32 results on '"Elham Beyranvand Nejad"'
Search Results
2. A third vaccination with a single T cell epitope confers protection in a murine model of SARS-CoV-2 infection
- Author
-
Iris N. Pardieck, Tetje C. van der Sluis, Esmé T. I. van der Gracht, Dominique M. B. Veerkamp, Felix M. Behr, Suzanne van Duikeren, Guillaume Beyrend, Jasper Rip, Reza Nadafi, Elham Beyranvand Nejad, Nils Mülling, Dena J. Brasem, Marcel G. M. Camps, Sebenzile K. Myeni, Peter J. Bredenbeek, Marjolein Kikkert, Yeonsu Kim, Luka Cicin-Sain, Tamim Abdelaal, Klaas P. J. M. van Gisbergen, Kees L. M. C. Franken, Jan Wouter Drijfhout, Cornelis J. M. Melief, Gerben C. M. Zondag, Ferry Ossendorp, and Ramon Arens
- Subjects
Science - Abstract
Vaccination regimens and the number of doses required for optimal immunity and protection are critical factors in the translation of vaccines. Here the authors show administration of a three dose protocol of a single T cell epitope to the SARS-CoV-2 spike protein induces a robust CD8+ T cell response and confers protection in a lethal murine challenge model of infection.
- Published
- 2022
- Full Text
- View/download PDF
3. Demarcated thresholds of tumor-specific CD8 T cells elicited by MCMV-based vaccine vectors provide robust correlates of protection
- Author
-
Elham Beyranvand Nejad, Robert B. Ratts, Eleni Panagioti, Christine Meyer, Jennifer D. Oduro, Luka Cicin-Sain, Klaus Früh, Sjoerd H. van der Burg, and Ramon Arens
- Subjects
T cells ,Cancer ,CMV-based vaccine vector ,Pre-existing immunity ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The capacity of cytomegalovirus (CMV) to elicit long-lasting strong T cell responses, and the ability to engineer the genome of this DNA virus positions CMV-based vaccine vectors highly suitable as a cancer vaccine platform. Defined immune thresholds for tumor protection and the factors affecting such thresholds have not well been investigated in cancer immunotherapy. We here determined using CMV as a vaccine platform whether critical thresholds of vaccine-specific T cell responses can be established that relate to tumor protection, and which factors control such thresholds. Methods We generated CMV-based vaccine vectors expressing the E7 epitope and tested these in preclinical models of HPV16-induced cancer. Vaccination was applied via different doses and routes (intraperitoneal (IP), subcutaneous (SC) and intranasal (IN)). The magnitude, kinetics and phenotype of the circulating tumor-specific CD8+ T cell response were determined. Mice were subsequently challenged with tumor cells, and the tumor protection was monitored. Results Immunization with CMV-based vaccines via the IP or SC route eliciting vaccine-induced CD8+ T cell responses of > 0.3% of the total circulating CD8 T cell population fully protects mice against lethal tumor challenge. However, low dose inoculations via the IP or SC route or IN vaccination elicited vaccine-induced CD8+ T cell responses that did not reach protective thresholds for tumor protection. In addition, whereas weak pre-existing immunity did not alter the protective thresholds of the vaccine-specific T cell response following subsequent immunization with CMV-based vaccine vectors, strong pre-existing immunity inhibited the development of vaccine-induced T cells and their control on tumor progression. Conclusions This study highlight the effectiveness of CMV-based vaccine vectors, and shows that demarcated thresholds of vaccine-specific T cells could be defined that correlate to tumor protection. Together, these results may hold importance for cancer vaccine development to achieve high efficacy in vaccine recipients.
- Published
- 2019
- Full Text
- View/download PDF
4. IL-6 signaling in macrophages is required for immunotherapy-driven regression of tumors
- Author
-
Sjoerd H van der Burg, Ramon Arens, Thorbald van Hall, Elham Beyranvand Nejad, Jan Willem Kleinovink, Camilla Labrie, Marit J van Elsas, Hans-Willi Mittrücker, Kees L M C Franken, Sylvia Heink, and Thomas Korn
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background High serum interleukin (IL-6) levels may cause resistance to immunotherapy by modulation of myeloid cells in the tumor microenvironment. IL-6 signaling blockade is tested in cancer, but as this inflammatory cytokine has pleiotropic effects, this treatment is not always effective.Methods IL-6 and IL-6R blockade was applied in an IL-6-mediated immunotherapy-resistant TC-1 tumor model (TC-1.IL-6) and immunotherapy-sensitive TC-1.control. Effects on therapeutic vaccination-induced tumor regression, recurrence and survival as well on T cells and myeloid cells in the tumor microenvironment were studied. The effects of IL-6 signaling in macrophages under therapy conditions were studied in Il6rafl/fl×LysMcre+ mice.Results Our therapeutic vaccination protocol elicits a strong tumor-specific CD8+ T-cell response, leading to enhanced intratumoral T-cell infiltration and recruitment of tumoricidal macrophages. Blockade of IL-6 signaling exacerbated tumor outgrowth, reflected by fewer complete regressions and more recurrences after therapeutic vaccination, especially in TC-1.IL-6 tumor-bearing mice. Early IL-6 signaling blockade partly inhibited the development of the vaccine-induced CD8+ T-cell response. However, the main mechanism was the malfunction of macrophages during therapy-induced tumor regression. Therapy efficacy was impaired in Il6rafl/fl×LysMcre+ but not cre-negative control mice, while no differences in the vaccine-induced CD8+ T-cell response were found between these mice. IL-6 signaling blockade resulted in decreased expression of suppressor of cytokine signaling 3, essential for effective M1-type function in macrophages, and increased expression of the phagocytic checkpoint molecule signal-regulatory protein alpha by macrophages.Conclusion IL-6 signaling is critical for macrophage function under circumstances of immunotherapy-induced tumor tissue destruction, in line with the acute inflammatory functions of IL-6 signaling described in infections.
