228 results on '"Storkus, Wj"'
Search Results
2. miR-29b and miR-198 overexpression in CD8+ T cells of renal cell carcinoma patients down-modulates JAK3 and MCL-1 leading to immune dysfunction
- Author
-
Gigante, M, Pontrelli, P, Herr, W, D'Avenia, M, Zaza, G, Cavalcanti, E, Accetturo, M, Lucarelli, G, Carrieri, G, Battaglia, M, Storkus, WJ, Gesualdo, L, Ranieri, E, Gigante, M, Pontrelli, P, Herr, W, D'Avenia, M, Zaza, G, Cavalcanti, E, Accetturo, M, Lucarelli, G, Carrieri, G, Battaglia, M, Storkus, WJ, Gesualdo, L, and Ranieri, E
- Abstract
Background: Mammalian microRNAs (miR) regulate the expression of genes relevant for the development of adaptive and innate immunity against cancer. Since T cell dysfunction has previously been reported in patients with renal cell carcinoma (RCC; clear cell type), we aimed to analyze these immune cells for genetic and protein differences when compared to normal donor T cells freshly after isolation and 35 days after in vitro stimulation (IVS) with HLA-matched RCC tumor cells. Methods: We investigated gene expression profiles of tumor-reactive CD8+ T cells obtained from RCC patient and compared with their HLA-matched healthy sibling donors using a microarray approach. In addition, miRNAs analysis was performed in a validation cohort of peripheral blood CD8+ T cells from 25 RCC patients compared to 15 healthy volunteers. Results: We observed that CD8+ T cells from RCC patients expressed reduced levels of anti-apoptotic and proliferation-associated gene products when compared with normal donor T cells both pre- and post-IVS. In particular, JAK3 and MCL-1 were down-regulated in patient CD8+ T cells versus their normal counterparts, likely due to defective suppressor activity of miR-29b and miR-198 in RCC CD8+ T cells. Indeed, specific inhibition of miR-29b or miR-198 in peripheral blood mononuclear cells (PBMCs) isolated from RCC patients, resulted in the up-regulation of JAK3 and MCL-1 proteins and significant improvement of cell survival in vitro. Conclusions: Our results suggest that miR-29b and miR-198 dysregulation in RCC patient CD8+ T cells is associated with dysfunctional immunity and foreshadow the development of miR-targeted therapeutics to correct such T cell defects in vivo.
- Published
- 2016
3. La regolazione genica attraverso i microRNA è associata ad alterazioni della risposta immunologica in pazienti con carcinoma renale (RCC): ruolo di JAK3 e MCL-1
- Author
-
Gigante, M, Pontrelli, P, Zaza, Gianluigi, Montemurno, E, Mancini, V, Battaglia, M, Storkus, Wj, Gesualdo, L, and Ranieri, E.
- Subjects
micro-RNA ,carcinoma renale ,JAK3 - Published
- 2010
4. Sensory nerves in melanoma impede the formation of tertiary lymphoid structures and anti-tumor immune responses
- Author
-
Kruglov, O, primary, Vats, K, additional, Sahoo, B, additional, Soman, V, additional, Chandran, UR, additional, Shurin, GV, additional, Skums, P, additional, Shurin, MR, additional, Zelikovsky, A, additional, Storkus, WJ, additional, and Bunimovich, YL, additional
- Full Text
- View/download PDF
5. JAK3/STAT5/6 pathway alterations are associated with immune deviation in CD8 T cells in renal cell carcinoma patients.
- Author
-
Cavalcanti E, Gigante M, Mancini V, Battaglia M, Ditonno P, Capobianco C, Cincione RI, Selvaggi FP, Herr W, Storkus WJ, Gesualdo L, and Ranieri E
- Abstract
To investigate the molecular mechanisms underlying altered T cell response in renal cell carcinoma (RCC) patients, we compared autologous and allogeneic CD8(+) T cell responses against RCC line from RCC patients and their HLA-matched donors, using mixed lymphocyte/tumor cell cultures (MLTCs). In addition, we analyzed the expression of molecules associated with cell cycle regulation. Autologous MLTC responder CD8(+) T cells showed cytotoxic activity against RCC cell lines; however the analysis of the distribution of CD8(+) T-cell subsets revealed that allogenic counterparts mediate superior antitumor efficacy. In RCC patients, a decreased proliferative response to tumor, associated with defects in JAK3/STAT5/6 expression that led to increased p27KIP1 expression and alterations in the cell cycle, was observed. These data define a molecular pathway involved in cell cycle regulation that is associated with the dysfunction of tumor-specific CD8(+) effector cells. If validated, this may define a therapeutic target in the setting of patients with RCC. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
6. NK Receptor Signaling Lowers TCR Activation Threshold, Enhancing Selective Recognition of Cancer Cells by TAA-Specific CTLs.
- Author
-
Dong B, Obermajer N, Tsuji T, Matsuzaki J, Bonura CM, Sander C, Withers H, Long MD, Chavel C, Olejniczak SH, Minderman H, Kirkwood JM, Edwards RP, Storkus WJ, Romero P, and Kalinski P
- Subjects
- Humans, Melanoma immunology, Melanoma metabolism, NK Cell Lectin-Like Receptor Subfamily K metabolism, Lymphocyte Activation immunology, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Antigens, Differentiation, T-Lymphocyte metabolism, Antigens, Differentiation, T-Lymphocyte immunology, Cell Line, Tumor, MART-1 Antigen immunology, MART-1 Antigen metabolism, Cytotoxicity, Immunologic, Receptors, Antigen, T-Cell metabolism, Receptors, Antigen, T-Cell immunology, Signal Transduction, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Cytotoxic metabolism, Antigens, Neoplasm immunology, Antigens, Neoplasm metabolism
- Abstract
Cytotoxic CD8+ T lymphocyte (CTL) recognition of non-mutated tumor-associated antigens (TAA), present on cancer cells and also in healthy tissues, is an important element of cancer immunity, but the mechanism of its selectivity for cancer cells and opportunities for its enhancement remain elusive. In this study, we found that CTL expression of the NK receptors (NKR) DNAM1 and NKG2D was associated with the effector status of CD8+ tumor-infiltrating lymphocytes and long-term survival of patients with melanoma. Using MART1 and NY-ESO-1 as model TAAs, we demonstrated that DNAM1 and NKG2D regulate T-cell receptor (TCR) functional avidity and set the threshold for TCR activation of human TAA-specific CTLs. Superior co-stimulatory effects of DNAM1 over CD28 involved enhanced TCR signaling, CTL killer function, and polyfunctionality. Double transduction of human CTLs with TAA-specific TCR and NKRs resulted in strongly enhanced antigen sensitivity, without a reduction in antigen specificity and selectivity of killer function. In addition, the elevation of NKR ligand expression on cancer cells due to chemotherapy also increased CTL recognition of cancer cells expressing low levels of TAAs. Our data help explain the ability of self-antigens to mediate tumor rejection in the absence of autoimmunity and support the development of dual-targeting adoptive T-cell therapies that use NKRs to enhance the potency and selectivity of recognition of TAA-expressing cancer cells., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
7. Therapeutic Anti-Tumor Efficacy of DC-Based Vaccines Targeting TME-Associated Antigens Is Improved When Combined with a Chemokine-Modulating Regimen and/or Anti-PD-L1.
- Author
-
Taylor JL, Kokolus KM, Basse PH, Filderman JN, Cosgrove CE, Watkins SC, Gambotto A, Lowe DB, Edwards RP, Kalinski P, and Storkus WJ
- Abstract
We previously reported that dendritic cell (DC)-based vaccines targeting antigens expressed by tumor-associated vascular endothelial cells (VECs) and pericytes effectively control tumor growth in translational mouse tumor models. In the current report, we examined whether the therapeutic benefits of such tumor blood vessel antigen (TBVA)-targeted vaccines could be improved by the cotargeting of tumor antigens in the s.c. B16 melanoma model. We also evaluated whether combination vaccines incorporating anti-PD-L1 checkpoint blockade and/or a chemokine-modulating (CKM; IFNα + TLR3-L [rintatolimod] + Celecoxib) regimen would improve T cell infiltration/functionality in tumors yielding enhanced treatment benefits. We report that DC-peptide or DC-tumor lysate vaccines coordinately targeting melanoma antigens and TBVAs were effective in slowing B16 growth in vivo and extending survival, with superior outcomes observed for DC-peptide-based vaccines. Peptide-based vaccines that selectively target either melanoma antigens or TBVAs elicited a CD8
+ T cell repertoire recognizing both tumor cells and tumor-associated VECs and pericytes in vitro, consistent with a treatment-induced epitope spreading mechanism. Notably, combination vaccines including anti-PD-L1 + CKM yielded superior therapeutic effects on tumor growth and animal survival, in association with the potentiation of polyfunctional CD8+ T cell reactivity against both tumor cells and tumor-associated vascular cells and a pro-inflammatory TME.- Published
- 2024
- Full Text
- View/download PDF
8. Antagonism of regulatory ISGs enhances the anti-melanoma efficacy of STING agonists.
- Author
-
Filderman JN, Taylor JL, Wang J, Zhang Y, Singh P, Ross MA, Watkins SC, Nedal Al Bzour A, Karapetyan L, Kalinski P, and Storkus WJ
- Subjects
- Mice, Animals, Proto-Oncogene Proteins B-raf, Cyclooxygenase 2, Cell Line, Tumor, Interferons, Tumor Microenvironment, B7-H1 Antigen, Melanoma, Experimental
- Abstract
Background: Stimulator of Interferon Genes (STING) is a dsDNA sensor that triggers type I inflammatory responses. Recent data from our group and others support the therapeutic efficacy of STING agonists applied intratumorally or systemically in a range of murine tumor models, with treatment benefits associated with tumor vascular normalization and improved immune cell recruitment and function within the tumor microenvironment (TME). However, such interventions are rarely curative and STING agonism coordinately upregulates expression of immunoregulatory interferon-stimulated genes (ISGs) including Arg2, Cox2, Isg15, Nos2 , and Pdl1 that may limit treatment benefits. We hypothesized that combined treatment of melanoma-bearing mice with STING agonist ADU-S100 together with antagonists of regulatory ISGs would result in improved control of tumor growth vs. treatment with ADU-S100 alone., Methods: Mice bearing either B16 (BRAF
WT PTENWT ) or BPR20 (BRAFV600E PTEN-/- ) melanomas were treated with STING agonist ADU-S100 plus various inhibitors of ARG2, COX2, NOS2, PD-L1, or ISG15. Tumor growth control and changes in the TME were evaluated for combination treatment vs ADU-S100 monotherapy by tumor area measurements and flow cytometry/transcriptional profiling, respectively., Results: In the B16 melanoma model, we noted improved antitumor efficacy only when ADU-S100 was combined with neutralizing/blocking antibodies against PD-L1 or ISG15, but not inhibitors of ARG2, COX2, or NOS2. Conversely, in the BPR20 melanoma model, improved tumor growth control vs. ADU-S100 monotherapy was only observed when combining ADU-S100 with ARG2i, COX2i, and NOS2i, but not anti-PD-L1 or anti-ISG15. Immune changes in the TME associated with improved treatment outcomes were subtle but included increases in proinflammatory innate immune cells and activated CD8+ CD69+ T cells and varied between the two tumor models., Conclusions: These data suggest contextual differences in the relative contributions of individual regulatory ISGs that serve to operationally limit the anti-tumor efficacy of STING agonists which should be considered in future design of novel combination protocols for optimal treatment benefit., 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. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Filderman, Taylor, Wang, Zhang, Singh, Ross, Watkins, Nedal Al Bzour, Karapetyan, Kalinski and Storkus.)- Published
- 2024
- Full Text
- View/download PDF
9. NK Receptors Replace CD28 As the Dominant Source of Signal 2 for Cognate Recognition of Cancer Cells by TAA-specific Effector CD8 + T Cells.
- Author
-
Dong B, Obermajer N, Tsuji T, Matsuzaki J, Bonura C, Withers H, Long M, Chavel C, Olejniczak SH, Minderman H, Edwards RP, Storkus WJ, Romero P, and Kalinski P
- Abstract
CD28-driven "signal 2" is critical for naïve CD8
+ T cell responses to dendritic cell (DC)-presented weak antigens, including non-mutated tumor-associated antigens (TAAs). However, it is unclear how DC-primed cytotoxic T lymphocytes (CTLs) respond to the same TAAs presented by cancer cells which lack CD28 ligands. Here, we show that NK receptors (NKRs) DNAM-1 and NKG2D replace CD28 during CTL re-activation by cancer cells presenting low levels of MHC I/TAA complexes, leading to enhanced proximal TCR signaling, immune synapse formation, CTL polyfunctionality, release of cytolytic granules and antigen-specific cancer cell killing. Double-transduction of T cells with recombinant TCR and NKR constructs or upregulation of NKR-ligand expression on cancer cells by chemotherapy enabled effective recognition and killing of poorly immunogenic tumor cells by CTLs. Operational synergy between TCR and NKRs in CTL recognition explains the ability of cancer-expressed self-antigens to serve as tumor rejection antigens, helping to develop more effective therapies., Competing Interests: Conflict of interest: The authors have declared that no conflict of interest exists.- Published
- 2023
- Full Text
- View/download PDF
10. Platelet STING agonism and venous thrombosis: translational implications for improved disease outcomes.
- Author
-
Filderman JN, Luke JJ, and Storkus WJ
- Subjects
- Humans, Blood Platelets, Neutrophils, Extracellular Traps, Venous Thrombosis drug therapy
- Published
- 2023
- Full Text
- View/download PDF
11. Expression of lymphoid structure-associated cytokine/chemokine gene transcripts in tumor and protein in serum are prognostic of melanoma patient outcomes.
