18 results on '"Zappasodi, Roberta"'
Search Results
2. Facts and Perspectives: Implications of tumor glycolysis on immunotherapy response in triple negative breast cancer.
- Author
-
Schreier, Ashley, Zappasodi, Roberta, Serganova, Inna, Brown, Kristy A., Demaria, Sandra, and Andreopoulou, Eleni
- Subjects
TRIPLE-negative breast cancer ,IMMUNE checkpoint inhibitors ,GLYCOLYSIS ,IMMUNE checkpoint proteins ,CANCER chemotherapy - Abstract
Triple negative breast cancer (TNBC) is an aggressive disease that is difficult to treat and portends a poor prognosis in many patients. Recent efforts to implement immune checkpoint inhibitors into the treatment landscape of TNBC have led to improved outcomes in a subset of patients both in the early stage and metastatic settings. However, a large portion of patients with TNBC remain resistant to immune checkpoint inhibitors and have limited treatment options beyond cytotoxic chemotherapy. The interplay between the anti-tumor immune response and tumor metabolism contributes to immunotherapy response in the preclinical setting, and likely in the clinical setting as well. Specifically, tumor glycolysis and lactate production influence the tumor immune microenvironment through creation of metabolic competition with infiltrating immune cells, which impacts response to immune checkpoint blockade. In this review, we will focus on how glucose metabolism within TNBC tumors influences the response to immune checkpoint blockade and potential ways of harnessing this information to improve clinical outcomes. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
3. Calreticulin mutant myeloproliferative neoplasms induce MHC-I skewing, which can be overcome by an optimized peptide cancer vaccine.
- Author
-
Gigoux, Mathieu, Holmström, Morten O., Zappasodi, Roberta, Park, Joseph J., Pourpe, Stephane, Bozkus, Cansu Cimen, Mangarin, Levi M. B., Redmond, David, Verma, Svena, Schad, Sara, George, Mariam M., Venkatesh, Divya, Ghosh, Arnab, Hoyos, David, Molvi, Zaki, Kamaz, Baransel, Marneth, Anna E., Duke, William, Leventhal, Matthew J., and Jan, Max
- Subjects
PEPTIDES ,T cell receptors ,CANCER vaccines ,MYELOPROLIFERATIVE neoplasms ,CALRETICULIN ,T cells ,MAJOR histocompatibility complex - Abstract
The majority of JAK2
V617F -negative myeloproliferative neoplasms (MPNs) have disease-initiating frameshift mutations in calreticulin (CALR), resulting in a common carboxyl-terminal mutant fragment (CALRMUT ), representing an attractive source of neoantigens for cancer vaccines. However, studies have shown that CALRMUT -specific T cells are rare in patients with CALRMUT MPN for unknown reasons. We examined class I major histocompatibility complex (MHC-I) allele frequencies in patients with CALRMUT MPN from two independent cohorts. We observed that MHC-I alleles that present CALRMUT neoepitopes with high affinity are underrepresented in patients with CALRMUT MPN. We speculated that this was due to an increased chance of immune-mediated tumor rejection by individuals expressing one of these MHC-I alleles such that the disease never clinically manifested. As a consequence of this MHC-I allele restriction, we reasoned that patients with CALRMUT MPN would not efficiently respond to a CALRMUT fragment cancer vaccine but would when immunized with a modified CALRMUT heteroclitic peptide vaccine approach. We found that heteroclitic CALRMUT peptides specifically designed for the MHC-I alleles of patients with CALRMUT MPN efficiently elicited a CALRMUT cross-reactive CD8+ T cell response in human peripheral blood samples but not to the matched weakly immunogenic CALRMUT native peptides. We corroborated this effect in vivo in mice and observed that C57BL/6J mice can mount a CD8+ T cell response to the CALRMUT fragment upon immunization with a CALRMUT heteroclitic, but not native, peptide. Together, our data emphasize the therapeutic potential of heteroclitic peptide–based cancer vaccines in patients with CALRMUT MPN. Producing peptides to activate T cells against MPN: Myeloproliferative neoplasms (MPNs) that have frameshift mutations in calreticulin (CALR) rarely have T cells that target this neoantigen. Gigoux et al. investigated the lack of these T cells by examining class I major histocompatiblity complex (MHC-I) allele frequencies in these patients. They saw a skewing of these alleles and thus developed a heteroclitic peptide vaccine to activate T cells against these tumors. They saw a response in patient samples ex vivo and in mice to immunization with this peptide, which represents a promising treatment for patients with this disease. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
