1
|
Lin Q, Olkowski CP, Choyke PL, Sato N. Tumor growth suppression in adoptive T cell therapy via IFN-γ targeting of tumor vascular endothelial cells. Theranostics 2024; 14:6897-6912. [PMID: 39629126 PMCID: PMC11610145 DOI: 10.7150/thno.101107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/27/2024] [Indexed: 12/06/2024] Open
Abstract
Rationale: In adoptive T cell therapy (ACT), the direct cytotoxic effects of CD8 T cells on tumor cells, including the release of interferon-gamma (IFN-γ), are considered the primary mechanism for tumor eradication. Cancer antigen escape diminishes the T cell responses, thereby limiting the therapeutic success. The impacts of IFN-γ targeting non-tumor cells in ACT, on the other hand, remains under-investigated. We hypothesized that IFN-γ action on non-tumor cells, particularly tumor vascular endothelial cells within the physiological tumor microenvironment, could influence therapeutic efficacy. Methods: ACT was performed against ovalbumin (OVA)- or OVA-peptide SIINFEKL-expressing syngeneic mouse tumors, MCA-205-OVA-GFP fibrosarcoma or MOC2-SIINFEKL oral squamous cell carcinoma, using ex vivo-activated OT-1 CD8 T cells expressing the T cell receptor against OVA. Efficacy was examined in wild-type mice, mice deficient for IFN-γ receptor 1 (IFN-γR1KO), and bone marrow chimeras lacking IFN-γR1 expression in endothelial cells. To exclude direct IFN-γ action against tumor cells, IFN-γR1KO-MCA-205-OVA-GFP tumors were used. IFN-γ production, STAT1 induction in its targets, and subsequent changes, especially in vasculatures in the tumor, were examined. Results: ACT suppressed the growth of MCA-205-OVA-GFP and MOC2-SIINFEKL tumors in wild-type mice but failed in IFNγR1KO mice. Furthermore, in the bone marrow chimeras lacking endothelial cell IFN-γR1, ACT efficacy was lost, thus implicating a vital role of IFN-γ action on the endothelium. IFN-γR1KO-MCA-205-OVA-GFP tumor growth was successfully suppressed by ACT in wild-type mice, suggesting that IFN-γ targeting of tumor cells may not be essential for ACT efficacy. OT-1 CD8 T cells interacted with endothelial cells or localized in proximity to the vessels on Day 1.5 after transfer, as observed by intravital microscopy. The OT-1 T cells found in tumors were limited in number but produced high levels of IFN-γ on Day 1.5, while their number peaked on Day 5.5 with negligible IFN-γ production. Together with IFN-γ production by endogenous lymphocytes, IFN-γ levels in the whole tumor peaked on Day 1.5, inducing IFN-γ/STAT1 signaling in endothelial cells. Early targeting of tumor vascular endothelial cells by IFN-γ led to endothelial regression, reduced perfusion, and tumor hypoxia/necrosis (Day 4.5-7). Conclusions: These findings highlight the critical role of T cell-derived IFN-γ action on endothelial cells early in ACT, emphasizing its dynamic influence on the tumor microenvironment, and offering insights into addressing antigen escape.
Collapse
Affiliation(s)
| | | | | | - Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Espinosa-Carrasco G, Chiu E, Scrivo A, Zumbo P, Dave A, Betel D, Kang SW, Jang HJ, Hellmann MD, Burt BM, Lee HS, Schietinger A. Intratumoral immune triads are required for immunotherapy-mediated elimination of solid tumors. Cancer Cell 2024; 42:1202-1216.e8. [PMID: 38906155 PMCID: PMC11413804 DOI: 10.1016/j.ccell.2024.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 03/11/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Tumor-specific CD8+ T cells are frequently dysfunctional and unable to halt tumor growth. We investigated whether tumor-specific CD4+ T cells can be enlisted to overcome CD8+ T cell dysfunction within tumors. We find that the spatial positioning and interactions of CD8+ and CD4+ T cells, but not their numbers, dictate anti-tumor responses in the context of adoptive T cell therapy as well as immune checkpoint blockade (ICB): CD4+ T cells must engage with CD8+ T cells on the same dendritic cell during the effector phase, forming a three-cell-type cluster (triad) to license CD8+ T cell cytotoxicity and cancer cell elimination. When intratumoral triad formation is disrupted, tumors progress despite equal numbers of tumor-specific CD8+ and CD4+ T cells. In patients with pleural mesothelioma treated with ICB, triads are associated with clinical responses. Thus, CD4+ T cells and triads are required for CD8+ T cell cytotoxicity during the effector phase and tumor elimination.
Collapse
Affiliation(s)
| | - Edison Chiu
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aurora Scrivo
- Department of Developmental and Molecular Biology, and Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Paul Zumbo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA; Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Asim Dave
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA; Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sung Wook Kang
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Hee-Jin Jang
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Matthew D Hellmann
- Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Bryan M Burt
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA; Division of Thoracic Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Hyun-Sung Lee
- Systems Onco-Immunology Laboratory, David J. Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
3
|
Hickey JW, Agmon E, Horowitz N, Tan TK, Lamore M, Sunwoo JB, Covert MW, Nolan GP. Integrating multiplexed imaging and multiscale modeling identifies tumor phenotype conversion as a critical component of therapeutic T cell efficacy. Cell Syst 2024; 15:322-338.e5. [PMID: 38636457 PMCID: PMC11030795 DOI: 10.1016/j.cels.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/07/2023] [Accepted: 03/19/2024] [Indexed: 04/20/2024]
Abstract
Cancer progression is a complex process involving interactions that unfold across molecular, cellular, and tissue scales. These multiscale interactions have been difficult to measure and to simulate. Here, we integrated CODEX multiplexed tissue imaging with multiscale modeling software to model key action points that influence the outcome of T cell therapies with cancer. The initial phenotype of therapeutic T cells influences the ability of T cells to convert tumor cells to an inflammatory, anti-proliferative phenotype. This T cell phenotype could be preserved by structural reprogramming to facilitate continual tumor phenotype conversion and killing. One takeaway is that controlling the rate of cancer phenotype conversion is critical for control of tumor growth. The results suggest new design criteria and patient selection metrics for T cell therapies, call for a rethinking of T cell therapeutic implementation, and provide a foundation for synergistically integrating multiplexed imaging data with multiscale modeling of the cancer-immune interface. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- John W Hickey
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Eran Agmon
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT 06032, USA
| | - Nina Horowitz
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Tze-Kai Tan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew Lamore
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - John B Sunwoo
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Otolaryngology, Head and Neck Surgery, Stanford Cancer Institute Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Markus W Covert
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| | - Garry P Nolan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
4
|
Hickey JW, Agmon E, Horowitz N, Lamore M, Sunwoo J, Covert M, Nolan GP. Integrating Multiplexed Imaging and Multiscale Modeling Identifies Tumor Phenotype Transformation as a Critical Component of Therapeutic T Cell Efficacy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.06.570168. [PMID: 38106218 PMCID: PMC10723382 DOI: 10.1101/2023.12.06.570168] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Cancer progression is a complex process involving interactions that unfold across molecular, cellular, and tissue scales. These multiscale interactions have been difficult to measure and to simulate. Here we integrated CODEX multiplexed tissue imaging with multiscale modeling software, to model key action points that influence the outcome of T cell therapies with cancer. The initial phenotype of therapeutic T cells influences the ability of T cells to convert tumor cells to an inflammatory, anti-proliferative phenotype. This T cell phenotype could be preserved by structural reprogramming to facilitate continual tumor phenotype conversion and killing. One takeaway is that controlling the rate of cancer phenotype conversion is critical for control of tumor growth. The results suggest new design criteria and patient selection metrics for T cell therapies, call for a rethinking of T cell therapeutic implementation, and provide a foundation for synergistically integrating multiplexed imaging data with multiscale modeling of the cancer-immune interface.
Collapse
Affiliation(s)
- John W Hickey
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Eran Agmon
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Nina Horowitz
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Matthew Lamore
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - John Sunwoo
- Department of Otolaryngology, Head and Neck Surgery, Stanford Cancer Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Markus Covert
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
5
|
Mani N, Andrews D, Obeng RC. Modulation of T cell function and survival by the tumor microenvironment. Front Cell Dev Biol 2023; 11:1191774. [PMID: 37274739 PMCID: PMC10232912 DOI: 10.3389/fcell.2023.1191774] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/02/2023] [Indexed: 06/06/2023] Open
Abstract
Cancer immunotherapy is shifting paradigms in cancer care. T cells are an indispensable component of an effective antitumor immunity and durable clinical responses. However, the complexity of the tumor microenvironment (TME), which consists of a wide range of cells that exert positive and negative effects on T cell function and survival, makes achieving robust and durable T cell responses difficult. Additionally, tumor biology, structural and architectural features, intratumoral nutrients and soluble factors, and metabolism impact the quality of the T cell response. We discuss the factors and interactions that modulate T cell function and survive in the TME that affect the overall quality of the antitumor immune response.
Collapse
Affiliation(s)
- Nikita Mani
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Dathan Andrews
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Rebecca C. Obeng
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
6
|
Ölmez F, Oğlak SC, Ölmez ÖF, Akbayır Ö, Yılmaz E, Akgöl S, Konal M, Seyhan NA, Kinter AK. High expression of CD8 in the tumor microenvironment is associated with PD-1 expression and patient survival in high-grade serous ovarian cancer. Turk J Obstet Gynecol 2022; 19:246-256. [PMID: 36149309 PMCID: PMC9511932 DOI: 10.4274/tjod.galenos.2022.59558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objective: The current study assesses programmed death-1 (PD-1) receptor expression and CD3, CD4, and CD8 tumor-infiltrating lymphocytes (TILs) in high-grade serous ovarian cancer (HGSOC) and associates our results with neoadjuvant chemotherapy history and disease prognosis. Materials and Methods: We included cases diagnosed with primary HGSOC with biopsy or surgical resection materials in this study. The immunoreactivity of CD3, CD4, CD8, and PD1 was assessed immunohistochemically in tumor tissue. We analyzed TILs in two predetermined groups of high and low TIL. The relationships between clinical characteristics, PD-1, and TIL were assessed. by the χ(2) test or Fisher’s Exact test. We used Kaplan-Meier survival analysis and Cox proportional hazards regression model to the connection between survival and the amounts of TIL, and PD1. Results: Univariate analysis demonstrated that optimal debulking (p<0.001), early International Federation of Gynecology and Obstetrics stage (p=0.046), and higher scores of stromal CD8+ TIL expression (p=0.028) in tumor cells were all substantially correlated with longer disease-free survival (DFS), whereas the remaining variables analyzed, including PD-1 positivity, stromal CD3+, and CD4+ TILs, and intraepithelial CD3+, CD4+, and CD8+ TILs, were not correlated with DFS. Also, univariate analysis revealed that optimal debulking (p=0.010), and higher scores of stromal CD8+ TIL expression (p=0.021) in tumor cells were all substantially correlated with longer overall survival (OS). Conclusion: Higher scores of stromal CD8+ TILs are substantially correlated with DFS and OS in univariate analyses, whereas scores of stromal CD3+ and CD4+ TILs, and intraepithelial CD3+, CD4+, and CD8+ TILs are not correlated with DFS and OS in both univariate and multivariate analyses. Also, we found a significant association between PD-1 positivity and the scores of stromal CD3+ TILs and intraepithelial CD8+ TILs. However, no remarkable relationship was revealed between PD-1 positivity and the survival of HGSOC cases.