- Author
-
Karapetyan L, AbuShukair HM, Li A, Knight A, Al Bzour AN, MacFawn IP, Thompson ZJ, Chen A, Yang X, Dadey R, Karunamurthy A, De Stefano DV, Sander C, Kunning SR, Najjar YG, Davar D, Luke JJ, Gooding W, Bruno TC, Kirkwood JM, and Storkus WJ
- Subjects
- Humans, Female, Middle Aged, Aged, Male, Prognosis, Cytokines, Gene Expression Profiling, Genomics, Tumor Microenvironment genetics, Melanoma genetics
- Abstract
Background: Proinflammatory chemokines/cytokines support development and maturation of tertiary lymphoid structures (TLS) within the tumor microenvironment (TME). In the current study, we sought to investigate the prognostic value of TLS-associated chemokines/cytokines (TLS-kines) expression levels in melanoma patients by performing serum protein and tissue transcriptomic analyses, and to then correlate these data with patients clinicopathological and TME characteristics., Methods: Levels of TLS-kines in patients' sera were quantitated using a custom Luminex Multiplex Assay. The Cancer Genomic Atlas melanoma cohort (TCGA-SKCM) and a Moffitt Melanoma cohort were used for tissue transcriptomic analyses. Associations between target analytes and survival outcomes, clinicopathological variables, and correlations between TLS-kines were statistically analyzed., Results: Serum of 95 patients with melanoma were evaluated; 48 (50%) female, median age of 63, IQR 51-70 years. Serum levels of APRIL/TNFSF13 were positively correlated with levels of both CXCL10 and CXCL13. In multivariate analyses, high levels of serum APRIL/TNFSF13 were associated with improved event-free survival after adjusting for age and stage (HR = 0.64, 95% CI 0.43-0.95; p = 0.03). High expression of APRIL/TNFSF13 tumor transcripts was significantly associated with improved OS in TCGA-SKCM (HR = 0.69, 95% CI 0.52-0.93; p = 0.01) and in Moffitt Melanoma patients (HR = 0.51, 95% CI: 0.32-0.82; p = 0.006). Further incorporation of CXCL13 and CXCL10 tumor transcript levels in a 3-gene index revealed that high APRIL/CXCL10/CXCL13 expression was associated with improved OS in the TCGA SKCM cohort (HR = 0.42, 95% CI 0.19-0.94; p = 0.035). Melanoma differentially expressed genes positively associated with high APRIL/CXCL10/CXCL13 tumor expression were linked to tumor infiltration by a diverse array of proinflammatory immune cell types., Conclusion: Serum protein and tumor transcript levels of APRIL/TNFSF13 are associated with improved survival outcomes. Patients exhibiting high coordinate expression of APRIL/CXCL10/CXCL13 transcripts in their tumors displayed superior OS. Further investigation of TLS-kine expression profiles related to clinical outcomes in larger cohort studies is warranted., Competing Interests: The authors report no competing interests relevant to this work. Outside of this work: JL reports the following disclosures: DSMB: Abbvie, Immutep, Evaxion; Scientific Advisory Board: no stock 7 Hills, Affivant, Bright Peak, Exo, Fstar, Inzen, RefleXion, Xilio stock Actym, Alphamab Oncology, Arch Oncology, Duke Street Bio, Kanaph, Mavu, NeoTx, Onc.AI, OncoNano, physIQ, Pyxis, Saros, STipe, Tempest; Consultancy with compensation: Abbvie, Agenus, Alnylam, Atomwise, Bayer, Bristol-Myers Squibb, Castle, Checkmate, Codiak, Crown, Cugene, Curadev, Day One, Eisai, EMD Serono, Endeavor, Flame, G1 Therapeutics, Genentech, Gilead, Glenmark, HotSpot, Kadmon, KSQ, Janssen, Ikena, Inzen, Immatics, Immunocore, Incyte, Instil, IO Biotech, Macrogenics, Merck, Mersana, Nektar, Novartis, Partner, Pfizer, Pioneering Medicines, PsiOxus, Regeneron, Replimmune, Ribon, Roivant, Servier, STINGthera, Synlogic, Synthekine; Research Support: all to institution for clinical trials unless noted AbbVie, Astellas, Astrazeneca, Bristol-Myers Squibb, Corvus, Day One, EMD Serono, Fstar, Genmab, Ikena, Immatics, Incyte, Kadmon, KAHR, Macrogenics, Merck, Moderna, Nektar, Next Cure, Numab, Palleon, Pfizer, Replimmune, Rubius, Servier, Scholar Rock, Synlogic, Takeda, Trishula, Tizona, Xencor; Patents: both provisional Serial #15/612,657 Cancer Immunotherapy, PCT/US18/36052 Microbiome Biomarkers for Anti-PD-1/PD-L1 Responsiveness: Diagnostic, Prognostic and Therapeutic Uses Thereof. DDa reports the following disclosures: Arcus, Bristol-Myers Squibb, Checkmate Pharmaceuticals, CellSight Technologies, Merck, GlaxoSmithKline/Tesaro research support; Array Biopharma, Checkmate Pharmaceuticals, Finch, Incyte, Immunocore, Merck; Shionogi consulting; and Vedanta Biosciences scientific advisory board. YN reports the following disclosures: Merck, Pfizer, and Bristol-Myers Squibb research support. Array Biopharma, Merck, Novartis, InterVenn Bio consulting/scientific advisory board. Pfizer, Immunocore speaker’s bureau. CE Speakers’ Bureau: Medical Learning Group MLG. JK reports the following disclosures: Honoraria: Bristol Myers Squibb Consulting or Advisory Role: Novartis, Amgen, Harbor BioMed, Istari Oncology, Scopus BioPharma, Pfizer, AXIO Research, Immunocore, Natera, DermTech, Ankyra Therapeutics, Becker Pharmaceutical Consulting, Fenix Group International, IQVIA, Merck, Replimune, SR One Capital Management, Iovance Biotherapeutics, Checkmate Pharmaceuticals, OncoSec, OncoCyte, Cancer Network, Takeda, Applied Clinical Intelligence. Research Funding: Amgen Inst, Bristol Myers Squibb Inst, Checkmate Pharmaceuticals Inst, Immunocore Inst, Iovance Biotherapeutics Inst, Novartis Inst, ImmVira Inst, Harbor BioMed Inst, Takeda Inst, Verastem Inst. TB reports the following disclosures: Walking Fish Therapeutics Scientific Advisory Board. The remaining 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. The reviewer PK declared a past collaboration with the author WJS to the handling editor., (Copyright © 2023 Karapetyan, AbuShukair, Li, Knight, Al Bzour, MacFawn, Thompson, Chen, Yang, Dadey, Karunamurthy, De Stefano, Sander, Kunning, Najjar, Davar, Luke, Gooding, Bruno, Kirkwood and Storkus.)
- Published
- 2023
- Full Text
- View/download PDF
12. Sentinel Lymph Node Gene Expression Signature Predicts Recurrence-Free Survival in Cutaneous Melanoma.
- Author
-
Karapetyan L, Gooding W, Li A, Yang X, Knight A, Abushukair HM, Vargas De Stefano D, Sander C, Karunamurthy A, Panelli M, Storkus WJ, Tarhini AA, and Kirkwood JM
- Abstract
We sought to develop a sentinel lymph node gene expression signature score predictive of disease recurrence in patients with cutaneous melanoma. Gene expression profiling was performed on SLN biopsies using U133A 2.0 Affymetrix gene chips. The top 25 genes associated with recurrence-free survival (RFS) were selected and a penalized regression function was used to select 12 genes with a non-zero coefficient. A proportional hazards regression model was used to evaluate the association between clinical covariates, gene signature score, and RFS. Among the 45 patients evaluated, 23 (51%) had a positive SLN. Twenty-one (46.7%) patients developed disease recurrence. For the top 25 differentially expressed genes (DEG), 12 non-zero penalized coefficients were estimated (CLGN, C1QTNF3, ADORA3, ARHGAP8, DCTN1, ASPSCR1, CHRFAM7A, ZNF223, PDE6G, CXCL3, HEXIM1, HLA-DRB). This 12-gene signature score was significantly associated with RFS (p < 0.0001) and produced a bootstrap C index of 0.888. In univariate analysis, Breslow thickness, presence of primary tumor ulceration, SLN positivity were each significantly associated with RFS. After simultaneously adjusting for these prognostic factors in relation to the gene signature, the 12-gene score remained a significant independent predictor for RFS (p < 0.0001). This SLN 12-gene signature risk score is associated with melanoma recurrence regardless of SLN status and may be used as a prognostic factor for RFS.
- Published
- 2022
- Full Text
- View/download PDF
13. Sensory Nerves Impede the Formation of Tertiary Lymphoid Structures and Development of Protective Antimelanoma Immune Responses.
- Author
-
Vats K, Kruglov O, Sahoo B, Soman V, Zhang J, Shurin GV, Chandran UR, Skums P, Shurin MR, Zelikovsky A, Storkus WJ, and Bunimovich YL
- Subjects
- Humans, Immunity, Tumor Microenvironment, Melanoma, Skin Neoplasms, Tertiary Lymphoid Structures
- Abstract
Peripheral neurons comprise a critical component of the tumor microenvironment (TME). The role of the autonomic innervation in cancer has been firmly established. However, the effect of the afferent (sensory) neurons on tumor progression remains unclear. Utilizing surgical and chemical skin sensory denervation methods, we showed that afferent neurons supported the growth of melanoma tumors in vivo and demonstrated that sensory innervation limited the activation of effective antitumor immune responses. Specifically, sensory ablation led to improved leukocyte recruitment into tumors, with decreased presence of lymphoid and myeloid immunosuppressive cells and increased activation of T-effector cells within the TME. Cutaneous sensory nerves hindered the maturation of intratumoral high endothelial venules and limited the formation of mature tertiary lymphoid-like structures containing organized clusters of CD4+ T cells and B cells. Denervation further increased T-cell clonality and expanded the B-cell repertoire in the TME. Importantly, CD8a depletion prevented denervation-dependent antitumor effects. Finally, we observed that gene signatures of inflammation and the content of neuron-associated transcripts inversely correlated in human primary cutaneous melanomas, with the latter representing a negative prognostic marker of patient overall survival. Our results suggest that tumor-associated sensory neurons negatively regulate the development of protective antitumor immune responses within the TME, thereby defining a novel target for therapeutic intervention in the melanoma setting., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
14. Editorial: Immunotherapy in renal cell carcinoma.
- Author
-
Gruenwald V and Storkus WJ
- Abstract
This collection contains 10 reports published In Frontiers in Oncology between August 2020 and March 2022 broadly focused on the immunobiology of renal cell carcinoma (RCC), the impact of immunotherapy in the setting of RCC, and the identification of biomarkers that are prognostic of RCC patient outcomes and response to immunotherapy., 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., (Copyright © 2022 Gruenwald and Storkus.)
- Published
- 2022
- Full Text
- View/download PDF
15. Finding the right help in the tumor microenvironment.
- Author
-
Filderman JN and Storkus WJ
- Subjects
- CD4-Positive T-Lymphocytes, CD8-Positive T-Lymphocytes, Humans, Lymphocytes, Tumor-Infiltrating, Prognosis, Head and Neck Neoplasms pathology, Tumor Microenvironment
- Abstract
Tumor-infiltrating lymphocytes (TILs) contain substantial numbers of CD4+ T cells mediating pro- and antitumor functions. While CD4+ Tregs are well characterized and known to promote tumor immune evasion, the fingerprint of CD4+ Th cells that recognizes tumor antigens and serves to restrict disease progression has remained poorly discriminated. In this issue of the JCI, Duhen et al. analyzed tumors from patients with head and neck squamous cell carcinoma or colon carcinoma and identified a unique programmed cell death 1-positive, ICOS1-positive (PD-1+ICOS1+) subpopulation of CD4+ TILs highly enriched for the ability to recognize tumor-associated antigens. These cells localized proximally to MHC II+ antigen-presenting cells and CD8+ T cells within tumors, where they appeared to proliferate and function almost exclusively as Th cells. These potentially therapeutic Th cells can be monitored for patient prognosis and are expected to have substantial utility in developing personalized adoptive cell- and vaccine-based immunotherapeutic approaches for improving patient outcomes.
