1
|
Xu J, Wang H, Zhang C, Jin SH, Chen X, Tan F, Frey B, Hecht M, Sun JG, Gaipl US, Ma H, Zhou JG. Efficacy of radiotherapy combined with atezolizumab or docetaxel in patients with previously treated NSCLC. iScience 2024; 27:111363. [PMID: 39640586 PMCID: PMC11617966 DOI: 10.1016/j.isci.2024.111363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/01/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024] Open
Abstract
Radiotherapy showed synergy with immunotherapy, yet the comparative effectiveness of combining immunotherapy (iRT) or chemotherapy (CRT) after platinum therapy failure in advanced non-small cell lung cancer (NSCLC) remains unexplored. We analyzed 163 patients (iRT: n = 120 vs. CRT: n = 43) eligible for combination radiotherapy. Before matching, median overall survival (OS) was significantly longer in iRT group (7.79 vs. 4.57 months, hazard ratio [HR]: 0.62, 95% confidence interval [CI]: 0.41-0.94, p = 0.024). After 1:2 propensity score matching (PSM) and inverse probability of treatment weighting (IPTW), iRT group showed improved OS, consistent with unmatched analysis (PSM, p = 0.033 and IPTW, p = 0.035). Exploratory analysis suggested that PD1+, central memory PD1+, and effector memory PD-L1+ CD4+ T cells were strong predictive biomarkers for iRT-treated patients (P OS = 0.025, P OS = 0.002, P OS = 0.010, respectively). Proliferative CD4+ T celllow was a prognostic (P OS = 0.008) and predictive biomarker for iRT (P OS < 0.001). Our work revealed iRT was prolonged OS in previously treated advanced NSCLC patients. Additionally, proliferative CD4+ T cell served as prognostic and predictive biomarkers.
Collapse
Affiliation(s)
- Junzhu Xu
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
| | - Haitao Wang
- Thoracic Surgery Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| | - Chi Zhang
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
| | - Su-Han Jin
- Department of Orthodontics, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaofei Chen
- Oncology Biometrics, AstraZeneca, Gaithersburg, MD 20850, USA
| | - Fangya Tan
- Harrisburg University of Science and Technology, Harrisburg, PA 17101, USA
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander- Universität Erlangen-Nurnberg, Erlangen, Germany
| | - Markus Hecht
- Department of Radiotherapy and Radiation Oncology, Saarland University Medical Center, Homburg, Germany
| | - Jian-Guo Sun
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Udo S. Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander- Universität Erlangen-Nurnberg, Erlangen, Germany
| | - Hu Ma
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
| | - Jian-Guo Zhou
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander- Universität Erlangen-Nurnberg, Erlangen, Germany
| |
Collapse
|
2
|
Sosa Cuevas E, Mouret S, Vayssière G, Kerboua S, Girard P, Molens JP, Manceau M, Charles J, Saas P, Aspord C. Circulating immune landscape in melanoma patients undergoing anti-PD1 therapy reveals key immune features according to clinical response to treatment. Front Immunol 2024; 15:1507938. [PMID: 39687620 PMCID: PMC11646980 DOI: 10.3389/fimmu.2024.1507938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Immune checkpoint blockers (ICB) bring unprecedented clinical success, yet many patients endure immune mediated adverse effects and/or fail to respond. Predictive signatures of response to ICB and mechanisms of clinical efficacy or failure remain understudied. DC subsets, in network with conventional αβ T (Tconv), NK, γδ T and iNKT cells, harbor pivotal roles in tumor control, yet their involvement in response to ICB remained underexplored. Methods We performed an extensive longitudinal monitoring of circulating immune cells from melanoma patients treated with first-line anti-PD1, before (T0) and during treatment. We assessed the phenotypic and functional features of DC and effector cells' subsets by multi-parametric flow cytometry and ProcartaPlex® dosages. Results We revealed differences according to response to treatment and modulations of patterns during treatment, highlighting a strong link between the immune landscape and the outcome of anti-PD1 therapy. Responders exhibited higher frequencies of circulating cDC1s, CD8+ T cells, and γδ2+ T cells in central memory (CM) stage. Notably, we observed a distinct remodeling of ICP expression profile, activation status and natural cytotoxicity receptor patterns of immune subsets during treatment. Anti-PD1 modulated DCs' functionality and triggered deep changes in the functional orientation of Tconv and γδT cells. Discussion Overall, our work provides new insights into the immunological landscape sustaining favorable clinical responses or resistance to first-line anti-PD1 therapy in melanoma patients. Such exploration participates in uncovering the mechanism of action of anti-PD1, discovering innovative predictive signatures of response, and paves the way to design pertinent combination strategies to improve patient clinical benefits in the future.
Collapse
Affiliation(s)
- Eleonora Sosa Cuevas
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Stéphane Mouret
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Univ. Grenoble Alpes, Grenoble, France
| | - Guillaume Vayssière
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Siham Kerboua
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Pauline Girard
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Jean-Paul Molens
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Marc Manceau
- Department of Medicine, Clinical Investigation Center, CHU Grenoble Alpes, Univ. Grenoble Alpes, Grenoble, France
| | - Julie Charles
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Univ. Grenoble Alpes, Grenoble, France
| | - Philippe Saas
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Caroline Aspord
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| |
Collapse
|
3
|
Kotrulev M, Gomez-Touriño I, Cordero OJ. Soluble CD26: From Suggested Biomarker for Cancer Diagnosis to Plausible Marker for Dynamic Monitoring of Immunotherapy. Cancers (Basel) 2024; 16:2427. [PMID: 39001488 PMCID: PMC11240764 DOI: 10.3390/cancers16132427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Soluble CD26 (sCD26), a glycoprotein with dipeptidyl peptidase (DPP4) enzymatic activity, can contribute to early diagnosis of colorectal cancer and advanced adenomas and has been studied, including for prognostic purposes, across various other types of cancer and disease. The latest research in this field has confirmed that most, though not all, serum/plasma sCD26 is related to inflammation. The shedding and/or secretion of sCD26 from different immune cells are being investigated, and blood DPP4 activity levels do not correlate very strongly with protein titers. Some of the main substrates of this enzyme are key chemokines involved in immune cell migration, and both soluble and cell-surface CD26 can bind adenosine deaminase (ADA), an enzyme involved in the metabolism of immunosuppressor extracellular adenosine. Of note, there are T cells enriched in CD26 expression and, in mice tumor models, tumor infiltrating lymphocytes exhibited heightened percentages of CD26+ correlating with tumor regression. We employed sCD26 as a biomarker in the follow-up after curative resection of colorectal cancer for the early detection of tumor recurrence. Changes after treatment with different biological disease-modifying antirheumatic drugs, including Ig-CTLA4, were also observed in rheumatoid arthritis. Serum soluble CD26/DPP4 titer variation has recently been proposed as a potential prognostic biomarker after a phase I trial in cancer immunotherapy with a humanized anti-CD26 antibody. We propose that dynamic monitoring of sCD26/DPP4 changes, in addition to well-known inflammatory biomarkers such as CRP already in use as informative for immune checkpoint immunotherapy, may indicate resistance or response during the successive steps of the treatment. As tumor cells expressing CD26 can also produce sCD26, the possibility of sorting immune- from non-immune-system-originated sCD26 is discussed.
Collapse
Affiliation(s)
- Martin Kotrulev
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.K.); (I.G.-T.)
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Iria Gomez-Touriño
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.K.); (I.G.-T.)
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Oscar J. Cordero
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
4
|
Zhao W, Liang Z, Yao Y, Ge Y, An G, Duan L, Yao J. GGT5: a potential immunotherapy response inhibitor in gastric cancer by modulating GSH metabolism and sustaining memory CD8+ T cell infiltration. Cancer Immunol Immunother 2024; 73:131. [PMID: 38748299 PMCID: PMC11096297 DOI: 10.1007/s00262-024-03716-3] [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: 11/03/2023] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
PURPOSE The variable responses to immunotherapy observed in gastric cancer (GC) patients can be attributed to the intricate nature of the tumor microenvironment. Glutathione (GSH) metabolism significantly influences the initiation and progression of gastric cancer. Consequently, targeting GSH metabolism holds promise for improving the effectiveness of Immune checkpoints inhibitors (ICIs). METHODS We investigated 16 genes related to GSH metabolism, sourced from the MSigDB database, using pan-cancer datasets from TCGA. The most representative prognosis-related gene was identified for further analysis. ScRNA-sequencing analysis was used to explore the tumor heterogeneity of GC, and the results were confirmed by Multiplex immunohistochemistry (mIHC). RESULTS Through DEGs, LASSO, univariate and multivariate Cox regression analyses, and survival analysis, we identified GGT5 as the hub gene in GSH metabolism with the potential to promote GC. Combining CIBERSORT, ssGSEA, and scRNA analysis, we constructed the immune architecture of GC. The subpopulations of T cells were isolated, revealing a strong association between GGT5 and memory CD8+ T cells. Furthermore, specimens from 10 GC patients receiving immunotherapy were collected. mIHC was used to assess the expression levels of GGT5 and memory CD8+ T cell markers. Our results established a positive correlation between GGT5 expression, the enrichment of memory CD8+ T cells, and a suboptimal response to immunotherapy. CONCLUSIONS Our study identifies GGT5, a hub gene in GSH metabolism, as a potential therapeutic target for inhibiting the response to immunotherapy in GC patients. These findings offer new insights into strategies for optimizing immunotherapy of GC.
Collapse
Affiliation(s)
- Wenjing Zhao
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ziwei Liang
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yongshi Yao
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yang Ge
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Guangyu An
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ling Duan
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiannan Yao
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Dong Y, Chen Z, Yang F, Wei J, Huang J, Long X. Prediction of immunotherapy responsiveness in melanoma through single-cell sequencing-based characterization of the tumor immune microenvironment. Transl Oncol 2024; 43:101910. [PMID: 38417293 PMCID: PMC10907870 DOI: 10.1016/j.tranon.2024.101910] [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: 11/27/2023] [Revised: 01/13/2024] [Accepted: 02/08/2024] [Indexed: 03/01/2024] Open
Abstract
Immune checkpoint inhibitors (ICB) therapy have emerged as effective treatments for melanomas. However, the response of melanoma patients to ICB has been highly heterogenous. Here, by analyzing integrated scRNA-seq datasets from melanoma patients, we revealed significant differences in the TiME composition between ICB-resistant and responsive tissues, with resistant or responsive tissues characterized by an abundance of myeloid cells and CD8+ T cells or CD4+ T cell predominance, respectively. Among CD4+ T cells, CD4+ CXCL13+ Tfh-like cells were associated with an immunosuppressive phenotype linked to immune escape-related genes and negative regulation of T cell activation. We also develop an immunotherapy response prediction model based on the composition of the immune compartment. Our predictive model was validated using CIBERSORTx on bulk RNA-seq datasets from melanoma patients pre- and post-ICB treatment and showed a better performance than other existing models. Our study presents an effective immunotherapy response prediction model with potential for further translation, as well as underscores the critical role of the TiME in influencing the response of melanomas to immunotherapy.
Collapse
Affiliation(s)
- Yucheng Dong
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Zhizhuo Chen
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Fan Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaxin Wei
- Department of Emergency Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiuzuo Huang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| | - Xiao Long
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
6
|
Su S, Chen F, Lv X, Qi L, Ding Z, Ren W, Wei M, Liu Y, Yu L, Liu B, Wang L. Predictive value of peripheral blood biomarkers in patients with non-small-cell lung cancer responding to anti-PD-1-based treatment. Cancer Immunol Immunother 2024; 73:12. [PMID: 38231411 PMCID: PMC10794255 DOI: 10.1007/s00262-023-03620-2] [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: 09/13/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND The introduction of the anti-PD-1 antibody has greatly improved the clinical outcomes of patients with non-small cell lung cancer (NSCLC). In this study, we retrospectively analyzed the efficacy of PD-1 antibody-based therapy in patients with locally advanced inoperable or metastatic NSCLC and reported an association between peripheral blood biomarkers and clinical response in these patients. METHODS This single-center study included medical record data of patients with NSCLC treated with the PD-1 antibody as a first-line or subsequent line of treatment, either as monotherapy or in combination with chemotherapy. The patients were enrolled from 2020 to 2022. We dynamically evaluated multiple Th1 and Th2 cytokines in the blood serum and analyzed the phenotype of T cells from the peripheral blood to explore the correlation between cytokine levels, T cell phenotypes, and clinical response. RESULTS A total of 88 patients with stage IIIA-IV NSCLC were enrolled, out of which 60 (68.18%) achieved a partial response (PR), 13 (14.77%) had stable disease (SD), and 15 (17.05%) experienced disease progression (PD). The disease control rate was 82.95%. Our results suggested a significant reduction (P = 0.002, P < 0.005) in lymphocyte absolute counts after treatment in patients with PD. Higher levels of IFN-γ (P = 0.023, P < 0.05), TNF-α (P = 0.00098, P < 0.005), IL-4 (P = 0.0031, P < 0.005), IL-5 (P = 0.0015, P < 0.005), and IL-10 (P = 0.036, P < 0.05) were detected in the peripheral blood before treatment in the PR group compared to the PD group. Moreover, patients with high levels of IL-5, IL-13, IL-4, IL-6, IFN-γ, and TNF-α (> 10 ng/mL) had superior progression-free survival compared to those with low levels (< 10 ng/mL). Furthermore, PD-1 expression on CD8+ T cells was higher in patients who showed a PR than in those who did not show a response (SD + PD; P = 0.042, P < 0.05). CONCLUSIONS The findings of this study imply that the decrease in absolute blood lymphocyte counts after treatment is correlated with disease progression. Serum cytokine levels may predict the effectiveness and survival rates of anti-PD-1 blockade therapy in patients with NSCLC. In addition, PD-1 expression on CD8+ T cells was positively associated with better clinical response. Our findings highlight the potential of peripheral blood biomarkers to predict the effectiveness of PD-1-targeted treatments in patients with NSCLC. Larger prospective studies are warranted to further clarify the value of these biomarkers.
