1
|
Zeng W, Wang J, Chen Z, Yang J, Zhu A, Zheng Y, Chen X, Liu Y, Wu L, Xie Y, Ju S, Chen J, Ding C, Li C, Tong X, Liu M, Zhao J. Efficient Predictor for Immunotherapy Efficacy: Detecting Pan-Clones Effector Tumor Antigen-Specific T Cells in Blood by Nanoparticles Loading Whole Tumor Antigens. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409913. [PMID: 39498880 PMCID: PMC11727128 DOI: 10.1002/advs.202409913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/09/2024] [Indexed: 11/07/2024]
Abstract
Cancer involves tumor cells and tumor-specific immunity. The ability to accurately quantify tumor-specific immunity is limited. Most immunotherapies function by activating new effector tumor antigen-specific T cells (ETASTs) or reactivating the pre-existing ETASTs repertoire. Therefore, the amount of ETASTs can be used to characterize immunotherapy efficacy. Tumor antigens are highly heterogeneous and detecting most ETASTs is challenging. Therefore, nanoparticles loading whole-cell tumor antigens are used to activate and detect pan-clones ETASTs in the blood. The differences between ETASTs and other T cells are transformed into activated and non-activated states. By measuring markers of the activated status and cytotoxic function of ETASTs, it can distinguish ETASTs from other T cells. ETASTs in patients with lung cancer are higher than those in healthy individuals and those with benign pulmonary nodules. Therapeutic efficacy positively correlated with the number of ETASTs in the blood. ETATS levels increase only in the blood of patients who respond to immunotherapy. Single-cell sequencing studies validated these findings. This study provides a highly accurate, specific, non-invasive, and efficient biomarker for predicting immunotherapy efficacy in lung and other cancers. This method can also be applied to evaluate the efficacy of other treatments, such as radiotherapy, oncolytic viruses, and nanomedicine-based therapies.
Collapse
Affiliation(s)
- Weibiao Zeng
- Institute of Thoracic SurgeryThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Department of PharmaceuticsCollege of Pharmaceutical SciencesSoochow UniversitySuzhouJiangsu215123P. R. China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Department of Thoracic SurgeryShanghai General HospitalShanghai Jiaotong University School of MedicineShanghai200080P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Jin Wang
- Department of PharmaceuticsCollege of Pharmaceutical SciencesSoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Zhike Chen
- Institute of Thoracic SurgeryThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Jian Yang
- Institute of Thoracic SurgeryThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Ao Zhu
- Department of PharmaceuticsCollege of Pharmaceutical SciencesSoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Yan Zheng
- Department of PharmaceuticsCollege of Pharmaceutical SciencesSoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Xianlan Chen
- Department of PharmaceuticsCollege of Pharmaceutical SciencesSoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Yuhan Liu
- Department of PharmaceuticsCollege of Pharmaceutical SciencesSoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Leilei Wu
- Department of RadiotherapyShanghai Pulmonary Hospital of Tongji UniversityShanghai200000P. R. China
| | - Yufeng Xie
- Institute of Thoracic SurgeryThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Sheng Ju
- Institute of Thoracic SurgeryThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Jun Chen
- Institute of Thoracic SurgeryThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Cheng Ding
- Institute of Thoracic SurgeryThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Chang Li
- Institute of Thoracic SurgeryThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Xin Tong
- Institute of Thoracic SurgeryThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| | - Mi Liu
- Department of PharmaceuticsCollege of Pharmaceutical SciencesSoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
- Suzhou Ersheng Biopharmaceutical Co., LtdSuzhou215123P. R. China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics DevelopmentSoochow UniversitySuzhou215123P. R. China
- Wuxi Boston Biopharmaceutical Co., LtdWuxi214125P. R. China
| | - Jun Zhao
- Institute of Thoracic SurgeryThe First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow UniversitySoochow UniversitySuzhouJiangsu215123P. R. China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational ResearchSoochow UniversitySuzhouJiangsu215123P. R. China
| |
Collapse
|
2
|
Pimentel JM, Zhou JY, Wu GS. Autophagy and cancer therapy. Cancer Lett 2024; 605:217285. [PMID: 39395780 DOI: 10.1016/j.canlet.2024.217285] [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: 08/02/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
Autophagy is an intracellular degradation process that sequesters cytoplasmic components in double-membrane vesicles known as autophagosomes, which are degraded upon fusion with lysosomes. This pathway maintains the integrity of proteins and organelles while providing energy and nutrients to cells, particularly under nutrient deprivation. Deregulation of autophagy can cause genomic instability, low protein quality, and DNA damage, all of which can contribute to cancer. Autophagy can also be overactivated in cancer cells to aid in cancer cell survival and drug resistance. Emerging evidence indicates that autophagy has functions beyond cargo degradation, including roles in tumor immunity and cancer stem cell survival. Additionally, autophagy can also influence the tumor microenvironment. This feature warrants further investigation of the role of autophagy in cancer, in which autophagy manipulation can improve cancer therapies, including cancer immunotherapy. This review discusses recent findings on the regulation of autophagy and its role in cancer therapy and drug resistance.
