1
|
Wang X, Wang X, Wang D, Zhou C, Lv K, Ma Y, Chang W, Wang B, Hu J, Ji Y, Dai Z, Ma Y. Interleukin-10 overexpression in 4T1 cells: A gateway to suppressing mammary carcinoma growth. Int Immunopharmacol 2024; 142:113089. [PMID: 39244897 DOI: 10.1016/j.intimp.2024.113089] [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] [Revised: 08/04/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
Interleukin-10 (IL-10) exerts complex effects on tumor growth, exhibiting both pro- and anti-tumor properties. Recent focus on the anti-inflammatory properties of IL-10 has highlighted its potential anti-tumor properties, particularly through the enhancement of CD8+ T cell activity. However, further research is needed to fully elucidate its other anti-tumor mechanisms. Our study investigates novel anti-tumor mechanisms of IL-10 in a murine mammary carcinoma model (4T1). We found that IL-10 overexpression in mouse 4T1 cells suppressed tumor growth in vivo. This suppression was accompanied by an increase in IFN-γ-secreting CD8+ T cells and a decrease in myeloid-derived suppressor cells (MDSCs) in tumor tissue. In vitro experiments showed that IL-10-rich tumor cell-derived supernatants inhibited myeloid cell differentiation into monocytic and granulocytic MDSCs while reducing MDSCs migration. In addition, IL-10 overexpression downregulated CXCL5 expression in 4T1 cells, resulting in decreased CXCR2+ MDSCs infiltration. Using RAG1-deficient mice and CXCL5 knockdown tumor models, we demonstrated that the anti-tumor effects of IL-10 depend on both CD8+ T cells and reduced MDSC infiltration. IL-10 attenuated the immunosuppressive tumor microenvironment by enhancing CD8+ T cell activity and inhibiting MDSCs infiltration. In human breast cancer, we observed a positive correlation between CXCL5 expression and MDSC infiltration. Our findings reveal a dual mechanism of IL-10-mediated tumor suppression: (1) direct enhancement of CD8+ T cell activity and (2) indirect reduction of immunosuppressive MDSCs through CXCL5 downregulation and inhibition of myeloid cell differentiation. This study provides new insights into the role of IL-10 in anti-tumor immunity and suggests potential strategies for breast cancer immunotherapy by modulating the IL-10-CXCL5-MDSCs axis.
Collapse
Affiliation(s)
- Xiaoqin Wang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoqian Wang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Dan Wang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Can Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Kaige Lv
- Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yanfen Ma
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Wenjing Chang
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Baofeng Wang
- Department of Radiotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jian Hu
- The Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yanhong Ji
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Yunfeng Ma
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China.
| |
Collapse
|
2
|
Rajkumari S, Singh J, Agrawal U, Agrawal S. Myeloid-derived suppressor cells in cancer: Current knowledge and future perspectives. Int Immunopharmacol 2024; 142:112949. [PMID: 39236460 DOI: 10.1016/j.intimp.2024.112949] [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: 05/30/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
MDSCs (myeloid-derived suppressor cells) are crucial for immune system evasion in cancer. They accumulate in peripheral blood and tumor microenvironment, suppressing immune cells like T-cells, natural killer cells and dendritic cells. They promote tumor angiogenesis and metastasis by secreting cytokines and growth factors and contribute to a tumor-promoting environment. The accumulation of MDSCs in cancer patients has been linked to poor prognosis and resistance to various cancer therapies. Targeting MDSCs and their immunosuppressive mechanisms may improve treatment outcomes and enhance immune surveillance by developing drugs that inhibit MDSC function, by preventing their accumulation and by disrupting the tumor-promoting environment. This review presents a detailed overview of the MDSC research in cancer with regulation of their development and function. The relevance of MDSC as a prognostic and predictive biomarker in different types of cancers, along with recent advancements on the therapeutic approaches to target MDSCs are discussed in detail.
Collapse
Affiliation(s)
- Sunanda Rajkumari
- ICMR National Institute of Medical Statistics, Ansari Nagar, New Delhi 110029, India
| | - Jaspreet Singh
- ICMR National Institute of Pathology, Safdarjung Hospital Campus, Ansari Nagar, New Delhi 110029, India
| | - Usha Agrawal
- Asian Institute of Public Health University (AIPH) University, 1001 Haridamada, Jatani, Near IIT Bhubaneswar, Bhubaneswar 751002, India
| | - Sandeep Agrawal
- Discovery Research Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India.
| |
Collapse
|
3
|
Kitelinger LE, Thim EA, Zipkowitz SY, Price RJ, Bullock TNJ. Tissue- and Temporal-Dependent Dynamics of Myeloablation in Response to Gemcitabine Chemotherapy. Cells 2024; 13:1317. [PMID: 39195207 DOI: 10.3390/cells13161317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/29/2024] Open
Abstract
For triple-negative breast cancer (TNBC), the most aggressive subset of breast cancer, immune cell infiltrates have prognostic implications. The presence of myeloid-derived suppressor cells supports tumor progression, while tumor-infiltrating lymphocytes (TILs) correlate with improved survival and responsiveness to immunotherapy. Manipulating the abundance of these populations may enhance tumor immunity. Gemcitabine (GEM), a clinically employed chemotherapeutic, is reported to be systemically myeloablative, and thus it is a potentially useful adjunct therapy for promoting anti-tumor immunity. However, knowledge about the immunological effects of GEM intratumorally is limited. Thus, we directly compared the impact of systemic GEM on immune cell presence and functionality in the tumor microenvironment (TME) to its effects in the periphery. We found that GEM is not myeloablative in the TME; rather, we observed sustained, significant reductions in TILs and dendritic cells-crucial components in initiating an adaptive immune response. We also performed bulk-RNA sequencing to identify immunological alterations transcriptionally induced by GEM. While we found evidence of upregulation in the interferon-gamma (IFN-γ) response pathway, we determined that GEM-mediated growth control is not dependent on IFN-γ. Overall, our findings yield new insights into the tissue- and temporal-dependent immune ablative effects of GEM, contrasting the paradigm that this therapy is specifically myeloablative.
Collapse
Affiliation(s)
- Lydia E Kitelinger
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Eric A Thim
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Sarah Y Zipkowitz
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Timothy N J Bullock
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
4
|
Li YL, Chen CH, Lai YS, Pan MR, Hung WC. Increased blood CSF3R + myeloid-derived suppressor cell is a predictor for breast cancer recurrence. Am J Cancer Res 2024; 14:3171-3185. [PMID: 39005677 PMCID: PMC11236774 DOI: 10.62347/mukd2745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
Early detection of cancer recurrence using specific biomarkers remains a clinically unmet need, although methodologies for monitoring tumor markers, cell-free DNA, and circulating tumor cells have been established for decades. Tumor recurrence develops in metastatic or dormant cancer cells under continuous immune surveillance. Alterations in the population and function of immune cells may contribute to cancer recurrence. Here, we utilized an animal model to imitate breast tumor recurrence after surgical resection and investigated the abundance and gene expression profiles of immune cells using NanoString analysis. Bioinformatic analysis of a published single-cell RNA sequencing database of myeloid-derived suppressor cells (MDSCs) was performed to identify common targets between the two studies. Identified biomarkers were validated using human peripheral blood mononuclear cell (PBMC) datasets. The inhibitory effect of MDSCs on T-cell proliferation was assessed in vitro. Our data demonstrated that the number of MDSCs significantly increased during recurrence. Comparison of our NanoString data with a single-cell RNA sequencing dataset of MDSCs in another spontaneous breast cancer model identified colony-stimulating factor 3 receptor (Csf3r)-positive MDSCs as a potential marker for predicting tumor relapse. We validated our findings using two previously published PBMC databases of patients with breast cancer with or without recurrence and confirmed the elevated MDSC gene signature and CSF3R expression in patients with tumor recurrence. 35 patients with breast cancer were also included in our study, that patients with higher levels of CSF3R had worse survival. In vitro experiments demonstrated that Csf3r + MDSCs exhibited enhanced reactive oxygen species (ROS) levels and robust T-cell suppression ability. We conclude that an increase in CSF3R + MDSCs is a potential biomarker for early detection of tumor recurrence in patients with breast cancer.
Collapse
Affiliation(s)
- Yen-Liang Li
- National Institute of Cancer Research, National Health Research InstitutesTainan 704, Taiwan
| | - Chung-Hsing Chen
- National Institute of Cancer Research, National Health Research InstitutesTainan 704, Taiwan
| | - You-Syuan Lai
- National Institute of Cancer Research, National Health Research InstitutesTainan 704, Taiwan
| | - Mei-Ren Pan
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical UniversityKaohsiung 807, Taiwan
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research InstitutesTainan 704, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical UniversityKaohsiung 807, Taiwan
| |
Collapse
|
5
|
Larson AC, Doty KR, Solheim JC. The double life of a chemotherapy drug: Immunomodulatory functions of gemcitabine in cancer. Cancer Med 2024; 13:e7287. [PMID: 38770637 PMCID: PMC11106691 DOI: 10.1002/cam4.7287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/22/2024] Open
Abstract
Although the development of immunotherapies has been revolutionary in the treatment of several cancers, many cancer types remain unresponsive to immune-based treatment and are largely managed by chemotherapy drugs. However, chemotherapeutics are not infallible and are frequently rendered ineffective as resistance develops from prolonged exposure. Recent investigations have indicated that some chemotherapy drugs have additional functions beyond their normative cytotoxic capacity and are in fact immune-modifying agents. Of the pharmaceuticals with identified immune-editing properties, gemcitabine is well-studied and of interest to clinicians and scientists alike. Gemcitabine is a chemotherapy drug approved for the treatment of multiple cancers, including breast, lung, pancreatic, and ovarian. Because of its broad applications, relatively low toxicity profile, and history as a favorable combinatory partner, there is promise in the recharacterization of gemcitabine in the context of the immune system. Such efforts may allow the identification of suitable immunotherapeutic combinations, wherein gemcitabine can be used as a priming agent to improve immunotherapy efficacy in traditionally insensitive cancers. This review looks to highlight documented immunomodulatory abilities of one of the most well-known chemotherapy agents, gemcitabine, relating to its influence on cells and proteins of the immune system.
Collapse
Affiliation(s)
- Alaina C. Larson
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Kenadie R. Doty
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Joyce C. Solheim
- Eppley Institute for Research in Cancer & Allied DiseasesUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Department of Biochemistry & Molecular BiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Department of Pathology, Microbiology, & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
6
|
Yokota S, Yonezawa T, Momoi Y, Maeda S. Myeloid derived suppressor cells in peripheral blood can be a prognostic factor in canine transitional cell carcinoma. Vet Immunol Immunopathol 2024; 269:110716. [PMID: 38308864 DOI: 10.1016/j.vetimm.2024.110716] [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: 11/14/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/05/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature cells with immunosuppressive properties found in the tumor microenvironment. MDSCs are divided into two major subsets: polymorphonuclear MDSCs (PMN-MDSCs) and monocytic MDSCs (M-MDSCs). Both MDSC subsets contribute to the creation of an immunosuppressive environment for tumor progression. In humans, patients with high levels of MDSCs show worse outcomes for several types of cancers. However, the association between MDSCs and clinical features has rarely been investigated in canine studies. In the present study, we measured the proportion of PMN-MDSCs and M-MDSCs in the peripheral blood and tumor tissue of dogs with hepatocellular carcinoma (HCC), prostate cancer (PC), transitional cell carcinoma (TCC), lymphoma, and pulmonary adenocarcinoma. Additionally, we examined immunosuppressive ability of PMN-MDSCs and M-MDSCs in peripheral blood mononuclear cells of TCC case on CD4+, CD8+ and interferon-γ+ cells and investigated the relationships of MDSCs with clinical features and outcomes. PMN-MDSCs increased in HCC, PC, TCC, and lymphoma. In contrast, M-MDSCs increased in the TCC. Both PMN-MDSCs and M-MDSCs exhibited immunosuppressive effects on CD8+, CD4+ and interferon-γ+ cells. In dogs with TCC, lymph node metastasis was associated with high level of PMN-MDSCs but not with M-MDSCs. High levels of both PMN-MDSCs and M-MDSCs were related to advanced tumor stage. Kaplan-Meier analysis revealed that high levels of both PMN-MDSCs and M-MDSCs were significantly associated with shorter overall survival. In addition, the Cox proportional hazard regression model showed that M-MDSCs and the tumor stage were independent prognostic factors for TCC. These results suggest that PMN-MDSCs and M-MDSCs may be involved in tumor progression and could be prognostic factors and promising therapeutic targets in dogs with TCC.