Collapse
|
7
|
Yuan C, Zhao X, Wangmo D, Alshareef D, Gates TJ, Subramanian S. Tumor models to assess immune response and tumor-microbiome interactions in colorectal cancer. Pharmacol Ther 2022; 231:107981. [PMID: 34480964 PMCID: PMC8844062 DOI: 10.1016/j.pharmthera.2021.107981] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023]
Abstract
Despite significant advances over the past 2 decades in preventive screening and therapy aimed at improving patient survival, colorectal cancer (CRC) remains the second most common cause of cancer death in the United States. The average 5-year survival rate of CRC patients with positive regional lymph nodes is only 40%, while less than 5% of patients with distant metastases survive beyond 5 years. There is a critical need to develop novel therapies that can improve overall survival in patients with poor prognoses, particularly since 60% of them are diagnosed at an advanced stage. Pertinently, immune checkpoint blockade therapy has dramatically changed how we treat CRC patients with microsatellite-instable high tumors. Furthermore, accumulating evidence shows that changes in gut microbiota are associated with the regulation of host antitumor immune response and cancer progression. Appropriate animal models are essential to deciphering the complex mechanisms of host antitumor immune response and tumor-gut microbiome metabolic interactions. Here, we discuss various mouse models of colorectal cancer that are developed to address key questions on tumor immune response and tumor-microbiota interactions. These CRC models will also serve as resourceful tools for effective preclinical studies.
Collapse
Affiliation(s)
- Ce Yuan
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Xianda Zhao
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Dechen Wangmo
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Duha Alshareef
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Travis J Gates
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Subbaya Subramanian
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
8
|
Johanson TM, Keenan CR, Allan RS. Shedding Structured Light on Molecular Immunity: The Past, Present and Future of Immune Cell Super Resolution Microscopy. Front Immunol 2021; 12:754200. [PMID: 34975842 PMCID: PMC8715013 DOI: 10.3389/fimmu.2021.754200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022] Open
Abstract
In the two decades since the invention of laser-based super resolution microscopy this family of technologies has revolutionised the way life is viewed and understood. Its unparalleled resolution, speed, and accessibility makes super resolution imaging particularly useful in examining the highly complex and dynamic immune system. Here we introduce the super resolution technologies and studies that have already fundamentally changed our understanding of a number of central immunological processes and highlight other immunological puzzles only addressable in super resolution.
Collapse
Affiliation(s)
- Timothy M. Johanson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Christine R. Keenan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Rhys S. Allan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
9
|
Designing of various biosensor devices for determination of apoptosis: A comprehensive review. Biochem Biophys Res Commun 2021; 578:42-62. [PMID: 34536828 DOI: 10.1016/j.bbrc.2021.08.089] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/29/2021] [Accepted: 08/29/2021] [Indexed: 12/30/2022]
Abstract
Apoptosis is a type of cell death caused by the occurrence of both pathological and physiological conditions triggered by ligation of death receptors outside the cell or triggered by DNA damage and/or cytoskeleton disruption. Timely monitoring of apoptosis can effectively help early diagnosis of related diseases and continuous assessment of the effectiveness of drugs. Detecting caspases, a protease family closely related to cellular apoptosis, and its identification as markers of apoptosis is a popular procedure. Biosensors are used for early diagnosis and play a very important role in preventing disease progression in various body sections. Recently, there has been a widespread increase in the desire to use materials made of paper (e.g. nitrocellulose membrane) for Point-of-Care (POC) testing systems since paper and paper-like materials are cheap, abundant and degradable. Microfluidic paper-based analytical devices (μPADs) are highly promising as they are cost-effective, easy to use, fast, precise and sustainable over time and under different environmental conditions. In this review, we focused our efforts on compiling the different approaches on identifying apoptosis pathway while giving brief information about apoptosis and biosensors. This review includes recent advantages in biosensing techniques to simply determine what happened in the cell life and which direction it would continue. As a conclusion, we believed that the review may help to researchers to compare/update the knowledge about diagnosis of the apoptosis pathway while reminding the basic definitions about the apoptosis and biosensor technologies.
Collapse
|
10
|
Beck RJ, Weigelin B, Beltman JB. Mathematical Modelling Based on In Vivo Imaging Suggests CD137-Stimulated Cytotoxic T Lymphocytes Exert Superior Tumour Control Due to an Enhanced Antimitotic Effect on Tumour Cells. Cancers (Basel) 2021; 13:cancers13112567. [PMID: 34073822 PMCID: PMC8197176 DOI: 10.3390/cancers13112567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 01/12/2023] Open
Abstract
Simple Summary Cytotoxic T lymphocytes (CTLs) play an important role in controlling tumours, and an improved understanding of how they accomplish this will benefit immunotherapeutic cancer treatment strategies. Stimulation of CTLs by targeting their CD137 receptor is a strategy currently under investigation for enhancing responses against tumours, yet so far only limited quantitative knowledge regarding the effects of such stimulation upon CTLs has been obtained. Here, we develop mathematical models to describe dynamic in vivo two-photon imaging of tumour infiltrating CTLs, to characterise differences in their function either in the presence or absence of a CD137 agonist antibody. We showed that an increased antiproliferative effect and a more sustained presence of CTLs within the tumour were the most important effects associated with anti-CD137 treatment. Abstract Several immunotherapeutic strategies for the treatment of cancer are under development. Two prominent strategies are adoptive cell transfer (ACT) of CTLs and modulation of CTL function with immune checkpoint inhibitors or with costimulatory antibodies. Despite some success with these approaches, there remains a lack of detailed and quantitative descriptions of the events following CTL transfer and the impact of immunomodulation. Here, we have applied ordinary differential equation models to two photon imaging data derived from a B16F10 murine melanoma. Models were parameterised with data from two different treatment conditions: either ACT-only, or ACT with intratumoural costimulation using a CD137 targeted antibody. Model dynamics and best fitting parameters were compared, in order to assess the mode of action of the CTLs and examine how the CD137 antibody influenced their activities. We found that the cytolytic activity of the transferred CTLs was minimal without CD137 costimulation, and that the CD137 targeted antibody did not enhance the per-capita killing ability of the transferred CTLs. Instead, the results of our modelling study suggest that an antiproliferative effect of CTLs exerted upon the tumour likely accounted for the majority of the reduction in tumour growth after CTL transfer. Moreover, we found that CD137 most likely improved tumour control via enhancement of this antiproliferative effect, as well as prolonging the period in which CTLs were inside the tumour, leading to a sustained duration of their antitumour effects following CD137 stimulation.
Collapse
Affiliation(s)
- Richard J. Beck
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden University, 2333 CC Leiden, The Netherlands;
| | - Bettina Weigelin
- Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany;
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
| | - Joost B. Beltman
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden University, 2333 CC Leiden, The Netherlands;
- Correspondence:
| |
Collapse
|
11
|
Lucca LE, Axisa PP, Lu B, Harnett B, Jessel S, Zhang L, Raddassi K, Zhang L, Olino K, Clune J, Singer M, Kluger HM, Hafler DA. Circulating clonally expanded T cells reflect functions of tumor-infiltrating T cells. J Exp Med 2021; 218:e20200921. [PMID: 33651881 PMCID: PMC7933991 DOI: 10.1084/jem.20200921] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/14/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Understanding the relationship between tumor and peripheral immune environments could allow longitudinal immune monitoring in cancer. Here, we examined whether T cells that share the same TCRαβ and are found in both tumor and blood can be interrogated to gain insight into the ongoing tumor T cell response. Paired transcriptome and TCRαβ repertoire of circulating and tumor-infiltrating T cells were analyzed at the single-cell level from matched tumor and blood from patients with metastatic melanoma. We found that in circulating T cells matching clonally expanded tumor-infiltrating T cells (circulating TILs), gene signatures of effector functions, but not terminal exhaustion, reflect those observed in the tumor. In contrast, features of exhaustion are displayed predominantly by tumor-exclusive T cells. Finally, genes associated with a high degree of blood-tumor TCR sharing were overexpressed in tumor tissue after immunotherapy. These data demonstrate that circulating TILs have unique transcriptional patterns that may have utility for the interrogation of T cell function in cancer immunotherapy.
Collapse
Affiliation(s)
- Liliana E. Lucca
- Department of Neurology and Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Pierre-Paul Axisa
- Department of Neurology and Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Benjamin Lu
- Department of Neurology and Department of Immunobiology, Yale School of Medicine, New Haven, CT
- Department of Medicine, Yale School of Medicine, New Haven, CT
| | - Brian Harnett
- Department of Neurology and Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Shlomit Jessel
- Department of Medicine, Yale School of Medicine, New Haven, CT
| | - Le Zhang
- Department of Neurology and Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Khadir Raddassi
- Department of Neurology and Department of Immunobiology, Yale School of Medicine, New Haven, CT
| | - Lin Zhang
- Department of Medicine, Yale School of Medicine, New Haven, CT
| | - Kelly Olino
- Department of Surgery, Yale School of Medicine, New Haven, CT
| | - James Clune
- Department of Surgery, Yale School of Medicine, New Haven, CT
| | - Meromit Singer
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | | | - David A. Hafler
- Department of Neurology and Department of Immunobiology, Yale School of Medicine, New Haven, CT
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| |
Collapse
|
12
|
Upadhyay R, Boiarsky JA, Pantsulaia G, Svensson-Arvelund J, Lin MJ, Wroblewska A, Bhalla S, Scholler N, Bot A, Rossi JM, Sadek N, Parekh S, Lagana A, Baccarini A, Merad M, Brown BD, Brody JD. A Critical Role for Fas-Mediated Off-Target Tumor Killing in T-cell Immunotherapy. Cancer Discov 2020; 11:599-613. [PMID: 33334730 DOI: 10.1158/2159-8290.cd-20-0756] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/11/2020] [Accepted: 11/19/2020] [Indexed: 12/15/2022]
Abstract
T cell-based therapies have induced cancer remissions, though most tumors ultimately progress, reflecting inherent or acquired resistance including antigen escape. Better understanding of how T cells eliminate tumors will help decipher resistance mechanisms. We used a CRISPR/Cas9 screen and identified a necessary role for Fas-FasL in antigen-specific T-cell killing. We also found that Fas-FasL mediated off-target "bystander" killing of antigen-negative tumor cells. This localized bystander cytotoxicity enhanced clearance of antigen-heterogeneous tumors in vivo, a finding that has not been shown previously. Fas-mediated on-target and bystander killing was reproduced in chimeric antigen receptor (CAR-T) and bispecific antibody T-cell models and was augmented by inhibiting regulators of Fas signaling. Tumoral FAS expression alone predicted survival of CAR-T-treated patients in a large clinical trial (NCT02348216). These data suggest strategies to prevent immune escape by targeting both the antigen expression of most tumor cells and the geography of antigen-loss variants. SIGNIFICANCE: This study demonstrates the first report of in vivo Fas-dependent bystander killing of antigen-negative tumors by T cells, a phenomenon that may be contributing to the high response rates of antigen-directed immunotherapies despite tumoral heterogeneity. Small molecules that target the Fas pathway may potentiate this mechanism to prevent cancer relapse.This article is highlighted in the In This Issue feature, p. 521.