Collapse
Affiliation(s)
- Shu Su
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210032, Jiangsu, China
| | - Fungjun Chen
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210032, Jiangsu, China
| | - Xin Lv
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210032, Jiangsu, China
| | - Liang Qi
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210032, Jiangsu, China
| | - Zhou Ding
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210032, Jiangsu, China
| | - Wei Ren
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210032, Jiangsu, China
| | - Ming Wei
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ye Liu
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lixia Yu
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210032, Jiangsu, China
| | - Baorui Liu
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210032, Jiangsu, China
| | - Lifeng Wang
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210032, Jiangsu, China.
| |
Collapse
|
7
|
Kverneland A, Thorsen S, Granhøj J, Hansen F, Konge M, Ellebæk E, Donia M, Svane I. Supervised clustering of peripheral immune cells associated with clinical response to checkpoint inhibitor therapy in patients with advanced melanoma. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 20:100396. [PMID: 37810199 PMCID: PMC10558712 DOI: 10.1016/j.iotech.2023.100396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Background and purpose Immune therapy with checkpoint inhibitors (CPIs) is a highly successful therapy in many cancers including metastatic melanoma. Still, many patients do not respond well to therapy and there are no blood-borne biomarkers available to assess the clinical outcome. Materials and methods To investigate cellular changes after CPI therapy, we carried out flow cytometry-based immune monitoring in a cohort of 90 metastatic melanoma patients before and after CPI therapy using the FlowSOM algorithm. To evaluate associations to the clinical outcome with therapy, we divided the patients based on progression-free survival. Results We found significant associations with CPI therapy in both peripheral blood mononuclear cell and T-cell subsets, but with the most pronounced effects in the latter. Particularly CD4+ effector memory T-cell subsets were associated with response with a positive correlation between CD27+HLA-DR+CD4+ effector memory T cells in a univariate (odds ratio: 1.07 [95% confidence interval 1.02-1.12]) and multivariate regression model (odds ratio: 1.08 [95% confidence interval 1.03-1.14]). We also found a trend towards stronger accumulation of CD57+CD8+ T cells in non-responding patients. Conclusion Our results show significant associations between immune monitoring and clinical outcome of therapy that could be evaluated as biomarkers in a clinical setting.
Collapse
Affiliation(s)
- A.H. Kverneland
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen
| | - S.U. Thorsen
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - J.S. Granhøj
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev
| | - F.S. Hansen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev
| | - M. Konge
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev
| | - E. Ellebæk
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev
| | - M. Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev
| | - I.M. Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev
| |
Collapse
|
8
|
Galati D, Zanotta S, Capone M, Madonna G, Mallardo D, Romanelli M, Simeone E, Festino L, Sparano F, Azzaro R, De Filippi R, Pinto A, Paulos CM, Ascierto PA. Potential clinical implications of CD4 +CD26 high T cells for nivolumab treated melanoma patients. J Transl Med 2023; 21:318. [PMID: 37170241 PMCID: PMC10176780 DOI: 10.1186/s12967-023-04184-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Nivolumab is an anti-PD1 antibody that has dramatically improved metastatic melanoma patients' outcomes. Nevertheless, many patients are resistant to PD-1 inhibition, occasionally experiencing severe off-target immune toxicity. In addition, no robust and reproducible biomarkers have yet been validated to identify the correct selection of patients who will benefit from anti-PD-1 treatment avoiding unwanted side effects. However, the strength of CD26 expression on CD4+ T lymphocytes permits the characterization of three subtypes with variable degrees of responsiveness to tumors, suggesting that the presence of CD26-expressing T cells in patients might be a marker of responsiveness to PD-1-based therapies. METHODS The frequency distribution of peripheral blood CD26-expressing cells was investigated employing multi-parametric flow cytometry in 69 metastatic melanoma patients along with clinical characteristics and blood count parameters at baseline (W0) and compared to 20 age- and sex-matched healthy controls. Percentages of baseline CD4+CD26high T cells were correlated with the outcome after nivolumab treatment. In addition, the frequency of CD4+CD26high T cells at W0 was compared with those obtained after 12 weeks (W1) of therapy in a sub-cohort of 33 patients. RESULTS Circulating CD4+CD26high T cells were significantly reduced in melanoma patients compared to healthy subjects (p = 0.001). In addition, a significant association was observed between a low baseline percentage of CD4+CD26high T cells (< 7.3%) and clinical outcomes, measured as overall survival (p = 0.010) and progression-free survival (p = 0.014). Moreover, patients with clinical benefit from nivolumab therapy had significantly higher frequencies of circulating CD4+CD26high T cells than patients with non-clinical benefit (p = 0.004) at 12 months. Also, a higher pre-treatment proportion of circulating CD4+CD26high T cells was correlated with Disease Control Rate (p = 0.014) and best Overall Response Rate (p = 0.009) at 12 months. Interestingly, after 12 weeks (W1) of nivolumab treatment, percentages of CD4+CD26high T cells were significantly higher in comparison with the frequencies measured at W0 (p < 0.0001), aligning the cell counts with the ranges seen in the blood of healthy subjects. CONCLUSIONS Our study firstly demonstrates that peripheral blood circulating CD4+CD26high T lymphocytes represent potential biomarkers whose perturbations are associated with reduced survival and worse clinical outcomes in melanoma patients.
Collapse
Affiliation(s)
- Domenico Galati
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Serena Zanotta
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Mariaelena Capone
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Gabriele Madonna
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Domenico Mallardo
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Marilena Romanelli
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Ester Simeone
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Lucia Festino
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Francesca Sparano
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Rosa Azzaro
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Rosaria De Filippi
- Dipartimento di Medicina Clinica e Chirurgia, Università Degli Studi Federico II, Naples, Italy
| | - Antonio Pinto
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Chrystal M. Paulos
- Division of Surgical Oncology, Department of Surgery, Emory University, Atlanta, GA USA
- Department of Microbiology and Immunology, Winship Cancer Institute, Emory University, Atlanta, GA USA
| | - Paolo A. Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| |
Collapse
|
9
|
van der Sluis TC, Beyrend G, van der Gracht ETI, Abdelaal T, Jochems SP, Belderbos RA, Wesselink TH, van Duikeren S, van Haften FJ, Redeker A, Ouboter LF, Beyranvand Nejad E, Camps M, Franken KLMC, Linssen MM, Hohenstein P, de Miranda NFCC, Mei H, Bins AD, Haanen JBAG, Aerts JG, Ossendorp F, Arens R. OX40 agonism enhances PD-L1 checkpoint blockade by shifting the cytotoxic T cell differentiation spectrum. Cell Rep Med 2023; 4:100939. [PMID: 36796366 PMCID: PMC10040386 DOI: 10.1016/j.xcrm.2023.100939] [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: 02/10/2022] [Revised: 10/07/2022] [Accepted: 01/20/2023] [Indexed: 02/17/2023]
Abstract
Immune checkpoint therapy (ICT) has the power to eradicate cancer, but the mechanisms that determine effective therapy-induced immune responses are not fully understood. Here, using high-dimensional single-cell profiling, we interrogate whether the landscape of T cell states in the peripheral blood predict responses to combinatorial targeting of the OX40 costimulatory and PD-1 inhibitory pathways. Single-cell RNA sequencing and mass cytometry expose systemic and dynamic activation states of therapy-responsive CD4+ and CD8+ T cells in tumor-bearing mice with expression of distinct natural killer (NK) cell receptors, granzymes, and chemokines/chemokine receptors. Moreover, similar NK cell receptor-expressing CD8+ T cells are also detected in the blood of immunotherapy-responsive cancer patients. Targeting the NK cell and chemokine receptors in tumor-bearing mice shows the functional importance of these receptors for therapy-induced anti-tumor immunity. These findings provide a better understanding of ICT and highlight the use and targeting of dynamic biomarkers on T cells to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Tetje C van der Sluis
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Guillaume Beyrend
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | | | - Tamim Abdelaal
- Department of Radiology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands; Systems and Biomedical Engineering Department, Faculty of Engineering, Cairo University, Giza 12613, Egypt; Pattern Recognition and Bioinformatics, Delft University of Technology, 2628XE Delft, the Netherlands
| | - Simon P Jochems
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Robert A Belderbos
- Department of Pulmonary Diseases, Erasmus Medical Center, 3015GD Rotterdam, the Netherlands
| | - Thomas H Wesselink
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Suzanne van Duikeren
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Floortje J van Haften
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Anke Redeker
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Laura F Ouboter
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Elham Beyranvand Nejad
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Marcel Camps
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Kees L M C Franken
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Margot M Linssen
- Central Animal and Transgenic Facility, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Peter Hohenstein
- Central Animal and Transgenic Facility, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Noel F C C de Miranda
- Department of Pathology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Hailiang Mei
- Department of Biomedical Data Sciences, Sequencing Analysis Support Core, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Adriaan D Bins
- Department of Internal Medicine, Amsterdam University Medical Center, 1105AZ Amsterdam, the Netherlands
| | - John B A G Haanen
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Joachim G Aerts
- Department of Pulmonary Diseases, Erasmus Medical Center, 3015GD Rotterdam, the Netherlands
| | - Ferry Ossendorp
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Ramon Arens
- Department of Immunology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands.
| |
Collapse
|
10
|
Fu Y, Huang Y, Li P, Wang L, Tang Z, Liu X, Bian X, Wu S, Wang X, Zhu B, Yu Y, Jiang J, Li C. Physical- and Chemical-Dually ROS-Responsive Nano-in-Gel Platforms with Sequential Release of OX40 Agonist and PD-1 Inhibitor for Augmented Combination Immunotherapy. NANO LETTERS 2023; 23:1424-1434. [PMID: 36779813 DOI: 10.1021/acs.nanolett.2c04767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Combination immunotherapy synergizing the PD-1 blockade with OX40 agonism has become a research hotspot, due to its enormous potential to overcome the restricted clinical objective response suffered by monotherapy. Questions of timing and sequence have been important aspects of immunotherapies when considering immunologic mechanisms; however, most of the time the straightforward additive approach was taken. Herein, our work is the first to investigate an alternative timing of aOX40 and aPD-1 treatment in melanoma-bearing mice, and it demonstrates that sequential administration (aOX40 first, then aPD-1 following) provided superior antitumor benefits than concurrent treatment. Based on that, to further avoid the limits suffered by solution forms, we adopted pharmaceutical technologies to construct an in situ-formed physical- and chemical-dually ROS-responsive nano-in-gel platform to implement sequential and prolonged release of aPD-1 and aOX40. Equipped with these advantages, the as-prepared (aPD-1NCs&aOX40)@Gels elicited augmented combination immunity and achieved great eradication of both primary and distant melanoma tumors in vivo.
Collapse
Affiliation(s)
- Yu Fu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yulan Huang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Pingrong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Luyao Wang
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX1 2JD, U.K
| | - Zhongjie Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xinlong Liu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xufei Bian
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Shuang Wu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xiaoyou Wang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Biyue Zhu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard University, Charlestown, Massachusetts 02138, United States
| | - Yang Yu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jiayun Jiang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University/Army Medical University, Chongqing 400038, P.R. China
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
11
|
Zhou N, Chen Y, Huang Q, Jiang L, Liao H, Gou H, Lu Y, Che G, Zhang Y. Pathological complete response to neoadjuvant tislelizumab plus chemotherapy in stage IIIB small cell lung cancer: A case report and literature review. Front Immunol 2023; 14:1111325. [PMID: 36911701 PMCID: PMC9992202 DOI: 10.3389/fimmu.2023.1111325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
Immunotherapy plus chemotherapy has been approved for the first-line treatment of extensive-stage small cell lung cancer (ES-SCLC, stage IV). Recently, the 2023 version of the National Comprehensive Cancer Network Guidelines recommended immunotherapy plus chemotherapy as the neoadjuvant regimen in patients with resectable non-small cell lung cancer (NSCLC). However, it is still unclear whether the combination regimen of immunotherapy plus chemotherapy is also beneficial for SCLC in the neoadjuvant context. Here, we report the case of a patient with stage IIIB SCLC who showed long-term survival and good tolerance to the neoadjuvant chemoimmunotherapy consisting of tislelizumab (an anti-PD-1 monoclonal antibody) plus etoposide-carboplatin. The patient achieved pathological complete response after receiving two cycles of neoadjuvant tislelizumab and chemotherapy followed by surgery. Two courses of post-operative tislelizumab and etoposide-carboplatin treatment were performed. The patient has survived for more than 23 months with no recurrence or metastases after neoadjuvant therapy. Multiplexed immunofluorescence and immunohistochemistry staining showed that the post-treatment specimens had remarkable immune cells infiltration, including CD3+ T cells, CD4+ T cells, and CD8+ T cells, which contrasted with very low levels of these cells in the pre-treatment samples. This study is, to the best of our knowledge, the first attempt to present the neoadjuvant chemoimmunotherapy of tislelizumab in combination with etoposide-carboplatin in SCLC. Our study suggested that neoadjuvant tislelizumab plus chemotherapy may facilitate radical resection and benefit patients with locally advanced (stage IIB-IIIC) SCLC.
Collapse
Affiliation(s)
- Nan Zhou
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Division of Medical Oncology, Cancer Center, Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuhong Chen
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Abdominal Oncology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Qian Huang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lili Jiang
- Department of Pathology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hu Liao
- Department of Thoracic surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hongfeng Gou
- Gastric Cancer Center, Division of Medical Oncology, Cancer Center, Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - You Lu
- Department of Thoracic Oncology, Cancer Center, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Guowei Che
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Zhang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
12
|
The tissue-resident marker CD103 on peripheral blood T cells predicts responses to anti-PD-1 therapy in gastric cancer. Cancer Immunol Immunother 2023; 72:169-181. [PMID: 35776160 DOI: 10.1007/s00262-022-03240-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/07/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment. Since clinical benefits are limited to a subset of patients, we aimed to identify peripheral blood biomarkers that predict the efficacy of the anti-programmed cell death protein 1 (PD-1) antibody (nivolumab) in patients with gastric cancer. METHODS We collected peripheral blood samples from gastric cancer patients (n = 29) before and after treatment with nivolumab and investigated the relationship between the frequency of surface or intracellular markers among nivolumab-binding PD-1+CD8+ T cells and treatment responses using multicolor flow cytometry. The tumors, lymph nodes, and peripheral blood of gastric cancer patients who underwent gastrectomy following nivolumab treatment were collected, and nivolumab-binding PD-1+CD8+ T cells in these tissue samples were characterized. RESULTS Patients with a high frequency of CD103 among PD-1+CD8+ T cells in peripheral blood 2 weeks after the start of treatment had significantly better progression-free survival than the low group (P = 0.032). This CD103+PD-1+CD8+ T cell population mainly consisted of central memory T cells, showing the high expression of Ki-67 and few cytotoxic granules. In contrast, effector memory T cells were more frequently observed among CD103+PD-1+CD8+ T cells in tumors, which implied a change in the differentiated status of central memory T cells in lymph nodes and peripheral blood to effector memory T cells in tumors during the treatment with ICIs. CONCLUSIONS A high frequency of CD103 among PD-1+CD8+ T cells 2 weeks after nivolumab treatment in patients with advanced gastric cancer may be a useful biomarker for predicting the efficacy of anti-PD-1 therapy.