Collapse
Affiliation(s)
- Shohei Yokota
- Department of Veterinary Clinical Pathobiology, Guraduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Tomohiro Yonezawa
- Department of Veterinary Clinical Pathobiology, Guraduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yasuyuki Momoi
- Department of Veterinary Clinical Pathobiology, Guraduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Shingo Maeda
- Department of Veterinary Clinical Pathobiology, Guraduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| |
Collapse
|
7
|
Sun SH, Angell CD, Savardekar H, Sundi D, Abood D, Benner B, DiVincenzo MJ, Duggan M, Choueiry F, Mace T, Trikha P, Lapurga G, Johnson C, Carlson EJ, Chung C, Peterson BR, Lianbo Yu, Zhao J, Kendra KL, Carson WE. BTK inhibition potentiates anti-PD-L1 treatment in murine melanoma: potential role for MDSC modulation in immunotherapy. Cancer Immunol Immunother 2023; 72:3461-3474. [PMID: 37528320 PMCID: PMC10592087 DOI: 10.1007/s00262-023-03497-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 07/07/2023] [Indexed: 08/03/2023]
Abstract
Myeloid-derived suppressor cells (MDSC) have been linked to loss of immune effector cell function through a variety of mechanisms such as the generation of reactive oxygen and nitrogen species and the production of inhibitory cytokines. Our group has shown that signaling through Bruton's tyrosine kinase (BTK) is important for MDSC function. Ibrutinib is an orally administered targeted agent that inhibits BTK activation and is currently used for the treatment of B cell malignancies. Using a syngeneic murine model of melanoma, the effect of BTK inhibition with ibrutinib on the therapeutic response to systemic PD-L1 blockade was studied. BTK was expressed by murine MDSC and their activation was inhibited by ibrutinib. Ibrutinib was not directly cytotoxic to cancer cells in vitro, but it inhibited BTK activation in MDSC and reduced expression of inducible nitric oxide synthase (NOS2) and production of nitric oxide. Ibrutinib treatments decreased the levels of circulating MDSC in vivo and increased the therapeutic efficacy of anti-PD-L1 antibody treatment. Gene expression profiling showed that ibrutinib decreased Cybb (NOX2) signaling, and increased IL-17 signaling (upregulating downstream targets Mmp9, Ptgs2, and S100a8). These results suggest that further exploration of MDSC inhibition could enhance the immunotherapy of advanced melanoma.PrécisInhibition of Bruton's tyrosine kinase, a key enzyme in myeloid cellular function, improves therapeutic response to an anti-PD-L1 antibody in an otherwise fairly resistant murine melanoma model.
Collapse
Affiliation(s)
- Steven H Sun
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Colin D Angell
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Himanshu Savardekar
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Debasish Sundi
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Urology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - David Abood
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Brooke Benner
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Mallory J DiVincenzo
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Megan Duggan
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Fouad Choueiry
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Thomas Mace
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Division of Gastrointestinal Oncology, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Prashant Trikha
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Gabriella Lapurga
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Courtney Johnson
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Erick J Carlson
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Catherine Chung
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Blake R Peterson
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Lianbo Yu
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jing Zhao
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kari L Kendra
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - William E Carson
- James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
8
|
Liu T, Rosek A, Gonzalez De Los Santos F, Phan SH. Detection of myeloid-derived suppressor cells by flow cytometry. Methods Cell Biol 2023; 184:1-15. [PMID: 38555150 DOI: 10.1016/bs.mcb.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Recently discovered heterogeneous myeloid-derived suppressor cells (MDSCs) are some of the most discussed immunosuppressive cells in contemporary immunology, especially in the tumor microenvironment, and are defined primarily by their T cell immunosuppressive function. The importance of these cells extend to other chronic pathological conditions as well, including chronic infection, inflammation, and tissue remodeling. In many of these conditions, their accumulation/expansion correlates with disease progression, poor prognosis, and reduced survival, which highlights the potential of how these cells may be used in a clinical setting as both prognostic factor and therapeutic target. In healthy individuals, these cells are usually not present in the circulation. Therefore, monitoring this cell population is of potential clinical significance, and utility in basic research. However, these cells have a complex phenotype without one single marker of sufficient specificity for their identification. Flow cytometry is a powerful tool allowing multi-parameter analysis of heterogeneous cell populations, which makes it ideally suitable for the complex phenotypic analysis essential for identification and enumeration of circulating MDSCs. This approach has the potential to provide a novel clinically useful tool for assessment of prognosis and treatment outcomes. The protocol in this chapter describes a flow cytometric analysis to identify and quantify MDSCs from human or mouse whole blood leukocytes and peripheral blood mononuclear cells, as well as a single cell suspension from solid tissue, by using multicolor fluorescence-conjugated antibodies against their surface markers.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States.
| | - Alyssa Rosek
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Sem H Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
9
|
Sooi K, Walsh R, Kumarakulasinghe N, Wong A, Ngoi N. A review of strategies to overcome immune resistance in the treatment of advanced prostate cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:656-673. [PMID: 37842236 PMCID: PMC10571060 DOI: 10.20517/cdr.2023.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/06/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023]
Abstract
Immunotherapy has become integral in cancer therapeutics over the past two decades and is now part of standard-of-care treatment in multiple cancer types. While various biomarkers and pathway alterations such as dMMR, CDK12, and AR-V7 have been identified in advanced prostate cancer to predict immunotherapy responsiveness, the vast majority of prostate cancer remain intrinsically immune-resistant, as evidenced by low response rates to anti-PD(L)1 monotherapy. Since regulatory approval of the vaccine therapy sipuleucel-T in the biomarker-unselected population, there has not been much success with immunotherapy treatment in advanced prostate cancer. Researchers have looked at various strategies to overcome immune resistance, including the identification of more biomarkers and the combination of immunotherapy with existing effective prostate cancer treatments. On the horizon, novel drugs using bispecific T-cell engager (BiTE) and chimeric antigen receptors (CAR) technology are being explored and have shown promising early efficacy in this disease. Here we discuss the features of the tumour microenvironment that predispose to immune resistance and rational strategies to enhance antitumour responsiveness in advanced prostate cancer.
Collapse
Affiliation(s)
| | | | | | | | - Natalie Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore 119228, Singapore
| |
Collapse
|
10
|
Fukui R, Watanabe T, Morimoto K, Fujimoto Y, Nagahashi M, Ishikawa E, Hirota S, Miyoshi Y. An increase in tumor-infiltrating lymphocytes after treatment is significantly associated with a poor response to neoadjuvant endocrine therapy for estrogen receptor-positive/HER2-negative breast cancers. Breast Cancer 2023; 30:703-713. [PMID: 37115435 PMCID: PMC10404203 DOI: 10.1007/s12282-023-01462-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND The reason for the poor prognosis of estrogen receptor (ER) + /human epidermal growth factor receptor 2 (HER2)- breast cancer patients with high levels of tumor-infiltrating lymphocytes (TILs) is poorly understood. The association between TILs and response to neoadjuvant endocrine therapy (NET) was examined. METHODS We recruited 170 patients with ER + /HER2- breast cancer who were treated with preoperative endocrine monotherapy. TILs were evaluated before and after NET, and their changes were noted. Furthermore, T cell subtypes were examined using CD8 and FOXP3 immunohistochemical analyses. Neutrophil and lymphocyte counts in the peripheral blood were analyzed with reference to TIL levels or changes. Responders were defined as Ki67 expression levels ≤ 2.7% after treatment. RESULTS Post-treatment (p = 0.016), but not pre-treatment (p = 0.464), TIL levels were significantly associated with the response to NET. TIL levels increased significantly after treatment among non-responders (p = 0.001). FOXP3 + T cell counts increased significantly after treatment in patients with increased TILs (p = 0.035), but not in those without increased TILs (p = 0.281). Neutrophil counts decreased significantly after treatment in patients without increased TILs (p = 0.026), but not in patients with increased TILs (p = 0.312). CONCLUSION An increase in TILs after NET was significantly associated with a poor response to NET. Given that FOXP3 + T-cell counts increased, and neutrophil counts did not decrease in patients with increased TILs after NET, the induction of an immunosuppressive microenvironment was speculated to play a role in the inferior efficacy. These data might partially indicate the involvement of the immune response in the efficacy of endocrine therapy.
Collapse
Affiliation(s)
- Reiko Fukui
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, Mukogawa-Cho 1-1, Nishinomiya City, Hyogo, 663-8501, Japan
| | - Takahiro Watanabe
- Department of Clinical Pathology, Chibune General Hospital, Osaka, Japan
| | - Koji Morimoto
- Department of Nutrition, College of Nutrition, Koshien University, Takarazuka, Hyogo, Japan
| | - Yukie Fujimoto
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, Mukogawa-Cho 1-1, Nishinomiya City, Hyogo, 663-8501, Japan
| | - Masayuki Nagahashi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, Mukogawa-Cho 1-1, Nishinomiya City, Hyogo, 663-8501, Japan
| | - Eri Ishikawa
- Department of Surgical Pathology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Seiichi Hirota
- Department of Surgical Pathology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Yasuo Miyoshi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, Mukogawa-Cho 1-1, Nishinomiya City, Hyogo, 663-8501, Japan.
| |
Collapse
|
11
|
Gjerstorff MF. Crosstalk between interferon and interleukin-1 antiviral signaling in cancer cells: implications for immune evasion and therapeutic resistance. Front Immunol 2023; 14:1219870. [PMID: 37359511 PMCID: PMC10285489 DOI: 10.3389/fimmu.2023.1219870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/29/2023] [Indexed: 06/28/2023] Open
Affiliation(s)
- Morten Frier Gjerstorff
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, ;Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| |
Collapse
|
12
|
Yokota S, Kaji K, Yonezawa T, Momoi Y, Maeda S. CD204⁺ tumor-associated macrophages are associated with clinical outcome in canine pulmonary adenocarcinoma and transitional cell carcinoma. Vet J 2023; 296-297:105992. [PMID: 37164121 DOI: 10.1016/j.tvjl.2023.105992] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 05/04/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
Tumor-associated macrophages are abundant infiltrating cells in the tumor microenvironment (TME). Macrophages can be classified into several types of subsets based on their immune responses. Among those subsets, M2 macrophages contribute to anti-inflammatory responses and create an immunosuppressive environment that promotes tumor cell proliferation. In a previous study, human cancer patients with high M2 macrophages showed a worse prognosis for many types of tumors. However, studies examining the relationship between M2 macrophages and clinical outcomes in canine tumors are limited. In the previous human and canine studies, CD204 has been used as the marker for detecting M2 macrophages. Then we evaluated CD204+ and total macrophages infiltration and its association with clinical outcomes in canine solid tumors. In this study, we examined dogs with oral malignant melanoma (OMM), pulmonary adenocarcinoma (PA), hepatocellular carcinoma (HCC), and transitional cell carcinoma (TCC). Compared to healthy tissues, CD204+ and total macrophages were increased in OMM, PA, and TCC, but not in HCC. High CD204+ macrophage levels were significantly associated with lung metastasis in TCC (P = 0.030). Kaplan-Meier analysis revealed that high CD204+ macrophage levels were associated with shorter overall survival (OS) in canine patients with PA (P = 0.012) and TCC (P = 0.0053). These results suggest that CD204+ macrophages contribute to tumor progression and could be a prognostic factor in dogs with PA and TCC.
Collapse
Affiliation(s)
- S Yokota
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - K Kaji
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - T Yonezawa
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Y Momoi
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - S Maeda
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| |
Collapse
|
13
|
Mehdizadeh R, Shariatpanahi SP, Goliaei B, Rüegg C. Targeting myeloid-derived suppressor cells in combination with tumor cell vaccination predicts anti-tumor immunity and breast cancer dormancy: an in silico experiment. Sci Rep 2023; 13:5875. [PMID: 37041172 PMCID: PMC10090155 DOI: 10.1038/s41598-023-32554-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/29/2023] [Indexed: 04/13/2023] Open
Abstract
Among the different breast cancer subsets, triple-negative breast cancer (TNBC) has the worst prognosis and limited options for targeted therapies. Immunotherapies are emerging as novel treatment opportunities for TNBC. However, the surging immune response elicited by immunotherapies to eradicate cancer cells can select resistant cancer cells, which may result in immune escape and tumor evolution and progression. Alternatively, maintaining the equilibrium phase of the immune response may be advantageous for keeping a long-term immune response in the presence of a small-size residual tumor. Myeloid-derived suppressor cells (MDSCs) are activated, expanded, and recruited to the tumor microenvironment by tumor-derived signals and can shape a pro-tumorigenic micro-environment by suppressing the innate and adaptive anti-tumor immune responses. We recently proposed a model describing immune-mediated breast cancer dormancy instigated by a vaccine consisting of dormant, immunogenic breast cancer cells derived from the murine 4T1 TNBC-like cell line. Strikingly, these 4T1-derived dormant cells recruited fewer MDSCs compared to aggressive 4T1 cells. Recent experimental studies demonstrated that inactivating MDSCs has a profound impact on reconstituting immune surveillance against the tumor. Here, we developed a deterministic mathematical model for simulating MDSCs depletion from mice bearing aggressive 4T1 tumors resulting in immunomodulation. Our computational simulations indicate that a vaccination strategy with a small number of tumor cells in combination with MDSC depletion can elicit an effective immune response suppressing the growth of a subsequent challenge with aggressive tumor cells, resulting in sustained tumor dormancy. The results predict a novel therapeutic opportunity based on the induction of effective anti-tumor immunity and tumor dormancy.