Collapse
Affiliation(s)
- Ranjan Upadhyay
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jonathan A Boiarsky
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gvantsa Pantsulaia
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Judit Svensson-Arvelund
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Matthew J Lin
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aleksandra Wroblewska
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sherry Bhalla
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | - Norah Sadek
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Samir Parekh
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alessandro Lagana
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alessia Baccarini
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Miriam Merad
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Brian D Brown
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joshua D Brody
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York. .,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
13
|
Zheng W, Ranoa DRE, Huang X, Hou Y, Yang K, Poli EC, Beckett MA, Fu YX, Weichselbaum RR. RIG-I-Like Receptor LGP2 Is Required for Tumor Control by Radiotherapy. Cancer Res 2020; 80:5633-5641. [PMID: 33087322 DOI: 10.1158/0008-5472.can-20-2324] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/11/2020] [Accepted: 10/15/2020] [Indexed: 11/16/2022]
Abstract
Dendritic cells (DC) play an essential role in innate immunity and radiation-elicited immune responses. LGP2 is a RIG-I-like receptor involved in cytoplasmic RNA recognition and antiviral responses. Although LGP2 has also been linked to cell survival of both tumor cells and T cells, the role of LGP2 in mediating DC function and antitumor immunity elicited by radiotherapy remains unclear. Here, we report that tumor DCs are linked to the clinical outcome of patients with breast cancer who received radiotherapy, and the presence of DC correlates with gene expression of LGP2 in the tumor microenvironment. In preclinical models, host LGP2 was essential for optimal antitumor control by ionizing radiation (IR). The absence of LGP2 in DC dampened type I IFN production and the priming capacity of DC. In the absence of LGP2, MDA5-mediated activation of type I IFN signaling was abrogated. The MDA5/LGP2 agonist high molecular weight poly I:C improved the antitumor effect of IR. This study reveals a previously undefined role of LGP2 in host immunity and provides a new strategy to improve the efficacy of radiotherapy. SIGNIFICANCE: These findings reveal an essential role of LGP2 in promoting antitumor immunity after radiotherapy and provide a new strategy to enhance radiotherapy.
Collapse
Affiliation(s)
- Wenxin Zheng
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Diana Rose E Ranoa
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Xiaona Huang
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Yuzhu Hou
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | | | - Michael A Beckett
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas.
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois.
| |
Collapse
|
14
|
Diehl MI, Wolf SP, Bindokas VP, Schreiber H. Automated cell cluster analysis provides insight into multi-cell-type interactions between immune cells and their targets. Exp Cell Res 2020; 393:112014. [PMID: 32439494 DOI: 10.1016/j.yexcr.2020.112014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/28/2020] [Accepted: 04/15/2020] [Indexed: 11/18/2022]
Abstract
Understanding interactions between immune cells and their targets is an important step on the path to fully characterizing the immune system, and in doing so, learning how it combats disease. Many studies of these interactions have a narrow focus, often looking only at a binary result of whether or not a specific treatment was successful or only focusing on the interactions between two individual cells. Therefore, in an effort to more comprehensively study multicellular interactions among immune cells and their targets, we used in vitro longitudinal time-lapse imaging and developed an automated cell cluster analysis tool, or macro, to investigate the formation of cell clusters. In particular, we investigated the behavior of cancer-specific CD8+ and CD4+ T cells on how they interact around their targets: cancer cells and antigen-presenting cells. The macro that we established allowed us to examine these large-scale clustering behaviors taking place between those four cell types. Thus, we were able to distinguish directed immune cell clustering from random cell movement. Furthermore, this macro can be generalized to be applicable to systems consisting of any number of differently labeled species and can be used to track clustering behaviors and compare them to randomized simulations.
Collapse
Affiliation(s)
- Markus I Diehl
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA
| | - Steven P Wolf
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA; Institute of Immunology, Campus Buch, Charité - Universitaetsmedizin Berlin, 13125, Berlin, Germany
| | - Vytas P Bindokas
- Integrated Microscopy Core, The University of Chicago, Chicago, IL, 60637, USA
| | - Hans Schreiber
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA; Committee on Cancer Biology and Committee on Immunology, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
15
|
Kimm MA, Tzoumas S, Glasl S, Omar M, Symvoulidis P, Olefir I, Rummeny EJ, Meier R, Ntziachristos V. Longitudinal imaging of T cell-based immunotherapy with multi-spectral, multi-scale optoacoustic tomography. Sci Rep 2020; 10:4903. [PMID: 32184401 PMCID: PMC7078227 DOI: 10.1038/s41598-020-61191-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/28/2020] [Indexed: 12/31/2022] Open
Abstract
Most imaging studies of immunotherapy have focused on tracking labeled T cell biodistribution in vivo for understanding trafficking and homing parameters and predicting therapeutic efficacy by the presence of transferred T cells at or in the tumour mass. Conversely, we investigate here a novel concept for longitudinally elucidating anatomical and pathophysiological changes of solid tumours after adoptive T cell transfer in a preclinical set up, using previously unexplored in-tandem macroscopic and mesoscopic optoacoustic (photoacoustic) imaging. We show non-invasive in vivo observations of vessel collapse during tumour rejection across entire tumours and observe for the first time longitudinal tumour rejection in a label-free manner based on optical absorption changes in the tumour mass due to cellular decline. We complement these observations with high resolution episcopic fluorescence imaging of T cell biodistribution using optimized T cell labeling based on two near-infrared dyes targeting the cell membrane and the cytoplasm. We discuss how optoacoustic macroscopy and mesoscopy offer unique contrast and immunotherapy insights, allowing label-free and longitudinal observations of tumour therapy. The results demonstrate optoacoustic imaging as an invaluable tool in understanding and optimizing T cell therapy.
Collapse
Affiliation(s)
- Melanie A Kimm
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stratis Tzoumas
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Sarah Glasl
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Murad Omar
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Panagiotis Symvoulidis
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ivan Olefir
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ernst J Rummeny
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Reinhard Meier
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Vasilis Ntziachristos
- Chair for Biological Imaging, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, Munich, Germany. .,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
16
|
Arina A, Beckett M, Fernandez C, Zheng W, Pitroda S, Chmura SJ, Luke JJ, Forde M, Hou Y, Burnette B, Mauceri H, Lowy I, Sims T, Khodarev N, Fu YX, Weichselbaum RR. Tumor-reprogrammed resident T cells resist radiation to control tumors. Nat Commun 2019; 10:3959. [PMID: 31477729 PMCID: PMC6718618 DOI: 10.1038/s41467-019-11906-2] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
Successful combinations of radiotherapy and immunotherapy depend on the presence of live T cells within the tumor; however, radiotherapy is believed to damage T cells. Here, based on longitudinal in vivo imaging and functional analysis, we report that a large proportion of T cells survive clinically relevant doses of radiation and show increased motility, and higher production of interferon gamma, compared with T cells from unirradiated tumors. Irradiated intratumoral T cells can mediate tumor control without newly-infiltrating T cells. Transcriptomic analysis suggests T cell reprogramming in the tumor microenvironment and similarities with tissue-resident memory T cells, which are more radio-resistant than circulating/lymphoid tissue T cells. TGFβ is a key upstream regulator of T cell reprogramming and contributes to intratumoral Tcell radio-resistance. These findings have implications for the design of radio-immunotherapy trials in that local irradiation is not inherently immunosuppressive, and irradiation of multiple tumors might optimize systemic effects of radiotherapy.
Collapse
Affiliation(s)
- Ainhoa Arina
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA.
| | - Michael Beckett
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Christian Fernandez
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Wenxin Zheng
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Sean Pitroda
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Steven J Chmura
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Jason J Luke
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Martin Forde
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Yuzhu Hou
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Byron Burnette
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Helena Mauceri
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Israel Lowy
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Tasha Sims
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Nikolai Khodarev
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
17
|
Wei D, Zeng X, Yang Z, Zhou Q, Weng X, He H, Gao W, Gu Z, Wei X. Visualizing Interactions of Circulating Tumor Cell and Dendritic Cell in the Blood Circulation Using In Vivo Imaging Flow Cytometry. IEEE Trans Biomed Eng 2019; 66:2521-2526. [DOI: 10.1109/tbme.2019.2891068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
18
|
Manaster Y, Shipony Z, Hutzler A, Kolesnikov M, Avivi C, Shalmon B, Barshack I, Besser MJ, Feferman T, Shakhar G. Reduced CTL motility and activity in avascular tumor areas. Cancer Immunol Immunother 2019; 68:1287-1301. [PMID: 31253998 PMCID: PMC11028152 DOI: 10.1007/s00262-019-02361-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 06/16/2019] [Indexed: 01/04/2023]
Abstract
Patchy infiltration of tumors by cytotoxic T cells (CTLs) predicts poorer prognosis for cancer patients. The factors limiting intratumoral CTL dissemination, though, are poorly understood. To study CTL dissemination in tumors, we histologically examined human melanoma samples and used mice to image B16-OVA tumors infiltrated by OT-I CTLs using intravital two-photon microscopy. In patients, most CTLs concentrated around peripheral blood vessels, especially in poorly infiltrated tumors. In mice, OT-I CTLs had to cluster around tumor cells to efficiently kill them in a contact-and perforin-dependent manner and cytotoxicity was strictly antigen-specific. OT-I CTLs as well as non-specific CTLs concentrated around peripheral vessels, and cleared the tumor cells around them. This was also the case when CTLs were injected directly into the tumors. CTLs crawled rapidly only in areas within 50 µm of flowing blood vessels and transient occlusion of vessels immediately, though reversibly, stopped their migration. In vitro, oxygen depletion and blockade of oxidative phosphorylation also reduced CTL motility. Taken together, these results suggest that hypoxia limits CTL migration away from blood vessels, providing immune-privileged niches for tumor cells to survive. Normalizing intratumoral vasculature may thus synergize with tumor immunotherapy.