Collapse
Affiliation(s)
- Julio M Pimentel
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA; Institutional Research Academic Career Development Award Program, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jun Ying Zhou
- Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, 48201, USA; Department of Oncology, Wayne State University, Detroit, MI, 48201, USA
| | - Gen Sheng Wu
- Molecular Therapeutics Program, Karmanos Cancer Institute, Detroit, MI, 48201, USA; Department of Oncology, Wayne State University, Detroit, MI, 48201, USA; Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
3
|
Wang J, Zeng W, Xue J, Zhu A, Chen X, Zheng Y, Liu Y, Qin S, Zhao J, Liu M. Efficient Biomarker for Immunotherapy: Measuring Broad Clones Effector Tumor Antigen-Specific T Cells in the Blood of Esophageal Cancer Patients. Anal Chem 2024. [PMID: 39561375 DOI: 10.1021/acs.analchem.4c04049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Cancer is the result of the interactions between tumor cells and tumor-specific immune responses. The current biomarkers detect tumor cells' properties, but accurate measurement of tumor-specific immunity is lacking. Most immunotherapies work by activating new effector tumor antigen-specific T cells (ETASTs) or reactivating pre-existing ETASTs' repertoire. The responses to immunotherapy depend on the increase of ETASTs. The amount of ETASTs, especially in blood, is critical for therapeutic efficacy. Distinguishing ETASTs from other T cells by their structural characteristics is difficult. Therefore, nanoparticles loading whole tumor antigens are utilized to activate broad clones ETASTs pre-existing in peripheral blood, followed by detecting them. Thus, the differences between ETASTs and other T cells are transformed to the differences between activated states and unactivated states. By measuring the markers of activated states and cytotoxic functions, we can distinguish ETASTs from other T cells. Nanoparticles loading mixed multiple allogeneic tumor tissue lysates or mixed multiple tumor cell lines can be utilized as universal nanoparticles to replace nanoparticles loading personalized tumor tissue. ETASTs (TATAN-activated CD8+IFN-γ+) in esophageal cancer patients are more than those in healthy people. Measurement of the ETASTs in the blood of esophageal cancer patients before and after ongoing therapy showed that ETATSs increased in the blood of patients who were responsive to immunotherapy but did not increase in the blood of nonresponders. These illustrated that therapeutic efficacy was positively correlated with the level of ETASTs in PBMC. Altogether, this study provides us a highly accurate and specific biomarker for predicting the therapeutic efficacy of cancer immunotherapy and potentially other therapies, such as radiotherapy.
Collapse
Affiliation(s)
- Jin Wang
- Department of Pharmaceutics, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational Research, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Weibiao Zeng
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Jiao Xue
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Ao Zhu
- Department of Pharmaceutics, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational Research, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Xianlan Chen
- Department of Pharmaceutics, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational Research, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Yan Zheng
- Department of Pharmaceutics, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational Research, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Yuhan Liu
- Department of Pharmaceutics, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational Research, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Songbing Qin
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Jun Zhao
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational Research, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
| | - Mi Liu
- Department of Pharmaceutics, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
- Institute of Minimally Invasive Thoracic Cancer Therapy and Translational Research, Soochow University, Suzhou, Jiangsu 215123, People's Republic of China
- Suzhou Ersheng Biopharmaceutical Co., Ltd., Suzhou 215123, People's Republic of China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China
- Wuxi Boston Biopharmaceutical Co., Ltd., Wuxi 214125, People's Republic of China
- Kunshan Hospital of Traditional Chinese Medicine, Kunshan 215300, People's Republic of China
| |
Collapse
|
4
|
Xiong A, Xu J, Wang S, Zhong R, Lu J, Chu T, Zhang W, Li Y, Zheng X, Han B, Nie W, Zhong H, Zhang X. On-treatment lung immune prognostic index is predictive for first-line PD-1 inhibitor combined with chemotherapy in patients with non-small cell lung cancer. Front Immunol 2023; 14:1173025. [PMID: 37304273 PMCID: PMC10247997 DOI: 10.3389/fimmu.2023.1173025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Background Inflammation is a factor that promotes tumor progression and immunosuppression. Lung immune prognostic index (LIPI) is a non-invasive and easily calculated indicator of inflammation. This study aimed to investigate whether continuous assessment of LIPI has predictive value for chemoimmunotherapy in non-small cell lung cancer (NSCLC) patients receiving first-line programmed cell death 1 (PD-1) inhibitor plus chemotherapy. In addition, the predictive value of LIPI in patients with the negative or low programmed death-ligand (PD-L1) expression level was also explored. Methods Totally, 146 stage IIIB to IV or recurrent NSCLC patients who received first-line PD-1 inhibitor combined with chemotherapy were enrolled in this study. The LIPI scores were calculated at baseline (PRE-LIPI) and after two cycles of the combined administration (POST-LIPI). This study analyzed the relationship between good/intermediate/poor PRE (POST)-LIPI and objective response rate (ORR), as well as progression-free survival (PFS) using logistic and Cox regression models. In addition, the predictive value of LIPI in patients with the negative or low PD-L1 expression level was explored. To further assess the potential predictive value of continuous assessment of LIPI, the association of sum (LIPI) [sum(LIPI) = PRE-LIPI + POST-LIPI] and PFS was analyzed in the 146 patients. Results Compared with good POST-LIPI group, significantly lower ORRs were found in intermediate POST-LIPI (P = 0.005) and poor POST-LIPI (P = 0.018) groups. Moreover, intermediate POST-LIPI (P =0.003) and poor POST-LIPI (P < 0.001) were significantly associated with a shorter PFS than good POST-LIPI. Additionally, a higher POST-LIPI score was still significantly associated with poorer treatment efficacy in patients with the negative or low PD-L1 expression level. Moreover, a higher sum (LIPI) score was significantly correlated with a shorter PFS (P = 0.001). Conclusion Continuous assessment of LIPI might be an effective method for predicting the efficacy of PD-1 inhibitor plus chemotherapy in NSCLC patients. In addition, in patients with the negative or low PD-L1 expression level, it might also have a potential predictive value for therapeutic efficacy to continuously assess LIPI during the treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Wei Nie