- Published
- 2021
- Full Text
- View/download PDF
5. Lack of myeloid cell infiltration as an acquired resistance strategy to immunotherapy
- Author
-
Sjoerd H van der Burg, Suzanne van Duikeren, Ramon Arens, Thorbald van Hall, Ziena Abdulrahman, Elham Beyranvand Nejad, Jan Willem Kleinovink, Noel F C C de Miranda, Camilla Labrie, Marit J van Elsas, Eva Rademaker, Tetje C van der Sluis, Amina F A.S Teunisse, Aart G Jochemsen, and Jan Oosting
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Immunotherapy of cancer is successful but tumor regression often is incomplete and followed by escape. Understanding the mechanisms underlying this acquired resistance will aid the development of more effective treatments.Methods We exploited a mouse model where tumor-specific therapeutic vaccination results in tumor regression, followed by local recurrence and resistance. In depth studies on systemic, local and tumor intrinsic changes were performed with flow and mass cytometry, immunohistochemistry, transcriptomics and several perturbation studies with inhibitors or agonistic antibodies in mice. Main findings were recapitulated in vaccinated patients.Results Full tumor regression and cure of tumor-bearing mice is dependent on the magnitude of the vaccine-induced T-cell response. Recurrence of tumors did not involve classical immune escape mechanisms, such as antigen-presentation alterations, immune checkpoint expression, resistance to killing or local immune suppression. However, the recurrent tumors displayed a changed transcriptome with alterations in p53, tumor necrosis factor-α and transforming growth factor-β signaling pathways and they became immunologically cold. Remarkably, ex vivo cell-sorted recurrent tumors, directly reinjected in naïve hosts retained their resistance to vaccination despite a strong infiltration with tumor-specific CD8+ T cells, similar to that of vaccine-responsive tumors. The influx of inflammatory mature myeloid effector cells in the resistant tumors, however, was impaired and this turned out to be the underlying mechanisms as restoration of inflammatory myeloid cell infiltration reinstated the sensitivity of these refractory tumors to vaccination. Notably, impaired myeloid cell infiltration after vaccination was also associated with vaccine resistance in patients.Conclusion An immunotherapy-induced disability of tumor cells to attract innate myeloid effector cells formed a major mechanism underlying immune escape and acquired resistance. These data not only stresses the importance of myeloid effector cells during immunotherapy but also demands for new studies to harness their tumoricidal activities.
- Published
- 2020
- Full Text
- View/download PDF
6. Supplemental figure 3 from Vaccine-Induced Tumor Necrosis Factor–Producing T Cells Synergize with Cisplatin to Promote Tumor Cell Death
- Author
-
Cornelis J.M. Melief, Sjoerd H. van der Burg, Ramon Arens, Bob van de Water, Ferry Ossendorp, Marij J.P. Welters, Vincent T.H.B.M. Smit, Louis Boon, Arjen Sloots, Elham Beyranvand Nejad, Ekaterina S. Jordanova, Suzanna Huppelschoten, Suzanne van Duikeren, and Tetje C. van der Sluis
- Abstract
Supplemental figure 3) Synergistic effects between cisplatin and peptide vaccination preserved with other treatment protocol
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 2 from Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells
- Author
-
Ramon Arens, Sjoerd H. van der Burg, Cornelis J.M. Melief, Peter A. van Veelen, George M. Janssen, Hideo Yagita, Suzanne van Duikeren, Tetje C. van der Sluis, and Elham Beyranvand Nejad
- Abstract
Induction of tumor-specific T-cell responses are dependent on cisplatin treated tumor cells.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Video 2 from Vaccine-Induced Tumor Necrosis Factor–Producing T Cells Synergize with Cisplatin to Promote Tumor Cell Death
- Author
-
Cornelis J.M. Melief, Sjoerd H. van der Burg, Ramon Arens, Bob van de Water, Ferry Ossendorp, Marij J.P. Welters, Vincent T.H.B.M. Smit, Louis Boon, Arjen Sloots, Elham Beyranvand Nejad, Ekaterina S. Jordanova, Suzanna Huppelschoten, Suzanne van Duikeren, and Tetje C. van der Sluis
- Abstract
Supplemental Video 2. TC-1 cells were in vitro cultured and treated with cisplatin (2 μg/ml). Cell death was followed for 22 hours using Annexin-V staining and automated imaging on a BD pathway 855 imager. Every hour a picture was taken.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplemental figure 2 from Vaccine-Induced Tumor Necrosis Factor–Producing T Cells Synergize with Cisplatin to Promote Tumor Cell Death
- Author
-
Cornelis J.M. Melief, Sjoerd H. van der Burg, Ramon Arens, Bob van de Water, Ferry Ossendorp, Marij J.P. Welters, Vincent T.H.B.M. Smit, Louis Boon, Arjen Sloots, Elham Beyranvand Nejad, Ekaterina S. Jordanova, Suzanna Huppelschoten, Suzanne van Duikeren, and Tetje C. van der Sluis
- Abstract
Supplemental figure 2) Activation markers on T cells are not affected by cisplatin treatment.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplemental figure 1 from Vaccine-Induced Tumor Necrosis Factor–Producing T Cells Synergize with Cisplatin to Promote Tumor Cell Death
- Author
-
Cornelis J.M. Melief, Sjoerd H. van der Burg, Ramon Arens, Bob van de Water, Ferry Ossendorp, Marij J.P. Welters, Vincent T.H.B.M. Smit, Louis Boon, Arjen Sloots, Elham Beyranvand Nejad, Ekaterina S. Jordanova, Suzanna Huppelschoten, Suzanne van Duikeren, and Tetje C. van der Sluis
- Abstract
Supplemental figure 1) Peptide vaccination synergizes with cisplatin, which allows lowering of the cisplatin dose.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplemental Video 4 from Vaccine-Induced Tumor Necrosis Factor–Producing T Cells Synergize with Cisplatin to Promote Tumor Cell Death
- Author
-
Cornelis J.M. Melief, Sjoerd H. van der Burg, Ramon Arens, Bob van de Water, Ferry Ossendorp, Marij J.P. Welters, Vincent T.H.B.M. Smit, Louis Boon, Arjen Sloots, Elham Beyranvand Nejad, Ekaterina S. Jordanova, Suzanna Huppelschoten, Suzanne van Duikeren, and Tetje C. van der Sluis
- Abstract
Supplemental Video 4. TC-1 cells were in vitro cultured and treated with cisplatin (2 μg/ml) and TNFα (250iU). Cell death was followed for 22 hours using Annexin-V staining and automated imaging on a BD pathway 855 imager. Every hour a picture was taken.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 5 from Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells
- Author
-
Ramon Arens, Sjoerd H. van der Burg, Cornelis J.M. Melief, Peter A. van Veelen, George M. Janssen, Hideo Yagita, Suzanne van Duikeren, Tetje C. van der Sluis, and Elham Beyranvand Nejad
- Abstract
Modulation of costimulatory molecule expression by HMGB1.