- Published
- 2022
- Full Text
- View/download PDF
16. Poorer survival outcomes in patients with multiple versus single primary melanoma.
- Author
-
Karapetyan L, Yang X, Knight AD, Huang Z, Wang H, Sander CA, Minnier CP, Wilson M, Li A, Karunamurthy A, Storkus WJ, and Kirkwood JM
- Subjects
- Female, Humans, Lymphocytes, Tumor-Infiltrating, Male, Middle Aged, Neoplasm Recurrence, Local, Prognosis, Retrospective Studies, Melanoma, Skin Neoplasms
- Abstract
Background: Given equivocal results related to overall survival (OS) for patients with multiple primary melanomas (MPMs) compared with those with single primary melanomas (SPMs) in previous reports, the authors sought to determine whether OS differs between these 2 cohorts in their center using their UPCI-96-99 database. Secondary aims were to assess the differences in recurrence-free survival (RFS). In a subset of patients, transcriptomic profiling of peripheral blood mononuclear cells (PBMCs) was performed to assess disease-associated genes of interest., Methods: This retrospective case-controlled study included patients with MPMs and age-, sex-, and stage-matched controls with SPMs at a 1:1 ratio. Cox regression models were used to evaluate the effect of the presence of MPMs on death and recurrence. NanoString PanCancer Immune Profiling was used to assess peripheral blood immune status in patients., Results: In total, 320 patients were evaluated. The mean patient age was 47 years; 43.8% were male. Patients with MPMs had worse RFS and OS (P = .023 and P = .0019, respectively). The presence of MPMs was associated with an increased risk of death (hazard ratio [HR], 4.52, P = .0006), and increased risk of disease recurrence (HR, 2.17; P = .004) after adjusting for age, sex, and stage. The degree of tumor-infiltrating lymphocytes (TILs) was different between the first melanoma of MPMs and SPMs. Expression of CXCL6 and FOXJ1 was increased in PBMCs isolated from patients with MPMs., Conclusions: Patients with MPMs had worse RFS and OS compared with patients with SPMs. Immunologic differences were also observed, including TIL content and expression of CXCL6/FOXJ1 in PBMCs of patients with MPMs, which warrant further investigation., (© 2022 American Cancer Society.)
- Published
- 2022
- Full Text
- View/download PDF
17. PET Imaging of VLA-4 in a New BRAF V600E Mouse Model of Melanoma.
- Author
-
Bellavia MC, Nyiranshuti L, Latoche JD, Ho KV, Fecek RJ, Taylor JL, Day KE, Nigam S, Pun M, Gallazzi F, Edinger RS, Storkus WJ, Patel RB, and Anderson CJ
- Subjects
- Animals, Cell Line, Tumor, Copper Radioisotopes, Disease Models, Animal, Humans, Mice, Positron Emission Tomography Computed Tomography, Positron-Emission Tomography methods, Prospective Studies, Proto-Oncogene Proteins B-raf genetics, Tissue Distribution, Integrin alpha4beta1 metabolism, Melanoma diagnostic imaging, Melanoma genetics
- Abstract
Purpose: Despite unprecedented responses to immune checkpoint inhibitors and targeted therapy in melanoma, a major subset of patients progresses and have few effective salvage options. We have previously demonstrated robust, selective uptake of the peptidomimetic LLP2A labeled with Cu-64 ([
64 Cu]-LLP2A) for positron emission tomography (PET) imaging in subcutaneous and metastatic models of B16F10 murine melanoma. LLP2A binds with high affinity to very late antigen-4 (VLA-4, integrin α4 β1 ), a transmembrane protein overexpressed in melanoma and other cancers that facilitates tumor growth and metastasis. Yet B16F10 fails to faithfully reflect human melanoma biology, as it lacks certain oncogenic driver mutations, including BRAF mutations found in ≥ 50 % of clinical specimens. Here, we evaluated the PET tracer [64 Cu]-CB-TE1A1P-PEG4 -LLP2A ([64 Cu]-LLP2A) in novel, translational BRAFV600E mutant melanoma models differing in VLA-4 expression-BPR (VLA-4- ) and BPRα (VLA-4+ )., Procedures: BPR cells were transduced with α4 (CD49d) to overexpress intact cell surface VLA-4 (BPRα). The binding affinity of [64 Cu]-LLP2A to BPR and BPRα cells was determined by saturation binding assays. [64 Cu]-LLP2A internalization into B16F10, BPR, and BPRα cells was quantified via a plate-based assay. Tracer biodistribution and PET/CT imaging were evaluated in mice bearing subcutaneous BPR and BPRα tumors., Results: [64 Cu]-LLP2A demonstrated high binding affinity to BPRα (Kd = 1.4 nM) but indeterminate binding to BPR cells. VLA-4+ BPRα and B16F10 displayed comparable time-dependent [64 Cu]-LLP2A internalization, whereas BPR internalization was undetectable. PET/CT showed increased tracer uptake in BPRα tumors vs. BPR tumors in vivo, which was validated by significantly greater (p < 0.0001) BPRα tumor uptake in biodistribution analyses., Conclusions: [64 Cu]-LLP2A discriminates BPRα (VLA-4+ ) vs. BPR (VLA-4- ) melanomas in vivo, supporting translation of these BRAF-mutated melanoma models via prospective imaging and theranostic studies. These results extend the utility of LLP2A to selectively target clinically relevant and therapy-resistant tumor variants toward its use for therapeutic patient care., (© 2021. World Molecular Imaging Society.)- Published
- 2022
- Full Text
- View/download PDF
18. Improved prognosis and evidence of enhanced immunogenicity in tumor and circulation of high-risk melanoma patients with unknown primary.
- Author
-
Tarhini AA, Lee SJ, Tan AC, El Naqa IM, Stephen Hodi F, Butterfield LH, LaFramboise WA, Storkus WJ, Karunamurthy AD, Conejo-Garcia JR, Hwu P, Streicher H, Sondak VK, and Kirkwood JM
- Subjects
- Adolescent, Adult, Child, Gene Expression Profiling, Humans, Melanoma immunology, Melanoma pathology, Neoplasms, Unknown Primary immunology, Neoplasms, Unknown Primary pathology, Skin Neoplasms immunology, Skin Neoplasms pathology, Young Adult, Melanoma mortality, Neoplasms, Unknown Primary mortality, Skin Neoplasms mortality
- Abstract
Background: Melanoma of unknown primary (MUP) represents a poorly understood group of patients both clinically and immunologically. We investigated differences in prognosis and candidate immune biomarkers in patients with unknown compared with those with known primary melanoma enrolled in the E1609 adjuvant trial that tested ipilimumab at 3 and 10 mg/kg vs high-dose interferon-alfa (HDI)., Patients and Methods: MUP status was defined as initial presentation with cutaneous, nodal or distant metastasis without a known primary. Relapse-free survival (RFS) and overall survival (OS) rates were estimated by the Kaplan-Meier method. Stratified (by stage) log-rank test was used to compare RFS and OS by primary tumor status. Gene expression profiling (GEP) was performed on the tumor biopsies of a subset of patients. Similarly, peripheral blood samples were tested for candidate soluble and cellular immune biomarkers., Results: MUP cases represented 12.8% of the total population (N=1699) including 11.7% on the ipilimumab arms and 14.7% on the HDI arm. Stratifying by stage, RFS (p=0.001) and overall survival (OS) (p=0.009) showed outcomes significantly better for patients with unknown primary. The primary tumor status remained prognostically significant after adjusting for treatment and stage in multivariate Cox proportional hazards models. Including only ipilimumab-treated patients, RFS (p=0.005) and OS (p=0.023) were significantly better in favor of those with unknown primary. Among patients with GEP data (n=718; 102 MUP, 616 known), GEP identified pathways and genes related to autoimmunity, inflammation, immune cell infiltration and immune activation that were significantly enriched in the MUP tumors compared with known primaries. Further investigation into infiltrating immune cell types estimated significant enrichment with CD8 +and CD4+T cells, B cells and NK cells as well as significantly higher major histocompatibility complex (MHC)-I and MHC-II scores in MUP compared with known primary. Among patients tested for circulating biomarkers (n=321; 66 unknown and 255 known), patients with MUP had significantly higher circulating levels of IL-2R (p=0.04)., Conclusion: Patients with MUP and high-risk melanoma had significantly better prognosis and evidence of significantly enhanced immune activation within the TME and the circulation, supporting the designation of MUP as a distinct prognostic marker in patients with high-risk melanoma., Competing Interests: Competing interests: AAT reports grants from National Cancer Institute, National Institute of Health, ECOG-ACRIN, grants from Bristol Myers Squibb, during the conduct of the study; grants from Bristol Myers Squibb, personal fees from Bristol Myers Squibb, grants from Merck, personal fees from Merck, personal fees from Novartis, personal fees from Genentech- Roche, grants from Genentech-Roche, personal fees from Array Biopharma, grants from Incyte, personal fees from Incyte, personal fees from NEWLINK Genetics, personal fees from HUYA, personal fees from BioNTech, grants from Prometheus, personal fees from Prometheus, personal fees from Immunocore, grants from Greenpeptide, grants from Amgen, grants from Clinigen, personal fees from Clinigen, personal fees from Partners Therapeutics, personal fees and grants from Regeneron, personal fees and grants from Sanofi-Genzyme outside the submitted work. SJL has nothing to disclose. A-CT reports has nothing to disclose. IMEN is a deputy editor for Medical Physics and reports relationship with Scientific Advisory Endectra, LLC. FSH reports clinical trial support from Eastern Cooperative oncology Group, during the conduct of the study; grants, personal fees and other from Bristol-Myers Squibb, personal fees from Merck, personal fees from EMD Serono, grants and personal fees from Novartis, personal fees from Takeda, personal fees from Surface, personal fees from Genentech/Roche, personal fees from Compass Therapeutics, personal fees from Apricity, personal fees from Bayer, personal fees from Aduro, personal fees from Partners Therapeutics, personal fees from Sanofi, personal fees from Pfizer, personal fees from Pionyr, from 7 Hills Pharma, personal fees from Verastem, other from Torque, personal fees from Rheos, outside the submitted work; in addition, FSH has a patent Methods for Treating MICA-Related Disorders (#20100111973) with royalties paid, a patent Tumor antigens and uses thereof (#7250291) issued, a patent Angiopoiten-2 Biomarkers Predictive of Anti-immune checkpoint response (#20170248603) pending, a patent Compositions and Methods for Identification, Assessment, Prevention, and Treatment of Melanoma using PD-L1 Isoforms (#20160340407) pending, a patent Therapeutic peptides (#20160046716) pending, a patent Therapeutic Peptides (#20140004112) pending, a patent Therapeutic Peptides (#20170022275) pending, a patent Therapeutic Peptides (#20170008962) pending, a patent THERAPEUTIC PEPTIDES Therapeutic Peptides Patent number: 9402905 issued, and a patent METHODS OF USING PEMBROLIZUMAB AND TREBANANIB pending. LHB declares the following unrelated advisory activities: StemImmune/Calidi Scientific and Medical Advisory Board, April 6, 2017-present; Western Oncolytics, Scientific Advisory Board, 2018-present; Torque Therapeutics, Scientific Advisory Board, 2018-2020; Khloris, Scientific Advisory Board, 2019-present; Pyxis, Scientific Advisory Board, 2019-present; Cytomix, Scientific Advisory Board, 2019-present; Vir, Scientific Advisory Board meeting, Feb. 2020; DCprime, Scientific Advisory Board meeting, Nov. 2020; RAPT, Scientific Advisory Board, 2020-present; Takeda, Scientific Advisor, 2020-present; EnaraBio scientific advisor, Feb. 2021. WAL has nothing to disclose. WJS has nothing to disclose. ADK has nothing to disclose. JRC-G has stock options with Compass Therapeutics, Anixa Biosciences and Alloy Therapeutics, receives honorarium from Anixa Biosciences, Alloy Therapeutics and Leidos, and has sponsored research with Anixa Biosciences. PH reports consulting fees consulting fees from Dragonfly and Immatics. HS has nothing to disclose. VKS reports personal fees from Merck, Bristol-Myers Squibb, Novartis, Array, Polynoma, Pfizer, and Regeneron, outside the submitted work. JMK reports grants and personal fees from Amgen, Bristol Myers Squibb, Castle Biosciences, Checkmate Pharmaceuticals, Immvira Pharma Co., Immunocore, Iovance Biotherapeutics, Lion Biotechnologies, Merck, Novartis Pharmaceuticals, Schering-Plough, personal fees Ankyra Therapeutics, Axio Research/Instil Bio, Becker Pharmaceutical Consulting, DermTech, Elsevier, Fenix Group International, Harbour BioMed, Intellisphere/Cancer Network, IQVIA, Istari Oncology, Millennium Pharmaceutical/Takeda Pharmaceutical, Natera, OncoCyte, OncoSec, Pfizer, Replimune, Scopus BioPharma, SR One Captital Management, outside the submitted work., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
19. Epigenetic modulation of antitumor immunity for improved cancer immunotherapy.
- Author
-
Dai E, Zhu Z, Wahed S, Qu Z, Storkus WJ, and Guo ZS
- Subjects
- Animals, Antineoplastic Agents, Immunological pharmacology, Antineoplastic Agents, Immunological therapeutic use, Biomarkers, Tumor, Clinical Trials as Topic, Combined Modality Therapy, Disease Management, Disease Susceptibility, Drug Development, Energy Metabolism, Gene Expression Regulation, Neoplastic, Humans, Neoplasms metabolism, Signal Transduction drug effects, Treatment Outcome, Tumor Microenvironment drug effects, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Epigenesis, Genetic, Immunity, Immunomodulation genetics, Immunotherapy methods, Neoplasms etiology, Neoplasms therapy
- Abstract
Epigenetic mechanisms play vital roles not only in cancer initiation and progression, but also in the activation, differentiation and effector function(s) of immune cells. In this review, we summarize current literature related to epigenomic dynamics in immune cells impacting immune cell fate and functionality, and the immunogenicity of cancer cells. Some important immune-associated genes, such as granzyme B, IFN-γ, IL-2, IL-12, FoxP3 and STING, are regulated via epigenetic mechanisms in immune or/and cancer cells, as are immune checkpoint molecules (PD-1, CTLA-4, TIM-3, LAG-3, TIGIT) expressed by immune cells and tumor-associated stromal cells. Thus, therapeutic strategies implementing epigenetic modulating drugs are expected to significantly impact the tumor microenvironment (TME) by promoting transcriptional and metabolic reprogramming in local immune cell populations, resulting in inhibition of immunosuppressive cells (MDSCs and Treg) and the activation of anti-tumor T effector cells, professional antigen presenting cells (APC), as well as cancer cells which can serve as non-professional APC. In the latter instance, epigenetic modulating agents may coordinately promote tumor immunogenicity by inducing de novo expression of transcriptionally repressed tumor-associated antigens, increasing expression of neoantigens and MHC processing/presentation machinery, and activating tumor immunogenic cell death (ICD). ICD provides a rich source of immunogens for anti-tumor T cell cross-priming and sensitizing cancer cells to interventional immunotherapy. In this way, epigenetic modulators may be envisioned as effective components in combination immunotherapy approaches capable of mediating superior therapeutic efficacy., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
20. Dendritic cell vaccines targeting tumor blood vessel antigens in combination with dasatinib induce therapeutic immune responses in patients with checkpoint-refractory advanced melanoma.