- *Correspondence: Xueyan Zhang, ; Wei Nie, ; Hua Zhong,
| | - Hua Zhong
- *Correspondence: Xueyan Zhang, ; Wei Nie, ; Hua Zhong,
| | - Xueyan Zhang
- *Correspondence: Xueyan Zhang, ; Wei Nie, ; Hua Zhong,
| |
Collapse
|
5
|
Hyun J, Kim SJ, Cho SD, Kim HW. Mechano-modulation of T cells for cancer immunotherapy. Biomaterials 2023; 297:122101. [PMID: 37023528 DOI: 10.1016/j.biomaterials.2023.122101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 03/12/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023]
Abstract
Immunotherapy, despite its promise for future anti-cancer approach, faces significant challenges, such as off-tumor side effects, innate or acquired resistance, and limited infiltration of immune cells into stiffened extracellular matrix (ECM). Recent studies have highlighted the importance of mechano-modulation/-activation of immune cells (mainly T cells) for effective caner immunotherapy. Immune cells are highly sensitive to the applied physical forces and matrix mechanics, and reciprocally shape the tumor microenvironment. Engineering T cells with tuned properties of materials (e.g., chemistry, topography, and stiffness) can improve their expansion and activation ex vivo, and their ability to mechano-sensing the tumor specific ECM in vivo where they perform cytotoxic effects. T cells can also be exploited to secrete enzymes that soften ECM, thus increasing tumor infiltration and cellular therapies. Furthermore, T cells, such as chimeric antigen receptor (CAR)-T cells, genomic engineered to be spatiotemporally controllable by physical stimuli (e.g., ultrasound, heat, or light), can mitigate adverse off-tumor effects. In this review, we communicate these recent cutting-edge endeavors devoted to mechano-modulating/-activating T cells for effective cancer immunotherapy, and discuss future prospects and challenges in this field.
Collapse
|
6
|
Cui Y, Shi J, Cui Y, Zhu Z, Zhu W. The relationship between autophagy and PD-L1 and their role in antitumor therapy. Front Immunol 2023; 14:1093558. [PMID: 37006252 PMCID: PMC10050383 DOI: 10.3389/fimmu.2023.1093558] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
Immune checkpoint blockade therapy is an important advance in cancer treatment, and the representative drugs (PD-1/PD-L1 antibodies) have greatly improved clinical outcomes in various human cancers. However, since many patients still experience primary resistance, they do not respond to anti-PD1/PD-L1 therapy, and some responders also develop acquired resistance after an initial response. Therefore, combined therapy with anti-PD-1/PD-L1 immunotherapy may result in better efficacy than monotherapy. In tumorigenesis and tumor development processes, the mutual regulation of autophagy and tumor immune escape is an intrinsic factor of malignant tumor progression. Understanding the correlation between the tumor autophagy pathway and tumor immune escape may help identify new clinical cancer treatment strategies. Since both autophagy and immune escape of tumor cells occur in a relatively complex microenvironmental network, autophagy affects the immune-mediated killing of tumor cells and immune escape. Therefore, comprehensive treatment targeting autophagy and immune escape to achieve “immune normalization” may be an important direction for future research and development. The PD-1/PD-L1 pathway is essential in tumor immunotherapy. High expression of PD-L1 in different tumors is closely related to poor survival rates, prognoses, and treatment effects. Therefore, exploring the mechanism of PD-L1 expression is crucial to improve the efficacy of tumor immunotherapy. Here, we summarize the mechanism and mutual relationship between autophagy and PD-L1 in antitumor therapy, which may help enhance current antitumor immunotherapy approaches.
Collapse
Affiliation(s)
- Yu Cui
- Department of Otolaryngology, Head & Neck Surgery, First Hospital of Jilin University, Changchun, China
| | - Jinfeng Shi
- Department of Otolaryngology, Head & Neck Surgery, First Hospital of Jilin University, Changchun, China
| | - Youbin Cui
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, China
| | - Zhanpeng Zhu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
- *Correspondence: Wei Zhu, ; Zhanpeng Zhu,
| | - Wei Zhu
- Department of Otolaryngology, Head & Neck Surgery, First Hospital of Jilin University, Changchun, China
- *Correspondence: Wei Zhu, ; Zhanpeng Zhu,
| |
Collapse
|
7
|
Knight A, Karapetyan L, Kirkwood JM. Immunotherapy in Melanoma: Recent Advances and Future Directions. Cancers (Basel) 2023; 15:1106. [PMID: 36831449 PMCID: PMC9954703 DOI: 10.3390/cancers15041106] [Citation(s) in RCA: 104] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
The use of immunotherapy in the treatment of advanced and high-risk melanoma has led to a striking improvement in outcomes. Although the incidence of melanoma has continued to rise, median survival has improved from approximately 6 months to nearly 6 years for patients with advanced inoperable stage IV disease. Recent understanding of the tumor microenvironment and its interplay with the immune system has led to the explosive development of novel immunotherapy treatments. Since the approval of the therapeutic cytokines interleukin-2 and interferon alfa-2 in the 1990s, the development of novel immune checkpoint inhibitors (ICIs), oncolytic virus therapy, and modulators of the tumor microenvironment have given way to a new era in melanoma treatment. Monoclonal antibodies directed at programmed cell death protein 1 receptor (PD-1) and its ligand (PDL-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and lymphocyte-activation gene 3 (LAG-3) have provided robust activation of the adaptive immune system, restoring immune surveillance leading to host tumor recognition and destruction. Multiple other immunomodulatory therapeutics are under investigation to overcome resistance to ICI therapy, including the toll-like receptor-9 (TLR-9) and 7/8 (TLR-7/8) agonists, stimulator of interferon genes (STING) agonists, and fecal microbiota transplantation. In this review, we focus on the recent advances in immunotherapy for the treatment of melanoma and provide an update on novel therapies currently under investigation.