Collapse
Affiliation(s)
- Reza Mehdizadeh
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | | | - Bahram Goliaei
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Curzio Rüegg
- Laboratory of Experimental and Translational Oncology, Pathology, Department of Oncology, Microbiology and Immunology, Faculty of Sciences and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
14
|
Wang HL, Ye ZM, He ZY, Huang L, Liu ZH. m6A-related lncRNA-based immune infiltration characteristic analysis and prognostic model for colonic adenocarcinoma. Hereditas 2023; 160:6. [PMID: 36755298 PMCID: PMC9909974 DOI: 10.1186/s41065-023-00267-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Colonic adenocarcinoma (COAD) is a common gastrointestinal tract tumor, and its occurrence and progression are typically associated with genomic instability, tumor-suppressor gene and oncogene mutations, and tumor mutational load. N6-methyladenosine (m6A) modification of RNAs and long non-coding RNA (lncRNA) expression are important in tumorigenesis and progression. However, the regulatory roles of m6A-associated lncRNAs in the tumor microenvironment, stratification of prognosis, and immunotherapy are unclear. METHODS We screened 43 prognostic lncRNAs linked to m6A and performed consistent molecular typing of COAD using consensus clustering. The single-sample Gene Set Enrichment Analysis and ESTIMATE algorithms were used to assess the immune characteristics of different subgroups. Covariation between methylation-related prognostic lncRNAs was eliminated by least absolute shrinkage and selection operator Cox regression. A nomogram was created and evaluated by combining the methylation-related prognostic lncRNA model with other clinical factors. The relationship between the prognostic model grouping and microsatellite instability, immunophenotype score, and tumor mutation burden was validated using R scripts. Finally, we used a linkage map to filter sensitive medicines to suppress the expression of high-risk genes. Three m6A-associated lncRNA modes were identified in 446 COAD specimens with different clinical endpoints and biological statuses. Risk scores were constructed based on the m6A-associated lncRNA signature genes. Patients with lower risk scores showed superior immunotherapy responses and clinical benefits compared to those with higher risk scores. Lower risk scores were also correlated with higher immunophenotype scores, tumor mutation burden, and mutation rates in significantly mutated genes (e.g., FAT4 and MUC16). Piperidolate, quinostatin, and mecamylamin were screened for their abilities to suppress the expression of high-risk genes in the model. CONCLUSIONS Quantitative assessment of m6A-associated lncRNAs in single tumors can enhance the understanding of tumor microenvironment profiles. The prognostic model constructed using m6A-associated lncRNAs may facilitate prognosis and immunotherapy stratification of patients with COAD; finally, three drugs with potential therapeutic value were screened based on the model.
Collapse
Affiliation(s)
- Hao-lun Wang
- grid.256607.00000 0004 1798 2653Graduate School of Guangxi Medical University, Nanning, 530021 China
| | - Zhuo-miao Ye
- grid.452223.00000 0004 1757 7615Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008 China
| | - Zi-yun He
- grid.256607.00000 0004 1798 2653Graduate School of Guangxi Medical University, Nanning, 530021 China
| | - Lu Huang
- grid.413431.0Day-Care Unit, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, 530021 China
| | - Zhi-hui Liu
- grid.413431.0Day-Care Unit, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, 530021 China
| |
Collapse
|
15
|
Amino Derivatives of Diaryl Pyrimidines and Azolopyrimidines as Protective Agents against LPS-Induced Acute Lung Injury. Molecules 2023; 28:molecules28020741. [PMID: 36677799 PMCID: PMC9863002 DOI: 10.3390/molecules28020741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/30/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
The problem of lung damage originating from excessive inflammation and cytokine release during various types of infections remains relevant and stimulates the search for highly effective and safe drugs. The biological activity of the latter may be associated with the regulation of hyperactivation of certain immune cells and enzymes. Here, we propose the design and synthesis of amino derivatives of 4,6- and 5,7-diaryl substituted pyrimidines and [1,2,4]triazolo[1,5-a]pyrimidines as promising double-acting pharmacophores inhibiting IL-6 and NO. The anti-inflammatory activity of 14 target compounds was studied on isolated primary murine macrophages after LPS stimulation. Seven compounds were identified to inhibit the synthesis of nitric oxide and interleukin 6 at a concentration of 100 µM. The most active compounds are micromolar inhibitors of IL-6 secretion and NO synthesis, showing a minimal impact on innate immunity, unlike the reference drug dexamethasone, along with acceptable cytotoxicity. Evaluation in an animal model of acute lung injury proved the protective activity of compound 6e, which was supported by biochemical, cytological and morphological markers.
Collapse
|
16
|
Nonami A, Matsuo R, Funakoshi K, Nakayama T, Goto S, Iino T, Takaishi S, Mizuno S, Akashi K, Eto M. Prospective study of adoptive activated αβT lymphocyte immunotherapy for refractory cancers: development and validation of a response scoring system. Cytotherapy 2023; 25:76-81. [PMID: 36253253 DOI: 10.1016/j.jcyt.2022.09.007] [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: 03/24/2022] [Revised: 09/03/2022] [Accepted: 09/25/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND AIMS This prospective clinical study aimed to determine the efficacy and prognostic factors of adoptive activated αβT lymphocyte immunotherapy for various refractory cancers. The primary endpoint was overall survival (OS), and the secondary endpoint was radiological response. METHODS The authors treated 96 patients. Activated αβT lymphocytes were infused every 2 weeks for a total of six times. Prognostic factors were identified by analyzing clinical and laboratory data obtained before therapy. RESULTS Median survival time (MST) was 150 days (95% confidence interval, 105-191), and approximately 20% of patients achieved disease control (complete response + partial response + stable disease). According to the multivariate Cox proportional hazards model with Akaike information criterion-best subset selection, sex, concurrent therapy, neutrophil/lymphocyte ratio, albumin, lactate dehydrogenase, CD4:CD8 ratio and T helper (Th)1:Th2 ratio were strong prognostic factors. Using parameter estimates of the Cox analysis, the authors developed a response scoring system. The authors then determined the threshold of the response score between responders and non-responders. This threshold was able to significantly differentiate OS of responders from that of non-responders. MST of responders was longer than that of non-responders (317.5 days versus 74 days). The validity of this response scoring system was then confirmed by internal validation. CONCLUSIONS Adoptive activated αβT lymphocyte immunotherapy has clinical efficacy in certain patients. The authors' scoring system is the first prognostic model reported for this therapy, and it is useful for selecting patients who might obtain a better prognosis through this modality.
Collapse
Affiliation(s)
- Atsushi Nonami
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan; Department of Medicine and Biosystemic Science, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Ryu Matsuo
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kouta Funakoshi
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| | - Tomohiro Nakayama
- Department of Radiology, Saiseikai Fukuoka General Hospital, Fukuoka, Japan
| | - Shigenori Goto
- Department of Next-Generation Cell and Immune Therapy, Juntendo University School of Medicine, Tokyo, Japan
| | - Tadafumi Iino
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Shigeo Takaishi
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan
| | - Shinichi Mizuno
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan; Division of Medical Sciences and Technology, Department of Health Sciences, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Akashi
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan; Department of Medicine and Biosystemic Science, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatoshi Eto
- Center for Advanced Medical Innovation, Kyushu University, Fukuoka, Japan; Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
17
|
PDX Models: A Versatile Tool for Studying the Role of Myeloid-Derived Suppressor Cells in Breast Cancer. Cancers (Basel) 2022; 14:cancers14246153. [PMID: 36551639 PMCID: PMC9777315 DOI: 10.3390/cancers14246153] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
The pivotal role of myeloid-derived suppressive cells (MDSCs) in cancer has become increasingly apparent over the past few years. However, to fully understand how MDSCs can promote human tumor progression and to develop strategies to target this cell type, relevant models that closely resemble the clinical complexity of human tumors are needed. Here, we show that mouse MDSCs of both the monocytic (M-MDCS) and the granulocytic (PMN-MDSC) lineages are recruited to human breast cancer patient-derived xenograft (PDX) tumors in mice. Transcriptomic analysis of FACS-sorted MDSC-subpopulations from the PDX tumors demonstrated the expression of several MDSC genes associated with both their mobilization and immunosuppressive function, including S100A8/9, Ptgs2, Stat3, and Cxcr2, confirming the functional identity of these cells. By combining FACS analysis, RNA sequencing, and immune florescence, we show that the extent and type of MDSC infiltration depend on PDX model intrinsic factors such as the expression of chemokines involved in mobilizing and recruiting tumor-promoting MDSCs. Interestingly, MDSCs have been shown to play a prominent role in breast cancer metastasis, and in this context, we demonstrate increased recruitment of MDSCs in spontaneous PDX lung metastases compared to the corresponding primary PDX tumors. We also demonstrate that T cell-induced inflammation enhances the recruitment of MDSC in experimental breast cancer metastases. In conclusion, breast cancer PDX models represent a versatile tool for studying molecular mechanisms that drive myeloid cell recruitment to primary and metastatic tumors and facilitate the development of innovative therapeutic strategies targeting these cells.
Collapse
|
18
|
Wang M, Wang Y, Liu R, Yu R, Gong T, Zhang Z, Fu Y. TLR4 Blockade Using Docosahexaenoic Acid Restores Vulnerability of Drug-Tolerant Tumor Cells and Prevents Breast Cancer Metastasis and Postsurgical Relapse. ACS BIO & MED CHEM AU 2022; 3:97-113. [PMID: 37101603 PMCID: PMC10125315 DOI: 10.1021/acsbiomedchemau.2c00061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
Nonmutational mechanisms were recently discovered leading to reversible drug tolerance. Despite the rapid elimination of a majority of tumor cells, a small subpopulation of "'drug-tolerant"' cells remain viable with lethal drug exposure, which may further lead to resistance or tumor relapse. Several signaling pathways are involved in the local or systemic inflammatory responses contributing to drug-induced phenotypic switch. Here, we report that Toll-like receptor 4 (TLR4)-interacting lipid docosahexaenoic acid (DHA) restores the cytotoxic effect of doxorubicin (DOX) in the lipopolysaccharide-treated breast tumor cell line 4T1, preventing the phenotypic switch to drug-tolerant cells, which significantly reduces primary tumor growth and lung metastasis in both 4T1 orthotopic and experimental metastasis models. Importantly, DHA in combination with DOX delays and inhibits tumor recurrence following surgical removal of the primary tumor. Furthermore, the coencapsulation of DHA and DOX in a nanoemulsion significantly prolongs the survival of mice in the postsurgical 4T1 tumor relapse model with significantly reduced systemic toxicity. The synergistic antitumor, antimetastasis, and antirecurrence effects of DHA + DOX combination are likely mediated by attenuating TLR4 activation, thus sensitizing tumor cells to standard chemotherapy.
Collapse
Affiliation(s)
- Mou Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Yuejing Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Renhe Liu
- The Scripps Research Institute, 10550 North Torrey Pines Road,
La Jolla, San Diego, California92037, United States
| | - Ruilian Yu
- Department of Oncology, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610072, China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Yao Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| |
Collapse
|
19
|
Lauder SN, Smart K, Bart VMT, Pires A, Scott J, Milutinovic S, Godkin A, Vanhaesebroeck B, Gallimore A. Treg-driven tumour control by PI3Kδ inhibition limits myeloid-derived suppressor cell expansion. Br J Cancer 2022; 127:1595-1602. [PMID: 35986086 PMCID: PMC9596434 DOI: 10.1038/s41416-022-01917-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 06/28/2022] [Accepted: 07/12/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Recent studies have demonstrated that blocking the PI3Kδ signalling enzyme (by administering a small molecule inhibitor, PI-3065) can potently improve the anti-tumour T-cell response through direct inhibition of Tregs. This treatment also has a negative impact on MDSC numbers but the primary mechanism driving this effect has remained unclear. METHODS The 4T1 breast cancer mouse model was used in combination with PI-3065 to gain insights into the effect of PI3Kδ inhibition on MDSCs. RESULTS PI-3065 treatment resulted in a concomitant reduction in MDSC expansion and tumour size. However, targeting Tregs independent of PI-3065 was also associated with reduced tumour volume and MDSC numbers. Surgical removal of tumours resulted in a rapid and significant decline in MDSC numbers, whilst ex vivo studies using cells from PI-3065-treated mice demonstrated no direct effect of the inhibitor on MDSC activity. CONCLUSIONS Our data suggest that MDSCs are not inhibited directly by PI-3065 treatment but that their reduced recruitment and immunosuppression within the tumour microenvironment is an indirect consequence of PI3Kδ-inhibition-driven tumour control. This indicates that PI3Kδ inhibition drives tumour immunity by breaking down multiple immunosuppressive pathways through both direct mechanisms (on Treg) and indirect mechanisms, secondary to tumour control (on MDSCs).