4. Fundamental immune–oncogenicity trade-offs define driver mutation fitness.
- Author
-
Hoyos, David, Zappasodi, Roberta, Schulze, Isabell, Sethna, Zachary, de Andrade, Kelvin César, Bajorin, Dean F., Bandlamudi, Chaitanya, Callahan, Margaret K., Funt, Samuel A., Hadrup, Sine R., Holm, Jeppe S., Rosenberg, Jonathan E., Shah, Sohrab P., Vázquez-García, Ignacio, Weigelt, Britta, Wu, Michelle, Zamarin, Dmitriy, Campitelli, Laura F., Osborne, Edward J., and Klinger, Mark
- Abstract
Missense driver mutations in cancer are concentrated in a few hotspots1. Various mechanisms have been proposed to explain this skew, including biased mutational processes2, phenotypic differences3–6 and immunoediting of neoantigens7,8; however, to our knowledge, no existing model weighs the relative contribution of these features to tumour evolution. We propose a unified theoretical ‘free fitness’ framework that parsimoniously integrates multimodal genomic, epigenetic, transcriptomic and proteomic data into a biophysical model of the rate-limiting processes underlying the fitness advantage conferred on cancer cells by driver gene mutations. Focusing on TP53, the most mutated gene in cancer1, we present an inference of mutant p53 concentration and demonstrate that TP53 hotspot mutations optimally solve an evolutionary trade-off between oncogenic potential and neoantigen immunogenicity. Our model anticipates patient survival in The Cancer Genome Atlas and patients with lung cancer treated with immunotherapy as well as the age of tumour onset in germline carriers of TP53 variants. The predicted differential immunogenicity between hotspot mutations was validated experimentally in patients with cancer and in a unique large dataset of healthy individuals. Our data indicate that immune selective pressure on TP53 mutations has a smaller role in non-cancerous lesions than in tumours, suggesting that targeted immunotherapy may offer an early prophylactic opportunity for the former. Determining the relative contribution of immunogenicity and oncogenic function to the selective advantage of hotspot mutations thus has important implications for both precision immunotherapies and our understanding of tumour evolution.A mathematical framework to estimate the fitness of cancer driver mutations by integrating mutational bias, oncogenicity and immunogenicity finds fundamental trade-offs in cancer evolution. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
5. MAIT and Vδ2 unconventional T cells are supported by a diverse intestinal microbiome and correlate with favorable patient outcome after allogeneic HCT.
- Author
-
Andrlová, Hana, Miltiadous, Oriana, Kousa, Anastasia I., Dai, Anqi, DeWolf, Susan, Violante, Sara, Park, Hee-Yon, Janaki-Raman, Sudha, Gardner, Rui, El Daker, Sary, Slingerland, John, Giardina, Paul, Clurman, Annelie, Gomes, Antonio L. C., Nguyen, Chi, da Silva, Marina Burgos, Armijo, Gabriel K., Lee, Nicole, Zappasodi, Roberta, and Chaligne, Ronan
- Subjects
GUT microbiome ,TREATMENT effectiveness ,HEMATOPOIETIC stem cell transplantation ,INTESTINAL physiology ,INTESTINAL mucosa ,GRAFT versus host disease ,MICROBIAL diversity - Abstract
Microbial diversity is associated with improved outcomes in recipients of allogeneic hematopoietic cell transplantation (allo-HCT), but the mechanism underlying this observation is unclear. In a cohort of 174 patients who underwent allo-HCT, we demonstrate that a diverse intestinal microbiome early after allo-HCT is associated with an increased number of innate-like mucosal-associated invariant T (MAIT) cells, which are in turn associated with improved overall survival and less acute graft-versus-host disease (aGVHD). Immune profiling of conventional and unconventional immune cell subsets revealed that the prevalence of Vδ2 cells, the major circulating subpopulation of γδ T cells, closely correlated with the frequency of MAIT cells and was associated with less aGVHD. Analysis of these populations using both single-cell transcriptomics and flow cytometry suggested a shift toward activated phenotypes and a gain of cytotoxic and effector functions after transplantation. A diverse intestinal microbiome with the capacity to produce activating ligands for MAIT and Vδ2 cells appeared to be necessary for the maintenance of these populations after allo-HCT. These data suggest an immunological link between intestinal microbial diversity, microbe-derived ligands, and maintenance of unconventional T cells. Unconventional transplant support: Numerous studies have suggested that a diverse intestinal microbiome is linked to better outcomes in recipients of allogeneic hematopoietic cell transplantation (allo-HCT). Andrlová et al. look to understand how unconventional T cell subsets facilitate this outcome in a cohort of 174 patients with allo-HCT. They observed that a diverse intestinal microbiome at early times after allo-HCT is associated with higher numbers of innate-like mucosal-associated invariant T (MAIT) cells, and this correlated with a higher prevalence of the Vδ2 subpopulation of γδ T cells and a reduced incidence of acute graft-versus-host disease (aGVHD). Further analysis indicated that a diverse intestinal microbiome could produce ligands that promote maintenance of activated and functional MAIT and Vδ2 cells following allo-HCT. These findings provide evidence of the immunological link between a diverse microbiome and maintenance of unconventional T cell subsets. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
