; Smyth, Mark J.; Anderson, AnaT-cell immunoglobulin mucin 3 (TIM3) is an inhibitory molecule that has emerged as a key regulator of dysfunctional or exhausted CD8+ T cells arising in chronic diseases such as cancer. In addition to exhausted CD8+ T cells, highly suppressive regulatory T cells (Tregs) represent a significant barrier against the induction of antitumor immunity. We have found that the majority of intratumoral FOXP3+ Tregs express TIM3. TIM3+ Tregs co-express PD-1, are highly suppressive and comprise a specialized subset of tissue Tregs that are rarely observed in the peripheral tissues or blood of tumor-bearing mice. The co-blockade of the TIM3 and PD-1 signaling pathways in vivo results in the downregulation of molecules associated with TIM3+ Treg suppressor functions. This suggests that the potent clinical efficacy of co-blocking TIM3 and PD-1 signal transduction cascades likely stems from the reversal of T-cell exhaustion combined with the inhibition of regulatory T-cell function in tumor tissues. Interestingly, we find that TIM3+ Tregs accumulate in the tumor tissue prior to the appearance of exhausted CD8+ T cells, and that the depletion of Tregs at this stage interferes with the development of the exhausted phenotype by CD8+ T cells. Collectively, our data indicate that TIM3 marks highly suppressive tissue-resident Tregs that play an important role in shaping the antitumor immune response in situ, increasing the value of TIM3-targeting therapeutic strategies against cancer.Publication Future perspectives in melanoma research: meeting report from the “Melanoma Bridge”, Napoli, December 5th-8th 2013(BioMed Central, 2014) Ascierto, Paolo A; Grimaldi, Antonio M; Anderson, Ana; Bifulco, Carlo; Cochran, Alistair; Garbe, Claus; Eggermont, Alexander M; Faries, Mark; Ferrone, Soldano; Gershenwald, Jeffrey E; Gajewski, Thomas F; Halaban, Ruth; Hodi, F Stephen; Kefford, Richard; Kirkwood, John M; Larkin, James; Leachman, Sancy; Maio, Michele; Marais, Richard; Masucci, Giuseppe; Melero, Ignacio; Palmieri, Giuseppe; Puzanov, Igor; Ribas, Antoni; Saenger, Yvonne; Schilling, Bastian; Seliger, Barbara; Stroncek, David; Sullivan, Ryan; Testori, Alessandro; Wang, Ena; Ciliberto, Gennaro; Mozzillo, Nicola; Marincola, Francesco M; Thurin, MagdalenaThe fourth “Melanoma Bridge Meeting” took place in Naples, December 5 to 8th, 2013. The four topics discussed at this meeting were: Diagnosis and New Procedures, Molecular Advances and Combination Therapies, News in Immunotherapy, and Tumor Microenvironment and Biomarkers. Until recently systemic therapy for metastatic melanoma patients was ineffective, but recent research in tumor biology and immunology has led to the development of new targeted and immunotherapeutic agents that prolong progression-free survival (PFS) and overall survival (OS). New therapies, such as mitogen-activated protein kinase (MAPK) pathway inhibitors, like BRAF and MEK inhibitors, as well as other signaling pathways inhibitors, are being tested in metastatic melanoma either as monotherapy or in combination, and have yielded promising results. Improved survival rates have also been observed with immune therapy for patients with metastatic melanoma. Immune-modulating antibodies came to the forefront with anti-CTLA-4, programmed cell death-1 (PD-1) and PD-1 ligand 1 (PD-L1) pathway blocking antibodies that result in durable responses in a subset of melanoma patients. Agents targeting other immune inhibitory (e.g., Tim-3) or immune stimulating (e.g., CD137) receptors and other approaches such as adoptive cell transfer demonstrate clinical benefit in melanoma as well. This meeting’s specific focus was on advances in targeted therapy and immunotherapy. Both combination targeted therapy approaches and different immunotherapies were discussed. Similarly to the previous meetings, the importance of biomarkers for clinical application as markers for diagnosis, prognosis and prediction of treatment response was an integral part of the meeting. Significant consideration was given to issues surrounding the development of novel therapeutic targets as further study of patterns of resistance to both immunologic and targeted drugs are paramount to future drug development to guide existing and future therapies. The overall emphasis on biomarkers supports novel concepts toward integrating biomarkers into contemporary clinical management of patients with melanoma across the entire spectrum of disease stage. Translation of the knowledge gained from the biology of tumor microenvironment across different tumors represents a bridge to impact on prognosis and response to therapy in melanoma.Publication Combination immunotherapy: Where do we go from here?