Collapse
|
13
|
Igase M, Inanaga S, Tani K, Nakaichi M, Sakai Y, Sakurai M, Kato M, Tsukui T, Mizuno T. Long-term survival of dogs with stage 4 oral malignant melanoma treated with anti-canine PD-1 therapeutic antibody: A follow-up case report. Vet Comp Oncol 2022; 20:901-905. [PMID: 35535636 DOI: 10.1111/vco.12829] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 11/29/2022]
Abstract
A monoclonal antibody targeting programmed cell death-1 (PD-1) is one of the most promising treatments for human cancers. Clinical studies in humans demonstrated that the anti-PD-1 antibody provides a long-lasting tumour response. Previously, we established an anti-canine PD-1 therapeutic antibody (ca-4F12-E6), and the pilot clinical study demonstrated that the antibody was effective in dogs with oral malignant melanoma (OMM). However, two OMM cases were still undergoing treatment when the pilot study was published. Here, we describe the long-term follow-up of those two cases. Although both cases showed long-term survival with complete response (CR), the tumour response differed; the effect onset was slow in one case and a durable response was observed in the second case even after treatment discontinuation. Secondary malignant tumours occurred during treatment in both cases. This follow-up study revealed that ca-4F12-E6 maintains CR in dogs for more than 1 year. In addition, the pattern of tumour response was unique compared to conventional therapy. These results indicate that new evaluation criteria for tumour response may be necessary for immunotherapy in veterinary medicine. Long-term follow-up is necessary regardless of the short-term treatment responsiveness.
Collapse
Affiliation(s)
- Masaya Igase
- Laboratory of Molecular Diagnostics and Therapeutics, The United Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Sakuya Inanaga
- Laboratory of Molecular Diagnostics and Therapeutics, The United Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Kenji Tani
- Laboratory of Veterinary Surgery, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Munekazu Nakaichi
- Laboratory of Veterinary Radiology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Yusuke Sakai
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masashi Sakurai
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masahiro Kato
- Nippon Zenyaku Kogyo Co., Ltd., Koriyama, Fukushima, Japan
| | | | - Takuya Mizuno
- Laboratory of Molecular Diagnostics and Therapeutics, The United Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
14
|
Systemic CD4 Immunity and PD-L1/PD-1 Blockade Immunotherapy. Int J Mol Sci 2022; 23:ijms232113241. [PMID: 36362027 PMCID: PMC9655397 DOI: 10.3390/ijms232113241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
PD-L1/PD-1 blockade immunotherapy has changed the therapeutic approaches for the treatment of many cancers. Nevertheless, the mechanisms underlying its efficacy or treatment failure are still unclear. Proficient systemic immunity seems to be a prerequisite for efficacy, as recently shown in patients and in mouse models. It is widely accepted that expansion of anti-tumor CD8 T cell populations is principally responsible for anti-tumor responses. In contrast, the role of CD4 T cells has been less studied. Here we review and discuss the evidence supporting the contribution of CD4 T cells to anti-tumor immunity, especially recent advances linking CD4 T cell subsets to efficacious PD-L1/PD-1 blockade immunotherapy. We also discuss the role of CD4 T cell memory subsets present in peripheral blood before the start of immunotherapies, and their utility as predictors of response.
Collapse
|
15
|
Cao L, Cao Z, Liu H, Liang N, Bing Z, Tian C, Li S. Detection of Potential Mutated Genes Associated with Common Immunotherapy Biomarkers in Non-Small-Cell Lung Cancer Patients. Curr Oncol 2022; 29:5715-5730. [PMID: 36005189 PMCID: PMC9406727 DOI: 10.3390/curroncol29080451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Microsatellite instability (MSI), high tumor mutation burden (TMB-H) and programmed cell death 1 ligand 1 (PD-L1) expression are hot biomarkers related to the improvement of immunotherapy response. Two cohorts of non-small-cell lung cancer (NSCLC) were collected and sequenced via targeted next-generation sequencing. Drug analysis was then performed on the shared genes using three different databases: Drugbank, DEPO and DRUGSURV. A total of 27 common genes were mutated in at least two groups of TMB-H-, MSI- and PD-L1-positive groups. AKT1, SMAD4, SCRIB and AXIN2 were severally involved in PI3K-activated, transforming growth factor beta (TGF-β)-activated, Hippo-repressed and Wnt-repressed pathways. This study provides an understanding of the mutated genes related to the immunotherapy biomarkers of NSCLC.
Collapse
Affiliation(s)
- Lei Cao
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Zhili Cao
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Hongsheng Liu
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Naixin Liang
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Zhongxing Bing
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Caijuan Tian
- Tianjin Marvel Medical Laboratory, Tianjin Marvelbio Technology Co., Ltd., Tianjin 300381, China
| | - Shanqing Li
- Department of Thoracic Surgery, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
- Correspondence: ; Tel./Fax: +86-010-6915-2630
| |
Collapse
|
16
|
Sung E, Ko M, Won JY, Jo Y, Park E, Kim H, Choi E, Jung UJ, Jeon J, Kim Y, Ahn H, Choi DS, Choi S, Hong Y, Park H, Lee H, Son YG, Park K, Won J, Oh SJ, Lee S, Kim KP, Yoo C, Song HK, Jin HS, Jung J, Park Y. LAG-3xPD-L1 bispecific antibody potentiates antitumor responses of T cells through dendritic cell activation. Mol Ther 2022; 30:2800-2816. [PMID: 35526096 PMCID: PMC9372323 DOI: 10.1016/j.ymthe.2022.05.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/08/2022] [Accepted: 05/03/2022] [Indexed: 11/26/2022] Open
Abstract
Several preclinical studies demonstrate that antitumor efficacy of programmed cell death-1 (PD-1)/programmed death-ligand 1 (PD-L1) blockade can be improved by combination with other checkpoint inhibitors. Lymphocyte-activation gene 3 (LAG-3) is an inhibitory checkpoint receptor involved in T cell exhaustion and tumor immune escape. Here, we describe ABL501, a bispecific antibody targeting LAG-3 and PD-L1 in modulating immune cell responses against tumors. ABL501 that efficiently inhibits both LAG-3 and PD-L1 pathways enhances the activation of effector CD4+ and CD8+ T cells with a higher degree than a combination of single anti-LAG-3 and anti-PD-L1. The augmented effector T cell responses by ABL501 resulted in mitigating regulatory-T-cell-mediated immunosuppression. Mechanistically, the simultaneous binding of ABL501 to LAG-3 and PD-L1 promotes dendritic cell (DC) activation and tumor cell conjugation with T cells that subsequently mounts effective CD8+ T cell responses. ABL501 demonstrates its potent in vivo antitumor efficacy in a humanized xenograft model and with knockin mice expressing human orthologs. The immune profiling analysis of peripheral blood reveals an increased abundance of LAG-3hiPD-1hi memory CD4+ T cell subset in relapsed cholangiocarcinoma patients after gemcitabine plus cisplatin therapy, which are more responsive to ABL501. This study supports the clinical evaluation of ABL501 as a novel cancer immunotherapeutic, and a first-in-human trial has started (NCT05101109).
Collapse
Affiliation(s)
| | - Minkyung Ko
- Theragnosis Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, South Korea
| | - Ju-Young Won
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Yunju Jo
- Theragnosis Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, South Korea; Department of Life Sciences, Korea University, Seoul 02481, South Korea
| | | | | | - Eunji Choi
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | | | | | | | - Hyejin Ahn
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Da-Som Choi
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Seunghyun Choi
- Theragnosis Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, South Korea
| | | | | | | | | | | | | | - Soo Jin Oh
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Seonmin Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Hyun Kyu Song
- Department of Life Sciences, Korea University, Seoul 02481, South Korea
| | - Hyung-Seung Jin
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, South Korea.
| | - Jaeho Jung
- ABL Bio Inc., Seongnam 13488, South Korea.
| | - Yoon Park
- Theragnosis Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, South Korea.
| |
Collapse
|
17
|
Lv B, Wang Y, Ma D, Cheng W, Liu J, Yong T, Chen H, Wang C. Immunotherapy: Reshape the Tumor Immune Microenvironment. Front Immunol 2022; 13:844142. [PMID: 35874717 PMCID: PMC9299092 DOI: 10.3389/fimmu.2022.844142] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
Tumor immune microenvironment (TIME) include tumor cells, immune cells, cytokines, etc. The interactions between these components, which are divided into anti-tumor and pro-tumor, determine the trend of anti-tumor immunity. Although the immune system can eliminate tumor through the cancer-immune cycle, tumors appear to eventually evade from immune surveillance by shaping an immunosuppressive microenvironment. Immunotherapy reshapes the TIME and restores the tumor killing ability of anti-tumor immune cells. Herein, we review the function of immune cells within the TIME and discuss the contribution of current mainstream immunotherapeutic approaches to remolding the TIME. Changes in the immune microenvironment in different forms under the intervention of immunotherapy can shed light on better combination treatment strategies.
Collapse
Affiliation(s)
- Bingzhe Lv
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yunpeng Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Dongjiang Ma
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wei Cheng
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jie Liu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Tao Yong
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Hao Chen
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China.,Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Chen Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
18
|
Evaluating the utility of an immune checkpoint-related lncRNA signature for identifying the prognosis and immunotherapy response of lung adenocarcinoma. Sci Rep 2022; 12:12785. [PMID: 35896612 PMCID: PMC9329438 DOI: 10.1038/s41598-022-16715-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 07/14/2022] [Indexed: 12/03/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is the most frequent subtype of lung cancer globally. However, the survival rate of lung adenocarcinoma patients remains low. Immune checkpoints and long noncoding RNAs are emerging as vital tools for predicting the immunotherapeutic response and outcomes of patients with lung adenocarcinoma. It is critical to identify lncRNAs associated with immune checkpoints in lung adenocarcinoma patients. In this study, immune checkpoint-related lncRNAs (IClncRNAs) were analysed and identified by coexpression. Based on the immune checkpoint-related lncRNAs, we divided patients with lung adenocarcinoma into two clusters and constructed a risk model. Kaplan–Meier analysis, Gene Set Enrichment Analysis, and nomogram analysis of the 2 clusters and the risk model were performed. Finally, the potential immunotherapeutic prediction value of this model was discussed. The risk model consisting of 6 immune checkpoint-related lncRNAs was an independent predictor of survival. Through regrouping the patients with this model, we can distinguish between them more effectively in terms of their immunotherapeutic response, tumour microenvironment, and chemotherapy response. This risk model based on immune checkpoint-based lncRNAs may have an excellent clinical value for predicting the immunotherapeutic response and outcomes of patients with LUAD.
Collapse
|
19
|
Chen S, Xie P, Cowan M, Huang H, Cardenas H, Keathley R, Tanner EJ, Fleming GF, Moroney JW, Pant A, Akasha AM, Davuluri RV, Kocherginsky M, Zhang B, Matei D. Epigenetic priming enhances antitumor immunity in platinum-resistant ovarian cancer. J Clin Invest 2022; 132:e158800. [PMID: 35671108 PMCID: PMC9282926 DOI: 10.1172/jci158800] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/02/2022] [Indexed: 11/17/2022] Open
Abstract
BackgroundImmune checkpoint inhibitors (ICIs) have modest activity in ovarian cancer (OC). To augment their activity, we used priming with the hypomethylating agent guadecitabine in a phase II study.MethodsEligible patients had platinum-resistant OC, normal organ function, measurable disease, and received up to 5 prior regimens. The treatment included guadecitabine (30 mg/m2) on days 1-4, and pembrolizumab (200 mg i.v.) on day 5, every 21 days. The primary endpoint was the response rate. Tumor biopsies, plasma, and PBMCs were obtained at baseline and after treatment.ResultsAmong 35 evaluable patients, 3 patients had partial responses (8.6%), and 8 (22.9%) patients had stable disease, resulting in a clinical benefit rate of 31.4% (95% CI: 16.9%-49.3%). The median duration of clinical benefit was 6.8 months. Long-interspersed element 1 (LINE1) was hypomethylated in post-treatment PBMCs, and methylomic and transcriptomic analyses showed activation of antitumor immunity in post-treatment biopsies. High-dimensional immune profiling of PBMCs showed a higher frequency of naive and/or central memory CD4+ T cells and of classical monocytes in patients with a durable clinical benefit or response (CBR). A higher baseline density of CD8+ T cells and CD20+ B cells and the presence of tertiary lymphoid structures in tumors were associated with a durable CBR.ConclusionEpigenetic priming using a hypomethylating agent with an ICI was feasible and resulted in a durable clinical benefit associated with immune responses in selected patients with recurrent OC.Trial registrationClinicalTrials.gov NCT02901899.FundingUS Army Medical Research and Material Command/Congressionally Directed Medical Research Programs (USAMRMC/CDMRP) grant W81XWH-17-0141; the Diana Princess of Wales Endowed Professorship and LCCTRAC funds from the Robert H. Lurie Comprehensive Cancer Center; Walter S. and Lucienne Driskill Immunotherapy Research funds; Astex Pharmaceuticals; Merck & Co.; National Cancer Institute (NCI), NIH grants CCSG P30 CA060553, CCSG P30 CA060553, and CA060553.