- Author
-
Storkus WJ, Maurer D, Lin Y, Ding F, Bose A, Lowe D, Rose A, DeMark M, Karapetyan L, Taylor JL, Chelvanambi M, Fecek RJ, Filderman JN, Looney TJ, Miller L, Linch E, Lowman GM, Kalinski P, Butterfield LH, Tarhini A, Tawbi H, and Kirkwood JM
- Subjects
- Antineoplastic Agents pharmacology, Cancer Vaccines pharmacology, Dasatinib pharmacology, Female, Humans, Male, Melanoma pathology, Pilot Projects, Prospective Studies, Antigens, Neoplasm therapeutic use, Antineoplastic Agents therapeutic use, Cancer Vaccines therapeutic use, Dasatinib therapeutic use, Dendritic Cells metabolism, Melanoma drug therapy
- Abstract
Background: A first-in-human, randomized pilot phase II clinical trial combining vaccines targeting overexpressed, non-mutated tumor blood vessel antigens (TBVA) and tyrosine kinase inhibitor dasatinib was conducted in human leukocyte antigen (HLA)-A2
+ patients with advanced melanoma., Methods: Patient monocyte-derived type-1-polarized dendritic cells were loaded with HLA-A2-presented peptides derived from TBVA (DLK1, EphA2, HBB, NRP1, RGS5, TEM1) and injected intradermally as a vaccine into the upper extremities every other week. Patients were randomized into one of two treatment arms receiving oral dasatinib (70 mg two times per day) beginning in week 5 (Arm A) or in week 1 (Arm B). Trial endpoints included T cell response to vaccine peptides (interferon-γ enzyme-linked immunosorbent spot), objective clinical response (Response Evaluation Criteria in Solid Tumors V.1.1) and exploratory tumor, blood and serum profiling of immune-associated genes/proteins., Results: Sixteen patients with advanced-stage cutaneous (n=10), mucosal (n=1) or uveal (n=5) melanoma were accrued, 15 of whom had previously progressed on programmed cell death protein 1 (PD-1) blockade. Of 13 evaluable patients, 6 patients developed specific peripheral blood T cell responses against ≥3 vaccine-associated peptides, with further evidence of epitope spreading. All six patients with specific CD8+ T cell response to vaccine-targeted antigens exhibited evidence of T cell receptor (TCR) convergence in association with preferred clinical outcomes (four partial response and two stabilization of disease (SD)). Seven patients failed to respond to vaccination (one SD and six progressive disease). Patients in Arm B (immediate dasatinib) outperformed those in Arm A (delayed dasatinib) for immune response rate (IRR; 66.7% vs 28.6%), objective response rate (ORR) (66.7% vs 0%), overall survival (median 15.45 vs 3.47 months; p=0.0086) and progression-free survival (median 7.87 vs 1.97 months; p=0.063). IRR (80% vs 25%) and ORR (60% vs 12.5%) was greater for females versus male patients. Tumors in patients exhibiting response to treatment displayed (1) evidence of innate and adaptive immune-mediated inflammation and TCR convergence at baseline, (2) on-treatment transcriptional changes associated with reduced hypoxia/acidosis/glycolysis, and (3) increased inflammatory immune cell infiltration and tertiary lymphoid structure neogenesis., Conclusions: Combined vaccination against TBVA plus dasatinib was safe and resulted in coordinating immunologic and/or objective clinical responses in 6/13 (46%) evaluable patients with melanoma, particularly those initiating treatment with both agents., Trial Registration Number: NCT01876212., Competing Interests: Competing interests: WJS, DM, YL, FD, LK, AB, DBL, AR, MD, JLT, MC, RJF, JNF and PK declare no competing interests. TJL was an employee of Thermo Fisher Scientific during the performance of this work. TJL, LM, EL and GML are/were employees of Thermo Fisher Scientific during the performance of this work. LHB declares the following unrelated advisory activities: StemImmune/Calidi Scientific and Medical Advisory Board, April 6, 2017–present; SapVax Advisory Board meetings November 15, 2017; December 6, 2018; NextCure, Scientific Advisory Board, 2018–2020; Western Oncolytics, Scientific Advisory Board, 2018–present; Torque Therapeutics, Scientific Advisory Board, 2018–2020; Khloris, Scientific Advisory Board, 2019–present; Pyxis, Scientific Advisory Board, 2019–present; Cytomix, Scientific Advisory Board, 2019–present; Vir, Scientific Advisory Board meeting, February 2020; DCprime, Scientific Advisory Board meeting, November 2020; RAPT, Scientific Advisory Board, 2020–present; Takeda, Scientific Advisor, 2020–present; EnaraBio scientific advisor, February 2021. AT declares the following unrelated advisory activities: Receipt of fees for consulting and/or advisory board participation from Partner Therapeutics, Merck, Bristol Myers Squibb, Novartis, Genentech-Roche, Array Biopharma, Sanofi-Genzyme/Regeneron, Pfizer, EMD Serono, NewLink Genetics, BioNTech, Immunocore, and Eisai; participation in a Data Safety Monitoring Board for Incyte; involvement with institution contracted research with Merck, OncoSec, Genentech-Roche, Bristol Myers Squibb, Amgen and Clinigen. HT declares the following unrelated consulting Honoraria: BMS, Novartis, Merck, Genentech, Eisai, Iovance, Boxer Capital, Karyopharm. Research Funding to Institution: BMS, Novartis, Merck, Genentech, GSK. JMK declares the following unrelated advisory activities and funding: Advisory Role: Bristol Myers Squibb, Novartis, Iovance Biotherapeutics, Elsevier, Amgen, Checkmate Pharmaceuticals, Harbour BioMed, Istari Oncology, OncoSec, Scopus BioPharma, Pfizer; Speakers’ Bureau: Bristol Myers Squibb unbranded IO; Research Funding: Amgen, Bristol Myers Squibb, Castle Biosciences, Checkmate Pharmaceuticals, Immunocore, Iovance Biotherapeutics, Novartis, Merck., (© 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
21. RGS5-TGFβ-Smad2/3 axis switches pro- to anti-apoptotic signaling in tumor-residing pericytes, assisting tumor growth.
- Author
-
Dasgupta S, Ghosh T, Dhar J, Bhuniya A, Nandi P, Das A, Saha A, Das J, Guha I, Banerjee S, Chakravarti M, Dasgupta PS, Alam N, Chakrabarti J, Majumdar S, Chakrabarti P, Storkus WJ, Baral R, and Bose A
- Subjects
- Animals, Female, Humans, Mice, Signal Transduction, Transfection, Neoplasms genetics, Pericytes metabolism, RGS Proteins metabolism, Smad2 Protein metabolism
- Abstract
Regulator-of-G-protein-signaling-5 (RGS5), a pro-apoptotic/anti-proliferative protein, is a signature molecule of tumor-associated pericytes, highly expressed in several cancers, and is associated with tumor growth and poor prognosis. Surprisingly, despite the negative influence of intrinsic RGS5 expression on pericyte survival, RGS5
high pericytes accumulate in progressively growing tumors. However, responsible factor(s) and altered-pathway(s) are yet to report. RGS5 binds with Gαi/q and promotes pericyte apoptosis in vitro, subsequently blocking GPCR-downstream PI3K-AKT signaling leading to Bcl2 downregulation and promotion of PUMA-p53-Bax-mediated mitochondrial damage. However, within tumor microenvironment (TME), TGFβ appeared to limit the cytocidal action of RGS5 in tumor-residing RGS5high pericytes. We observed that in the presence of high RGS5 concentrations, TGFβ-TGFβR interactions in the tumor-associated pericytes lead to the promotion of pSmad2-RGS5 binding and nuclear trafficking of RGS5, which coordinately suppressed RGS5-Gαi/q and pSmad2/3-Smad4 pairing. The RGS5-TGFβ-pSmad2 axis thus mitigates both RGS5- and TGFβ-dependent cellular apoptosis, resulting in sustained pericyte survival/expansion within the TME by rescuing PI3K-AKT signaling and preventing mitochondrial damage and caspase activation. This study reports a novel mechanism by which TGFβ fortifies and promotes survival of tumor pericytes by switching pro- to anti-apoptotic RGS5 signaling in TME. Understanding this altered RGS5 signaling might prove beneficial in designing future cancer therapy., (© 2021. The Author(s), under exclusive licence to ADMC Associazione Differenziamento e Morte Cellulare.)- Published
- 2021
- Full Text
- View/download PDF
22. CD40 Cross-Linking Induces Migration of Renal Tumor Cell through Nuclear Factor of Activated T Cells (NFAT) Activation.
- Author
-
Pontrelli P, Gigante M, Spadaccino F, Netti GS, Saldarelli M, Balducci L, Gigante M, Battaglia M, Storkus WJ, Castellano G, Stallone G, Gesualdo L, and Ranieri E
- Subjects
- Aged, Apoptosis, Biomarkers, Tumor genetics, CD40 Antigens genetics, CD40 Ligand genetics, Cell Movement, Cell Proliferation, Cross-Linking Reagents, Female, Humans, Kidney Neoplasms genetics, Kidney Neoplasms metabolism, Male, Middle Aged, NFATC Transcription Factors genetics, Neoplasm Metastasis, Prognosis, Tumor Cells, Cultured, Biomarkers, Tumor metabolism, CD40 Antigens metabolism, CD40 Ligand metabolism, Gene Expression Regulation, Neoplastic, Kidney Neoplasms pathology, NFATC Transcription Factors metabolism
- Abstract
CD40 crosslinking plays an important role in regulating cell migration, adhesion and proliferation in renal cell carcinoma (RCC). CD40/CD40L interaction on RCC cells activates different intracellular pathways but the molecular mechanisms leading to cell scattering are not yet clearly defined. Aim of our study was to investigate the main intracellular pathways activated by CD40 ligation and their specific involvement in RCC cell migration. CD40 ligation increased the phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun NH (2)-terminal kinase (JNK) and p38 MAPK. Furthermore, CD40 crosslinking activated different transcriptional factors on RCC cell lines: AP-1, NFkB and some members of the Nuclear Factor of Activated T cells (NFAT) family. Interestingly, the specific inhibition of NFAT factors by cyclosporine A, completely blocked RCC cell motility induced by CD40 ligation. In tumor tissue, we observed a higher expression of NFAT factors and in particular an increased activation and nuclear migration of NFATc4 on RCC tumor tissues belonging to patients that developed metastases when compared to those who did not. Moreover, CD40-CD40L interaction induced a cytoskeleton reorganization and increased the expression of integrin β1 on RCC cell lines, and this effect was reversed by cyclosporine A and NFAT inhibition. These data suggest that CD40 ligation induces the activation of different intracellular signaling pathways, in particular the NFATs factors, that could represent a potential therapeutic target in the setting of patients with metastatic RCC.
- Published
- 2021
- Full Text
- View/download PDF
23. Unbiased High-Throughput Drug Combination Pilot Screening Identifies Synergistic Drug Combinations Effective against Patient-Derived and Drug-Resistant Melanoma Cell Lines.
- Author
-
Close DA, Kirkwood JM, Fecek RJ, Storkus WJ, and Johnston PA
- Subjects
- Animals, Antineoplastic Agents chemistry, Cell Line, Tumor, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor standards, Drug Synergism, High-Throughput Screening Assays standards, Humans, Melanoma drug therapy, Mice, Reproducibility of Results, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm drug effects, Drug Screening Assays, Antitumor methods, High-Throughput Screening Assays methods
- Abstract
We describe the development, optimization, and validation of 384-well growth inhibition assays for six patient-derived melanoma cell lines (PDMCLs), three wild type (WT) for BRAF and three with V600E- BRAF mutations. We conducted a pilot drug combination (DC) high-throughput screening (HTS) of 45 pairwise 4×4 DC matrices prepared from 10 drugs in the PDMCL assays: two B-Raf inhibitors (BRAFi), a MEK inhibitor (MEKi), and a methylation agent approved for melanoma; cytotoxic topoisomerase II and DNA methyltransferase chemotherapies; and drugs targeting the base excision DNA repair enzyme APE1 (apurinic/apyrimidinic endonuclease-1/redox effector factor-1), SRC family tyrosine kinases, the heat shock protein 90 (HSP90) molecular chaperone, and histone deacetylases.Pairwise DCs between dasatinib and three drugs approved for melanoma therapy-dabrafenib, vemurafenib, or trametinib-were flagged as synergistic in PDMCLs. Exposure to fixed DC ratios of the SRC inhibitor dasatinib with the BRAFis or MEKis interacted synergistically to increase PDMCL sensitivity to growth inhibition and enhance cytotoxicity independently of PDMCL BRAF status. These DCs synergistically inhibited the growth of mouse melanoma cell lines that either were dabrafenib-sensitive or had acquired resistance to dabrafenib with cross resistance to vemurafenib, trametinib, and dasatinib. Dasatinib DCs with dabrafenib, vemurafenib, or trametinib activated apoptosis and increased cell death in melanoma cells independently of their BRAF status or their drug resistance phenotypes. These preclinical in vitro studies provide a data-driven rationale for the further investigation of DCs between dasatinib and BRAFis or MEKis as candidates for melanoma combination therapies with the potential to improve outcomes and/or prevent or delay the emergence of disease resistance.
