86 results on '"Zappasodi R"'
Search Results
2. Correction to: Toward a comprehensive view of cancer immune responsiveness: A synopsis from the SITC workshop (Journal for ImmunoTherapy of Cancer (2020) 7 (131) DOI: 10.1186/s40425-019-0602-4)
- Author
-
Bedognetti, D., Ceccarelli, M., Galluzzi, L., Lu, R., Palucka, K., Samayoa, J., Spranger, S., Warren, S., Wong, K. -K., Ziv, E., Chowell, D., Coussens, L. M., De Carvalho, D. D., Denardo, D. G., Galon, J., Kaufman, H. L., Kirchhoff, T., Lotze, M. T., Luke, J. J., Minn, A. J., Politi, K., Shultz, L. D., Simon, R., Thorsson, V., Weidhaas, J. B., Ascierto, M. L., Ascierto, P. A., Barnes, J. M., Barsan, V., Bommareddy, P. K., Bot, A., Church, S. E., Ciliberto, G., De Maria, A., Draganov, D., W. S., Ho, Mcgee, H. M., Monette, A., Murphy, J. F., Nistico, P., Park, W., Patel, M., Quigley, M., Radvanyi, L., Raftopoulos, H., Rudqvist, N. -P., Snyder, A., Sweis, R. F., Valpione, S., Zappasodi, R., Butterfield, L. H., Disis, M. L., Fox, B. A., Cesano, A., and Marincola, F. M.
- Published
- 2019
3. Pleiotropic antitumor effects of the pan-HDAC inhibitor ITF2357 against c-Myc-overexpressing human B-cell non-Hodgkin lymphomas
- Author
-
Zappasodi, R., Cavanè, A., Iorio, M., Tortoreto, M., Guarnotta, C., Ruggiero, G., Piovan, C., Magni, M., Zaffaroni, N., Tagliabue, E., Croce, C., Zunino, F., Gianni, A., Di Nicola, M., Zappasodi, R, Cavanè, A, Iorio, MV, Tortoreto, M, Guarnotta, C, Ruggiero, G, Piovan, C, Magni, M, Zaffaroni, N, Tagliabue, E, Croce, CM, Zunino, F, Gianni, AM, and Di Nicola, M
- Subjects
c-Myc ,microRNA ,non-Hodgkin lymphoma ,histone deacetylase inhibitors ,histone deacetylase inhibitor - 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.
- Published
- 2014
4. Tenth annual meeting of the Italian Network for Tumor Biotherapy (NIBIT), SIENA, Italy, November 5-7, 2012
- Author
-
Maio, M., Nicolay, H. J. M., Ascierto, P. A., Belardelli, F., Camerini, R., Colombo, M. P., Queirolo, P., Ridolfi, R., Russo, V., Parisi, G., Cutaia, O., Fonsatti, E., Parmiani, G., Mennonna, D., Carluccio, S., Bellone, M., Maccalli, C., Brendolan, A., Mondino, A., Corti, A., Bondanza, A., Locatelli, F., Seliger, B., Filaci, G., Rosato, A., Pittoni, P., Tazzari, M., Rivoltini, L., Anichini, A., Vallacchi, V., Zappasodi, R., Quaglino, E., Moresco, R. M., Camisaschi, C., Calabro, L., Ferrucci, P. F., Fratta, E., Ugel, S., van Baren, N., Guidoboni, M., Covre, A., Carbone, E., Aurisicchio, L., Palmieri, G., Di Giacomo, A. M., Volonte, A., Jachetti, E., and Sangiolo, D.
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Immunology ,Cancer ,medicine.disease ,NIBIT ,Oncology ,Family medicine ,Immunotherapy ,Networks ,Cancer, Immunology, Immunotherapy, Networks, NIBIT ,Immunology and Allergy ,Medicine ,business - Published
- 2013
5. The histone deacetylase inhibitor ITF2357 (Givinostat) promotes Burkitt’s lymphoma cell line death modulating micro-RNA and Tissue Transglutaminase 2 expression
- Author
-
Di Nicola, M., Zappasodi, R., Baldan, F., Iorio, M. V., Magni, M., Tagliabue, E., Giacomini, A, Croce, C. M., Carlo- Stella, C., and Gianni, A. M.
- Published
- 2009
6. Sialidase NEU4 is involved in glioblastoma stem cell survival
- Author
-
Silvestri, I, primary, Testa, F, additional, Zappasodi, R, additional, Cairo, C W, additional, Zhang, Y, additional, Lupo, B, additional, Galli, R, additional, Di Nicola, M, additional, Venerando, B, additional, and Tringali, C, additional
- Published
- 2014
- Full Text
- View/download PDF
7. 281 Delta15HER2 – a Player in HER2-driven Tumor Progression
- Author
-
Ghedini, G.C., primary, Marzano, G., additional, Castagnoli, L., additional, Ciravolo, V., additional, Amici, A., additional, Zappasodi, R., additional, Santilli, G., additional, Iezzi, M., additional, Tagliabue, E., additional, and Pupa, S.M., additional
- Published
- 2012
- Full Text
- View/download PDF
8. 602 Identification of HSP105 as a novel non-Hodgkin lymphoma restricted antigen
- Author
-
Pupa, S.M., primary, Zappasodi, R., additional, Ghedini, G.C., additional, Castagnoli, L., additional, Aiello, P., additional, Miccichè, F., additional, Cabras, A.D., additional, Bongarzone, I., additional, and Gianni, A.M., additional
- Published
- 2010
- Full Text
- View/download PDF
9. 537 Potent in vitro and in vivo anti-tumor activity of ITF2357 by modulation of c-myc related miRNA signature in human Burkitt's lymphoma
- Author
-
Zappasodi, R., primary, Iorio, M.V., additional, Cavanè, A., additional, Magni, M., additional, Ruggiero, G., additional, Carlo-Stella, C., additional, Croce, C.M., additional, Gianni, A.M., additional, and Nicola, M. Di, additional
- Published
- 2010
- Full Text
- View/download PDF
10. Identification of HSP105 as a novel B-cell non-Hodgkin lymphoma (NHL) antigen (ag).
- Author
-
Zappasodi, R., primary, Pupa, S., additional, Bongarzone, I., additional, Ghedini, G. C., additional, Castagnoli, L., additional, Miccichè, F., additional, Cabras, A., additional, Carlo-Stella, C., additional, Gianni, A. M., additional, and Di Nicola, M. A., additional
- Published
- 2010
- Full Text
- View/download PDF
11. The effect of artificial antigen-presenting cells with preclustered anti-CD28/-CD3/-LFA-1 monoclonal antibodies on the induction of ex vivo expansion of functional human antitumor T cells
- Author
-
Zappasodi, R., primary, Di Nicola, M., additional, Carlo-Stella, C., additional, Mortarini, R., additional, Molla, A., additional, Vegetti, C., additional, Albani, S., additional, Anichini, A., additional, and Gianni, A. M., additional
- Published
- 2008
- Full Text
- View/download PDF
12. Immunization of indolent non-Hodgkin’s lymphoma patients with autologous monocyte-derived dendritic cells loaded with heat shocked and killed autologous tumor cells: A phase I study
- Author
-
Di Nicola, M. A., primary, Carlo-Stella, C., additional, Zappasodi, R., additional, Passoni, L., additional, Liliana, D., additional, Magni, M., additional, Matteucci, P., additional, Mortarini, R., additional, Anichini, A., additional, and Gianni, A. M., additional
- Published
- 2007
- Full Text
- View/download PDF
13. HSP105 inhibition hampers the growth of human aggressive B-cell non-Hodgkin lymphoma by counteracting key oncogenic pathways
- Author
-
Zappasodi, R., Cavane, A., Tortoreto, M., Cristina Alessandra Tringali, Cabras, A. D., Ruggiero, G., Castagnoli, L., Venerando, B., Zaffaroni, N., Pupa, S. M., Gianni, A. M., and Di Nicola, M.
14. SEROLOGICAL IDENTIFICATION OF HSP105 AS A NOVEL NON-HODGKIN LYMPHOMA ASSOCIATED ANTIGEN
- Author
-
Di Nicola, M., Zappasodi, R., Ghedini, G. C., Bongarzone, I., Lorenzo Castagnoli, Cabras, A. D., Tripodo, C., Messina, A., Tortoreto, M., Pupa, S. M., and Gianni, A. M.
15. Involvement of sialidase NEU4 expression in glioblastoma stem cells malignancy
- Author
-
Cristina Alessandra Tringali, Zappasodi, R., Lupo, B., Anastasia, L., Papini, N., Tettamanti, G., Di Nicola, M., and Venerando, B.
16. Rational design of anti-GITR-based combination immunotherapy
- Author
-
Zappasodi, R, primary, Sirard, C, additional, Li, Y, additional, Budhu, S, additional, Abu-Akeel, M, additional, Liu, C, additional, Yang, X, additional, Zhong, H, additional, Newman, W, additional, Qi, J, additional, Wong, P, additional, Schaer, D, additional, Koon, H, additional, Velcheti, V, additional, Hellman, MD, additional, Postow, MA, additional, Callahan, MK, additional, Wolchok, JD, additional, and Merghoub, T, additional
- Full Text
- View/download PDF
17. HSPH1 inhibition downregulates Bcl-6 and c-Myc and hampers the growth of human aggressive B-cell non-Hodgkin lymphoma
- Author
-
Giusi Ruggiero, Antonello Cabras, Serenella M. Pupa, Monica Tortoreto, Claudio Tripodo, Alessandro M. Gianni, Carla Guarnotta, Cristina Tringali, Massimo Di Nicola, Bruno Venerando, Filippo de Braud, Alessandra Cavanè, Roberta Zappasodi, Nadia Zaffaroni, Lorenzo Castagnoli, Zappasodi, R., Ruggiero, G., Guarnotta, C., Tortoreto, M., Tringali, C., Cavanè, A., Cabras, A., Castagnoli, L., Venerando, B., Zaffaroni, N., Gianni, A., De Braud, F., Tripodo, C., Pupa, S., and Di Nicola, M.