Collapse
Affiliation(s)
- Siqi Chen
- Department of Medicine, Hematology/Oncology Division
| | - Ping Xie
- Department of Medicine, Hematology/Oncology Division
| | | | - Hao Huang
- Department of Obstetrics and Gynecology
| | | | - Russell Keathley
- Department of Obstetrics and Gynecology
- Driskill Graduate Training Program in Life Sciences, and
| | - Edward J. Tanner
- Department of Obstetrics and Gynecology
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Gini F. Fleming
- Department of Medicine, Hematology/Oncology Division, University of Chicago, Chicago, Illinois, USA
| | - John W. Moroney
- Department of Medicine, Hematology/Oncology Division, University of Chicago, Chicago, Illinois, USA
| | - Alok Pant
- Northwestern Medicine, Lake Forest Hospital, Lake Forest, Illinois, USA
| | - Azza M. Akasha
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ramana V. Davuluri
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York, USA
| | - Masha Kocherginsky
- Department of Obstetrics and Gynecology
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Division of Biostatistics, Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Bin Zhang
- Department of Medicine, Hematology/Oncology Division
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Daniela Matei
- Department of Obstetrics and Gynecology
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Jesse Brown VA Medical Center, Chicago, Illinois, USA
| |
Collapse
|
20
|
Zhang H, Liu M, Du G, Yu B, Ma X, Gui Y, Cao L, Li X, Tan B. Immune checkpoints related-LncRNAs can identify different subtypes of lung cancer and predict immunotherapy and prognosis. J Cancer Res Clin Oncol 2022; 148:1597-1612. [PMID: 35296921 DOI: 10.1007/s00432-022-03940-3] [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: 06/14/2021] [Accepted: 02/02/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Non-small cell lung cancer is the most common subtype of lung cancer in the world. However, the survival rate of non-small cell lung cancer patients remains low currently. Immune checkpoint and long non-coding RNAs are emerging as critical roles in prognostic significance and the immunotherapeutic response of non-small cell lung cancer. It is critical to discern LncRNAs related with immune checkpoints in patients with Non-small cell lung cancer. METHODS In this study, immune checkpoint-linked LncRNAs were determined and achieved by the co-expression analysis. Immune checkpoint-linked LncRNAs with noteworthy prognostic value (P < 0.05) gained were next utilized to separate into two cluster by non-negative matrix factorization (NMF). Univariate and a least absolute shrinkage and selection operator were applied to construct an immune checkpoint-linked LncRNAs model. Kaplan-Meier analysis, Gene Set Enrichment Analysis, and the nomogram were utilized to investigate the LncRNAs model. Lastly, the capability immunotherapy and chemotherapy prediction value of this risk model were also estimated. RESULTS The model consisting of ten immune checkpoint-related LncRNAs was acknowledged to be a self-determining predictor of prognosis. Through regrouping the NSCLC patients by this model, difference between them more efficiently on immunotherapeutic response, tumor microenvironment and chemotherapy response could be discovered. This risk model related to the immune checkpoint-based LncRNAs may have an excellent clinical prediction for prognosis and the immunotherapeutic response in patients with NSCLC. CONCLUSIONS We performed an integrative analysis of LncRNAs linked with immune checkpoints and emphasized the significance of NSCLC subtypes classification, immune checkpoints related LncRNAs in estimating the tumor microenvironment score, immune cell infiltration of the tumor, immunotherapy, and chemotherapy response.
Collapse
Affiliation(s)
- Hongpan Zhang
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, No. 1, Maoyuan south road, Shunqing District, Nanchong City, Sichuan Province, 637000, People's Republic of China
- North Sichuan Medical College, No. 55 Dongshun road, Gaoping district, Nanchong, Sichuan province, People's Republic of China
| | - Meihan Liu
- North Sichuan Medical College, No. 55 Dongshun road, Gaoping district, Nanchong, Sichuan province, People's Republic of China
| | - Guobo Du
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, No. 1, Maoyuan south road, Shunqing District, Nanchong City, Sichuan Province, 637000, People's Republic of China
- North Sichuan Medical College, No. 55 Dongshun road, Gaoping district, Nanchong, Sichuan province, People's Republic of China
| | - Bin Yu
- Guangyuan Central Hospital, No. 16 Jingxiangzi, Lizhou district, Guangyuan, Sichuan province, People's Republic of China
| | - Xiaojie Ma
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, No. 1, Maoyuan south road, Shunqing District, Nanchong City, Sichuan Province, 637000, People's Republic of China
- North Sichuan Medical College, No. 55 Dongshun road, Gaoping district, Nanchong, Sichuan province, People's Republic of China
| | - Yan Gui
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, No. 1, Maoyuan south road, Shunqing District, Nanchong City, Sichuan Province, 637000, People's Republic of China
- North Sichuan Medical College, No. 55 Dongshun road, Gaoping district, Nanchong, Sichuan province, People's Republic of China
| | - Lu Cao
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, No. 1, Maoyuan south road, Shunqing District, Nanchong City, Sichuan Province, 637000, People's Republic of China
- North Sichuan Medical College, No. 55 Dongshun road, Gaoping district, Nanchong, Sichuan province, People's Republic of China
| | - Xianfu Li
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, No. 1, Maoyuan south road, Shunqing District, Nanchong City, Sichuan Province, 637000, People's Republic of China.
- North Sichuan Medical College, No. 55 Dongshun road, Gaoping district, Nanchong, Sichuan province, People's Republic of China.
| | - Bangxian Tan
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, No. 1, Maoyuan south road, Shunqing District, Nanchong City, Sichuan Province, 637000, People's Republic of China.
- North Sichuan Medical College, No. 55 Dongshun road, Gaoping district, Nanchong, Sichuan province, People's Republic of China.
| |
Collapse
|
21
|
Kumagai S, Koyama S, Itahashi K, Tanegashima T, Lin YT, Togashi Y, Kamada T, Irie T, Okumura G, Kono H, Ito D, Fujii R, Watanabe S, Sai A, Fukuoka S, Sugiyama E, Watanabe G, Owari T, Nishinakamura H, Sugiyama D, Maeda Y, Kawazoe A, Yukami H, Chida K, Ohara Y, Yoshida T, Shinno Y, Takeyasu Y, Shirasawa M, Nakama K, Aokage K, Suzuki J, Ishii G, Kuwata T, Sakamoto N, Kawazu M, Ueno T, Mori T, Yamazaki N, Tsuboi M, Yatabe Y, Kinoshita T, Doi T, Shitara K, Mano H, Nishikawa H. Lactic acid promotes PD-1 expression in regulatory T cells in highly glycolytic tumor microenvironments. Cancer Cell 2022; 40:201-218.e9. [PMID: 35090594 DOI: 10.1016/j.ccell.2022.01.001] [Citation(s) in RCA: 368] [Impact Index Per Article: 122.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/07/2021] [Accepted: 01/05/2022] [Indexed: 12/13/2022]
Abstract
The balance of programmed death-1 (PD-1)-expressing CD8+ T cells and regulatory T (Treg) cells in the tumor microenvironment (TME) determines the clinical efficacy of PD-1 blockade therapy through the competition of their reactivation. However, factors that determine this balance remain unknown. Here, we show that Treg cells gain higher PD-1 expression than effector T cells in highly glycolytic tumors, including MYC-amplified tumors and liver tumors. Under low-glucose environments via glucose consumption by tumor cells, Treg cells actively absorbed lactic acid (LA) through monocarboxylate transporter 1 (MCT1), promoting NFAT1 translocation into the nucleus, thereby enhancing the expression of PD-1, whereas PD-1 expression by effector T cells was dampened. PD-1 blockade invigorated the PD-1-expressing Treg cells, resulting in treatment failure. We propose that LA in the highly glycolytic TME is an active checkpoint for the function of Treg cells in the TME via upregulation of PD-1 expression.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cell Line, Tumor
- Disease Models, Animal
- Fluorescent Antibody Technique
- Gene Expression Regulation, Neoplastic/drug effects
- Glycolysis
- Humans
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Immune Checkpoint Proteins/metabolism
- Immunophenotyping
- Lactic Acid/metabolism
- Lactic Acid/pharmacology
- Lymphocyte Activation
- Lymphocyte Count
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Mice
- Molecular Targeted Therapy
- Prognosis
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Treatment Outcome
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
Collapse
Affiliation(s)
- Shogo Kumagai
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo 104-0045, Japan; Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan; Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shohei Koyama
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan; Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan.
| | - Kota Itahashi
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Tokiyoshi Tanegashima
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Yi-Tzu Lin
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan; Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yosuke Togashi
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Takahiro Kamada
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Takuma Irie
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Genki Okumura
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Hidetoshi Kono
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Daisuke Ito
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Rika Fujii
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Sho Watanabe
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Atsuo Sai
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan; Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shota Fukuoka
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Eri Sugiyama
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Go Watanabe
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Takuya Owari
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Hitomi Nishinakamura
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Daisuke Sugiyama
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yuka Maeda
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan
| | - Akihito Kawazoe
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Hiroki Yukami
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Keigo Chida
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Yuuki Ohara
- Pathology and Clinical Laboratories, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yuki Shinno
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yuki Takeyasu
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Masayuki Shirasawa
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Kenta Nakama
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Keiju Aokage
- Department of Thoracic Surgery, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Jun Suzuki
- Department of Thoracic Surgery, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Genichiro Ishii
- Pathology and Clinical Laboratories, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Takeshi Kuwata
- Pathology and Clinical Laboratories, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Naoya Sakamoto
- Pathology and Clinical Laboratories, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Masahito Kawazu
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Toshihide Ueno
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Taisuke Mori
- Department of Pathology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Masahiro Tsuboi
- Department of Thoracic Surgery, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Yasushi Yatabe
- Department of Pathology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Takahiro Kinoshita
- Department of Gastric Surgery, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Toshihiko Doi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045/Chiba 277-8577, Japan; Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
22
|
Shi J, Liu J, Tu X, Li B, Tong Z, Wang T, Zheng Y, Shi H, Zeng X, Chen W, Yin W, Fang W. Single-cell immune signature for detecting early-stage HCC and early assessing anti-PD-1 immunotherapy efficacy. J Immunother Cancer 2022; 10:jitc-2021-003133. [PMID: 35101942 PMCID: PMC8804705 DOI: 10.1136/jitc-2021-003133] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2021] [Indexed: 12/30/2022] Open
Abstract
Background The early diagnosis of hepatocellular carcinoma (HCC) can greatly improve patients’ 5-year survival rate, and the early efficacy assessment is important for oncologists to harness the anti-programmed cell death protein 1 (PD-1) immunotherapy in patients with advanced HCC. The lack of effective predicting biomarkers not only leads to delayed detection of the disease but also results in ineffective immunotherapy and limited clinical survival benefit. Methods We exploited the single-cell approach (cytometry by time of flight (CyTOF)) to analyze peripheral blood mononuclear cells from multicohorts of human samples. Immune signatures for different stages of patients with HCC were systematically profiled and statistically compared. Furthermore, the dynamic changes of peripheral immune compositions for both first-line and second-line patients with HCC after anti-PD-1 monotherapy were also evaluated and systematically compared. Results We identified stage-specific immune signatures for HCC and constructed a logistic AdaBoost-SVM classifier based on these signatures. The classifier provided superior performance in predicting early-stage HCC over the commonly used serum alpha-fetoprotein level. We also revealed the treatment stage-specific immune signatures from peripheral blood and their dynamical changing patterns, all of which were integrated to achieve early discrimination of patients with non-durable benefit for both first-line and second-line anti-PD-1 monotherapies. Conclusions Our newly identified single-cell peripheral immune signatures provide promising non-invasive biomarkers for early detection of HCC and early assessment for anti-PD-1 immunotherapy efficacy in patients with advanced HCC. These new findings can potentially facilitate early diagnosis and novel immunotherapy for patients with HCC in future practice and further guide the utility of CyTOF in clinical translation of cancer research. Trial registration numbers NCT02576509 and NCT02989922.
Collapse
Affiliation(s)
- Jiawei Shi
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, People's Republic of China.,Departments of Cell Biology and Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Junwei Liu
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, People's Republic of China.,Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Xiaoxuan Tu
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou, People's Republic of China
| | - Bin Li
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou, People's Republic of China
| | - Zhou Tong
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Tian Wang
- Zhejiang Puluoting Health Technology Co., Ltd, Hangzhou, People's Republic of China
| | - Yi Zheng
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Zhejiang University, Hangzhou, People's Republic of China
| | - Hongyu Shi
- Zhejiang Puluoting Health Technology Co., Ltd, Hangzhou, People's Republic of China
| | - Xun Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Wei Chen
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, People's Republic of China .,Departments of Cell Biology and Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Weiwei Yin
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, People's Republic of China .,Department of Thoracic Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, People's Republic of China.,Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, College of Biomedical Engineering and Instrument of Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou, People's Republic of China
| |
Collapse
|
23
|
T cell subtype profiling measures exhaustion and predicts anti-PD-1 response. Sci Rep 2022; 12:1342. [PMID: 35079117 PMCID: PMC8789795 DOI: 10.1038/s41598-022-05474-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/05/2022] [Indexed: 12/11/2022] Open
Abstract
Anti-PD-1 therapy can provide long, durable benefit to a fraction of patients. The on-label PD-L1 test, however, does not accurately predict response. To build a better biomarker, we created a method called T Cell Subtype Profiling (TCSP) that characterizes the abundance of T cell subtypes (TCSs) in FFPE specimens using five RNA models. These TCS RNA models are created using functional methods, and robustly discriminate between naïve, activated, exhausted, effector memory, and central memory TCSs, without the reliance on non-specific, classical markers. TCSP is analytically valid and corroborates associations between TCSs and clinical outcomes. Multianalyte biomarkers based on TCS estimates predicted response to anti-PD-1 therapy in three different cancers and outperformed the indicated PD-L1 test, as well as Tumor Mutational Burden. Given the utility of TCSP, we investigated the abundance of TCSs in TCGA cancers and created a portal to enable researchers to discover other TCSP-based biomarkers.
Collapse
|
24
|
Loo K, Smithy JW, Postow MA, Betof Warner A. Factors Determining Long-Term Antitumor Responses to Immune Checkpoint Blockade Therapy in Melanoma. Front Immunol 2022; 12:810388. [PMID: 35087529 PMCID: PMC8787112 DOI: 10.3389/fimmu.2021.810388] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
With the increasing promise of long-term survival with immune checkpoint blockade (ICB) therapies, particularly for patients with advanced melanoma, clinicians and investigators are driven to identify prognostic and predictive factors that may help to identify individuals who are likely to experience durable benefit. Several ICB combinations are being actively developed to expand the armamentarium of treatments for patients who may not achieve long-term responses to ICB single therapies alone. Thus, negative predictive markers are also of great interest. This review seeks to deepen our understanding of the mechanisms underlying the durability of ICB treatments. We will discuss the currently available long-term data from the ICB clinical trials and real-world studies describing the survivorship of ICB-treated melanoma patients. Additionally, we explore the current treatment outcomes in patients rechallenged with ICB and the patterns of ICB resistance based on sites of disease, namely, liver or CNS metastases. Lastly, we discuss the landscape in melanoma in the context of prognostic or predictive factors as markers of long-term response to ICB.
