1
|
Nelissen S, Miller AD. Assessment of SOX10 expression in 437 canine neoplasms of different embryologic origins. Vet Pathol 2024; 61:704-711. [PMID: 38366813 DOI: 10.1177/03009858241231562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
Several members of the SRY-related HMG-box (SOX) protein family are implicated in tumorigenesis, metastasis, and regulation of the tumor microenvironment. SOX10, which is involved in neural crest cell migration and differentiation, has long been recognized a sensitive and specific immunohistochemical (IHC) marker in the diagnosis of melanoma in humans. However, expression of SOX10 in other tumor types has infrequently been evaluated in humans until recently and has not been thoroughly investigated in the dog. Our aim was to characterize the expression of SOX10 in canine neoplasms to objectively assess its value as a diagnostic IHC marker. Immunohistochemistry for SOX10 was performed on 437 archived, formalin-fixed paraffin-embedded tissues from representative canine neoplasms of ectodermal (15 tumor types), mesodermal (13 tumor types), endodermal (8 tumor types), and mixed/unknown (7 tumor types) embryologic origin. Oral and cutaneous tumors of melanocytic origin were used as positive controls. Intense SOX10 immunolabeling was observed in most tumors of ectodermal origin, including consistent expression in mammary carcinomas, and gliomas. Embryonal and hair follicle neoplasms inconsistently exhibited strong nuclear immunolabeling. Oral fibrosarcomas and undifferentiated oral sarcomas both inconsistently exhibited moderate to strong nuclear immunolabeling. Neoplasms of mesodermal and endodermal origin lacked immunolabeling. Salivary carcinomas, representing an unknown/mixed embryologic origin, were strongly labeled. SOX10 expression is not limited to melanomas, but is expressed by canine tumors of diverse tissues and embryologic derivation. Importantly, expression of SOX10 by a subset of oral sarcomas impairs its value as a marker for spindle cell oral melanomas.
Collapse
Affiliation(s)
- Sophie Nelissen
- Cornell University College of Veterinary Medicine, Ithaca, NY, USA
| | - Andrew D Miller
- Cornell University College of Veterinary Medicine, Ithaca, NY, USA
| |
Collapse
|
2
|
Shao Q, Lundgren M, Lynch J, Jiang M, Mir M, Bischof J, Nelson M. Tumor therapeutic response monitored by telemetric temperature sensing, a preclinical study on immunotherapy and chemotherapy. Sci Rep 2023; 13:7727. [PMID: 37173516 PMCID: PMC10182083 DOI: 10.1038/s41598-023-34919-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 05/10/2023] [Indexed: 05/15/2023] Open
Abstract
Temperature in the body and the tumor reflects physiological and pathological conditions. A reliable, contactless, and simplistic measurement system can be used for long-term monitoring of disease progression and therapy response. In this study, miniaturized battery-free wireless chips implanted into growing tumors on small animals were used to capture both basal and tumor temperature dynamics. Three preclinical models: melanoma (B16), breast cancer (4T1), and colon cancer (MC-38), were treated with adoptive T cell transfer, AC-T chemotherapy, and anti-PD-1 immunotherapy respectively. Each model presents a distinctive pattern of temperature history dependent on the tumor characteristic and influenced by the administered therapy. Certain features are associated with positive therapeutic response, for instance the transient reduction of body and tumor temperature following adaptive T cell transfer, the elevation of tumor temperature following chemotherapy, and a steady decline of body temperature following anti-PD-1 therapy. Tracking in vivo thermal activity by cost-effective telemetric sensing has the potential of offering earlier treatment assessment to patients without requiring complex imaging or lab testing. Multi-parametric on-demand monitoring of tumor microenvironment by permanent implants and its integration into health information systems could further advance cancer management and reduce patient burden.
Collapse
Affiliation(s)
- Qi Shao
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, USA.
| | - Mia Lundgren
- Department of Radiology, University of Minnesota, Minneapolis, USA
| | - Justin Lynch
- School of Medicine, University of Minnesota, Minneapolis, USA
| | - Minhan Jiang
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, USA
| | - Mikael Mir
- School of Medicine, University of Minnesota, Minneapolis, USA
| | - John Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, USA
| | - Michael Nelson
- Department of Radiology, University of Minnesota, Minneapolis, USA
| |
Collapse
|
3
|
Immunomodulation of Melanoma by Chemo-Thermo-Immunotherapy Using Conjugates of Melanogenesis Substrate NPrCAP and Magnetite Nanoparticles: A Review. Int J Mol Sci 2022; 23:ijms23126457. [PMID: 35742905 PMCID: PMC9223671 DOI: 10.3390/ijms23126457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 12/30/2022] Open
Abstract
A major advance in drug discovery and targeted therapy directed at cancer cells may be achieved by the exploitation and immunomodulation of their unique biological properties. This review summarizes our efforts to develop novel chemo-thermo-immunotherapy (CTI therapy) by conjugating a melanogenesis substrate, N-propionyl cysteaminylphenol (NPrCAP: amine analog of tyrosine), with magnetite nanoparticles (MNP). In our approach, NPrCAP provides a unique drug delivery system (DDS) because of its selective incorporation into melanoma cells. It also functions as a melanoma-targeted therapeutic drug because of its production of highly reactive free radicals (melanoma-targeted chemotherapy). Moreover, the utilization of MNP is a platform to develop thermo-immunotherapy because of heat shock protein (HSP) expression upon heat generation in MNP by exposure to an alternating magnetic field (AMF). This comprehensive review covers experimental in vivo and in vitro mouse melanoma models and preliminary clinical trials with a limited number of advanced melanoma patients. We also discuss the future directions of CTI therapy.