Collapse
Affiliation(s)
- Yoav Manaster
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel
| | - Zohar Shipony
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel
| | - Anat Hutzler
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel
| | - Masha Kolesnikov
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel
| | - Camila Avivi
- Department of Pathology, Sheba Medical Center, Derech Sheba 2, 52621, Ramat Gan, Israel
| | - Bruria Shalmon
- Department of Pathology, Sheba Medical Center, Derech Sheba 2, 52621, Ramat Gan, Israel
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Derech Sheba 2, 52621, Ramat Gan, Israel
| | - Michal J Besser
- Ella Institute, Sheba Medical Center, Derech Sheba 2, 52621, Ramat Gan, Israel
- Department of Human Microbiology and Immunology, Sackler Medical School, Tel-Aviv University, 35 Klachkin st, 6997801, Tel Aviv, Israel
| | - Tali Feferman
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel.
| | - Guy Shakhar
- Department of Immunology, Weizmann Institute of Science, Wolfson Bldg., 234 Herzl St., 76100, Rehovot, Israel.
| |
Collapse
|
19
|
Hwang C, Lee SJ, Lee JH, Kim KH, Suh DS, Kwon BS, Choi KU. Stromal tumor-infiltrating lymphocytes evaluated on H&E-stained slides are an independent prognostic factor in epithelial ovarian cancer and ovarian serous carcinoma. Oncol Lett 2019; 17:4557-4565. [PMID: 30944645 PMCID: PMC6444488 DOI: 10.3892/ol.2019.10095] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/01/2019] [Indexed: 11/30/2022] Open
Abstract
Studies on tumor-infiltrating lymphocytes (TILs) in epithelial ovarian cancer (EOC) have focused on the clinical significance of inflammatory cells of specific subtypes that are identifiable using immunohistochemistry. However, the subtypes of inflammatory cells that reportedly affect patient survival and prognosis have differed from study to study, and few studies have examined TILs using hematoxylin and eosin (H&E) staining. Therefore, the present study aimed to identify the clinical importance of general stromal TILs in EOC by using H&E staining to apply breast cancer recommendations from the International TILs Working Group 2014 on breast cancer using H&E staining. Stromal TILs in 256 EOC cases from Pusan National University Hospital and 475 cases of high-grade serous carcinoma from The Cancer Genome Atlas dataset were assessed. Stromal TILs were evaluated using H&E-stained slides according to the breast cancer recommendations of the International Working Group 2014, and patients were classified into low and high stromal TIL groups according to their stromal TIL values. The associations of these groups with clinicopathologic factors were assessed, and it was confirmed that group membership correlated with survival and prognosis. According to the χ2 assessment, the stromal TIL group was associated with tumor grade. Furthermore, the stromal TIL group was associated with overall survival according to Kaplan-Meier analysis with the log-rank test. Finally, the stromal TIL group was an independent prognostic factor according to univariate and multivariate Cox regression analyses. In cases of EOC, the evaluation of general stromal TILs on H&E-stained slides could be used to predict prognosis.
Collapse
Affiliation(s)
- Chungsu Hwang
- Department of Pathology, Pusan National University Yangsan Hospital, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - So Jung Lee
- Department of Pathology, Pusan National University Hospital, Seo-Gu, Busan 602-739, Republic of Korea
| | - Jung Hee Lee
- Department of Pathology, Pusan National University Yangsan Hospital, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
| | - Ki Hyung Kim
- Department of Obstetrics and Gynecology, Pusan National University Hospital, Seo-Gu, Busan 602-739, Republic of Korea
| | - Dong Soo Suh
- Department of Obstetrics and Gynecology, Pusan National University Hospital, Seo-Gu, Busan 602-739, Republic of Korea
| | - Byung-Su Kwon
- Department of Obstetrics and Gynecology, Pusan National University Hospital, Seo-Gu, Busan 602-739, Republic of Korea
| | - Kyung Un Choi
- Department of Pathology, Pusan National University Hospital, Seo-Gu, Busan 602-739, Republic of Korea
| |
Collapse
|
20
|
Beck RJ, Slagter M, Beltman JB. Contact-Dependent Killing by Cytotoxic T Lymphocytes Is Insufficient for EL4 Tumor Regression In Vivo. Cancer Res 2019; 79:3406-3416. [PMID: 31040155 DOI: 10.1158/0008-5472.can-18-3147] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 02/08/2019] [Accepted: 04/25/2019] [Indexed: 11/16/2022]
Abstract
Immunotherapies are an emerging strategy for treatment of solid tumors. Improved understanding of the mechanisms employed by cytotoxic T lymphocytes (CTL) to control tumors will aid in the development of immunotherapies. CTLs can directly kill tumor cells in a contact-dependent manner or may exert indirect effects on tumor cells via secretion of cytokines. Here, we aim to quantify the importance of these mechanisms in murine thymoma EL4/EG7 cells. We developed an agent-based model (ABM) and an ordinary differential equation model of tumor regression after adoptive transfer of a population of CTLs. Models were parameterized based on in vivo measurements of CTL infiltration and killing rates applied to EL4/EG7 tumors and OTI T cells. We quantified whether infiltrating CTLs are capable of controlling tumors through only direct, contact-dependent killing. Both models agreed that the low measured killing rate of CTLs in vivo was insufficient to cause tumor regression. In our ABM, we also simulated CTL production of the cytokine IFNγ in order to explore how an antiproliferative effect of IFNγ might aid CTLs in tumor control. In this model, IFNγ substantially reduced tumor growth compared with direct killing alone. Collectively, these data demonstrate that contact-dependent killing is insufficient for EL4 regression in vivo and highlight the potential importance of cytokine-induced antiproliferative effects in T-cell-mediated tumor control. SIGNIFICANCE: Computational modeling highlights the importance of cytokine-induced antiproliferative effects in T-cell-mediated control of tumor progression.
Collapse
Affiliation(s)
- Richard J Beck
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Maarten Slagter
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Joost B Beltman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
21
|
Johansson-Percival A, He B, Ganss R. Immunomodulation of Tumor Vessels: It Takes Two to Tango. Trends Immunol 2018; 39:801-814. [DOI: 10.1016/j.it.2018.08.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/03/2018] [Accepted: 08/03/2018] [Indexed: 12/25/2022]
|
22
|
Zheng W, Skowron KB, Namm JP, Burnette B, Fernandez C, Arina A, Liang H, Spiotto MT, Posner MC, Fu YX, Weichselbaum RR. Combination of radiotherapy and vaccination overcomes checkpoint blockade resistance. Oncotarget 2018; 7:43039-43051. [PMID: 27343548 PMCID: PMC5190006 DOI: 10.18632/oncotarget.9915] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/20/2016] [Indexed: 01/19/2023] Open
Abstract
The majority of cancer patients respond poorly to either vaccine or checkpoint blockade, and even to the combination of both. They are often resistant to high doses of radiation therapy as well. We examined prognostic markers of immune cell infiltration in pancreatic cancer. Patients with low CD8+ T cell infiltration and high PD-L1 expression (CD8+ TloPD-L1hi) experienced poor outcomes. We developed a mouse tumor fragment model with a trackable model antigen (SIYRYYGL or SIY) to mimic CD8+ TloPD-L1hi cancers. Tumors arising from fragments contained few T cells, even after vaccination. Fragment tumors responded poorly to PD-L1 blockade, SIY vaccination or radiation individually. By contrast, local ionizing radiation coupled with vaccination increased CD8+ T cell infiltration that was associated with upregulation of CXCL10 and CCL5 chemokines in the tumor, but demonstrated modest inhibition of tumor growth. The addition of an anti-PD-L1 antibody enhanced the effector function of tumor-infiltrating T cells, leading to significantly improved tumor regression and increased survival compared to vaccination and radiation. These results indicate that sequential combination of radiation, vaccination and checkpoint blockade converts non-T cell-inflamed cancers to T cell-inflamed cancers, and mediates regression of established pancreatic tumors with an initial CD8+ TloPD-L1hi phenotype. This study has opened a new strategy for shifting “cold” to hot tumors that will respond to immunotherapy.
Collapse
Affiliation(s)
- Wenxin Zheng
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA.,The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Kinga B Skowron
- Department of Surgery, University of Chicago, Chicago, IL, USA.,The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Jukes P Namm
- Department of Surgery, University of Chicago, Chicago, IL, USA.,The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA.,Department of Surgery, Loma Linda University Health, Loma Linda, CA, USA
| | - Byron Burnette
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA.,The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Christian Fernandez
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA.,The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Ainhoa Arina
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA.,The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Hua Liang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA.,The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Michael T Spiotto
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA.,The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | | | - Yang-Xin Fu
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA.,Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA.,The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| |
Collapse
|
23
|
Spranger S, Dai D, Horton B, Gajewski TF. Tumor-Residing Batf3 Dendritic Cells Are Required for Effector T Cell Trafficking and Adoptive T Cell Therapy. Cancer Cell 2017; 31:711-723.e4. [PMID: 28486109 PMCID: PMC5650691 DOI: 10.1016/j.ccell.2017.04.003] [Citation(s) in RCA: 983] [Impact Index Per Article: 140.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 02/16/2017] [Accepted: 04/04/2017] [Indexed: 12/18/2022]
Abstract
Effector T cells have the capability of recognizing and killing cancer cells. However, whether tumors can become immune resistant through exclusion of effector T cells from the tumor microenvironment is not known. By using a tumor model resembling non-T cell-inflamed human tumors, we assessed whether adoptive T cell transfer might overcome failed spontaneous priming. Flow cytometric assays combined with intra-vital imaging indicated failed trafficking of effector T cells into tumors. Mechanistically, this was due to the absence of CXCL9/10, which we found to be produced by CD103+ dendritic cells (DCs) in T cell-inflamed tumors. Our data indicate that lack of CD103+ DCs within the tumor microenvironment dominantly resists the effector phase of an anti-tumor T cell response, contributing to immune escape.