Collapse
Affiliation(s)
- Andrew Knight
- Department of Medicine, Division of General Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Lilit Karapetyan
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - John M. Kirkwood
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
8
|
Umeda K, Tanaka N, Yasumizu Y, Takeda T, Matsumoto K, Morita S, Kosaka T, Mizuno R, Oya M. Site-Specific Differences in PD-1 Blockade Success and Biomarkers in Urothelial Carcinoma Treated with Pembrolizumab. Clin Genitourin Cancer 2023; 21:128-135. [PMID: 36058808 DOI: 10.1016/j.clgc.2022.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Since tumors in different human organs may have different tumor microenvironments, we evaluate time-course changes in all tumor locations after pembrolizumab treatment in urothelial carcinoma (UC) to examine the differences in efficacy of pembrolizumab per organ. Further, we uncover the usefulness of inflammatory markers such as neutrophil-to-lymphocyte ratio (NLR), CRP, and kinetics of these markers to predict pembrolizumab success and relation to overall survival (OS) in UC. PATIENTS AND METHODS A total of 115 cancerous lesions from 44 UC patients were measurable based on RECIST 1.1 criteria. The serum CRP and NLR levels were measured at baseline prior to pembrolizumab treatment and at least every 3 weeks just prior to pembrolizumab administration. RESULTS Site-specific success (ie, patients with CR/PR/SD by RESIST 1.1) rates for pembrolizumab treatments were as follows: primary tumors: 67%, lymph node: 70%, lung: 44%, liver: 40%, and peritoneal metastasis: 56%. Focusing on the major metastasis sites, lymph nodes and lungs, we examined the relationships between NLR, CRP, or that kinetics and pembrolizumab success. In lymph nodes, both early NLR kinetics (P = .005) and CRP kinetics (P = .035) was a predictor for pembrolizumab success. On the other hand, none of 4 was not in lung metastases. Regarding to the mortality, the multivariate analysis revealed that early NLR kinetics was a prognostic biomarker for OS among the 4, independent of performance status and liver metastasis. CONCLUSION We revealed that site-specific pembrolizumab success in UC. Early NLR kinetics was a predictor for lymph node pembrolizumab success and OS in our cohorts.
Collapse
Affiliation(s)
- Kota Umeda
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Nobuyuki Tanaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan.
| | - Yota Yasumizu
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Toshikazu Takeda
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | | | - Shinya Morita
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Ryuichi Mizuno
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
9
|
Da L, Qu Z, Zhang C, Shen Y, Huang W, Zhang Y, Gu K. Prognostic value of inflammatory markers and clinical features for survival in advanced or metastatic esophageal squamous cell carcinoma patients receiving anti-programmed death 1 treatment. Front Oncol 2023; 13:1144875. [PMID: 37035159 PMCID: PMC10076857 DOI: 10.3389/fonc.2023.1144875] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Purpose This study aims to assess the prognostic value of inflammatory markers and clinical features in advanced or metastatic esophageal squamous cell carcinoma (ESCC) patients receiving anti-programmed death 1 (PD-1) treatment. Methods Based on receiver operating characteristic curve (ROC) analysis, Youden's indexes were applied to determine the cut-off values for inflammatory markers, including neutrophil-to-lymphocyte ratio (NLR), derived neutrophil-to-lymphocye ratio (dNLR), monocyte-to-lymphocyte ratio (MLR), platelet-to-lymphocyte ratio (PLR), and systemic immune-inflammation index (SII). Wilcoxon test was conducted to evaluate the changes in above inflammatory markers. Kaplan-Meier method was utilized to estimate progression-free survival (PFS) and overall survival (OS), and the Log-rank test was used to compare the different survival between groups. Univariate and multivariate Cox regression analyses were performed to assess the prognostic value of inflammatory markers and clinical features. Results 162 advanced or metastatic ESCC patients receiving anti-PD-1 treatment were enrolled in this retrospective study. The cut-off values of NLR, dNLR, MLR, PLR, and SII were 4.748, 2.214, 0.309, 250.505, and 887.895, respectively. NLR, dNLR, PLR, and SII declined significantly among the partial response (PR) (P<0.001, P<0.001, P=0.036, P<0.001), objective response rate (ORR) (P<0.001, P<0.001, P=0.036, P<0.001), and disease control rate (DCR) (P<0.001, P<0.001, P=0.038, P<0.001) groups, respectively. Significant increases were found in NLR (P<0.001), dNLR (P<0.001), MLR (P=0.001), and SII (P=0.024) when anti-PD-1 treatment failed. Multivariate Cox regression analysis indicated that NLR (P<0.001, P=0.002), lymph node metastasis (P=0.013, P=0.001), Eastern Cooperative Oncology Group Performance Status (ECOG PS) (P=0.008, P=0.002), and treatment lines (P=0.037, P=0.048) were significant prognostic indicators of PFS and OS. Additionally, SII (P=0.016) was also significantly related to OS in ESCC patients. The risk score model showed that low risk patients prolonged PFS and OS than those with middle or high risk (P<0.001, P<0.001). Conclusion Inflammatory markers can reflect short-term outcomes of anti-PD-1 treatment for ESCC patients. NLR, lymph node metastases, ECOG PS, and treatment lines are significant prognostic indicators for PFS and OS. And the risk score model constructed based on the above factors has favourable prognostic predictive value.