Collapse
|
4
|
Strobel SB, Machiraju D, Hülsmeyer I, Becker JC, Paschen A, Jäger D, Wels WS, Bachmann M, Hassel JC. Expression of Potential Targets for Cell-Based Therapies on Melanoma Cells. Life (Basel) 2021; 11:life11040269. [PMID: 33805080 PMCID: PMC8064084 DOI: 10.3390/life11040269] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 01/17/2023] Open
Abstract
Tumor antigen-specific redirection of cytotoxic T cells (CTLs) or natural killer (NK) cells including chimeric antigen receptor (CAR-) and T cell receptor (TCR-) cell therapy is currently being evaluated in different tumor entities including melanoma. Expression of melanoma-specific antigen recognized by the respective CAR or TCR directly or presented by HLA molecules is an indispensable prerequisite for this innovative therapy. In this study, we investigated in 168 FFPE tumor specimens of patients with stage I-IV melanoma the protein expression of HER2, TRP2, ABCB5, gp100, p53, and GD2 by immunohistochemistry (IHC). These results were correlated with clinical parameters. Membrane expression of HER2 and GD2 was also investigated in ten melanoma cell lines by flow cytometry for which corresponding tumors were analyzed by IHC. Our results demonstrated that gp100 was the most frequently overexpressed protein (61%), followed by TRP2 (50%), GD2 (38%), p53 (37%), ABCB5 (17%), and HER2 (3%). TRP2 expression was higher in primary tumors compared to metastases (p = 0.005). Accordingly, TRP2 and ABCB5 expression was significantly associated with lower tumor thickness of the primary (p = 0.013 and p = 0.025). There was no association between protein expression levels and survival in advanced melanoma patients. Flow cytometric analysis revealed abundant surface expression of GD2 and HER2 in all melanoma cell lines. The discordant HER2 expression in situ and in vitro suggests a tissue culture associated induction. In summary, our data support the use of gp100 and GD2 as a potential target for developing engineered TCR- or CAR-cell therapies, respectively, against melanoma.
Collapse
Affiliation(s)
- Sophia B. Strobel
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; (S.B.S.); (D.M.)
| | - Devayani Machiraju
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; (S.B.S.); (D.M.)
| | - Ingrid Hülsmeyer
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (I.H.); (J.C.B.); (A.P.); (W.S.W.)
| | - Jürgen C. Becker
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (I.H.); (J.C.B.); (A.P.); (W.S.W.)
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), 45141 Essen, Germany
- Department of Dermatology, University Hospital Essen, 45147 Essen, Germany
| | - Annette Paschen
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (I.H.); (J.C.B.); (A.P.); (W.S.W.)
- Department of Dermatology, University Hospital Essen, 45147 Essen, Germany
| | - Dirk Jäger
- National Center for Tumor Diseases (NCT) Heidelberg, Department of Medical Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany;
- National Center for Tumor Diseases, German Cancer Research Center, Clinical Cooperation Unit Applied Tumor Immunity, 69120 Heidelberg, Germany
| | - Winfried S. Wels
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (I.H.); (J.C.B.); (A.P.); (W.S.W.)
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60590 Frankfurt am Main, Germany
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany;
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), University Hospital ‘Carl Gustav Carus’, TU Dresden, 01307 Dresden, Germany
- Tumor Immunology, University Cancer Center (UCC) ‘Carl Gustav Carus’, TU Dresden, 01307 Dresden, Germany
| | - Jessica C. Hassel
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany; (S.B.S.); (D.M.)
- Correspondence:
| |
Collapse
|
5
|
Tucker CG, Mitchell JS, Martinov T, Burbach BJ, Beura LK, Wilson JC, Dwyer AJ, Singh LM, Mescher MF, Fife BT. Adoptive T Cell Therapy with IL-12-Preconditioned Low-Avidity T Cells Prevents Exhaustion and Results in Enhanced T Cell Activation, Enhanced Tumor Clearance, and Decreased Risk for Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2020; 205:1449-1460. [PMID: 32737148 DOI: 10.4049/jimmunol.2000007] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 06/28/2020] [Indexed: 12/16/2022]
Abstract
Optimal ex vivo expansion protocols of tumor-specific T cells followed by adoptive cell therapy must yield T cells able to home to tumors and effectively kill them. Our previous study demonstrated ex vivo activation in the presence of IL-12-induced optimal CD8+ T cell expansion and melanoma regression; however, adverse side effects, including autoimmunity, can occur. This may be due to transfer of high-avidity self-specific T cells. In this study, we compared mouse low- and high-avidity T cells targeting the tumor Ag tyrosinase-related protein 2 (TRP2). Not surprisingly, high-avidity T cells provide superior tumor control, yet low-avidity T cells can promote tumor regression. The addition of IL-12 during in vitro expansion boosts low-avidity T cell responsiveness, tumor regression, and prevents T cell exhaustion. In this study, we demonstrate that IL-12-primed T cells are resistant to PD-1/PD-L1-mediated suppression and retain effector function. Importantly, IL-12 preconditioning prevented exhaustion as LAG-3, PD-1, and TOX were decreased while simultaneously increasing KLRG1. Using intravital imaging, we also determined that high-avidity T cells have sustained contacts with intratumoral dendritic cells and tumor targets compared with low-avidity T cells. However, with Ag overexpression, this defect is overcome, and low-avidity T cells control tumor growth. Taken together, these data illustrate that low-avidity T cells can be therapeutically beneficial if cocultured with IL-12 cytokine during in vitro expansion and highly effective in vivo if Ag is not limiting. Clinically, low-avidity T cells provide a safer alternative to high-avidity, TCR-engineered T cells, as IL-12-primed, low-avidity T cells cause less autoimmune vitiligo.
Collapse
Affiliation(s)
- Christopher G Tucker
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Jason S Mitchell
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Brandon J Burbach
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Lalit K Beura
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Joseph C Wilson
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Alexander J Dwyer
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Lovejot M Singh
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Matthew F Mescher
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455;
| |
Collapse
|
6
|
Hotblack A, Holler A, Piapi A, Ward S, Stauss HJ, Bennett CL. Tumor-Resident Dendritic Cells and Macrophages Modulate the Accumulation of TCR-Engineered T Cells in Melanoma. Mol Ther 2018; 26:1471-1481. [PMID: 29628306 PMCID: PMC5986719 DOI: 10.1016/j.ymthe.2018.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 12/17/2022] Open
Abstract
Ongoing clinical trials explore T cell receptor (TCR) gene therapy as a treatment option for cancer, but responses in solid tumors are hampered by the immunosuppressive microenvironment. The production of TCR gene-engineered T cells requires full T cell activation in vitro, and it is currently unknown whether in vivo interactions with conventional dendritic cells (cDCs) regulate the accumulation and function of engineered T cells in tumors. Using the B16 melanoma model and the inducible depletion of CD11c+ cells in CD11c.diphtheria toxin receptor (DTR) mice, we analyzed the interaction between tumor-resident cDCs and engineered T cells expressing the melanoma-specific TRP-2 TCR. We found that depletion of CD11c+ cells triggered the recruitment of cross-presenting cDC1 into the tumor and enhanced the accumulation of TCR-engineered T cells. We show that the recruited tumor cDCs present melanoma tumor antigen, leading to enhanced activation of TCR-engineered T cells. In addition, detailed analysis of the tumor myeloid compartment revealed that the depletion of a population of DT-sensitive macrophages can contribute to the accumulation of tumor-infiltrating T cells. Together, these data suggest that the relative frequency of tumor-resident cDCs and macrophages may impact the therapeutic efficacy of TCR gene therapy in solid tumors.