- Subjects
Lymphoma, B-Cell ,Xenograft Model Antitumor Assay ,DNA-Binding Protein ,Immunology ,Down-Regulation ,Mice, SCID ,Settore MED/08 - Anatomia Patologica ,Biology ,Biochemistry ,HSP110 Heat-Shock Protein ,Proto-Oncogene Proteins c-myc ,Mice ,Downregulation and upregulation ,immune system diseases ,Cell Line, Tumor ,hemic and lymphatic diseases ,Heat shock protein ,Gene Knockdown Techniques ,medicine ,Animals ,Humans ,Gene silencing ,HSP110 Heat-Shock Proteins ,DNA-Binding Proteins ,Xenograft Model Antitumor Assays ,Medicine (all) ,Hematology ,Cell Biology ,Animal ,medicine.disease ,In vitro ,Lymphoma ,Cell culture ,Gene Knockdown Technique ,Proto-Oncogene Proteins c-bcl-6 ,Cancer research ,B-Cell Non-Hodgkin Lymphoma ,Human - Abstract
We have shown that human B-cell non-Hodgkin lymphomas (B-NHLs) express heat shock protein (HSP)H1/105 in function of their aggressiveness. Here, we now clarify its role as a functional B-NHL target by testing the hypothesis that it promotes the stabilization of key lymphoma oncoproteins. HSPH1 silencing in 4 models of aggressive B-NHLs was paralleled by Bcl-6 and c-Myc downregulation. In vitro and in vivo analysis of HSPH1-silenced Namalwa cells showed that this effect was associated with a significant growth delay and the loss of tumorigenicity when 10(4) cells were injected into mice. Interestingly, we found that HSPH1 physically interacts with c-Myc and Bcl-6 in both Namalwa cells and primary aggressive B-NHLs. Accordingly, expression of HSPH1 and either c-Myc or Bcl-6 positively correlated in these diseases. Our study indicates that HSPH1 concurrently favors the expression of 2 key lymphoma oncoproteins, thus confirming its candidacy as a valuable therapeutic target of aggressive B-NHLs.
- Published
- 2015
18. Immunotherapy advances in uro-genital malignancies
- Author
-
Elena Verzoni, Massimo Di Nicola, Giuseppe Procopio, Roberto Salvioni, RAFFAELE RATTA, Andrea Necchi, Raffaele Ratta, Paolo Grassi, Filippo Guglielmo Maria De Braud, Daniele Raggi, Roberta Zappasodi, Ratta, R, Zappasodi, R, Raggi, D, Grassi, P, Verzoni, E, Necchi, A, Di Nicola, M, Salvioni, R, de Braud, F, and Procopio, G
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Antineoplastic Agents ,Pembrolizumab ,Cancer Vaccines ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,Atezolizumab ,Internal medicine ,medicine ,Animals ,Humans ,Brentuximab vedotin ,business.industry ,Cancer ,Hematology ,Immunotherapy ,medicine.disease ,Immune checkpoint ,030104 developmental biology ,030220 oncology & carcinogenesis ,Nivolumab ,business ,Urogenital Neoplasms ,medicine.drug - Abstract
Immunotherapy for the treatment of cancer has made significant progresses over the last 20 years. Multiple efforts have been attempted to restore immune-mediated tumor elimination, leading to the development of several targeted immunotherapies. Data from recent clinical trials suggest that these agents might improve the prognosis of patients with advanced genito-urinary (GU) malignancies. Nivolumab has been the first immune checkpoint-inhibitor approved for pre-treated patients with metastatic renal cell carcinoma. Pembrolizumab and atezolizumab have shown promising results in both phase I and II trials in urothelial carcinoma. Brentuximab vedotin has demonstrated early signals of clinical activity and immunomodulatory effects in highly pre-treated patients with testicular germ cell tumors. In this review, we have summarized the major clinical achievements of immunotherapy in GU cancers, focusing on immune checkpoint blockade as well as the new immunomodulatory monoclonal antibodies (mAbs) under clinical evaluation for these malignancies. (C) 2016 Elsevier Ireland Ltd. All rights reserved.
- Published
- 2016
19. Activated d16HER2 homodimers and src kinase mediate optimal efficacy for trastuzumab
- Author
-
Patrizia Gasparini, Giulia Marzano, Patrizia Nanni, Manuela Campiglio, Elda Tagliabue, Arianna Palladini, Pier Luigi Lollini, Claudia Chiodoni, Manuela Iezzi, Alessia Lamolinara, Lorenzo Castagnoli, Valentina Ciravolo, Tiziana Triulzi, Serenella M. Pupa, Gaia C. Ghedini, Augusto Amici, Roberta Zappasodi, Sylvie Ménard, Castagnoli, L, Iezzi, M, Ghedini, Gc, Ciravolo, V, Marzano, G, Lamolinara, A, Zappasodi, R, Gasparini, P, Campiglio, M, Amici, A, Chiodoni, C, Palladini, Arianna, Lollini, PIER LUIGI, Triulzi, T, Menard, S, Nanni, Patrizia, Tagliabue, E, and Pupa, Sm
- Subjects
Genetically modified mouse ,Cancer Research ,Receptor, ErbB-2 ,Transgene ,Mice, Transgenic ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Mice ,mammary cancer ,Trastuzumab ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Isoforms ,skin and connective tissue diseases ,Receptor ,neoplasms ,Effector ,business.industry ,Exons ,src-Family Kinases ,Oncology ,HER-2 ,Drug Resistance, Neoplasm ,Monoclonal ,Cancer research ,Female ,Signal transduction ,Neoplasm Recurrence, Local ,Protein Multimerization ,business ,Delta16-HER-2 ,medicine.drug ,Proto-oncogene tyrosine-protein kinase Src ,Signal Transduction - Abstract
A splice isoform of the HER2 receptor that lacks exon 16 (d16HER2) is expressed in many HER2-positive breast tumors, where it has been linked with resistance to the HER2-targeting antibody trastuzumab, but the impact of d16HER2 on tumor pathobiology and therapeutic response remains uncertain. Here, we provide genetic evidence in transgenic mice that expression of d16HER2 is sufficient to accelerate mammary tumorigenesis and improve the response to trastuzumab. A comparative analysis of effector signaling pathways activated by d16HER2 and wild-type HER2 revealed that d16HER2 was optimally functional through a link to SRC activation (pSRC). Clinically, HER2-positive breast cancers from patients who received trastuzumab exhibited a positive correlation in d16HER2 and pSRC abundance, consistent with the mouse genetic results. Moreover, patients expressing high pSRC or an activated “d16HER2 metagene” were found to derive the greatest benefit from trastuzumab treatment. Overall, our results establish the d16HER2 signaling axis as a signature for decreased risk of relapse after trastuzumab treatment. Cancer Res; 74(21); 6248–59. ©2014 AACR.
- Published
- 2014
20. Serological identification of HSP105 as a novel non-Hodgkin lymphoma therapeutic target
- Author
-
Italia Bongarzone, Antonella Messina, Claudio Tripodo, Roberta Zappasodi, Carmelo Carlo-Stella, Monica Tortoreto, Michele Magni, Alessandro M. Gianni, Gaia C. Ghedini, Massimo Di Nicola, Lorenzo Castagnoli, Antonello Cabras, Serenella M. Pupa, Zappasodi, R, Bongarzone, I, Ghedini, GC, Castagnoli, L, Cabras, AD, Messina, A, Tortoreto, M, Tripodo, C, Magni, M, Carlo-Stella, C, Gianni, AM, Pupa, SM, and Di Nicola, M.
- Subjects
Immunology ,Mice, SCID ,Biochemistry ,Antibodies ,Flow cytometry ,Antigen-Antibody Reactions ,Cohort Studies ,HSP105 ,Mice ,Antigen ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Serologic Tests ,HSP110 Heat-Shock Proteins ,medicine.diagnostic_test ,biology ,business.industry ,Lymphoma, Non-Hodgkin ,non-Hodgkin lymphoma ,Cell Biology ,Hematology ,Cell cycle ,medicine.disease ,Immunohistochemistry ,Lymphoma ,Granzyme B ,Gene Expression Regulation, Neoplastic ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,biology.protein ,Antibody ,business ,Diffuse large B-cell lymphoma - Abstract
We reported that the clinical efficacy of dendritic cell–based vaccination is strongly associated with immunologic responses in relapsed B-cell non-Hodgkin lymphoma (B-NHL) patients. We have now investigated whether postvaccination antibodies from responders recognize novel shared NHL-restricted antigens. Immunohistochemistry and flow cytometry showed that they cross-react with allogeneic B-NHLs at significantly higher levels than their matched prevaccination samples or nonresponders' antibodies. Western blot analysis of DOHH-2 lymphoma proteome revealed a sharp band migrating at approximately 100 to 110 kDa only with postvaccine repertoires from responders. Mass spectrometry identified heat shock protein-105 (HSP105) in that molecular weight interval. Flow cytometry and immunohistochemistry disclosed HSP105 on the cell membrane and in the cytoplasm of B-NHL cell lines and 97 diagnostic specimens. A direct correlation between HSP105 expression and lymphoma aggressiveness was also apparent. Treatment of aggressive human B-NHL cell lines with an anti-HSP105 antibody had no direct effects on cell cycle or apoptosis but significantly reduced the tumor burden in xenotransplanted immunodeficient mice. In vivo antilymphoma activity of HSP105 engagement was associated with a significant local increase of Granzyme B+ killer cells that very likely contributed to the tumor-restricted necrosis. Our study adds HSP105 to the list of nononcogenes that can be exploited as antilymphoma targets.
- Published
- 2011
21. Pharmacologic LDH inhibition redirects intratumoral glucose uptake and improves antitumor immunity in solid tumor models.
- Author
-
Verma S, Budhu S, Serganova I, Dong L, Mangarin LM, Khan JF, Bah MA, Assouvie A, Marouf Y, Schulze I, Zappasodi R, Wolchok JD, and Merghoub T
- Subjects
- Animals, Mice, Humans, Glucose Transporter Type 1 metabolism, Glucose Transporter Type 1 antagonists & inhibitors, Glucose Transporter Type 1 immunology, Glucose Transporter Type 1 genetics, Cell Line, Tumor, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory drug effects, Melanoma, Experimental immunology, Melanoma, Experimental pathology, Melanoma, Experimental drug therapy, Melanoma, Experimental metabolism, Glycolysis drug effects, Female, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating drug effects, Colonic Neoplasms immunology, Colonic Neoplasms drug therapy, Colonic Neoplasms pathology, Colonic Neoplasms metabolism, Enzyme Inhibitors pharmacology, Immunotherapy, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Glucose metabolism, Tumor Microenvironment immunology, Tumor Microenvironment drug effects, L-Lactate Dehydrogenase metabolism, L-Lactate Dehydrogenase antagonists & inhibitors, L-Lactate Dehydrogenase immunology
- Abstract
Tumor reliance on glycolysis is a hallmark of cancer. Immunotherapy is more effective in controlling glycolysis-low tumors lacking lactate dehydrogenase (LDH) due to reduced tumor lactate efflux and enhanced glucose availability within the tumor microenvironment (TME). LDH inhibitors (LDHi) reduce glucose uptake and tumor growth in preclinical models, but their impact on tumor-infiltrating T cells is not fully elucidated. Tumor cells have higher basal LDH expression and glycolysis levels compared with infiltrating T cells, creating a therapeutic opportunity for tumor-specific targeting of glycolysis. We demonstrate that LDHi treatment (a) decreases tumor cell glucose uptake, expression of the glucose transporter GLUT1, and tumor cell proliferation while (b) increasing glucose uptake, GLUT1 expression, and proliferation of tumor-infiltrating T cells. Accordingly, increasing glucose availability in the microenvironment via LDH inhibition leads to improved tumor-killing T cell function and impaired Treg immunosuppressive activity in vitro. Moreover, combining LDH inhibition with immune checkpoint blockade therapy effectively controls murine melanoma and colon cancer progression by promoting effector T cell infiltration and activation while destabilizing Tregs. Our results establish LDH inhibition as an effective strategy for rebalancing glucose availability for T cells within the TME, which can enhance T cell function and antitumor immunity.
