1
|
Jiménez-Cortegana C, Gutiérrez-García C, Sánchez-Jiménez F, Vilariño-García T, Flores-Campos R, Pérez-Pérez A, Garnacho C, Sánchez-León ML, García-Domínguez DJ, Hontecillas-Prieto L, Palazón-Carrión N, De La Cruz-Merino L, Sánchez-Margalet V. Impact of obesity‑associated myeloid‑derived suppressor cells on cancer risk and progression (Review). Int J Oncol 2024; 65:79. [PMID: 38940351 PMCID: PMC11251741 DOI: 10.3892/ijo.2024.5667] [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] [Received: 04/09/2024] [Accepted: 06/12/2024] [Indexed: 06/29/2024] Open
Abstract
Obesity is a chronic disease caused by the accumulation of excessive adipose tissue. This disorder is characterized by chronic low‑grade inflammation, which promotes the release of proinflammatory mediators, including cytokines, chemokines and leptin. Simultaneously, chronic inflammation can predispose to cancer development, progression and metastasis. Proinflammatory molecules are involved in the recruitment of specific cell populations in the tumor microenvironment. These cell populations include myeloid‑derived suppressor cells (MDSCs), a heterogeneous, immature myeloid population with immunosuppressive abilities. Obesity‑associated MDSCs have been linked with tumor dissemination, progression and poor clinical outcomes. A comprehensive literature review was conducted to assess the impact of obesity‑associated MDSCs on cancer in both preclinical models and oncological patients with obesity. A secondary objective was to examine the key role that leptin, the most important proinflammatory mediator released by adipocytes, plays in MDSC‑driven immunosuppression Finally, an overview is provided of the different therapeutic approaches available to target MDSCs in the context of obesity‑related cancer.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Cristian Gutiérrez-García
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Flora Sánchez-Jiménez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Teresa Vilariño-García
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Rocio Flores-Campos
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Antonio Pérez-Pérez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Carmen Garnacho
- Department of Normal and Pathological Histology and Cytology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Maria L. Sánchez-León
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Daniel J. García-Domínguez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Natalia Palazón-Carrión
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Luis De La Cruz-Merino
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, Seville 41013, Spain
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, Seville 41013, Spain
| |
Collapse
|
2
|
Lian T, Zhang W, Su H, Yu Q, Zhang H, Zou Q, Chen H, Xiong W, Zhang N, Wang K, Zhao L, Fu ZF, Cui M. TLR9 promotes monocytic myeloid-derived suppressor cell induction during JEV infection. Microbes Infect 2024; 26:105336. [PMID: 38724001 DOI: 10.1016/j.micinf.2024.105336] [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] [Received: 07/26/2023] [Revised: 03/18/2024] [Accepted: 04/08/2024] [Indexed: 05/16/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of heterologous populations of immature bone marrow cells consisting of progenitor cells of macrophages, dendritic cells and granulocytes. Recent studies have revealed that the accumulation of MDSCs in the mouse spleen plays a pivotal role in suppressing the immune response following JEV infection. However, the mechanisms by which JEV induces MDSCs are poorly understood. Here, it was found that JEV infection induces mitochondrial damage and the release of mitochondrial DNA (mtDNA), which further leads to the activation of TLR9. TLR9 deficiency decreases the M-MDSCs population and their suppressive function both in vitro and in vivo. Moreover, the increase of MHCⅡ expression on antigen-presenting cells and CD28 expression on T cells in TLR9-/- mice was positively correlated with M-MDSCs reduction. Accordingly, the survival rate of TLR9-/- mice dramatically increased after JEV infection. These findings reveal the connections of mitochondrial damage and TLR9 activation to the induction of M-MDSCs during JEV infection.
Collapse
Affiliation(s)
- Tingting Lian
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Weijia Zhang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Haoran Su
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Qing Yu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Hongxin Zhang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Qingcui Zou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Haowei Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Wenjing Xiong
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Nan Zhang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Ke Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Ling Zhao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Zhen F Fu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Min Cui
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China; International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China.
| |
Collapse
|
3
|
Bausart M, Rodella G, Dumont M, Ucakar B, Vanvarenberg K, Malfanti A, Préat V. Combination of local immunogenic cell death-inducing chemotherapy and DNA vaccine increases the survival of glioblastoma-bearing mice. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 50:102681. [PMID: 37105343 DOI: 10.1016/j.nano.2023.102681] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/22/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023]
Abstract
Immunotherapy efficacy as monotherapy is negligible for glioblastoma (GBM). We hypothesized that combining therapeutic vaccination using a plasmid encoding an epitope derived from GBM-associated antigen (pTOP) with local delivery of immunogenic chemotherapy using mitoxantrone-loaded PEGylated PLGA-based nanoparticles (NP-MTX) would improve the survival of GBM-bearing mice by stimulating an antitumor immune response. We first proved that MTX retained its ability to induce cytotoxicity and immunogenic cell death of GBM cells after encapsulation. Intratumoral delivery of MTX or NP-MTX increased the frequency of IFN-γ-secreting CD8 T cells. NP-MTX mixed with free MTX in combination with pTOP DNA vaccine increased the median survival of GL261-bearing mice and increased M1-like macrophages in the brain. The addition of CpG to this combination abolished the survival benefit but led to increased M1 to M2 macrophage ratio and IFN-γ-secreting CD4 T cell frequency. These results highlight the benefits of combination strategies to potentiate immunotherapy and improve GBM outcome.
Collapse
Affiliation(s)
- Mathilde Bausart
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Giulia Rodella
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Mathilde Dumont
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Bernard Ucakar
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Kevin Vanvarenberg
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium.
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium.
| |
Collapse
|
4
|
Local and systemic inflammation triggers different outcomes of tumor growth related to infiltration of anti-tumor or pro-tumor macrophages. Chin Med J (Engl) 2022; 135:1821-1828. [PMID: 35903953 PMCID: PMC9521782 DOI: 10.1097/cm9.0000000000001775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Previous evidence suggests inflammation may be a double-edged sword with cancer-promoting and cancer suppressing function. In this study, we explore the impact of local and systemic inflammation on cancer growth. METHODS Female BALB/C mice were subcutaneously implanted with foreign body (plastic plates) to build up a local inflammation and intraperitoneally injected with PolyIC or lipopolysaccharides (LPS) to build up a systemic inflammation, followed by subcutaneous injection of 5 × 10 5 colon cancer cells. Immunohistochemistry and enzyme linked immunosorbent assay were utilized to detect the Ki67 and interleukin (IL) 6, IL-1β, and monocyte chemoattractant protein-1 expression in the tumor tissues and serum, respectively. The distributions of immune cells and expression of toll-like receptors (TLRs) were evaluated by flow cytometry (FCM) and quantitative real time-polymerase chain reaction. RESULTS The results showed that local inflammation induced by foreign body implantation suppressed tumor growth with decreased tumor weight ( P = 0.001), volume ( P = 0.004) and Ki67 index ( P < 0.001). Compared with the control group, myeloid-derived suppressive cells sharply decreased ( P = 0.040), while CD4 + T cells slightly increased in the tumor tissues of the group of foreign body-induced local inflammation ( P = 0.035). Moreover, the number of M1 macrophages ( P = 0.040) and expression of TLRs, especially TLR3 ( P < 0.001) and TLR4 ( P < 0.001), were significantly up-regulated in the foreign body group. Contrarily, tumor growth was significantly promoted in LPS or PolyIC-induced systemic inflammation ( P = 0.009 and 0.006). FCM results showed M1 type macrophages ( P = 0.017 and 0.006) and CD8 + T cells ( P = 0.031 and 0.023) were decreased, while M2 type macrophages ( P = 0.002 and 0.007) were significantly increased in tumor microenvironment of LPS or PolyIC-induced systemic inflammation group. In addition, the decreased expression of TLRs was detected in LPS or PolyIC group. CONCLUSIONS The foreign body-induced local inflammation inhibited tumor growth, while LPS or PolyIC- induced systemic inflammation promoted tumor growth. The results suggested that the different outcomes of tumor growth might be attributed to the infiltration of anti-tumor or pro-tumor immune cells, especially M1 or M2 type macrophages into tumor microenvironment.