- Published
- 2021
- Full Text
- View/download PDF
24. STING Agonists as Cancer Therapeutics.
- Author
-
Amouzegar A, Chelvanambi M, Filderman JN, Storkus WJ, and Luke JJ
- Abstract
The interrogation of intrinsic and adaptive resistance to cancer immunotherapy has identified lack of antigen presentation and type I interferon signaling as biomarkers of non-T-cell-inflamed tumors and clinical progression. A myriad of pre-clinical studies have implicated the cGAS/stimulator of interferon genes (STING) pathway, a cytosolic DNA-sensing pathway that drives activation of type I interferons and other inflammatory cytokines, in the host immune response against tumors. The STING pathway is also increasingly understood to have other anti-tumor functions such as modulation of the vasculature and augmentation of adaptive immunity via the support of tertiary lymphoid structure development. Many natural and synthetic STING agonists have entered clinical development with the first generation of intra-tumor delivered cyclic dinucleotides demonstrating safety but only modest systemic activity. The development of more potent and selective STING agonists as well as novel delivery systems that would allow for sustained inflammation in the tumor microenvironment could potentially augment response rates to current immunotherapy approaches and overcome acquired resistance. In this review, we will focus on the latest developments in STING-targeted therapies and provide an update on the clinical development and application of STING agonists administered alone, or in combination with immune checkpoint blockade or other approaches.
- Published
- 2021
- Full Text
- View/download PDF
25. STINGing the Tumor Microenvironment to Promote Therapeutic Tertiary Lymphoid Structure Development.
- Author
-
Filderman JN, Appleman M, Chelvanambi M, Taylor JL, and Storkus WJ
- Subjects
- Animals, Cytokines metabolism, Humans, Immunotherapy, Inflammation Mediators metabolism, Membrane Proteins metabolism, Neoplasms immunology, Neoplasms metabolism, Neoplasms pathology, Signal Transduction, Tertiary Lymphoid Structures metabolism, Tertiary Lymphoid Structures pathology, Antineoplastic Agents therapeutic use, Membrane Proteins agonists, Neoplasms drug therapy, Tertiary Lymphoid Structures immunology, Tumor Microenvironment immunology
- Abstract
Tertiary lymphoid structures (TLS), also known as ectopic lymphoid structures (ELS) or tertiary lymphoid organs (TLO), represent a unique subset of lymphoid tissues noted for their architectural similarity to lymph nodes, but which conditionally form in peripheral tissues in a milieu of sustained inflammation. TLS serve as regional sites for induction and expansion of the host B and T cell repertoires via an operational paradigm involving mature dendritic cells (DC) and specialized endothelial cells (i.e. high endothelial venules; HEV) in a process directed by TLS-associated cytokines and chemokines. Recent clinical correlations have been reported for the presence of TLS within tumor biopsies with overall patient survival and responsiveness to interventional immunotherapy. Hence, therapeutic strategies to conditionally reinforce TLS formation within the tumor microenvironment (TME) via the targeting of DC, vascular endothelial cells (VEC) and local cytokine/chemokine profiles are actively being developed and tested in translational tumor models and early phase clinical trials. In this regard, a subset of agents that promote tumor vascular normalization (VN) have been observed to coordinately support the development of a pro-inflammatory TME, maturation of DC and VEC, local production of TLS-inducing cytokines and chemokines, and therapeutic TLS formation. This mini-review will focus on STING agonists, which were originally developed as anti-angiogenic agents, but which have recently been shown to be effective in promoting VN and TLS formation within the therapeutic TME. Future application of these drugs in combination immunotherapy approaches for greater therapeutic efficacy is further discussed., 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., (Copyright © 2021 Filderman, Appleman, Chelvanambi, Taylor and Storkus.)
- Published
- 2021
- Full Text
- View/download PDF
26. Thirty years of therapeutic innovation in melanoma research.
- Author
-
Lejeune FJ, Storkus WJ, and Riley PA
- Subjects
- Biomedical Research, Humans, Time Factors, Melanoma drug therapy, Skin Neoplasms drug therapy
- Published
- 2021
- Full Text
- View/download PDF
27. Cutaneous Melanoma: Mutational Status and Potential Links to Tertiary Lymphoid Structure Formation.
- Author
-
Salem D, Chelvanambi M, Storkus WJ, and Fecek RJ
- Subjects
- Animals, Humans, Immunotherapy, Melanoma immunology, Melanoma pathology, Melanoma therapy, Prognosis, Skin Neoplasms immunology, Skin Neoplasms pathology, Skin Neoplasms therapy, Tertiary Lymphoid Structures pathology, Tumor Microenvironment, Biomarkers, Tumor genetics, Melanoma genetics, Mutation, Skin Neoplasms genetics, Tertiary Lymphoid Structures immunology
- Abstract
Recent advances in immunotherapy have enabled rapid evolution of novel interventional approaches designed to reinvigorate and expand patient immune responses against cancer. An emerging approach in cancer immunology involves the conditional induction of tertiary lymphoid structures (TLS), which are non-encapsulated ectopic lymphoid structures forming at sites of chronic, pathologic inflammation. Cutaneous melanoma (CM), a highly-immunogenic form of solid cancer, continues to rise in both incidence and mortality rate, with recent reports supporting a positive correlation between the presence of TLS in melanoma and beneficial treatment outcomes amongst advanced-stage patients. In this context, TLS in CM are postulated to serve as dynamic centers for the initiation of robust anti-tumor responses within affected regions of active disease. Given their potential importance to patient outcome, significant effort has been recently devoted to gaining a better understanding of TLS neogenesis and the influence these lymphoid organs exert within the tumor microenvironment. Here, we briefly review TLS structure, function, and response to treatment in the setting of CM. To uncover potential tumor-intrinsic mechanisms that regulate TLS formation, we have taken the novel perspective of evaluating TLS induction in melanomas impacted by common driver mutations in BRAF, PTEN, NRAS, KIT, PRDM1, and MITF. Through analysis of The Cancer Genome Atlas (TCGA), we show expression of DNA repair proteins (DRPs) including BRCA1, PAXIP, ERCC1, ERCC2, ERCC3, MSH2, and PMS2 to be negatively correlated with expression of pro-TLS genes, suggesting DRP loss may favor TLS development in support of improved patient outcome and patient response to interventional immunotherapy., 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., (Copyright © 2021 Salem, Chelvanambi, Storkus and Fecek.)
- Published
- 2021
- Full Text
- View/download PDF
28. STING agonist-based treatment promotes vascular normalization and tertiary lymphoid structure formation in the therapeutic melanoma microenvironment.
- Author
-
Chelvanambi M, Fecek RJ, Taylor JL, and Storkus WJ
- Subjects
- Angiogenic Proteins genetics, Angiogenic Proteins metabolism, Animals, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Cell Line, Tumor, Cytokines genetics, Cytokines metabolism, Dendritic Cells immunology, Dendritic Cells metabolism, Female, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating immunology, Melanoma, Experimental immunology, Melanoma, Experimental metabolism, Melanoma, Experimental pathology, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Signal Transduction, Skin Neoplasms immunology, Skin Neoplasms metabolism, Skin Neoplasms pathology, Tertiary Lymphoid Structures immunology, Tertiary Lymphoid Structures pathology, Tumor Burden drug effects, Tumor Microenvironment, Mice, Antineoplastic Agents pharmacology, CD8-Positive T-Lymphocytes drug effects, Dendritic Cells drug effects, Lymphocytes, Tumor-Infiltrating metabolism, Melanoma, Experimental drug therapy, Membrane Proteins agonists, Neovascularization, Pathologic, Skin Neoplasms drug therapy, Tertiary Lymphoid Structures metabolism
- Abstract
Background: The degree of immune infiltration in tumors, especially CD8
+ T cells, greatly impacts patient disease course and response to interventional immunotherapy. Enhancement of tumor infiltrating lymphocyte (TIL) is a critical element of efficacious therapy and one that may be achieved via administration of agents that promote tumor vascular normalization (VN) and/or induce the development of tertiary lymphoid structures (TLS) within the tumor microenvironment (TME)., Methods: Low-dose stimulator of interferon genes (STING) agonist ADU S-100 (5 µg/mouse) was delivered intratumorally to established subcutaneous B16.F10 melanomas on days 10, 14 and 17 post-tumor inoculation. Treated and control tumors were isolated at various time points to assess transcriptional changes associated with VN and TLS formation via quantitative PCR (qPCR), with corollary immune cell composition changes in isolated tissues determined using flow cytometry and immunofluorescence microscopy. In vitro assays were performed on CD11c+ BMDCs treated with 2.5 µg/mL ADU S-100 or CD11c+ DCs isolated from tumor digests and associated transcriptional changes analyzed via qPCR or profiled using DNA microarrays. For T cell repertoireβ-CDR3 analyses, T cell CDR3 was sequenced from gDNA isolated from splenocytes and enzymatically digested tumors., Results: We report that activation of STING within the TME leads to slowed melanoma growth in association with increased production of antiangiogenic factors including Tnfsf15 (Vegi) and Cxcl10, and TLS-inducing factors including Ccl19 , Ccl21 , Lta , Ltb and Light . Therapeutic responses resulting from intratumoral STING activation were characterized by improved VN, enhanced tumor infiltration by CD8+ T cells and CD11c+ DCs and local TLS neogenesis, all of which were dependent on host expression of STING. Consistent with a central role for DC in TLS formation, ADU S-100-activated mCD11c+ DCs also exhibited upregulated expression of TLS promoting factors including lymphotoxin-α (LTA), interleukin (IL)-36, inflammatory chemokines and type I interferons in vitro and in vivo. TLS formation in ADU S-100-treated mice was associated with the development of a highly oligoclonal TIL repertoire enriched in expanded T cell clonotypes unique to the TME and not detected in the periphery., Conclusions: Our data support the premise that i.t. delivery of low-dose STING agonist promotes VN and a proinflammatory TME supportive of TLS formation, enrichment in the TIL repertoire and tumor growth control., 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
29. Skin immunization for effective treatment of multifocal melanoma refractory to PD1 blockade and Braf inhibitors.
- Author
-
Hao X, Falo Iii LD, Chen G, Zhang J, Carey CD, Storkus WJ, Falo LD Jr, and You Z
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, Drug Synergism, Female, Humans, Immune Checkpoint Inhibitors therapeutic use, Immunization, Intramolecular Oxidoreductases genetics, Intramolecular Oxidoreductases immunology, Male, Melanoma genetics, Melanoma immunology, Mice, Mutation, PTEN Phosphohydrolase genetics, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Proto-Oncogene Proteins B-raf genetics, Tamoxifen adverse effects, Tamoxifen analogs & derivatives, Treatment Outcome, Vaccines, DNA genetics, Vaccines, DNA immunology, Xenograft Model Antitumor Assays, Drug Resistance, Neoplasm drug effects, Immune Checkpoint Inhibitors administration & dosage, Intramolecular Oxidoreductases administration & dosage, Melanoma drug therapy, Protein Kinase Inhibitors administration & dosage, Vaccines, DNA administration & dosage
- Abstract
Background: Despite the remarkable benefits associated with the interventional treatment of melanomas (and other solid cancers) with immune checkpoint and Braf inhibitors (Brafi), most patients ultimately progress on therapy. The presence of multifocal/disseminated disease in patients increases their mortality risk. Hence, the development of novel strategies to effectively treat patients with melanomas that are resistant to anti-PD1 mAb (αPD1) and/or Brafi, particularly those with multifocal/disseminated disease remains a major unmet clinical need., Methods: Mice developing induced/spontaneous Braf
V600E /Pten-/- melanomas were treated by cutaneous immunization with a DNA vaccine encoding the melanoma-associated antigen TRP2, with Brafi or αPD1 alone, or with a combination of these treatments. Tumor progression, tumor-infiltration by CD4+ and CD8+ T cells, and the development of TRP2-specific CD8+ T cells were then monitored over time., Results: Vaccination led to durable antitumor immunity against PD1/Brafi-resistant melanomas in both single lesion and multifocal disease models, and it sensitized PD1-resistant melanomas to salvage therapy with αPD1. The therapeutic efficacy of the vaccine was associated with host skin-resident cells, the induction of a systemic, broadly reactive IFNγ+ CD8+ T cell repertoire, increased frequencies of CD8+ TIL and reduced levels of PD1hi/int CD8+ T cells. Extended survival was associated with improved TIL functionality, exemplified by the presence of enhanced levels of IFNγ+ CD8+ TIL and IL2+ CD4+ TIL., Conclusions: These data support the use of a novel genetic vaccine for the effective treatment of localized or multifocal melanoma refractory to conventional αPD1-based and/or Brafi-based (immune)therapy., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
30. Dysregulated NF-κB-Dependent ICOSL Expression in Human Dendritic Cell Vaccines Impairs T-cell Responses in Patients with Melanoma.