- Published
- 2024
- Full Text
- View/download PDF
22. Loss of CREBBP and KMT2D cooperate to accelerate lymphomagenesis and shape the lymphoma immune microenvironment.
- Author
-
Li J, Chin CR, Ying HY, Meydan C, Teater MR, Xia M, Farinha P, Takata K, Chu CS, Jiang Y, Eagles J, Passerini V, Tang Z, Rivas MA, Weigert O, Pugh TJ, Chadburn A, Steidl C, Scott DW, Roeder RG, Mason CE, Zappasodi R, Béguelin W, and Melnick AM
- Subjects
- Animals, Mice, B-Lymphocytes metabolism, Chromatin genetics, Chromatin metabolism, Germinal Center metabolism, Mutation, Tumor Microenvironment genetics, Lymphoma, Large B-Cell, Diffuse genetics
- Abstract
Despite regulating overlapping gene enhancers and pathways, CREBBP and KMT2D mutations recurrently co-occur in germinal center (GC) B cell-derived lymphomas, suggesting potential oncogenic cooperation. Herein, we report that combined haploinsufficiency of Crebbp and Kmt2d induces a more severe mouse lymphoma phenotype (vs either allele alone) and unexpectedly confers an immune evasive microenvironment manifesting as CD8
+ T-cell exhaustion and reduced infiltration. This is linked to profound repression of immune synapse genes that mediate crosstalk with T-cells, resulting in aberrant GC B cell fate decisions. From the epigenetic perspective, we observe interaction and mutually dependent binding and function of CREBBP and KMT2D on chromatin. Their combined deficiency preferentially impairs activation of immune synapse-responsive super-enhancers, pointing to a particular dependency for both co-activators at these specialized regulatory elements. Together, our data provide an example where chromatin modifier mutations cooperatively shape and induce an immune-evasive microenvironment to facilitate lymphomagenesis., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
23. Germinal Center Dark Zone harbors ATR-dependent determinants of T-cell exclusion that are also identified in aggressive lymphoma.
- Author
-
Cancila V, Morello G, Bertolazzi G, Chan AS, Bastianello G, Paysan D, Jaynes PW, Schiavoni G, Mattei F, Piconese S, Revuelta MV, Noto F, De Ninno A, Cammarata I, Pagni F, Venkatachalapathy S, Sangaletti S, Di Napoli A, Vacca D, Lonardi S, Lorenzi L, Ferreri AJM, Belmonte B, Varano G, Colombo MP, Bicciato S, Inghirami G, Cerchietti L, Ponzoni M, Zappasodi R, Facchetti F, Foiani M, Casola S, Jeyasekharan AD, and Tripodo C
- Abstract
The germinal center (GC) dark zone (DZ) and light zone (LZ) regions spatially separate expansion and diversification from selection of antigen-specific B-cells to ensure antibody affinity maturation and B cell memory. The DZ and LZ differ significantly in their immune composition despite the lack of a physical barrier, yet the determinants of this polarization are poorly understood. This study provides novel insights into signals controlling asymmetric T-cell distribution between DZ and LZ regions. We identify spatially-resolved DNA damage response and chromatin compaction molecular features that underlie DZ T-cell exclusion. The DZ spatial transcriptional signature linked to T-cell immune evasion clustered aggressive Diffuse Large B-cell Lymphomas (DLBCL) for differential T cell infiltration. We reveal the dependence of the DZ transcriptional core signature on the ATR kinase and dissect its role in restraining inflammatory responses contributing to establishing an immune-repulsive imprint in DLBCL. These insights may guide ATR-focused treatment strategies bolstering immunotherapy in tumors marked by DZ transcriptional and chromatin-associated features., Competing Interests: CONFLICT OF INTERESTS ADJ has received consultancy fees from DKSH/Beigene, Roche, Gilead, Turbine Ltd, AstraZeneca, Antengene, Janssen, MSD and IQVIA; and research funding from Janssen and AstraZeneca.
- Published
- 2024
- Full Text
- View/download PDF
24. Modulating Treg stability to improve cancer immunotherapy.
- Author
-
Kang JH and Zappasodi R
- Subjects
- Humans, T-Lymphocytes, Regulatory, Tumor Escape, Immunotherapy, Tumor Microenvironment, Neoplasms pathology
- Abstract
Immunosuppressive regulatory T cells (Tregs) provide a main mechanism of tumor immune evasion. Targeting Tregs, especially in the tumor microenvironment (TME), continues to be investigated to improve cancer immunotherapy. Recent studies have unveiled intratumoral Treg heterogeneity and plasticity, furthering the complexity of the role of Tregs in tumor immunity and immunotherapy response. The phenotypic and functional diversity of intratumoral Tregs can impact their response to therapy and may offer new targets to modulate specific Treg subsets. In this review we provide a unifying framework of critical factors contributing to Treg heterogeneity and plasticity in the TME, and we discuss how this information can guide the development of more specific Treg-targeting therapies for cancer immunotherapy., Competing Interests: Declaration of interests R.Z. is inventor on patent applications related to work on GITR, PD-1, and CTLA-4. R.Z. is scientific advisory board member of iTEOS Therapeutics, has consulted for Leap Therapeutics, and receives grant support from AstraZeneca and Bristol Myers Squibb., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
25. APR-246 increases tumor antigenicity independent of p53.
- Author
-
Michels J, Venkatesh D, Liu C, Budhu S, Zhong H, George MM, Thach D, Yao ZK, Ouerfelli O, Liu H, Stockwell BR, Campesato LF, Zamarin D, Zappasodi R, Wolchok JD, and Merghoub T
- Subjects
- Mice, Animals, Tumor Suppressor Protein p53 genetics, Antigens, Neoplasm, CD8-Positive T-Lymphocytes, Melanoma
- Abstract
We previously reported that activation of p53 by APR-246 reprograms tumor-associated macrophages to overcome immune checkpoint blockade resistance. Here, we demonstrate that APR-246 and its active moiety, methylene quinuclidinone (MQ) can enhance the immunogenicity of tumor cells directly. MQ treatment of murine B16F10 melanoma cells promoted activation of melanoma-specific CD8
+ T cells and increased the efficacy of a tumor cell vaccine using MQ-treated cells even when the B16F10 cells lacked p53. We then designed a novel combination of APR-246 with the TLR-4 agonist, monophosphoryl lipid A, and a CD40 agonist to further enhance these immunogenic effects and demonstrated a significant antitumor response. We propose that the immunogenic effect of MQ can be linked to its thiol-reactive alkylating ability as we observed similar immunogenic effects with the broad-spectrum cysteine-reactive compound, iodoacetamide. Our results thus indicate that combination of APR-246 with immunomodulatory agents may elicit effective antitumor immune response irrespective of the tumor's p53 mutation status., (© 2023 Michels et al.)- Published
- 2023
- Full Text
- View/download PDF
26. Updates on radiotherapy-immunotherapy combinations: Proceedings of 6 th annual ImmunoRad conference.
- Author
-
Gregucci F, Spada S, Barcellos-Hoff MH, Bhardwaj N, Chan Wah Hak C, Fiorentino A, Guha C, Guzman ML, Harrington K, Herrera FG, Honeychurch J, Hong T, Iturri L, Jaffee E, Karam SD, Knott SRV, Koumenis C, Lyden D, Marciscano AE, Melcher A, Mondini M, Mondino A, Morris ZS, Pitroda S, Quezada SA, Santambrogio L, Shiao S, Stagg J, Telarovic I, Timmerman R, Vozenin MC, Weichselbaum R, Welsh J, Wilkins A, Xu C, Zappasodi R, Zou W, Bobard A, Demaria S, Galluzzi L, Deutsch E, and Formenti SC
- Subjects
- Humans, Combined Modality Therapy, Immunotherapy, Neoplasms radiotherapy, Neoplasms drug therapy
- Abstract
Focal radiation therapy (RT) has attracted considerable attention as a combinatorial partner for immunotherapy (IT), largely reflecting a well-defined, predictable safety profile and at least some potential for immunostimulation. However, only a few RT-IT combinations have been tested successfully in patients with cancer, highlighting the urgent need for an improved understanding of the interaction between RT and IT in both preclinical and clinical scenarios. Every year since 2016, ImmunoRad gathers experts working at the interface between RT and IT to provide a forum for education and discussion, with the ultimate goal of fostering progress in the field at both preclinical and clinical levels. Here, we summarize the key concepts and findings presented at the Sixth Annual ImmunoRad conference., Competing Interests: MHBH is or has have been a recipient of research grants paid to UCSF or in-kind resources from Roche-Genentech, Varian Medical Systems, Eli Lilly, Pathway Innovations and has received fees for consulting from EMD-Serono, Varian Medical Systems, Genentech, Pathway Innovation, Scholar Rock. KHhas Honoraria: Arch Oncology (Inst), AstraZeneca (Inst), BMS (Inst), Boehringer Ingelheim (Inst), Codiak Biosciences (Inst), F-Star Therapeutics (Inst), Inzen Therapeutics (Inst), Merck Serono (Inst), MSD (Inst), Oncolys Biopharma (Inst), Pfizer (Inst), Replimune (Inst), VacV Biotherapeutics (Inst); Consulting or Advisory Role: Arch Oncology (Inst), AstraZeneca (Inst), BMS (Inst), Boehringer Ingelheim (Inst), Inzen Therapeutics (Inst), Merck Serono (Inst), MSD (Inst), Oncolys BioPharma (Inst), Replimune (Inst); Speakers’ Bureau: BMS (Inst), Merck Serono (Inst), MSD (Inst); Research Funding: AstraZeneca (Inst), Boehringer Ingelheim (Inst), Merck Sharp & Dohme (Inst), Replimune (Inst). FGH received Grant or Research Support Companies from Accuray inc, Bioprotect, Bristol-Myers Squibb, Roche-ImFlame/ImCore, Nanobiotix, AstraZeneca, Debio Pharmaceuticals, Seagen, Eisai, MSD; Grant or Research Support Foundations from Prostate Cancer Foundation, San Salvatore Foundation; Investigator or Co-Investigator Clinical Trials in Bristol-Myers Squibb; Consultations: Johnson & Johnson; Academic Collaborations: EORTC chairman Gynecology Cancer Group, ESMO Scientific Committee member for drug development, ASTRO Scientific Committee Annual Meeting. TH has Consulting: Synthetic Biologics, Novocure, Boston Scientific, Inivata, Merck, GSK; Scientific Advisory Board: PanTher Therapeutics (Equity), Lustgarten; Research Funding (Clinical Trials): Taiho, AstraZeneca, BMS, GSK, IntraOp, Ipsen. EJ reports other support from Abmeta and Adventris, personal fees from Achilles, Dragonfly, Mestag, The Medical Home Group, and