6. CTLA-4 blockade drives loss of Treg stability in glycolysis-low tumours.
- Author
-
Zappasodi, Roberta, Serganova, Inna, Cohen, Ivan J., Maeda, Masatomo, Shindo, Masahiro, Senbabaoglu, Yasin, Watson, McLane J., Leftin, Avigdor, Maniyar, Rachana, Verma, Svena, Lubin, Matthew, Ko, Myat, Mane, Mayuresh M., Zhong, Hong, Liu, Cailian, Ghosh, Arnab, Abu-Akeel, Mohsen, Ackerstaff, Ellen, Koutcher, Jason A., and Ho, Ping-Chih
- Abstract
Limiting metabolic competition in the tumour microenvironment may increase the effectiveness of immunotherapy. Owing to its crucial role in the glucose metabolism of activated T cells, CD28 signalling has been proposed as a metabolic biosensor of T cells1. By contrast, the engagement of CTLA-4 has been shown to downregulate T cell glycolysis1. Here we investigate the effect of CTLA-4 blockade on the metabolic fitness of intra-tumour T cells in relation to the glycolytic capacity of tumour cells. We found that CTLA-4 blockade promotes metabolic fitness and the infiltration of immune cells, especially in glycolysis-low tumours. Accordingly, treatment with anti-CTLA-4 antibodies improved the therapeutic outcomes of mice bearing glycolysis-defective tumours. Notably, tumour-specific CD8
+ T cell responses correlated with phenotypic and functional destabilization of tumour-infiltrating regulatory T (Treg ) cells towards IFNγ- and TNF-producing cells in glycolysis-defective tumours. By mimicking the highly and poorly glycolytic tumour microenvironments in vitro, we show that the effect of CTLA-4 blockade on the destabilization of Treg cells is dependent on Treg cell glycolysis and CD28 signalling. These findings indicate that decreasing tumour competition for glucose may facilitate the therapeutic activity of CTLA-4 blockade, thus supporting its combination with inhibitors of tumour glycolysis. Moreover, these results reveal a mechanism by which anti-CTLA-4 treatment interferes with Treg cell function in the presence of glucose.CTLA-4 promotes glucose uptake by tumour-infiltrating regulatory T cells, making them unstable. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
7. Blockade of the AHR restricts a Treg-macrophage suppressive axis induced by L-Kynurenine.
- Author
-
Campesato, Luis Felipe, Budhu, Sadna, Tchaicha, Jeremy, Weng, Chien-Huan, Gigoux, Mathieu, Cohen, Ivan Jose, Redmond, David, Mangarin, Levi, Pourpe, Stephane, Liu, Cailian, Zappasodi, Roberta, Zamarin, Dmitriy, Cavanaugh, Jill, Castro, Alfredo C., Manfredi, Mark G., McGovern, Karen, Merghoub, Taha, and Wolchok, Jedd D.