Collapse
Affiliation(s)
- Stefani Spranger
- Department of Pathology, The University of Chicago, 5841 South Maryland Avenue, MC2115, Chicago, IL 60637, USA
| | - Daisy Dai
- Department of Pathology, The University of Chicago, 5841 South Maryland Avenue, MC2115, Chicago, IL 60637, USA
| | - Brendan Horton
- Department of Pathology, The University of Chicago, 5841 South Maryland Avenue, MC2115, Chicago, IL 60637, USA
| | - Thomas F Gajewski
- Department of Pathology, The University of Chicago, 5841 South Maryland Avenue, MC2115, Chicago, IL 60637, USA; Department of Medicine, The University of Chicago, 5841 South Maryland Avenue, MC2115, Chicago, IL 60637, USA.
| |
Collapse
|
24
|
Kammertoens T, Friese C, Arina A, Idel C, Briesemeister D, Rothe M, Ivanov A, Szymborska A, Patone G, Kunz S, Sommermeyer D, Engels B, Leisegang M, Textor A, Fehling HJ, Fruttiger M, Lohoff M, Herrmann A, Yu H, Weichselbaum R, Uckert W, Hübner N, Gerhardt H, Beule D, Schreiber H, Blankenstein T. Tumour ischaemia by interferon-γ resembles physiological blood vessel regression. Nature 2017; 545:98-102. [PMID: 28445461 PMCID: PMC5567674 DOI: 10.1038/nature22311] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/30/2017] [Indexed: 12/11/2022]
Abstract
The relative contribution of the effector molecules produced by T cells to tumour rejection is unclear, but interferon-γ (IFNγ) is critical in most of the analysed models. Although IFNγ can impede tumour growth by acting directly on cancer cells, it must also act on the tumour stroma for effective rejection of large, established tumours. However, which stroma cells respond to IFNγ and by which mechanism IFNγ contributes to tumour rejection through stromal targeting have remained unknown. Here we use a model of IFNγ induction and an IFNγ-GFP fusion protein in large, vascularized tumours growing in mice that express the IFNγ receptor exclusively in defined cell types. Responsiveness to IFNγ by myeloid cells and other haematopoietic cells, including T cells or fibroblasts, was not sufficient for IFNγ-induced tumour regression, whereas responsiveness of endothelial cells to IFNγ was necessary and sufficient. Intravital microscopy revealed IFNγ-induced regression of the tumour vasculature, resulting in arrest of blood flow and subsequent collapse of tumours, similar to non-haemorrhagic necrosis in ischaemia and unlike haemorrhagic necrosis induced by tumour necrosis factor. The early events of IFNγ-induced tumour ischaemia resemble non-apoptotic blood vessel regression during development, wound healing or IFNγ-mediated, pregnancy-induced remodelling of uterine arteries. A better mechanistic understanding of how solid tumours are rejected may aid the design of more effective protocols for adoptive T-cell therapy.
Collapse
Affiliation(s)
- Thomas Kammertoens
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Christian Friese
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Ainhoa Arina
- Department of Radiation and Cellular Oncology, Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637, USA
| | - Christian Idel
- Department of Pathology, The University of Chicago, Chicago, Illinois 60637, USA
| | - Dana Briesemeister
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Michael Rothe
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Andranik Ivanov
- Berlin Institute of Health, 10117 Berlin, Germany
- Charité - Universitätsmedizin, 10117 Berlin, Germany
| | - Anna Szymborska
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Giannino Patone
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Severine Kunz
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | | | - Boris Engels
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Matthias Leisegang
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health, 10117 Berlin, Germany
| | - Ana Textor
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | | | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Michael Lohoff
- Institute for Medical Microbiology, University of Marburg, 35032 Marburg, Germany
| | - Andreas Herrmann
- Beckman Research Institute at the Comprehensive Cancer Center City of Hope, Los Angeles, California 91010-3000, USA
| | - Hua Yu
- Beckman Research Institute at the Comprehensive Cancer Center City of Hope, Los Angeles, California 91010-3000, USA
| | - Ralph Weichselbaum
- Department of Radiation and Cellular Oncology, Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637, USA
| | - Wolfgang Uckert
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health, 10117 Berlin, Germany
| | - Norbert Hübner
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Charité - Universitätsmedizin, 10117 Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, 13347 Berlin, Germany
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health, 10117 Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, 13347 Berlin, Germany
| | - Dieter Beule
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health, 10117 Berlin, Germany
| | - Hans Schreiber
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Department of Pathology, The University of Chicago, Chicago, Illinois 60637, USA
- Berlin Institute of Health, 10117 Berlin, Germany
| | - Thomas Blankenstein
- Institute of Immunology, Charité Campus Buch, 13125 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health, 10117 Berlin, Germany
| |
Collapse
|
25
|
Torcellan T, Stolp J, Chtanova T. In Vivo Imaging Sheds Light on Immune Cell Migration and Function in Cancer. Front Immunol 2017; 8:309. [PMID: 28382036 PMCID: PMC5360706 DOI: 10.3389/fimmu.2017.00309] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/06/2017] [Indexed: 01/04/2023] Open
Abstract
There is ample evidence for both beneficial and harmful involvement of the immune system in tumor development and spread. Immune cell recruitment to tumors is essential not only for the success of anticancer immune therapies but also for tumor-induced immune suppression. Now that immune-based therapies are playing an increasingly important role in treatment of solid tumors such as metastatic melanomas, precise analysis of the in vivo contributions of different leukocyte subsets in tumor immunity has become an even greater priority. Recently, this goal has been markedly facilitated by the use of intravital microscopy, which has enabled us to visualize the dynamic interactions between cells of the immune system and tumor targets in the context of the tumor microenvironment. For example, intravital imaging techniques have shed new light on T cell infiltration of tumors, the mechanisms of cancer cell killing, and how myeloid cells contribute to tumor tolerance and spread. This mini-review summarizes the recent advances made to our understanding of the roles of innate and adaptive immune cells in cancer based on the use of these in vivo imaging approaches.
Collapse
Affiliation(s)
- Tommaso Torcellan
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Jessica Stolp
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Tatyana Chtanova
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
26
|
Qi S, Li H, Lu L, Qi Z, Liu L, Chen L, Shen G, Fu L, Luo Q, Zhang Z. Long-term intravital imaging of the multicolor-coded tumor microenvironment during combination immunotherapy. eLife 2016; 5. [PMID: 27855783 PMCID: PMC5173323 DOI: 10.7554/elife.14756] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 10/17/2016] [Indexed: 12/22/2022] Open
Abstract
The combined-immunotherapy of adoptive cell therapy (ACT) and cyclophosphamide (CTX) is one of the most efficient treatments for melanoma patients. However, no synergistic effects of CTX and ACT on the spatio-temporal dynamics of immunocytes in vivo have been described. Here, we visualized key cell events in immunotherapy-elicited immunoreactions in a multicolor-coded tumor microenvironment, and then established an optimal strategy of metronomic combined-immunotherapy to enhance anti-tumor efficacy. Intravital imaging data indicated that regulatory T cells formed an 'immunosuppressive ring' around a solid tumor. The CTX-ACT combined-treatment elicited synergistic immunoreactions in tumor areas, which included relieving the immune suppression, triggering the transient activation of endogenous tumor-infiltrating immunocytes, increasing the accumulation of adoptive cytotoxic T lymphocytes, and accelerating the infiltration of dendritic cells. These insights into the spatio-temporal dynamics of immunocytes are beneficial for optimizing immunotherapy and provide new approaches for elucidating the mechanisms underlying the involvement of immunocytes in cancer immunotherapy. DOI:http://dx.doi.org/10.7554/eLife.14756.001 Melanoma is a form of skin cancer that is particularly difficult to treat. A new approach that has shown a lot of promise in treating many different cancers, including melanoma, is called “immunotherapy”. This technique harnesses the immune system – the body’s natural defences that help to protect against infections and disease – to combat cancer. One powerful type of immunotherapy involves injecting patients with cells called lymphocytes, which form part of the immune system. This is known as adoptive cell therapy and can activate the immune system to fight cancer, helping to shrink tumors. This treatment can be made even more powerful by combining it with a drug called cyclophosphamide and this combination, known as CTX-ACT, is currently one of the most efficient treatments for melanoma. Yet, little information is available to indicate why this treatment is so effective. Using mice implanted with melanoma cells, Qi, Li et al. sought to understand how CTX-ACT treatment works, with the goal of optimising it to increase its success. The results showed that a protective barrier of immune cells that suppresses the anti-tumor immune response – called an “immunosuppressive ring” – surrounds untreated tumors. CTX-ACT treatment can breakdown these rings, helping to reactivate the anti-tumor immune reaction in the tumors. This allows both the injected and mouse’s own immune cells to move into the tumor and destroy cancer cells. Qi, Li et al. used their findings to optimise treatment and succeeded in controlling tumor growth in the mice for several weeks. These new insights could be used to improve current immunotherapies, and offer new approaches for investigating the involvement of immune cells in the treatment of a wide range of different cancers. DOI:http://dx.doi.org/10.7554/eLife.14756.002
Collapse
Affiliation(s)
- Shuhong Qi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Lisen Lu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Zhongyang Qi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China.,Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanxin Shen
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Fu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihong Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Textor A, Schmidt K, Kloetzel PM, Weißbrich B, Perez C, Charo J, Anders K, Sidney J, Sette A, Schumacher TNM, Keller C, Busch DH, Seifert U, Blankenstein T. Preventing tumor escape by targeting a post-proteasomal trimming independent epitope. J Exp Med 2016; 213:2333-2348. [PMID: 27697836 PMCID: PMC5068242 DOI: 10.1084/jem.20160636] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/31/2016] [Indexed: 02/05/2023] Open
Abstract
Blankenstein and colleagues describe a novel strategy to avoid tumor escape from adoptive T cell therapy. Adoptive T cell therapy (ATT) can achieve regression of large tumors in mice and humans; however, tumors frequently recur. High target peptide-major histocompatibility complex-I (pMHC) affinity and T cell receptor (TCR)-pMHC affinity are thought to be critical to preventing relapse. Here, we show that targeting two epitopes of the same antigen in the same cancer cells via monospecific T cells, which have similar pMHC and pMHC-TCR affinity, results in eradication of large, established tumors when targeting the apparently subdominant but not the dominant epitope. Only the escape but not the rejection epitope required postproteasomal trimming, which was regulated by IFN-γ, allowing IFN-γ–unresponsive cancer variants to evade. The data describe a novel immune escape mechanism and better define suitable target epitopes for ATT.