Collapse
Affiliation(s)
- Alastair Hotblack
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK
| | - Angelika Holler
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK
| | - Alice Piapi
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK
| | - Sophie Ward
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK; Cancer Institute, Division of Cancer Studies, University College London, London WC1E 6DD, UK
| | - Hans J Stauss
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK.
| | - Clare L Bennett
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK; Cancer Institute, Division of Cancer Studies, University College London, London WC1E 6DD, UK.
| |
Collapse
|
7
|
Wu S, Zhu W, Peng Y, Wang L, Hong Y, Huang L, Dong D, Xie J, Merchen T, Kruse E, Guo ZS, Bartlett D, Fu N, He Y. The Antitumor Effects of Vaccine-Activated CD8 + T Cells Associate with Weak TCR Signaling and Induction of Stem-Like Memory T Cells. Cancer Immunol Res 2017; 5:908-919. [PMID: 28851693 DOI: 10.1158/2326-6066.cir-17-0016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/02/2017] [Accepted: 08/22/2017] [Indexed: 02/04/2023]
Abstract
To understand why vaccine-activated tumor-specific T cells often fail to generate antitumor effects, we studied two α-fetoprotein-specific CD8+ T cells (Tet499 and Tet212) that had different antitumor effects. We found that Tet499 required high antigen doses for reactivation, but could survive persistent antigen stimulation and maintain their effector functions. In contrast, Tet212 had a low threshold of reactivation, but underwent exhaustion and apoptosis in the presence of persistent antigen. In vivo, Tet499 cells expanded more than Tet212 upon reencountering antigen and generated stronger antitumor effects. The different antigen responsiveness and antitumor effects of Tet212 and Tet499 cells correlated with their activation and differentiation states. Compared with Tet212, the population of Tet499 cells was less activated and contained more stem-like memory T cells (Tscm) that could undergo expansion in vivo The TCR signaling strength on Tet499 was weaker than Tet212, correlating with more severe Tet499 TCR downregulation. Weak TCR signaling may halt T-cell differentiation at the Tscm stage during immune priming and also explains why Tet499 reactivation requires a high antigen dose. Weak TCR signaling of Tet499 cells in the effector stage will also protect them from exhaustion and apoptosis when they reencounter persistent antigen in tumor lesion, which generates antitumor effects. Further investigation of TCR downregulation and manipulation of TCR signaling strength may help design cancer vaccines to elicit a mix of tumor-specific CD8+ T cells, including Tscm, capable of surviving antigen restimulation to generate antitumor effects. Cancer Immunol Res; 5(10); 908-19. ©2017 AACR.
Collapse
Affiliation(s)
- Sha Wu
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia.,Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wei Zhu
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia.,Division of Laboratory Medicine of Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yibing Peng
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Lan Wang
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yuan Hong
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Lei Huang
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Dayong Dong
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Junping Xie
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Todd Merchen
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia.,Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Edward Kruse
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia.,Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zong Sheng Guo
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - David Bartlett
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Ning Fu
- Division of Laboratory Medicine of Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Yukai He
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia. .,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
8
|
Thompson MG, Navarro F, Chitsike L, Ramirez L, Kovacs EJ, Watkins SK. Alcohol exposure differentially effects anti-tumor immunity in females by altering dendritic cell function. Alcohol 2016; 57:1-8. [PMID: 27916138 DOI: 10.1016/j.alcohol.2016.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 10/17/2016] [Accepted: 10/17/2016] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DCs) are a critical component of anti-tumor immunity due to their ability to induce a robust immune response to antigen (Ag). Alcohol was previously shown to reduce DC ability to present foreign Ag and promote pro-inflammatory responses in situations of infection and trauma. However the impact of alcohol exposure on generation of an anti-tumor response, especially in the context of generation of an immune vaccine has not been examined. In the clinic, DC vaccines are typically generated from autologous blood, therefore prior exposure to substances such as alcohol may be a critical factor to consider regarding the effectiveness in generating an immune response. In this study, we demonstrate for the first time that ethanol differentially affects DC and tumor Ag-specific T cell responses depending on sex. Signaling pathways were found to be differentially regulated in DC in females compared to males and these differences were exacerbated by ethanol treatment. DC from female mice treated with ethanol were unable to activate Ag-specific cytotoxic T cells (CTL) as shown by reduced expression of CD44, CD69, and decreased production of granzyme B and IFNγ. Furthermore, although FOXO3, an immune suppressive mediator of DC function, was found to be upregulated in DC from female mice, ethanol related suppression was independent of FOXO3. These findings demonstrate for the first time differential impacts of alcohol on the immune system of females compared to males and may be a critical consideration for determining the effectiveness of an immune based therapy for cancer in patients that consume alcohol.
Collapse
Affiliation(s)
- Matthew G Thompson
- Loyola University Chicago, Department of Surgery, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA
| | - Flor Navarro
- Loyola University Chicago, Department of Surgery, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA
| | - Lennox Chitsike
- Loyola University Chicago, Department of Surgery, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA
| | - Luis Ramirez
- Loyola University Chicago, Department of Surgery, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA
| | - Elizabeth J Kovacs
- Loyola University Chicago, Department of Surgery, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA; University of Colorado Denver, Department of Surgery, Aurora, CO, USA
| | - Stephanie K Watkins
- Loyola University Chicago, Department of Surgery, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA.
| |
Collapse
|
9
|
Abstract
Cellular stress or apoptosis triggers the release of ATP, ADP and other nucleotides into the extracellular space. Extracellular nucleotides function as autocrine and paracrine signalling molecules by activating cell-surface P2 purinergic receptors that elicit pro-inflammatory immune responses. Over time, extracellular nucleotides are metabolized to adenosine, leading to reduced P2 signalling and increased signalling through anti-inflammatory adenosine (P1 purinergic) receptors. Here, we review how local purinergic signalling changes over time during tissue responses to injury or disease, and we discuss the potential of targeting purinergic signalling pathways for the immunotherapeutic treatment of ischaemia, organ transplantation, autoimmunity or cancer.