- Subjects
IMMUNE checkpoint inhibitors ,ARYL hydrocarbon receptors ,SUPPRESSOR cells ,INDOLEAMINE 2,3-dioxygenase ,PROGRAMMED cell death 1 receptors ,CYTOTOXIC T cells - Abstract
Tryptophan catabolism by the enzymes indoleamine 2,3-dioxygenase 1 and tryptophan 2,3-dioxygenase 2 (IDO/TDO) promotes immunosuppression across different cancer types. The tryptophan metabolite L-Kynurenine (Kyn) interacts with the ligand-activated transcription factor aryl hydrocarbon receptor (AHR) to drive the generation of Tregs and tolerogenic myeloid cells and PD-1 up-regulation in CD8
+ T cells. Here, we show that the AHR pathway is selectively active in IDO/TDO-overexpressing tumors and is associated with resistance to immune checkpoint inhibitors. We demonstrate that IDO-Kyn-AHR-mediated immunosuppression depends on an interplay between Tregs and tumor-associated macrophages, which can be reversed by AHR inhibition. Selective AHR blockade delays progression in IDO/TDO-overexpressing tumors, and its efficacy is improved in combination with PD-1 blockade. Our findings suggest that blocking the AHR pathway in IDO/TDO expressing tumors would overcome the limitation of single IDO or TDO targeting agents and constitutes a personalized approach to immunotherapy, particularly in combination with immune checkpoint inhibitors. The tryptophan metabolite kynurenine is an endogenous ligand of the aryl hydrocarbon receptor (AHR). Here, the authors show that AHR targeting in IDO/TDO-expressing tumours counteracts a regulatory T cell/macrophage suppressive axis and synergizes with immune checkpoint blockade to hinder tumour growth. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
8. Strategies for Predicting Response to Checkpoint Inhibitors.
- Author
-
Zappasodi, Roberta, Wolchok, Jedd D., and Merghoub, Taha
- Abstract
Purpose of Review: Despite the clinical successes of immune checkpoint blockade across multiple tumor types, many patients do not respond to these therapies or become resistant after an initial response. This underscores the need to improve our understanding of the molecular determinants of response to guide more personalized and rational utilization of these therapies. Here, we describe available biomarkers of checkpoint blockade activity by classifying them into four major categories: tumor-intrinsic, immune microenvironmental, host-related, and dynamic factors.Recent Findings: The clinical experience accumulated thus far with checkpoint blockade now offers the opportunity to comprehensively study the molecular and immune features associated with response. This is yielding a growing set of biomarkers whose integration will be key to more accurately predict clinical outcome.Summary: We propose a model for systematic assessment of available baseline and dynamic biomarkers in relationship with patients’ outcomes. This will improve our understanding of the tumor-immune interactions and dynamics that predict a clinical response and will provide key information to develop more personalized and effective treatment strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
9. Editorial: Factors determining long term anti-tumor responses to immune checkpoint blockade therapy.
- Author
-
Zappasodi, Roberta, Cook, Graham P., and Taylor, Alison
- Subjects
IMMUNE checkpoint proteins ,IMMUNE response - Published
- 2022
- Full Text
- View/download PDF
10. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer.
- Author
-
Balachandran, Vinod P., Łuksza, Marta, Zhao, Julia N., Makarov, Vladimir, Moral, John Alec, Remark, Romain, Herbst, Brian, Askan, Gokce, Bhanot, Umesh, Senbabaoglu, Yasin, Wells, Daniel K., Cary, Charles Ian Ormsby, Grbovic-Huezo, Olivera, Attiyeh, Marc, Medina, Benjamin, Zhang, Jennifer, Loo, Jennifer, Saglimbeni, Joseph, Abu-Akeel, Mohsen, and Zappasodi, Roberta
- Abstract
Pancreatic ductal adenocarcinoma is a lethal cancer with fewer than 7% of patients surviving past 5 years. T-cell immunity has been linked to the exceptional outcome of the few long-term survivors, yet the relevant antigens remain unknown. Here we use genetic, immunohistochemical and transcriptional immunoprofiling, computational biophysics, and functional assays to identify T-cell antigens in long-term survivors of pancreatic cancer. Using whole-exome sequencing and in silico neoantigen prediction, we found that tumours with both the highest neoantigen number and the most abundant CD8
+ T-cell infiltrates, but neither alone, stratified patients with the longest survival. Investigating the specific neoantigen qualities promoting T-cell activation in long-term survivors, we discovered that these individuals were enriched in neoantigen qualities defined by a fitness model, and neoantigens in the tumour antigen MUC16 (also known as CA125). A neoantigen quality fitness model conferring greater immunogenicity to neoantigens with differential presentation and homology to infectious disease-derived peptides identified long-term survivors in two independent datasets, whereas a neoantigen quantity model ascribing greater immunogenicity to increasing neoantigen number alone did not. We detected intratumoural and lasting circulating T-cell reactivity to both high-quality and MUC16 neoantigens in long-term survivors of pancreatic cancer, including clones with specificity to both high-quality neoantigens and predicted cross-reactive microbial epitopes, consistent with neoantigen molecular mimicry. Notably, we observed selective loss of high-quality and MUC16 neoantigenic clones on metastatic progression, suggesting neoantigen immunoediting. Our results identify neoantigens with unique qualities as T-cell targets in pancreatic ductal adenocarcinoma. More broadly, we identify neoantigen quality as a biomarker for immunogenic tumours that may guide the application of immunotherapies. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