Surgtx, other support from Parker Institute, grants and other support from the Lustgarten Foundation, Genentech, BMS, and Break Through Cancer outside the submitted work. SDK receives clinical funding from AstraZeneca, Genentech, and Ionis; she also receives preclinical research funding from Roche. KS is founder and consultant for Faeth Therapeutics and Transomic Technologies. CK is the co-recipient of a Sponsored Research Agreement from Ion Beam Applications (IBA). AM is funded by the Associazione Italiana per la Ricerca sul Cancro (AIRC IG 2018 Id.21763 and AIRC Programma di ricerca 5 per Mille 2019 Id.22737). MM declare grants from Boehringer Ingelheim, AC Biosciences and MSD outside the submitted work. ZSM has Scientific Advisory Board roles and equity options with Archeus Technologies and Seneca Therapeutics. JS owns stock and is a member of the Scientific Advisory Board of Surface Oncology, and is a member of the Scientific Advisory Board of Domain Therapeutics. RT has research grants to his institution from: Varian Medical Systems, Elekta Oncology, Accuray, Inc; scientific advisory board member for: Reflexion Medical, ImmuneSensor Therapeutics. AW acknowledge funding from AstraZeneca and imCORE. RW has stock and other ownership interests with Boost Therapeutics, Immvira LLC, Reflexion Pharmaceuticals, Coordination Pharmaceuticals Inc., Magi Therapeutics, Oncosenescence, Aqualung Therapeutics Corporation, and Cyntegron; he has served in a consulting or advisory role for Aettis Inc., AstraZeneca, Coordination Pharmaceuticals, Genus, Merck Serono S.A., Nano Proteagen, NKGen Biotech, Shuttle Pharmaceuticals, Highlight Therapeutics, S.L., Aqualung Therapeutics Corporation; he has research grants with Varian and Regeneron. RZ is scientific advisory board member of iTeos Therapeutics, receives research grant support from Bristol Myers Squibb and AstraZeneca, and is inventor on patent applications related to work on GITR, CTLA-4, and PD-1 (patent numbers: US20180244793A1; US10323091B2; WO2018106864A1; WO2019094352A1). SD has received compensation for consultant/advisory services from Lytix Biopharma, Mersana Therapeutics, EMD Serono, Ono Pharmaceutical, and Genentech, and research support from Lytix Biopharma and Boehringer-Ingelheim for unrelated projects. LG is/has been holding research contracts with Lytix Biopharma, Promontory and Onxeo, has received consulting/advisory honoraria from Boehringer Ingelheim, AstraZeneca, OmniSEQ, Onxeo, The Longevity Labs, Inzen, Imvax, Sotio, Promontory, Noxopharm, EduCom, and the Luke Heller TECPR2 Foundation, and holds Promontory stock options. ED reports grants and personal fees from Roche Genentech; grants from Servier; grants from AstraZeneca; grants and personal fees from Merck-Serono; grants from BMS; and grants from MSD outside the submitted work. SCF has Consultant: Bayer, Bristol Myers Squibb, Varian, ViewRay, Accuray, Elekta, Janssen, Regeneron, GlaxoSmithKline, Eisai, Astra Zeneca, MedImmune, Merck US, EMD Serono/Merck, Genentech/ROCHE, Boehringer Ingelheim, Nanobiotix and Grant/Research: support from: Bristol Myers Squibb, Varian, Regeneron, Merck, Celldex. All other authors have no conflict of interest to declare., (© 2023 The Author(s). Published with license by Taylor & Francis Group, LLC.)
- Published
- 2023
- Full Text
- View/download PDF
27. Targeting GITR in cancer immunotherapy - there is no perfect knowledge.
- Author
-
Davar D and Zappasodi R
- Subjects
- Humans, Glucocorticoid-Induced TNFR-Related Protein, Immunosuppression Therapy, T-Lymphocytes, Regulatory, Immunotherapy, Neoplasms therapy
- Abstract
Glucocorticoid-induced TNFR-related protein (GITR) belongs to the TNFR superfamily (TNFRSF) and stimulates both the acquired and innate immunity. GITR is broadly expressed on immune cells, particularly regulatory T cells (Tregs) and natural killer (NK) cells. Given its potential to promote T effector function and impede Treg immune suppression, GITR is an attractive target for cancer immunotherapy. Preclinically, GITR agonists have demonstrated potent anti-tumor efficacy singly and in combination with a variety of agents, including PD-1 blockade. Multiple GITR agonists have been advanced into the clinic, although the experience with these agents has been disappointing. Recent mechanistic insights into the roles of antibody structure, valency, and Fc functionality in mediating anti-tumor efficacy may explain some of the apparent inconsistency or discordance between preclinical data and observed clinical efficacy.
- Published
- 2023
- Full Text
- View/download PDF
28. Society for Immunotherapy of Cancer (SITC) consensus definitions for resistance to combinations of immune checkpoint inhibitors.
- Author
-
Kluger H, Barrett JC, Gainor JF, Hamid O, Hurwitz M, LaVallee T, Moss RA, Zappasodi R, Sullivan RJ, Tawbi H, and Sharon E
- Subjects
- Humans, Consensus, Immunotherapy, Societies, Medical, Immune Checkpoint Inhibitors therapeutic use, Neoplasms drug therapy
- Abstract
Immunotherapy is the standard of care for several cancers and the field continues to advance at a rapid pace, with novel combinations leading to indications in an increasing number of disease settings. Durable responses and long-term survival with immunotherapy have been demonstrated in some patients, though lack of initial benefit and recurrence after extended disease control remain major hurdles for the field. Many new combination regimens are in development for patients whose disease progressed on initial immunotherapy. To guide clinical trial design and support analyses of emerging molecular and cellular data surrounding mechanisms of resistance, the Society for Immunotherapy of Cancer (SITC) previously generated consensus clinical definitions for resistance to single-agent anti-PD-1 immune checkpoint inhibitors (ICIs) in three distinct scenarios: primary resistance, secondary resistance, and progression after treatment discontinuation. An unmet need still exists, however, for definitions of resistance to ICI-based combinations, which represent an expanding frontier in the immunotherapy treatment landscape. In 2021, SITC convened a workshop including stakeholders from academia, industry, and government to develop consensus definitions for resistance to ICI-based combination regimens for improved outcome assessment, trial design and drug development. This manuscript reports the minimum drug exposure requirements and time frame for progression that define resistance in both the metastatic setting and the perioperative setting, as well as key caveats and areas for future research with ICI/ICI combinations. Definitions for resistance to ICIs in combination with chemotherapy and targeted therapy will be published in companion volumes to this paper., Competing Interests: Competing interests: HK—consulting fees: Iovance, Immunocore, Celldex, Array Biopharma, Merck, Elevate Bio, Instil Bio, Bristol Myers Squibb, Clinigen, Shionogi, Chemocentryx, Calithera, Signatero. JCB—salary and employment: AstraZeneca; Ownership interest less than 5%: AstraZeneca. JFG—consulting fees: Bristol-Myers Squibb, Genentech/Roche, Takeda, Loxo/Lilly, Blueprint, Oncorus, Regeneron, Gilead, Mirati, Moderna, AstraZeneca, Pfizer, Novartis, Merck, iTeos, Karyopharm, Jazz Pharmaceuticals, Silverback Therapeutics, and GlydeBio; Contracted research: Novartis, Genentech/Roche, Takeda, Bristol-Myers Squibb, Tesaro, Moderna, Blueprint, Jounce, Array Biopharma, Merck, Adaptimmune, Novartis, and Alexo; Ownership interest less than 5%: Ironwood Pharmaceuticals; Spousal employment: Ironwood Pharmaceuticals. MH—consulting fees: Bristol Myers Squibb, CRISPR Therapeutics, Exelixis, Nektar Therapeutics, Janssen; Spousal employment: Arvinas. OH—consulting fees: Aduro, Akeso, Amgen, Beigene, Bioatla, Bristol Myers Squibb, Genentech/Roche, GSK, Immunocore, Idera, Incyte, Iovance, Instil Bio, Janssen, Merck, Nextcure, Novartis, Pfizer, Sanofi Regeneron, SeaGen, Tempus, Zelluna; Fees for non-CME services: Bristol Myers Squibb, Novartis, Pfizer, Sanofi Regeneron; Contracted research: Arcus, Aduro, Akeso, Amgen, Bioatla, Bristol Myers Squibb, Cytomx, Exelixis, Genentech/Roche, GSK, Immunocore, Idera, Incyte, Iovance, Merck, Moderna, Merck Serono, NextCure, Novartis, Pfizer, Rubius, Sanofi Regeneron, SeaGen, Torque, Zelluna. RAM—salary and employment: Bristol Myers Squibb; Ownership interest less than 5%: Bristol Myers Squibb. RZ—IP rights: Memorial Sloan Kettering, Weill Cornell Medical College; Consulting fees: Leap Therapeutics, iTEOS. TL—Salary and employment: Coherus Biosciences; IP rights: AstraZeneca, Parker Institute for Cancer Immunotherapy, Celldex, EntreMed; Consulting fees: TRex Bio, Grey Wolf Therapeutics, Exosis, LisCure Biosciences, BiOne Cure, Inovio, 1440 Foundation; Ownership interest less than 5%: AstraZeneca, Coherus Biosciences. RJS—consulting fees: Asana Biosciences, AstraZeneca, Bristol Myers Squibb, Eisai, Iovance, Merck, Novartis, OncoSec, Pfizer, Replimune; Contracted research: Merck, Amgen. HT—consulting fees: Genentech/Roche, Bristol Myers Squibb, Novartis, Merck, Pfizer, Eisai, Karyopharm, Boxer Capital; Contracted research: Genentech/Roche, Bristol Myers Squibb, Novartis, Merck, GSK. ES—nothing to disclose. SITC Staff: SMW, CG, PJI—nothing to disclose., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
29. Facts and Perspectives: Implications of tumor glycolysis on immunotherapy response in triple negative breast cancer.
- Author
-
Schreier A, Zappasodi R, Serganova I, Brown KA, Demaria S, and Andreopoulou E
- 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., Competing Interests: RZ is an inventor on patent applications related to work on GITR, PD-1, and CTLA-4 (patent numbers: US20180244793A1; US10323091B2; WO2018106864A1; WO2019094352A1). RZ is a scientific advisory board member for iTEOS Belgium SA. SD has received compensation for consultancy/advisory services from Lytix Biopharma, EMD Serono, Ono Pharmaceutical, and Genentech, and research support from Lytix Biopharma and Boehringer-Ingelheim for unrelated projects. AS, IS, KB, and EA do not have any conflicts of interest to declare., (Copyright © 2023 Schreier, Zappasodi, Serganova, Brown, Demaria and Andreopoulou.)