Collapse
Affiliation(s)
- Sarah N Lauder
- Division of Infection and Immunity, Cardiff University School of Medicine, SIURI, Cardiff, CF14 4XN, UK.
| | - Kathryn Smart
- Division of Infection and Immunity, Cardiff University School of Medicine, SIURI, Cardiff, CF14 4XN, UK
| | - Valentina M T Bart
- Division of Infection and Immunity, Cardiff University School of Medicine, SIURI, Cardiff, CF14 4XN, UK
| | - Ana Pires
- Division of Infection and Immunity, Cardiff University School of Medicine, SIURI, Cardiff, CF14 4XN, UK
| | - Jake Scott
- Division of Infection and Immunity, Cardiff University School of Medicine, SIURI, Cardiff, CF14 4XN, UK
| | - Stefan Milutinovic
- Division of Infection and Immunity, Cardiff University School of Medicine, SIURI, Cardiff, CF14 4XN, UK
| | - Andrew Godkin
- Division of Infection and Immunity, Cardiff University School of Medicine, SIURI, Cardiff, CF14 4XN, UK
| | - Bart Vanhaesebroeck
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street, London, WC1E 6BT, UK
| | - Awen Gallimore
- Division of Infection and Immunity, Cardiff University School of Medicine, SIURI, Cardiff, CF14 4XN, UK
| |
Collapse
|
20
|
Pomatto-Watson LCD, Bodogai M, Carpenter M, Chowdhury D, Krishna P, Ng S, Bosompra O, Kato J, Wong S, Reyes-Sepulveda C, Bernier M, Price NL, Biragyn A, de Cabo R. Replenishment of myeloid-derived suppressor cells (MDSCs) overrides CR-mediated protection against tumor growth in a murine model of triple-negative breast cancer. GeroScience 2022; 44:2471-2490. [PMID: 35996062 PMCID: PMC9768076 DOI: 10.1007/s11357-022-00635-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/28/2022] [Indexed: 01/06/2023] Open
Abstract
Caloric restriction (CR) is the leading non-pharmacological intervention to delay induced and spontaneous tumors in pre-clinical models. These effects of CR are largely attributed to canonical inhibition of pro-growth pathways. However, our recent data suggest that CR impairs primary tumor growth and cancer progression in the murine 4T1 model of triple negative breast cancer (TNBC), at least in part, through reduced frequency of the myeloid-derived suppressor cells (MDSC). In the present study, we sought to determine whether injection of excess MDSCs could block regression in 4T1 tumor growth and metastatic spread in BALB/cJ female mice undergoing daily CR. Our findings show that MDSC injection impeded CR-mediated protection against tumor growth without increasing lung metastatic burden. Overall, these results reveal that CR can slow cancer progression by affecting immune suppressive cells.Impact statement: Inoculation of MDSCs from donor mice effectively impedes the ability of calorie restriction to protect against primary tumor growth without impacting lung metastatic burden in recipient animals.
Collapse
Affiliation(s)
- Laura C D Pomatto-Watson
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Monica Bodogai
- Immunoregulation Section, Laboratory of Molecular Biology and Immunology, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Melissa Carpenter
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Dolly Chowdhury
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Priya Krishna
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Sandy Ng
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Oye Bosompra
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Jonathan Kato
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Sarah Wong
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Carlos Reyes-Sepulveda
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Michel Bernier
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Nathan L Price
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Molecular Biology and Immunology, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
21
|
Comprehensive Multiomics Analysis Identified IQGAP3 as a Potential Prognostic Marker in Pan-Cancer. DISEASE MARKERS 2022; 2022:4822964. [PMID: 36164370 PMCID: PMC9508463 DOI: 10.1155/2022/4822964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/01/2022] [Indexed: 12/24/2022]
Abstract
Background IQGAP3 has important function in cancer progression and has become a potential therapeutic target as a transmembrane protein. But its role in tumor immunity and pan-cancer was not systematically investigated. This study evaluated the potential role of IQGAP3 and clinical significance in pan-cancer through combined multiomics analysis. Methods From Genotype Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) databases, transcriptomic datasets were first obtained, and from Gene Expression Omnibus (GEO), expression profiling microarray data were acquired and integrated to systematically assess the expression differences and prognostic relevance of IQGAP3 in pancreatic cancer. Immunohistochemical data were obtained from Human Protein Atlas (HPA) to assess IQGAP3 protein expression differences, and exome data from TCGA were used to analyze IQGAP3 expression in relation to tumor mutational burden (TMB), microsatellite instability (MSI), and mutation. Additionally, we also analyzed the relationship between IQGAP3 expression and immune checkpoints, mismatch repair (MMR), and IQGAP3 relationship with methylation and copy number variation based on expression profiles. Results Microsatellite instability (MSI), immune checkpoints, mismatch repair (MMR), and tumor mutational burden (TMB) all closely interacted with IQGAP3 mRNA. In addition, detailed relationships between the immune microenvironment and IQGAP3 mRNA as well as immune cell CD4+ Th2 and myeloid-derived suppressor cells (MDSCs) were determined. Mechanistically, IQGAP3 was involved in cytoskeleton formation, T cell receptor signaling pathways, DNA damage, cell cycle, P53 pathway, Fc gamma R-mediated phagocytosis, and apoptosis. Conclusion IQGAP3 could serve as an effective prognostic biomarker for pan-cancer immune-related therapy.
Collapse
|
22
|
Baterdene O, Miura K, Ueno W, Watanabe S, Tsukui M, Nomoto H, Goka R, Maeda H, Yamamoto H, Morimoto N. A successful case of transarterial chemoembolization for hyperprogressive disease induced by immunotherapy in a patient with unresectable hepatocellular carcinoma. Clin J Gastroenterol 2022; 15:1101-1107. [DOI: 10.1007/s12328-022-01697-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022]
|
23
|
Liu T, Gonzalez De Los Santos F, Rinke AE, Fang C, Flaherty KR, Phan SH. B7H3-dependent myeloid-derived suppressor cell recruitment and activation in pulmonary fibrosis. Front Immunol 2022; 13:901349. [PMID: 36045668 PMCID: PMC9420866 DOI: 10.3389/fimmu.2022.901349] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/22/2022] [Indexed: 11/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease without effective curative therapy. Recent evidence shows increased circulating myeloid-derived suppressor cells (MDSCs) in cancer, inflammation, and fibrosis, with some of these cells expressing B7H3. We sought to investigate the role of MDSCs in IPF and its potential mediation via B7H3. Here we prospectively collected peripheral blood samples from IPF patients to analyze for circulating MDSCs and B7H3 expression to assess their clinical significance and potential impact on co-cultured lung fibroblasts and T-cell activation. In parallel, we assess MDSC recruitment and potential B7H3 dependence in a mouse model of pulmonary fibrosis. Expansion of MDSCs in IPF patients correlated with disease severity. Co-culture of soluble B7H3 (sB7H3)-treated mouse monocytic MDSCs (M-MDSCs), but not granulocytic MDSCs (G-MDSCs), activated lung fibroblasts and myofibroblast differentiation. Additionally, sB7H3 significantly enhanced MDSC suppression of T-cell proliferation. Activated M-MDSCs displayed elevated TGFβ and Arg1 expression relative to that in G-MDSCs. Treatment with anti-B7H3 antibodies inhibited bone marrow-derived MDSC recruitment into the bleomycin-injured lung, accompanied by reduced expression of inflammation and fibrosis markers. Selective telomerase reverse transcriptase (TERT) deficiency in myeloid cells also diminished MDSC recruitment associated with the reduced plasma level of sB7H3, lung recruitment of c-Kit+ hematopoietic progenitors, myofibroblast differentiation, and fibrosis. Lung single-cell RNA sequencing (scRNA-seq) revealed fibroblasts as a predominant potential source of sB7H3, and indeed the conditioned medium from activated mouse lung fibroblasts had a chemotactic effect on bone marrow (BM)-MDSC, which was abolished by B7H3 blocking antibody. Thus, in addition to their immunosuppressive activity, TERT and B7H3-dependent MDSC expansion/recruitment from BM could play a paracrine role to activate myofibroblast differentiation during pulmonary fibrosis with potential significance for disease progression mediated by sB7H3.
Collapse
Affiliation(s)
- Tianju Liu
- Departments of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
- *Correspondence: Sem H. Phan, ; Tianju Liu,
| | | | - Andrew E. Rinke
- Departments of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Chuling Fang
- Departments of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Kevin R. Flaherty
- Division of Pulmonary/Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Sem H. Phan
- Departments of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
- *Correspondence: Sem H. Phan, ; Tianju Liu,
| |
Collapse
|
24
|
Kadiyala P, Elhossiny AM, Carpenter ES. Using Single Cell Transcriptomics to Elucidate the Myeloid Compartment in Pancreatic Cancer. Front Oncol 2022; 12:881871. [PMID: 35664793 PMCID: PMC9161632 DOI: 10.3389/fonc.2022.881871] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/08/2022] [Indexed: 11/25/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a dismal disease with a 5-year survival rate of 10%. A hallmark feature of this disease is its abundant microenvironment which creates a highly immunosuppressive milieu. This is, in large part, mediated by an abundant infiltration of myeloid cells in the PDAC tumor microenvironment. Consequently, therapies that modulate myeloid function may augment the efficacy of standard of care for PDAC. Unfortunately, there is limited understanding about the various subsets of myeloid cells in PDAC, particularly in human studies. This review highlights the application of single-cell RNA sequencing to define the myeloid compartment in human PDAC and elucidate the crosstalk between myeloid cells and the other components of the tumor immune microenvironment.
Collapse
Affiliation(s)
- Padma Kadiyala
- Department of Immunology, University of Michigan, Ann Arbor, MI, United States
| | - Ahmed M. Elhossiny
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
| | - Eileen S. Carpenter
- Department of Intenal Medicine, Division of Gastroenterology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Eileen S. Carpenter,
| |
Collapse
|
25
|
Tengesdal IW, Li S, Powers NE, May M, Neff CP, Joosten LAB, Marchetti C, Dinarello CA. Activation of Host-NLRP3 Inflammasome in Myeloid Cells Dictates Response to Anti-PD-1 Therapy in Metastatic Breast Cancers. Pharmaceuticals (Basel) 2022; 15:574. [PMID: 35631400 PMCID: PMC9144656 DOI: 10.3390/ph15050574] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/22/2022] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
Tumor-associated inflammation leads to dysregulated cytokine production that promotes tumor immune evasion and anti-tumor immunity dysfunction. In advanced stage breast cancer, the proinflammatory cytokine IL-1β is overexpressed due to large proportions of activated myeloid cells in the tumor microenvironment (TME). Here, we demonstrate the role of the host nucleotide-binding domain, leucine-rich containing family, pyrin domain-containing 3 (NLRP3) inflammasome in metastatic breast cancer. In vitro, we show that stimulation of THP-1 cells with conditioned media collected from MDA-MB-468 cells induced NLRP3 activation and increased Pdcd1l1 expression. In vivo, mice deficient in NLRP3 orthotopically implanted with metastatic breast cancer cell line (E0771) showed significant reduction in tumor growth (p < 0.05) and increased survival (p < 0.01). Inhibition of NLRP3 with the small molecule OLT1177® reduced expression of Pdcd1l1 (p < 0.001), Casp1 (p < 0.01) and Il1b (p < 0.01) in primary tumors. Furthermore, tumor-bearing mice receiving OLT1177® showed reduced infiltration of myeloid-derived suppressor cells (MDSCs) (p < 0.001) and increased CD8+ T cells (p < 0.05) and NK cells (p < 0.05) in the TME. NLRP3 inhibition in addition to anti-PD-1 treatment significantly reduced tumor growth from the monotherapies (p < 0.05). These data define NLRP3 activation as a key driver of immune suppression in metastatic breast cancers. Furthermore, this study suggests NLRP3 as a valid target to increase efficacy of immunotherapy with checkpoint inhibitor in metastatic breast cancers.
Collapse
Affiliation(s)
- Isak W. Tengesdal
- Department of Medicine, Radboud University Medical Center, 6525 Nijmegen, The Netherlands; (I.W.T.); (L.A.B.J.); (C.A.D.)
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (S.L.); (N.E.P.); (M.M.); (C.P.N.)
| | - Suzhao Li
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (S.L.); (N.E.P.); (M.M.); (C.P.N.)
| | - Nicholas E. Powers
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (S.L.); (N.E.P.); (M.M.); (C.P.N.)
| | - Makenna May
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (S.L.); (N.E.P.); (M.M.); (C.P.N.)
| | - Charles P. Neff
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (S.L.); (N.E.P.); (M.M.); (C.P.N.)
| | - Leo A. B. Joosten
- Department of Medicine, Radboud University Medical Center, 6525 Nijmegen, The Netherlands; (I.W.T.); (L.A.B.J.); (C.A.D.)
| | - Carlo Marchetti
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (S.L.); (N.E.P.); (M.M.); (C.P.N.)
| | - Charles A. Dinarello
- Department of Medicine, Radboud University Medical Center, 6525 Nijmegen, The Netherlands; (I.W.T.); (L.A.B.J.); (C.A.D.)