Collapse
|
5
|
Xu D, Jiang S, He Y, Jin X, Zhao G, Wang B. Development of a therapeutic vaccine targeting Merkel cell polyomavirus capsid protein VP1 against Merkel cell carcinoma. NPJ Vaccines 2021; 6:119. [PMID: 34611173 PMCID: PMC8492671 DOI: 10.1038/s41541-021-00382-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Merkel cell carcinoma (MCC) is a rare but aggressive skin cancer with a high mortality rate, while Merkel cell polyomavirus (MCV) has been pointed as the causative agent of MCC. A better prognosis of MCC associated with a high level of antibodies against the capsid protein VP1 suggests that anti-VP1 immune response might be essential against MCC growth. In the current study, we developed a VP1-target vaccine formulated with CRA. Using a tumorigenic CMS5-VP1 tumor model, the vaccine-induced a potent antitumor efficacy in a dose-dependent manner was evidently demonstrated and mainly mediated by both VP1-specific CD4+ and CD8+ T-cell responses against the growth of CMS5-VP1 tumors in vaccinated BALB/c mice since the depletion of CD4+ and CD8+ T cells reverse the antitumor effects. Thus, immunotherapy with this vaccine represents a novel approach for the clinical treatment of aggressive MCV-related MCC in humans.
Collapse
Affiliation(s)
- Dan Xu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China.,Shanghai Zerun Biotech Co., LTD, Shanghai, China
| | - Sheng Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yue He
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China.,Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou, China
| | - Xiang Jin
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | | | - Bin Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Farag KI, Makkouk A, Norian LA. Re-Evaluating the Effects of Obesity on Cancer Immunotherapy Outcomes in Renal Cancer: What Do We Really Know? Front Immunol 2021; 12:668494. [PMID: 34421889 PMCID: PMC8374888 DOI: 10.3389/fimmu.2021.668494] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Obesity has reached global epidemic proportions and its effects on interactions between the immune system and malignancies, particularly as related to cancer immunotherapy outcomes, have come under increasing scrutiny. Although the vast majority of pre-clinical murine studies suggest that host obesity should have detrimental effects on anti-tumor immunity and cancer immunotherapy outcomes, the opposite has been found in multiple retrospective human studies. As a result, acceptance of the "obesity paradox" paradigm, wherein obesity increases cancer risk but then improves patient outcomes, has become widespread. However, results to the contrary do exist and the biological mechanisms that promote beneficial obesity-associated outcomes remain unclear. Here, we highlight discrepancies in the literature regarding the obesity paradox for cancer immunotherapy outcomes, with a particular focus on renal cancer. We also discuss multiple factors that may impact research findings and warrant renewed research attention in future studies. We propose that specific cancer patient populations may be affected in fundamentally different ways by host obesity, leading to divergent effects on anti-tumor immunity and/or immunotherapy outcomes. Continued, thoughtful analysis of this critical issue is therefore needed to permit a more nuanced understanding of the complex effects of host obesity on cancer immunotherapy outcomes in patients with renal cancer or other malignancies.
Collapse
Affiliation(s)
- Kristine I Farag
- Science and Technology Honors Program, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Amani Makkouk
- Department of Pharmacology, Adicet Bio, Menlo Park, CA, United States
| | - Lyse A Norian
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, United States.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States.,Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
7
|
Diet-Induced Obesity Impairs Outcomes and Induces Multi-Factorial Deficiencies in Effector T Cell Responses Following Anti-CTLA-4 Combinatorial Immunotherapy in Renal Tumor-Bearing Mice. Cancers (Basel) 2021; 13:cancers13102295. [PMID: 34064933 PMCID: PMC8151089 DOI: 10.3390/cancers13102295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 01/04/2023] Open
Abstract
Simple Summary Immunotherapy use has become standard for many patients with advanced kidney cancer; unfortunately, <50% of patients experience durable responses. Mounting evidence suggests that modifiable factors, such as diet and obesity, impact immunotherapy outcomes. Obesity, a major U.S. health epidemic, blunts anti-tumor immunity and promotes tumor growth in multiple preclinical models. However, the full biological impact of obesity on the T cell responses needed to achieve positive immunotherapy outcomes remains unclear. Here, we studied the effects of obesity on T cell responses following combinatorial immunotherapy in a mouse model of kidney cancer. We found that obesity is associated with blunted effector T cell responses, resulting in diminished immunotherapy outcomes. This therapy produces sustained T cell responses and robust tumor control in obese-resistant mice fed the same high-fat diet. Finding ways to amplify T cell responses within renal tumors from hosts with obesity will be critical for achieving optimal immunotherapy outcomes. Abstract Associations between modifiable factors and the efficacy of cancer immunotherapies remain uncertain. We found previously that diet-induced obesity (DIO) reduces the efficacy of an immunotherapy consisting of adenovirus-encoded TRAIL plus CpG oligonucleotide (AdT/CpG) in mice with renal tumors. To eliminate confounding effects of diet and determine whether outcomes could be improved in DIO mice, we evaluated AdT/CpG combined with anti-CTLA-4 in diet-matched, obese-resistant (OB-RES) versus DIO tumor-bearing mice. Therapy-treated OB-RES mice displayed effective renal tumor control and sustained CD4+ and CD8+ T cell responses. In contrast, therapy-treated DIO mice exhibited progressive tumor outgrowth and blunted T cell responses, characterized by reduced intratumoral frequencies of IFNγ+ CD4+ and CD8+ T cells. Weak effector T cell responses in therapy-treated DIO mice were accompanied by low intratumoral concentrations of the T cell chemoattractant CCL5, heightened concentrations of pro-tumorigenic GM-CSF, and impaired proliferative capacity of CD44+CD8+ T cells in tumor-draining lymph nodes. Our findings demonstrate that in lean mice with renal tumors, combining in situ T cell priming upstream of anti-CTLA-4 enhances outcomes versus anti-CTLA-4 alone. However, host obesity is associated with heightened immunotherapy resistance, characterized by multi-factorial deficiencies in effector CD4+ and CD8+ T cell responses that extend beyond the tumor microenvironment.
Collapse
|
8
|
Park JS, Lee ME, Kim SH, Jang WS, Ham WS. Development of a highly pulmonary metastatic orthotopic renal cell carcinoma murine model. Biol Open 2021; 10:256557. [PMID: 33913471 PMCID: PMC8084570 DOI: 10.1242/bio.058566] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 03/25/2021] [Indexed: 01/01/2023] Open
Abstract
The incidence of renal cell carcinoma (RCC) is high, and its outcomes remain poor. Mortality is attributable largely to metastatic disease and a dearth of effective therapeutic interventions. The lungs are the most common metastatic site. To elucidate the biological mechanisms underlying pulmonary metastasis and identify superior therapeutic strategies, we developed a novel and clinically relevant murine RCC model exhibiting enhanced pulmonary metastasis. Mice underwent intrarenal implantation using luciferase-expressing Renca, a murine renal adenocarcinoma cell line. Primary renal tumor progression and development of metastatic lung lesions were monitored in live mice using bioluminescent imaging, followed by post-mortem organ assessment. Cells were isolated from pulmonary metastases for reimplantation, followed by repeat monitoring and assessment. This process was repeated once more for a total of two in vivo passages to select for pulmonary metastatic Renca cell subpopulations. However, a single round of in vivo selection was sufficient to produce a near-maximally metastatic subpopulation. Relative to Renca cell-implanted mice, subpopulation-implanted mice exhibited shorter implantation-metastasis intervals (5 days), shorter implantation-moribundity intervals (sacrificed at 18.6±2.9 versus 22.3±1.1 days), a higher number of metastatic lung lesions at 23 days (183.9±39.0 versus 172.6±38.2) and poorer survival. Implantation of cells derived from the second round of in vivo selection produced no further significant differences in the above metrics. This model consistently and efficiently recapitulates RCC pulmonary metastasis while allowing in vivo monitoring of tumor progression, thereby facilitating elucidation of the molecular mechanisms underlying pulmonary metastasis and evaluation of therapeutic modalities.