Collapse
Affiliation(s)
- Caglar Cekic
- Department of Molecular Biology and Genetics, Bilkent University, Ankara 06800, Turkey
| | - Joel Linden
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| |
Collapse
|
10
|
Zhu Z, Cuss SM, Singh V, Gurusamy D, Shoe JL, Leighty R, Bronte V, Hurwitz AA. CD4+ T Cell Help Selectively Enhances High-Avidity Tumor Antigen-Specific CD8+ T Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:3482-9. [PMID: 26320256 DOI: 10.4049/jimmunol.1401571] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 07/31/2015] [Indexed: 01/18/2023]
Abstract
Maintaining antitumor immunity remains a persistent impediment to cancer immunotherapy. We and others have previously reported that high-avidity CD8(+) T cells are more susceptible to tolerance induction in the tumor microenvironment. In the present study, we used a novel model where T cells derived from two independent TCR transgenic mouse lines recognize the same melanoma antigenic epitope but differ in their avidity. We tested whether providing CD4(+) T cell help would improve T cell responsiveness as a function of effector T cell avidity. Interestingly, delivery of CD4(+) T cell help during in vitro priming of CD8(+) T cells improved cytokine secretion and lytic capacity of high-avidity T cells, but not low-avidity T cells. Consistent with this observation, copriming with CD4(+) T cells improved antitumor immunity mediated by higher avidity, melanoma-specific CD8(+) T cells, but not T cells with similar specificity but lower avidity. Enhanced tumor immunity was associated with improved CD8(+) T cell expansion and reduced tolerization, and it was dependent on presentation of both CD4(+) and CD8(+) T cell epitopes by the same dendritic cell population. Our findings demonstrate that CD4(+) T cell help preferentially augments high-avidity CD8(+) T cells and provide important insight for understanding the requirements to elicit and maintain durable tumor immunity.
Collapse
Affiliation(s)
- Ziqiang Zhu
- Tumor Immunity and Tolerance Section, Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702
| | - Steven M Cuss
- Tumor Immunity and Tolerance Section, Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702
| | - Vinod Singh
- Tumor Immunity and Tolerance Section, Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702
| | - Devikala Gurusamy
- Tumor Immunity and Tolerance Section, Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702
| | - Jennifer L Shoe
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick, MD 21072
| | - Robert Leighty
- Data Management Services, National Cancer Institute, Frederick MD, 21702; and
| | - Vincenzo Bronte
- Department of Pathology and Diagnostics, Immunology Section, University of Verona, 37134 Verona, Italy
| | - Arthur A Hurwitz
- Tumor Immunity and Tolerance Section, Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702;
| |
Collapse
|
11
|
Zhang Y, Chen G, Liu Z, Tian S, Zhang J, Carey CD, Murphy KM, Storkus WJ, Falo LD, You Z. Genetic vaccines to potentiate the effective CD103+ dendritic cell-mediated cross-priming of antitumor immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:5937-47. [PMID: 25972487 PMCID: PMC4458448 DOI: 10.4049/jimmunol.1500089] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/16/2015] [Indexed: 12/24/2022]
Abstract
The development of effective cancer vaccines remains an urgent, but as yet unmet, clinical need. This deficiency is in part due to an incomplete understanding of how to best invoke dendritic cells (DC) that are crucial for the induction of tumor-specific CD8(+) T cells capable of mediating durable protective immunity. In this regard, elevated expression of the transcription factor X box-binding protein 1 (XBP1) in DC appears to play a decisive role in promoting the ability of DC to cross-present Ags to CD8(+) T cells in the therapeutic setting. Delivery of DNA vaccines encoding XBP1 and tumor Ag to skin DC resulted in increased IFN-α production by plasmacytoid DC (pDC) from skin/tumor draining lymph nodes and the cross-priming of Ag-specific CD8(+) T cell responses associated with therapeutic benefit. Antitumor protection was dependent on cross-presenting Batf3(+) DC, pDC, and CD8(+) T cells. CD103(+) DC from the skin/tumor draining lymph nodes of the immunized mice appeared responsible for activation of Ag-specific naive CD8(+) T cells, but were dependent on pDC for optimal effectiveness. Similarly, human XBP1 improved the capacity of human blood- and skin-derived DC to activate human T cells. These data support an important intrinsic role for XBP1 in DC for effective cross-priming and orchestration of Batf3(+) DC-pDC interactions, thereby enabling effective vaccine induction of protective antitumor immunity.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Guo Chen
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Zuqiang Liu
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Shenghe Tian
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Jiying Zhang
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Cara D Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110
| | - Walter J Storkus
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213
| | - Zhaoyang You
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213
| |
Collapse
|
12
|
Ngiow SF, von Scheidt B, Möller A, Smyth MJ, Teng MWL. The interaction between murine melanoma and the immune system reveals that prolonged responses predispose for autoimmunity. Oncoimmunology 2014; 2:e23036. [PMID: 23524369 PMCID: PMC3601172 DOI: 10.4161/onci.23036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
An assessment of antitumor immunity versus autoimmunity as provoked by the specific depletion of Foxp3+ Tregs is now possible with the development of Foxp3-diphtheria toxin receptor-like transgenic mouse models. We have used the poorly immunogenic B16F10 melanoma model to characterize a very heterogeneous antitumor effect of the immune response induced by Treg depletion. Depletion and neutralization studies demonstrated the importance of host T cells and interferon γ (IFNγ) in mediating the antitumor response developing in Treg-depleted mice. Such a response correlated with increased proliferation of granzyme B- and IFNγ-producing T cells in the tumor. Furthermore, enhanced antitumor immunity modulated the expression of MHC Class I molecules by B16F10 melanoma cells in Treg-depleted mice. Since Foxp3+ Treg depletion induced a significantly heterogeneous antitumor response, for the first time we were able to assess antitumor immunity and autoimmunity across different groups of responding mice. Strikingly, the duration of the tumor-immune system interaction provoked in individual Treg-depleted mice positively correlated with their propensity to develop vitiligo. A rapid complete tumor rejection was not associated with the development of autoimmunity, however, a proportion of mice that suppressed, but did not effectively clear, B16F10 melanoma did develop vitiligo. The significant implication is that approaches that combine with Treg depletion to rapidly reject tumors may also diminish autoimmune toxicities.
Collapse
Affiliation(s)
- Shin Foong Ngiow
- Cancer Immunology Program; Trescowthick Laboratories; Peter MacCallum Cancer Centre; East Melbourne, Australia ; Department of Pathology; University of Melbourne; Parkville, Australia
| | | | | | | | | |
Collapse
|
13
|
T cell avidity and tumor immunity: problems and solutions. CANCER MICROENVIRONMENT 2013; 7:1-9. [PMID: 24357332 DOI: 10.1007/s12307-013-0143-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/05/2013] [Indexed: 12/16/2022]
Abstract
A potent T cell response is an important component of durable anti-tumor immunity. The quality of the T cell response can, in-part, be measured by the avidity of the T cell for its tumor antigen-expressing target. While convention suggests that raising the avidity of the responding T cells may make for a more potent anti-tumor immune response, the threshold for effective tumor immunity remains unclear, as do some of the adverse effects of an inappropriately high avidity response. In this review, we discuss the relationship between T cell avidity and anti-tumor immunity, considering both experimental model systems as well as human clinical trials.