Collapse
Affiliation(s)
- Kimberly Loo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Internal Medicine, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY, United States
| | - James W. Smithy
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Michael A. Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Allison Betof Warner
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
25
|
Wu J, Zhang T, Xiong H, Zeng L, Wang Z, Peng Y, Chen W, Hu X, Su T. Tumor-Infiltrating CD4 + Central Memory T Cells Correlated with Favorable Prognosis in Oral Squamous Cell Carcinoma. J Inflamm Res 2022; 15:141-152. [PMID: 35035226 PMCID: PMC8754505 DOI: 10.2147/jir.s343432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) is the most frequent oral malignancy with a poor prognosis, in which tumor-infiltrating immune cells may play a critical role. Therefore, our study aims to screen potential immune cells and immune-related genes for predicting OSCC prognosis. METHODS A total of 310 OSCC patients with full transcriptional data and clinical characteristics were extracted from the TCGA database. Then, we obtained their abundance of tumor-infiltrating immune cells on TIMER 2.0 and analyzed them using xCell method. Univariate and multivariate Cox regressions were applied successively to identify the immune cells associated with overall survival of OSCC patients. Furthermore, we screened the prognostic genes that related to the identified immune cells and validated their expressions by immunohistochemistry. RESULTS CD4+ central memory T (TCM) cell was recognized as the sole independent immune cell correlated with OSCC prognosis (p = 0.0085). A novel nomogram based on CD4+ TCM cell abundance was established for predicting the prognosis of OSCC patients, with calibration plots showing good performance for 1-, 3-, 5-year overall survival. Thirty-four related prognostic genes were screened according to the differential abundance of CD4+ TCM cell infiltration. In immunohistochemistry analysis, DEFB1 showed a significant positive relationship with the density of CD4+ TCM cells (p = 0.0075). CONCLUSION CD4+ central memory T cell was proposed as an independent prognostic biomarker for OSCC patients. DEFB1 might positively regulate the abundance of tumor-infiltrating CD4+ TCM cells, thus improving OSCC prognosis. Our findings may provide a new insight into better prognosis prediction and precise medicine for OSCC.
Collapse
Affiliation(s)
- Jin Wu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Tianyi Zhang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Haofeng Xiong
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Liujun Zeng
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Zijia Wang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Ying Peng
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Weijun Chen
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Xin Hu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Tong Su
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People’s Republic of China
- Institute of Oral Cancer and Precancerous Lesions, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| |
Collapse
|
26
|
Fujioka Y, Sugiyama D, Matsumura I, Minami Y, Miura M, Atsuta Y, Ohtake S, Kiyoi H, Miyazaki Y, Nishikawa H, Takahashi N. Regulatory T Cell as a Biomarker of Treatment-Free Remission in Patients with Chronic Myeloid Leukemia. Cancers (Basel) 2021; 13:5904. [PMID: 34885012 PMCID: PMC8657169 DOI: 10.3390/cancers13235904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 01/09/2023] Open
Abstract
Treatment-free remission (TFR) has become a therapeutic goal in chronic myeloid leukemia (CML), and approximately half of the patients with chronic phase-CML (CML-CP) with deep molecular remission (DMR) by tyrosine-kinase inhibitors (TKIs) have achieved TFR. However, the mechanism of continuous TFR is still unclear, as there are "fluctuate" patients who have BCR-ABL-positive leukemia cells but do not observe obvious relapse. We focused on the immune response and conducted an immune analysis using clinical samples from the imatinib discontinuation study, JALSG-STIM213. The results showed that, in the group that maintained TFR for 3 years, changes in regulatory T (Treg) cells were observed early after stopping imatinib treatment. The effector Treg (eTreg) cells increased transiently at 1 month after stopping imatinib and then returned to baseline at 3 months after stopping imatinib treatment. There was no difference in the Treg phenotype, and CD8+ T cells in the TFR group were relatively activated. High concentrations of imatinib before stopping were negatively correlated with eTreg cells after stopping imatinib. These data suggest immunological involvement in the maintenance of the TFR, and that Treg cells after stopping imatinib might be a biomarker for TFR. Furthermore, high imatinib exposure may have a negative immunological impact on the continuous TFR.
Collapse
Affiliation(s)
- Yuki Fujioka
- Department of Hematology, Nephrology and Rheumatology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan;
| | - Daisuke Sugiyama
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan;
| | - Itaru Matsumura
- Department of Hematology and Oncology, Kinki University Hospital, Osaka 589-8511, Japan;
| | - Yosuke Minami
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-0882, Japan;
| | - Masatomo Miura
- Department of Pharmacy, Akita University Hospital, Akita 010-8543, Japan;
| | - Yoshiko Atsuta
- The Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya 461-0047, Japan;
| | | | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University, Nagoya 464-8601, Japan;
| | - Yasushi Miyazaki
- Department of Hematology, Nagasaki University, Nagasaki 852-8521, Japan;
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan;
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan;
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
| | - Naoto Takahashi
- Department of Hematology, Nephrology and Rheumatology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan
| |
Collapse
|
27
|
Takahashi H, Sakakura K, Ida S, Kawabata-Iwakawa R, Matsuyama T, Tada H, Mito I, Chikamatsu K. Circulating naïve and effector memory T cells correlate with prognosis in head and neck squamous cell carcinoma. Cancer Sci 2021; 113:53-64. [PMID: 34751489 PMCID: PMC8748237 DOI: 10.1111/cas.15195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 12/01/2022] Open
Abstract
T‐cell memory is an important mechanism for long‐term protection against diverse pathogens. Generation and persistence of memory T cells are vital components of anti‐tumor immunity, given their ability to persist for prolonged durations, as well as activate and migrate rapidly. In the present study, we investigated the clinical and prognostic significance of T‐cell subsets in the peripheral circulation of patients with head and neck squamous cell carcinoma (HNSCC). Moreover, we calculated the enrichment scores of T‐cell subsets in primary tumor tissues and compared their clinical characteristics using a public database. Multivariate survival analyses of circulating T‐cell parameters revealed that clinical parameters, except M factor, were not independent prognostic factors, whereas proportions of CD8+ T cells, naïve T cells (TNs), effector memory T cells (TEMs), and CD38+CD8+ T cells were independent prognostic factors, suggesting the importance of these peripheral T‐cell parameters as independent prognostic biomarkers. Consistent with these results, the T‐cell enrichment analysis indicated that enrichment of CD8+ TNs in the tumor microenvironment was an independent prognostic factor. Moreover, an ex vivo experiment demonstrated significantly less cytotoxic activity in CD38+ T cells than in CD38− T cells. These findings suggest that T‐cell memory‐related parameters in both systemic immunity and the tumor microenvironment could be used as prognostic biomarkers regardless of clinical characteristics. Further characterization of circulating T cells would lead to the development of novel biomarkers for patients with HNSCC.
Collapse
Affiliation(s)
- Hideyuki Takahashi
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Koichi Sakakura
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Shota Ida
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, Maebashi, Japan
| | - Toshiyuki Matsuyama
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hiroe Tada
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ikko Mito
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kazuaki Chikamatsu
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
28
|
Yasuda Y, Iwama S, Sugiyama D, Okuji T, Kobayashi T, Ito M, Okada N, Enomoto A, Ito S, Yan Y, Sugiyama M, Onoue T, Tsunekawa T, Ito Y, Takagi H, Hagiwara D, Goto M, Suga H, Banno R, Takahashi M, Nishikawa H, Arima H. CD4 + T cells are essential for the development of destructive thyroiditis induced by anti-PD-1 antibody in thyroglobulin-immunized mice. Sci Transl Med 2021; 13:13/593/eabb7495. [PMID: 33980577 DOI: 10.1126/scitranslmed.abb7495] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 01/07/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022]
Abstract
Immune-related adverse events induced by anti-programmed cell death-1 antibodies (PD-1-Ab), including destructive thyroiditis (thyroid-irAE), are thought to be caused by activated T cells. However, the T cell subsets that are directly responsible for damaging self-organs remain unclear. To clarify which T cell subsets are involved in the development of thyroid-irAE, a mouse model of thyroid-irAE was analyzed. PD-1-Ab administration 2.5 months after immunization with thyroglobulin caused destructive thyroiditis. Thyroiditis was completely prevented by previous depletion of CD4+ T cells and partially prevented by depleting CD8+ T cells. The frequencies of central and effector memory CD4+ T cell subsets and the secretion of interferon-γ after stimulation with thyroglobulin were increased in the cervical lymph nodes of mice with thyroid-irAE compared with controls. Histopathological analysis revealed infiltration of CD4+ T cells expressing granzyme B in thyroid glands and major histocompatibility complex class II expression on thyrocytes in mice with thyroid-irAE. Adoptive transfer of CD4+ T cells from cervical lymph nodes in mice with thyroid-irAE caused destruction of thyroid follicular architecture in the irradiated recipient mice. Flow cytometric analyses showed that the frequencies of central and effector memory CD4+ T cells expressing the cytotoxic marker CD27 were higher in peripheral blood mononuclear cells collected from patients with thyroid-irAE induced by PD-1-Ab versus those without. These data suggest a critical role for cytotoxic memory CD4+ T cells activated by PD-1-Ab in the pathogenesis of thyroid-irAE.
Collapse
Affiliation(s)
- Yoshinori Yasuda
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Daisuke Sugiyama
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Takayuki Okuji
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Tomoko Kobayashi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Masaaki Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Norio Okada
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Sachiko Ito
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yue Yan
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Taku Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yoshihiro Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.,Department of CKD Initiatives/Nephrology, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
| | - Hiroshi Takagi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Motomitsu Goto
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.,Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya 464-8601, Japan
| | - Masahide Takahashi
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.,Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Tokyo 104-0045, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
29
|
Jin P, Li J, Meng Y, Wu L, Bai M, Yu J, Meng X. PET/CT metabolic patterns in systemic immune activation: A new perspective on the assessment of immunotherapy response and efficacy. Cancer Lett 2021; 520:91-99. [PMID: 34237407 DOI: 10.1016/j.canlet.2021.06.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/24/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023]
Abstract
Despite advances in immunotherapy, extensive challenges remain in its clinical application. Positron emission tomography (PET)/computed tomography (CT) is widely used in the diagnosis and follow-up of malignant tumors and in the prediction of treatment outcomes. Successful cancer immunotherapy requires systemic immune activation. In addition to local immune responses, a systemic antitumor response involving primary and secondary lymphoid organs is required for tumor eradication. Immune-related adverse events (IRAEs) are considered to be a manifestation of excessive immune activation. PET/CT can monitor the metabolic changes in peripheral lymphoid organs and related organs. Thus, it can identify patients with effective immune activation and predict the efficacy and outcomes of immunotherapy. This review aimed to investigate the theoretical basis and feasibility of applying PET/CT for monitoring the immune activation status of peripheral lymphoid organs after immunotherapy and predict its effectiveness. Towards this goal, we reviewed the cellular components and structural composition of peripheral lymphoid organs, as well as their functions in the systemic immune response. We analyzed the theoretical basis and feasibility of applying PET/CT to monitor the immune activation status of peripheral lymphoid organs after immunotherapy to predict the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Peng Jin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jianing Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yingtao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Leilei Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Department of Radiation Oncology, School of Medicine, Shandong University, Jinan, China
| | - Menglin Bai
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Department of Radiation Oncology, School of Medicine, Shandong University, Jinan, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
30
|
Early memory differentiation and cell death resistance in T cells predicts melanoma response to sequential anti-CTLA4 and anti-PD1 immunotherapy. Genes Immun 2021; 22:108-119. [PMID: 34079092 DOI: 10.1038/s41435-021-00138-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/11/2021] [Accepted: 05/21/2021] [Indexed: 11/09/2022]
Abstract
Immune checkpoint blockers (ICBs)-based immunotherapy has revolutionised oncology. However, the benefits of ICBs are limited to only a subset of patients. Herein, the biomarkers-driven application of ICBs promises to increase their efficacy. Such biomarkers include lymphocytic IFNγ-signalling and/or cytolytic activity (granzymes and perforin-1) footprints, whose levels in pre-treatment tumours can predict favourable patient survival following ICB-treatment. However, it is not clear whether such biomarkers have the same value in predicting survival of patients receiving first-line anti-CTLA4 ICB-therapy, and subsequently anti-PD1 ICB-therapy (i.e., sequential ICB-immunotherapy regimen). To address this, we applied highly integrated systems/computational immunology approaches to existing melanoma bulk-tumour transcriptomic and single-cell (sc)RNAseq data originating from immuno-oncology clinical studies applying ICB-treatment. Interestingly, we observed that CD8+/CD4+T cell-associated IFNγ-signalling or cytolytic activity signatures fail to predict tumour response in patients treated with anti-CTLA4 ICB-therapy as a first-line and anti-PD1 ICB-therapy in the second-line setting. On the contrary, signatures associated with early memory CD8+/CD4+T cells (integrating TCF1-driven stem-like transcriptional programme), capable of resisting cell death/apoptosis, better predicted objective response rates to ICB-immunotherapy, and favourable survival in the setting of sequential ICB-immunotherapy. These observations suggest that sequencing of ICB-therapy might have a specific impact on the T cell-repertoire and may influence the predictive value of tumoural immune biomarkers.
Collapse
|
31
|
Bai Z, Su G, Fan R. Single-cell Analysis Technologies for Immuno-oncology Research: from Mechanistic Delineation to Biomarker Discovery. GENOMICS, PROTEOMICS & BIOINFORMATICS 2021; 19:191-207. [PMID: 34000441 PMCID: PMC8602396 DOI: 10.1016/j.gpb.2021.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 12/10/2020] [Accepted: 03/06/2021] [Indexed: 11/29/2022]
Abstract
The successes with immune checkpoint blockade (ICB) and chimeric antigen receptor (CAR)-T-cell therapy in treating multiple cancer types have established immunotherapy as a powerful curative option for patients with advanced cancers. Unfortunately, many patients do not derive benefit or long-term responses, highlighting a pressing need to perform complete investigation of the underlying mechanisms and the immunotherapy-induced tumor regression or rejection. In recent years, a large number of single-cell technologies have leveraged advances in characterizing immune system, profiling tumor microenvironment, and identifying cellular heterogeneity, which establish the foundations for lifting the veil on the comprehensive crosstalk between cancer and immune system during immunotherapies. In this review, we introduce the applications of the most widely used single-cell technologies in furthering our understanding of immunotherapies in terms of underlying mechanisms and their association with therapeutic outcomes. We also discuss how single-cell analyses help to deliver new insights into biomarker discovery to predict patient response rate, monitor acquired resistance, and support prophylactic strategy development for toxicity management. Finally, we provide an overview of applying cutting-edge single-cell spatial-omics to point out the heterogeneity of tumor-immune interactions at higher level that can ultimately guide to the rational design of next-generation immunotherapies.