- Published
- 2023
- Full Text
- View/download PDF
30. Editorial: Factors determining long term anti-tumor responses to immune checkpoint blockade therapy.
- Author
-
Zappasodi R, Cook GP, and Taylor A
- Subjects
- Humans, Radioimmunotherapy, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Neoplasms drug therapy
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
31. Increased p53 expression induced by APR-246 reprograms tumor-associated macrophages to augment immune checkpoint blockade.
- Author
-
Ghosh A, Michels J, Mezzadra R, Venkatesh D, Dong L, Gomez R, Samaan F, Ho YJ, Campesato LF, Mangarin L, Fak J, Suek N, Holland A, Liu C, Abu-Akeel M, Bykov Y, Zhong H, Fitzgerald K, Budhu S, Chow A, Zappasodi R, Panageas KS, de Henau O, Ruscetti M, Lowe SW, Merghoub T, and Wolchok JD
- Subjects
- Animals, Mice, Quinuclidines, Tumor Microenvironment, Tumor Suppressor Protein p53 genetics, Immune Checkpoint Inhibitors pharmacology, Tumor-Associated Macrophages
- Abstract
In addition to playing a major role in tumor cell biology, p53 generates a microenvironment that promotes antitumor immune surveillance via tumor-associated macrophages. We examined whether increasing p53 signaling in the tumor microenvironment influences antitumor T cell immunity. Our findings indicate that increased p53 signaling induced either pharmacologically with APR-246 (eprenetapopt) or in p53-overexpressing transgenic mice can disinhibit antitumor T cell immunity and augment the efficacy of immune checkpoint blockade. We demonstrated that increased p53 expression in tumor-associated macrophages induces canonical p53-associated functions such as senescence and activation of a p53-dependent senescence-associated secretory phenotype. This was linked with decreased expression of proteins associated with M2 polarization by tumor-associated macrophages. Our preclinical data led to the development of a clinical trial in patients with solid tumors combining APR-246 with pembrolizumab. Biospecimens from select patients participating in this ongoing trial showed that there was a suppression of M2-polarized myeloid cells and increase in T cell proliferation with therapy in those who responded to the therapy. Our findings, based on both genetic and a small molecule-based pharmacological approach, suggest that increasing p53 expression in tumor-associated macrophages reprograms the tumor microenvironment to augment the response to immune checkpoint blockade.
- Published
- 2022
- Full Text
- View/download PDF
32. Phase IB Study of GITR Agonist Antibody TRX518 Singly and in Combination with Gemcitabine, Pembrolizumab, or Nivolumab in Patients with Advanced Solid Tumors.
- Author
-
Davar D, Zappasodi R, Wang H, Naik GS, Sato T, Bauer T, Bajor D, Rixe O, Newman W, Qi J, Holland A, Wong P, Sifferlen L, Piper D, Sirard CA, Merghoub T, Wolchok JD, and Luke JJ
- Subjects
- Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal, Humanized administration & dosage, Deoxycytidine analogs & derivatives, Humans, Nivolumab administration & dosage, Gemcitabine, Antineoplastic Agents therapeutic use, Neoplasms pathology
- Abstract
Purpose: TRX518 is a mAb engaging the glucocorticoid-induced TNF receptor-related protein (GITR). This open-label, phase I study (TRX518-003) evaluated the safety and efficacy of repeated dose TRX518 monotherapy and in combination with gemcitabine, pembrolizumab, or nivolumab in advanced solid tumors., Patients and Methods: TRX518 monotherapy was dose escalated (Part A) and expanded (Part B) up to 4 mg/kg loading, 1 mg/kg every 3 weeks. Parts C-E included dose-escalation (2 and 4 mg/kg loading followed by 1 mg/kg) and dose-expansion (4 mg/kg loading) phases with gemcitabine (Part C), pembrolizumab (Part D), or nivolumab (Part E). Primary endpoints included incidence of dose-limiting toxicities (DLT), serious adverse events (SAE), and pharmacokinetics. Secondary endpoints were efficacy and pharmacodynamics., Results: A total of 109 patients received TRX518: 43 (Parts A+B), 30 (Part C), 26 (Part D), and 10 (Part E), respectively. A total of 67% of patients in Parts D+E had received prior anti-PD(L)1 or anti-CTLA-4. No DLTs, treatment-related SAEs, and/or grade 4 or 5 AEs were observed with TRX518 monotherapy. In Parts C-E, no DLTs were observed, although TRX518-related SAEs were reported in 3.3% (Part C) and 10.0% (Part E), respectively. Objective response rate was 3.2%, 3.8%, 4%, and 12.5% in Parts A+B, C, D, and E, respectively. TRX518 affected peripheral and intratumoral regulatory T cells (Treg) with different kinetics depending on the combination regimen. Responses with TRX518 monotherapy+anti-PD1 combination were associated with intratumoral Treg reductions and CD8 increases and activation after treatment., Conclusions: TRX518 showed an acceptable safety profile with pharmacodynamic activity. Repeated dose TRX518 monotherapy and in combination resulted in limited clinical responses associated with immune activation. See related commentary by Hernandez-Guerrero and Moreno, p. 3905., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
33. Calreticulin mutant myeloproliferative neoplasms induce MHC-I skewing, which can be overcome by an optimized peptide cancer vaccine.
- Author
-
Gigoux M, Holmström MO, Zappasodi R, Park JJ, Pourpe S, Bozkus CC, Mangarin LMB, Redmond D, Verma S, Schad S, George MM, Venkatesh D, Ghosh A, Hoyos D, Molvi Z, Kamaz B, Marneth AE, Duke W, Leventhal MJ, Jan M, Ho VT, Hobbs GS, Knudsen TA, Skov V, Kjær L, Larsen TS, Hansen DL, Lindsley RC, Hasselbalch H, Grauslund JH, Lisle TL, Met Ö, Wilkinson P, Greenbaum B, Sepulveda MA, Chan T, Rampal R, Andersen MH, Abdel-Wahab O, Bhardwaj N, Wolchok JD, Mullally A, and Merghoub T
- Subjects
- Animals, Calreticulin genetics, Humans, Janus Kinase 2 genetics, Major Histocompatibility Complex, Mice, Mice, Inbred C57BL, Mutation genetics, Peptides, Vaccines, Subunit, Cancer Vaccines, Myeloproliferative Disorders genetics, Neoplasms genetics
- 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.- Published
- 2022
- Full Text
- View/download PDF
34. Tumor-induced double positive T cells display distinct lineage commitment mechanisms and functions.
- Author
-
Schad SE, Chow A, Mangarin L, Pan H, Zhang J, Ceglia N, Caushi JX, Malandro N, Zappasodi R, Gigoux M, Hirschhorn D, Budhu S, Amisaki M, Arniella M, Redmond D, Chaft J, Forde PM, Gainor JF, Hellmann MD, Balachandran V, Shah S, Smith KN, Pardoll D, Elemento O, Wolchok JD, and Merghoub T
- Subjects
- Animals, CD4 Antigens metabolism, CD8 Antigens metabolism, CD8-Positive T-Lymphocytes, Cell Differentiation, Cell Lineage genetics, Mice, T-Lymphocyte Subsets, CD4-Positive T-Lymphocytes, Melanoma metabolism
- Abstract
Transcription factors ThPOK and Runx3 regulate the differentiation of "helper" CD4+ and "cytotoxic" CD8+ T cell lineages respectively, inducing single positive (SP) T cells that enter the periphery with the expression of either the CD4 or CD8 co-receptor. Despite the expectation that these cell fates are mutually exclusive and that mature CD4+CD8+ double positive (DP) T cells are present in healthy individuals and augmented in the context of disease, yet their molecular features and pathophysiologic role are disputed. Here, we show DP T cells in murine and human tumors as a heterogenous population originating from SP T cells which re-express the opposite co-receptor and acquire features of the opposite cell type's phenotype and function following TCR stimulation. We identified distinct clonally expanded DP T cells in human melanoma and lung cancer by scRNA sequencing and demonstrated their tumor reactivity in cytotoxicity assays. Our findings indicate that antigen stimulation induces SP T cells to differentiate into DP T cell subsets gaining in polyfunctional characteristics., (© 2022 Schad et al.)
- Published
- 2022
- Full Text
- View/download PDF
35. Author Correction: Fundamental immune-oncogenicity trade-offs define driver mutation fitness.
- Author
-
Hoyos D, Zappasodi R, Schulze I, Sethna Z, de Andrade KC, Bajorin DF, Bandlamudi C, Callahan MK, Funt SA, Hadrup SR, Holm JS, Rosenberg JE, Shah SP, Vázquez-García I, Weigelt B, Wu M, Zamarin D, Campitelli LF, Osborne EJ, Klinger M, Robins HS, Khincha PP, Savage SA, Balachandran VP, Wolchok JD, Hellmann MD, Merghoub T, Levine AJ, Łuksza M, and Greenbaum BD
- Published
- 2022
- Full Text
- View/download PDF
36. Fundamental immune-oncogenicity trade-offs define driver mutation fitness.
- Author
-
Hoyos D, Zappasodi R, Schulze I, Sethna Z, de Andrade KC, Bajorin DF, Bandlamudi C, Callahan MK, Funt SA, Hadrup SR, Holm JS, Rosenberg JE, Shah SP, Vázquez-García I, Weigelt B, Wu M, Zamarin D, Campitelli LF, Osborne EJ, Klinger M, Robins HS, Khincha PP, Savage SA, Balachandran VP, Wolchok JD, Hellmann MD, Merghoub T, Levine AJ, Łuksza M, and Greenbaum BD
- Subjects
- Datasets as Topic, Genes, p53, Genetic Fitness, Genomics, Healthy Volunteers, Humans, Immunotherapy, Mutation, Missense, Reproducibility of Results, Carcinogenesis genetics, Carcinogenesis immunology, Evolution, Molecular, Lung Neoplasms genetics, Lung Neoplasms therapy, Mutation genetics
- Abstract
Missense driver mutations in cancer are concentrated in a few hotspots
1 . 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., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
37. MAIT and Vδ2 unconventional T cells are supported by a diverse intestinal microbiome and correlate with favorable patient outcome after allogeneic HCT.
- Author
-
Andrlová H, Miltiadous O, Kousa AI, Dai A, DeWolf S, Violante S, Park HY, Janaki-Raman S, Gardner R, El Daker S, Slingerland J, Giardina P, Clurman A, Gomes ALC, Nguyen C, da Silva MB, Armijo GK, Lee N, Zappasodi R, Chaligne R, Masilionis I, Fontana E, Ponce D, Cho C, Bush A, Hill L, Chao N, Sung AD, Giralt S, Vidal EH, Hosszu KK, Devlin SM, Peled JU, Cross JR, Perales MA, Godfrey DI, van den Brink MRM, and Markey KA
- Subjects
- Humans, Ligands, Gastrointestinal Microbiome, Graft vs Host Disease, Hematopoietic Stem Cell Transplantation, Mucosal-Associated Invariant T Cells
- 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.