Collapse
|
14
|
Bot A, Marincola F, Smith KA. Repositioning therapeutic cancer vaccines in the dawning era of potent immune interventions. Expert Rev Vaccines 2013; 12:1219-34. [PMID: 24099049 DOI: 10.1586/14760584.2013.836908] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Based on lessons learned with various immune interventions, this review aims to provide a constructive framework for repositioning therapeutic cancer vaccination. Intensive research throughout the past decade has identified key hurdles interfering with the efficacy of cancer vaccines. The vaccination concept still holds promise if positioned appropriately in minimal residual disease and select early disease stage cancer indications. However, in advanced cancer, it must be integrated with complementary immune interventions to ensure reconstruction of a functional immune repertoire and simultaneous blockade of immune inhibiting mechanisms. Vaccination could render complex and integrative immune interventions simpler, safer and more effective. The near future will witness an explosion of activities in the cancer immunotherapy arena, witnessing a rational repositioning of vaccines rather than their extinction.
Collapse
|
15
|
Chinnasamy D, Tran E, Yu Z, Morgan RA, Restifo NP, Rosenberg SA. Simultaneous targeting of tumor antigens and the tumor vasculature using T lymphocyte transfer synergize to induce regression of established tumors in mice. Cancer Res 2013; 73:3371-80. [PMID: 23633494 PMCID: PMC3686092 DOI: 10.1158/0008-5472.can-12-3913] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Most systemic cancer therapies target tumor cells directly, although there is increasing interest in targeting the tumor stroma that can comprise a substantial portion of the tumor mass. We report here a synergy between two T-cell therapies, one directed against the stromal tumor vasculature and the other directed against antigens expressed on the tumor cell. Simultaneous transfer of genetically engineered syngeneic T cells expressing a chimeric antigen receptor targeting the VEGF receptor-2 (VEGFR2; KDR) that is overexpressed on tumor vasculature and T-cells specific for the tumor antigens gp100 (PMEL), TRP-1 (TYRP1), or TRP-2 (DCT) synergistically eradicated established B16 melanoma tumors in mice and dramatically increased the tumor-free survival of mice compared with treatment with either cell type alone or T cells coexpressing these two targeting molecules. Host lymphodepletion before cell transfer was required to mediate the antitumor effect. The synergistic antitumor response was accompanied by a significant increase in the infiltration and expansion and/or persistence of the adoptively transferred tumor antigen-specific T cells in the tumor microenvironment and thus enhanced their antitumor potency. The data presented here emphasize the possible beneficial effects of combining antiangiogenic with tumor-specific immunotherapeutic approaches for the treatment of patients with cancer.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Disease Models, Animal
- Immunotherapy, Adoptive/methods
- Lymphocyte Activation/immunology
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/therapy
- Receptors, Antigen/genetics
- Receptors, Antigen/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- T-Lymphocytes/immunology
- Vascular Endothelial Growth Factor Receptor-2/immunology
Collapse
Affiliation(s)
- Dhanalakshmi Chinnasamy
- Surgery Branch, National Cancer Institute, Clinical Research Center, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
16
|
Xiao H, Peng Y, Hong Y, Huang L, Guo ZS, Bartlett DL, Fu N, Munn DH, Mellor A, He Y. Local administration of TLR ligands rescues the function of tumor-infiltrating CD8 T cells and enhances the antitumor effect of lentivector immunization. THE JOURNAL OF IMMUNOLOGY 2013; 190:5866-73. [PMID: 23610140 DOI: 10.4049/jimmunol.1203470] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cancer vaccines, to date, have shown limited effect to control the growth of established tumors due largely to effector failure of the antitumor immune responses. Tumor lesion is characterized as chronic indolent inflammation in which the effector function of tumor-infiltrating lymphocytes (TILs) is severely impaired. In this study, we investigated whether the effector function of CD8 TILs could be rescued by converting the chronic inflammation milieu to acute inflammation within tumors. We found that injection of TLR3/9 ligands (polyI:C/CpG) into a tumor during the effector phase of lentivector (lv) immunization effectively rescued the function of lv-activated CD8 TILs and decreased the percentage of T regulatory within the tumor, resulting in a marked improvement in the antitumor efficacy of lv immunization. Mechanistically, rescue of the effector function of CD8 TILs by TLR3/9 ligands is most likely dependent on production, within a tumor, of type-1 IFN that can mature and activate tumor-infiltrating dendritic cells. The effector function of CD8 TILs could not be rescued in mice lacking intact type I IFN signaling. These findings have important implications for tumor immunotherapy, suggesting that type I IFN-mediated activation of tumor-infiltrating dendritic cells within a tumor will most likely restore/enhance the effector function of CD8 TILs and thus improve the antitumor efficacy of current cancer vaccines.
Collapse
Affiliation(s)
- Haiyan Xiao
- Immunology/Immunotherapy Program, Georgia Regents University Cancer Center, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Zhu Z, Singh V, Watkins SK, Bronte V, Shoe JL, Feigenbaum L, Hurwitz AA. High-avidity T cells are preferentially tolerized in the tumor microenvironment. Cancer Res 2012. [PMID: 23204239 DOI: 10.1158/0008-5472.can-12-1123] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
One obstacle in eliciting potent antitumor immune responses is the induction of tolerance to tumor antigens. TCR(lo) mice bearing a TCR transgene specific for the melanoma antigen tyrosinase-related protein-2 (TRP-2, Dct) harbor T cells that maintain tumor antigen responsiveness but lack the ability to control melanoma outgrowth. We used this model to determine whether higher avidity T cells could control tumor growth without becoming tolerized. As a part of the current study, we developed a second TRP-2-specific TCR transgenic mouse line (TCR(hi)) that bears higher avidity T cells and spontaneously developed autoimmune depigmentation. In contrast to TCR(lo) T cells, which were ignorant of tumor-derived antigen, TCR(hi) T cells initially delayed subcutaneous B16 melanoma tumor growth. However, persistence in the tumor microenvironment resulted in reduced IFN-γ production and CD107a (Lamp1) mobilization, hallmarks of T-cell tolerization. IFN-γ expression by TCR(hi) T cells was critical for upregulation of MHC-I on tumor cells and control of tumor growth. Blockade of PD-1 signals prevented T-cell tolerization and restored tumor immunity. Depletion of tumor-associated dendritic cells (TADC) reduced tolerization of TCR(hi) T cells and enhanced their antitumor activity. In addition, TADCs tolerized TCR(hi) T cells but not TCR(lo) T cells in vitro. Our findings show that T-cell avidity is a critical determinant of not only tumor control but also susceptibility to tolerization in the tumor microenvironment. For this reason, care should be exercised when considering T-cell avidity in designing cancer immunotherapeutics.