Collapse
Affiliation(s)
| | | | | | | | | | - Yiyin Zhang
- *Correspondence: Yiyin Zhang, ; Kangsheng Gu,
| | | |
Collapse
|
10
|
Sun X, Zhang J, Xiao C, Ge Z. Expression profile and prognostic values of LSM family in skin cutaneous melanoma. BMC Med Genomics 2022; 15:238. [DOI: 10.1186/s12920-022-01395-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Background
The like-Smith (LSM) family is a group of RNA-binding proteins involved in RNA metabolism. However, their involvement in tumors, particularly skin cutaneous melanoma (SKCM), is not fully understood. In this study, we focused on the expression profiles and prognostic values of the LSM family in SKCM.
Methods
Raw data were downloaded from The Cancer Genome Atlas. The expression profile and prognostic value of LSM genes in SKCM were explored using the GEPIA, cBioPortal, and HPA databases. Protein–protein and gene–gene interaction analyses were performed using STRING and GeneMANIA. Enrichment and Cox regression analysis were conducted using R software. The TISIDB database was used to explore the relationship between LSMs and immunomodulators. Receiver operating characteristic curves and nomogram models were constructed to validate prognostic values.
Results
mRNA and protein expression levels of LSM2, LSM4, and LSM12 were significantly elevated in SKCM. The upregulated mRNA expression of LSM2 (p = 0.0013) and LSM4 (p = 0.0043) was significantly correlated with poor overall survival in patients with SKCM, whereas only LSM2 (p = 0.049) overexpression was markedly associated with worse disease-free survival. LSM2 overexpression was an independent risk factor (p = 0.013) and was confirmed to have a high prognostic value in SKCM using the receiver operating characteristic curve (AUC = 0.942) and nomogram models. All LSM genes were identified as genomic mutations, whereas alteration of LSM2 (p = 0.0153) significantly affected the overall survival in patients with SKCM. Significant correlations were observed between LSM family expression, immune cell infiltration, and immunomodulator. Furthermore, function and pathway enrichment analysis showed that the LSM family was mainly RNA binding proteins and involved in RNA splicing and degradation.
Conclusion
Expression profiles and prognostic values of LSM in SKCM were inconsistent. Among the LSM family, only LSM2 may serve as a potential poor prognosticator and immunotherapeutic target of SKCM.
Collapse
|
11
|
Tumor infiltrating lymphocytes (TILs) as a predictive biomarker of response to checkpoint blockers in solid tumors: a systematic review. Crit Rev Oncol Hematol 2022; 177:103773. [PMID: 35917885 DOI: 10.1016/j.critrevonc.2022.103773] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/05/2022] [Accepted: 07/29/2022] [Indexed: 11/20/2022] Open
Abstract
Immunotherapy is a standard of care in many solid tumors but many patients derive limited benefit from it. There is increasing interest toward tumor infiltrating lymphocytes (TILs) since their presence may be related with good outcomes from treatment with immune checkpoint blockers. We aimed at systematically reviewing existing evidence about the role of TILs as possible predictors of response to immunotherapy in solid tumors. We reviewed 1193 records published from January 2010 until December 2021. Associations between TILs and outcomes were observed mainly in melanoma and breast cancer. Overall survival and overall response rate for advanced disease and pathological complete response for early-phase tumors were the most commonly assessed endpoints. No definitive conclusion can be drawn on the predictive role of TILs. Additional studies, exploiting data from prospective, randomized clinical trials should further evaluate TILs also with the aim of identifying standard cut-off to differentiate between high and low TILs.
Collapse
|
12
|
Peng J, Xiao L, Zou D, Han L. A Somatic Mutation Signature Predicts the Best Overall Response to Anti-programmed Cell Death Protein-1 Treatment in Epidermal Growth Factor Receptor/Anaplastic Lymphoma Kinase-Negative Non-squamous Non-small Cell Lung Cancer. Front Med (Lausanne) 2022; 9:808378. [PMID: 35592856 PMCID: PMC9112854 DOI: 10.3389/fmed.2022.808378] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/25/2022] [Indexed: 12/02/2022] Open
Abstract
Background We aimed to exploit a somatic mutation signature (SMS) to predict the best overall response to anti-programmed cell death protein-1 (PD-1) therapy in non-small cell lung cancer (NSCLC). Methods Tumor samples of 248 patients with epidermal growth factor receptor (EGFR)/anaplastic lymphoma kinase (ALK)-negative non-squamous NSCLC treated with anti-PD-1 were molecularly tested by targeted next-generation sequencing or whole exome sequencing. On the basis of machine learning, we developed and validated a predictive model named SMS using the training (n = 83) and validation (n = 165) cohorts. Results The SMS model comprising a panel of 15 genes (TP53, PTPRD, SMARCA4, FAT1, MGA, NOTCH1, NTRK3, INPP4B, KMT2A, PAK1, ATRX, BCOR, KDM5C, DDR2, and ARID1B) was built to predict best overall response in the training cohort. The areas under the curves of the training and validation cohorts were higher than those of tumor mutational burden and PD-L1 expression. Patients with SMS-high in the training and validation cohorts had poorer progression-free survival [hazard ratio (HR) = 6.01, P < 0.001; HR = 3.89, P < 0.001] and overall survival (HR = 7.60, P < 0.001; HR = 2.82, P < 0.001) than patients with SMS-low. SMS was an independent factor in multivariate analyses of progression-free survival and overall survival (HR = 4.32, P < 0.001; HR = 3.07, P < 0.001, respectively). Conclusion This study revealed the predictive value of SMS for immunotherapy best overall response and prognosis in EGFR/ALK-negative non-squamous NSCLC as a potential biomarker in anti-PD-1 therapy.