Collapse
Affiliation(s)
- Zhiliang Bai
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA; State Key Laboratory of Precision Measurement Technology and Instrument, Tianjin University, Tianjin 300072, China
| | - Graham Su
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA; Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT 06511, USA; Human and Translational Immunology, Yale School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
32
|
Araujo B de Lima V, Hansen M, Spanggaard I, Rohrberg K, Reker Hadrup S, Lassen U, Svane IM. Immune Cell Profiling of Peripheral Blood as Signature for Response During Checkpoint Inhibition Across Cancer Types. Front Oncol 2021; 11:558248. [PMID: 33842304 PMCID: PMC8027233 DOI: 10.3389/fonc.2021.558248] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Despite encouraging results with immune checkpoint inhibition (ICI), a large fraction of cancer patients still does not achieve clinical benefit. Finding predictive markers in the complexity of the tumor microenvironment is a challenging task and often requires invasive procedures. In our study, we looked for putative variables related to treatment benefit among immune cells in peripheral blood across different tumor types treated with ICIs. For that, we included 33 patients with different solid tumors referred to our clinical unit for ICI. Peripheral blood mononuclear cells were isolated at baseline, 6 and 20 weeks after treatment start. Characterization of immune cells was carried out by multi-color flow cytometry. Response to treatment was assessed radiologically by RECIST 1.1. Clinical outcome correlated with a shift towards an effector-like T cell phenotype, PD-1 expression by CD8+T cells, low levels of myeloid-derived suppressor cells and classical monocytes. Dendritic cells seemed also to play a role in terms of survival. From these findings, we hypothesized that patients responding to ICI had already at baseline an immune profile, here called ‘favorable immune periphery’, providing a higher chance of benefitting from ICI. We elaborated an index comprising cell types mentioned above. This signature correlated positively with the likelihood of benefiting from the treatment and ultimately with longer survival. Our study illustrates that patients responding to ICI seem to have a pre-existing immune profile in peripheral blood that favors good outcome. Exploring this signature can help to identify patients likely to achieve benefit from ICI.
Collapse
Affiliation(s)
| | - Morten Hansen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Iben Spanggaard
- Department of Oncology, Phase 1 Unit, Rigshospitalet, Copenhagen, Denmark
| | | | - Sine Reker Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Ulrik Lassen
- Department of Oncology, Phase 1 Unit, Rigshospitalet, Copenhagen, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| |
Collapse
|
33
|
Hong A, Piva M, Liu S, Hugo W, Lomeli SH, Zoete V, Randolph CE, Yang Z, Wang Y, Lee JJ, Lo SJ, Sun L, Vega-Crespo A, Garcia AJ, Shackelford DB, Dubinett SM, Scumpia PO, Byrum SD, Tackett AJ, Donahue TR, Michielin O, Holmen SL, Ribas A, Moriceau G, Lo RS. Durable Suppression of Acquired MEK Inhibitor Resistance in Cancer by Sequestering MEK from ERK and Promoting Antitumor T-cell Immunity. Cancer Discov 2021; 11:714-735. [PMID: 33318037 PMCID: PMC7933113 DOI: 10.1158/2159-8290.cd-20-0873] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/05/2020] [Accepted: 11/23/2020] [Indexed: 12/19/2022]
Abstract
MAPK targeting in cancer often fails due to MAPK reactivation. MEK inhibitor (MEKi) monotherapy provides limited clinical benefits but may serve as a foundation for combination therapies. Here, we showed that combining a type II RAF inhibitor (RAFi) with an allosteric MEKi durably prevents and overcomes acquired resistance among cancers with KRAS, NRAS, NF1, BRAF non-V600, and BRAF V600 mutations. Tumor cell-intrinsically, type II RAFi plus MEKi sequester MEK in RAF complexes, reduce MEK/MEK dimerization, and uncouple MEK from ERK in acquired-resistant tumor subpopulations. Immunologically, this combination expands memory and activated/exhausted CD8+ T cells, and durable tumor regression elicited by this combination requires CD8+ T cells, which can be reinvigorated by anti-PD-L1 therapy. Whereas MEKi reduces dominant intratumoral T-cell clones, type II RAFi cotreatment reverses this effect and promotes T-cell clonotypic expansion. These findings rationalize the clinical development of type II RAFi plus MEKi and their further combination with PD-1/L1-targeted therapy. SIGNIFICANCE: Type I RAFi + MEKi are indicated only in certain BRAF V600MUT cancers. In contrast, type II RAFi + MEKi are durably active against acquired MEKi resistance across broad cancer indications, which reveals exquisite MAPK addiction. Allosteric modulation of MAPK protein/protein interactions and temporal preservation of intratumoral CD8+ T cells are mechanisms that may be further exploited.This article is highlighted in the In This Issue feature, p. 521.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm/drug effects
- Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors
- GTP Phosphohydrolases/genetics
- GTP Phosphohydrolases/metabolism
- Humans
- Immunity, Cellular/drug effects
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors
- Mutation
- Neoplasms/drug therapy
- Neoplasms/etiology
- Neoplasms/metabolism
- Neoplasms/pathology
- Protein Binding
- Protein Kinase Inhibitors/pharmacology
- Protein Stability
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Treatment Outcome
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Aayoung Hong
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Marco Piva
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Sixue Liu
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Willy Hugo
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Shirley H Lomeli
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Vincent Zoete
- Department of Fundamental Oncology, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | - Zhentao Yang
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Yan Wang
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Jordan J Lee
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Skylar J Lo
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Lu Sun
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Agustin Vega-Crespo
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Alejandro J Garcia
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - David B Shackelford
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Steven M Dubinett
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Philip O Scumpia
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California
- Department of Dermatology, Veteran's Administration Greater Los Angeles Healthcare System, Los Angeles, California
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Timothy R Donahue
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Olivier Michielin
- Department of Oncology, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Sheri L Holmen
- Huntsman Cancer Institute and Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Gatien Moriceau
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California.
| | - Roger S Lo
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California
| |
Collapse
|
34
|
Lee L, Alrasheed N, Khandelwal G, Fitzsimons E, Richards H, Wilson W, Chavda SJ, Henry J, Conde L, De Massy MR, Chin M, Galas-Filipowicz D, Herrero J, Chain B, Quezada SA, Yong K. Increased Immune-Regulatory Receptor Expression on Effector T Cells as Early Indicators of Relapse Following Autologous Stem Cell Transplantation for Multiple Myeloma. Front Immunol 2021; 12:618610. [PMID: 33717112 PMCID: PMC7946836 DOI: 10.3389/fimmu.2021.618610] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 01/21/2021] [Indexed: 12/18/2022] Open
Abstract
The benefit of autologous stem cell transplantation (ASCT) in newly diagnosed myeloma patients, apart from supporting high dose chemotherapy, may include effects on T cell function in the bone marrow (BM). We report our exploratory findings on marrow infiltrating T cells early post-ASCT (day+100), examining phenotype and T cell receptor (TCR) repertoire, seeking correlations with timing of relapse. Compared to healthy donors (HD), we observed an increase in regulatory T cells (CD4+FoxP3+, Tregs) with reduction in CD4 T cells, leading to lower CD4:8 ratios. Compared to paired pre-treatment marrow, both CD4 and CD8 compartments showed a reduction in naïve, and increase in effector memory subsets, suggestive of a more differentiated phenotype. This was supported by increased levels of several immune-regulatory and activation proteins (ICOS, PD-1, LAG-3, CTLA-4 and GzmB) when compared with HD. Unsupervised analysis identified a patient subgroup with shorter PFS (p=0.031) whose BM contained increased Tregs, and higher immune-regulatory markers (ICOS, PD-1, LAG-3) on effector T cells. Using single feature analysis, higher frequencies of marrow PD-1+ on CD4+FoxP3- cells and Ki67+ on CD8 cells were independently associated with early relapse. Finally, studying paired pre-treatment and post-ASCT BM (n=5), we note reduced abundance of TCR sequences at day+100, with a greater proportion of expanded sequences indicating a more focused persistent TCR repertoire. Our findings indicate that, following induction chemotherapy and ASCT, marrow T cells demonstrate increased activation and differentiation, with TCR repertoire focusing. Pending confirmation in larger series, higher levels of immune-regulatory proteins on T cell effectors at day+100 may indicate early relapse.
Collapse
Affiliation(s)
- Lydia Lee
- Research Department of Hematology, Cancer Institute, University College London, London, United Kingdom
| | - Nouf Alrasheed
- Research Department of Hematology, Cancer Institute, University College London, London, United Kingdom
| | - Garima Khandelwal
- Bill Lyons Informatics Centre, Cancer Institute, University College London, London, United Kingdom
| | - Evelyn Fitzsimons
- Research Department of Hematology, Cancer Institute, University College London, London, United Kingdom
| | - Huw Richards
- Research Department of Hematology, Cancer Institute, University College London, London, United Kingdom
| | - William Wilson
- Cancer Research UK & UCL Cancer Trials Centre, London, United Kingdom
| | - Selina J. Chavda
- Research Department of Hematology, Cancer Institute, University College London, London, United Kingdom
| | - Jake Henry
- Research Department of Hematology, Cancer Institute, University College London, London, United Kingdom
- Cancer Immunology Unit, Research Department of Hematology, University College London Cancer Institute, London, United Kingdom
| | - Lucia Conde
- Bill Lyons Informatics Centre, Cancer Institute, University College London, London, United Kingdom
| | - Marc Robert De Massy
- Research Department of Hematology, Cancer Institute, University College London, London, United Kingdom
- Cancer Immunology Unit, Research Department of Hematology, University College London Cancer Institute, London, United Kingdom
- Department of Immunology, University College London, London, United Kingdom
| | - Melody Chin
- Research Department of Hematology, Cancer Institute, University College London, London, United Kingdom
| | - Daria Galas-Filipowicz
- Research Department of Hematology, Cancer Institute, University College London, London, United Kingdom
| | - Javier Herrero
- Bill Lyons Informatics Centre, Cancer Institute, University College London, London, United Kingdom
| | - Benny Chain
- Department of Immunology, University College London, London, United Kingdom
| | - Sergio A. Quezada
- Research Department of Hematology, Cancer Institute, University College London, London, United Kingdom
- Cancer Immunology Unit, Research Department of Hematology, University College London Cancer Institute, London, United Kingdom
| | - Kwee Yong
- Research Department of Hematology, Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
35
|
Saito R, Sawada Y, Nakamura M. Immune Profile Analysis in Peripheral Blood and Tumor in Patients with Malignant Melanoma. Int J Mol Sci 2021; 22:ijms22041957. [PMID: 33669410 PMCID: PMC7920420 DOI: 10.3390/ijms22041957] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Melanoma is a severe and life-threatening malignancy derived from melanocytes. The traditional treatment for melanoma could not sustain satisfactory outcomes long term; however, the recent immune checkpoint treatment has made a breakthrough in these problems. Nivolumab is a representative immune checkpoint treatment, and this PD-1-targeted therapy has evolutionally developed and improved the clinical outcome in a recent decade. On the other hand, the clinical application of immune checkpoint treatment presents clinicians with novel questions, especially how to obtain additional efficacy and overcome the disadvantage by using this treatment. To answer these problems, we first investigated the distribution of PD-L1 in various organs to clarify the organs most affected by anti-PD-1 antibody treatment. Among various organs, lung, placenta, spleen, heart, and thyroid highly expressed PD-L1, while skin, thalamus, hippocampus, ovary, stomach, testis, and prostate showed lower expressions of PD-L1. Furthermore, the immune profiles were also examined in tumors and peripheral blood in patients with melanoma. PD-1 was highly expressed in CD8 and CD4 cells, and B cells also highly expressed PD-1 compared with NK cells. However, there was no significant difference in Th1/Th2/Th17 cytokines and inhibitory cytokine IL-10. Although nevus showed a low expression of PD-L1 compared with healthy skin, PD-L1 expression was increased in growth-phase melanoma. Finally, we analyzed the peripheral blood profiles in patients treated with nivolumab. PD-1-bearing dendritic cells (DCs) were increased during nivolumab treatment and Lin-CD11c+HLA-DR+ cells were highly increased during nivolumab treatment. These findings indicate a clue to answering the problems during nivolumab treatment and suggest to us the importance of multiple aspect observation during immune checkpoint treatment.
Collapse
Affiliation(s)
| | - Yu Sawada
- Correspondence: ; Tel.: +81-093-691-7445
| | | |
Collapse
|
36
|
Aggen DH, Ager CR, Obradovic A, Chowdhury N, Ghasemzadeh A, Mao W, Chaimowitz M, Lopez-Bujanda ZA, Spina CS, Hawley JE, Dallos MC, Zhang C, Wang V, Li H, Guo X, Drake CG. Blocking IL1 Beta Promotes Tumor Regression and Remodeling of the Myeloid Compartment in a Renal Cell Carcinoma Model: Multidimensional Analyses. Clin Cancer Res 2021; 27:608-621. [PMID: 33148676 PMCID: PMC7980495 DOI: 10.1158/1078-0432.ccr-20-1610] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/11/2020] [Accepted: 10/29/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Intratumoral immunosuppression mediated by myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) represents a potential mechanism of immune checkpoint inhibitor (ICI) resistance in solid tumors. By promoting TAM and MDSC infiltration, IL1β may drive adaptive and innate immune resistance in renal cell carcinoma (RCC) and in other tumor types. EXPERIMENTAL DESIGN Using the RENCA model of RCC, we evaluated clinically relevant combinations of anti-IL1β plus either anti-PD-1 or the multitargeted tyrosine kinase inhibitor (TKI), cabozantinib. We performed comprehensive immune profiling of established RENCA tumors via multiparameter flow cytometry, tumor cytokine profiling, and single-cell RNA sequencing (RNA-seq). Similar analyses were extended to the MC38 tumor model. RESULTS Analyses via multiparameter flow cytometry, tumor cytokine profiling, and single-cell RNA-seq showed that anti-IL1β reduces infiltration of polymorphonuclear MDSCs and TAMs. Combination treatment with anti-IL1β plus anti-PD-1 or cabozantinib showed increased antitumor activity that was associated with decreases in immunosuppressive MDSCs and increases in M1-like TAMs. CONCLUSIONS Single-cell RNA-seq analyses show that IL1β blockade and ICI or TKI remodel the myeloid compartment through nonredundant, relatively T-cell-independent mechanisms. IL1β is an upstream mediator of adaptive myeloid resistance and represents a potential target for kidney cancer immunotherapy.
Collapse
Affiliation(s)
- David H. Aggen
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Current Address: Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
- Department of Hematology Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Casey R. Ager
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Aleksandar Obradovic
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Nivedita Chowdhury
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Ali Ghasemzadeh
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Wendy Mao
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Current Address: Kite Pharma, 930 Clopper Rd Suite 200, Gaithersburg, MD USA 20878
| | - Matthew Chaimowitz
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Zoila A. Lopez-Bujanda
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21205
| | - Catherine S. Spina
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Jessica E. Hawley
- Department of Hematology Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Matthew C. Dallos
- Department of Hematology Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA 55905
| | - Vinson Wang
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Department of Urology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Hu Li
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Xinzheng Guo
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Charles G. Drake
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Department of Hematology Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
- Department of Urology, Columbia University Irving Medical Center, New York, NY USA 10032
| |
Collapse
|
37
|
Potentiality of multiple modalities for single-cell analyses to evaluate the tumor microenvironment in clinical specimens. Sci Rep 2021; 11:341. [PMID: 33431933 PMCID: PMC7801605 DOI: 10.1038/s41598-020-79385-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022] Open
Abstract
Single-cell level analysis is powerful tool to assess the heterogeneity of cellular components in tumor microenvironments (TME). In this study, we investigated immune-profiles using the single-cell analyses of endoscopically- or surgically-resected tumors, and peripheral blood mononuclear cells from gastric cancer patients. Furthermore, we technically characterized two distinct platforms of the single-cell analysis; RNA-seq-based analysis (scRNA-seq), and mass cytometry-based analysis (CyTOF), both of which are broadly embraced technologies. Our study revealed that the scRNA-seq analysis could cover a broader range of immune cells of TME in the biopsy-resected small samples of tumors, detecting even small subgroups of B cells or Treg cells in the tumors, although CyTOF could distinguish the specific populations in more depth. These findings demonstrate that scRNA-seq analysis is a highly-feasible platform for elucidating the complexity of TME in small biopsy tumors, which would provide a novel strategies to overcome a therapeutic difficulties against cancer heterogeneity in TME.