Collapse
Affiliation(s)
- Ziqiang Zhu
- Tumor Immunity and Tolerance Section, Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Frederick National Laboratory for Cancer Research, NCI, Frederick, MD 21702, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Ito A, Yamaguchi M, Okamoto N, Sanematsu Y, Kawabe Y, Wakamatsu K, Ito S, Honda H, Kobayashi T, Nakayama E, Tamura Y, Okura M, Yamashita T, Jimbow K, Kamihira M. T-cell receptor repertoires of tumor-infiltrating lymphocytes after hyperthermia using functionalized magnetite nanoparticles. Nanomedicine (Lond) 2012; 8:891-902. [PMID: 23066648 DOI: 10.2217/nnm.12.142] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM Accumulating evidence has indicated that hyperthermia using magnetite nanoparticles induces antitumor immunity. This study investigated the diversity of T-cell receptors (TCRs) in tumor-infiltrating lymphocytes after hyperthermia using magnetite nanoparticles. MATERIALS & METHODS Functionalized magnetite nanoparticles, N-propionyl-4-S-cysteaminylphenol (NPrCAP)/magnetite, were synthesized by conjugating the melanogenesis substrate NPrCAP with magnetite nanoparticles. NPrCAP/magnetite nanoparticles were injected into B16 melanomas in C57BL/6 mice, which were subjected to an alternating magnetic field for hyperthermia treatment. RESULTS Enlargement of the tumor-draining lymph nodes was observed after hyperthermia. The TCR repertoire was restricted in tumor-infiltrating lymphocytes, and expansion of Vβ11(+) T cells was preferentially found. DNA sequences of the third complementaritydetermining regions revealed the presence of clonally expanded T cells. CONCLUSION These results indicate that the T-cell response in B16 melanomas after hyperthermia is dominated by T cells directed toward a limited number of epitopes and that epitope-specific T cells frequently use a restricted TCR repertoire.
Collapse
Affiliation(s)
- Akira Ito
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Miska J, Bas E, Devarajan P, Chen Z. Autoimmunity-mediated antitumor immunity: tumor as an immunoprivileged self. Eur J Immunol 2012; 42:2584-96. [PMID: 22777737 DOI: 10.1002/eji.201242590] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 05/22/2012] [Accepted: 06/20/2012] [Indexed: 11/09/2022]
Abstract
The association of autoimmunity with antitumor immunity challenges a paradigm of selective surveillance against tumors. Aided with well-characterized models of robust autoimmunity, we show that self-antigen-specific effector T (Teff) cell clones could eradicate tumor cells. However, a tumor microenvironment reinforced by Treg cells and myeloid-derived suppressor cells (MDSCs) presented a barrier to the autoimmune effectors, more so in tumors than in healthy tissues. This barrier required optimal CTLA4 expression in Teff cells. In a spontaneous model of breast cancer, subtle reductions in CTLA4 expression impeded tumor onset and progression, providing the first direct evidence that CTLA4 inhibits spontaneous tumor development. In an adoptive therapy model of lymphoma, self-antigen-specific Teff cells were potentiated by even a modest reduction of CTLA4. A subtle reduction of CTLA4 did not curtail Treg-cell suppression. Thus, Teff cells had an exquisite sensitivity to physiological levels of CTLA4 variations. However, both Treg and Teff cells were impacted by anti-CTLA4 antibody blockade. Therefore, whether CTLA4 impacts through Treg cells or Teff cells depends on its expression level. Overall, the results suggest that the tumor microenvironment represents an "immunoprivileged self" that could be overcome practically and at least partially by RNAi silencing of CTLA4 in Teff cells.
Collapse
Affiliation(s)
- Jason Miska
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | |
Collapse
|
20
|
Abstract
Tumors create a unique immunosuppressive microenvironment (tumor microenvironment, TME) whereby leukocytes are recruited into the tumor by various chemokines and growth factors. However, once in the TME, these cells lose the ability to promote anti-tumor immunity and begin to support tumor growth and down-regulate anti-tumor immune responses. Studies on tumor-associated leukocytes have mainly focused on cells isolated from tumor-draining lymph nodes or spleen due to the inherent difficulties in obtaining sufficient cell numbers and purity from the primary tumor. While identifying the mechanisms of cell activation and trafficking through the lymphatic system of tumor bearing mice is important and may give insight to the kinetics of immune responses to cancer, in our experience, many leukocytes, including dendritic cells (DCs), in tumor-draining lymph nodes have a different phenotype than those that infiltrate tumors. Furthermore, we have previously demonstrated that adoptively-transferred T cells isolated from the tumor-draining lymph nodes are not tolerized and are capable of responding to secondary stimulation in vitro unlike T cells isolated from the TME, which are tolerized and incapable of proliferation or cytokine production. Interestingly, we have shown that changing the tumor microenvironment, such as providing CD4(+) T helper cells via adoptive transfer, promotes CD8(+) T cells to maintain pro-inflammatory effector functions. The results from each of the previously mentioned studies demonstrate the importance of measuring cellular responses from TME-infiltrating immune cells as opposed to cells that remain in the periphery. To study the function of immune cells which infiltrate tumors using the Miltenyi Biotech isolation system, we have modified and optimized this antibody-based isolation procedure to obtain highly enriched populations of antigen presenting cells and tumor antigen-specific cytotoxic T lymphocytes. The protocol includes a detailed dissection of murine prostate tissue from a spontaneous prostate tumor model (TRansgenic Adenocarcinoma of the Mouse Prostate -TRAMP) and a subcutaneous melanoma (B16) tumor model followed by subsequent purification of various leukocyte populations.