Collapse
Affiliation(s)
- Jie Peng
- Department of Medical Oncology, The Second Affiliated Hospital, Guizhou Medical University, Kaili City, China
| | - Lushan Xiao
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dan Zou
- Department of Medical Oncology, The Second Affiliated Hospital, Guizhou Medical University, Kaili City, China
| | - Lijie Han
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Analysis of Spatial Heterogeneity of Responses in Metastatic Sites in Renal Cell Carcinoma Patients Treated with Nivolumab. Tomography 2022; 8:1363-1373. [PMID: 35645396 PMCID: PMC9149819 DOI: 10.3390/tomography8030110] [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: 04/09/2022] [Revised: 05/14/2022] [Accepted: 05/18/2022] [Indexed: 12/04/2022] Open
Abstract
Background: The purpose was to determine whether tumor response to CPI varies by organ and to characterize response patterns in a group of surgically treated metastatic RCC patients treated with Nivolumab. Methods: A retrospective analysis was undertaken between January 2016 and March 2020 on patients receiving Nivolumab for metastatic RCC, following first-line therapy and having at least one baseline and two follow-up scans. A Fisher’s exact test was used to compare categorical variables, and a Kruskal–Wallis test was used to compare continuous variables. Results: Twenty-one out of thirty patients evaluated were eligible, and they were divided into two groups: responders (n = 11) and non-responders (n = 10). According to all iRECIST standards, 18 (85.7 percent) of the 21 patients had PD (10 patients), PR (3 patients), or SD (8 patients). At baseline, 7, 15, 4, 13, 7, and 7 patients, respectively, had detectable hepatic metastasis and lung, brain, lymph node, soft tissue, and other intra-abdominal metastases; these patients were evaluated for organ-specific response. The ORRs for hepatic metastasis and lung, brain, lymph node, soft tissue, adrenals, and other intraperitoneal metastases were correspondingly 10%, 20%, 35%, 0%, and 25%. In total, 13 (61.9%) of them demonstrated varied responses to CPI therapy, with 6 (28.5%) demonstrating intra-organ differential responses. The lymph nodes (35%) had the best objective response (BOR), followed by the adrenals and peritoneum (both 25%), the brain (20%), and the lung (20%). The response rate was highest in adrenal gland lesions (2/4; 50%), followed by lymph nodes (13/19; 68.4 percent) and liver (5/10; 50%), whereas rates were lowest for lesions in the lung (9/25; 36%), intraperitoneal metastases (1/4; 25%), and brain (1/5; 20%). Conclusions: In renal cell carcinoma, checkpoint inhibitors have a variable response at different metastatic sites, with the best response occurring in lymph nodes and the least occurring in soft tissue.
Collapse
|
14
|
Shimizu T, Miyake M, Nishimura N, Inoue K, Fujii K, Iemura Y, Ichikawa K, Omori C, Tomizawa M, Maesaka F, Oda Y, Miyamoto T, Sakamoto K, Kiba K, Tanaka M, Oyama N, Okajima E, Fujimoto K, Hori S, Morizawa Y, Gotoh D, Nakai Y, Torimoto K, Tanaka N, Fujimoto K. Organ-Specific and Mixed Responses to Pembrolizumab in Patients with Unresectable or Metastatic Urothelial Carcinoma: A Multicenter Retrospective Study. Cancers (Basel) 2022; 14:cancers14071735. [PMID: 35406508 PMCID: PMC8997142 DOI: 10.3390/cancers14071735] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 11/16/2022] Open
Abstract
To investigate the organ-specific response and clinical outcomes of mixed responses (MRs) to immune checkpoint inhibitors (ICIs) for unresectable or metastatic urothelial carcinoma (ur/mUC), we retrospectively analyzed 136 patients who received pembrolizumab. The total objective response rate (ORR) and organ-specific ORR were determined for each lesion according to the Response Evaluation Criteria in Solid Tumors version 1.1 as follows: (i) complete response (CR), (ii) partial response (PR), (iii) stable disease (SD), and (iv) progressive disease (PD). Most of the organ-specific ORR was 30−40%, but bone metastasis was only 5%. There was a significant difference in overall survival (OS) between responders and non-responders with locally advanced lesions and lymph node, lung, or liver metastases (HR 9.02 (3.63−22.4) p < 0.0001; HR 3.63 (1.97−6.69), p < 0.0001; HR 2.75 (1.35−5.59), p = 0.0053; and HR 3.17 (1.00−10.0), p = 0.049, respectively). MR was defined as occurring when PD happened in one lesion plus either CR or PR occurred in another lesion simultaneously, and 12 cases were applicable. MR was significantly associated with a poorer prognosis than that of the responder group (CR or PR; HR 0.09 (0.02−0.35), p = 0.004). Patients with bone metastases benefitted less. Care may be needed to treat patients with MR as well as patients with pure PD. Further studies should be conducted in the future.