Collapse
Affiliation(s)
- Jee Soo Park
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Myung Eun Lee
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Seung Hwan Kim
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Won Sik Jang
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Won Sik Ham
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
9
|
Zhou H, Jiang M, Yuan H, Ni W, Tai G. Dual roles of myeloid-derived suppressor cells induced by Toll-like receptor signaling in cancer. Oncol Lett 2020; 21:149. [PMID: 33552267 PMCID: PMC7798029 DOI: 10.3892/ol.2020.12410] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 12/03/2020] [Indexed: 12/16/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are one of the major components of the tumor microenvironment (TME), and are the main mediators of tumor-induced immunosuppression. Recent studies have reported that the survival, differentiation and immunosuppressive activity of MDSCs are affected by the Toll-like receptor (TLR) signaling pathway. However, the regulatory effect of TLR signaling on MDSCs remains controversial. TLR-induced MDSC can acquire different immunosuppressive activities to influence the immune response that can be either beneficial or detrimental to cancer immunotherapy. The present review summarizes the effects of TLR signals on the number, phenotype and inhibitory activity of MDSCs, and their role in cancer immunotherapy, which cannot be ignored if effective cancer immunotherapies are to be developed for the immunosuppression of the TME.
Collapse
Affiliation(s)
- Hongyue Zhou
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Mengyu Jiang
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hongyan Yuan
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Weihua Ni
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guixiang Tai
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
10
|
Boi SK, Orlandella RM, Gibson JT, Turbitt WJ, Wald G, Thomas L, Buchta Rosean C, Norris KE, Bing M, Bertrand L, Gross BP, Makkouk A, Starenki D, Farag KI, Sorge RE, Brown JA, Gordetsky J, Yasin H, Garje R, Nandagopal L, Weiner GJ, Lubaroff DM, Arend RC, Li P, Zakharia Y, Yang E, Salem AK, Nepple K, Marquez-Lago TT, Norian LA. Obesity diminishes response to PD-1-based immunotherapies in renal cancer. J Immunother Cancer 2020; 8:e000725. [PMID: 33427691 PMCID: PMC7757487 DOI: 10.1136/jitc-2020-000725] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Obesity is a major risk factor for renal cancer, yet our understanding of its effects on antitumor immunity and immunotherapy outcomes remains incomplete. Deciphering these associations is critical, given the growing clinical use of immune checkpoint inhibitors for metastatic disease and mounting evidence for an obesity paradox in the context of cancer immunotherapies, wherein obese patients with cancer have improved outcomes. METHODS We investigated associations between host obesity and anti-programmed cell death (PD-1)-based outcomes in both renal cell carcinoma (RCC) subjects and orthotopic murine renal tumors. Overall survival (OS) and progression-free survival (PFS) were determined for advanced RCC subjects receiving standard of care anti-PD-1 who had ≥6 months of follow-up from treatment initiation (n=73). Renal tumor tissues were collected from treatment-naive subjects categorized as obese (body mass index, 'BMI' ≥30 kg/m2) or non-obese (BMI <30 kg/m2) undergoing partial or full nephrectomy (n=19) then used to evaluate the frequency and phenotype of intratumoral CD8+ T cells, including PD-1 status, by flow cytometry. In mice, antitumor immunity and excised renal tumor weights were evaluated ±administration of a combinatorial anti-PD-1 therapy. For a subset of murine renal tumors, immunophenotyping was performed by flow cytometry and immunogenetic profiles were evaluated via nanoString. RESULTS With obesity, RCC patients receiving anti-PD-1 administration exhibited shorter PFS (p=0.0448) and OS (p=0.0288). Treatment-naive renal cancer subjects had decreased frequencies of tumor-infiltrating PD-1highCD8+ T cells, a finding recapitulated in our murine model. Following anti-PD-1-based immunotherapy, both lean and obese mice possessed distinct populations of treatment responders versus non-responders; however, obesity reduced the frequency of treatment responders (73% lean vs 44% obese). Tumors from lean and obese treatment responders displayed similar immunogenetic profiles, robust infiltration by PD-1int interferon (IFN)γ+CD8+ T cells and reduced myeloid-derived suppressor cells (MDSC), yielding favorable CD44+CD8+ T cell to MDSC ratios. Neutralizing interleukin (IL)-1β in obese mice improved treatment response rates to 58% and reduced MDSC accumulation in tumors. CONCLUSIONS We find that obesity is associated with diminished efficacy of anti-PD-1-based therapies in renal cancer, due in part to increased inflammatory IL-1β levels, highlighting the need for continued study of this critical issue.
Collapse
Affiliation(s)
- Shannon K Boi
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rachael M Orlandella
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Justin Tyler Gibson
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - William James Turbitt
- Department of Nutrition Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Gal Wald
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Lewis Thomas
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Claire Buchta Rosean
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Katlyn E Norris
- Honors Undergraduate Research Program, School of Health Professions, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Megan Bing
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Laura Bertrand
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Brett P Gross
- Interdisciplinary Program in Immunology, The University of Iowa, Iowa City, Iowa, USA
| | - Amani Makkouk
- Interdisciplinary Program in Immunology, The University of Iowa, Iowa City, Iowa, USA
| | - Dmytro Starenki
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Kristine I Farag
- Science and Technology Honors Program, College of Arts and Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Robert E Sorge
- Department of Psychology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James A Brown
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
| | - Jennifer Gordetsky
- Departments of Pathology and Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hesham Yasin
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
- Department of Internal Medicine, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Rohan Garje
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
- Genitourinary Oncology Program, Division of Hematology, Oncology and Blood and Marrow Transplantation, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - Lakshminarayanan Nandagopal
- Division of Hematology and Oncology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
- The University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama, USA
| | - George J Weiner
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
- Department of Internal Medicine, Division of General Medicine, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
| | - David M Lubaroff
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
| | - Rebecca C Arend
- The University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama, USA
- Department of Obstetrics and Gynecology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Peng Li
- Department of Biostatistics, The University of Alabama at Birmingham School of Nursing, Birmingham, Alabama, USA
| | - Yousef Zakharia
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
- Genitourinary Oncology Program, Division of Hematology, Oncology and Blood and Marrow Transplantation, The University of Iowa, Iowa City, Iowa, USA
| | - Eddy Yang
- The University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama, USA
- Department of Radiation Oncology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Aliasger K Salem
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
- Division of Pharmaceutics and Translational Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA
| | - Kenneth Nepple
- Department of Urology, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, Iowa, USA
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
| | - Tatiana T Marquez-Lago
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lyse A Norian
- Department of Nutrition Sciences, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- The University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama, USA
| |
Collapse
|
11
|
The Antidiabetic Agent Acarbose Improves Anti-PD-1 and Rapamycin Efficacy in Preclinical Renal Cancer. Cancers (Basel) 2020; 12:cancers12102872. [PMID: 33036247 PMCID: PMC7601245 DOI: 10.3390/cancers12102872] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Although immune-stimulatory and targeted therapies benefit many patients with metastatic kidney cancer, a sizeable proportion of patients fail to respond. Recent studies in mice demonstrate that nutrient-limiting dietary interventions can improve responses to chemotherapy. However, these studies did not investigate effects on metastasis, and the impact of these interventions on the response to immunotherapy or targeted therapies in kidney cancer is unknown. We therefore studied the effects of a glucose-limiting drug called acarbose, which is used to treat type 2 diabetes, in a spontaneously-metastasizing mouse model of kidney cancer. We found that acarbose slowed kidney cancer growth and promoted protective immune responses. In combination with either an immunotherapy or a targeted therapy used clinically to treat kidney cancer, acarbose led to improved treatment outcomes and reduced lung metastases. Our findings contribute to the emerging idea of using nutrition-based interventions to enhance responses to cancer treatments. Abstract Although immune checkpoint inhibitors and targeted therapeutics have changed the landscape of treatment for renal cell carcinoma (RCC), most patients do not experience significant clinical benefits. Emerging preclinical studies report that nutrition-based interventions and glucose-regulating agents can improve therapeutic efficacy. However, the impact of such agents on therapeutic efficacy in metastatic kidney cancer remains unclear. Here, we examined acarbose, an alpha-glucosidase inhibitor and antidiabetic agent, in a preclinical model of metastatic kidney cancer. We found that acarbose blunted postprandial blood glucose elevations in lean, nondiabetic mice and impeded the growth of orthotopic renal tumors, an outcome that was reversed by exogenous glucose administration. Delayed renal tumor outgrowth in mice on acarbose occurred in a CD8 T cell-dependent manner. Tumors from these mice exhibited increased frequencies of CD8 T cells that retained production of IFNγ, TNFα, perforin, and granzyme B. Combining acarbose with either anti-PD-1 or the mammalian target of rapamycin inhibitor, rapamycin, significantly reduced lung metastases relative to control mice on the same therapies. Our findings in mice suggest that combining acarbose with current RCC therapeutics may improve outcomes, warranting further study to determine whether acarbose can achieve similar responses in advanced RCC patients in a safe and likely cost-effective manner.