- Published
- 2023
- Full Text
- View/download PDF
13. Data from Vaccine-Induced Tumor Necrosis Factor–Producing T Cells Synergize with Cisplatin to Promote Tumor Cell Death
- Author
-
Cornelis J.M. Melief, Sjoerd H. van der Burg, Ramon Arens, Bob van de Water, Ferry Ossendorp, Marij J.P. Welters, Vincent T.H.B.M. Smit, Louis Boon, Arjen Sloots, Elham Beyranvand Nejad, Ekaterina S. Jordanova, Suzanna Huppelschoten, Suzanne van Duikeren, and Tetje C. van der Sluis
- Abstract
Purpose: Cancer immunotherapy, such as vaccination, is an increasingly successful treatment modality, but its interaction with chemotherapy remains largely undefined. Therefore, we explored the mechanism of synergy between vaccination with synthetic long peptides (SLP) of human papillomavirus type 16 (HPV16) and cisplatin in a preclinical tumor model for HPV16.Experimental Design: SLP vaccination in this preclinical tumor model allowed the elucidation of novel mechanisms of synergy between chemo- and immunotherapy. By analyzing the tumor immune infiltrate, we focused on the local intratumoral effects of chemotherapy, vaccination, or the combination.Results: Of several chemotherapeutic agents, cisplatin synergized best with SLP vaccination in tumor eradication, without requirement for the maximum-tolerated dose (MTD). Upon SLP vaccination, tumors were highly infiltrated with HPV-specific, tumor necrosis factor-α (TNFα)- and interferon-γ (IFNγ)–producing T cells. Upon combined treatment, tumor cell proliferation was significantly decreased compared with single treated and untreated tumors. Furthermore, we showed that TNFα strongly enhanced cisplatin-induced apoptotic tumor cell death in a JNK-dependent manner. This is consistent with upregulation of proapoptotic molecules and with enhanced cell death in vivo upon combined SLP vaccination and cisplatin treatment. In vivo neutralization of TNFα significantly reduced the antitumor responses induced by the combined treatment.Conclusion: Taken together, our data show that peptide vaccination with cisplatin treatment leads to decreased tumor cell proliferation and TNFα-induced enhanced cisplatin-mediated killing of tumor cells, together resulting in superior tumor eradication. Clin Cancer Res; 21(4); 781–94. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure 3 from Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells
- Author
-
Ramon Arens, Sjoerd H. van der Burg, Cornelis J.M. Melief, Peter A. van Veelen, George M. Janssen, Hideo Yagita, Suzanne van Duikeren, Tetje C. van der Sluis, and Elham Beyranvand Nejad
- Abstract
Cisplatin-induced cell death enhances T-cell activation by upregulating CD70, CD80 and CD86 costimulatory molecules.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure 4 from Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells
- Author
-
Ramon Arens, Sjoerd H. van der Burg, Cornelis J.M. Melief, Peter A. van Veelen, George M. Janssen, Hideo Yagita, Suzanne van Duikeren, Tetje C. van der Sluis, and Elham Beyranvand Nejad
- Abstract
Categorizations of proteins released upon cisplatin treatment of tumor cells.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplemental Video 3 from Vaccine-Induced Tumor Necrosis Factor–Producing T Cells Synergize with Cisplatin to Promote Tumor Cell Death
- Author
-
Cornelis J.M. Melief, Sjoerd H. van der Burg, Ramon Arens, Bob van de Water, Ferry Ossendorp, Marij J.P. Welters, Vincent T.H.B.M. Smit, Louis Boon, Arjen Sloots, Elham Beyranvand Nejad, Ekaterina S. Jordanova, Suzanna Huppelschoten, Suzanne van Duikeren, and Tetje C. van der Sluis
- Abstract
Supplemental Video 3. TC-1 cells were in vitro cultured wer treated with TNFα (250iU). Cell death was followed for 22 hours using Annexin-V staining and automated imaging on a BD pathway 855 imager. Every hour a picture was taken.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplemental Video 1 from Vaccine-Induced Tumor Necrosis Factor–Producing T Cells Synergize with Cisplatin to Promote Tumor Cell Death
- Author
-
Cornelis J.M. Melief, Sjoerd H. van der Burg, Ramon Arens, Bob van de Water, Ferry Ossendorp, Marij J.P. Welters, Vincent T.H.B.M. Smit, Louis Boon, Arjen Sloots, Elham Beyranvand Nejad, Ekaterina S. Jordanova, Suzanna Huppelschoten, Suzanne van Duikeren, and Tetje C. van der Sluis
- Abstract
Supplemental Video 1. TC-1 cells were in vitro cultured and left untreated.Cell death was followed for 22 hours using Annexin-V staining and automated imaging on a BD pathway 855 imager. Every hour a picture was taken.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure 7 from Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells
- Author
-
Ramon Arens, Sjoerd H. van der Burg, Cornelis J.M. Melief, Peter A. van Veelen, George M. Janssen, Hideo Yagita, Suzanne van Duikeren, Tetje C. van der Sluis, and Elham Beyranvand Nejad
- Abstract
Cisplatin treatment does not affect the expression of costimulatory molecules on tumor cells, total myeloid cells of spleen and TDLN in vivo.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure 1 from Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells
- Author
-
Ramon Arens, Sjoerd H. van der Burg, Cornelis J.M. Melief, Peter A. van Veelen, George M. Janssen, Hideo Yagita, Suzanne van Duikeren, Tetje C. van der Sluis, and Elham Beyranvand Nejad
- Abstract
The maximum tolerated dose (MTD) of cisplatin does not induce liver or organ toxicity.