Collapse
|
38
|
Meneveau MO, Sahli ZT, Lynch KT, Mauldin IS, Slingluff CL. Immunotyping and Quantification of Melanoma Tumor-Infiltrating Lymphocytes. Methods Mol Biol 2021; 2265:515-528. [PMID: 33704737 DOI: 10.1007/978-1-0716-1205-7_36] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The density of tumour-infiltrating lymphocytes (TILs) in melanoma is correlated with improved clinical prognosis; however, standardized TIL immunotyping and quantification protocols are lacking. Herein, we provide a review of the technologies being utilized for the immunotyping and quantification of melanoma TILs.
Collapse
Affiliation(s)
- Max O Meneveau
- Department of Surgery, The University of Virginia Health System, Charlottesville, VA, USA
| | - Zeyad T Sahli
- Department of Surgery, The University of Virginia Health System, Charlottesville, VA, USA
| | - Kevin T Lynch
- Department of Surgery, The University of Virginia Health System, Charlottesville, VA, USA
| | - Ileana S Mauldin
- Department of Surgery, The University of Virginia Health System, Charlottesville, VA, USA
| | - Craig L Slingluff
- Department of Surgery, The University of Virginia Health System, Charlottesville, VA, USA.
| |
Collapse
|
39
|
Zuazo M, Arasanz H, Bocanegra A, Fernandez G, Chocarro L, Vera R, Kochan G, Escors D. Systemic CD4 Immunity as a Key Contributor to PD-L1/PD-1 Blockade Immunotherapy Efficacy. Front Immunol 2020; 11:586907. [PMID: 33329566 PMCID: PMC7734243 DOI: 10.3389/fimmu.2020.586907] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/30/2020] [Indexed: 01/22/2023] Open
Abstract
PD-L1/PD-1 blockade immunotherapy has significantly improved treatment outcome for several cancer types compared to conventional cytotoxic therapies. However, the specific molecular and cellular mechanisms behind its efficacy are currently unclear. There is increasing evidence in murine models and in patients that unveil the key importance of systemic immunity to achieve clinical responses under several types of immunotherapy. Indeed, PD-L1/PD-1 blockade induces the expansion of systemic CD8+ PD-1+ T cell subpopulations which might be responsible for direct anti-tumor responses. However, the role of CD4+ T cells in PD-L1/PD-1 blockade-induced anti-tumor responses has been less documented. In this review we focus on the experimental data supporting the “often suspected” indispensable helper function of CD4 T cells towards CD8 effector anti-tumor responses in cancer; and particularly, we highlight the recently published studies uncovering the key contribution of systemic CD4 T cells to clinical efficacy in PD-L1/PD-1 blockade therapies. We conclude and propose that the presence of specific CD4 T cell memory subsets in peripheral blood before the initiation of treatments is a strong predictor of responses in non-small cell lung cancer patients. Therefore, development of new approaches to improve CD4 responses before PD-L1/PD-1 blockade therapy could be the solution to increase response rates and survival of patients.
Collapse
Affiliation(s)
- Miren Zuazo
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Complejo Hospitalario de Navarra-UPNA-IdISNA, Pamplona, Spain
| | - Hugo Arasanz
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Complejo Hospitalario de Navarra-UPNA-IdISNA, Pamplona, Spain
| | - Ana Bocanegra
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Complejo Hospitalario de Navarra-UPNA-IdISNA, Pamplona, Spain
| | - Gonzalo Fernandez
- Department of Oncology, Complejo Hospitalario de Navarra-IdISNA, Pamplona, Spain
| | - Luisa Chocarro
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Complejo Hospitalario de Navarra-UPNA-IdISNA, Pamplona, Spain
| | - Ruth Vera
- Department of Oncology, Complejo Hospitalario de Navarra-IdISNA, Pamplona, Spain
| | - Grazyna Kochan
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Complejo Hospitalario de Navarra-UPNA-IdISNA, Pamplona, Spain
| | - David Escors
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Complejo Hospitalario de Navarra-UPNA-IdISNA, Pamplona, Spain
| |
Collapse
|
40
|
Xiao X, Mo H, Tu K. CTNNB1 mutation suppresses infiltration of immune cells in hepatocellular carcinoma through miRNA-mediated regulation of chemokine expression. Int Immunopharmacol 2020; 89:107043. [PMID: 33039961 DOI: 10.1016/j.intimp.2020.107043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/06/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
Mutations in the CTNNB1 gene in hepatocellular carcinoma (HCC) are related to immune exclusion, and HCC patients with CTNNB1 mutations tend to be primarily resistant to anti-PD1 therapy. However, systemic evaluation of immune cell infiltration in HCC with mutant CTNNB1 is lacking, and the mechanism of immune exclusion resulting from CTNNB1 mutations remains unclear. Based on CTNNB1 mutation status in HCC, we investigated RNA and miRNA expression and infiltration of immune cells. Data downloaded from TCGA showed that HCC with CTNNB1 mutation had an increased expression of CTNNB1. HCC with CTNNB1 mutation showed a reduction in infiltration score as well as in abundance of certain kinds of immune cells, including CD4 naïve T cells, CD4+ T cells, Tex cells, Th2 cells, Tfh cells, B cells, macrophages, and NK cells. Furthermore, there were 13 chemokines downregulated among all the 14 differentially expressed chemokines (DE-CKs) in CTNNB1 mutants compared to those in the wild type. A positive correlation was found between the expression of DE-CKs and infiltration score, as well as infiltration level of 6 types of immune cells, namely B cells, CD8+ cells, CD4+ cells, macrophages, neutrophils, and dendritic cells. Additionally, 302 differentially expressed immune-related genes (DE-IRGs) were involved mainly in the human immune response and cytokine-cytokine receptor interaction. The target DE-IRGs of differentially expressed miRNAs (DE-miRNAs) were identified and used to construct a network with DE-miRNAs and DE-CKs. Overall, CTNNB1 mutation in HCC led to a decrease in chemokine expression and subsequent suppression of immune cell infiltration partly through regulating specific miRNA-IRG-CK axes, pointing to a potential combination of interference of Wnt/β-catenin signaling with immunotherapy in HCC with CTNNB1 mutation.
Collapse
Affiliation(s)
- Xuelian Xiao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, Shaanxi Province 710061, China
| | - Huanye Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, Shaanxi Province 710061, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, Shaanxi Province 710061, China.
| |
Collapse
|
41
|
Sugiyama E, Togashi Y, Takeuchi Y, Shinya S, Tada Y, Kataoka K, Tane K, Sato E, Ishii G, Goto K, Shintani Y, Okumura M, Tsuboi M, Nishikawa H. Blockade of EGFR improves responsiveness to PD-1 blockade in EGFR-mutated non-small cell lung cancer. Sci Immunol 2020; 5:5/43/eaav3937. [PMID: 32005679 DOI: 10.1126/sciimmunol.aav3937] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/09/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022]
Abstract
The clinical efficacy of anti-PD-1 (programmed cell death-1) monoclonal antibody (mAb) against cancers with oncogenic driver gene mutations, which often harbor a low tumor mutation burden, is variable, suggesting different contributions of each driver mutation to immune responses. Here, we investigated the immunological phenotypes in the tumor microenvironment (TME) of epidermal growth factor receptor (EGFR)-mutated lung adenocarcinomas, for which anti-PD-1 mAb is largely ineffective. Whereas EGFR-mutated lung adenocarcinomas had a noninflamed TME, CD4+ effector regulatory T cells, which are generally present in the inflamed TME, showed high infiltration. The EGFR signal activated cJun/cJun N-terminal kinase and reduced interferon regulatory factor-1; the former increased CCL22, which recruits CD4+ regulatory T cells, and the latter decreased CXCL10 and CCL5, which induce CD8+ T cell infiltration. The EGFR inhibitor erlotinib decreased CD4+ effector regulatory T cells infiltration in the TME and in combination with anti-PD-1 mAb showed better antitumor effects than either treatment alone. Our results suggest that EGFR inhibitors when used in conjunction with anti-PD-1 mAb could increase the efficacy of immunotherapy in lung adenocarcinomas.
Collapse
Affiliation(s)
- Eri Sugiyama
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.,Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
| | - Yosuke Togashi
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
| | - Yoshiko Takeuchi
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
| | - Sayoko Shinya
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
| | - Yasuko Tada
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
| | - Keisuke Kataoka
- Division of Molecular Oncology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
| | | | - Eiichi Sato
- Department of Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo 160-0023, Japan
| | | | - Koichi Goto
- Division of Thoracic Oncology, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Yasushi Shintani
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Meinoshin Okumura
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | | | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan. .,Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
| |
Collapse
|
42
|
Han X, Alu A, Xiao Y, Wei Y, Wei X. Hyperprogression: A novel response pattern under immunotherapy. Clin Transl Med 2020; 10:e167. [PMID: 32997401 PMCID: PMC7510779 DOI: 10.1002/ctm2.167] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 02/05/2023] Open
Abstract
Checkpoint blockade therapy has shown significant therapeutic benefits and resulted in durable responses in patients with various tumors. However, accumulating evidence has demonstrated that 4-29% of all patients with cancers with various histologies may suffer from tumor flare following such therapy. This novel tumor response pattern, termed hyperprogression, is a potentially deleterious side effect of checkpoint blockade therapy that accelerates disease progression in a subset of patients. In this review, we describe possible immune checkpoint blockade biomarkers and the epidemiology, different definitions, and predictors of hyperprogression based on the research findings and further present the available evidence supporting pathophysiological hypotheses that might explain hyperprogression during checkpoint blockade therapy. We also compare hyperprogression and pseudoprogression. Finally, we discuss areas requiring further study.
Collapse
Affiliation(s)
- Xue‐jiao Han
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Aqu Alu
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Yi‐nan Xiao
- West China School of MedicineWest China HospitalSichuan UniversityChengduChina
| | - Yu‐quan Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Xia‐wei Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
43
|
The PD-1 expression balance between effector and regulatory T cells predicts the clinical efficacy of PD-1 blockade therapies. Nat Immunol 2020; 21:1346-1358. [DOI: 10.1038/s41590-020-0769-3] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 07/28/2020] [Indexed: 02/06/2023]
|
44
|
Kumagai S, Togashi Y, Sakai C, Kawazoe A, Kawazu M, Ueno T, Sato E, Kuwata T, Kinoshita T, Yamamoto M, Nomura S, Tsukamoto T, Mano H, Shitara K, Nishikawa H. An Oncogenic Alteration Creates a Microenvironment that Promotes Tumor Progression by Conferring a Metabolic Advantage to Regulatory T Cells. Immunity 2020; 53:187-203.e8. [PMID: 32640259 DOI: 10.1016/j.immuni.2020.06.016] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 03/31/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022]
Abstract
Only a small percentage of patients afflicted with gastric cancer (GC) respond to immune checkpoint blockade (ICB). To study the mechanisms underlying this resistance, we examined the immune landscape of GC. A subset of these tumors was characterized by high frequencies of regulatory T (Treg) cells and low numbers of effector T cells. Genomic analyses revealed that these tumors bore mutations in RHOA that are known to drive tumor progression. RHOA mutations in cancer cells activated the PI3K-AKT-mTOR signaling pathway, increasing production of free fatty acids that are more effectively consumed by Treg cells than effector T cells. RHOA mutant tumors were resistant to PD-1 blockade but responded to combination of PD-1 blockade with inhibitors of the PI3K pathway or therapies targeting Treg cells. We propose that the metabolic advantage conferred by RHOA mutations enables Treg cell accumulation within GC tumors, generating an immunosuppressive TME that underlies resistance to ICB.
Collapse
Affiliation(s)
- Shogo Kumagai
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan; Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yosuke Togashi
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan.
| | - Chika Sakai
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan
| | - Akihito Kawazoe
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Masahito Kawazu
- Division of Cellular Signaling, Group for Cancer Development and Progression, National Cancer Center Research Institute, Tokyo, Japan
| | - Toshihide Ueno
- Division of Cellular Signaling, Group for Cancer Development and Progression, National Cancer Center Research Institute, Tokyo, Japan
| | - Eiichi Sato
- Department of Pathology, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Takeshi Kuwata
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Chiba, Japan
| | - Takahiro Kinoshita
- Department of Gastric Surgery, National Cancer Center Hospital East, Chiba, Japan
| | - Masami Yamamoto
- Division of Physiological Pathology, Department of Applied Science, School of Veterinary Nursing and Technology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuya Tsukamoto
- Department of Pathology, Graduate School of Medicine, Fujita Health University, Aichi, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, Group for Cancer Development and Progression, National Cancer Center Research Institute, Tokyo, Japan
| | - Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan; Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
45
|
Rochigneux P, Garcia AJ, Chanez B, Madroszyk A, Olive D, Garon EB. Medical Treatment of Lung Cancer: Can Immune Cells Predict the Response? A Systematic Review. Front Immunol 2020; 11:1036. [PMID: 32670271 PMCID: PMC7327092 DOI: 10.3389/fimmu.2020.01036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/29/2020] [Indexed: 01/23/2023] Open
Abstract
The landscape for medical treatment of lung cancer has irreversibly changed since the development of immuno-oncology (IO). Yet, while immune checkpoint blockade (ICB) revealed that T lymphocytes play a major role in lung cancer, the precise dynamic of innate and adaptive immune cells induced by anticancer treatments including chemotherapy, targeted therapy, and/or ICB is poorly understood. In lung cancer, studies evaluating specific immune cell populations as predictors of response to medical treatment are scarce, and knowledge is fragmented. Here, we review the different techniques allowing the detection of immune cells in the tumor and blood (multiplex immunohistochemistry and immunofluorescence, RNA-seq, DNA methylation pattern, mass cytometry, functional tests). In addition, we present data that consider different baseline immune cell populations as predictors of response to medical treatments of lung cancer. We also review the potential for assessing dynamic changes in cell populations during treatment as a biomarker. As powerful tools for immune cell detection and data analysis are available, clinicians and researchers could increase understanding of mechanisms of efficacy and resistance in addition to identifying new targets for IO by developing translational studies that decipher the role of different immune cell populations during lung cancer treatments.