Collapse
|
12
|
Turbitt WJ, Orlandella RM, Gibson JT, Peterson CM, Norian LA. Therapeutic Time-restricted Feeding Reduces Renal Tumor Bioluminescence in Mice but Fails to Improve Anti-CTLA-4 Efficacy. Anticancer Res 2020; 40:5445-5456. [PMID: 32988866 PMCID: PMC7957951 DOI: 10.21873/anticanres.14555] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND/AIM Dietary interventions like time-restricted feeding (TRF) show promising anti-cancer properties. We examined whether therapeutic TRF alone or combined with immunotherapy would diminish renal tumor growth in mice of varying body weights. MATERIALS AND METHODS Young (7 week) chow-fed or older (27 week) high-fat diet (HFD)-fed BALB/c mice were orthotopically injected with renal tumor cells expressing luciferase. After tumor establishment, mice were randomized to ad libitum feeding or TRF +/- anti-CTLA-4. Body composition, tumor viability and growth, and immune responses were quantified. RESULTS TRF alone reduced renal tumor bioluminescence in older HFD-fed, but not young chow-fed mice. In the latter, TRF mitigated tumor-induced loss of lean- and fat-mass. However, TRF did not alter excised renal tumor weights or intratumoral immune responses and failed to improve anti-CTLA-4 outcomes in any mice. CONCLUSION Therapeutic TRF exhibits modest anti-cancer properties but fails to improve anti-CTLA-4 immune checkpoint blockade in murine renal cancer.
Collapse
Affiliation(s)
- William J Turbitt
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, U.S.A
| | - Rachael M Orlandella
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, U.S.A
| | - Justin T Gibson
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, U.S.A
| | - Courtney M Peterson
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, U.S.A
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, U.S.A
| | - Lyse A Norian
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, U.S.A.
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, U.S.A
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, U.S.A
| |
Collapse
|
13
|
Is Host Metabolism the Missing Link to Improving Cancer Outcomes? Cancers (Basel) 2020; 12:cancers12092338. [PMID: 32825010 PMCID: PMC7564800 DOI: 10.3390/cancers12092338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
For the past 100 years, oncologists have relentlessly pursued the destruction of tumor cells by surgical, chemotherapeutic or radiation oncological means. Consistent with this focus, treatment plans are typically based on key characteristics of the tumor itself such as disease site, histology and staging based on local, regional and systemic dissemination. Precision medicine is similarly built on the premise that detailed knowledge of molecular alterations of tumor cells themselves enables better and more effective tumor cell destruction. Recently, host factors within the tumor microenvironment including the vasculature and immune systems have been recognized as modifiers of disease progression and are being targeted for therapeutic gain. In this review, we argue that—to optimize the impact of old and new treatment options—we need to take account of an epidemic that occurs independently of—but has major impact on—the development and treatment of malignant diseases. This is the rapidly increasing number of patients with excess weight and its’ attendant metabolic consequences, commonly described as metabolic syndrome. It is well established that patients with altered metabolism manifesting as obesity, metabolic syndrome and chronic inflammation have an increased incidence of cancer. Here, we focus on evidence that these patients also respond differently to cancer therapy including radiation and provide a perspective how exercise, diet or pharmacological agents may be harnessed to improve therapeutic responses in this patient population.
Collapse
|
14
|
Sivagnanalingam U, Beatty PL, Finn OJ. Myeloid derived suppressor cells in cancer, premalignancy and inflammation: A roadmap to cancer immunoprevention. Mol Carcinog 2020; 59:852-861. [PMID: 32333615 DOI: 10.1002/mc.23206] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/04/2020] [Accepted: 04/05/2020] [Indexed: 12/15/2022]
Abstract
The ultimate success of any form of cancer therapy or cancer prevention depends on its ability to engage the power of the immune system to completely eliminate a growing tumor, lower the life-time tumor risk and establish long-term memory to prevent recurrence or future tumors. For that reason, all therapies but especially immunotherapies depend on the immune health (immunocompetence) of each treated individual. Cancer and chronic illnesses, combined with a usually more advanced age of cancer patients or those at risk for cancer are known to severely suppress multiple antitumor functions of the immune system. Understanding the critical mechanisms controlling and mediating immune suppression can lead to additional therapies to alleviate the effects of those mechanisms and improve the outcome of cancer therapy and prevention. We introduce and review here a highly immunosuppressive cell population found in cancer, precancer, and chronic inflammatory diseases, myeloid derived suppressor cells (MDSC). First described in the setting of advanced cancer, their presence and immunosuppressive activity has been seen more recently in early premalignant lesions and in chronic inflammatory diseases leading to cancer. We describe the detrimental effects of their presence on cancer immunotherapy, immunosurveillance and immunoprevention and review early attempts to develop drugs to eliminate them or reduce their negative impact.
Collapse
Affiliation(s)
- Umayal Sivagnanalingam
- School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Pamela L Beatty
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Olivera J Finn
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
15
|
Hutchison S, Sahay B, de Mello SC, Sayour EJ, Lejeune A, Szivek A, Livaccari AM, Fox-Alvarez S, Salute M, Powers L, Milner RJ. Characterization of myeloid-derived suppressor cells and cytokines GM-CSF, IL-10 and MCP-1 in dogs with malignant melanoma receiving a GD3-based immunotherapy. Vet Immunol Immunopathol 2019; 216:109912. [PMID: 31446208 DOI: 10.1016/j.vetimm.2019.109912] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 07/23/2019] [Accepted: 07/30/2019] [Indexed: 01/04/2023]
Abstract
Melanoma in humans and canines is an aggressive and highly metastatic cancer. The mucosal forms in both species share genetic and histopathologic features, making dogs a valuable spontaneous disease animal model. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells of myeloid origin with immunosuppressive capabilities, which are increased in many human cancers and contribute to tumor immune evasion. They are a possible target to improve immunotherapy outcomes. Current information regarding MDSCs in canines is minimal, limiting their use as translational model for the study of MDSCs. The objective of this study was to characterize major MDSCs subsets (monocytic and polymorphonuclear) and the cytokines granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin 10 (IL-10) and monocyte chemoattractant protein-1 (MCP-1) in canines with malignant melanoma and to evaluate changes in MDSCs and the cytokines over time in response to a GD3-based active immunotherapy. Whole blood and serum collected from 30 healthy controls and 33 patients enrolled in the University of Florida melanoma vaccine trial were analyzed by flow cytometry with canine specific CD11b, MHCII and anti-human CD14 antibodies to assess ostensibly polymorphonuclear-MDSC (CD11b+ MHCII- CD14-) and monocytic-MDSC (CD11b+ MHCII- CD14+) subsets. IL-10, MCP-1 and both MDSCs subsets were significantly elevated in melanoma dogs versus controls. Both MDSCs subsets decreased significantly following GD3-based immunotherapy administration but no significant changes in cytokines were seen over time. To our knowledge, this is the first report documenting increased monocytic-MDSCs in canine melanoma. This is consistent with human malignant melanoma data, supporting dogs as a valuable model for therapeutic intervention studies.