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure 6 from Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells
- Author
-
Ramon Arens, Sjoerd H. van der Burg, Cornelis J.M. Melief, Peter A. van Veelen, George M. Janssen, Hideo Yagita, Suzanne van Duikeren, Tetje C. van der Sluis, and Elham Beyranvand Nejad
- Abstract
Effects of cisplatin on the tumor microenvironment.
- Published
- 2023
- Full Text
- View/download PDF
21. Data from Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells
- Author
-
Ramon Arens, Sjoerd H. van der Burg, Cornelis J.M. Melief, Peter A. van Veelen, George M. Janssen, Hideo Yagita, Suzanne van Duikeren, Tetje C. van der Sluis, and Elham Beyranvand Nejad
- Abstract
Certain cytotoxic chemotherapeutic drugs are immunogenic, stimulating tumor immunity through mechanisms that are not completely understood. Here we show how the DNA-damaging drug cisplatin modulates tumor immunity. At the maximum tolerated dose (MTD), cisplatin cured 50% of mice with established murine TC-1 or C3 tumors, which are preclinical models of human papillomavirus (HPV)-associated cancer. Notably, the curative benefit of cisplatin relied entirely upon induction of tumor-specific CD8+ T cells. Mechanistic investigations showed that cisplatin stimulated tumor infiltration of inflammatory antigen-presenting cells (APC) expressing relatively higher levels of the T-cell costimulatory ligands CD70, CD80, and CD86. Cell death triggered by cisplatin was associated with the release of at least 19 proteins in the tumor environment that could act as damage-associated molecular patterns and upregulate costimulatory molecules, either alone or in concert, but the responsible proteins remain unknown. Essentially, the curative effect of cisplatin was abrogated in mice lacking expression of CD80 and CD86 on APCs. Furthermore, cisplatin treatment was improved by CTLA-4 blockade, which increases the availability of CD80/86 to bind to CD28. In contrast, there was no effect of CD27 stimulation, which replaces CD70 interaction. At the cisplatin MTD, cure rates could also be increased by vaccination with synthetic long peptides, whereas cures could also be achieved at similar rates at 80% of the MTD with reduced side effects. Our findings reveal an essential basis for the immunogenic properties of cisplatin, which are mediated by the induction of costimulatory signals for CD8+ T-cell–dependent tumor destruction. Cancer Res; 76(20); 6017–29. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Figure Legends and Methods from Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells
- Author
-
Ramon Arens, Sjoerd H. van der Burg, Cornelis J.M. Melief, Peter A. van Veelen, George M. Janssen, Hideo Yagita, Suzanne van Duikeren, Tetje C. van der Sluis, and Elham Beyranvand Nejad
- Abstract
Supplementary Figure Legends and Supplementary Materials and Methods
- Published
- 2023
- Full Text
- View/download PDF
23. OX40 agonism enhances PD-L1 checkpoint blockade by shifting the cytotoxic T cell differentiation spectrum
- Author
-
Tetje C. van der Sluis, Guillaume Beyrend, Esmé T.I. van der Gracht, Tamim Abdelaal, Simon P. Jochems, Robert A. Belderbos, Thomas H. Wesselink, Suzanne van Duikeren, Floortje J. van Haften, Anke Redeker, Laura F. Ouboter, Elham Beyranvand Nejad, Marcel Camps, Kees L.M.C. Franken, Margot M. Linssen, Peter Hohenstein, Noel F.C.C. de Miranda, Hailiang Mei, Adriaan D. Bins, John B.A.G. Haanen, Joachim G. Aerts, Ferry Ossendorp, Ramon Arens, and Pulmonary Medicine
- Subjects
mass cytometry ,predictive biomarkers ,SDG 3 - Good Health and Well-being ,T cells ,systemic immune activation ,immunotherapy ,General Biochemistry, Genetics and Molecular Biology ,single-cell RNA sequencing ,immune checkpoint therapy - Abstract
Immune checkpoint therapy (ICT) has the power to eradicate cancer, but the mechanisms that determine effective therapy-induced immune responses are not fully understood. Here, using high-dimensional single-cell profiling, we interrogate whether the landscape of T cell states in the peripheral blood predict responses to combinatorial targeting of the OX40 costimulatory and PD-1 inhibitory pathways. Single-cell RNA sequencing and mass cytometry expose systemic and dynamic activation states of therapy-responsive CD4+ and CD8+ T cells in tumor-bearing mice with expression of distinct natural killer (NK) cell receptors, granzymes, and chemokines/chemokine receptors. Moreover, similar NK cell receptor-expressing CD8+ T cells are also detected in the blood of immunotherapy-responsive cancer patients. Targeting the NK cell and chemokine receptors in tumor-bearing mice shows the functional importance of these receptors for therapy-induced anti-tumor immunity. These findings provide a better understanding of ICT and highlight the use and targeting of dynamic biomarkers on T cells to improve cancer immunotherapy.