(BioMed Central, 2015) Overacre, Abigail E.; Kurtulus, Sema; Sznol, Mario; Pardoll, Drew M.; Anderson, Ana; Vignali, Dario A. A.The remarkable clinical success of cancer immunotherapies targeting the checkpoint receptors CTLA-4 and PD-1 has generated considerable excitement and emboldened efforts to build on this important foundation. Research efforts are now focused on understanding the mechanism of action of these immunotherapies, identifying new inhibitory mechanisms that could be targeted to achieve responses in patients with refractory cancers, and developing approaches that might exhibit efficacy against “immunologically inert” tumors. The outstanding challenges in moving forward are developing reliable strategies for determining which patients will respond optimally to a given immunotherapy, and what combination of immunotherapies and conventional therapies will prove beneficial against each tumor type. These issues were discussed in a one-day workshop at the SITC meeting in November 2014.Publication TIM3 Mediates T Cell Exhaustion during Mycobacterium tuberculosis Infection(Public Library of Science, 2016) Jayaraman, Pushpa; Jacques, Miye K.; Zhu, Chen; Steblenko, Katherine M.; Stowell, Britni L.; Madi, Asaf; Anderson, Ana; Kuchroo, Vijay; Behar, Samuel M.While T cell immunity initially limits Mycobacterium tuberculosis infection, why T cell immunity fails to sterilize the infection and allows recrudescence is not clear. One hypothesis is that T cell exhaustion impairs immunity and is detrimental to the outcome of M. tuberculosis infection. Here we provide functional evidence for the development T cell exhaustion during chronic TB. Second, we evaluate the role of the inhibitory receptor T cell immunoglobulin and mucin domain–containing-3 (TIM3) during chronic M. tuberculosis infection. We find that TIM3 expressing T cells accumulate during chronic infection, co-express other inhibitory receptors including PD1, produce less IL-2 and TNF but more IL-10, and are functionally exhausted. Finally, we show that TIM3 blockade restores T cell function and improves bacterial control, particularly in chronically infected susceptible mice. These data show that T cell immunity is suboptimal during chronic M. tuberculosis infection due to T cell exhaustion. Moreover, in chronically infected mice, treatment with anti-TIM3 mAb is an effective therapeutic strategy against tuberculosis.Publication The Non-Obese Diabetic Mouse Strain as a Model to Study CD8+ T Cell Function in Relapsing and Progressive Multiple Sclerosis(Frontiers Media S.A., 2015) Ignatius Arokia Doss, Prenitha Mercy; Roy, Andrée-Pascale; Wang, AiLi; Anderson, Ana; Rangachari, ManuMultiple sclerosis (MS) is a neurodegenerative disease resulting from an autoimmune attack on central nervous system (CNS) myelin. Although CD4+ T cell function in MS pathology has been extensively studied, there is also strong evidence that CD8+ T lymphocytes play a key role. Intriguingly, CD8+ T cells accumulate in great numbers in the CNS in progressive MS, a form of the disease that is refractory to current disease-modifying therapies that target the CD4+ T cell response. Here, we discuss the function of CD8+ T cells in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. In particular, we describe EAE in non-obese diabetic (NOD) background mice, which develop a pattern of disease characterized by multiple attacks and remissions followed by a progressively worsening phase. This is highly reminiscent of the pattern of disease observed in nearly half of MS patients. Particular attention is paid to a newly described transgenic mouse strain (1C6) on the NOD background whose CD4+ and CD8+ T cells are directed against the encephalitogenic peptide MOG[35–55]. Use of this model will give us a more complete picture of the role(s) played by distinct T cell subsets in