Collapse
Affiliation(s)
- S Hutchison
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - B Sahay
- Department of Infectious Disease and Immunology, University of Florida, Gainesville, FL, USA
| | - Souza Ch de Mello
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - E J Sayour
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida Brain Tumor Immunotherapy Program, McKnight Brain Institute, Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - A Lejeune
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - A Szivek
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - A M Livaccari
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - S Fox-Alvarez
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - M Salute
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - L Powers
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, FL, USA
| | - R J Milner
- Department of Small Animal Clinical Sciences, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
16
|
Cao Y, He Y, Wang X, Liu Y, Shi K, Zheng Z, Su X, Lei A, He J, Zhou J. Polymorphonuclear myeloid-derived suppressor cells attenuate allergic airway inflammation by negatively regulating group 2 innate lymphoid cells. Immunology 2019; 156:402-412. [PMID: 30575026 DOI: 10.1111/imm.13040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/20/2018] [Accepted: 12/06/2018] [Indexed: 01/17/2023] Open
Abstract
Hyperactivation of the type 2 immune response is the major mechanism of allergic asthma, in which both group 2 innate lymphoid cells (ILC2s) and type 2 helper T (Th2) cells participate. Myeloid-derived suppressor cells (MDSCs) alleviate asthma by suppressing Th2 cells. However, the potential effects of MDSCs on the biological functions of ILC2s remain largely unknown. Here, we examined the roles of MDSCs (MDSCs) in the modulation of ILC2 function. Our results showed that polymorphonuclear (PMN)-MDSCs, but not monocytic (M-) MDSCs, effectively suppressed the cytokine production of ILC2s both in vitro and in vivo, thereby alleviating airway inflammation. Further analyses showed that cyclo-oxygenase-1 may mediate the suppressive effects of PMN-MDSCs on ILC2 responses. Our findings demonstrated that PMN-MDSCs may serve as a potent therapeutic target for the treatment of ILC2-driven allergic asthma.
Collapse
Affiliation(s)
- Yingjiao Cao
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yumei He
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiangyang Wang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yongdong Liu
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kun Shi
- Department of Obstetrics and Gynaecology, Guangzhou Women and Children's Medical Centre, Guangzhou, China
| | - Zheng Zheng
- Department of Obstetrics and Gynaecology, Guangzhou Women and Children's Medical Centre, Guangzhou, China
| | - Xue Su
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Aihua Lei
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Juan He
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jie Zhou
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Orillion A, Damayanti NP, Shen L, Adelaiye-Ogala R, Affronti H, Elbanna M, Chintala S, Ciesielski M, Fontana L, Kao C, Elzey BD, Ratliff TL, Nelson DE, Smiraglia D, Abrams SI, Pili R. Dietary Protein Restriction Reprograms Tumor-Associated Macrophages and Enhances Immunotherapy. Clin Cancer Res 2018; 24:6383-6395. [PMID: 30190370 DOI: 10.1158/1078-0432.ccr-18-0980] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/03/2018] [Accepted: 08/31/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE Diet and healthy weight are established means of reducing cancer incidence and mortality. However, the impact of diet modifications on the tumor microenvironment and antitumor immunity is not well defined. Immunosuppressive tumor-associated macrophages (TAMs) are associated with poor clinical outcomes and are potentially modifiable through dietary interventions. We tested the hypothesis that dietary protein restriction modifies macrophage function toward antitumor phenotypes. EXPERIMENTAL DESIGN Macrophage functional status under different tissue culture conditions and in vivo was assessed by Western blot, immunofluorescence, qRT-PCR, and cytokine array analyses. Tumor growth in the context of protein or amino acid (AA) restriction and immunotherapy, namely, a survivin peptide-based vaccine or a PD-1 inhibitor, was examined in animal models of prostate (RP-B6Myc) and renal (RENCA) cell carcinoma. All tests were two-sided. RESULTS Protein or AA-restricted macrophages exhibited enhanced tumoricidal, proinflammatory phenotypes, and in two syngeneic tumor models, protein or AA-restricted diets elicited reduced TAM infiltration, tumor growth, and increased response to immunotherapies. Further, we identified a distinct molecular mechanism by which AA-restriction reprograms macrophage function via a ROS/mTOR-centric cascade. CONCLUSIONS Dietary protein restriction alters TAM activity and enhances the tumoricidal capacity of this critical innate immune cell type, providing the rationale for clinical testing of this supportive tool in patients receiving cancer immunotherapies.
Collapse
Affiliation(s)
- Ashley Orillion
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana.,Department of Cellular and Molecular Biology, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Nur P Damayanti
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Li Shen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Remi Adelaiye-Ogala
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana.,Department of Cancer Pathology and Prevention, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Hayley Affronti
- Department of Cellular and Molecular Biology, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - May Elbanna
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Sreenivasulu Chintala
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Michael Ciesielski
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Luigi Fontana
- Charles Perkins Centre and Central Clinical School, The University of Sydney, New South Wales, Australia
| | - Chinghai Kao
- Department of Urology, Indiana University, Indianapolis, Indiana
| | - Bennett D Elzey
- Department of Urology, Indiana University, Indianapolis, Indiana.,Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
| | - Timothy L Ratliff
- Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - David E Nelson
- Department of Microbiology and Immunology, Indiana University, Indianapolis, Indiana
| | - Dominic Smiraglia
- Department of Cellular and Molecular Biology, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Scott I Abrams
- Department of Immunology, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York.
| | - Roberto Pili
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana.
| |
Collapse
|
18
|
Murphy KA, James BR, Sjaastad FV, Kucaba TA, Kim H, Brincks EL, Chua SC, Wilber A, Griffith TS. Cutting Edge: Elevated Leptin during Diet-Induced Obesity Reduces the Efficacy of Tumor Immunotherapy. THE JOURNAL OF IMMUNOLOGY 2018; 201:1837-1841. [PMID: 30135180 DOI: 10.4049/jimmunol.1701738] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 07/27/2018] [Indexed: 12/12/2022]
Abstract
Various malignancies are reproducibly cured in mouse models, but most cancer immunotherapies show objective responses in a fraction of treated patients. One reason for this disconnect may be the use of young, lean mice lacking immune-altering comorbidities present in cancer patients. Although many cancer patients are overweight or obese, the effect of obesity on antitumor immunity is understudied in preclinical tumor models. We examined the effect of obesity on two immunotherapeutic models: systemic anti-CTLA-4 mAb and intratumoral delivery of a TRAIL-encoding adenovirus plus CpG. Both therapies were effective in lean mice, but neither provided a survival benefit to diet-induced obese BALB/c mice. Interestingly, tumor-bearing leptin-deficient (ob/ob) obese BALB/c mice did respond to treatment. Moreover, reducing systemic leptin with soluble leptin receptor:Fc restored the antitumor response in diet-induced obese mice. These data demonstrate the potential of targeting leptin to improve tumor immunotherapy when immune-modulating comorbidities are present.