- Published
- 2023
24. Interleukin‐6‐mediated resistance to immunotherapy is linked to impaired myeloid cell function
- Author
-
Camilla Labrie, Elham Beyranvand Nejad, Kees L. M. C. Franken, Sjoerd H. van der Burg, Tetje C. van der Sluis, Rueshandra Roosenhoff, Suzanne van Duikeren, Thorbald van Hall, and Ramon Arens
- Subjects
Cancer Research ,Myeloid ,medicine.medical_treatment ,Tumor Immunology and Microenvironment ,chemotherapy ,Major histocompatibility complex ,Cancer Vaccines ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,tumor microenvironment ,Myeloid Cells ,Cisplatin ,Human papillomavirus 16 ,MHC class II ,Chemotherapy ,Tumor microenvironment ,biology ,business.industry ,interleukin-6 ,Papillomavirus Infections ,Histocompatibility Antigens Class II ,Cancer ,Immunotherapy ,medicine.disease ,Disease Models, Animal ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,interleukin‐6 ,030220 oncology & carcinogenesis ,Vaccines, Subunit ,biology.protein ,Cancer research ,Female ,immunotherapy ,business ,medicine.drug - Abstract
High serum levels of interleukin‐6 (IL‐6) correlate with poor prognosis and chemotherapy resistance in several cancers. The underlying mechanisms and its effects on immunotherapy are largely unknown. To address this, we developed a human papillomavirus type 16 (HPV16)‐associated tumor model expressing IL‐6 to investigate the impact of tumor‐expressed IL‐6 during cisplatin chemotherapy and HPV16 synthetic long peptide vaccination as immunotherapy. The effects of tumor‐produced IL‐6 on tumor growth, survival and the tumor microenvironment were analyzed. Our data demonstrated that tumor‐produced IL‐6 conferred resistance to cisplatin and therapeutic vaccination. This was not caused by a changed in vitro or in vivo growth rate of tumor cells, or a changed sensitivity of tumor cells to chemotherapy or T‐cell‐mediated killing. Furthermore, no overt differences in the frequencies of tumor‐infiltrating subsets of T cells or CD11b+ myeloid cells were observed. IL‐6, however, affected the systemic and local function of myeloid cells, reflected by a strong reduction of major histocompatibility complex (MHC) class II expression on all major myeloid cell subtypes. Resistance to both therapies was associated with a changed intratumoral influx of MHC class II+ myeloid cells toward myeloid cells with no or lower MHC class II expression. Importantly, while these IL‐6‐mediated effects provided resistance to the immunotherapy and chemotherapy as single therapies, their combination still successfully mediated tumor control. In conclusion, IL‐6‐mediated therapy resistance is caused by an extrinsic mechanism involving an impaired function of intratumoral myeloid cells. The fact that resistance can be overcome by combination therapies provides direction to more effective therapies for cancer., What's new? Interleukin‐6 (IL‐6) cytokine has multiple effects on hematopoiesis and immune function and typically circulates at low levels. In cancer, however, IL‐6 serum levels are significantly elevated, with suspected impacts on tumor behavior. In this study, using a mouse model of human papillomavirus‐induced cancer with IL‐6 expression, the authors show that tumor‐produced IL‐6 confers resistance to both chemotherapy and immunotherapy. Resistance was associated with impaired myeloid cell maturation, with no evidence of involvement of mechanisms intrinsic to tumor cells. Resistance was overcome by combining chemotherapy and immunotherapy, providing insight into a potentially effective therapeutic approach for cancers with IL‐6‐mediated resistance.
- Published
- 2020
- Full Text
- View/download PDF
25. OX40 Agonism Enhances Efficacy of PD-L1 Checkpoint Blockade by Shifting the Cytotoxic T Cell Differentiation Spectrum
- Author
-
Guillaume Beyrend, Tetje C. van der Sluis, Esmé T.I. van der Gracht, Tamim Abdelaal, Simon P. Jochems, Robert A. Belderbos, Thomas H. Wesselink, Suzanne van Duikeren, Floortje J. van Haften, Anke Redeker, Elham Beyranvand Nejad, Marcel Camps, Kees LMC Franken, Margot M. Linssen, Peter Hohenstein, Noel F.C.C. de Miranda, Hailiang Mei, Adriaan D. Bins, John B.A.G. Haanen, Joachim G. Aerts, Ferry Ossendorp, and Ramon Arens
- Abstract
Immune checkpoint therapy (ICT) has the potency to eradicate cancer but the mechanisms that determine effective versus non-effective therapy-induced immune responses are not fully understood. Here, using high-dimensional single-cell profiling we examined whether T cell states in the blood circulation could predict responsiveness to a combined ICT, sequentially targeting OX40 costimulatory and PD-1 inhibitory pathways, which effectively eradicated syngeneic mouse tumors. Unbiased assessment of transcriptomic alterations by single-cell RNA sequencing and profiling of cell-surface protein expression by mass cytometry revealed unique activation states for therapy-responsive CD4+ and CD8+ T cells. Effective ICT elicited T cells with dynamic expression of distinct NK cell and chemokine receptors, and these cells were systemically present in lymphoid tissues and in the tumor. Moreover, NK cell receptor-expressing CD8+ T cells were also present in the peripheral blood of immunotherapy-responsive cancer patients. Targeting of the NK cell and chemokine receptors in tumor-bearing mice showed their functional importance for therapy-induced anti-tumor immunity. These findings provide a better understanding of ICT and highlight the use of dynamic biomarkers on effector CD4+ and CD8+ T cells to improve cancer immunotherapy.
- Published
- 2022
- Full Text
- View/download PDF
26. A third vaccination with a single T cell epitope protects against SARS-CoV-2 infection in the absence of neutralizing antibodies
- Author
-
Iris N. Pardieck, Esmé T.I. van der Gracht, Dominique M.B. Veerkamp, Felix M. Behr, Suzanne van Duikeren, Guillaume Beyrend, Jasper Rip, Reza Nadafi, Tetje C. van der Sluis, Elham Beyranvand Nejad, Nils Mülling, Dena J. Brasem, Marcel G.M. Camps, Sebenzile K. Myeni, Peter J. Bredenbeek, Marjolein Kikkert, Yeonsu Kim, Luka Cicin-Sain, Tamim Abdelaal, Klaas P.J.M. van Gisbergen, Kees L.M.C. Franken, Jan Wouter Drijfhout, Cornelius J.M. Melief, Gerben C.M. Zondag, Ferry Ossendorp, and Ramon Arens
- Abstract
Understanding the mechanisms and impact of booster vaccinations can facilitate decisions on vaccination programmes. This study shows that three doses of the same synthetic peptide vaccine eliciting an exclusive CD8+ T cell response against one SARS-CoV-2 Spike epitope protected all mice against lethal SARS-CoV-2 infection in the K18-hACE2 transgenic mouse model in the absence of neutralizing antibodies, while only a second vaccination with this T cell vaccine was insufficient to provide protection. The third vaccine dose of the single T cell epitope peptide resulted in superior generation of effector-memory T cells in the circulation and tissue-resident memory T (TRM) cells, and these tertiary vaccine-specific CD8+ T cells were characterized by enhanced polyfunctional cytokine production. Moreover, fate mapping showed that a substantial fraction of the tertiary effector-memory CD8+ T cells developed from remigrated TRM cells. Thus, repeated booster vaccinations quantitatively and qualitatively improve the CD8+ T cell response leading to protection against otherwise lethal SARS-CoV-2 infection.SummaryA third dose with a single T cell epitope-vaccine promotes a strong increase in tissue-resident memory CD8+ T cells and fully protects against SARS-CoV-2 infection, while single B cell epitope-eliciting vaccines are unable to provide protection.