- Published
- 2022
- Full Text
- View/download PDF
38. A decade of checkpoint blockade immunotherapy in melanoma: understanding the molecular basis for immune sensitivity and resistance.
- Author
-
Huang AC and Zappasodi R
- Subjects
- Humans, Immunotherapy, Ipilimumab therapeutic use, Tumor Microenvironment, Immune Checkpoint Inhibitors, Melanoma drug therapy
- Abstract
Ten years since the immune checkpoint inhibitor ipilimumab was approved for advanced melanoma, it is time to reflect on the lessons learned regarding modulation of the immune system to treat cancer and on novel approaches to further extend the efficacy of current and emerging immunotherapies. Here, we review the studies that led to our current understanding of the melanoma immune microenvironment in humans and the mechanistic work supporting these observations. We discuss how this information is guiding more precise analyses of the mechanisms of action of immune checkpoint blockade and novel immunotherapeutic approaches. Lastly, we review emerging evidence supporting the negative impact of melanoma metabolic adaptation on anti-tumor immunity and discuss how to counteract such mechanisms for more successful use of immunotherapy., (© 2022. Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
39. Hallmarks of Resistance to Immune-Checkpoint Inhibitors.
- Author
-
Karasarides M, Cogdill AP, Robbins PB, Bowden M, Burton EM, Butterfield LH, Cesano A, Hammer C, Haymaker CL, Horak CE, McGee HM, Monette A, Rudqvist NP, Spencer CN, Sweis RF, Vincent BG, Wennerberg E, Yuan J, Zappasodi R, Lucey VMH, Wells DK, and LaVallee T
- Subjects
- Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Antineoplastic Agents, Immunological adverse effects, Neoplasms
- Abstract
Immune-checkpoint inhibitors (ICI), although revolutionary in improving long-term survival outcomes, are mostly effective in patients with immune-responsive tumors. Most patients with cancer either do not respond to ICIs at all or experience disease progression after an initial period of response. Treatment resistance to ICIs remains a major challenge and defines the biggest unmet medical need in oncology worldwide. In a collaborative workshop, thought leaders from academic, biopharma, and nonprofit sectors convened to outline a resistance framework to support and guide future immune-resistance research. Here, we explore the initial part of our effort by collating seminal discoveries through the lens of known biological processes. We highlight eight biological processes and refer to them as immune resistance nodes. We examine the seminal discoveries that define each immune resistance node and pose critical questions, which, if answered, would greatly expand our notion of immune resistance. Ultimately, the expansion and application of this work calls for the integration of multiomic high-dimensional analyses from patient-level data to produce a map of resistance phenotypes that can be utilized to guide effective drug development and improved patient outcomes., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
40. Therapeutic antibody activation of the glucocorticoid-induced TNF receptor by a clustering mechanism.
- Author
-
He C, Maniyar RR, Avraham Y, Zappasodi R, Rusinova R, Newman W, Heath H, Wolchok JD, Dahan R, Merghoub T, and Meyerson JR
- Abstract
GITR is a TNF receptor, and its activation promotes immune responses and drives antitumor activity. The receptor is activated by the GITR ligand (GITRL), which is believed to cluster receptors into a high-order array. Immunotherapeutic agonist antibodies also activate the receptor, but their mechanisms are not well characterized. We solved the structure of full-length mouse GITR bound to Fabs from the antibody DTA-1. The receptor is a dimer, and each subunit binds one Fab in an orientation suggesting that the antibody clusters receptors. Binding experiments with purified proteins show that DTA-1 IgG and GITRL both drive extensive clustering of GITR. Functional data reveal that DTA-1 and the anti-human GITR antibody TRX518 activate GITR in their IgG forms but not as Fabs. Thus, the divalent character of the IgG agonists confers an ability to mimic GITRL and cluster and activate GITR. These findings will inform the clinical development of this class of antibodies for immuno-oncology.
- Published
- 2022
- Full Text
- View/download PDF
41. Epigenetic, Metabolic, and Immune Crosstalk in Germinal-Center-Derived B-Cell Lymphomas: Unveiling New Vulnerabilities for Rational Combination Therapies.
- Author
-
Serganova I, Chakraborty S, Yamshon S, Isshiki Y, Bucktrout R, Melnick A, Béguelin W, and Zappasodi R
- Abstract
B-cell non-Hodgkin lymphomas (B-NHLs) are highly heterogenous by genetic, phenotypic, and clinical appearance. Next-generation sequencing technologies and multi-dimensional data analyses have further refined the way these diseases can be more precisely classified by specific genomic, epigenomic, and transcriptomic characteristics. The molecular and genetic heterogeneity of B-NHLs may contribute to the poor outcome of some of these diseases, suggesting that more personalized precision-medicine approaches are needed for improved therapeutic efficacy. The germinal center (GC) B-cell like diffuse large B-cell lymphomas (GCB-DLBCLs) and follicular lymphomas (FLs) share specific epigenetic programs. These diseases often remain difficult to treat and surprisingly do not respond advanced immunotherapies, despite arising in secondary lymphoid organs at sites of antigen recognition. Epigenetic dysregulation is a hallmark of GCB-DLBCLs and FLs, with gain-of-function (GOF) mutations in the histone methyltransferase EZH2 , loss-of-function (LOF) mutations in histone acetyl transferases CREBBP and EP300 , and the histone methyltransferase KMT2D representing the most prevalent genetic lesions driving these diseases. These mutations have the common effect to disrupt the interactions between lymphoma cells and the immune microenvironment, via decreased antigen presentation and responsiveness to IFN-γ and CD40 signaling pathways. This indicates that immune evasion is a key step in GC B-cell lymphomagenesis. EZH2 inhibitors are now approved for the treatment of FL and selective HDAC3 inhibitors counteracting the effects of CREBBP LOF mutations are under development. These treatments can help restore the immune control of GCB lymphomas, and may represent optimal candidate agents for more effective combination with immunotherapies. Here, we review recent progress in understanding the impact of mutant chromatin modifiers on immune evasion in GCB lymphomas. We provide new insights on how the epigenetic program of these diseases may be regulated at the level of metabolism, discussing the role of metabolic intermediates as cofactors of epigenetic enzymes. In addition, lymphoma metabolic adaptation can negatively influence the immune microenvironment, further contributing to the development of immune cold tumors, poorly infiltrated by effector immune cells. Based on these findings, we discuss relevant candidate epigenetic/metabolic/immune targets for rational combination therapies to investigate as more effective precision-medicine approaches for GCB lymphomas., Competing Interests: AM has research funds from Janssen, Astra Zeneca, Epizyme, Daiichi Sankyo, and Sanofi, and has consulted for Astra Zeneca, Bristol Myers Squibb, Epizyme, Constellation, ExoTherapeutics, Daiichi Sankyo, and Janssen. W.B. is consulting for Eisai Co., Lrd. RZ is inventor on patent applications related to work on GITR, PD-1, and CTLA-4. RZ is scientific advisory board member of iTEOS Therapeutics, has consulted for Leap Therapeutics and receives grant support from Astra Zeneca and Bristol Myers Squibb.The other authors have no conflicts of interest to disclose., (Copyright © 2022 Serganova, Chakraborty, Yamshon, Isshiki, Bucktrout, Melnick, Béguelin and Zappasodi.)
- Published
- 2022
- Full Text
- View/download PDF
42. Supporting the Next Generation of Scientists to Lead Cancer Immunology Research.
- Author
-
Alspach E, Chow RD, Demehri S, Guerriero JL, Gujar S, Hartmann FJ, Helmink BA, Hudson WH, Ho WJ, Ma L, Maier BB, Maltez VI, Miller BC, Moran AE, Parry EM, Pillai PS, Rafiq S, Reina-Campos M, Rosato PC, Rudqvist NP, Ruhland MK, Sagiv-Barfi I, Sahu AD, Samstein RM, Schürch CM, Sen DR, Thommen DS, Wolf Y, and Zappasodi R
- Subjects
- Humans, Leadership, Allergy and Immunology education, Biomedical Research methods, Neoplasms epidemiology, Physicians organization & administration
- Abstract
Recent success in the use of immunotherapy for a broad range of cancers has propelled the field of cancer immunology to the forefront of cancer research. As more and more young investigators join the community of cancer immunologists, the Arthur L. Irving Family Foundation Cancer Immunology Symposium provided a platform to bring this expanding and vibrant community together and support the development of the future leaders in the field. This commentary outlines the lessons that emerged from the inaugural symposium highlighting the areas of scientific and career development that are essential for professional growth in the field of cancer immunology and beyond. Leading scientists and clinicians in the field provided their experience on the topics of scientific trajectory, career trajectory, publishing, fundraising, leadership, mentoring, and collaboration. Herein, we provide a conceptual and practical framework for career development to the broader scientific community., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
43. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8 + T cells in tumors.