Collapse
Affiliation(s)
- Philippe Rochigneux
- Department of Medical Oncology, Paoli-Calmettes Institute, Marseille, France.,Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France.,Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA, United States
| | - Alejandro J Garcia
- Cytometry Core Laboratory, David Geffen School of Medicine at the University of California, Los Angeles, CA, United States
| | - Brice Chanez
- Department of Medical Oncology, Paoli-Calmettes Institute, Marseille, France
| | - Anne Madroszyk
- Department of Medical Oncology, Paoli-Calmettes Institute, Marseille, France
| | - Daniel Olive
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Edward B Garon
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, CA, United States
| |
Collapse
|
46
|
Russano M, Napolitano A, Ribelli G, Iuliani M, Simonetti S, Citarella F, Pantano F, Dell'Aquila E, Anesi C, Silvestris N, Argentiero A, Solimando AG, Vincenzi B, Tonini G, Santini D. Liquid biopsy and tumor heterogeneity in metastatic solid tumors: the potentiality of blood samples. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:95. [PMID: 32460897 PMCID: PMC7254767 DOI: 10.1186/s13046-020-01601-2] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022]
Abstract
In a large number of cancer types, treatment selection depends on the presence of specific tumor biomarkers. Due to the dynamic nature of cancer, very often these predictive biomarkers are not uniformly present in all cancer cells. Tumor heterogeneity represents indeed one of the main causes of therapeutic failure, and its decoding remains a major ongoing challenge in the field. Liquid biopsy is the sampling and analysis of non-solid biological tissue often through rapid and non-invasive methods, which allows the assessment in real-time of the evolving landscape of cancer. Samples can be obtained from blood and most other bodily fluids. A blood-based liquid biopsy can capture circulating tumor cells and leukocytes, as well as circulating tumor-derived nucleic acids. In this review, we discuss the current and possibly future applications of blood-based liquid biopsy in oncology, its advantages and its limitations in clinical practice. We specifically focused on its role as a tool to capture tumor heterogeneity in metastatic cancer patients.
Collapse
Affiliation(s)
- Marco Russano
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy
| | - Andrea Napolitano
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy
| | - Giulia Ribelli
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy.
| | - Michele Iuliani
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy
| | - Sonia Simonetti
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy
| | - Fabrizio Citarella
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy
| | - Francesco Pantano
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy
| | - Emanuela Dell'Aquila
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy
| | - Cecilia Anesi
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, IRCCS-Istituto Tumori "Giovanni Paolo II" of Bari, 70124, Bari, Italy.,Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', 70124, Bari, Italy
| | - Antonella Argentiero
- Medical Oncology Unit, IRCCS-Istituto Tumori "Giovanni Paolo II" of Bari, 70124, Bari, Italy
| | - Antonio Giovanni Solimando
- Medical Oncology Unit, IRCCS-Istituto Tumori "Giovanni Paolo II" of Bari, 70124, Bari, Italy.,Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine 'G. Baccelli', University of Bari Medical School, 70124, Bari, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy
| | - Giuseppe Tonini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy
| | - Daniele Santini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Álvaro del Portillo, 21, 00128, Rome, Italy
| |
Collapse
|
47
|
Chornoguz O, Leettola CN, Leander K, Brosnan K, Emmell E, Chiu ML, Santulli-Marotto S. Characterization of a Novel Bispecific Antibody That Activates T Cells In Vitro and Slows Tumor Growth In Vivo. Monoclon Antib Immunodiagn Immunother 2020; 38:242-254. [PMID: 31825302 PMCID: PMC6918852 DOI: 10.1089/mab.2019.0035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Although CD3 T cell redirecting antibodies have been successfully utilized for the treatment of hematological malignancies (blinatumomab), the T cell signaling pathways induced by these molecules are incompletely understood. To gain insight into the mechanism of action for T cell redirection antibodies, we created a novel murine CD3xEpCAM bispecific antibody that incorporates a silent Fc to dissect function and signaling of murine CD8 OT1 T cells upon stimulation. T cell-mediated cytotoxicity, cytokine secretion, expression of activation markers, and proliferation were directly induced in T cells treated with the novel CD3xEpCAM bispecific molecule in vitro in the presence of epithelial cell adhesion molecule (EpCAM) expressing tumor cells. Nanostring analysis showed that CD3xEpCAM induced a gene expression profile that resembled antigen-mediated activation, although the magnitude was lower than that of the antigen-induced response. In addition, this CD3xEpCAM bispecific antibody exhibited in vivo efficacy. This is the first study that investigates both in vitro and in vivo murine CD8 T cell function and signaling induced by a CD3xEpCAM antibody having a silent Fc to delineate differences between antigen-independent and antigen-specific T cell activation. These findings expand the understanding of T cell function and signaling induced by CD3 redirection bispecific antibodies and may help to develop more efficacious CD3 redirection therapeutics for cancer treatment, particularly for solid tumors.
Collapse
Affiliation(s)
- Olesya Chornoguz
- Janssen Biotherapeutics, Janssen R&D, Spring House, Pennsylvania
| | | | - Karen Leander
- Janssen Biotherapeutics, Janssen R&D, Spring House, Pennsylvania
| | - Kerry Brosnan
- Janssen Biotherapeutics, Janssen R&D, Spring House, Pennsylvania
| | - Eva Emmell
- Janssen Biotherapeutics, Janssen R&D, Spring House, Pennsylvania
| | - Mark L Chiu
- Janssen Biotherapeutics, Janssen R&D, Spring House, Pennsylvania
| | | |
Collapse
|
48
|
Hatae R, Chamoto K, Kim YH, Sonomura K, Taneishi K, Kawaguchi S, Yoshida H, Ozasa H, Sakamori Y, Akrami M, Fagarasan S, Masuda I, Okuno Y, Matsuda F, Hirai T, Honjo T. Combination of host immune metabolic biomarkers for the PD-1 blockade cancer immunotherapy. JCI Insight 2020; 5:133501. [PMID: 31855576 DOI: 10.1172/jci.insight.133501] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/10/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUNDCurrent clinical biomarkers for the programmed cell death 1 (PD-1) blockade therapy are insufficient because they rely only on the tumor properties, such as programmed cell death ligand 1 expression frequency and tumor mutation burden. Identifying reliable, responsive biomarkers based on the host immunity is necessary to improve the predictive values.METHODSWe investigated levels of plasma metabolites and T cell properties, including energy metabolism markers, in the blood of patients with non-small cell lung cancer before and after treatment with nivolumab (n = 55). Predictive values of combination markers statistically selected were evaluated by cross-validation and linear discriminant analysis on discovery and validation cohorts, respectively. Correlation between plasma metabolites and T cell markers was investigated.RESULTSThe 4 metabolites derived from the microbiome (hippuric acid), fatty acid oxidation (butyrylcarnitine), and redox (cystine and glutathione disulfide) provided high response probability (AUC = 0.91). Similarly, a combination of 4 T cell markers, those related to mitochondrial activation (PPARγ coactivator 1 expression and ROS), and the frequencies of CD8+PD-1hi and CD4+ T cells demonstrated even higher prediction value (AUC = 0.96). Among the pool of selected markers, the 4 T cell markers were exclusively selected as the highest predictive combination, probably because of their linkage to the abovementioned metabolite markers. In a prospective validation set (n = 24), these 4 cellular markers showed a high accuracy rate for clinical responses of patients (AUC = 0.92).CONCLUSIONCombination of biomarkers reflecting host immune activity is quite valuable for responder prediction.FUNDINGAMED under grant numbers 18cm0106302h0003, 18gm0710012h0105, and 18lk1403006h0002; the Tang Prize Foundation; and JSPS KAKENHI grant numbers JP16H06149, 17K19593, and 19K17673.
Collapse
Affiliation(s)
| | | | | | - Kazuhiro Sonomura
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Life Science Research Center, Technology Research Laboratory, Shimadzu Corporation, Kyoto, Japan
| | - Kei Taneishi
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,RIKEN Cluster for Science, Technology and Innovation Hub, Kobe, Japan
| | - Shuji Kawaguchi
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | - Sidonia Fagarasan
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan
| | - Izuru Masuda
- Medical Examination Center, Takeda Hospital, Kyoto, Japan
| | - Yasushi Okuno
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | |
Collapse
|
49
|
Spassova I, Ugurel S, Terheyden P, Sucker A, Hassel JC, Ritter C, Kubat L, Habermann D, Farahpour F, Saeedghalati M, Peiffer L, Kumar R, Schrama D, Hoffmann D, Schadendorf D, Becker JC. Predominance of Central Memory T Cells with High T-Cell Receptor Repertoire Diversity is Associated with Response to PD-1/PD-L1 Inhibition in Merkel Cell Carcinoma. Clin Cancer Res 2020; 26:2257-2267. [PMID: 31932494 DOI: 10.1158/1078-0432.ccr-19-2244] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/06/2019] [Accepted: 01/08/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Merkel cell carcinoma (MCC) is an aggressive neuroendocrine skin cancer, which can be effectively controlled by immunotherapy with PD-1/PD-L1 checkpoint inhibitors. However, a significant proportion of patients are characterized by primary therapy resistance. Predictive biomarkers for response to immunotherapy are lacking. EXPERIMENTAL DESIGN We applied Bayesian inference analyses on 41 patients with MCC testing various clinical and biomolecular characteristics to predict treatment response. Further, we performed a comprehensive analysis of tumor tissue-based immunologic parameters including multiplexed immunofluorescence for T-cell activation and differentiation markers, expression of immune-related genes and T-cell receptor (TCR) repertoire analyses in 18 patients, seven objective responders, and 11 nonresponders. RESULTS Bayesian inference analyses demonstrated that among currently discussed biomarkers only unimpaired overall performance status and absence of immunosuppression were associated with response to therapy. However, in responders, a predominance of central memory T cells and expression of genes associated with lymphocyte attraction and activation was evident. In addition, TCR repertoire usage of tumor-infiltrating lymphocytes (TILs) demonstrated low T-cell clonality, but high TCR diversity in responding patients. In nonresponders, terminally differentiated effector T cells with a constrained TCR repertoire prevailed. Sequential analyses of tumor tissue obtained during immunotherapy revealed a more pronounced and diverse clonal expansion of TILs in responders indicating an impaired proliferative capacity among TILs of nonresponders upon checkpoint blockade. CONCLUSIONS Our explorative study identified new tumor tissue-based molecular characteristics associated with response to anti-PD-1/PD-L1 therapy in MCC. These observations warrant further investigations in larger patient cohorts to confirm their potential value as predictive markers.
Collapse
Affiliation(s)
- Ivelina Spassova
- Translational Skin Cancer Research, German Consortium for Translational Cancer Research (Deutsches Konsortium für Translationale Krebsforschung; DKTK), Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital of Essen, Essen, Germany
| | - Patrick Terheyden
- Department of Dermatology, University Hospital of Lübeck, Lübeck, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital of Essen, Essen, Germany
| | - Jessica C Hassel
- Department of Dermatology, University Hospital of Heidelberg, Germany
| | - Cathrin Ritter
- Translational Skin Cancer Research, German Consortium for Translational Cancer Research (Deutsches Konsortium für Translationale Krebsforschung; DKTK), Essen, Germany
| | - Linda Kubat
- Translational Skin Cancer Research, German Consortium for Translational Cancer Research (Deutsches Konsortium für Translationale Krebsforschung; DKTK), Essen, Germany
| | - Daniel Habermann
- Bioinformatics and Computational Biophysics, University of Duisburg-Essen, Essen, Germany
| | - Farnoush Farahpour
- Bioinformatics and Computational Biophysics, University of Duisburg-Essen, Essen, Germany
| | | | - Lukas Peiffer
- Translational Skin Cancer Research, German Consortium for Translational Cancer Research (Deutsches Konsortium für Translationale Krebsforschung; DKTK), Essen, Germany.,German Cancer Research Center (Deutsches Krebsforschungs Zentrum, DKFZ), Heidelberg, Germany
| | - Rajiv Kumar
- German Cancer Research Center (Deutsches Krebsforschungs Zentrum, DKFZ), Heidelberg, Germany.,Division of Molecular Genetic Epidemiology, Heidelberg, Germany
| | - David Schrama
- Department of Dermatology, University Hospital of Würzburg, Germany
| | - Daniel Hoffmann
- Bioinformatics and Computational Biophysics, University of Duisburg-Essen, Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital of Essen, Essen, Germany
| | - Jürgen C Becker
- Translational Skin Cancer Research, German Consortium for Translational Cancer Research (Deutsches Konsortium für Translationale Krebsforschung; DKTK), Essen, Germany. .,Department of Dermatology, University Hospital of Essen, Essen, Germany.,German Cancer Research Center (Deutsches Krebsforschungs Zentrum, DKFZ), Heidelberg, Germany
| |
Collapse
|
50
|
Chamoto K, Hatae R, Honjo T. Current issues and perspectives in PD-1 blockade cancer immunotherapy. Int J Clin Oncol 2020; 25:790-800. [PMID: 31900651 PMCID: PMC7192862 DOI: 10.1007/s10147-019-01588-7] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 11/14/2019] [Indexed: 12/21/2022]
Abstract
Programmed cell death 1 (PD-1) signal receptor blockade has revolutionized the field of cancer therapy. Despite their considerable potential for treating certain cancers, drugs targeting PD-1 still present two main drawbacks: the substantial number of unresponsive patients and/or patients showing recurrences, and side effects associated with the autoimmune response. These drawbacks highlight the need for further investigation of the mechanisms underlying the therapeutic effects, as well as the need to develop novel biomarkers to predict the lack of treatment response and to monitor potential adverse events. Combination therapy is a promising approach to improve the efficacy of PD-1 blockade therapy. Considering the increasing number of patients with cancer worldwide, solving the above issues is central to the field of cancer immunotherapy. In this review, we discuss these issues and clinical perspectives associated with PD-1 blockade cancer immunotherapy.
Collapse
Affiliation(s)
- Kenji Chamoto
- Department of Immunology and Genomic Medicine, Kyoto University Graduate School of Medicine, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Ryusuke Hatae
- Department of Immunology and Genomic Medicine, Kyoto University Graduate School of Medicine, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Tasuku Honjo
- Department of Immunology and Genomic Medicine, Kyoto University Graduate School of Medicine, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|