- Published
- 2021
- Full Text
- View/download PDF
27. IL-6 signaling in macrophages is required for immunotherapy-driven regression of tumors
- Author
-
Jan Willem Kleinovink, Thorbald van Hall, Marit J van Elsas, Kees L. M. C. Franken, Sjoerd H. van der Burg, Hans-Willi Mittrücker, Elham Beyranvand Nejad, Ramon Arens, Thomas Korn, Sylvia Heink, and Camilla Labrie
- Subjects
Cytotoxicity, Immunologic ,Cancer Research ,medicine.medical_treatment ,Injections, Subcutaneous ,Papillomavirus E7 Proteins ,Immunology ,active ,CD8-Positive T-Lymphocytes ,Cancer Vaccines ,Lymphocytes, Tumor-Infiltrating ,Cell Line, Tumor ,Neoplasms ,Tumor-Associated Macrophages ,medicine ,Immunology and Allergy ,Macrophage ,Animals ,SOCS3 ,RC254-282 ,Pharmacology ,Mice, Knockout ,Tumor microenvironment ,business.industry ,Interleukin-6 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Interleukin ,Basic Tumor Immunology ,Immunotherapy ,adaptive immunity ,Acquired immune system ,Receptors, Interleukin-6 ,Blockade ,Tumor Burden ,macrophages ,ddc ,Mice, Inbred C57BL ,Cytokine ,Phenotype ,Oncology ,Oligodeoxyribonucleotides ,Cancer research ,Molecular Medicine ,Female ,immunotherapy ,business ,Signal Transduction ,immune evation - Abstract
BackgroundHigh serum interleukin (IL-6) levels may cause resistance to immunotherapy by modulation of myeloid cells in the tumor microenvironment. IL-6 signaling blockade is tested in cancer, but as this inflammatory cytokine has pleiotropic effects, this treatment is not always effective.MethodsIL-6 and IL-6R blockade was applied in an IL-6-mediated immunotherapy-resistant TC-1 tumor model (TC-1.IL-6) and immunotherapy-sensitive TC-1.control. Effects on therapeutic vaccination-induced tumor regression, recurrence and survival as well on T cells and myeloid cells in the tumor microenvironment were studied. The effects of IL-6 signaling in macrophages under therapy conditions were studied in Il6rafl/fl×LysMcre+ mice.ResultsOur therapeutic vaccination protocol elicits a strong tumor-specific CD8+ T-cell response, leading to enhanced intratumoral T-cell infiltration and recruitment of tumoricidal macrophages. Blockade of IL-6 signaling exacerbated tumor outgrowth, reflected by fewer complete regressions and more recurrences after therapeutic vaccination, especially in TC-1.IL-6 tumor-bearing mice. Early IL-6 signaling blockade partly inhibited the development of the vaccine-induced CD8+ T-cell response. However, the main mechanism was the malfunction of macrophages during therapy-induced tumor regression. Therapy efficacy was impaired in Il6rafl/fl×LysMcre+ but not cre-negative control mice, while no differences in the vaccine-induced CD8+ T-cell response were found between these mice. IL-6 signaling blockade resulted in decreased expression of suppressor of cytokine signaling 3, essential for effective M1-type function in macrophages, and increased expression of the phagocytic checkpoint molecule signal-regulatory protein alpha by macrophages.ConclusionIL-6 signaling is critical for macrophage function under circumstances of immunotherapy-induced tumor tissue destruction, in line with the acute inflammatory functions of IL-6 signaling described in infections.
- Published
- 2020
28. Lack of myeloid cell infiltration as an acquired resistance strategy to immunotherapy
- Author
-
Jan Willem Kleinovink, Jan Oosting, Elham Beyranvand Nejad, Suzanne van Duikeren, Aart G. Jochemsen, Thorbald van Hall, Camilla Labrie, Eva Rademaker, Tetje C. van der Sluis, Ziena Abdulrahman, Sjoerd H. van der Burg, Ramon Arens, Marit J van Elsas, Amina F A S Teunisse, and Noel F C C de Miranda
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,medicine.medical_treatment ,Immunology ,active ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Immunology and Allergy ,Medicine ,Animals ,Humans ,tumor microenvironment ,Myeloid Cells ,RC254-282 ,Pharmacology ,Tumor microenvironment ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Basic Tumor Immunology ,tumor escape ,Immunotherapy ,vaccination ,Immune checkpoint ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Tumor Escape ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Tumor necrosis factor alpha ,immunotherapy ,business ,CD8 - Abstract
BackgroundImmunotherapy of cancer is successful but tumor regression often is incomplete and followed by escape. Understanding the mechanisms underlying this acquired resistance will aid the development of more effective treatments.MethodsWe exploited a mouse model where tumor-specific therapeutic vaccination results in tumor regression, followed by local recurrence and resistance. In depth studies on systemic, local and tumor intrinsic changes were performed with flow and mass cytometry, immunohistochemistry, transcriptomics and several perturbation studies with inhibitors or agonistic antibodies in mice. Main findings were recapitulated in vaccinated patients.ResultsFull tumor regression and cure of tumor-bearing mice is dependent on the magnitude of the vaccine-induced T-cell response. Recurrence of tumors did not involve classical immune escape mechanisms, such as antigen-presentation alterations, immune checkpoint expression, resistance to killing or local immune suppression. However, the recurrent tumors displayed a changed transcriptome with alterations in p53, tumor necrosis factor-α and transforming growth factor-β signaling pathways and they became immunologically cold. Remarkably, ex vivo cell-sorted recurrent tumors, directly reinjected in naïve hosts retained their resistance to vaccination despite a strong infiltration with tumor-specific CD8+ T cells, similar to that of vaccine-responsive tumors. The influx of inflammatory mature myeloid effector cells in the resistant tumors, however, was impaired and this turned out to be the underlying mechanisms as restoration of inflammatory myeloid cell infiltration reinstated the sensitivity of these refractory tumors to vaccination. Notably, impaired myeloid cell infiltration after vaccination was also associated with vaccine resistance in patients.ConclusionAn immunotherapy-induced disability of tumor cells to attract innate myeloid effector cells formed a major mechanism underlying immune escape and acquired resistance. These data not only stresses the importance of myeloid effector cells during immunotherapy but also demands for new studies to harness their tumoricidal activities.