- Author
-
Xu S, Chaudhary O, Rodríguez-Morales P, Sun X, Chen D, Zappasodi R, Xu Z, Pinto AFM, Williams A, Schulze I, Farsakoglu Y, Varanasi SK, Low JS, Tang W, Wang H, McDonald B, Tripple V, Downes M, Evans RM, Abumrad NA, Merghoub T, Wolchok JD, Shokhirev MN, Ho PC, Witztum JL, Emu B, Cui G, and Kaech SM
- Subjects
- Animals, Biological Transport physiology, Cell Line, Tumor, HEK293 Cells, Humans, Leukocytes, Mononuclear metabolism, Lymphocytes, Tumor-Infiltrating metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Tumor Microenvironment physiology, CD36 Antigens metabolism, CD8-Positive T-Lymphocytes metabolism, Lipid Peroxidation physiology, Lipoproteins, LDL metabolism, Neoplasms metabolism, Receptors, Scavenger metabolism
- Abstract
A common metabolic alteration in the tumor microenvironment (TME) is lipid accumulation, a feature associated with immune dysfunction. Here, we examined how CD8
+ tumor infiltrating lymphocytes (TILs) respond to lipids within the TME. We found elevated concentrations of several classes of lipids in the TME and accumulation of these in CD8+ TILs. Lipid accumulation was associated with increased expression of CD36, a scavenger receptor for oxidized lipids, on CD8+ TILs, which also correlated with progressive T cell dysfunction. Cd36-/- T cells retained effector functions in the TME, as compared to WT counterparts. Mechanistically, CD36 promoted uptake of oxidized low-density lipoproteins (OxLDL) into T cells, and this induced lipid peroxidation and downstream activation of p38 kinase. Inhibition of p38 restored effector T cell functions in vitro, and resolution of lipid peroxidation by overexpression of glutathione peroxidase 4 restored functionalities in CD8+ TILs in vivo. Thus, an oxidized lipid-CD36 axis promotes intratumoral CD8+ T cell dysfunction and serves as a therapeutic avenue for immunotherapies., Competing Interests: Declaration of interests G.C. receives research funding from Bayer AG and Boehringer Ingelheim, but the funding is not relevant to the current study. J.L.W. and X.S. are named inventors on patent applications or patents related to the use of oxidation-specific antibodies held by UCSD. R.Z. is an inventor on patent applications related to work on GITR, PD-1, and CTLA-4. R.Z. is a consultant for Leap Therapeutics and iTEOS. T.M. is a cofounder and holds equity in IMVAQ Therapeutics. T.M. is a consultant for Immunos Therapeutics, Pfizer, and Immunogenesis. T.M. has research support from Bristol-Myers Squibb; Surface Oncology; Kyn Therapeutics; Infinity Pharmaceuticals, Inc.; Peregrine Pharmaceuticals, Inc.; Adaptive Biotechnologies; Leap Therapeutics, Inc.; and Aprea. T.M. has patents on applications related to work on oncolytic viral therapy, alpha virus-based vaccines, neoantigen modeling, CD40, GITR, OX40, PD-1, and CTLA-4. J.D.W. is a consultant for Adaptive Biotech, Amgen, Apricity, Ascentage Pharma, Astellas, AstraZeneca, Bayer, Beigene, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Chugai, Elucida, Eli Lilly, F Star, Georgiamune, Imvaq, Kyowa Hakko Kirin, Linneaus, Merck Pharmaceuticals, Neon Therapeutics, Polynoma, Psioxus, Recepta, Takara Bio, Trieza, Truvax, Sellas Life Sciences, Serametrix, Surface Oncology, Syndax, Syntalogic, and Werewolf Therapeutics. J.D.W. reports grants from Bristol Myers Squibb and Sephora. J.D.W. has equity in Tizona Pharmaceuticals, Adaptive Biotechnologies, Imvaq, Beigene, Linneaus, Apricity, Arsenal IO, and Georgiamune. J.D.W. is an inventor on patent applications related to work on DNA vaccines in companion animals with cancer, assays for suppressive myeloid cells in blood, oncolytic viral therapy, alphavirus-based vaccines, neo-antigen modeling, CD40, GITR, OX40, PD-1, and CTLA-4., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
44. Fifteen-year follow-up of relapsed indolent non-Hodgkin lymphoma patients vaccinated with tumor-loaded dendritic cells.
- Author
-
Fucà G, Ambrosini M, Agnelli L, Brich S, Sgambelluri F, Mortarini R, Pupa SM, Magni M, Devizzi L, Matteucci P, Cabras A, Zappasodi R, De Santis F, Anichini A, De Braud F, Gianni AM, and Di Nicola M
- Subjects
- Cancer Vaccines pharmacology, Female, Follow-Up Studies, Humans, Male, Neoplasm Recurrence, Local, Recurrence, Time Factors, Cancer Vaccines therapeutic use, Dendritic Cells transplantation, Immunotherapy methods, Lymphoma, Non-Hodgkin therapy
- Abstract
We previously published the results of a pilot study showing that vaccination with tumor-loaded dendritic cells (DCs) induced both T and B cell response and produced clinical benefit in the absence of toxicity in patients with relapsed, indolent non-Hodgkin lymphoma (iNHL). The purpose of the present short report is to provide a 15-year follow-up of our study and to expand the biomarker analysis previously performed. The long-term follow-up highlighted the absence of particular or delayed toxicity and the benefit of active immunization with DCs loaded with autologous, heat-shocked and UV-C treated tumor cells in relapsed iNHL (5-year and 10-year progression-free survival (PFS) rates: 55.6% and 33.3%, respectively; 10-year overall survival (OS) rate: 83.3%). Female patients experienced a better PFS (p=0.016) and a trend towards a better OS (p=0.185) compared with male patients. Of note, we observed a non-negligible fraction of patients (22%) who experienced a long-lasting complete response. In a targeted gene expression profiling of pre-treatment tumor biopsies in 11 patients with available formalin-fixed, paraffin-embedded tissue, we observed that KIT , ATG12 , TNFRSF10C , PBK , ITGA2 , GATA3 , CLU , NCAM1 , SYT17 and LTK were differentially expressed in patients with responder versus non-responder tumors. The characterization of peripheral monocytic cells in a subgroup of 14 patients with available baseline blood samples showed a higher frequency of the subset of CD14
++ CD16+ cells (intermediate monocytes) in patients with responding tumors. Since in patients with relapsed iNHL the available therapeutic options are often incapable of inducing a long-lasting complete remission and can be sometimes characterized by intolerable toxicity, we think that the encouraging results of our long-term follow-up analysis represent a stimulus to further investigate the role of active vaccination in this specific setting and in earlier lines of therapy and to explore novel combinatorial strategies encompassing other innovative immunotherapy agents, such as immune-checkpoint inhibitors., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
45. To Go or Not to Go?-Targeting Tregs Traveling in Tumors.
- Author
-
Chakraborty S and Zappasodi R
- Subjects
- Animals, Humans, Mice, Receptors, G-Protein-Coupled, Neoplasms therapy
- Abstract
Regulatory T cells (Treg) are one of the major impediments to effective antitumor immunity and successful immunotherapy. Elevated intratumoral Treg frequencies, observed in a variety of malignancies, have been associated with poor prognosis. In this issue of Cancer Research , two studies underscore the potential of harnessing the unique migratory profile of tumor-infiltrating Tregs to selectively eliminate these cells without compromising peripheral tolerance. Both studies identify surface migratory receptors, CCR8 by Campbell and colleagues and GPR15 by Adamczyk and colleagues, as selective markers of intratumoral Tregs in tumor-bearing mice and patients with cancer. Genetic deletion of GPR15 or antibody-mediated depletion of CCR8 was found to preferentially decrease tumor-infiltrating Tregs and substantially delayed tumor progression. Together, these two studies highlight the significance of migratory molecules in intratumoral Tregs and propose two potential selective targets for preferential elimination of tumor-associated "pathogenic" Tregs, which can be hijacked to enhance the response to immunotherapy. See related articles by Adamczyk et al., p. 2970 and Campbell et al., p. 2983 ., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
46. CTLA-4 blockade drives loss of T reg stability in glycolysis-low tumours.
- Author
-
Zappasodi R, Serganova I, Cohen IJ, Maeda M, Shindo M, Senbabaoglu Y, Watson MJ, Leftin A, Maniyar R, Verma S, Lubin M, Ko M, Mane MM, Zhong H, Liu C, Ghosh A, Abu-Akeel M, Ackerstaff E, Koutcher JA, Ho PC, Delgoffe GM, Blasberg R, Wolchok JD, and Merghoub T
- Subjects
- Animals, Breast Neoplasms immunology, Breast Neoplasms metabolism, Cell Line, Tumor, Female, Humans, Melanoma genetics, Melanoma immunology, Melanoma metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, CTLA-4 Antigen antagonists & inhibitors, Glycolysis, Neoplasms immunology, Neoplasms metabolism, T-Lymphocytes, Regulatory immunology
- 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 cells
1 . 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.- Published
- 2021
- Full Text
- View/download PDF
47. Targeting Phosphatidylserine Enhances the Anti-tumor Response to Tumor-Directed Radiation Therapy in a Preclinical Model of Melanoma.
- Author
-
Budhu S, Giese R, Gupta A, Fitzgerald K, Zappasodi R, Schad S, Hirschhorn D, Campesato LF, De Henau O, Gigoux M, Liu C, Mazo G, Deng L, Barker CA, Wolchok JD, and Merghoub T
- Subjects
- Animals, Disease Models, Animal, Humans, Melanoma pathology, Mice, Tumor Microenvironment, Melanoma radiotherapy, Phosphatidylserines metabolism
- 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., Competing Interests: Declaration of Interests All authors concur with the submission of this manuscript and have no financial or other interests related to the submitted work. T.M. is a cofounder and holds an equity in IMVAQ Therapeutics. He is a consultant of Immunos Therapeutics and Pfizer. He has research support from Bristol Myers Squibb; Surface Oncology; Kyn Therapeutics; Infinity Pharmaceuticals, Inc.; Peregrine Pharmaceuticals, Inc.; Adaptive Biotechnologies; Leap Therapeutics, Inc.; and Aprea. He has patents on applications related to work on oncolytic viral therapy, alpha virus-based vaccine, neoantigen modeling, CD40, GITR, OX40, PD-1, and CTLA-4. J.D.W. is a consultant for Adaptive Biotech, Advaxis, Amgen, Apricity, Array BioPharma, Ascentage Pharma, Astellas, Bayer, Beigene, Bristol Myers Squibb, Celgene, Chugai, Elucida, Eli Lilly, F Star, Genentech, Imvaq, Janssen, Kleo Pharma, Linnaeus, MedImmune, Merck, Neon Therapeutics, Ono, Polaris Pharma, Polynoma, Psioxus, Puretech, Recepta, Trieza, Sellas Life Sciences, Serametrix, Surface Oncology, and Syndax. Research support: Bristol Myers Squibb, Medimmune, Merck Pharmaceuticals, and Genentech. Equity: Potenza Therapeutics, Tizona Pharmaceuticals, Adaptive Biotechnologies, Elucida, Imvaq, Beigene, Trieza, and Linnaeus. Honorarium: Esanex. Patents: xenogeneic DNA vaccines, alphavirus replicon particles expressing TRP2, MDSC assay, Newcastle disease viruses for cancer therapy, genomic signature to identify responders to ipilimumab in melanoma, engineered vaccinia viruses for cancer immunotherapy, anti-CD40 agonist monoclonal antibody (mAb) fused to monophosphoryl lipid A (MPL) for cancer therapy, CAR+ T cells targeting differentiation antigens as means to treat cancer, anti-PD-1 antibody, anti-CTLA-4 antibodies, and anti-GITR antibodies and methods of use thereof. L.D. is a cofounder and holds equity in IMVAQ Therapeutics. She has patents on applications related to work on oncolytic viral therapy. R.Z. is inventor on patent applications related to work on GITR, PD-1, and CTLA-4. R.Z. is consultant for Leap Therapeutics and iTEOS Belgium SA. C.A.B. is a consultant of Regeneron. He has research support from Amgen and Merck., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
48. Silibinin down-regulates PD-L1 expression in nasopharyngeal carcinoma by interfering with tumor cell glycolytic metabolism.