CNS autoimmunity.Publication An IL-27/NFIL3 signaling axis drives Tim-3 and IL-10 expression and T cell dysfunction(2015) Zhu, Chen; Sakuishi, Kaori; Xiao, Sheng; Sun, Zhiyi; Zaghouani, Sarah; Gu, Guangxiang; Wang, Chao; Tan, Dewar J.; Wu, Chuan; Rangachari, Manu; Pertel, Thomas; Jin, Hyun-Tak; Ahmed, Rafi; Anderson, Ana; Kuchroo, VijaySUMMARY The inhibitory receptor Tim-3 has emerged as a critical regulator of the T cell dysfunction that develops in chronic viral infections and cancers. However, little is known regarding the signaling pathways that drive Tim-3 expression. Here, we demonstrate that IL-27 induces NFIL3, which promotes permissive chromatin remodeling of the Tim-3 locus and induces Tim-3 expression together with the immunosuppressive cytokine IL-10. We further show that the IL-27/NFIL3 signaling axis is crucial for the induction of Tim-3 in vivo. IL-27-conditioned Th1 cells exhibit reduced effector function and are poor mediators of intestinal inflammation. This inhibitory effect is NFIL3 dependent. In contrast, tumor-infiltrating lymphocytes (TILs) from IL-27R−/− mice exhibit reduced NFIL3, less Tim-3 expression and failure to develop dysfunctional phenotype, resulting in better tumor growth control. Thus, our data identify an IL-27/NFIL3 signaling axis as a key regulator of effector T cell responses via induction of Tim-3, IL-10, and T cell dysfunction.Publication Targeting Tim-3 and PD-1 Pathways to Reverse T Cell Exhaustion and Restore Anti-Tumor Immunity(The Rockefeller University Press, 2010) Sakuishi, Kaori; Apetoh, Lionel; Sullivan, Jenna M.; Blazar, Bruce R.; Kuchroo, Vijay; Anderson, AnaThe immune response plays an important role in staving off cancer; however, mechanisms of immunosuppression hinder productive anti-tumor immunity. T cell dysfunction or exhaustion in tumor-bearing hosts is one such mechanism. PD-1 has been identified as a marker of exhausted T cells in chronic disease states, and blockade of PD-1–PD-1L interactions has been shown to partially restore T cell function. We have found that T cell immunoglobulin mucin (Tim) 3 is expressed on CD8\(^+\) tumor-infiltrating lymphocytes (TILs) in mice bearing solid tumors. All Tim-3\(^+\) TILs coexpress PD-1, and Tim-3\(^+\)PD-1\(^+\) TILs repre- sent the predominant fraction of T cells infiltrating tumors. Tim-3\(^+\)PD-1\(^+\) TILs exhibit the most severe exhausted phenotype as defined by failure to proliferate and produce IL-2, TNF, and IFN-\(\gamma\). We further find that combined targeting of the Tim-3 and PD-1 pathways is more effective in controlling tumor growth than targeting either pathway alone.Publication Targeting PGLYRP1 promotes antitumor immunity while inhibiting autoimmune neuroinflammation(Springer Science and Business Media LLC, 2023-10-12) Schnell, Alexandra; Huang, Linglin; Regan, Brianna; Vonficht, Dominik; Bollhagen, Alina; Wang, Mona; Hou, Yu; Bod, Lloyd; Chihara, Norio; Madi, Asaf; Anderson, Ana; Kuchroo, VijayCo-inhibitory and checkpoint molecules suppress T-cell function in the tumor microenvironment, thereby rendering T cells dysfunctional. While immune checkpoint blockade (ICB) has emerged as a successful treatment option for multiple human cancers, severe autoimmune-like side effects limit its application. Here we found the gene encoding the Peptidoglycan Recognition Protein 1 (PGLYRP1) to be highly co-expressed with co-inhibitory molecules and hypothesized that it might be a promising target for cancer immunotherapy. Indeed, genetic deletion of PGLYRP1 in mice led to decreased tumor growth and an increased activation/effector phenotype in CD8+ T cells, suggesting an inhibitory function of PGLYRP1 in CD8+ T cells. Surprisingly, the genetic deletion of PGLYRP1 strongly protected against the development of experimental autoimmune encephalomyelitis (EAE), a model of autoimmune disease in the central nervous system (CNS). Pglyrp1-deficient myeloid cells had a defect in antigen presentation and T-cell activation, indicating that PGLYRP1 might act as a proinflammatory molecule in myeloid cells during autoimmunity. Our results highlight PGLYRP1 as a promising target for immunotherapy that, when targeted, elicits a potent antitumor immune response while protecting against some forms of tissue inflammation and autoimmunity.