- Published
- 2020
29. Demarcated thresholds of tumor-specific CD8 T cells elicited by MCMV-based vaccine vectors provide robust correlates of protection
- Author
-
Sjoerd H. van der Burg, Luka Cicin-Sain, Robert B. Ratts, Jennifer D. Oduro, Eleni Panagioti, Ramon Arens, Christine Meyer, Elham Beyranvand Nejad, Klaus Früh, and HZI,Helmholtz-Zentrum für Infektionsforschung GmbH, Inhoffenstr. 7,38124 Braunschweig, Germany.
- Subjects
Cancer Research ,Papillomavirus E7 Proteins ,medicine.medical_treatment ,T cell ,CMV-based vaccine vector ,Pre-existing immunity ,Immunology ,T cells ,Cytomegalovirus ,CD8-Positive T-Lymphocytes ,Biology ,Cancer Vaccines ,lcsh:RC254-282 ,Epitopes ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Cancer ,Pharmacology ,Human papillomavirus 16 ,Papillomavirus Infections ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Mice, Inbred C57BL ,Vaccination ,medicine.anatomical_structure ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Molecular Medicine ,Cancer vaccine ,CD8 ,Research Article ,030215 immunology - Abstract
Background The capacity of cytomegalovirus (CMV) to elicit long-lasting strong T cell responses, and the ability to engineer the genome of this DNA virus positions CMV-based vaccine vectors highly suitable as a cancer vaccine platform. Defined immune thresholds for tumor protection and the factors affecting such thresholds have not well been investigated in cancer immunotherapy. We here determined using CMV as a vaccine platform whether critical thresholds of vaccine-specific T cell responses can be established that relate to tumor protection, and which factors control such thresholds. Methods We generated CMV-based vaccine vectors expressing the E7 epitope and tested these in preclinical models of HPV16-induced cancer. Vaccination was applied via different doses and routes (intraperitoneal (IP), subcutaneous (SC) and intranasal (IN)). The magnitude, kinetics and phenotype of the circulating tumor-specific CD8+ T cell response were determined. Mice were subsequently challenged with tumor cells, and the tumor protection was monitored. Results Immunization with CMV-based vaccines via the IP or SC route eliciting vaccine-induced CD8+ T cell responses of > 0.3% of the total circulating CD8 T cell population fully protects mice against lethal tumor challenge. However, low dose inoculations via the IP or SC route or IN vaccination elicited vaccine-induced CD8+ T cell responses that did not reach protective thresholds for tumor protection. In addition, whereas weak pre-existing immunity did not alter the protective thresholds of the vaccine-specific T cell response following subsequent immunization with CMV-based vaccine vectors, strong pre-existing immunity inhibited the development of vaccine-induced T cells and their control on tumor progression. Conclusions This study highlight the effectiveness of CMV-based vaccine vectors, and shows that demarcated thresholds of vaccine-specific T cells could be defined that correlate to tumor protection. Together, these results may hold importance for cancer vaccine development to achieve high efficacy in vaccine recipients. Electronic supplementary material The online version of this article (10.1186/s40425-019-0500-9) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
30. The importance of correctly timing cancer immunotherapy
- Author
-
Marij J. P. Welters, Elham Beyranvand Nejad, Sjoerd H. van der Burg, and Ramon Arens
- Subjects
0301 basic medicine ,Time Factors ,Combination therapy ,medicine.medical_treatment ,T-Lymphocytes ,Clinical Biochemistry ,Antineoplastic Agents ,Disease ,Bioinformatics ,Cancer Vaccines ,combination therapy ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,immunomonitoring ,Neoplasms ,Drug Discovery ,medicine ,timing ,Animals ,Humans ,Immunologic Factors ,Cancer ,Pharmacology ,Modalities ,business.industry ,Vaccination ,Immunotherapy ,medicine.disease ,Radiation therapy ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,business - Abstract
The treatment options for cancer-surgery, radiotherapy and chemotherapy-are now supplemented with immunotherapy. Previously underappreciated but now gaining strong interest are the immune modulatory properties of the three conventional modalities. Moreover, there is a better understanding of the needs and potential of the different immune therapeutic platforms. Key to improved treatment will be the combinations of modalities that complete each other's shortcomings. Area covered: Tumor-specific T-cells are required for optimal immunotherapy. In this review, the authors focus on the correct timing of different types of chemotherapeutic agents or immune modulators and immunotherapeutic drugs, not only for the activation and expansion of tumor-specific T-cells but also to support and enhance their anti-tumor efficacy. Expert opinion: At an early phase of disease, clinical success can be obtained using single treatment modalities but at later disease stages, combinations of several modalities are required. The gain in success is determined by a thorough understanding of the direct and indirect immune effects of the modalities used. Profound knowledge of these effects requires optimal tuning of immunomonitoring. This will guide the appropriate combination of treatments and allow for correct sequencing the order and interval of the different therapeutic modalities.