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (S.L.); (N.E.P.); (M.M.); (C.P.N.)
| |
Collapse
|
26
|
Tan Q, Yin S, Zhou D, Chi Y, Man X, Li H. Potential Predictive and Prognostic Value of Biomarkers Related to Immune Checkpoint Inhibitor Therapy of Triple-Negative Breast Cancer. Front Oncol 2022; 12:779786. [PMID: 35646659 PMCID: PMC9134495 DOI: 10.3389/fonc.2022.779786] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
As an aggressive subtype of breast cancer, triple-negative breast cancer (TNBC) is associated with poor prognosis and lack of effective therapy, except chemotherapy. In recent years, immunotherapy based on immune checkpoint (IC) inhibition has emerged as a promising therapeutic strategy in TNBC. TNBC has more tumor-infiltrating lymphocytes (TILs) and higher rate of mutation and programmed cell death ligand-1 (PD-L1) expression than other subtypes of breast cancer have. However, previous studies have shown that monotherapy has little efficacy and only some TNBC patients can benefit from immunotherapy. Therefore, it is important to identify biomarkers that can predict the efficacy of IC inhibitors (ICIs) in TNBC. Recently, various biomarkers have been extensively explored, such as PD-L1, TILs and tumor mutational burden (TMB). Clinical trials have shown that PD-L1-positive patients with advanced TNBC benefit from ICIs plus chemotherapy. However, in patients with early TNBC receiving neoadjuvant therapy, PD-L1 cannot predict the efficacy of ICIs. These inconsistent conclusions suggest that PD-L1 is the best to date but an imperfect predictive biomarker for efficacy of ICIs. Other studies have shown that advanced TNBC patients with TMB ≥10 mutations/Mb can achieve clinical benefits from pembrolizumab. TILs also have potential predictive value in TNBC. Here, we select some biomarkers related to ICIs and discuss their potential predictive and prognostic value in TNBC. We hope these biomarkers could help to identify suitable patients and realize precision immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Huihui Li
- Department of Breast Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
27
|
Murad LD, Silva TDQ, Schilithz AOC, Fernandes PV, Monteiro M, Murad LB, Fialho E. Low body mass index is associated with reduced intratumoral CD4+ T-lymphocyte infiltration in laryngeal squamous cell carcinoma patients. Nutr Res 2022; 102:1-12. [DOI: 10.1016/j.nutres.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 10/19/2022]
|
28
|
Li L, Ai L, Jia L, Zhang L, Lei B, Zhang Q. High score of LDH plus dNLR predicts poor survival in patients with HER2-positive advanced breast cancer treated with trastuzumab emtansine. BMC Cancer 2022; 22:29. [PMID: 34980025 PMCID: PMC8722106 DOI: 10.1186/s12885-021-09131-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 12/20/2021] [Indexed: 12/02/2022] Open
Abstract
Objective To investigate the prognostic value of derived neutrophil to lymphocyte ratio (dNLR) and lactate dehydrogenase (LDH) in patients with advanced HER2 positive breast cancer treated with trastuzumab emtansine. Methods Fifty one patients with advanced HER2 positive breast cancer who received T-DM1 treatment in Harbin Medical University Cancer Hospital were selected. The clinical data and blood test indexes were collected, and the ROC curve determined the optimal cut-off value. Kaplan-Meier survival curve and Cox regression model was used to analyze the effect of different levels of dNLR,LDH,LNI (dNLR combined with LDH index) before and after T-DM1 treatment on the survival of patients. Results The median PFS and OS of the patients with advanced HER2 positive breast cancer who received T-DM1 treatment were 6.9 months and 22.2 months, respectively. The optimal cut-off value of LDH and dNLR before T-DM1 treatment was 244 U / L (P = 0.003) and 1.985 (P = 0.013), respectively. Higher LDH and dNLR were significantly correlated with shorter median PFS and OS (P < 0.05). The median PFS of patients with LNI (0), LNI (1) and LNI (2) were 8.1 months, 5.5 months and 2.3 months, respectively, P = 0.007. Univariate and multivariate analysis showed that LDH > 244 U / L, dNLR > 1.985, LNI > 0, ECOG ≥1 and HER-2 (IHC2 +, FISH+) before the T-DM1 treatment were the poor prognostic factors. LDH uptrend after the T-DM1 treatment also predicted poor prognosis. Conclusion Serum LDH > 244 U / L and dNLR > 1.985 before the T-DM1 treatment were prognostic risk factors for patients with advanced HER2 positive breast cancer receiving T-DM1 treatment. The higher LNI score was significantly associated with shorter PFS and OS. LDH uptrend after T-DM1 treatment was also related to the poor prognosis.
Collapse
Affiliation(s)
- Liru Li
- Internal Medicine-Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Lin Ai
- Internal Medicine-Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Lin Jia
- Internal Medicine-Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Lei Zhang
- Internal Medicine-Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Boya Lei
- Internal Medicine-Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Qingyuan Zhang
- Internal Medicine-Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
29
|
He N, Jiang J. Contribution of immune cells to bone metastasis pathogenesis. Front Endocrinol (Lausanne) 2022; 13:1019864. [PMID: 36246916 PMCID: PMC9556850 DOI: 10.3389/fendo.2022.1019864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Bone metastasis is closely related to the survival rate of cancer patients and reduces their quality of life. The bone marrow microenvironment contains a complex immune cell component with a local microenvironment that is conducive to tumor formation and growth. In this unique immune environment, a variety of immune cells, including T cells, natural killer cells, macrophages, dendritic cells, and myeloid-derived suppressor cells, participate in the process of bone metastasis. In this review, we will introduce the interactions between immune cells and cancer cells in the bone microenvironment, obtain the details of their contributions to the implications of bone metastasis, and discuss immunotherapeutic strategies targeting immune cells in cancer patients with bone metastasis.
Collapse
Affiliation(s)
- Ningning He
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Oncology, Yangzhou University, Yangzhou, China
- Department of Oncology, First People’s Hospital of Changzhou, Changzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Oncology, First People’s Hospital of Changzhou, Changzhou, China
- *Correspondence: Jingting Jiang,
| |
Collapse
|
30
|
Beyond immune checkpoint blockade: emerging immunological strategies. Nat Rev Drug Discov 2021; 20:899-919. [PMID: 33686237 DOI: 10.1038/s41573-021-00155-y] [Citation(s) in RCA: 214] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2021] [Indexed: 02/07/2023]
Abstract
The success of checkpoint inhibitors has accelerated the clinical implementation of a vast mosaic of single agents and combination immunotherapies. However, the lack of clinical translation for a number of immunotherapies as monotherapies or in combination with checkpoint inhibitors has clarified that new strategies must be employed to advance the field. The next chapter of immunotherapy should examine the immuno-oncology therapeutic failures, and consider the complexity of immune cell-cancer cell interactions to better design more effective anticancer drugs. Herein, we briefly review the history of immunotherapy and checkpoint blockade, highlighting important clinical failures. We discuss the critical aspects - beyond T cell co-receptors - of immune processes within the tumour microenvironment (TME) that may serve as avenues along which new therapeutic strategies in immuno-oncology can be forged. Emerging insights into tumour biology suggest that successful future therapeutics will focus on two key factors: rescuing T cell homing and dysfunction in the TME, and reappropriating mononuclear phagocyte function for TME inflammatory remodelling. New drugs will need to consider the complex cell networks that exist within tumours and among cancer types.
Collapse
|
31
|
Welte T, Mai J, Zhang Z, Tian S, Zhang G, Xu Y, Zhang L, Chen SS, Wang T, Shen H. A heparan-sulfate-bearing syndecan-1 glycoform is a distinct surface marker for intra-tumoral myeloid-derived suppressor cells. iScience 2021; 24:103349. [PMID: 34825135 PMCID: PMC8603209 DOI: 10.1016/j.isci.2021.103349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/17/2021] [Accepted: 10/22/2021] [Indexed: 11/19/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) infiltrate cancer tissue, promote tumor growth, and are associated with resistance to cancer therapies. However, there is no practical approach available to distinguish MDSCs from mature counterparts inside tumors. Here, we show that a recently isolated thioaptamer probe (T1) binds to MDSC subsets in colorectal and pancreatic tumors with high specificity. Whole transcriptome and functional analysis revealed that T1-binding cells contain polymorphonuclear (PMN)-MDSCs characterized by several immunosuppression pathways, ROS production, and T cell suppression activity, whereas T1-non-binding PMNs were mature and nonsuppressive. We identified syndecan-1 as the T1-interacting protein on MDSCs and chronic myelogenous leukemia K562 cell line. Heparan sulfate chains were essential in T1-binding. Inside tumors PMN-MDSCs expressed heparan sulfate biogenesis enzymes at higher levels. Tumor-cell-derived soluble factor(s) enhanced MDSCs' affinity for T1. Overall, we uncovered heparan-sulfate-dependent MDSC modulation in the tumor microenvironment and identified T1 as tool preferentially targeting tumor-promoting myeloid cell subsets.
Collapse
Affiliation(s)
- Thomas Welte
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Zhe Zhang
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Shaohui Tian
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Guodong Zhang
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Yitian Xu
- Center for Immunotherapy Research, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Licheng Zhang
- Center for Immunotherapy Research, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Shu-shia Chen
- Center for Immunotherapy Research, Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Tian Wang
- Department of Microbiology & Immunology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX 77030, USA
- Innovative Therapeutic Program, Houston Methodist Cancer Center, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
32
|
Hao Z, Li R, Wang Y, Li S, Hong Z, Han Z. Landscape of Myeloid-derived Suppressor Cell in Tumor Immunotherapy. Biomark Res 2021; 9:77. [PMID: 34689842 PMCID: PMC8543853 DOI: 10.1186/s40364-021-00333-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/26/2021] [Indexed: 02/08/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are a group of immature cells that produced by emergency myelopoiesis. Emerging evidences have identified the vital role of MDSC in cancer microenvironment, in which MDSC exerts both immunological and non-immunological activities to assist the progression of cancer. Advances in pre-clinical research have provided us the understanding of MDSC in cancer context from the perspective of molecular mechanism. In clinical scenario, MDSC and its subsets have been discovered to exist in peripheral blood and tumor site of patients from various types of cancers. In this review, we highlight the clinical value of MDSC in predicting prognosis of cancer patients and the responses of immunotherapies, therefore to propose the MDSC-inhibiting strategy in the scenario of cancer immunotherapies. Phenotypes and biological functions of MDSC in cancer microenvironment are comprehensively summarized to provide potential targets of MDSC-inhibiting strategy from the aspect of molecular mechanisms.
Collapse
Affiliation(s)
- Zhaonian Hao
- Department of Neurosurgery, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Ruyuan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.,Department of Gynecology and Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuanyuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Shuangying Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhenya Hong
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Zhiqiang Han
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
33
|
Hattori M, Novick D, Takaura K, Tanizawa Y, Kawaguchi T, Haro JM, Monistrol-Mula A, Onishi A, Iwata H. A systematic literature review of prognostic factors in patients with HR+/HER2- advanced breast cancer in Japan. Jpn J Clin Oncol 2021; 51:1498-1508. [PMID: 34417808 PMCID: PMC8491537 DOI: 10.1093/jjco/hyab131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/28/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Breast cancer is the most prevalent cancer in women in Japan and the fifth in mortality. This systematic review summarized the evidence for prognostic factors for patients with HR+/HER2- advanced and metastatic breast cancer in Japan. METHODS MEDLINE and EMBASE were searched with keywords 'breast neoplasms' AND 'Japan' AND 'advanced' or equivalent, and Japan Medical Abstract Society database with 'breast cancer' AND 'advanced/metastatic' for publications from January 2010 to October 2019. ASCO, ESMO, ABC4 abstracts and WHO website were hand searched. The endpoints of interest were overall survival, progression-free survival, tumour response and post-progression survival. Factors were evaluated based on the consistency in direction and the strength (hazard ratios) of association. RESULTS Searches identified 4530 publications, of which 27 were eligible. All were observational studies. Among the endpoints, overall survival was the most commonly assessed (n = 22) and evaluated further. Ki-67 expression, progesterone receptor expression status, tumour grade and lymph node metastases were consistently associated with poor overall survival in univariate analysis but not in multivariate analysis. Short disease-free interval, the number of metastatic organs and liver metastasis were consistently associated with poor overall survival in both of univariate and multivariate analysis. The association was strong for liver metastasis (hazard ratio ≥2.8 in the majority of studies) and moderate for disease-free interval and the number of metastatic organs (hazard ratio 1.3-2.8 in the majority of studies). CONCLUSIONS Disease-free interval, the number of metastatic organs and liver metastasis were identified as independent prognostic factors for overall survival. These findings may help clinical decision-making to improve outcomes in patients with HR+/HER2- advanced and metastatic breast cancer.
Collapse
Affiliation(s)
- Masaya Hattori
- Department of Breast Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Diego Novick
- For reprints and all correspondence: Diego Novick, Eli Lilly and Company, Windlesham, UK. E-mail:
| | | | | | | | - Josep Maria Haro
- Group of Epidemiology of Mental Disorders and Ageing, Institut de Recerca Sant Joan de Déu (IRSJD), CIBERSAM, Universitat de Barcelona, Barcelona, Spain
| | - Anna Monistrol-Mula
- Group of Epidemiology of Mental Disorders and Ageing, Institut de Recerca Sant Joan de Déu (IRSJD), CIBERSAM, Universitat de Barcelona, Barcelona, Spain
| | - Akira Onishi
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center, Nagoya, Japan
| |
Collapse
|
34
|
Zhao N, Zhu W, Wang J, Liu W, Kang L, Yu R, Liu B. Group 2 innate lymphoid cells promote TNBC lung metastasis via the IL-13-MDSC axis in a murine tumor model. Int Immunopharmacol 2021; 99:107924. [PMID: 34217145 DOI: 10.1016/j.intimp.2021.107924] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 11/23/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) are reportedly associated with the progression of many tumors. However, the role of ILC2s in triple-negative breast cancer (TNBC) lung metastasis remains unclear. In this study, we found that ILC2s may be a key element in the process of TNBC lung metastasis since the adoptive transfer of pulmonary ILC2s increased the numbers of metastatic lung nodules and reduced the survival of tumor-bearing mice. ILC2-promoted 4 T1 lung metastasis appears to be related to ILC2-derived IL-13. An expansion of IL-13-producing ILC2s and an elevated expression of IL-13 mRNA in pulmonary ILC2s were determined in tumor-bearing mice, in parallel with an increase in the levels of local IL-13 by ILC2 transfer. The neutralization of IL-13 reduced the increased pulmonary metastatic nodules and improved the decreased survival rate caused by ILC2-adoptive transfer. Interestingly, adoptive transfer of ILC2s elevated IL-13Ra1 expression in myeloid-derived suppressor cells (MDSCs). Treatment of ILC2-transferred tumor-bearing mice with anti-IL-13 antibodies significantly diminished the number of pulmonary MDSCs and inhibited MDSC activation. Moreover, when pulmonary MDSCs were cocultured with ILC2s in the presence of an anti-IL-13 mAb, the number and activation of MDSCs were reduced. Depletion of MDSCs may promote the proliferation of CD4+ T cells and CD8+ T cells, but reduce the expansion of regulatory T cells (Tregs) in the lungs of ILC2-transferred tumor-bearing mice. Our results suggest that pulmonary ILC2s may promote TNBC lung metastasis via the ILC2-derived IL-13-activated MDSC pathway.