Collapse
Affiliation(s)
- Stephanie K Watkins
- Tumor Immunity and Tolerance Section, Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, National Cancer Institute - Frederick, USA
| | | | | | | |
Collapse
|
21
|
Hong Y, Peng Y, Xiao H, Mi M, Munn D, He Y. Immunoglobulin Fc fragment tagging allows strong activation of endogenous CD4 T cells to reshape the tumor milieu and enhance the antitumor effect of lentivector immunization. THE JOURNAL OF IMMUNOLOGY 2012; 188:4819-27. [PMID: 22504640 DOI: 10.4049/jimmunol.1103512] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A major problem with current cancer vaccines is that the induction of CD8 immune responses is rarely associated with antitumor benefits, mainly owing to multiple immune suppressions in established tumor lesions. In this study, we investigated if and how activation of endogenous CD4 T cells could be achieved to influence the suppressive tumor milieu and antitumor effect. We engineered a lentivector (lv) to express a nominal fusion Ag composed of hepatitis B surface protein and IgG2a Fc fragment (HBS-Fc-lv) to increase the magnitude of CD8 response but, more importantly, to induce effective coactivation of CD4 T cells. We found that, remarkably, immunization with HBS-Fc-lv caused significant regression of established tumors. Immunologic analysis revealed that, compared with HBS-lv without Fc fragment, immunization with HBS-Fc-lv markedly increased the number of functional CD8 and CD4 T cells and the level of Th1/Tc1-like cytokines in the tumor while substantially decreasing the regulatory T cell ratio. The favorable immunologic changes in tumor lesions and the improvement of antitumor effects from HBS-Fc-lv immunization were dependent on the CD4 activation, which was Fc receptor mediated. Adoptive transfer of CD4 T cells from the HBS-Fc-lv-immunized mice could activate endogenous CD8 T cells in an IFN-γ-dependent manner. We conclude that endogenous CD4 T cells can be activated by lv expressing Fc-tagged Ag to provide another layer of help--that is, creating a Th1/Tc1-like proinflammatory milieu within the tumor lesion to boost the effector phase of immune responses in enhancing the antitumor effect.
Collapse
Affiliation(s)
- Yuan Hong
- Immunology/Immunotherapy Program, Cancer Center, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | | | |
Collapse
|
22
|
Träger U, Sierro S, Djordjevic G, Bouzo B, Khandwala S, Meloni A, Mortensen M, Simon AK. The immune response to melanoma is limited by thymic selection of self-antigens. PLoS One 2012; 7:e35005. [PMID: 22506061 PMCID: PMC3323626 DOI: 10.1371/journal.pone.0035005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 03/12/2012] [Indexed: 12/18/2022] Open
Abstract
The expression of melanoma-associated antigens (MAA) being limited to normal melanocytes and melanomas, MAAs are ideal targets for immunotherapy and melanoma vaccines. As MAAs are derived from self, immune responses to these may be limited by thymic tolerance. The extent to which self-tolerance prevents efficient immune responses to MAAs remains unknown. The autoimmune regulator (AIRE) controls the expression of tissue-specific self-antigens in thymic epithelial cells (TECs). The level of antigens expressed in the TECs determines the fate of auto-reactive thymocytes. Deficiency in AIRE leads in both humans (APECED patients) and mice to enlarged autoreactive immune repertoires. Here we show increased IgG levels to melanoma cells in APECED patients correlating with autoimmune skin features. Similarly, the enlarged T cell repertoire in AIRE−/− mice enables them to mount anti-MAA and anti-melanoma responses as shown by increased anti-melanoma antibodies, and enhanced CD4+ and MAA-specific CD8+ T cell responses after melanoma challenge. We show that thymic expression of gp100 is under the control of AIRE, leading to increased gp100-specific CD8+ T cell frequencies in AIRE−/− mice. TRP-2 (tyrosinase-related protein), on the other hand, is absent from TECs and consequently TRP-2 specific CD8+ T cells were found in both AIRE−/− and AIRE+/+ mice. This study emphasizes the importance of investigating thymic expression of self-antigens prior to their inclusion in vaccination and immunotherapy strategies.
Collapse
Affiliation(s)
- Ulrike Träger
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
| | - Sophie Sierro
- Ludwig Institute for Cancer Research, Epalinges, Switzerland
| | - Gordana Djordjevic
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
| | - Basma Bouzo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
| | - Shivani Khandwala
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
| | - Antonella Meloni
- Pediatric Clinic II, Ospedale Microcitemico and Department of Biomedical and Biotechnological Science, University of Cagliari, Cagliari, Italy
| | - Monika Mortensen
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
- Apoptosis Department and Center for Genotoxic Stress Research, Institute of Cancer Biology, Danish Cancer Society, Copenhagen, Denmark
| | - Anna Katharina Simon
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Biomedical Centre, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
23
|
Sevko A, Kremer V, Falk C, Umansky L, Shurin MR, Shurin GV, Umansky V. Application of paclitaxel in low non-cytotoxic doses supports vaccination with melanoma antigens in normal mice. J Immunotoxicol 2012; 9:275-81. [PMID: 22449053 DOI: 10.3109/1547691x.2012.655343] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Chemotherapeutic agents such as paclitaxel applied in ultra-low, non-cytotoxic doses were previously shown to stimulate dendritic cell activity and anti-tumor immune responses upon vaccination in mouse transplantable tumor models. However, the mechanisms of these alterations-termed chemoimmunomodulation or chemomodulation-are still not clear. This study investigated the effect of paclitaxel applied in ultra-low, non-cytotoxic doses on the efficiency of immunization of healthy C57BL/6 mice with the peptide derived from tyrosinase related protein (TRP)-2 as a model melanoma antigen. Using an IFNγ ELISPOT assay, it was found that administration of 1 mg paclitaxel/kg in combination with the peptide vaccination strongly increased the frequencies of TRP-2 specific spleen T-cells as compared to levels due to the vaccination alone. This was associated with a significant decrease in the levels of regulatory T-cells (T(reg)) and immature myeloid cells (known as a counterpart of myeloid derived suppressor cells [MDSC] in healthy mice). Such impairments of potential immunosuppressive cells were found to correlate with a strong increase in the amount of effector CD8+ and CD4+ T-cells in the bone marrow and spleen. Furthermore, in paclitaxel-treated mice, a significant augmentation of natural killer (NK) cell numbers in the bone marrow and their ability to produce IFNγ were observed. In addition, the level of NK-T-cells in the lymph nodes was also increased. It is suggested that paclitaxel applied in ultra-low, non-cytotoxic doses may potentially enhance the efficacy of anti-tumor vaccinations by neutralizing immunosuppressive T(reg) and MDSC populations in tumor-bearing hosts.