- Published
- 2016
31. Tumor Eradication by Cisplatin Is Sustained by CD80/86-Mediated Costimulation of CD8+ T Cells
- Author
-
Hideo Yagita, Suzanne van Duikeren, Elham Beyranvand Nejad, George M.C. Janssen, Peter A. van Veelen, Cornelis J. M. Melief, Ramon Arens, Sjoerd H. van der Burg, and Tetje C. van der Sluis
- Subjects
0301 basic medicine ,Cancer Research ,Antigen-Presenting Cells ,chemical and pharmacologic phenomena ,Antineoplastic Agents ,Pharmacology ,Biology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,03 medical and health sciences ,Interferon-gamma ,Mice ,0302 clinical medicine ,medicine ,Cytotoxic T cell ,Animals ,Antigen-presenting cell ,CD86 ,Cisplatin ,Myeloid-Derived Suppressor Cells ,Vaccination ,CD28 ,Neoplasms, Experimental ,Mice, Inbred C57BL ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Myeloid-derived Suppressor Cell ,B7-1 Antigen ,B7-2 Antigen ,CD80 ,CD8 ,medicine.drug ,CD27 Ligand - Abstract
Certain cytotoxic chemotherapeutic drugs are immunogenic, stimulating tumor immunity through mechanisms that are not completely understood. Here we show how the DNA-damaging drug cisplatin modulates tumor immunity. At the maximum tolerated dose (MTD), cisplatin cured 50% of mice with established murine TC-1 or C3 tumors, which are preclinical models of human papillomavirus (HPV)-associated cancer. Notably, the curative benefit of cisplatin relied entirely upon induction of tumor-specific CD8+ T cells. Mechanistic investigations showed that cisplatin stimulated tumor infiltration of inflammatory antigen-presenting cells (APC) expressing relatively higher levels of the T-cell costimulatory ligands CD70, CD80, and CD86. Cell death triggered by cisplatin was associated with the release of at least 19 proteins in the tumor environment that could act as damage-associated molecular patterns and upregulate costimulatory molecules, either alone or in concert, but the responsible proteins remain unknown. Essentially, the curative effect of cisplatin was abrogated in mice lacking expression of CD80 and CD86 on APCs. Furthermore, cisplatin treatment was improved by CTLA-4 blockade, which increases the availability of CD80/86 to bind to CD28. In contrast, there was no effect of CD27 stimulation, which replaces CD70 interaction. At the cisplatin MTD, cure rates could also be increased by vaccination with synthetic long peptides, whereas cures could also be achieved at similar rates at 80% of the MTD with reduced side effects. Our findings reveal an essential basis for the immunogenic properties of cisplatin, which are mediated by the induction of costimulatory signals for CD8+ T-cell–dependent tumor destruction. Cancer Res; 76(20); 6017–29. ©2016 AACR.
- Published
- 2016
32. Vaccine-Induced Tumor Necrosis Factor-Producing T Cells Synergize with Cisplatin to Promote Tumor Cell Death
- Author
-
Cornelis J. M. Melief, Louis Boon, Ramon Arens, Suzanne van Duikeren, Sjoerd H. van der Burg, Ferry Ossendorp, Elham Beyranvand Nejad, Marij J. P. Welters, Arjen Sloots, Ekaterina S. Jordanova, Vincent T.H.B.M. Smit, Bob van de Water, Tetje C. van der Sluis, Suzanna Huppelschoten, Obstetrics and gynaecology, and CCA - Innovative therapy
- Subjects
Cancer Research ,medicine.medical_treatment ,Uterine Cervical Neoplasms ,Antineoplastic Agents ,CD8-Positive T-Lymphocytes ,Real-Time Polymerase Chain Reaction ,Cancer Vaccines ,Mice ,Viral Proteins ,Lymphocytes, Tumor-Infiltrating ,Cancer immunotherapy ,medicine ,Animals ,Humans ,Cisplatin ,Human papillomavirus 16 ,Chemotherapy ,Cell Death ,Tumor Necrosis Factor-alpha ,business.industry ,Papillomavirus Infections ,Cancer ,Immunotherapy ,medicine.disease ,Adoptive Transfer ,Mice, Inbred C57BL ,Vaccination ,Disease Models, Animal ,Oncology ,Apoptosis ,Immunology ,Cancer research ,Female ,Tumor necrosis factor alpha ,Peptides ,business ,medicine.drug - Abstract
Purpose: Cancer immunotherapy, such as vaccination, is an increasingly successful treatment modality, but its interaction with chemotherapy remains largely undefined. Therefore, we explored the mechanism of synergy between vaccination with synthetic long peptides (SLP) of human papillomavirus type 16 (HPV16) and cisplatin in a preclinical tumor model for HPV16. Experimental Design: SLP vaccination in this preclinical tumor model allowed the elucidation of novel mechanisms of synergy between chemo- and immunotherapy. By analyzing the tumor immune infiltrate, we focused on the local intratumoral effects of chemotherapy, vaccination, or the combination. Results: Of several chemotherapeutic agents, cisplatin synergized best with SLP vaccination in tumor eradication, without requirement for the maximum-tolerated dose (MTD). Upon SLP vaccination, tumors were highly infiltrated with HPV-specific, tumor necrosis factor-α (TNFα)- and interferon-γ (IFNγ)–producing T cells. Upon combined treatment, tumor cell proliferation was significantly decreased compared with single treated and untreated tumors. Furthermore, we showed that TNFα strongly enhanced cisplatin-induced apoptotic tumor cell death in a JNK-dependent manner. This is consistent with upregulation of proapoptotic molecules and with enhanced cell death in vivo upon combined SLP vaccination and cisplatin treatment. In vivo neutralization of TNFα significantly reduced the antitumor responses induced by the combined treatment. Conclusion: Taken together, our data show that peptide vaccination with cisplatin treatment leads to decreased tumor cell proliferation and TNFα-induced enhanced cisplatin-mediated killing of tumor cells, together resulting in superior tumor eradication. Clin Cancer Res; 21(4); 781–94. ©2014 AACR.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.