Collapse
Affiliation(s)
- Na Zhao
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang 110001, China; Department of Medical Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Wenwen Zhu
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang 110001, China
| | - Jia Wang
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang 110001, China
| | - Weiwei Liu
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang 110001, China
| | - Longdan Kang
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang 110001, China
| | - Rui Yu
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang 110001, China
| | - Beixing Liu
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang 110001, China.
| |
Collapse
|
35
|
DeNardo DG, Galkin A, Dupont J, Zhou L, Bendell J. GB1275, a first-in-class CD11b modulator: rationale for immunotherapeutic combinations in solid tumors. J Immunother Cancer 2021; 9:jitc-2021-003005. [PMID: 34452928 PMCID: PMC8404448 DOI: 10.1136/jitc-2021-003005] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 12/20/2022] Open
Abstract
Resistance to immune checkpoint inhibitors (ICI) and other anticancer therapies is often associated with the accumulation of myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) in the tumor microenvironment (TME). Therefore, targeting MDSC recruitment or function is of significant interest as a strategy to treat patients with ICI-resistant cancer. The migration and recruitment of MDSCs to the TME is mediated in part by the CD11b/CD18 integrin heterodimer (Mac-1; αMβ2), expressed on both MDSCs and TAMs. However, inhibition or blockade of CD11b/CD18 has had limited success in clinical trials to date, likely since saturation of CD11b requires doses that are not clinically tolerable with the agents tested so far. Interestingly, activation of CD11b with leukadherin-1 was found to reduce macrophage and neutrophil migration in animal models of inflammatory conditions. Preclinical studies with GB1275, a salt form of leukadherin-1, demonstrated that activation of CD11b improves the antitumor immune response and enhances the response to immunotherapy in mouse models of pancreatic adenocarcinoma, breast cancer and lung cancer. Based on the promising results from preclinical studies, a phase 1/2 clinical study (NCT04060342) of GB1275 in patients with advanced solid tumor types known to be resistant or less likely responsive to immuno-oncology therapies, including pancreatic, breast, prostate, and microsatellite-stable colorectal cancer, is ongoing. In this review, we examine targeting MDSCs as a therapeutic approach in cancer therapy, with a special focus on GB1275 preclinical studies laying the rationale for the phase 1/2 clinical study.
Collapse
Affiliation(s)
- David G DeNardo
- Department of Medicine, ICCE Institute, Department of Pathology and Immunology, Siteman Cancer Center, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | | | | | - Lei Zhou
- Gossamer Bio, San Diego, California, USA
| | - Johanna Bendell
- Sarah Cannon Research Institute, Tennessee Oncology, Nashville, Tennessee, USA
| |
Collapse
|
36
|
Gonda K, Horita S, Maejima Y, Takenoshita S, Shimomura K. Soluble interleukin-2 receptor as a predictive and prognostic marker for patients with familial breast cancer. Sci Prog 2021; 104:368504211039590. [PMID: 34559590 PMCID: PMC10461470 DOI: 10.1177/00368504211039590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The incidence of breast cancer increases annually, and it has become common within families of breast cancer patients. Interleukin-2 activates cytotoxic T lymphocytes, which are important for cancer immunity. To identify markers of increased familial breast cancer risk, soluble interleukin-2 receptor levels and immunologic factors were investigated in familial breast cancer and non-familial breast cancer patients. Of 106 untreated breast cancer patients in this study, 24 had familial breast cancer and 82 had non-familial breast cancer. The patients' soluble interleukin-2 receptor, interleukin-10, vascular endothelial growth factor, interleukin-17, regulatory T cell, myeloid-derived suppressor cell, white blood cell, and C-reactive protein levels, and their neutrophil-to-lymphocyte ratios were measured, and their prognoses were compared according to the soluble interleukin-2 receptor levels. Additionally, postoperative tissues from the patients with high soluble interleukin-2 receptor levels were stained with programmed cell death ligand 1 and cluster of differentiation 8. The soluble interleukin-2 receptor level in the familial breast cancer patients was significantly higher, and it showed significantly stronger correlations with the neutrophil-to-lymphocyte ratio and the interleukin-10, vascular endothelial growth factor, interleukin-17, regulatory T cell, myeloid-derived suppressor cell, white blood cell, and C-reactive protein levels, than in the non-familial breast cancer patients. The regulatory T cell and myeloid-derived suppressor cell levels were significantly higher in the patients with high soluble interleukin-2 receptor levels, and the overall survival and disease-free-survival rates were significantly worse for the familial breast cancer patients than for the non-familial breast cancer patients. Triple-negative breast cancer tissues from the familial breast cancer patients with high soluble interleukin-2 receptor levels stained well for programmed cell death ligand 1 and cluster of differentiation 8. Soluble interleukin-2 receptor levels can be used to predict the prognosis of familial breast cancer patients. Prospectively identifying patients who are less likely to have non-familial breast cancer is vital for improving their overall survival.
Collapse
Affiliation(s)
- Kenji Gonda
- Department of Medicine, Daido Obesity
and Metabolism Research Center, Japan
- Department of Medicine, Daido Central
Hospital, Japan
- Department of Bioregulation and
Pharmacological Medicine, Fukushima Medical
University, Japan
- Department of Surgery, Fukushima Medical
University, Japan
| | - Shoichiro Horita
- Department of Bioregulation and
Pharmacological Medicine, Fukushima Medical
University, Japan
| | - Yuko Maejima
- Department of Bioregulation and
Pharmacological Medicine, Fukushima Medical
University, Japan
| | | | - Kenju Shimomura
- Department of Bioregulation and
Pharmacological Medicine, Fukushima Medical
University, Japan
| |
Collapse
|
37
|
Baram T, Erlichman N, Dadiani M, Balint-Lahat N, Pavlovski A, Meshel T, Morzaev-Sulzbach D, Gal-Yam EN, Barshack I, Ben-Baruch A. Chemotherapy Shifts the Balance in Favor of CD8+ TNFR2+ TILs in Triple-Negative Breast Tumors. Cells 2021; 10:cells10061429. [PMID: 34201054 PMCID: PMC8229590 DOI: 10.3390/cells10061429] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is primarily treated via chemotherapy; in parallel, efforts are made to introduce immunotherapies into TNBC treatment. CD4+ TNFR2+ lymphocytes were reported as Tregs that contribute to tumor progression. However, our published study indicated that TNFR2+ tumor-infiltrating lymphocytes (TNFR2+ TILs) were associated with improved survival in TNBC patient tumors. Based on our analyses of the contents of CD4+ and CD8+ TILs in TNBC patient tumors, in the current study, we determined the impact of chemotherapy on CD4+ and CD8+ TIL subsets in TNBC mouse tumors. We found that chemotherapy led to (1) a reduction in CD4+ TNFR2+ FOXP3+ TILs, indicating that chemotherapy decreased the content of CD4+ TNFR2+ Tregs, and (2) an elevation in CD8+ TNFR2+ and CD8+ TNFR2+ PD-1+ TILs; high levels of these two subsets were significantly associated with reduced tumor growth. In spleens of tumor-bearing mice, chemotherapy down-regulated CD4+ TNFR2+ FOXP3+ cells but the subset of CD8+ TNFR2+ PD-1+ was not present prior to chemotherapy and was not increased by the treatment. Thus, our data suggest that chemotherapy promotes the proportion of protective CD8+ TNFR2+ TILs and that, unlike other cancer types, therapeutic strategies directed against TNFR2 may be detrimental in TNBC.
Collapse
Affiliation(s)
- Tamir Baram
- George S. Wise Faculty of Life Sciences, The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv 69978-01, Israel; (T.B.); (N.E.); (T.M.)
| | - Nofar Erlichman
- George S. Wise Faculty of Life Sciences, The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv 69978-01, Israel; (T.B.); (N.E.); (T.M.)
| | - Maya Dadiani
- Sheba Medical Center, Breast Oncology Institute, Ramat Gan 5211401, Israel; (M.D.); (D.M.-S.); (E.N.G.-Y.)
| | - Nora Balint-Lahat
- Sheba Medical Center, Pathology Institute, Ramat Gan 5211401, Israel; (N.B.-L.); (A.P.); (I.B.)
| | - Anya Pavlovski
- Sheba Medical Center, Pathology Institute, Ramat Gan 5211401, Israel; (N.B.-L.); (A.P.); (I.B.)
| | - Tsipi Meshel
- George S. Wise Faculty of Life Sciences, The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv 69978-01, Israel; (T.B.); (N.E.); (T.M.)
| | - Dana Morzaev-Sulzbach
- Sheba Medical Center, Breast Oncology Institute, Ramat Gan 5211401, Israel; (M.D.); (D.M.-S.); (E.N.G.-Y.)
| | - Einav Nili Gal-Yam
- Sheba Medical Center, Breast Oncology Institute, Ramat Gan 5211401, Israel; (M.D.); (D.M.-S.); (E.N.G.-Y.)
| | - Iris Barshack
- Sheba Medical Center, Pathology Institute, Ramat Gan 5211401, Israel; (N.B.-L.); (A.P.); (I.B.)
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978-01, Israel
| | - Adit Ben-Baruch
- George S. Wise Faculty of Life Sciences, The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv 69978-01, Israel; (T.B.); (N.E.); (T.M.)
- Correspondence: ; Tel.: +972-3-6407933 or +972-3-6405491; Fax: +972-3-6422046
| |
Collapse
|
38
|
Effects of Chemotherapy Agents on Circulating Leukocyte Populations: Potential Implications for the Success of CAR-T Cell Therapies. Cancers (Basel) 2021; 13:cancers13092225. [PMID: 34066414 PMCID: PMC8124952 DOI: 10.3390/cancers13092225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/25/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary CAR-T cell therapy is a new approach to cancer treatment that is based on manipulating a patient’s own T cells such that they become able to seek and destroy cancer cells in a highly specific manner. This approach is showing remarkable efficacy in treating some types of blood cancers but so far has been much less effective against solid cancers. Here, we review the diverse effects of chemotherapy agents on circulating leukocyte populations and find that, despite some negative effects over the short term, chemotherapy can favourably modulate the immune systems of cancer patients over the longer term. Since blood is the starting material for CAR-T cell production, we propose that these effects could significantly influence the success of manufacturing, and anti-cancer activity, of CAR-T cells. Thus, if timed correctly, chemotherapy-induced changes to circulating immune cells could allow CAR-T cells to unleash more effective anti-tumour responses. Abstract Adoptive T-cell therapy using autologous T cells genetically modified to express cancer-specific chimeric antigen receptors (CAR) has emerged as a novel approach for cancer treatment. CAR-T cell therapy has been approved in several major jurisdictions for treating refractory or relapsed cases of B-cell precursor acute lymphoblastic leukaemia and diffuse large B-cell lymphoma. However, in solid cancer patients, several clinical studies of CAR-T cell therapy have demonstrated minimal therapeutic effects, thus encouraging interest in better integrating CAR-T cells with other treatments such as conventional cytotoxic chemotherapy. Increasing evidence shows that not only do chemotherapy drugs have tumoricidal effects, but also significantly modulate the immune system. Here, we discuss immunomodulatory effects of chemotherapy drugs on circulating leukocyte populations, including their ability to enhance cytotoxic effects and preserve the frequency of CD8+ T cells and to deplete immunosuppressive populations including regulatory T cells and myeloid-derived suppressor cells. By modulating the abundance and phenotype of leukocytes in the blood (the ‘raw material’ for CAR-T cell manufacturing), we propose that prior chemotherapy could facilitate production of the most effective CAR-T cell products. Further research is required to directly test this concept and identify strategies for the optimal integration of CAR-T cell therapies with cytotoxic chemotherapy for solid cancers.
Collapse
|
39
|
Mandula JK, Rodriguez PC. Tumor-related stress regulates functional plasticity of MDSCs. Cell Immunol 2021; 363:104312. [PMID: 33652258 PMCID: PMC8026602 DOI: 10.1016/j.cellimm.2021.104312] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/15/2021] [Accepted: 01/29/2021] [Indexed: 12/15/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) impair protective anti-tumor immunity and remain major obstacles that stymie the effectiveness of promising cancer therapies. Diverse tumor-derived stressors galvanize the differentiation, intra-tumoral expansion, and immunomodulatory function of MDSCs. These tumor-associated 'axes of stress' underwrite the immunosuppressive programming of MDSCs in cancer and contribute to the phenotypic/functional heterogeneity that characterize tumor-MDSCs. This review discusses various tumor-associated axes of stress that direct MDSC development, accumulation, and immunosuppressive function, as well as current strategies aimed at overcoming the detrimental impact of MDSCs in cancer. To better understand the constellation of signals directing MDSC biology, we herein summarize the pivotal roles, signaling mediators, and effects of reactive oxygen/nitrogen species-related stress, chronic inflammatory stress, hypoxia-linked stress, endoplasmic reticulum stress, metabolic stress, and therapy-associated stress on MDSCs. Although therapeutic targeting of these processes remains mostly pre-clinical, intercepting signaling through the axes of stress could overcome MDSC-related immune suppression in tumor-bearing hosts.