Collapse
Affiliation(s)
- Takuto Shimizu
- Department of Urology, Nara Medical University, Kashihara 634-8522, Japan; (T.S.); (K.I.); (K.S.); (S.H.); (Y.M.); (D.G.); (Y.N.); (K.T.); (K.F.)
| | - Makito Miyake
- Department of Urology, Nara Medical University, Kashihara 634-8522, Japan; (T.S.); (K.I.); (K.S.); (S.H.); (Y.M.); (D.G.); (Y.N.); (K.T.); (K.F.)
- Correspondence: ; Tel.: +81-744-22-3051; Fax: +81-744-22-9282
| | - Nobutaka Nishimura
- Department of Urology, Okanami General Hospital, Iga 518-0842, Japan; (N.N.); (K.F.)
| | - Kuniaki Inoue
- Department of Urology, Nara Medical University, Kashihara 634-8522, Japan; (T.S.); (K.I.); (K.S.); (S.H.); (Y.M.); (D.G.); (Y.N.); (K.T.); (K.F.)
| | - Koyo Fujii
- Department of Urology, Osaka Gyoumeikan Hospital, Osaka 554-0012, Japan;
| | - Yusuke Iemura
- Department of Urology, Hirao Hospital, Kashihara 634-0076, Japan;
| | - Kazuki Ichikawa
- Department of Urology, Takai Hospital, Tenri 632-0372, Japan;
| | - Chihiro Omori
- Department of Urology, Nara Prefecture General Medical Center, Nara 630-8581, Japan;
| | - Mitsuru Tomizawa
- Department of Urology, Yamato Takada Municipal Hospital, Yamato Takada 635-8501, Japan;
| | - Fumisato Maesaka
- Department of Urology, Nara City Hospital, Nara 630-8305, Japan; (F.M.); (E.O.)
| | - Yuki Oda
- Department of Urology, Nara Prefecture Seiwa Medical Center, Ikoma 636-0802, Japan; (Y.O.); (N.O.)
| | - Tatsuki Miyamoto
- Department of Urology, Hoshigaoka Medical Center, Hirakata 573-8511, Japan;
| | - Keiichi Sakamoto
- Department of Urology, Nara Medical University, Kashihara 634-8522, Japan; (T.S.); (K.I.); (K.S.); (S.H.); (Y.M.); (D.G.); (Y.N.); (K.T.); (K.F.)
| | - Keisuke Kiba
- Department of Urology, Kindai University Nara Hospital, Ikoma 630-0293, Japan;
| | - Masahiro Tanaka
- Department of Urology, Osaka Kaisei Hospital, Osaka 532-0003, Japan;
| | - Nobuo Oyama
- Department of Urology, Nara Prefecture Seiwa Medical Center, Ikoma 636-0802, Japan; (Y.O.); (N.O.)
| | - Eijiro Okajima
- Department of Urology, Nara City Hospital, Nara 630-8305, Japan; (F.M.); (E.O.)
| | - Ken Fujimoto
- Department of Urology, Okanami General Hospital, Iga 518-0842, Japan; (N.N.); (K.F.)
| | - Shunta Hori
- Department of Urology, Nara Medical University, Kashihara 634-8522, Japan; (T.S.); (K.I.); (K.S.); (S.H.); (Y.M.); (D.G.); (Y.N.); (K.T.); (K.F.)
| | - Yosuke Morizawa
- Department of Urology, Nara Medical University, Kashihara 634-8522, Japan; (T.S.); (K.I.); (K.S.); (S.H.); (Y.M.); (D.G.); (Y.N.); (K.T.); (K.F.)
| | - Daisuke Gotoh
- Department of Urology, Nara Medical University, Kashihara 634-8522, Japan; (T.S.); (K.I.); (K.S.); (S.H.); (Y.M.); (D.G.); (Y.N.); (K.T.); (K.F.)
| | - Yasushi Nakai
- Department of Urology, Nara Medical University, Kashihara 634-8522, Japan; (T.S.); (K.I.); (K.S.); (S.H.); (Y.M.); (D.G.); (Y.N.); (K.T.); (K.F.)
| | - Kazumasa Torimoto
- Department of Urology, Nara Medical University, Kashihara 634-8522, Japan; (T.S.); (K.I.); (K.S.); (S.H.); (Y.M.); (D.G.); (Y.N.); (K.T.); (K.F.)
| | - Nobumichi Tanaka
- Department of Brachytherapy, Nara Medical University, Kashihara 634-8522, Japan;
| | - Kiyohide Fujimoto
- Department of Urology, Nara Medical University, Kashihara 634-8522, Japan; (T.S.); (K.I.); (K.S.); (S.H.); (Y.M.); (D.G.); (Y.N.); (K.T.); (K.F.)