- Author
-
Maurer DM, Adamik J, Santos PM, Shi J, Shurin MR, Kirkwood JM, Storkus WJ, and Butterfield LH
- Subjects
- Adult, Aged, Aged, 80 and over, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Cancer Vaccines administration & dosage, Cancer Vaccines genetics, Combined Modality Therapy methods, Dendritic Cells immunology, Dendritic Cells metabolism, Female, Humans, Immunogenicity, Vaccine, Inducible T-Cell Co-Stimulator Ligand genetics, Interferon-alpha administration & dosage, Male, Melanoma immunology, Melanoma mortality, Middle Aged, NF-kappa B p50 Subunit genetics, Prognosis, Progression-Free Survival, Signal Transduction drug effects, Skin Neoplasms immunology, Skin Neoplasms mortality, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Cancer Vaccines immunology, Dendritic Cells transplantation, Inducible T-Cell Co-Stimulator Ligand metabolism, Melanoma therapy, NF-kappa B p50 Subunit metabolism, Skin Neoplasms therapy
- Abstract
Therapeutic cancer vaccines targeting melanoma-associated antigens are commonly immunogenic but are rarely effective in promoting objective clinical responses. To identify critical molecules for activation of effective antitumor immunity, we have profiled autologous dendritic cell (DC) vaccines used to treat 35 patients with melanoma. We showed that checkpoint molecules induced by ex vivo maturation correlated with in vivo DC vaccine activity. Melanoma patient DCs had reduced expression of cell surface inducible T-cell costimulator ligand (ICOSL) and had defective intrinsic NF-κB signaling. Chromatin immunoprecipitation assays revealed NF-κB-dependent transcriptional regulation of ICOSL expression by DCs. Blockade of ICOSL on DCs reduced priming of antigen-specific CD8
+ and CD4+ T cells from naïve donors in vitro Concentration of extracellular/soluble ICOSL released from vaccine DCs positively correlated with patient clinical outcomes, which we showed to be partially regulated by ADAM10/17 sheddase activity. These data point to the critical role of canonical NF-κB signaling, the regulation of matrix metalloproteinases, and DC-derived ICOSL in the specific priming of cognate T-cell responses in the cancer setting. This study supports the implementation of targeted strategies to augment these pathways for improved immunotherapeutic outcomes in patients with cancer., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
31. Actin-binding protein profilin1 promotes aggressiveness of clear-cell renal cell carcinoma cells.
- Author
-
Allen A, Gau D, Francoeur P, Sturm J, Wang Y, Martin R, Maranchie J, Duensing A, Kaczorowski A, Duensing S, Wu L, Lotze MT, Koes D, Storkus WJ, and Roy P
- Subjects
- Actins antagonists & inhibitors, Actins metabolism, Animals, CapZ Actin Capping Protein genetics, CapZ Actin Capping Protein metabolism, Carcinoma, Renal Cell metabolism, Cell Line, Tumor, Cell Movement, Cell Proliferation, Cofilin 1 genetics, Cofilin 1 metabolism, Databases, Genetic, Endothelial Cells cytology, Endothelial Cells metabolism, Humans, Kidney Neoplasms metabolism, Mice, Mice, Inbred BALB C, Profilins antagonists & inhibitors, Profilins genetics, Prognosis, RNA Interference, RNA, Small Interfering metabolism, Tumor Microenvironment, Up-Regulation, Carcinoma, Renal Cell pathology, Kidney Neoplasms pathology, Profilins metabolism
- Abstract
Clear-cell renal cell carcinoma (ccRCC), the most common subtype of renal cancer, has a poor clinical outcome. A hallmark of ccRCC is genetic loss-of-function of VHL (von Hippel-Lindau) that leads to a highly vascularized tumor microenvironment. Although many ccRCC patients initially respond to antiangiogenic therapies, virtually all develop progressive, drug-refractory disease. Given the role of dysregulated expressions of cytoskeletal and cytoskeleton-regulatory proteins in tumor progression, we performed analyses of The Cancer Genome Atlas (TCGA) transcriptome data for different classes of actin-binding proteins to demonstrate that increased mRNA expression of profilin1 (Pfn1), Arp3, cofilin1, Ena/VASP, and CapZ, is an indicator of poor prognosis in ccRCC. Focusing further on Pfn1, we performed immunohistochemistry-based classification of Pfn1 staining in tissue microarrays, which indicated Pfn1 positivity in both tumor and stromal cells; however, the vast majority of ccRCC tumors tend to be Pfn1-positive selectively in stromal cells only. This finding is further supported by evidence for dramatic transcriptional up-regulation of Pfn1 in tumor-associated vascular endothelial cells in the clinical specimens of ccRCC. In vitro studies support the importance of Pfn1 in proliferation and migration of RCC cells and in soluble Pfn1's involvement in vascular endothelial cell tumor cell cross-talk. Furthermore, proof-of-concept studies demonstrate that treatment with a novel computationally designed Pfn1-actin interaction inhibitor identified herein reduces proliferation and migration of RCC cells in vitro and RCC tumor growth in vivo Based on these findings, we propose a potentiating role for Pfn1 in promoting tumor cell aggressiveness in the setting of ccRCC., Competing Interests: Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article., (© 2020 Allen et al.)
- Published
- 2020
- Full Text
- View/download PDF
32. Neoepitopes as Difference Makers for General Cancer Vaccines?
- Author
-
Filderman JN and Storkus WJ
- Subjects
- Antigens, Neoplasm genetics, Humans, Immunotherapy, Mutation, Cancer Vaccines, Neoplasms therapy
- Abstract
The cancer mutanome has been associated with disease prognosis as well as response to interventional immunotherapy and provides a substrate for development of personalized vaccines targeting tumor neoepitopes. Recent findings suggest that neoantigen-based vaccines may represent general interventional approaches for patients with solid cancers, regardless of their inherent mutational burden. See related article by Fang et al., p. 4511 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
33. Bi- and Tri-Specific T Cell Engager-Armed Oncolytic Viruses: Next-Generation Cancer Immunotherapy.
- Author
-
Guo ZS, Lotze MT, Zhu Z, Storkus WJ, and Song XT
- Abstract
Oncolytic viruses (OVs) are potent anti-cancer biologics with a bright future, having substantial evidence of efficacy in patients with cancer. Bi- and tri-specific antibodies targeting tumor antigens and capable of activating T cell receptor signaling have also shown great promise in cancer immunotherapy. In a cutting-edge strategy, investigators have incorporated the two independent anti-cancer modalities, transforming them into bi- or tri-specific T cell engager (BiTE or TriTE)-armed OVs for targeted immunotherapy. Since 2014, multiple research teams have studied this combinatorial strategy, and it showed substantial efficacy in various tumor models. Here, we first provide a brief overview of the current status of oncolytic virotherapy and the use of multi-specific antibodies for cancer immunotherapy. We then summarize progress on BiTE and TriTE antibodies as a novel class of cancer therapeutics in preclinical and clinical studies, followed by a discussion of BiTE- or TriTE-armed OVs for cancer therapy in translational models. In addition, T cell receptor mimics (TCRm) have been developed into BiTEs and are expected to greatly expand the application of BiTEs and BiTE-armed OVs for the effective targeting of intracellular tumor antigens. Future applications of such innovative combination strategies are emerging as precision cancer immunotherapies.
- Published
- 2020
- Full Text
- View/download PDF
34. Tumor-derived exosomes promote carcinogenesis of murine oral squamous cell carcinoma.
- Author
-
Razzo BM, Ludwig N, Hong CS, Sharma P, Fabian KP, Fecek RJ, Storkus WJ, and Whiteside TL
- Subjects
- 4-Nitroquinoline-1-oxide toxicity, Animals, Apoptosis, B7-H1 Antigen metabolism, Carcinogenesis chemically induced, Carcinogenesis metabolism, Carcinogens toxicity, Carcinoma, Squamous Cell chemically induced, Carcinoma, Squamous Cell metabolism, Exosomes drug effects, Exosomes metabolism, Female, Humans, Mice, Mice, Inbred C57BL, Mouth Neoplasms chemically induced, Mouth Neoplasms metabolism, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Carcinogenesis pathology, Carcinoma, Squamous Cell pathology, Cell Proliferation, Exosomes pathology, Mouth Neoplasms pathology
- Abstract
Circulating tumor-derived exosomes (TEX) interact with a variety of cells in cancer-bearing hosts, leading to cellular reprogramming which promotes disease progression. To study TEX effects on the development of solid tumors, immunosuppressive exosomes carrying PD-L1 and FasL were isolated from supernatants of murine or human HNSCC cell lines. TEX were delivered (IV) to immunocompetent C57BL/6 mice bearing premalignant oral/esophageal lesions induced by the carcinogen, 4-nitroquinoline 1-oxide (4NQO). Progression of the premalignant oropharyngeal lesions to malignant tumors was monitored. A single TEX injection increased the number of developing tumors (6.2 versus 3.2 in control mice injected with phosphate-buffered saline; P < 0.0002) and overall tumor burden per mouse (P < 0.037). The numbers of CD4+ and CD8+ T lymphocytes infiltrating the developing tumors were coordinately reduced (P < 0.01) in mice injected with SCCVII-derived TEX relative to controls. Notably, TEX isolated from mouse or human tumors had similar effects on tumor development and immune cells. A single IV injection of TEX was sufficient to condition mice harboring premalignant OSCC lesions for accelerated tumor progression in concert with reduced immune cell migration to the tumor., (© The Author(s) 2019. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
35. Tumor Arrests DN2 to DN3 Pro T Cell Transition and Promotes Its Conversion to Thymic Dendritic Cells by Reciprocally Regulating Notch1 and Ikaros Signaling.
- Author
-
Guha I, Bhuniya A, Shukla D, Patidar A, Nandi P, Saha A, Dasgupta S, Ganguly N, Ghosh S, Nair A, Majumdar S, Saha B, Storkus WJ, Baral R, and Bose A
- Subjects
- Animals, Cell Cycle Checkpoints, Cell Differentiation, Cell Line, Tumor, Cell Proliferation, Gene Expression Regulation, Neoplastic, Humans, Ikaros Transcription Factor genetics, Interleukin-10 genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptor, Notch1 genetics, Signal Transduction, Dendritic Cells physiology, Ikaros Transcription Factor metabolism, Lymphoid Progenitor Cells physiology, Receptor, Notch1 metabolism, Sarcoma immunology, Skin Neoplasms immunology, T-Lymphocytes physiology, Thymus Gland cytology
- Abstract
Tumor progression in the host leads to severe impairment of intrathymic T-cell differentiation/maturation, leading to the paralysis of cellular anti-tumor immunity. Such suppression manifests the erosion of CD4
+ CD8+ double-positive (DP) immature thymocytes and a gradual increase in CD4- CD8- double negative (DN) early T-cell progenitors. The impact of such changes on the T-cell progenitor pool in the context of cancer remains poorly investigated. Here, we show that tumor progression blocks the transition of Lin- Thy1.2+ CD25+ CD44+ c-Kitlow DN2b to Lin- Thy1.2+ CD25+ CD44- c-Kit- DN3 in T-cell maturation, instead leading to DN2-T-cell differentiation into dendritic cells (DC). We observed that thymic IL-10 expression is upregulated, particularly at cortico-medullary junctions (CMJ), under conditions of progressive disease, resulting in the termination of IL-10Rhigh DN2-T-cell maturation due to dysregulated expression of Notch1 and its target, CCR7 (thus restricting these cells to the CMJ). Intrathymic differentiation of T-cell precursors in IL-10-/- mice and in vitro fetal thymic organ cultures revealed that IL-10 promotes the interaction between thymic stromal cells and Notch1low DN2-T cells, thus facilitating these DN2-T cells to differentiate toward CD45+ CD11c+ MHC-II+ thymic DCs as a consequence of activating the Ikaros/IRF8 signaling axis. We conclude that a novel function of thymically-expressed IL-10 in the tumor-bearing host diverts T-cell differentiation toward a DC pathway, thus limiting the protective adaptive immune repertoire., (Copyright © 2020 Guha, Bhuniya, Shukla, Patidar, Nandi, Saha, Dasgupta, Ganguly, Ghosh, Nair, Majumdar, Saha, Storkus, Baral and Bose.)- Published
- 2020
- Full Text
- View/download PDF
36. Single injection of IL-12 coacervate as an effective therapy against B16-F10 melanoma in mice.
- Author
-
Hwang MP, Fecek RJ, Qin T, Storkus WJ, and Wang Y
- Subjects
- Animals, Interleukin-12, Killer Cells, Natural, Mice, Mice, Inbred C57BL, Tumor Microenvironment, Melanoma, Experimental drug therapy, Skin Neoplasms drug therapy
- Abstract
Melanoma is the deadliest type of skin cancer with one of the fastest increasing incidence rates among solid tumors. The use of checkpoint inhibitors (e.g. αPD-1 antibody) has recently emerged as a viable alternative to conventional modes of therapy. However, increasing evidence points towards the need for a tumor priming step to improve intratumoral immune cell infiltration. IL-12 is an immune-activating cytokine with such potential and was explored in earlier clinical trials as a highly concentrated systemic infusion. This unfortunately led to severe adverse effects. From this perspective, the localization and gradual release of such a potent immunotherapeutic agent in the tumor microenvironment is desired. This manuscript reports the use of a heparin-based complex coacervate to deliver IL-12, in which heparin-binding motifs on IL-12 allow for its effective encapsulation. IL-12-encapsulated complex coacervates significantly improved the bioactivity of IL-12 and provided protection from proteolytic cleavage in-vitro. Indeed, a single injection of IL-12 coacervate significantly inhibits the in-vivo growth of treated and untreated, contralateral tumor growth in a syngeneic B16F10 mouse melanoma model. Furthermore, tumors in mice receiving IL-12 complex coacervate treatment displayed increased infiltration by natural killer (NK) cells and CD8α
+ T cells, and a decreased presence of CD4+ Foxp3+ regulatory T cells. This study provides proof-of-concept data supporting the use of complex coacervates for sustained delivery of immunostimulatory proteins as an effective therapeutic strategy against disseminated tumors., (Copyright © 2019 Elsevier B.V. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
37. IL-36 Signaling in the Tumor Microenvironment.
- Author
-
Chelvanambi M, Weinstein AM, and Storkus WJ
- Subjects
- Animals, Dendritic Cells immunology, Humans, T-Lymphocytes immunology, Interleukin-1 immunology, Interleukin-1 metabolism, Neoplasms immunology, Tumor Microenvironment
- Abstract
The ability of the immune system to prevent or control the growth of tumor cells is critically dependent on inflammatory processes that lead to the activation, expansion, and recruitment of antitumor effector cells into the tumor microenvironment (TME). These processes are orchestrated by soluble cytokines produced in tissues that alarm local immune surveillance cells (such as dendritic cells, DCs) to mobilize protective antitumor immune populations (B cells, T cells). The interleukin (IL)-36 family of pro-inflammatory cytokines plays an important role in multiple disease processes, ranging from an instigator of autoimmune psoriasis to an initiator of therapeutic immune responses against tumor cells. This chapter will focus on the biologic role of immunomodulatory IL-36 family cytokines in the cancer setting and their potential utility in the design of effective interventional therapies. (127 words).