Collapse
Affiliation(s)
| | - Britnie R James
- Department of Urology, University of Minnesota, Minneapolis, MN 55455
| | - Frances V Sjaastad
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN 55455
| | - Tamara A Kucaba
- Department of Urology, University of Minnesota, Minneapolis, MN 55455
| | - Hyunjoon Kim
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455
| | - Erik L Brincks
- Department of Urology, University of Minnesota, Minneapolis, MN 55455
| | - Streamson C Chua
- Division of Endocrinology and Diabetes, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Andrew Wilber
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702.,Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL 62702
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN 55455; .,Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN 55455.,Center for Immunology, University of Minnesota, Minneapolis, MN 55455; and.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
19
|
Abstract
Current therapies of renal cell carcinoma (RCC), a highly vascularised tumour, mostly rely on anti-angiogenic treatment options. These include tyrosine kinase inhibitors (TKIs) and anti-VEGF monoclonal antibodies. Although these strategies aim at restraining vascularisation to control tumour growth, the effects of such therapies are much wider, as affecting the vessel structure deeply modifies the microenvironment of the tumour mass. The aim of this review is to provide an overview of current knowledge on the global effects of anti-angiogenic treatment, mostly TKIs, on the shaping of the immune component of the RCC microenvironment. The data supporting the modification of immunity by anti-angiogenic therapies are collected to reveal the potential of angiogenesis modulation as a strategy for the adjuvant anti-cancer approach in immunotherapy.
Collapse
|
20
|
He YM, Li X, Perego M, Nefedova Y, Kossenkov AV, Jensen EA, Kagan V, Liu YF, Fu SY, Ye QJ, Zhou YH, Wei L, Gabrilovich DI, Zhou J. Transitory presence of myeloid-derived suppressor cells in neonates is critical for control of inflammation. Nat Med 2018; 24:224-231. [PMID: 29334374 PMCID: PMC5803434 DOI: 10.1038/nm.4467] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 12/12/2017] [Indexed: 12/15/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) are pathologically activated and relatively immature myeloid cells, which are implicated in the immune regulation of many pathologic conditions1,2. Phenotypically and morphologically MDSC are similar to neutrophils (PMN-MDSC) and monocytes (M-MDSC). However, they have potent suppressive activity, a distinct gene expression profile, and biochemical characteristics3. None or very few MDSC are observed in steady state physiological conditions. Therefore, until recently, accumulation of MDSC was considered as a consequence of pathological process or pregnancy. Here, we report that MDSC with a potent ability to suppress T cells are present during the first weeks of life in mice and humans. MDSC suppressive activity was triggered by lactoferrin and mediated by nitric oxide, PGE2, and S100A9/A8 proteins. Newborn MDSC had a transcriptome similar to that of tumor MDSC, but with a strong up-regulation of an antimicrobial gene network and had potent antibacterial activity. MDSC played a critical role in control of experimental necrotizing enterocolitis (NEC) in newborn mice. MDSC in infants with very low-weight, which are prone to the development of NEC, had lower MDSC levels and suppressive activity than infants with normal weight. Thus, the transitory presence of MDSC may be critical for regulation of inflammation in newborns.
Collapse
Affiliation(s)
- Yu-Mei He
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Guangzhou, China
| | - Xing Li
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Guangzhou, China.,Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | | | - Erik A Jensen
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Valerian Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yu-Feng Liu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shu-Yu Fu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qing-Jian Ye
- Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan-Hong Zhou
- Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Lai Wei
- Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Dmitry I Gabrilovich
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Guangzhou, China.,Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Jie Zhou
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Guangzhou, China.,Guangzhou Women and Children's Medical Center, Guangzhou, China
| |
Collapse
|
21
|
Bruchard M, Ghiringhelli F. Effect of Pharmaceutical Compounds on Myeloid-Derived Suppressor Cells. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
22
|
|
23
|
Orillion A, Hashimoto A, Damayanti N, Shen L, Adelaiye-Ogala R, Arisa S, Chintala S, Ordentlich P, Kao C, Elzey B, Gabrilovich D, Pili R. Entinostat Neutralizes Myeloid-Derived Suppressor Cells and Enhances the Antitumor Effect of PD-1 Inhibition in Murine Models of Lung and Renal Cell Carcinoma. Clin Cancer Res 2017; 23:5187-5201. [PMID: 28698201 DOI: 10.1158/1078-0432.ccr-17-0741] [Citation(s) in RCA: 289] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/18/2017] [Accepted: 06/12/2017] [Indexed: 12/16/2022]
Abstract
PURPOSE Recent advances in immunotherapy highlight the antitumor effects of immune checkpoint inhibition despite a relatively limited subset of patients receiving clinical benefit. The selective class I histone deacetylase inhibitor entinostat has been reported to have immunomodulatory activity including targeting of immune suppressor cells in the tumor microenvironment. Thus, we decided to assess whether entinostat could enhance anti-PD-1 treatment and investigate those alterations in the immunosuppressive tumor microenvironment that contribute to the combined antitumor activity. EXPERIMENTAL DESIGN We utilized syngeneic mouse models of lung (LLC) and renal cell (RENCA) carcinoma and assessed immune correlates, tumor growth, and survival following treatment with entinostat (5 or 10 mg/kg, p.o.) and a PD-1 inhibitor (10 and 20 mg/kg, s.c.). RESULTS Entinostat enhanced the antitumor effect of PD-1 inhibition in two syngeneic mouse tumor models by reducing tumor growth and increasing survival. Entinostat inhibited the immunosuppressive function of both polymorphonuclear (PMN)- and monocytic-myeloid derived suppressor cell (M-MDSC) populations. Analysis of MDSC response to entinostat revealed significantly reduced arginase-1, iNOS, and COX-2 levels, suggesting potential mechanisms for the altered function. We also observed significant alterations in cytokine/chemokine release in vivo with a shift toward a tumor-suppressive microenvironment. CONCLUSIONS Our results demonstrate that entinostat enhances the antitumor effect of PD-1 targeting through functional inhibition of MDSCs and a transition away from an immune-suppressive tumor microenvironment. These data provide a mechanistic rationale for the clinical testing and potential markers of response of this novel combination in solid tumor patients.
Collapse
Affiliation(s)
- Ashley Orillion
- Gentourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana.,Department of Cellular and Molecular Biology, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | | | - Nur Damayanti
- Gentourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Li Shen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Remi Adelaiye-Ogala
- Gentourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana.,Department of Cancer Pathology and Prevention, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Sreevani Arisa
- Gentourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Sreenivasulu Chintala
- Gentourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | | | - Chingai Kao
- Department of Urology, Indiana University, Indianapolis, Indiana
| | - Bennett Elzey
- Department of Urology, Indiana University, Indianapolis, Indiana.,Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | | | - Roberto Pili
- Gentourinary Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana. .,Department of Urology, Indiana University, Indianapolis, Indiana
| |
Collapse
|
24
|
Buchta CM, Boi SK, Miller BJ, Milhem MM, Norian LA. Obesity Does Not Exacerbate the Protumorigenic Systemic Environment in Sarcoma Subjects. Immunohorizons 2017; 1:20-28. [PMID: 29202127 PMCID: PMC5711445 DOI: 10.4049/immunohorizons.1700001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sarcomas are a rare but fatal tumor type that accounts for <1% of adult solid malignancies and ~15% of childhood malignancies. Although the use of immunotherapy is being actively investigated for other solid tumors, advances in immunotherapy for sarcoma patients are lacking. To better understand the systemic immune environment in sarcoma patients, we performed a detailed multiplex analysis of serum cytokines, chemokines, and protumorigenic factors from treatment-naive subjects with localized, high-grade sarcoma. Because obesity is a major healthcare issue in the United States, we additionally examined the effects of obesity on serum protein profiles in our sarcoma subject cohort. We found that the systemic host environment is profoundly altered to favor tumor progression, with epidermal growth factor, angiopoietin-2, vascular endothelial growth factor A, IL-6, IL-8, and MIP-1β all increased relative to tumor-free controls (all p < 0.05). Surprisingly, we found that obesity did not exacerbate this protumorigenic profile, as epidermal growth factor and IL-8 decreased with increasing subject body mass index (both p < 0.05 versus normal or overweight subjects). The Th2-related cytokines IL-4, IL-5, and IL-13 were also decreased in the presence of obesity. Thus, although the systemic environment in sarcoma subjects favors tumor progression, obesity does not further aggravate the production of protumorigenic factors.