- Author
-
Sellam LS, Zappasodi R, Chettibi F, Djennaoui D, Yahi-Ait Mesbah N, Amir-Tidadini ZC, Touil-Boukoffa C, Ouahioune W, Merghoub T, and Bourouba M
- Subjects
- Adolescent, Adult, Antineoplastic Agents, Phytogenic pharmacology, B7-H1 Antigen metabolism, Biopsy, Cell Line, Tumor, Cell Proliferation drug effects, Citric Acid Cycle, Down-Regulation drug effects, Drug Discovery, Gene Expression Regulation, Neoplastic drug effects, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Lactate Dehydrogenase 5 metabolism, Middle Aged, Oxidative Phosphorylation, Signal Transduction, Silybin pharmacology, Antineoplastic Agents, Phytogenic metabolism, B7-H1 Antigen genetics, Glycolysis drug effects, Nasopharyngeal Carcinoma drug therapy, Nasopharyngeal Neoplasms drug therapy, Silybin metabolism
- Abstract
The upregulation of checkpoint inhibitor PD-L1 expression has recently been associated with nasopharyngeal carcinoma (NPC) resistance to therapy. The mechanism of induction of PD-L1 has also been linked to enhanced aerobic glycolysis promoted by HIF1-α dysregulation and LDH-A activity in cancer. Here, we investigated the effect of the anti-tumoral compound Silibinin on HIF-1α/LDH-A mediated cancer cell metabolism and PD-L1 expression in NPC. Our results demonstrate that exposure to Silibinin potently inhibits tumor growth and promotes a shift from aerobic glycolysis toward oxidative phosphorylation. The EBV + NPC cell line C666-1 and glycolytic human tumor explants treated with Silibinin displayed a reduction in LDH-A activity which consistently associated with a reduction in lactate levels. This effect was accompanied by an increase in intracellular citrate levels in C666-1 cells. Accordingly, expression of HIF-1α, a critical regulator of glycolysis, was down-regulated after treatment. This event associated with a down-regulation in PD-L1. Altogether, our results provide evidence that silibinin can alter PD-L1 expression by interfering with HIF-1α/LDH-A mediated cell metabolism in NPC. These results provide a new perspective for Silibinin use to overcome PD-L1 mediated NPC resistance to therapy., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
49. Blockade of the AHR restricts a Treg-macrophage suppressive axis induced by L-Kynurenine.
- Author
-
Campesato LF, Budhu S, Tchaicha J, Weng CH, Gigoux M, Cohen IJ, Redmond D, Mangarin L, Pourpe S, Liu C, Zappasodi R, Zamarin D, Cavanaugh J, Castro AC, Manfredi MG, McGovern K, Merghoub T, and Wolchok JD
- Subjects
- Animals, Drug Resistance, Neoplasm, Humans, Immune Tolerance, Immunotherapy, Indoleamine-Pyrrole 2,3,-Dioxygenase genetics, Indoleamine-Pyrrole 2,3,-Dioxygenase metabolism, Mice, Neoplasms immunology, Neoplasms therapy, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor immunology, Receptors, Aryl Hydrocarbon genetics, Receptors, Aryl Hydrocarbon metabolism, Signal Transduction, Tryptophan Oxygenase genetics, Tryptophan Oxygenase metabolism, Tumor Cells, Cultured, Tumor Microenvironment, Kynurenine immunology, Macrophages immunology, Receptors, Aryl Hydrocarbon antagonists & inhibitors, T-Lymphocytes, Regulatory immunology
- 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.- Published
- 2020
- Full Text
- View/download PDF
50. Defining tumor resistance to PD-1 pathway blockade: recommendations from the first meeting of the SITC Immunotherapy Resistance Taskforce.
- Author
-
Kluger HM, Tawbi HA, Ascierto ML, Bowden M, Callahan MK, Cha E, Chen HX, Drake CG, Feltquate DM, Ferris RL, Gulley JL, Gupta S, Humphrey RW, LaVallee TM, Le DT, Hubbard-Lucey VM, Papadimitrakopoulou VA, Postow MA, Rubin EH, Sharon E, Taube JM, Topalian SL, Zappasodi R, Sznol M, and Sullivan RJ
- Subjects
- Biomarkers, Tumor, Female, Humans, Male, Neoplasms immunology, Neoplasms therapy, Immunotherapy methods, Programmed Cell Death 1 Receptor antagonists & inhibitors
- Abstract
As the field of cancer immunotherapy continues to advance at a fast pace, treatment approaches and drug development are evolving rapidly to maximize patient benefit. New agents are commonly evaluated for activity in patients who had previously received a programmed death receptor 1 (PD-1)/programmed death-ligand 1 (PD-L1) inhibitor as standard of care or in an investigational study. However, because of the kinetics and patterns of response to PD-1/PD-L1 blockade, and the lack of consistency in the clinical definitions of resistance to therapy, the design of clinical trials of new agents and interpretation of results remains an important challenge. To address this unmet need, the Society for Immunotherapy of Cancer convened a multistakeholder taskforce-consisting of experts in cancer immunotherapy from academia, industry, and government-to generate consensus clinical definitions for resistance to PD-(L)1 inhibitors in three distinct scenarios: primary resistance, secondary resistance, and progression after treatment discontinuation. The taskforce generated consensus on several key issues such as the timeframes that delineate each type of resistance, the necessity for confirmatory scans, and identified caveats for each specific resistance classification. The goal of this effort is to provide guidance for clinical trial design and to support analyses of emerging molecular and cellular data surrounding mechanisms of resistance., Competing Interests: Competing interests: HMK has served as a consultant for Corvus, Nektar, Biodesix, Genentech, Pfizer, Merck & Co., Immunocore, Array Biopharma and Celldex, and has received research support from Merck & Co., Apexigen and Bristol-Myers Squibb. HAT has served as a consultant or an advisory board member for Bristol-Myers Squibb, Novartis, Merck & Co., Genentech, and Array, and has received commercial research grants from Bristol-Myers Squibb, Merck & Co., Genentech, GlaxoSmithKline and Celgene. MLA is a full-time employee of AstraZeneca/MedImmune. MB is a full-time employee of Bristol-Myers Squibb. MKC has served as a consultant and/or advisory board member for AstraZeneca/MedImmune, Incyte, Moderna and Merck &. Co. She has also received research grant funding from Bristol-Myers Squibb, and reports that a family member is currently employeed by Bristol-Myers Squibb. EC is an employee of and has stock in Roche Genentech. CGD has received research funding Aduro Biotech, Bristol-Myers Squibb, Janssen, royalties from Bristol-Myers Squibb, AstraZeneca, and Janssen, has served as a consultant for Agenus, Dendreon, Janssen, Eli Lilly, Merck & Co., Medimmune, Pierre Fabre, and Roche Genentech, and has ownership interest in Compugen, Harpoon, and Kleo. DMF is a full-time employee of Bristol-Myers Squibb. RLF has served as a consultant for Aduro Biotech, Bain Capital Life Sciences, Bristol-Myers Squibb, Iovance Biotherapeutics, Nanobiotix, Ono Pharmaceutical CO. Ltd, Torque Therapeutics, and TTMS, has served on an advisory board for Amgen, Bristol-Myers Squibb, EMD Serono, GlaxoSmithKline, Eli Lilly, Merck & Co., Numab Therapeutics AG, Oncorus, Pfizerv, PPD (Benitec, Immunicum), Regeneron Pharmaceuticals, and Tesaro, has conducted clinical trials in collaboration with AstraZeneca/MedImmune, Bristol-Myers Squibb, Merck & Co., and has received research funding from AstraZeneca/MedImmune, Bristol-Myers Squibb, Tesaro, TTMS, and VentriRx Pharmaaceuticals. SG has served as a consultant and/or an advisory board member for Exelixis, Janssen Biotech, and AstraZeneca. RWH is a full-time employee and stockholder of CytomX Therapeutics. TML holds stock in AstraZeneca, and has institutional support from Bristol-Myers Squibb. DTL has served on an advisory board for Merck & Co. and Bristol-Myers Squibb, has received research funding from Merck & Co., Bristol-Myers Squibb, Aduro Biotech, Curegenix, and Medivir, has received speaking honoraria from Merck & Co., and is an inventor of licensed intellectual property related to technology for mismatch repair deficiency for diagnosis and therapy (WO2016077553A1) from Johns Hopkins University. The terms of these arrangements are being managed by Johns Hopkins. VML has served on an advisory board for FXBiopharma. VAP has received research funding from Checkmate and Incyte, has received personal fees from LOXO Oncology, Araxes Pharma, Takeda, AbbVie, Tesaro, Exelixis, has received both grants and personal fees from Nektar Therapeutics, AstraZeneca, Eli Lilly, Roche, Merck & Co., Bristol-Myers Squibb, Novartis, Janssen, Checkmate, Incyte, and has served on an advisory board for Arrys Therapeutics. MAP has received personal fees from Merck & Co., Bristol-Myers Squibb, Novartis, Array BioPharma, Aduro, Incytem NewLink Genetics, has received non-financial support from Merck & Co., Bristol-Myers Squibb, RGenix, Infinity, AstraZeneca, Novartis, and Array Bio Pharma. ER is an employee of Merck & Co. JMT has received research funding from Bristol-Myers Squibb, has served on an advisory board for Bristol-Myers Squibb, Merck & Co., AstraZeneca, and Amgen. SLT reports grants and non-financial support from Bristol-Myers Squibb; personal fees from AbbVie, ImaginAb, Immunocore, Avidity NanoMedicines LLC, and Merck; and personal fees and non-financial support from Five Prime Therapeutics and Dragonfly Therapeutics, outside the submitted work. In addition, SLT has patents pending. SLT’s spouse has financial relationships with the following entities: Aduro, Amgen, Bayer, Camden Nexus, Compugen, DNAtrix, Dynavax Technolgies, Ervaxx, FLX Bio, Immunomic Therapeutics, Janssen Pharmaceuticals, Jounce Therapeutics, MedImmune/AstraZeneca, Pfizer, Potenza Therapeutics, Rock Springs Capital, Tizona LLC, Trieza Therapeutics, and WindMIL. RZ is the inventor on patent applications related to work on GITR, PD-1 and CTLA-4, and consultant for Leap Therapeutics. MS has received consulting fees from Roche Genentech, Bristol-Myers Squibb, AstraZeneca/Medimmune, Novartis, Seattle Genetics, Nektar Therapeutics, Eli Lilly, Biodesix, Modulate Therapeutics, Newlink Genetics, Molecular Partners, Innate Pharma, Abbvie, Immunocore, Genmab, Almac, Hinge, Allakos, Anaeropharma, and Array, has served on an advisory board for Symphogen, Adaptimmune, Omniox, Lycera, Pieris, and Torque, and holds stock options in Torque. RS has received personal fees from Amgen, Merck & Co., Genentech, Novartis, Compugen, Replimmune, Array, and Syndax, has recieved research funding from Amgen and Merck & Co., has recieved clinical trial support from Merck & Co., Tesaro, Sanofi, Genentech and Novartis., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.