Collapse
Affiliation(s)
- Alexandra Sevko
- Skin Cancer Unit, German Cancer Research Center and University Hospital Mannheim, Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
24
|
Vasievich EA, Ramishetti S, Zhang Y, Huang L. Trp2 peptide vaccine adjuvanted with (R)-DOTAP inhibits tumor growth in an advanced melanoma model. Mol Pharm 2011; 9:261-8. [PMID: 22142394 DOI: 10.1021/mp200350n] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Previously we have shown cationic lipid (R)-DOTAP as the immunologically active enantiomer of the DOTAP racemic mixture, initiating complete tumor regression in an exogenous antigen model (murine cervical cancer model). Here, we investigate the use of (R)-DOTAP as an efficacious adjuvant delivering an endogenous antigen in an aggressive murine solid tumor melanoma model. (R)-DOTAP/Trp2 peptide complexes showed decreasing size and charge with increasing peptide concentration, taking a rod shape at highest concentrations. The particles were stable for 2 weeks at 4 °C. A dose of 75 nmol of Trp2 (formulated in (R)-DOTAP) was able to show statistically significant tumor growth delay compared to lower doses of 5 and 25 nmol, which were no different than untreated tumors. (R)-DOTAP/Trp2 (75 nmol) treated mice also showed increased T cell IFN-γ secretion after restimulation with Trp2, as well as CTL activity in vivo. This vaccination group also showed the highest population of functionally active tumor-infiltrating lymphocytes, indicated by IFN-γ secretion after restimulation with Trp2. Thus, (R)-DOTAP has shown the ability to break tolerance as an adjuvant. Its activity to enhance immunogenicity of other tumor associated antigens should be studied further.
Collapse
Affiliation(s)
- Elizabeth A Vasievich
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
25
|
Byrne KT, Turk MJ. New perspectives on the role of vitiligo in immune responses to melanoma. Oncotarget 2011; 2:684-94. [PMID: 21911918 PMCID: PMC3248219 DOI: 10.18632/oncotarget.323] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 09/09/2011] [Indexed: 01/09/2023] Open
Abstract
Melanoma-associated vitiligo is the best-studied example of the linkage between tumor immunity and autoimmunity. Although vitiligo is an independent positive prognostic factor for melanoma patients, the autoimmune destruction of melanocytes was long thought to be merely a side effect of robust anti-tumor immunity. However, new data reveal a key role for vitiligo in supporting T cell responses to melanoma. This research perspective reviews the history of melanoma-associated vitiligo in patients, the experimental studies that form the basis for understanding this relationship, and the unique characteristics of melanoma-specific CD8 T cells found in hosts with vitiligo. We also discuss the implications of our recent findings for the interpretation of patient responses, and the design of next-generation cancer immunotherapies.
Collapse
Affiliation(s)
- Katelyn T Byrne
- Dartmouth Medical School and the Norris Cotton Cancer Center, Lebanon, NH, USA
| | | |
Collapse
|
26
|
Sierro SR, Donda A, Perret R, Guillaume P, Yagita H, Levy F, Romero P. Combination of lentivector immunization and low-dose chemotherapy or PD-1/PD-L1 blocking primes self-reactive T cells and induces anti-tumor immunity. Eur J Immunol 2011; 41:2217-28. [DOI: 10.1002/eji.201041235] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 03/14/2011] [Accepted: 04/27/2011] [Indexed: 11/08/2022]
|
27
|
Watkins SK, Zhu Z, Riboldi E, Shafer-Weaver KA, Stagliano KER, Sklavos MM, Ambs S, Yagita H, Hurwitz AA. FOXO3 programs tumor-associated DCs to become tolerogenic in human and murine prostate cancer. J Clin Invest 2011; 121:1361-72. [PMID: 21436588 DOI: 10.1172/jci44325] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 01/26/2011] [Indexed: 01/05/2023] Open
Abstract
The limited success of cancer immunotherapy is often attributed to the loss of antigen-specific T cell function in situ. However, the mechanism for this loss of function is unknown. In this study, we describe a population of tumor-associated DCs (TADCs) in both human and mouse prostate cancer that tolerizes and induces suppressive activity in tumor-specific T cells. In tumors from human prostate cancer patients and transgenic adenocarcinoma of the mouse prostate (TRAMP) mice, TADCs expressed elevated levels of FOXO3 and Foxo3, respectively, which correlated with expression of suppressive genes that negatively regulate T cell function. Silencing FOXO3 and Foxo3 with siRNAs abrogated the ability of human and mouse TADCs, respectively, to tolerize and induce suppressive activity by T cells. Silencing Foxo3 in mouse TADCs was also associated with diminished expression of tolerogenic mediators, such as indoleamine-2,3-dioxygenase, arginase, and TGF-β, and upregulated expression of costimulatory molecules and proinflammatory cytokines. Importantly, transfer of tumor-specific CD4+ Th cells into TRAMP mice abrogated TADC tolerogenicity, which was associated with reduced Foxo3 expression. These findings demonstrate that FOXO3 may play a critical role in mediating TADC-induced immune suppression. Moreover, our results identify what we believe to be a novel target for preventing CTL tolerance and enhancing immune responses to cancer by modulating the immunosuppressive activity of TADCs found in the tumor microenvironment.
Collapse
Affiliation(s)
- Stephanie K Watkins
- Tumor Immunity and Tolerance Section, Laboratory of Molecular Immunoregulation, National Cancer Institute-Frederick, Frederick, Maryland 21702, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Shafer-Weaver KA, Anderson MJ, Stagliano K, Malyguine A, Greenberg NM, Hurwitz AA. Cutting Edge: Tumor-specific CD8+ T cells infiltrating prostatic tumors are induced to become suppressor cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:4848-52. [PMID: 19801511 DOI: 10.4049/jimmunol.0900848] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We previously reported that naive, tumor-specific CD8(+) (TcR-I) T cells transferred into prostate tumor-bearing mice traffic to the prostate where they become tolerized. We now report that TcR-I cells suppress the proliferation of naive T cells. This suppression is mediated at least in part by secreted factors, and the suppressive activity can be blocked by Abs directed against TGF-beta. We further report that TcR-I cells must infiltrate the prostate to acquire suppressive activity. Delivery of tumor-specific CD4(+) T cells prevents the conversion of TcR-I cells into suppressor cells. Taken together, our findings may have critical implications for sustaining T cell responsiveness during immunotherapy, as the development of suppressor cells in the tumor microenvironment may eliminate the potency of T cells primed in the periphery or delivered during adoptive immunotherapy.
Collapse
Affiliation(s)
- Kimberly A Shafer-Weaver
- Tumor Immunity and Tolerance Section, Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, National Cancer Institute-Frederick, Frederick, MD 21702, USA
| | | | | | | | | | | |
Collapse
|