Collapse
Affiliation(s)
- Ana Textor
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Karin Schmidt
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany.,Institute for Biochemistry, Charité, Campus Mitte, 10117 Berlin, Germany
| | - Peter-M Kloetzel
- Institute for Biochemistry, Charité, Campus Mitte, 10117 Berlin, Germany.,Berlin Institute of Health, 10117 Berlin, Germany
| | - Bianca Weißbrich
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University, 81675 Munich, Germany
| | - Cynthia Perez
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Jehad Charo
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Kathleen Anders
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | - John Sidney
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Ton N M Schumacher
- The Division of Immunology, The Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Christin Keller
- Institute for Biochemistry, Charité, Campus Mitte, 10117 Berlin, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University, 81675 Munich, Germany
| | - Ulrike Seifert
- Institute for Biochemistry, Charité, Campus Mitte, 10117 Berlin, Germany.,Institute for Molecular and Clinical Immunology, Otto-von-Guericke-Universität, 39120 Magdeburg, Germany.,Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Thomas Blankenstein
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany .,Berlin Institute of Health, 10117 Berlin, Germany.,Institute of Immunology, Charité, Campus Buch, 13125 Berlin, Germany
| |
Collapse
|
28
|
Teijeira A, Etxeberria I, Ponz-Sarvise M, Melero I. Immunotherapy of Cancer Visualized by Live Microscopy: Seeing Is Believing. Clin Cancer Res 2016; 22:4277-9. [PMID: 27330056 DOI: 10.1158/1078-0432.ccr-16-1072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 05/13/2016] [Indexed: 11/16/2022]
Abstract
The success of immunotherapy of cancer depends on several cellular events in the tumors that can be visualized by live microscopy strategies in experimental models. Taking advantage of advanced microscopy techniques, Lehmann and colleagues explore in this issue of CCR the mechanism of action of a novel bispecific mAb (TCB-CEA) that targets membrane-bound CEA and CD3ε. Clin Cancer Res; 22(17); 4277-9. ©2016 AACRSee related article by Lehmann et al., p. 4417.
Collapse
Affiliation(s)
- Alvaro Teijeira
- Immunology and Immunotherapy Department, CIMA, Pamplona, Spain
| | | | - Mariano Ponz-Sarvise
- Oncology Department, University Clinic, University of Navarra and Instituto de Investigacion Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Ignacio Melero
- Immunology and Immunotherapy Department, CIMA, Pamplona, Spain. Immunology Department, University Clinic, University of Navarra and Instituto de Investigacion Sanitaria de Navarra (IdISNA), Pamplona, Spain.
| |
Collapse
|
29
|
Tumor-associated fibroblasts predominantly come from local and not circulating precursors. Proc Natl Acad Sci U S A 2016; 113:7551-6. [PMID: 27317748 DOI: 10.1073/pnas.1600363113] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Fibroblasts are common cell types in cancer stroma and lay down collagen required for survival and growth of cancer cells. Although some cancer therapy strategies target tumor fibroblasts, their origin remains controversial. Multiple publications suggest circulating mesenchymal precursors as a source of tumor-associated fibroblasts. However, we show by three independent approaches that tumor fibroblasts derive primarily from local, sessile precursors. First, transplantable tumors developing in a mouse expressing green fluorescent reporter protein (EGFP) under control of the type I collagen (Col-I) promoter (COL-EGFP) had green stroma, whereas we could not find COL-EGFP(+) cells in tumors developing in the parabiotic partner lacking the fluorescent reporter. Lack of incorporation of COL-EGFP(+) cells from the circulation into tumors was confirmed in parabiotic pairs of COL-EGFP mice and transgenic mice developing autochthonous intestinal adenomas. Second, transplantable tumors developing in chimeric mice reconstituted with bone marrow cells from COL-EGFP mice very rarely showed stromal fibroblasts expressing EGFP. Finally, cancer cells injected under full-thickness COL-EGFP skin grafts transplanted in nonreporter mice developed into tumors containing green stromal cells. Using multicolor in vivo confocal microscopy, we found that Col-I-expressing fibroblasts constituted approximately one-third of the stromal mass and formed a continuous sheet wrapping the tumor vessels. In summary, tumors form their fibroblastic stroma predominantly from precursors present in the local tumor microenvironment, whereas the contribution of bone marrow-derived circulating precursors is rare.
Collapse
|
30
|
Sobolik T, Su YJ, Ashby W, Schaffer DK, Wells S, Wikswo JP, Zijlstra A, Richmond A. Development of novel murine mammary imaging windows to examine wound healing effects on leukocyte trafficking in mammary tumors with intravital imaging. INTRAVITAL 2016; 5:e1125562. [PMID: 28243517 DOI: 10.1080/21659087.2015.1125562] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 11/12/2015] [Accepted: 11/18/2015] [Indexed: 01/25/2023]
Abstract
We developed mammary imaging windows (MIWs) to evaluate leukocyte infiltration and cancer cell dissemination in mouse mammary tumors imaged by confocal microscopy. Previous techniques relied on surgical resection of a skin flap to image the tumor microenvironment restricting imaging time to a few hours. Utilization of mammary imaging windows offers extension of intravital imaging of the tumor microenvironment. We have characterized strengths and identified some previously undescribed potential weaknesses of MIW techniques. Through iterative enhancements of a transdermal portal we defined conditions for improved quality and extended confocal imaging time for imaging key cell-cell interactions in the tumor microenvironment.
Collapse
Affiliation(s)
- Tammy Sobolik
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ying-Jun Su
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Will Ashby
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - David K Schaffer
- Department of Physics and Astronomy and the Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University , Nashville, TN, USA
| | - Sam Wells
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine , Nashville, TN, USA
| | - John P Wikswo
- Department of Physics and Astronomy and the Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Andries Zijlstra
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ann Richmond
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
31
|
Leisegang M, Engels B, Schreiber K, Yew PY, Kiyotani K, Idel C, Arina A, Duraiswamy J, Weichselbaum RR, Uckert W, Nakamura Y, Schreiber H. Eradication of Large Solid Tumors by Gene Therapy with a T-Cell Receptor Targeting a Single Cancer-Specific Point Mutation. Clin Cancer Res 2015; 22:2734-43. [PMID: 26667491 DOI: 10.1158/1078-0432.ccr-15-2361] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/07/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Cancers usually contain multiple unique tumor-specific antigens produced by single amino acid substitutions (AAS) and encoded by somatic nonsynonymous single nucleotide substitutions. We determined whether adoptively transferred T cells can reject large, well-established solid tumors when engineered to express a single type of T-cell receptor (TCR) that is specific for a single AAS. EXPERIMENTAL DESIGN By exome and RNA sequencing of an UV-induced tumor, we identified an AAS in p68 (mp68), a co-activator of p53. This AAS seemed to be an ideal tumor-specific neoepitope because it is encoded by a trunk mutation in the primary autochthonous cancer and binds with highest affinity to the MHC. A high-avidity mp68-specific TCR was used to genetically engineer T cells as well as to generate TCR-transgenic mice for adoptive therapy. RESULTS When the neoepitope was expressed at high levels and by all cancer cells, their direct recognition sufficed to destroy intratumor vessels and eradicate large, long-established solid tumors. When the neoepitope was targeted as autochthonous antigen, T cells caused cancer regression followed by escape of antigen-negative variants. Escape could be thwarted by expressing the antigen at increased levels in all cancer cells or by combining T-cell therapy with local irradiation. Therapeutic efficacies of TCR-transduced and TCR-transgenic T cells were similar. CONCLUSIONS Gene therapy with a single TCR targeting a single AAS can eradicate large established cancer, but a uniform expression and/or sufficient levels of the targeted neoepitope or additional therapy are required to overcome tumor escape. Clin Cancer Res; 22(11); 2734-43. ©2015 AACRSee related commentary by Liu, p. 2602.
Collapse
Affiliation(s)
| | - Boris Engels
- Department of Pathology, The University of Chicago, Illinois
| | - Karin Schreiber
- Department of Pathology, The University of Chicago, Illinois
| | - Poh Yin Yew
- Department of Medicine, The University of Chicago, Illinois
| | | | - Christian Idel
- Department of Pathology, The University of Chicago, Illinois
| | - Ainhoa Arina
- Department of Pathology, The University of Chicago, Illinois
| | | | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, The Ludwig Center for Metastasis Research, The University of Chicago, Illinois
| | - Wolfgang Uckert
- Molecular Cell Biology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany. Institute of Biology, Humboldt University Berlin, Berlin, Germany
| | | | - Hans Schreiber
- Institute of Immunology, Charité, Campus Buch, Berlin, Germany. Department of Pathology, The University of Chicago, Illinois
| |
Collapse
|
32
|
Nahas GR, Walker ND, Bryan M, Rameshwar P. A Perspective of Immunotherapy for Breast Cancer: Lessons Learned and Forward Directions for All Cancers. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2015; 9:35-43. [PMID: 26568682 PMCID: PMC4631157 DOI: 10.4137/bcbcr.s29425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/09/2015] [Accepted: 07/12/2015] [Indexed: 12/14/2022]
Abstract
Immunotherapy for cancer has been a focus 50 years ago. At the time, this treatment was developed prior to cloning of the cytokines, no knowledge of regulatory T-cells, and very little information that mesenchymal stem cells (MSCs) (originally colony forming unit-fibroblasts [CFU-F]) could be licensed by the inflammatory microenvironment to suppress an immune response. Given the information available at that time, mononuclear cells from the peripheral blood were activated ex vivo and then replaced in the patients with tumor. The intent was to harness these activated immune cells to target the cancer cells. These studies did not lead to long-term responses because the activated cells when reinfused into the patients were an advantage to the resident MSCs, which can home the tumor and then become suppressive in the presence of the immune cells. The immune suppression caused by MSCs would also expand regulatory T-cells, resulting instead in tumor protection. As time progressed, these different fields converged into a new approach to use immunotherapy for cancer. This article discusses these approaches and also reviews chimeric antigen receptor in the context of future treatments for solid tumors, including breast cancer.
Collapse
Affiliation(s)
| | - Nykia D Walker
- Rutgers New Jersey Medical School, Newark, NJ, USA. ; Rutgers Graduate School of Biomedical Sciences, Newark, NJ, USA
| | | | - Pranela Rameshwar
- Rutgers New Jersey Medical School, Newark, NJ, USA. ; Rutgers Graduate School of Biomedical Sciences, Newark, NJ, USA
| |
Collapse
|
33
|
Leignadier J, Favre S, Luther SA, Luescher IF. CD8 engineered cytotoxic T cells reprogram melanoma tumor environment. Oncoimmunology 2015; 5:e1086861. [PMID: 27141342 DOI: 10.1080/2162402x.2015.1086861] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 08/18/2015] [Accepted: 08/20/2015] [Indexed: 10/23/2022] Open
Abstract
Cytotoxic T lymphocytes (CTL) from CD8β-deficient mice have powerful FasL-mediated cytotoxicity and IFNγ responses, but ablated Ca2+ and NFAT signaling, which can be restored by transduction with CD8β. Upon infection with lymphocytic choriomeningitis virus (LCMV), these cells yielded GP33-specific CTL (CD8βR) that exhibited high FasL/Fas-mediated cytotoxicity, IFNγ CXCL9 and 10 chemokine responses. Transfer of these cells in B16-GP33 tumor bearing mice resulted in (i) massive T cell tumor infiltration, (ii) strong reduction of myeloid-derived suppressor cells (MDSCs), regulatory T cells (Treg) and IL-17-expressing T helper cells, (iii) maturation of tumor-associated antigen-presenting cells and (iv) production of endogenous, B16 melanoma-specific CTL that eradicated the tumor long after the transferred CD8βR CTL perished. Our study demonstrates that the synergistic combination of strong Fas/FasL mediated cytotoxicity, IFNγ and CXCL9 and 10 responses endows adoptively transferred CTL to reprogram the tumor environment and to thus enable the generation of endogenous, tumoricidal immunity.