- Published
- 2020
- Full Text
- View/download PDF
38. Impact of combination immunochemotherapies on progression of 4NQO-induced murine oral squamous cell carcinoma.
- Author
-
Ludwig S, Hong CS, Razzo BM, Fabian KPL, Chelvanambi M, Lang S, Storkus WJ, and Whiteside TL
- Subjects
- 4-Nitroquinoline-1-oxide toxicity, Animals, Cisplatin therapeutic use, Combined Modality Therapy methods, Female, Mice, Mice, Inbred C57BL, Mouth Neoplasms chemically induced, Mouth Neoplasms immunology, Mouth Neoplasms pathology, Neoplasms, Experimental chemically induced, Neoplasms, Experimental immunology, Neoplasms, Experimental pathology, Squamous Cell Carcinoma of Head and Neck chemically induced, Squamous Cell Carcinoma of Head and Neck immunology, Squamous Cell Carcinoma of Head and Neck pathology, Treatment Outcome, Triazines therapeutic use, Triazoles therapeutic use, Vaccines, Subunit therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cancer Vaccines therapeutic use, Immunotherapy methods, Mouth Neoplasms therapy, Neoplasms, Experimental therapy, Squamous Cell Carcinoma of Head and Neck therapy
- Abstract
Advanced oral squamous cell carcinomas (OSCC) have limited therapeutic options. Although immune therapies are emerging as a potentially effective alternative or adjunct to chemotherapies, the therapeutic efficacy of combination immune chemotherapies has yet to be determined. Using a 4-nitroquinolone-N-oxide (4NQO) orthotopic model of OSCC in immunocompetent mice, we evaluated the therapeutic efficacy of single- and combined-agent treatment with a poly-epitope tumor peptide vaccine, cisplatin and/or an A
2A R inhibitor, ZM241385. The monotherapies or their combinations resulted in a partial inhibition of tumor growth and, in some cases, a significant but transient upregulation of systemic anti-tumor CD8+ T cell responses. These responses eroded in the face of expanding immunoregulatory cell populations at later stages of tumor progression. Our findings support the need for the further development of combinatorial therapeutic approaches that could more effectively silence dominant immune inhibitory pathways operating in OSCC and provide novel, more beneficial treatment options for this tumor.- Published
- 2019
- Full Text
- View/download PDF
39. Inhibiting Autophagy in Renal Cell Cancer and the Associated Tumor Endothelium.
- Author
-
Russell KL, Gorgulho CM, Allen A, Vakaki M, Wang Y, Facciabene A, Lee D, Roy P, Buchser WJ, Appleman LJ, Maranchie J, Storkus WJ, and Lotze MT
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Biomarkers, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell etiology, Carcinoma, Renal Cell pathology, Clinical Trials as Topic, Cytoskeleton metabolism, Disease Susceptibility, Drug Discovery, Drug Screening Assays, Antitumor, Endothelium drug effects, Endothelium pathology, High-Throughput Nucleotide Sequencing, Humans, Kidney Neoplasms drug therapy, Kidney Neoplasms etiology, Kidney Neoplasms pathology, Mitochondria drug effects, Mitochondria genetics, Mitochondria metabolism, Mitophagy drug effects, Mitophagy genetics, Molecular Imaging, Molecular Targeted Therapy, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic genetics, Neovascularization, Pathologic metabolism, Von Hippel-Lindau Tumor Suppressor Protein genetics, Von Hippel-Lindau Tumor Suppressor Protein metabolism, Autophagy drug effects, Autophagy genetics, Carcinoma, Renal Cell metabolism, Endothelium metabolism, Kidney Neoplasms metabolism
- Abstract
The clear cell subtype of kidney cancer encompasses most renal cell carcinoma cases and is associated with the loss of von Hippel-Lindau gene function or expression. Subsequent loss or mutation of the other allele influences cellular stress responses involving nutrient and hypoxia sensing. Autophagy is an important regulatory process promoting the disposal of unnecessary or degraded cellular components, tightly linked to almost all cellular processes. Organelles and proteins that become damaged or that are no longer needed in the cell are sequestered and digested in autophagosomes upon fusing with lysosomes, or alternatively, released via vesicular exocytosis. Tumor development tends to disrupt the regulation of the balance between this process and apoptosis, permitting prolonged cell survival and increased replication. Completed trials of autophagic inhibitors using hydroxychloroquine in combination with other anticancer agents including rapalogues and high-dose interleukin 2 have now been reported. The complex nature of autophagy and the unique biology of clear cell renal cell carcinoma warrant further understanding to better develop the next generation of relevant anticancer agents.
- Published
- 2019
- Full Text
- View/download PDF
40. Vaccinia virus-mediated cancer immunotherapy: cancer vaccines and oncolytics.
- Author
-
Guo ZS, Lu B, Guo Z, Giehl E, Feist M, Dai E, Liu W, Storkus WJ, He Y, Liu Z, and Bartlett DL
- Subjects
- Animals, Cancer Vaccines administration & dosage, Cancer Vaccines genetics, Cancer Vaccines immunology, Clinical Studies as Topic, Dendritic Cells immunology, Dendritic Cells metabolism, Genetic Engineering, Genetic Therapy, Genetic Vectors administration & dosage, Host-Pathogen Interactions immunology, Humans, Membrane Glycoproteins administration & dosage, Membrane Glycoproteins immunology, Neoplasms genetics, Neoplasms immunology, Oncolytic Viruses physiology, T-Lymphocytes immunology, T-Lymphocytes metabolism, Vaccinia virus physiology, Genetic Vectors genetics, Immunotherapy, Neoplasms therapy, Oncolytic Virotherapy, Oncolytic Viruses genetics, Vaccinia virus genetics
- Abstract
Cancer vaccines and oncolytic immunotherapy are promising treatment strategies with potential to provide greater clinical benefit to patients with advanced-stage cancer. In particular, recombinant vaccinia viruses (VV) hold great promise as interventional agents. In this article, we first summarize the current understanding of virus biology and viral genes involved in host-virus interactions to further improve the utility of these agents in therapeutic applications. We then discuss recent findings from basic and clinical studies using VV as cancer vaccines and oncolytic immunotherapies. Despite encouraging results gleaned from translational studies in animal models, clinical trials implementing VV vectors alone as cancer vaccines have yielded largely disappointing results. However, the combination of VV vaccines with alternate forms of standard therapies has resulted in superior clinical efficacy. For instance, combination regimens using TG4010 (MVA-MUC1-IL2) with first-line chemotherapy in advanced-stage non-small cell lung cancer or combining PANVAC with docetaxel in the setting of metastatic breast cancer have clearly provided enhanced clinical benefits to patients. Another novel cancer vaccine approach is to stimulate anti-tumor immunity via STING activation in Batf3-dependent dendritic cells (DC) through the use of replication-attenuated VV vectors. Oncolytic VVs have now been engineered for improved safety and superior therapeutic efficacy by arming them with immune-stimulatory genes or pro-apoptotic molecules to facilitate tumor immunogenic cell death, leading to enhanced DC-mediated cross-priming of T cells recognizing tumor antigens, including neoantigens. Encouraging translational and early phase clinical results with Pexa-Vec have matured into an ongoing global phase III trial for patients with hepatocellular carcinoma. Combinatorial approaches, most notably those using immune checkpoint blockade, have produced exciting pre-clinical results and warrant the development of innovative clinical studies. Finally, we discuss major hurdles that remain in the field and offer some perspectives regarding the development of next generation VV vectors for use as cancer therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
41. Association of IL-36γ with tertiary lymphoid structures and inflammatory immune infiltrates in human colorectal cancer.
- Author
-
Weinstein AM, Giraldo NA, Petitprez F, Julie C, Lacroix L, Peschaud F, Emile JF, Marisa L, Fridman WH, Storkus WJ, and Sautès-Fridman C
- Subjects
- Aged, Aged, 80 and over, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic immunology, Humans, Interleukin-1 metabolism, Male, Middle Aged, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Colorectal Neoplasms immunology, Inflammation immunology, Interleukin-1 immunology, Tertiary Lymphoid Structures immunology
- Abstract
IL-1 family cytokines play a dual role in the gut, with different family members contributing either protective or pathogenic effects. IL-36γ is an IL-1 family cytokine involved in polarizing type-1 immune responses. However, its function in the gut, including in colorectal cancer pathogenesis, is not well appreciated. In a murine model of colon carcinoma, IL-36γ controls tertiary lymphoid structure formation and promotes a type-1 immune response concurrently with a decrease in expression of immune checkpoint molecules in the tumor microenvironment. Here, we demonstrate that IL-36γ plays a similar role in driving a pro-inflammatory phenotype in human colorectal cancer. We analyzed a cohort of 33 primary colorectal carcinoma tumors using imaging, flow cytometry, and transcriptomics to determine the pattern and role of IL-36γ expression in this disease. In the colorectal tumor microenvironment, we observed IL-36γ to be predominantly expressed by M1 macrophages and cells of the vasculature, including smooth muscle cells and high endothelial venules. This pattern of IL-36γ expression is associated with a CD4
+ central memory T cell infiltrate and an increased density of B cells in tertiary lymphoid structures, as well as with markers of fibrosis. Conversely, expression of the antagonist to IL-36 signaling, IL-1F5, was associated with intratumoral expression of checkpoint molecules, including PD-1, PD-L1, and CTLA4, which can suppress the immune response. These data support a role for IL-36γ in the physiologic immune response to colorectal cancer by sustaining inflammation within the tumor microenvironment.- Published
- 2019
- Full Text
- View/download PDF
42. Combined VLA-4-Targeted Radionuclide Therapy and Immunotherapy in a Mouse Model of Melanoma.
- Author
-
Choi J, Beaino W, Fecek RJ, Fabian KPL, Laymon CM, Kurland BF, Storkus WJ, and Anderson CJ
- Subjects
- Animals, B7-H1 Antigen antagonists & inhibitors, CTLA-4 Antigen antagonists & inhibitors, Cell Line, Tumor, Dipeptides chemistry, Female, Gallium Radioisotopes, Heterocyclic Compounds, 1-Ring chemistry, Humans, Lutetium pharmacokinetics, Male, Melanoma, Experimental diagnostic imaging, Mice, Mice, Inbred C57BL, Molecular Targeted Therapy methods, Phenylurea Compounds chemistry, Polyethylene Glycols chemistry, Programmed Cell Death 1 Receptor antagonists & inhibitors, Radioisotopes pharmacokinetics, Radiopharmaceuticals chemistry, Radiopharmaceuticals pharmacokinetics, Radiotherapy Dosage, Tumor Protein, Translationally-Controlled 1, Integrin alpha4beta1 antagonists & inhibitors, Lutetium therapeutic use, Melanoma, Experimental immunology, Melanoma, Experimental radiotherapy, Radioimmunotherapy methods, Radioisotopes therapeutic use, Radiopharmaceuticals therapeutic use
- Abstract
Very late antigen-4 (VLA-4; also known as integrin α
4 β1 ) is expressed at high levels in aggressive and metastatic melanoma tumors and may provide an ideal target for imaging and targeted radionuclide therapy (TRT).177 Lu-DOTA-PEG4 -LLP2A (177 Lu-LLP2A) is a TRT that shows high affinity for VLA-4 and high uptake in B16F10 mouse melanoma tumors in vivo. Here, we report efficacy studies of177 Lu-LLP2A, alone and combined with immune checkpoint inhibitors (ICIs) (anti-PD-1, anti-PD-L1, and anti-CTLA-4 antibodies), in B16F10 tumor-bearing mice. Methods: Tumor cells (1 × 106 ) were implanted subcutaneously in C57BL/6 mice. After 8-10 d, the mice were randomized into 8 groups.177 Lu-LLP2A was injected intravenously on day 8 or 9 (single dose), and ICI antibodies were administered intraperitoneally in 3 doses. Tumor growth was monitored over time via calipers. Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining for apoptosis was performed on fixed tumors. In a separate study, Cy3-LLP2A or Cy3-scrambled LLP2A was injected in tumor-bearing mice, and tumors were collected 4 h after injection and then analyzed by flow cytometry and immunofluorescence microscopy using different immune cell markers. Results: TRT alone showed efficacy comparable to the dual-ICI anti-PD-1 + anti-CTLA-4 or anti-PD-L1 + anti-CTLA-4, whereas TRT + ICIs significantly enhanced survival. TUNEL staining showed that the highest levels of apoptosis were in the TRT + ICI groups. In addition to targeting tumor cells, TRT also bound immune cells in the tumor microenvironment. Flow cytometry data showed that the tumors consisted of about 77% tumor cells and fibroblasts (CD45-negative/CD49d-positive) and about 23% immune cells (CD45-positive/CD49d-positive) and that immune cells expressed higher levels of VLA-4. Cy3-LLP2A and CD49d colocalized with macrophages (CD68), T cells (CD8, CD4), and B cells (CD19). Immunohistochemical analysis identified a significant colocalization of Cy3-LLP2A and CD68. Conclusion: Combination treatment with TRT + ICIs targets both tumor cells and immune cells and has potential as a therapeutic agent in patients with metastatic melanoma., (© 2018 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2018