Collapse
Affiliation(s)
- Claire M Buchta
- Department of Urology, University of Iowa, Iowa City, IA 52242
| | - Shannon K Boi
- Graduate Biomedical Sciences, Immunology Theme, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Benjamin J Miller
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242
| | - Mohammed M Milhem
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242.,Division of Hematology and Oncology, University of Iowa, Iowa City, IA 52242
| | - Lyse A Norian
- Department of Urology, University of Iowa, Iowa City, IA 52242.,Department of Nutrition Sciences, Nutrition Obesity Research Center, and Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233
| |
Collapse
|
25
|
Murphy KA, James BR, Wilber A, Griffith TS. A Syngeneic Mouse Model of Metastatic Renal Cell Carcinoma for Quantitative and Longitudinal Assessment of Preclinical Therapies. J Vis Exp 2017. [PMID: 28448047 DOI: 10.3791/55080] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Renal cell carcinoma (RCC) affects > 60,000 people in the United States annually, and ~ 30% of RCC patients have multiple metastases at the time of diagnosis. Metastatic RCC (mRCC) is incurable, with a median survival time of only 18 months. Immune-based interventions (e.g., interferon (IFN) and interleukin (IL)-2) induce durable responses in a fraction of mRCC patients, and multikinase inhibitors (e.g., sunitinib or sorafenib) or anti-VEGF receptor monoclonal antibodies (mAb) are largely palliative, as complete remissions are rare. Such shortcomings in current therapies for mRCC patients provide the rationale for the development of novel treatment protocols. A key component in the preclinical testing of new therapies for mRCC is a suitable animal model. Beneficial features that recapitulate the human condition include a primary renal tumor, renal tumor metastases, and an intact immune system to investigate any therapy-driven immune effector responses and the formation of tumor-induced immunosuppressive factors. This report describes an orthotopic mRCC mouse model that has all of these features. We describe an intrarenal implantation technique using the mouse renal adenocarcinoma cell line Renca, followed by the assessment of tumor growth in the kidney (primary site) and lungs (metastatic site).
Collapse
Affiliation(s)
- Katherine A Murphy
- Department of Urology, University of Minnesota; Masonic Cancer Center, University of Minnesota
| | - Britnie R James
- Department of Urology, University of Minnesota; Masonic Cancer Center, University of Minnesota; Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota
| | - Andrew Wilber
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine; Simmons Cancer Institute
| | - Thomas S Griffith
- Department of Urology, University of Minnesota; Masonic Cancer Center, University of Minnesota; Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota;
| |
Collapse
|
26
|
Sher YP, Lin SI, Chen IH, Liu HY, Lin CY, Chiang IP, Roffler S, Chen HW, Liu SJ. A HLA-A2-restricted CTL epitope induces anti-tumor effects against human lung cancer in mouse xenograft model. Oncotarget 2016; 7:671-83. [PMID: 26621839 PMCID: PMC4808025 DOI: 10.18632/oncotarget.6400] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 11/16/2015] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapy is attractive for antigen-specific T cell-mediated anti-tumor therapy, especially in induction of cytotoxic T lymphocytes. In this report, we evaluated human CTL epitope-induced anti-tumor effects in human lung cancer xenograft models. The tumor associated antigen L6 (TAL6) is highly expressed in human lung cancer cell lines and tumor specimens as compared to normal lung tissues. TAL6 derived peptides strongly inhibited tumor growth, cancer metastasis and prolonged survival time in HLA-A2 transgenic mice immunized with a formulation of T-helper (Th) peptide, synthetic CpG ODN, and adjuvant Montanide ISA-51 (ISA-51). Adoptive transfer of peptide-induced CTL cells from HLA-A2 transgenic mice into human tumor xenograft SCID mice significantly inhibited tumor growth. Furthermore, combination of CTL-peptide immunotherapy and gemcitabine additively improved the therapeutic effects. This pre-clinical evaluation model provides a useful platform to develop efficient immunotherapeutic drugs to treat lung cancer and demonstrates a promising strategy with benefit of antitumor immune responses worthy of further development in clinical trials.
Collapse
Affiliation(s)
- Yuh-Pyng Sher
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Su-I Lin
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli, Taiwan.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - I-Hua Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Hsin-Yu Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Chen-Yuan Lin
- Division of Hematology and Oncology, China Medical University Hospital, Taichung, Taiwan
| | - I-Ping Chiang
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Steve Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsin-Wei Chen
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan.,National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Shih-Jen Liu
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan.,National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli, Taiwan.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
27
|
CD8 T Cell-Independent Antitumor Response and Its Potential for Treatment of Malignant Gliomas. Cancers (Basel) 2016; 8:cancers8080071. [PMID: 27472363 PMCID: PMC4999780 DOI: 10.3390/cancers8080071] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/30/2016] [Accepted: 07/19/2016] [Indexed: 01/06/2023] Open
Abstract
Malignant brain tumors continue to represent a devastating diagnosis with no real chance for cure. Despite an increasing list of potential salvage therapies, standard-of-care for these patients has not changed in over a decade. Immunotherapy has been seen as an exciting option, with the potential to offer specific and long lasting tumor clearance. The “gold standard” in immunotherapy has been the development of a tumor-specific CD8 T cell response to potentiate tumor clearance and immunological memory. While many advances have been made in the field of immunotherapy, few therapies have seen true success. Many of the same principles used to develop immunotherapy in tumors of the peripheral organs have been applied to brain tumor immunotherapy. The immune-specialized nature of the brain should call into question whether this approach is appropriate. Recent results from our own experiments require a rethinking of current dogma. Perhaps a CD8 T cell response is not sufficient for an organ as immunologically unique as the brain. Examination of previously elucidated principles of the brain’s immune-specialized status and known immunological preferences should generate discussion and experimentation to address the failure of current therapies.
Collapse
|
28
|
Murphy KA, James BR, Guan Y, Torry DS, Wilber A, Griffith TS. Exploiting natural anti-tumor immunity for metastatic renal cell carcinoma. Hum Vaccin Immunother 2016; 11:1612-20. [PMID: 25996049 DOI: 10.1080/21645515.2015.1035849] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Clinical observations of spontaneous disease regression in some renal cell carcinoma (RCC) patients implicate a role for tumor immunity in controlling this disease. Puzzling, however, are findings that high levels of tumor infiltrating lymphocytes (TIL) are common to RCC. Despite expression of activation markers by TILs, functional impairment of innate and adaptive immune cells has been consistently demonstrated contributing to the failure of the immune system to control RCC. Immunotherapy can overcome the immunosuppressive effects of the tumor and provide an opportunity for long-term disease free survival. Unfortunately, complete response rates remain sub-optimal indicating the effectiveness of immunotherapy remains limited by tumor-specific factors and/or cell types that inhibit antitumor immune responses. Here we discuss immunotherapies and the function of multiple immune system components to achieve an effective response. Understanding these complex interactions is essential to rationally develop novel therapies capable of renewing the immune system's ability to respond to these tumors.