Collapse
Affiliation(s)
- Jessica K Mandula
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
40
|
The Pan-Immune-Inflammation-Value Predicts the Survival of Patients with Human Epidermal Growth Factor Receptor 2 (HER2)-Positive Advanced Breast Cancer Treated with First-Line Taxane-Trastuzumab-Pertuzumab. Cancers (Basel) 2021; 13:cancers13081964. [PMID: 33921727 PMCID: PMC8073809 DOI: 10.3390/cancers13081964] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/01/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Although taxane-trastuzumab-pertuzumab combinations in the first-line treatment setting significantly improved clinical outcomes in patients with Human Epidermal growth factor Receptor 2 positive (HER2+) advanced breast cancer (aBC), their clinical efficacy is highly heterogeneous, and reliable biomarkers of benefit from this treatment are lacking. Different peripheral blood parameters have emerged as prognostic biomarkers in breast cancer, but their predictive role in HER2+ BC patients receiving dual anti-HER2 blockade remains unclear. In this work, we identified the Pan-Immune-Inflammatory Value (PIV), a recently defined parameter, taking into account peripheral blood neutrophil, platelet, monocyte and lymphocyte counts, as an independent predictor of worse OS in patients with HER2+ aBC receiving first line trastuzumab-pertuzumab biochemotherapy. The PIV outperforms other well-known peripheral blood parameters, thus potentially representing a new tool to improve the prognostic stratification of HER2+ aBC patients in a first-line treatment setting. Abstract Different peripheral blood parameters have emerged as prognostic biomarkers in breast cancer (BC), but their predictive role in Human Epidermal growth factor Receptor 2 positive (HER2+) advanced BC (aBC) patients receiving dual anti-HER2 blockade remains unclear. We evaluated the impact of the Pan-Immune-Inflammatory Value (PIV), defined as the product of peripheral blood neutrophil, platelet, and monocyte counts divided by lymphocyte counts, on the prognosis of HER2+ aBC patients treated with first line trastuzumab-pertuzumab-based biochemotherapy. We also evaluated the association between the neutrophil-to-lymphocyte ratio (NLR), the platelet-to-lymphocyte ratio (PLR), and the monocyte to lymphocyte ratio (MLR) and clinical outcomes. Cox regression models were used to estimate the impact of these variables, as well as of other clinically relevant covariates, on patient survival. We included 57 HER2+ aBC patients treated with taxane-trastuzumab-pertuzumab in our Institution. High baseline MLR, PLR, and PIV were similarly predictive of worse PFS at univariate analysis, but only high PIV was associated with a trend toward worse PFS at multivariable analysis. Regarding OS, both high PIV and MLR were associated with significantly worse patient survival at univariate analysis, but only the PIV was statistically significantly associated with worse overall survival at multivariable analysis (HR 7.96; 95% CI: 2.18–29.09). Our study reveals the PIV as a new and potent predictor of OS in HER2+ aBC patients treated with first line trastuzumab-pertuzumab-containing biochemotherapy. Prospective studies are needed to validate this new prognostic parameter in HER2+ aBC.
Collapse
|
41
|
Wang Z, Jiang R, Li Q, Wang H, Tao Q, Zhai Z. Elevated M-MDSCs in Circulation Are Indicative of Poor Prognosis in Diffuse Large B-Cell Lymphoma Patients. J Clin Med 2021; 10:jcm10081768. [PMID: 33921711 PMCID: PMC8074013 DOI: 10.3390/jcm10081768] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/28/2021] [Accepted: 04/09/2021] [Indexed: 12/20/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are defined as negative regulators that suppress the immune response through a variety of mechanisms, which usually cluster in cancer, inflammation, and autoimmune diseases. This study aims to investigate the correlation between M-MDSCs and the clinical features of diffuse large B-cell lymphoma (DLBCL) patients, as well as the possible accumulation mechanism of M-MDSCs. The level of M-MDSCs is significantly increased in newly diagnosed and relapsed DLBCL patients. Regarding newly diagnosed DLBCL patients, the frequency of M-MDSCs is positively correlated with tumor progression and negatively correlated with overall survival (OS). More importantly, the level of M-MDSCs can be defined as a biomarker for a poor prognosis in DLBCL patients. Additionally, interleukin-35 (IL-35) mediates the accumulation of M-MDSCs in DLBCL patients. Anti-IL-35 treatment significantly reduces levels of M-MDSCs in Ly8 tumor-bearing mice. Thus, M-MDSCs are involved in the pathological process of DLBCL. Targeting M-MDSCs may be a promising therapeutic strategy for the treatment of DLBCL patients.
Collapse
Affiliation(s)
- Zhitao Wang
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (Z.W.); (Q.L.); (H.W.); (Q.T.)
| | - Rui Jiang
- Department of Hematology, The 901st Hospital of the Joint Logistics Support Force of PLA, Hefei 230032, China;
| | - Qian Li
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (Z.W.); (Q.L.); (H.W.); (Q.T.)
| | - Huiping Wang
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (Z.W.); (Q.L.); (H.W.); (Q.T.)
| | - Qianshan Tao
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (Z.W.); (Q.L.); (H.W.); (Q.T.)
| | - Zhimin Zhai
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (Z.W.); (Q.L.); (H.W.); (Q.T.)
- Correspondence:
| |
Collapse
|
42
|
Bagherifar R, Kiaie SH, Hatami Z, Ahmadi A, Sadeghnejad A, Baradaran B, Jafari R, Javadzadeh Y. Nanoparticle-mediated synergistic chemoimmunotherapy for tailoring cancer therapy: recent advances and perspectives. J Nanobiotechnology 2021; 19:110. [PMID: 33865432 PMCID: PMC8052859 DOI: 10.1186/s12951-021-00861-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022] Open
Abstract
Nowadays, a potent challenge in cancer treatment is considered the lack of efficacious strategy, which has not been able to significantly reduce mortality. Chemoimmunotherapy (CIT) as a promising approach in both for the first-line and relapsed therapy demonstrated particular benefit from two key gating strategies, including chemotherapy and immunotherapy to cancer therapy; therefore, the discernment of their participation and role of potential synergies in CIT approach is determinant. In this study, in addition to balancing the pros and cons of CIT with the challenges of each of two main strategies, the recent advances in the cancer CIT have been discussed. Additionally, immunotherapeutic strategies and the immunomodulation effect induced by chemotherapy, which boosts CIT have been brought up. Finally, harnessing and development of the nanoparticles, which mediated CIT have expatiated in detail.
Collapse
Affiliation(s)
- Rafieh Bagherifar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Hossein Kiaie
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Hatami
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Armin Ahmadi
- Department of Chemical & Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Jafari
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Shafa St, Ershad Blvd., P.O. BoX: 1138, 57147, Urmia, Iran.
- Department of Immunology and Genetics, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Yousef Javadzadeh
- Biotechnology Research Center, and Faculty of Pharmacy, Tabriz University of Medical Science, 5166-15731, Tabriz, Iran.
| |
Collapse
|
43
|
Lan HR, Du WL, Liu Y, Mao CS, Jin KT, Yang X. Role of immune regulatory cells in breast cancer: Foe or friend? Int Immunopharmacol 2021; 96:107627. [PMID: 33862552 DOI: 10.1016/j.intimp.2021.107627] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/26/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022]
Abstract
Breast cancer (BC) is the most common cancer among women between the ages of 20 and 50, affecting more than 2.1 million people and causing the annual death of more than 627,000 women worldwide. Based on the available knowledge, the immune system and its components are involved in the pathogenesis of several malignancies, including BC. Cancer immunobiology suggests that immune cells can play a dual role and induce anti-tumor or immunosuppressive responses, depending on the tumor microenvironment (TME) signals. The most important effector immune cells with anti-tumor properties are natural killer (NK) cells, B, and T lymphocytes. On the other hand, immune and non-immune cells with regulatory/inhibitory phenotype, including regulatory T cells (Tregs), regulatory B cells (Bregs), tolerogenic dendritic cells (tDCs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), mesenchymal stem cells (MSCs), and regulatory natural killer cells (NKregs), can promote the growth and development of tumor cells by inhibiting anti-tumor responses, inducing angiogenesis and metastasis, as well as the expression of inhibitory molecules and suppressor mediators of the immune system. However, due to the complexity of the interaction and the modification in the immune cells' phenotype and the networking of the immune responses, the exact mechanism of action of the immunosuppressive and regulatory cells is not yet fully understood. This review article reviews the immune responses involved in BC as well as the role of regulatory and inhibitory cells in the pathogenesis of the disease. Finally, therapeutic approaches based on inhibition of immunosuppressive responses derived from regulatory cells are discussed.
Collapse
Affiliation(s)
- Huan-Rong Lan
- Department of Breast and Thyroid Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, PR China
| | - Wen-Lin Du
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, PR China; Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, PR China
| | - Yuyao Liu
- Department of Colorectal Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, PR China
| | - Chun-Sen Mao
- Department of Colorectal Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, PR China
| | - Ke-Tao Jin
- Department of Colorectal Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, PR China
| | - Xue Yang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, PR China.
| |
Collapse
|
44
|
Lo YW, Lee AYL, Liu YC, Ko HH, Peng HH, Lee HC, Pan PY, Chiang CP, Cheng SJ. β-glucan therapy converts the inhibition of myeloid-derived suppressor cells in oral cancer patients. Oral Dis 2021; 28:1484-1495. [PMID: 33655573 DOI: 10.1111/odi.13827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/21/2021] [Accepted: 02/08/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The myeloid-derived suppressor cells (MDSCs) frequently have a high expansion in cancer patients. This research explored whether administration of β-glucan could increase anti-tumor immunity in oral squamous cell carcinoma (OSCC) patients. MATERIALS AND METHODS This study evaluated the MDSC level of circulating blood as CD33+ /CD11b+ /HLA-DR-/low by flow cytometry in 30 healthy donors (HDs, group I), in 48 oral squamous cell carcinoma (OSCC) patients before and after 14-day preoperative administration of β-glucan (group II), and in 52 OSCC patients without taking β-glucan (group III). RESULTS A significantly higher mean MDSC level was observed in 100 OSCC patients than in 30 HDs (p < .001). There was a significant reduction of the mean MDSC level in group II patients after taking β-glucan (p < .001). Moreover, we discovered a significantly higher recurrence-free survival (RFS) in group II than in group III patients (p = .026). Finally, the multivariate Cox regression further identified the MDSC level ≤1% and administration of β-glucan as more favorable prognostic factors for OSCC patients. CONCLUSION Preoperative administration of β-glucan can augment anti-tumor immunity and increase RFS rate via subversion of suppressive function of MDSC in OSCC patients.
Collapse
Affiliation(s)
- Ya-Wen Lo
- School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Alan Yueh-Luen Lee
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan.,Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ching Liu
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan.,Department of Dentistry, China Medical University Hospital, Taichung, Taiwan
| | - Hui-Hsin Ko
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan.,Department of Dentistry, College of Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Dentistry, College of Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Hsin-Hui Peng
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan.,Department of Dentistry, College of Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Dentistry, College of Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Hsiang-Chieh Lee
- Graduate Institute of Photonics and Optoelectronics, National Taiwan University, Taipei, Taiwan
| | - Pei-Yao Pan
- Department of Dentistry, College of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Pin Chiang
- School of Dentistry, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan.,Department of Dentistry, College of Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Dentistry, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Shih-Jung Cheng
- School of Dentistry, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan.,Department of Dentistry, College of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
45
|
Bansal D, Reimers MA, Knoche EM, Pachynski RK. Immunotherapy and Immunotherapy Combinations in Metastatic Castration-Resistant Prostate Cancer. Cancers (Basel) 2021; 13:cancers13020334. [PMID: 33477569 PMCID: PMC7831137 DOI: 10.3390/cancers13020334] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/01/2021] [Accepted: 01/14/2021] [Indexed: 12/21/2022] Open
Abstract
Although most prostate cancers are localized, and the majority are curable, recurrences occur in approximately 35% of men. Among patients with prostate-specific antigen (PSA) recurrence and PSA doubling time (PSADT) less than 15 months after radical prostatectomy, prostate cancer accounted for approximately 90% of the deaths by 15 years after recurrence. An immunosuppressive tumor microenvironment (TME) and impaired cellular immunity are likely largely responsible for the limited utility of checkpoint inhibitors (CPIs) in advanced prostate cancer compared with other tumor types. Thus, for immunologically "cold" malignancies such as prostate cancer, clinical trial development has pivoted towards novel approaches to enhance immune responses. Numerous clinical trials are currently evaluating combination immunomodulatory strategies incorporating vaccine-based therapies, checkpoint inhibitors, and chimeric antigen receptor (CAR) T cells. Other trials evaluate the efficacy and safety of these immunomodulatory agents' combinations with standard approaches such as androgen deprivation therapy (ADT), taxane-based chemotherapy, radiotherapy, and targeted therapies such as tyrosine kinase inhibitors (TKI) and poly ADP ribose polymerase (PARP) inhibitors. Here, we will review promising immunotherapies in development and ongoing trials for metastatic castration-resistant prostate cancer (mCRPC). These novel trials will build on past experiences and promise to usher a new era to treat patients with mCRPC.