- Full Text
- View/download PDF
43. Tumor-Derived α-Fetoprotein Directly Drives Human Natural Killer-Cell Activation and Subsequent Cell Death.
- Author
-
Vujanovic L, Stahl EC, Pardee AD, Geller DA, Tsung A, Watkins SC, Gibson GA, Storkus WJ, and Butterfield LH
- Subjects
- Cell Death, Cell Line, Tumor, Cell Proliferation, Cells, Cultured, Cytokines immunology, Humans, Carcinoma, Hepatocellular immunology, Killer Cells, Natural immunology, Liver Neoplasms immunology, alpha-Fetoproteins immunology
- Abstract
Hepatocellular carcinoma (HCC) patients with reduced natural killer (NK)-cell numbers and function have been shown to have a poor disease outcome. Mechanisms underlying NK-cell deficiency and dysfunction in HCC patients remain largely unresolved. α-Fetoprotein (AFP) is an oncofetal antigen produced by HCC. Previous studies demonstrated that tumor-derived AFP (tAFP) can indirectly impair NK-cell activity by suppressing dendritic cell function. However, a direct tAFP effect on NK cells remains unexplored. The purpose of this study was to examine the ability of cord blood-derived AFP (nAFP) and that of tAFP to directly modulate human NK-cell activity and longevity in vitro Short-term exposure to tAFP and, especially, nAFP proteins induced a unique proinflammatory, IL2-hyperresponsive phenotype in NK cells as measured by IL1β, IL6, and TNF secretion, CD69 upregulation, and enhanced tumor cell killing. In contrast, extended coculture with tAFP, but not nAFP, negatively affected long-term NK-cell viability. NK-cell activation was directly mediated by the AFP protein itself, whereas their viability was affected by hydrophilic components within the low molecular mass cargo that copurified with tAFP. Identification of the distinct impact of circulating tAFP on NK-cell function and viability may be crucial to developing a strategy to ameliorate HCC patient NK-cell functional deficits. Cancer Immunol Res; 5(6); 493-502. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
44. Oncolytic Immunotherapy: Conceptual Evolution, Current Strategies, and Future Perspectives.
- Author
-
Guo ZS, Liu Z, Kowalsky S, Feist M, Kalinski P, Lu B, Storkus WJ, and Bartlett DL
- Abstract
The concept of oncolytic virus (OV)-mediated cancer therapy has been shifted from an operational virotherapy paradigm to an immunotherapy. OVs often induce immunogenic cell death (ICD) of cancer cells, and they may interact directly with immune cells as well to prime antitumor immunity. We and others have developed a number of strategies to further stimulate antitumor immunity and to productively modulate the tumor microenvironment (TME) for potent and sustained antitumor immune cell activity. First, OVs have been engineered or combined with other ICD inducers to promote more effective T cell cross-priming, and in many cases, the breaking of functional immune tolerance. Second, OVs may be armed to express Th1-stimulatory cytokines/chemokines or costimulators to recruit and sustain the potent antitumor immunity into the TME to focus their therapeutic activity within the sites of disease. Third, combinations of OV with immunomodulatory drugs or antibodies that recondition the TME have proven to be highly promising in early studies. Fourth, combinations of OVs with other immunotherapeutic regimens (such as prime-boost cancer vaccines, CAR T cells; armed with bispecific T-cell engagers) have also yielded promising preliminary findings. Finally, OVs have been combined with immune checkpoint blockade, with robust antitumor efficacy being observed in pilot evaluations. Despite some expected hurdles for the rapid translation of OV-based state-of-the-art protocols, we believe that a cohort of these novel approaches will join the repertoire of standard cancer treatment options in the near future.
- Published
- 2017
- Full Text
- View/download PDF
45. Tbet and IL-36γ cooperate in therapeutic DC-mediated promotion of ectopic lymphoid organogenesis in the tumor microenvironment.
- Author
-
Weinstein AM, Chen L, Brzana EA, Patil PR, Taylor JL, Fabian KL, Wallace CT, Jones SD, Watkins SC, Lu B, Stroncek DF, Denning TL, Fu YX, Cohen PA, and Storkus WJ
- Abstract
We have previously reported that direct injection of dendritic cells (DC) engineered to express the Type-1 transactivator Tbet (i.e., DC.Tbet) into murine tumors results in antitumor efficacy in association with the development of structures resembling tertiary lymphoid organs (TLO) in the tumor microenvironment (TME). These TLO contained robust infiltrates of B cells, DC, NK cells, and T cells in proximity to PNAd
+ blood vessels; however, they were considered incomplete, since the recruited B cells failed to organize into classic germinal center-like structures. We now report that antitumor efficacy and TLO-inducing capacity of DC.Tbet-based i.t. therapy is operational in peripheral lymph node-deficient LTA-/- mice, and that it is highly dependent upon a direct Tbet target gene product, IL-36γ/IL-1F9. Intratumoral DC.Tbet fails to provide protection to tumor-bearing IL-36R-/- hosts, or to tumor-bearing wild-type recipient mice co-administered rmIL-1F5/IL-36RN, a natural IL-36R antagonist. Remarkably, the injection of tumors with DC engineered to secrete a bioactive form of mIL-36γ (DC.IL36γ) also initiated therapeutic TLO and slowed tumor progression in vivo . Furthermore, DC.IL36γ cells strongly upregulated their expression of Tbet, suggesting that Tbet and IL-36γ cooperate to reinforce each other's expression in DC, rendering them competent to promote TLO formation in an "immunologically normalized," therapeutic TME.- Published
- 2017
- Full Text
- View/download PDF
46. Intratumoral delivery of tumor antigen-loaded DC and tumor-primed CD4 + T cells combined with agonist α-GITR mAb promotes durable CD8 + T-cell-dependent antitumor immunity.
- Author
-
Liu Z, Hao X, Zhang Y, Zhang J, Carey CD, Falo LD Jr, Storkus WJ, and You Z
- Abstract
The progressive tumor microenvironment (TME) coordinately supports tumor cell expansion and metastasis, while it antagonizes the survival and (poly-)functionality of antitumor T effector cells. There remains a clear need to develop novel therapeutic strategies that can transform the TME into a pro-inflammatory niche that recruits and sustains protective immune cell populations. While intravenous treatment with tumor-primed CD4
+ T cells combined with intraperitoneal delivery of agonist anti-glucocorticoid-induced TNF receptor (α-GITR) mAb results in objective antitumor responses in murine early stage disease models, this approach is ineffective against more advanced tumors. Further subcutaneous co-administration of a vaccine consisting of tumor antigen-loaded dendritic cells (DC) failed to improve the antitumor efficacy of this approach. Remarkably, these same three therapeutic agents elicited significant antitumor benefits when the antitumor CD4+ T cells and tumor antigen-loaded DC were co-injected directly into tumors along with intratumoral or intraperitoneal delivery of α-GITR mAb. This latter protocol induced the production of an array of antitumor cytokines and chemokines within the TME, supporting increased tumor-infiltration by antitumor CD8+ T cells capable of mediating tumor regression and extended overall survival.- Published
- 2017
- Full Text
- View/download PDF
47. Therapeutic efficacy of combined vaccination against tumor pericyte-associated antigens DLK1 and DLK2 in mice.
- Author
-
Fabian KP, Chi-Sabins N, Taylor JL, Fecek R, Weinstein A, and Storkus WJ
- Abstract
When compared with vascular cells in normal tissues, pericytes and vascular endothelial cells (VEC) in tumor blood vessels exhibit altered morphology and epigenetic programming that leads to the expression of unique antigens that allow for differential recognition by CD8
+ T cells. We have previously shown that the Notch antagonist delta-like homolog 1 (DLK1) is a tumor pericyte-associated antigen expressed in setting of melanoma and a range of carcinomas. In this report, we show that therapeutic vaccination against DLK1 in murine models results in slowed tumor growth, but also to the compensatory expression of the DLK1 homolog, DLK2, by tumor-associated pericytes. Vaccines targeting both DLK1 and DLK2 resulted in superior antitumor benefits in association with improved activation and recruitment of antigen-specific Type 1 CD8+ T cells, reduced presence of myeloid-derived suppressive cells, T regulatory cell and tumor vascular normalization. The antitumor efficacy of vaccines coordinately targeting DLK1 and DLK2 was further improved by inclusion of PD-L1 blockade, thus defining a combination immunotherapy theoretically suitable for the treatment of a broad range of solid (vascularized) cancers.- Published
- 2017
- Full Text
- View/download PDF
48. Immunotherapeutic Targeting of Tumor-Associated Blood Vessels.
- Author
-
Fabian KL and Storkus WJ
- Subjects
- Animals, Blood Vessels pathology, Humans, Immunotherapy methods, Neoplasms blood supply, Neoplasms immunology, Neoplasms therapy, Neovascularization, Pathologic immunology, Neovascularization, Pathologic pathology, Neovascularization, Pathologic therapy
- Abstract
Pathological angiogenesis occurs during tumor progression and leads in the formation of an abnormal vasculature in the tumor microenvironment (TME). The tumor vasculature is disorganized, tortuous and leaky, resulting in high interstitial pressure and hypoxia in the TME, all of which are events that support tumor growth and survival. Given the sustaining role of the tumor vasculature, it has become an increasingly attractive target for the development of anti-cancer therapies. Antibodies, tyrosine kinase inhibitors and cancer vaccines that target pro-angiogenic factors, angiogenesis-associated receptors or tumor blood vessel-associated antigens continue to be developed and tested for therapeutic efficacy. Preferred anti-angiogenic protocols include those that "normalize" the tumor-associated vasculature which reduce hypoxia and improve tumor blood perfusion, resulting in tumor cell apoptosis, decreased immunosuppression, and enhanced effector immune cell infiltration/tumoricidal action within the TME.
- Published
- 2017
- Full Text
- View/download PDF
49. Vaccine therapy + dasatinib for the treatment of patients with stage IIIB-IV melanoma.
- Author
-
Tarhini AA, Tawbi H, and Storkus WJ
- Abstract
Competing Interests: Financial & competing interests disclosure This work was supported by NIH R01 CA169118 (WJ Storkus) and the University of Pittsburgh Melanoma and Skin Cancer Specialized Programs of Research Excellence (SPORE) NIH P50 CA121973 (A Tarhini, WJ Storkus). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.
- Published
- 2016
- Full Text
- View/download PDF
50. Tumor-associated mesenchymal stem cells inhibit naïve T cell expansion by blocking cysteine export from dendritic cells.
- Author
-
Ghosh T, Barik S, Bhuniya A, Dhar J, Dasgupta S, Ghosh S, Sarkar M, Guha I, Sarkar K, Chakrabarti P, Saha B, Storkus WJ, Baral R, and Bose A
- Subjects
- Animals, Cell Differentiation, Cell Line, Cell Proliferation, Female, Humans, Interferon-gamma metabolism, Interleukin-10 metabolism, Interleukin-2 metabolism, Lymphocyte Activation, Mesenchymal Stem Cells cytology, Mice, STAT3 Transcription Factor, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Cysteine metabolism, Dendritic Cells metabolism, Mesenchymal Stem Cells pathology
- Abstract
Mesenchymal stem cells (MSCs) represent an important cellular constituent of the tumor microenvironment, which along with tumor cells themselves, serve to regulate protective immune responses in support of progressive disease. We report that tumor MSCs prevent the ability of dendritic cells (DC) to promote naïve CD4(+) and CD8(+) T cell expansion, interferon gamma secretion and cytotoxicity against tumor cells, which are critical to immune-mediated tumor eradication. Notably, tumor MSCs fail to prevent DC-mediated early T cell activation events or the ability of responder T cells to produce IL-2. The immunoregulatory activity of tumor MSCs is IL-10- and STAT3-dependent, with STAT3 repressing DC expression of cystathionase, a critical enzyme that converts methionine-to-cysteine. Under cysteine-deficient priming conditions, naïve T cells exhibit defective cellular metabolism and proliferation. Bioinformatics analyses as well as in vitro observations suggest that STAT3 may directly bind to a GAS-like motif within the cystathionase promoter (-269 to -261) leading to IL-10-STAT3 mediated repression of cystathionase gene transcription. Our collective results provide evidence for a novel mechanism of tumor MSC-mediated T cell inhibition within tumor microenvironment., (© 2016 UICC.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.