Collapse
Affiliation(s)
- Katherine A Murphy
- a Department of Urology; University of Minnesota ; Minneapolis , MN , USA
| | | | | | | | | | | |
Collapse
|
29
|
De Vlaeminck Y, González-Rascón A, Goyvaerts C, Breckpot K. Cancer-Associated Myeloid Regulatory Cells. Front Immunol 2016; 7:113. [PMID: 27065074 PMCID: PMC4810015 DOI: 10.3389/fimmu.2016.00113] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/14/2016] [Indexed: 12/25/2022] Open
Abstract
Myeloid cells are critically involved in the pathophysiology of cancers. In the tumor microenvironment (TME), they comprise tumor-associated macrophages (TAMs), neutrophils (TANs), dendritic cells, and myeloid-derived suppressor cells, which are further subdivided into a monocytic subset and a granulocytic subset. Some of these myeloid cells, in particular TAMs and TANs, are divided into type 1 or type 2 cells, according to the paradigm of T helper type 1 or type 2 cells. Type 1-activated cells are generally characterized as cells that aid tumor rejection, while all other myeloid cells are shown to favor tumor progression. Moreover, these cells are often at the basis of resistance to various therapies. Much research has been devoted to study the biology of myeloid cells. This endeavor has proven to be challenging, as the markers used to categorize myeloid cells in the TME are not restricted to particular subsets. Also from a functional and metabolic point of view, myeloid cells share many features. Finally, myeloid cells are endowed with a certain level of plasticity, which further complicates studying them outside their environment. In this article, we challenge the exclusive use of cell markers to unambiguously identify myeloid cell subsets in the TME. We further propose to divide myeloid cells into myeloid regulatory or stimulatory cells according to their pro- or antitumor function, because we contend that for therapeutic purposes it is not targeting the cell subsets but rather targeting their protumor traits; hence, myeloid regulatory cells will push antitumor immunotherapy to the next level.
Collapse
Affiliation(s)
- Yannick De Vlaeminck
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel , Brussels , Belgium
| | - Anna González-Rascón
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium; Centro de Investigación en Alimentación y Desarrollo, Hermosillo, Mexico
| | - Cleo Goyvaerts
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel , Brussels , Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel , Brussels , Belgium
| |
Collapse
|
30
|
Brincks EL, Kucaba TA, James BR, Murphy KA, Schwertfeger KL, Sangwan V, Banerjee S, Saluja AK, Griffith TS. Triptolide enhances the tumoricidal activity of TRAIL against renal cell carcinoma. FEBS J 2015; 282:4747-4765. [PMID: 26426449 DOI: 10.1111/febs.13532] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 08/19/2015] [Accepted: 09/23/2015] [Indexed: 12/11/2022]
Abstract
Renal cell carcinoma (RCC) is resistant to traditional cancer therapies, and metastatic RCC (mRCC) is incurable. The shortcomings in current therapeutic options for patients with mRCC provide the rationale for the development of novel treatment protocols. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has proven to be a potent inducer of tumor cell death in vitro and in vivo, and a number of TRAIL death receptor agonists (recombinant TRAIL or TRAIL death receptor-specific mAb) have been developed and tested clinically. Unfortunately the clinical efficacy of TRAIL has been underwhelming and is likely due to a number of possible mechanisms that render tumors resistant to TRAIL, prompting the search for drugs that increase tumor cell susceptibility to TRAIL. The objective of this study was to determine the effectiveness of combining the diterpene triepoxide triptolide, or its water-soluble prodrug, Minnelide, with TRAIL receptor agonists against RCC in vitro or in vivo, respectively. TRAIL-induced apoptotic death of human RCC cells was increased in the presence of triptolide. The triptolide-induced sensitization was accompanied by increased TRAIL-R2 (DR5) and decreased heat shock protein 70 expression. In vivo treatment of mice bearing orthotopic RCC (Renca) tumors showed the combination of Minnelide and agonistic anti-DR5 mAb significantly decreased tumor burden and increased animal survival compared to either therapy alone. Our data suggest triptolide/Minnelide sensitizes RCC cells to TRAIL-induced apoptosis through altered TRAIL death receptor and heat shock protein expression.
Collapse
Affiliation(s)
- Erik L Brincks
- Department of Urology, University of Minnesota, Minneapolis, MN 55455
| | - Tamara A Kucaba
- Department of Urology, University of Minnesota, Minneapolis, MN 55455
| | - Britnie R James
- Department of Urology, University of Minnesota, Minneapolis, MN 55455
| | | | - Kathryn L Schwertfeger
- Department of Lab Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455.,Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN 55455.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
| | - Veena Sangwan
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455
| | - Sulagna Banerjee
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455
| | - Ashok K Saluja
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455.,Department of Surgery, University of Minnesota, Minneapolis, MN 55455
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN 55455.,Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN 55455.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455.,Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
31
|
Hale M, Itani F, Buchta CM, Wald G, Bing M, Norian LA. Obesity triggers enhanced MDSC accumulation in murine renal tumors via elevated local production of CCL2. PLoS One 2015; 10:e0118784. [PMID: 25769110 PMCID: PMC4358922 DOI: 10.1371/journal.pone.0118784] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 01/21/2015] [Indexed: 01/02/2023] Open
Abstract
Obesity is one of the leading risk factors for developing renal cell carcinoma, an immunogenic tumor that is treated clinically with immunostimulatory therapies. Currently, however, the mechanisms linking obesity with renal cancer incidence are unclear. Using a model of diet-induced obesity, we found that obese BALB/c mice with orthotopic renal tumors had increased total frequencies of myeloid-derived suppressor cells (MDSC) in renal tumors and spleens by d14 post-tumor challenge, relative to lean counterparts. Renal tumors from obese mice had elevated concentrations of the known myeloid cell chemoattractant CCL2, which was produced locally by increased percentages of dendritic cells, macrophages, B cells, and CD45- cells in tumors. MDSC expression of the CCL2 receptor, CCR2, was unaltered by obesity but greater percentages of CCR2+ MDSCs were present in renal tumors from obese mice. Of note, the intracellular arginase levels and per-cell suppressive capacities of tumor-infiltrating and splenic MDSCs were unchanged in obese mice relative to lean controls. Thus, our findings suggest that obesity promotes renal tumor progression via development of a robust immunosuppressive environment that is characterized by heightened local and systemic MDSC prevalence. Targeted intervention of the CCL2/CCR2 pathway may facilitate immune-mediated renal tumor clearance in the obese.
Collapse
Affiliation(s)
- Malika Hale
- Department of Urology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Farah Itani
- Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Claire M. Buchta
- Department of Urology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Gal Wald
- Department of Urology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Megan Bing
- Department of Urology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Lyse A. Norian
- Department of Urology, The University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, Iowa, United States of America
- Fraternal Order of Eagles Diabetes Research Center, The University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
32
|
Messmer MN, Netherby CS, Banik D, Abrams SI. Tumor-induced myeloid dysfunction and its implications for cancer immunotherapy. Cancer Immunol Immunother 2014; 64:1-13. [PMID: 25432147 DOI: 10.1007/s00262-014-1639-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 11/19/2014] [Indexed: 01/27/2023]
Abstract
Immune function relies on an appropriate balance of the lymphoid and myeloid responses. In the case of neoplasia, this balance is readily perturbed by the dramatic expansion of immature or dysfunctional myeloid cells accompanied by a reciprocal decline in the quantity/quality of the lymphoid response. In this review, we seek to: (1) define the nature of the atypical myelopoiesis observed in cancer patients and the impact of this perturbation on clinical outcomes; (2) examine the potential mechanisms underlying these clinical manifestations; and (3) explore potential strategies to restore normal myeloid cell differentiation to improve activation of the host antitumor immune response. We posit that fundamental alterations in myeloid homeostasis triggered by the neoplastic process represent critical checkpoints that govern therapeutic efficacy, as well as offer novel cellular-based biomarkers for tracking changes in disease status or relapse.
Collapse
Affiliation(s)
- Michelle N Messmer
- Department of Immunology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | | | | | | |
Collapse
|