Collapse
|
46
|
Immunological status of peripheral blood is associated with prognosis in patients with bone and soft-tissue sarcoma. Oncol Lett 2021; 21:212. [PMID: 33510813 PMCID: PMC7836390 DOI: 10.3892/ol.2021.12473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/23/2020] [Indexed: 12/22/2022] Open
Abstract
Immune-checkpoint inhibitors have shown promising antitumor effects against certain types of cancer. However, specific immune-checkpoint inhibitors for patients with sarcoma have yet to be identified, whereas the immunological status of peripheral blood in patients with bone sarcoma and soft-tissue sarcoma (STS) remains unknown. In addition, it is unclear whether the immunological status from the peripheral blood could be used as a prognostic indicator. Therefore, the present study aimed to clarify the immunological status of peripheral blood samples derived from patients with bone sarcoma and STS. Immune monitoring was performed using the peripheral blood samples of 61 patients with no metastasis of high-grade sarcoma. A total of 25 patients with metastatic sarcoma were used for comparison. A total of 41 immune cell subsets were analyzed using multicolor-flow cytometry. The patients that did not have metastasis demonstrated higher quantities of monocytic myeloid-derived suppressor cells (M-MDSCs) and T cell immunoglobulin and mucin domain-3 (Tim-3)+ CD8+ T cells, which were significantly associated with poor disease-free survival (DFS) time, while higher quantities of NKG2D+ CD8+ T cells were significantly associated with improved DFS time. Multivariate Cox regression analysis demonstrated that the number of Tim-3+ CD8+ T cells was associated with lower DFS time. A significant association was also found between the number of M-MDSCs and progression-free survival (PFS) time in patients with metastasis. The results suggested the occurrence of immune surveillance, which indicated that the host immune reaction against cancer existed in patients with bone sarcoma and STS. Notably, a high number of M-MDSCs was associated with both DFS and PFS time, suggesting a strong prognostic value. The data suggested that the immune status of peripheral blood was associated with the prognosis in patients with sarcoma, as previously reported in patients with other cancer types. In summary, the results may assist with the development of novel strategies for sarcoma treatment, based on the use of biomarkers or immunotherapy.
Collapse
|
47
|
Liu F, Wu H. Identification of Prognostic Biomarkers and Molecular Targets Among JAK Family in Breast Cancer. J Inflamm Res 2021; 14:97-114. [PMID: 33469338 PMCID: PMC7813467 DOI: 10.2147/jir.s284889] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Background Janus kinases (JAKs) are a family of non-receptor tyrosine kinases involved in multiple malignancies. However, clinical values of JAKs as prognostic markers and potential mechanism as molecular targets in breast invasive carcinoma (BC) are not completely clarified. Methodology TIMER, UALCAN and GEPIA were used to assess the expression and methylation levels of JAKs in BC. Kaplan–Meier Plotter, bc-GenExMiner, SurvExpress, TRGAted, MethSurv, and SurvivalMeth were used to assess the multilevel prognostic significance of JAKs in breast cancer patients. And cBioPortal, TIMER, STRING, GeneMANIA, NetworkAnalysis, LinkedOmics, DAVID 6.8, and Metascape were applied for multilayer networks and functional enrichment analyses. Correlations between immune cell infiltrates/their gene markers and JAKs were evaluated by TIMER. Results We first explored the expression and methylation level of JAKs in breast cancer and found significantly reduced JAK1 and JAK2 expression at mRNA and protein levels, significantly higher JAK3 protein expression, and significantly increased TYK2 expression at mRNA level but decreased at protein level. In addition, hypermethylation of JAK3 and TYK2 and hypomethylation of JAK1 were found in tumor samples. In terms of prognostic values of JAKs in BC patients, low transcriptional levels of JAK1, JAK2, JAK3, and TYK2 indicated worse OS/DMFS/PPS/RFS/DFS, inferior DFS, worse RFS, and shorter OS/DMFS/RFS, respectively. The mRNA signature analysis showed that high-risk group had unfavorable OS/RFS/MFS. Low JAK2 protein level indicated unfavorable DSS/PFS in BC patients. Five CpGs of JAK1, four CpGs of JAK2, 20 CpGs of JAK3, and 13 CpGs of TYK2 were significantly associated with prognosis in BC patients. The DNA methylation signature analysis also suggested worse prognosis in the high-risk group. For potential biological roles of JAKs, interaction analyses, functional enrichment analyses for biological process, cellular component, molecular function, and KEGG pathway analyses of JAKs and their neighbor genes in BC were conducted. Kinase targets, gene–miRNA interactions, and transcription factor–gene interactions of JAKs were also identified. Furthermore, JAKs were found to be significantly related to immune infiltrates as well as the expression levels of multiple immune markers in BC. Conclusion JAKs showed multilevel prognostic value and important biological roles in BC. They might serve as promising prognostic markers and possible targets in breast cancer.
Collapse
Affiliation(s)
- Fangteng Liu
- Department of Breast Surgery, The Third Hospital of Nanchang, Nanchang 330009, Jiangxi, People's Republic of China.,Faculty of Medicine, University of Munich, Munich 80336, Germany
| | - Hengyu Wu
- Department of Breast Surgery, The Third Hospital of Nanchang, Nanchang 330009, Jiangxi, People's Republic of China
| |
Collapse
|
48
|
Linares-Galiana I, Berenguer-Frances MA, Cañas-Cortés R, Pujol-Canadell M, Comas-Antón S, Martínez E, Laplana M, Pérez-Montero H, Pla-Farnós MJ, Navarro-Martin A, Nuñez M, Both B, Guedea F. Changes in peripheral immune cells after intraoperative radiation therapy in low-risk breast cancer. JOURNAL OF RADIATION RESEARCH 2021; 62:110-118. [PMID: 33006364 PMCID: PMC7779348 DOI: 10.1093/jrr/rraa083] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/07/2020] [Indexed: 05/15/2023]
Abstract
A detailed understanding of the interactions and the best dose-fractionation scheme of radiation to maximize antitumor immunity have not been fully established. In this study, the effect on the host immune system of a single dose of 20 Gy through intraoperative radiation therapy (IORT) on the surgical bed in low-risk breast cancer patients undergoing conserving breast cancer has been assessed. Peripheral blood samples from 13 patients were collected preoperatively and at 48 h and 3 and 10 weeks after the administration of radiation. We performed a flow cytometry analysis for lymphocyte subpopulations, natural killer cells (NK), regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSCs). We observed that the subpopulation of NK CD56+high CD16+ increased significantly at 3 weeks after IORT (0.30-0.42%, P < 0.001), while no changes were found in immunosuppressive profile, CD4+CD25+Foxp3+Helios+ Treg cells, granulocytic MDSCs (G-MDSCs) and monocytic MDSCs (Mo-MDSCs). A single dose of IORT may be an effective approach to improve antitumor immunity based on the increase in NK cells and the non-stimulation of immunosuppressive cells involved in immune escape. These findings support future combinations of IORT with immunotherapy, if they are confirmed in a large cohort of breast cancer patients.
Collapse
Affiliation(s)
- Isabel Linares-Galiana
- Radiation Oncology Department, Hospital Duran i Reynals, Institut Català d'Oncologia (ICO), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
- Radiobiology and Cancer Group, ONCOBELL Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
| | - Miguel Angel Berenguer-Frances
- Radiation Oncology Department, Hospital Duran i Reynals, Institut Català d'Oncologia (ICO), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
- Radiobiology and Cancer Group, ONCOBELL Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
| | - Rut Cañas-Cortés
- Radiobiology and Cancer Group, ONCOBELL Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
| | - Monica Pujol-Canadell
- Radiobiology and Cancer Group, ONCOBELL Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
| | - Silvia Comas-Antón
- Radiation Oncology Department, Hospital Germans Trias i Pujol, Institut Català d'Oncologia (ICO), Carretera de Canyet, s/n, 08916 Badalona, Spain
| | - Evelyn Martínez
- Radiation Oncology Department, Hospital Duran i Reynals, Institut Català d'Oncologia (ICO), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
| | - Maria Laplana
- Radiation Oncology Department, Hospital Duran i Reynals, Institut Català d'Oncologia (ICO), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
| | - Héctor Pérez-Montero
- Radiation Oncology Department, Hospital Duran i Reynals, Institut Català d'Oncologia (ICO), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
| | - María Jesús Pla-Farnós
- Gynecology Department, Hospital Universitari de Bellvitge, Carrer de la Feixa Llarga, s/n, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Arturo Navarro-Martin
- Radiation Oncology Department, Hospital Duran i Reynals, Institut Català d'Oncologia (ICO), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
- Radiobiology and Cancer Group, ONCOBELL Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
| | - Miriam Nuñez
- Radiation Oncology Department, Hospital Duran i Reynals, Institut Català d'Oncologia (ICO), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
- Radiobiology and Cancer Group, ONCOBELL Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
| | - Brigitte Both
- Medical Affairs & Professional Education, Business Sector Radiotherapy, Medical Technology Business Group, Carl Zeiss Meditec AG, ZEISS Group, Rudolf-Eber-Straße 11 Oberkochen, Germany
| | - Ferran Guedea
- Radiation Oncology Department, Hospital Duran i Reynals, Institut Català d'Oncologia (ICO), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
- Radiobiology and Cancer Group, ONCOBELL Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Avinguda de la Gran Via de l'Hospitalet 199-203, L'Hospitalet de Llobregat, 08098 Barcelona, Spain
| |
Collapse
|
49
|
Mehta AK, Cheney EM, Hartl CA, Pantelidou C, Oliwa M, Castrillon JA, Lin JR, Hurst KE, de Oliveira Taveira M, Johnson NT, Oldham WM, Kalocsay M, Berberich MJ, Boswell SA, Kothari A, Johnson S, Dillon DA, Lipschitz M, Rodig S, Santagata S, Garber JE, Tung N, Yélamos J, Thaxton JE, Mittendorf EA, Sorger PK, Shapiro GI, Guerriero JL. Targeting immunosuppressive macrophages overcomes PARP inhibitor resistance in BRCA1-associated triple-negative breast cancer. NATURE CANCER 2021; 2:66-82. [PMID: 33738458 PMCID: PMC7963404 DOI: 10.1038/s43018-020-00148-7] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/27/2020] [Indexed: 12/26/2022]
Abstract
Despite objective responses to PARP inhibition and improvements in progression-free survival compared to standard chemotherapy in patients with BRCA-associated triple-negative breast cancer (TNBC), benefits are transitory. Using high dimensional single-cell profiling of human TNBC, here we demonstrate that macrophages are the predominant infiltrating immune cell type in BRCA-associated TNBC. Through multi-omics profiling we show that PARP inhibitors enhance both anti- and pro-tumor features of macrophages through glucose and lipid metabolic reprogramming driven by the sterol regulatory element-binding protein 1 (SREBP-1) pathway. Combined PARP inhibitor therapy with CSF-1R blocking antibodies significantly enhanced innate and adaptive anti-tumor immunity and extends survival in BRCA-deficient tumors in vivo and is mediated by CD8+ T-cells. Collectively, our results uncover macrophage-mediated immune suppression as a liability of PARP inhibitor treatment and demonstrate combined PARP inhibition and macrophage targeting therapy induces a durable reprogramming of the tumor microenvironment, thus constituting a promising therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Anita K Mehta
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Emily M Cheney
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Christina A Hartl
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Constantia Pantelidou
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Madisson Oliwa
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jessica A Castrillon
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jia-Ren Lin
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Katie E Hurst
- Department of Orthopedics and Physical Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Mateus de Oliveira Taveira
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Department of Imaging, AC Camargo Cancer Center, São Paulo, Brazil
| | - Nathan T Johnson
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - William M Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Marian Kalocsay
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Matthew J Berberich
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Sarah A Boswell
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Aditi Kothari
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Shawn Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Deborah A Dillon
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mikel Lipschitz
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Scott Rodig
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sandro Santagata
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Nadine Tung
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - José Yélamos
- Cancer Research Program, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Jessica E Thaxton
- Department of Orthopedics and Physical Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Charleston, SC, USA
| | - Elizabeth A Mittendorf
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, MA, USA
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jennifer L Guerriero
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA.
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA.
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
50
|
Furukawa N, Popel AS. Peptides that immunoactivate the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2021; 1875:188486. [PMID: 33276025 PMCID: PMC8369429 DOI: 10.1016/j.bbcan.2020.188486] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/04/2020] [Accepted: 11/21/2020] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy has achieved positive clinical outcomes and is revolutionizing cancer treatment. However, cancer immunotherapy has thus far failed to improve outcomes for most "cold tumors", which are characterized by low infiltration of immune cells and immunosuppressive tumor microenvironment. Enhancing the responsiveness of cold tumors to cancer immunotherapy by stimulating the components of the tumor microenvironment is a strategy pursued in the last decade. Currently, most of the agents used to modify the tumor microenvironment are small molecules or antibodies. Small molecules exhibit low affinity and specificity towards the target and antibodies have shortcomings such as poor tissue penetration and high production cost. Peptides may overcome these drawbacks and therefore are promising materials for immunomodulating agents. Here we systematically summarize the currently developed immunoactivating peptides and discuss the potential of peptide therapeutics in cancer immunology.
Collapse
Affiliation(s)
- Natsuki Furukawa
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, USA.
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, USA; The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|