11. Microenvironment modulation and enhancement of antilymphoma therapy by the heparanase inhibitor roneparstat.
- Author
-
Rossini, Anna, Zunino, Franco, Ruggiero, Giusi, De Cesare, Michelandrea, Cominetti, Denis, Tortoreto, Monica, Lanzi, Cinzia, Cassinelli, Giuliana, Zappasodi, Roberta, Tripodo, Claudio, Gulino, Alessandro, Zaffaroni, Nadia, and Di Nicola, Massimo
- Published
- 2018
- Full Text
- View/download PDF
12. Lymphoma immunotherapy: current status.
- Author
-
Zappasodi, Roberta, de Braud, Filippo, Di Nicola, Massimo, Umansky, Viktor, and Berraondo, Pedro
- Subjects
LYMPHOMA treatment ,IMMUNOTHERAPY ,IMMUNE response - Abstract
The rationale to treat lymphomas with immunotherapy comes from long-standing evidence on their distinctive immune responsiveness. Indolent B-cell non-Hodgkin lymphomas, in particular, establish key interactions with the immune microenvironment to ensure prosurvival signals and prevent antitumor immune activation. However, reports of spontaneous regressions indicate that, under certain circumstances, patients develop therapeutic antitumor immunity. Several immunotherapeutic approaches have been thus developed to boost these effects in all patients. To date, targeting CD20 on malignant B cells with the antibody rituximab has been the most clinically effective strategy. However, relapse and resistance prevent to cure approximately half of B-NHL patients, underscoring the need of more effective therapies. The recognition of B-cell receptor variable regions as B-NHL unique antigens promoted the development of specific vaccines to immunize patients against their own tumor. Despite initial promising results, this strategy has not yet demonstrated a sufficient clinical benefit to reach the regulatory approval. Several novel agents are now available to stimulate immune effector functions or counteract immunosuppressive mechanisms, such as engineered antitumor T cells, co-stimulatory receptor agonist, and immune checkpoint-blocking antibodies. Thus, multiple elements can now be exploited in more effective combinations to break the barriers for the induction of anti-lymphoma immunity. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
13. Alphavirus-based vaccines in melanoma: rationale and potential improvements in immunotherapeutic combinations.
- Author
-
Zappasodi, Roberta and Merghoub, Taha
- Published
- 2015
- Full Text
- View/download PDF
14. Non-Hodgkin's Lymphomas.
- Author
-
Zappasodi, Roberta and Di Nicola, Massimo
- Published
- 2012
- Full Text
- View/download PDF
15. Pleiotropic antitumor effects of the pan-HDAC inhibitor ITF2357 against c-Myc-overexpressing human B-cell non-Hodgkin lymphomas.
- Author
-
Zappasodi, Roberta, Cavanè, Alessandra, Iorio, Marilena V., Tortoreto, Monica, Guarnotta, Carla, Ruggiero, Giusi, Piovan, Claudia, Magni, Michele, Zaffaroni, Nadia, Tagliabue, Elda, Croce, Carlo M., Zunino, Franco, Gianni, Alessandro M., and Di Nicola, Massimo
- Abstract
Histone deacetylases (HDAC) extensively contribute to the c-Myc oncogenic program, pointing to their inhibition as an effective strategy against c-Myc-overexpressing cancers. We, thus, studied the therapeutic activity of the new-generation pan-HDAC inhibitor ITF2357 (Givinostat®) against c-Myc-overexpressing human B-cell non-Hodgkin lymphomas (B-NHLs). ITF2357 anti-proliferative and pro-apoptotic effects were analyzed in B-NHL cell lines with c-Myc translocations (Namalwa, Raji and DOHH-2), stabilizing mutations (Raji) or post-transcriptional alterations (SU-DHL-4) in relationship to c-Myc modulation. ITF2357 significantly delayed the in vitro growth of all B-NHL cell lines by inducing G1 cell-cycle arrest, eventually followed by cell death. These events correlated with the extent of c-Myc protein, but not mRNA, downregulation, indicating the involvement of post-transcriptional mechanisms. Accordingly, c-Myc-targeting microRNAs let-7a and miR-26a were induced in all treated lymphomas and the cap-dependent translation machinery components 4E-BP1, eIF4E and eIF4G, as well as their upstream regulators, Akt and PIM kinases, were inhibited in function of the cell sensitivity to ITF2357, and, in turn, c-Myc downregulation. In vivo, ITF2357 significantly hampered the growth of Namalwa and Raji xenografts in immunodeficient mice. Noteworthy, its combination with suboptimal cyclophosphamide, achieved complete remissions in most animals and equaled or even exceeded the activity of optimal cyclophosphamide. Collectively, our findings provide the rationale for testing the clinical advantages of adding ITF2357 to current therapies for the still very ominous c-Myc-overexpressing lymphomas. They equally provide the proof-of-concept for its clinical evaluation in rational combination with the promising inhibitors of B-cell receptor and PI3K/Akt/mTOR axis currently in the process of development. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