Collapse
Affiliation(s)
- Julie Leignadier
- Ludwig Center for Cancer Research, University of Lausanne , Epalinges, Switzerland
| | - Stephanie Favre
- Department of Biochemistry, University of Lausanne , Epalinges, Switzerland
| | - Sanjiv A Luther
- Department of Biochemistry, University of Lausanne , Epalinges, Switzerland
| | - Immanuel F Luescher
- Ludwig Center for Cancer Research, University of Lausanne , Epalinges, Switzerland
| |
Collapse
|
34
|
Maeda A, Kulbatski I, DaCosta RS. Emerging Applications for Optically Enabled Intravital Microscopic Imaging in Radiobiology. Mol Imaging 2015. [DOI: 10.2310/7290.2015.00022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Azusa Maeda
- From the Princess Margaret Cancer Centre, University Health Network, MaRS Centre; Techna Institute for Advancement of Technologies for Health; and Department of Medical Biophysics, University of Toronto, MaRS Centre, Toronto, ON
| | - Iris Kulbatski
- From the Princess Margaret Cancer Centre, University Health Network, MaRS Centre; Techna Institute for Advancement of Technologies for Health; and Department of Medical Biophysics, University of Toronto, MaRS Centre, Toronto, ON
| | - Ralph S. DaCosta
- From the Princess Margaret Cancer Centre, University Health Network, MaRS Centre; Techna Institute for Advancement of Technologies for Health; and Department of Medical Biophysics, University of Toronto, MaRS Centre, Toronto, ON
| |
Collapse
|
35
|
Abstract
Radiation has been a staple of cancer therapy since the early 20th century and is implemented in nearly half of current cancer treatment plans. Originally, the genotoxic function of radiation led to a focus on damage and repair pathways associated with deoxyribonucleic acid as important therapeutic targets to augment radiation efficacy. However, in recent decades, the participation of endogenous immune responses in modifying radiation effects have been widely documented and exploited in both preclinical and clinical settings. In particular, preclinical studies have highlighted the capacity of hypofractionated-radiation dose schedules to modify endogenous immune responses raising interest in the use of hypofractionation in the clinical setting to harness the indirect immune effects of radiation and improve clinical responses. We review the current literature regarding the immunomodulatory effects of hypofractionated "ablative" radiation with a primary focus on the preclinical literature but also highlight examples from the clinical literature.
Collapse
Affiliation(s)
- Byron Burnette
- Department of Radiation and Cellular Oncology, The Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL.
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, The Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL
| |
Collapse
|
36
|
|
37
|
Pachynski RK, Scholz A, Monnier J, Butcher EC, Zabel BA. Evaluation of Tumor-infiltrating Leukocyte Subsets in a Subcutaneous Tumor Model. J Vis Exp 2015:52657. [PMID: 25938949 PMCID: PMC4541547 DOI: 10.3791/52657] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Specialized immune cells that infiltrate the tumor microenvironment regulate the growth and survival of neoplasia. Malignant cells must elude or subvert anti-tumor immune responses in order to survive and flourish. Tumors take advantage of a number of different mechanisms of immune "escape," including the recruitment of tolerogenic DC, immunosuppressive regulatory T cells (Tregs), and myeloid-derived suppressor cells (MDSC) that inhibit cytotoxic anti-tumor responses. Conversely, anti-tumor effector immune cells can slow the growth and expansion of malignancies: immunostimulatory dendritic cells, natural killer cells which harbor innate anti-tumor immunity, and cytotoxic T cells all can participate in tumor suppression. The balance between pro- and anti-tumor leukocytes ultimately determines the behavior and fate of transformed cells; a multitude of human clinical studies have borne this out. Thus, detailed analysis of leukocyte subsets within the tumor microenvironment has become increasingly important. Here, we describe a method for analyzing infiltrating leukocyte subsets present in the tumor microenvironment in a mouse tumor model. Mouse B16 melanoma tumor cells were inoculated subcutaneously in C57BL/6 mice. At a specified time, tumors and surrounding skin were resected en bloc and processed into single cell suspensions, which were then stained for multi-color flow cytometry. Using a variety of leukocyte subset markers, we were able to compare the relative percentages of infiltrating leukocyte subsets between control and chemerin-expressing tumors. Investigators may use such a tool to study the immune presence in the tumor microenvironment and when combined with traditional caliper size measurements of tumor growth, will potentially allow them to elucidate the impact of changes in immune composition on tumor growth. Such a technique can be applied to any tumor model in which the tumor and its microenvironment can be resected and processed.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Female
- Flow Cytometry
- Killer Cells, Natural/immunology
- Leukocytes/immunology
- Leukocytes/pathology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Tumor Microenvironment
Collapse
Affiliation(s)
- Russell K Pachynski
- Division of Oncology, Department of Medicine, Washington University School of Medicine;
| | - Alexander Scholz
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine
| | - Justin Monnier
- Palo Alto Institute for Research and Education, Veterans Affairs Palo Alto Health Care System; Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine
| | - Brian A Zabel
- Palo Alto Institute for Research and Education, Veterans Affairs Palo Alto Health Care System
| |
Collapse
|
38
|
Targeting cancer-specific mutations by T cell receptor gene therapy. Curr Opin Immunol 2015; 33:112-9. [PMID: 25728991 DOI: 10.1016/j.coi.2015.02.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 01/22/2015] [Accepted: 02/10/2015] [Indexed: 12/30/2022]
Abstract
The ease of sequencing the cancer genome, identifying all somatic mutations and grafting mutation-specific T cell receptor (TCR) genes into T cells for adoptive transfer allow, for the first time, a truly tumor-specific and effective therapy. Mutation-specific TCR gene therapy might achieve optimal efficacy with least possible toxicity. Recent clinical data confirm the long-standing evidence from experimental cancer models that antigens encoded by the tumor-specific somatic mutations are potentially the best targets for adoptive T cell therapy. Open questions are, how many somatic mutations create suitable epitopes, whether only individual-specific or also recurrent somatic mutations qualify as suitable epitopes and how neoantigen-specific TCRs are most efficiently obtained. Tumor heterogeneity needs to be considered; therefore, it will be important to identify immunogenic driver mutations that occurred early, are essential for cancer cell survival and present in all cancer cells.
Collapse
|
39
|
Arina A, Bronte V. Myeloid-derived suppressor cell impact on endogenous and adoptively transferred T cells. Curr Opin Immunol 2015; 33:120-5. [PMID: 25728992 DOI: 10.1016/j.coi.2015.02.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 01/23/2015] [Accepted: 02/10/2015] [Indexed: 12/14/2022]
Abstract
Novel models of autochthonous tumorigenesis and adoptive T cell therapy (ATT) are providing new clues regarding the pro-tumorigenic and immunosuppressive effects of myeloid-derived suppressor cells (MDSC), and their interaction with T cells. New findings are shifting the perception of the main level at which MDSC act, from direct cell-to-cell suppression to others, such as limiting T cell infiltration. Adoptively transferred, high-avidity T cells recognizing peptides with high-affinity for MHC-I eliminated large tumors. However, low-avidity T cells or low-affinity peptides resulted in failure to eradicate tumors. Manipulation of intratumoral myeloid cells improved the outcome of otherwise unsuccessful ATT. Therefore, therapeutic intervention directed at the tumor stroma might be required when using suboptimal T cells for ATT.
Collapse
Affiliation(s)
- Ainhoa Arina
- Department of Radiation and Cellular Oncology, The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA.
| | - Vincenzo Bronte
- Verona University Hospital, Department of Pathology and Diagnostics, 37134 Verona, Italy.
| |
Collapse
|
40
|
Apetoh L, Smyth MJ, Drake CG, Abastado JP, Apte RN, Ayyoub M, Blay JY, Bonneville M, Butterfield LH, Caignard A, Castelli C, Cavallo F, Celis E, Chen L, Colombo MP, Comin-Anduix B, Coukos G, Dhodapkar MV, Dranoff G, Frazer IH, Fridman WH, Gabrilovich DI, Gilboa E, Gnjatic S, Jäger D, Kalinski P, Kaufman HL, Kiessling R, Kirkwood J, Knuth A, Liblau R, Lotze MT, Lugli E, Marincola F, Melero I, Melief CJ, Mempel TR, Mittendorf EA, Odun K, Overwijk WW, Palucka AK, Parmiani G, Ribas A, Romero P, Schreiber RD, Schuler G, Srivastava PK, Tartour E, Valmori D, van der Burg SH, van der Bruggen P, van den Eynde BJ, Wang E, Zou W, Whiteside TL, Speiser DE, Pardoll DM, Restifo NP, Anderson AC. Consensus nomenclature for CD8 + T cell phenotypes in cancer. Oncoimmunology 2015; 4:e998538. [PMID: 26137416 DOI: 10.1080/2162402x.2014.998538] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 12/10/2014] [Indexed: 10/23/2022] Open
Abstract
Whereas preclinical investigations and clinical studies have established that CD8+ T cells can profoundly affect cancer progression, the underlying mechanisms are still elusive. Challenging the prevalent view that the beneficial effect of CD8+ T cells in cancer is solely attributable to their cytotoxic activity, several reports have indicated that the ability of CD8+ T cells to promote tumor regression is dependent on their cytokine secretion profile and their ability to self-renew. Evidence has also shown that the tumor microenvironment can disarm CD8+ T cell immunity, leading to the emergence of dysfunctional CD8+ T cells. The existence of different types of CD8+ T cells in cancer calls for a more precise definition of the CD8+ T cell immune phenotypes in cancer and the abandonment of the generic terms "pro-tumor" and "antitumor." Based on recent studies investigating the functions of CD8+ T cells in cancer, we here propose some guidelines to precisely define the functional states of CD8+ T cells in cancer.
Collapse
Affiliation(s)
- Lionel Apetoh
- INSERM; UMR 866 , Dijon, France ; Centre Georges François Leclerc , Dijon, France ; Université de Bourgogne , Dijon, France
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute , Herston, Queensland, Australia
| | - Charles G Drake
- Sidney Kimmel Comprehensive Cancer Center; Johns Hopkins University School of Medicine , Baltimore, MD, USA
| | - Jean-Pierre Abastado
- Institut de Recherches Internationales Servier ; 53, rue Carnot , Suresnes, France
| | - Ron N Apte
- The Shraga Segal Department of Microbiology; Immunology and Genetics ; The Faculty of Health Sciences, Ben Gurion University of the Negev , Beer Sheva, Israel
| | - Maha Ayyoub
- INSERM, Unité1102; Equipe Labellisée Ligue Contre le Cancer ; Institut de Cancérologie de l'Ouest , Nantes-Saint Herblain; France
| | - Jean-Yves Blay
- Cancer Research Center of Lyon; INSERM UMR 1052 ; CNRS UMR 5286 , Centre Leon Berard, Lyon, France ; Medical Oncology Department , Lyon, France
| | - Marc Bonneville
- CRCNA, INSERM U892; CNRS UMR 6299 , Nantes, France ; Institut Mérieux , Lyon, France
| | - Lisa H Butterfield
- University of Pittsburgh Cancer Institute, Departments of Medicine, Surgery, and Immunology , Pittsburgh, PA, USA
| | | | - Chiara Castelli
- Unit of Immunotherapy of Human Tumor; Department of Experimental Oncology and Molecular Medicine ; Fondazione IRCCS Istituto Nazionale dei Tumori , Milan, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences; Molecular Biotechnology Center, University of Torino , Italy
| | - Esteban Celis
- Cancer Immunology; Inflammation and Tolerance Program; Georgia Regents University Cancer Center ; Augusta, GA, USA
| | - Lieping Chen
- Department of Immunobiology and Yale Cancer Center; Yale University School of Medicine , New Haven, CT, USA
| | - Mario P Colombo
- Molecular Immunology Unit; Department of Experimental Oncology and Molecular Medicine ; Fondazione IRCCS Istituto Nazionale dei Tumori ; Milan, Italy
| | - Begoña Comin-Anduix
- UCLA School of Medicine ; Jonsson Comprehensive Cancer Center Los Angeles , CA, USA
| | - Georges Coukos
- Ludwig Center for Cancer Research; Department of Oncology; University of Lausanne , Switzerland
| | - Madhav V Dhodapkar
- Department of Immunobiology and Yale Cancer Center; Yale University School of Medicine , New Haven, CT, USA
| | - Glenn Dranoff
- Department of Medical Oncology and Cancer Vaccine Center; Dana-Farber Cancer Institute and Department of Medicine ; Brigham and Women's Hospital and Harvard Medical School , Boston, MA, USA
| | - Ian H Frazer
- The University of Queensland , Queensland, Australia
| | - Wolf-Hervé Fridman
- Cordeliers Research Centre, University of Paris-Descartes , Paris, France
| | | | - Eli Gilboa
- Department of Microbiology & Immunology; Dodson Interdisciplinary Immunotherapy Institute ; Sylvester Comprehensive Cancer Center; Miller School of Medicine ; University of Miami , Miami, FL, USA
| | - Sacha Gnjatic
- Tisch Cancer Institute; Icahn School of Medicine at Mount Sinai , New York, NY, USA
| | - Dirk Jäger
- Department of Medical Oncology; National Center for Tumor Diseases ; Internal Medicine VI; Heidelberg University Hospital , Heidelberg, Germany
| | - Pawel Kalinski
- Department of Surgery; University of Pittsburgh ; Pittsburgh, PA, USA
| | | | - Rolf Kiessling
- Department of Oncology/Pathology; Karolinska Institutet , Stockholm, Sweden
| | - John Kirkwood
- Division of Hematology/Oncology; Department of Medicine ; School of Medicine; University of Pittsburgh , Pittsburgh; PA; USA ; Melanoma and Skin Cancer Program; University of Pittsburgh Cancer Institute , Pittsburgh, PA, USA
| | | | - Roland Liblau
- INSERM-UMR 1043 ; Toulouse, France ; CNRS ; U5282 , Toulouse, France ; Universite de Toulouse; UPS ; Centre de Physiopathologie Toulouse Purpan (CPTP) ; Toulouse, France ; CHU Toulouse Purpan ; Toulouse, France
| | - Michael T Lotze
- Hillman Cancer Center; University of Pittsburgh Schools of Health Sciences , Pittsburgh, PA, USA
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology; Humanitas Clinical and Research Center , Rozzano, Italy
| | | | - Ignacio Melero
- Division of Oncology; Center for Applied Medical Research and Clinica Universidad de Navarra , Pamploma, Spain
| | | | - Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases; Massachusetts General Hospital ; Harvard Medical School , Boston, MA, USA
| | - Elizabeth A Mittendorf
- Deparment of Surgical Oncology; University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Kunle Odun
- Departments of Gynecologic Oncology and Immunology; Roswell Park Cancer Institute , Buffalo, NY, USA
| | - Willem W Overwijk
- Department of Melanoma Medical Oncology; University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | | | - Giorgio Parmiani
- Division of Medical Oncology and Immunotherapy; University Hospital , Siena, Italy
| | - Antoni Ribas
- UCLA School of Medicine ; Jonsson Comprehensive Cancer Center Los Angeles , CA, USA
| | - Pedro Romero
- Ludwig Center for Cancer Research; Department of Oncology; University of Lausanne , Switzerland
| | - Robert D Schreiber
- Department of Pathology and Immunology; Washington University School of Medicine , St. Louis, MO USA
| | - Gerold Schuler
- Department of Dermatology; Universitatsklinikum Erlangen , Erlangen, Germany
| | - Pramod K Srivastava
- Center for Immunotherapy of Cancer and Infectious Diseases; Carole and Ray Neag Comprehensive Cancer Center ; University of Connecticut Health Center , Farmington, CT, USA
| | - Eric Tartour
- Department of Clinical Oncology, INSERM U970; Universite Paris Descartes ; Sorbonne Paris-Cité; Paris ; France; Hôpital Européen Georges Pompidou ; Service d'Immunologie Biologique ; Paris, France
| | - Danila Valmori
- INSERM, Unité1102; Equipe Labellisée Ligue Contre le Cancer ; Institut de Cancérologie de l'Ouest , Nantes-Saint Herblain; France ; Faculty of Medicine, University of Nantes, 44035 Nantes, France
| | | | - Pierre van der Bruggen
- Ludwig Institute for Cancer Research; BrusselsBranch de Duve Institute ; Université Catholique de Louvain , Brussels, Blegium
| | - Benoît J van den Eynde
- Ludwig Institute for Cancer Research; BrusselsBranch de Duve Institute ; Université Catholique de Louvain , Brussels, Blegium
| | - Ena Wang
- Research Branch; Sidra Medical and Research Centre , Doha, Qatar
| | - Weiping Zou
- Department of Surgery; University of Michigan School of Medicine , Ann Arbor , MI, USA
| | - Theresa L Whiteside
- Department of Pathology; Immunology, and Otolaryngology ; University of Pittsburgh Cancer Institute , Pittsburgh, PA, USA
| | - Daniel E Speiser
- Ludwig Center for Cancer Research; Department of Oncology; University of Lausanne , Switzerland
| | - Drew M Pardoll
- Sidney Kimmel Comprehensive Cancer Center; Johns Hopkins University School of Medicine , Baltimore, MD, USA
| | - Nicholas P Restifo
- National Cancer Institute; National Institutes of Health , Bethesda, MD, USA
| | - Ana C Anderson
- Evergrande Center for Immunologic Diseases; Ann Romney Center for Neurologic Diseases ; Brigham and Women's Hospital and Harvard Medical School , Boston, MA USA
| |
Collapse
|
41
|
|
42
|
Abstract
T cells are a crucial component of the immune response to infection and cancer. In addition to coordinating immunity in lymphoid tissue, T cells play a vital role at the disease site, which relies on their efficient and specific trafficking capabilities. The process of T-cell trafficking is highly dynamic, involving a series of distinct processes, which include rolling, adhesion, extravasation, and chemotaxis. Trafficking of T cells to the tumor microenvironment is critical for the success of cancer immunotherapies such as adoptive cellular transfer. Although this approach has achieved some remarkable responses in patients with advanced melanoma and hematologic malignancy, the success against solid cancers has been more moderate. One of the major challenges for adoptive immunotherapy is to be able to effectively target a higher frequency of T cells to the tumor microenvironment, overcoming hurdles associated with immunosuppression and aberrant vasculature. This review summarizes recent advances in our understanding of T-cell migration in solid cancer and immunotherapy based on the adoptive transfer of natural or genetically engineered tumor-specific T cells and discusses new strategies that may enhance the trafficking of these cells, leading to effective eradication of solid cancer and metastases.
Collapse
Affiliation(s)
- Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. Department of Pathology, University of Melbourne, Parkville, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia.
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. Department of Pathology, University of Melbourne, Parkville, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia. Department of Immunology, Monash University, Clayton, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. Department of Pathology, University of Melbourne, Parkville, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia. Department of Immunology, Monash University, Clayton, Australia.
| |
Collapse
|
43
|
Arina A. Rethinking the role of myeloid-derived suppressor cells in adoptive T-cell therapy for cancer. Oncoimmunology 2014; 3:e28464. [PMID: 25050213 PMCID: PMC4063155 DOI: 10.4161/onci.28464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 03/07/2014] [Indexed: 11/19/2022] Open
Abstract
The expansion of cancer-induced myeloid cells is thought to be one of the main obstacles to successful immunotherapy. Nevertheless, in murine tumors undergoing immune-mediated destruction by adoptively transferred T cells, we have recently shown that such cells maintain their immunosuppressive properties. Therefore, adoptive T-cell therapy can, under certain conditions, overcome myeloid cell immunosuppression.
Collapse
Affiliation(s)
- Ainhoa Arina
- Department of Pathology; The University of Chicago; Chicago, IL USA
| |
Collapse
|
44
|
Arina A, Schreiber K, Binder DC, Karrison TG, Liu RB, Schreiber H. Adoptively transferred immune T cells eradicate established tumors despite cancer-induced immune suppression. THE JOURNAL OF IMMUNOLOGY 2013; 192:1286-93. [PMID: 24367029 DOI: 10.4049/jimmunol.1202498] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Myeloid-derived CD11b(+)Gr1(+) suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) are considered a major obstacle for effective adoptive T cell therapy. Myeloid cells suppress naive T cell proliferation ex vivo and can prevent the generation of T cell responses in vivo. We find, however, that adoptively transferred immune T cells eradicate well-established tumors in the presence of MDSCs and TAMs, which are strongly immunosuppressive ex vivo. These MDSCs and TAMs were comparable in numbers and immunosuppressive capacity among different tumor models. Longitudinal microscopy of tumors in vivo revealed that after T cell transfer, tumor vasculature and cancer cells disappeared simultaneously. During T cell-mediated tumor destruction, the tumor stroma contained abundant myeloid cells (mainly TAMs) that retained their suppressive properties. Preimmunized but not naive mice resisted immune suppression caused by an unrelated tumor burden, supporting the idea that in vivo, myeloid immunosuppressive cells can suppress naive but not memory T cell responses.
Collapse
Affiliation(s)
- Ainhoa Arina
- Department of Pathology, University of Chicago, Chicago, IL 60637
| | | | | | | | | | | |
Collapse
|