16. Author Correction: Fundamental immune–oncogenicity trade-offs define driver mutation fitness.
- Author
-
Hoyos, David, Zappasodi, Roberta, Schulze, Isabell, Sethna, Zachary, de Andrade, Kelvin César, Bajorin, Dean F., Bandlamudi, Chaitanya, Callahan, Margaret K., Funt, Samuel A., Hadrup, Sine R., Holm, Jeppe S., Rosenberg, Jonathan E., Shah, Sohrab P., Vázquez-García, Ignacio, Weigelt, Britta, Wu, Michelle, Zamarin, Dmitriy, Campitelli, Laura F., Osborne, Edward J., and Klinger, Mark
- Published
- 2022
- Full Text
- View/download PDF
17. Targeting Phosphatidylserine Enhances the Anti-tumor Response to Tumor-Directed Radiation Therapy in a Preclinical Model of Melanoma.
- Author
-
Budhu, Sadna, Giese, Rachel, Gupta, Aditi, Fitzgerald, Kelly, Zappasodi, Roberta, Schad, Sara, Hirschhorn, Daniel, Campesato, Luis Felipe, De Henau, Olivier, Gigoux, Mathieu, Liu, Cailian, Mazo, Gregory, Deng, Liang, Barker, Christopher A., Wolchok, Jedd D., and Merghoub, Taha
- Abstract
Phosphatidylserine (PS) is exposed on the surface of apoptotic cells and is known to promote immunosuppressive signals in the tumor microenvironment (TME). Antibodies that block PS interaction with its receptors have been shown to repolarize the TME into a proinflammatory state. Radiation therapy (RT) is an effective focal treatment of isolated solid tumors but is less effective at controlling metastatic cancers. We found that tumor-directed RT caused an increase in expression of PS on the surface of viable immune infiltrates in mouse B16 melanoma. We hypothesize that PS expression on immune cells may provide negative feedback to immune cells in the TME. Treatment with an antibody that targets PS (mch1N11) enhanced the anti-tumor efficacy of tumor-directed RT and improved overall survival. This combination led to an increase in proinflammatory tumor-associated macrophages. The addition of anti-PD-1 to RT and mch1N11 led to even greater anti-tumor efficacy and overall survival. We found increased PS expression on several immune subsets in the blood of patients with metastatic melanoma after receiving tumor-directed RT. These findings highlight the potential of combining PS targeting with RT and PD-1 pathway blockade to improve outcomes in patients with advanced-stage cancers. • PS expression on immune cells in murine melanoma is increased after tumor-directed RT • Blocking PS interaction with its receptors enhances the anti-tumor efficacy of RT • PD-1 blockade further potentiates the anti-tumor responses • Melanoma patients' PBMCs exhibit an increase in PS expression after RT Budhu et al. show that tumor-directed irradiation of murine B16 melanoma causes an increase in PS on the surface of infiltrating immune cells. Blocking PS and RT improves the anti-tumor efficacy and overall survival, which can be further improved with the addition of anti-PD-1. Melanoma patients exhibit increased PS on their PBMCs after RT. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
18. Correction to: Strategies for Predicting Response to Checkpoint Inhibitors.
- Author
-
Zappasodi, Roberta, Wolchok, Jedd D., and Merghoub, Taha
- Abstract
The original version of this article unfortunately contained a mistake. The conflict of interest statement was incorrect. The corrected statement is given below. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.