| |
Collapse
|
15
|
Predictive Biomarkers for Outcomes of Immune Checkpoint Inhibitors (ICIs) in Melanoma: A Systematic Review. Cancers (Basel) 2021; 13:cancers13246366. [PMID: 34944986 PMCID: PMC8699321 DOI: 10.3390/cancers13246366] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have strongly improved the survival of melanoma patients. However, as durable response to ICIs are only seen in a minority, there is an unmet need to identify biomarkers that predict response. Therefore, we provide a systematic review that evaluates all biomarkers studied in association with outcomes of melanoma patients receiving ICIs. We searched Pubmed, COCHRANE Library, Embase, Emcare, and Web of Science for relevant articles that were published before June 2020 and studied blood, tumor, or fecal biomarkers that predicted response or survival in melanoma patients treated with ICIs. Of the 2536 identified reports, 177 were included in our review. Risk of bias was high in 40%, moderate in 50% and low in 10% of all studies. Biomarkers that correlated with response were myeloid-derived suppressor cells (MDSCs), circulating tumor cells (CTCs), CD8+ memory T-cells, T-cell receptor (TCR) diversity, tumor-infiltrating lymphocytes (TILs), gene expression profiling (GEP), and a favorable gut microbiome. This review shows that biomarkers for ICIs in melanoma patients are widely studied, but heterogeneity between studies is high, average sample sizes are low, and validation is often lacking. Future studies are needed to further investigate the predictive utility of some promising candidate biomarkers.
Collapse
|
16
|
Jones D, Wang Z, Chen IX, Zhang S, Banerji R, Lei PJ, Zhou H, Xiao V, Kwong C, van Wijnbergen JWM, Pereira ER, Vakoc BJ, Huang P, Nia HT, Padera TP. Solid stress impairs lymphocyte infiltration into lymph-node metastases. Nat Biomed Eng 2021; 5:1426-1436. [PMID: 34282290 PMCID: PMC8678215 DOI: 10.1038/s41551-021-00766-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 06/17/2021] [Indexed: 02/06/2023]
Abstract
Strong and durable anticancer immune responses are associated with the generation of activated cancer-specific T cells in the draining lymph nodes. However, cancer cells can colonize lymph nodes and drive tumour progression. Here, we show that lymphocytes fail to penetrate metastatic lesions in lymph nodes. In tissue from patients with breast, colon, and head and neck cancers, as well as in mice with spontaneously developing breast-cancer lymph-node metastases, we found that lymphocyte exclusion from nodal lesions is associated with the presence of solid stress caused by lesion growth, that solid stress induces reductions in the number of functional high endothelial venules in the nodes, and that relieving solid stress in the mice increased the presence of lymphocytes in lymph-node lesions by about 15-fold. Solid-stress-mediated impairment of lymphocyte infiltration into lymph-node metastases suggests a therapeutic route for overcoming T-cell exclusion during immunotherapy.
Collapse
Affiliation(s)
- Dennis Jones
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA.
| | - Zixiong Wang
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Ivy X Chen
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sue Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Rohin Banerji
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Pin-Ji Lei
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Hengbo Zhou
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Victoria Xiao
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Cecilia Kwong
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Jan Willem M van Wijnbergen
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ethel R Pereira
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Benjamin J Vakoc
- Harvard Medical School, Boston, MA, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Peigen Huang
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Hadi T Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Timothy P Padera
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Gao L, Chen Y. Autophagy controls programmed death-ligand 1 expression on cancer cells (Review). Biomed Rep 2021; 15:84. [PMID: 34512972 DOI: 10.3892/br.2021.1460] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/29/2021] [Indexed: 12/17/2022] Open
Abstract
Programmed death-ligand 1 (PD-L1) is a transmembrane protein mainly located on cancer cells, including renal cell carcinoma, breast, colorectal, gastric and non-small cell lung cancer. PD-L1 binds to the PD-1 receptor expressed on T lymphocytes to inhibit the activation of T lymphocytes, thus allowing tumour cells to escape immune surveillance, leading to tumour growth and the poor prognosis of patients with cancer. Inhibitors targeting the programmed death-1/PD-L1 axis have been widely used in the clinical treatment of a variety of solid tumours in recent years. However, the clinical efficacy of these inhibitors varies. Studies have demonstrated that the effect of the targeted drug is positively associated with the expression of PD-L1 on the tumour membrane. Hence, exploring the mechanism of PD-L1 expression is very important for the treatment of tumours. Autophagy is a physiological process that maintains the stability of the internal environment. Autophagy degrades aging organelles and long-lived proteins and produces nutrients for cell recycling. To the best of our knowledge, the present review is the first to summarize the research that has been conducted on autophagy-regulated PD-L1 expression, which may provide new avenues for tumour immunotherapy.
Collapse
Affiliation(s)
- Lijuan Gao
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,The First Clinical College of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yongshun Chen
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,The First Clinical College of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
18
|
Zhu G, Shi R, Li Y, Zhang Z, Xu S, Chen C, Cao P, Zhang H, Liu M, Pan Z, Liu H, Chen J. ARID1A, ARID1B, and ARID2 Mutations Serve as Potential Biomarkers for Immune Checkpoint Blockade in Patients With Non-Small Cell Lung Cancer. Front Immunol 2021; 12:670040. [PMID: 34512623 PMCID: PMC8426508 DOI: 10.3389/fimmu.2021.670040] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023] Open
Abstract
Worldwide, non-small cell lung cancer (NSCLC) has the highest morbidity and mortality of all malignancies. The lack of responsiveness to checkpoint inhibitors is a central problem in the modern era of cancer immunotherapy, with the rapid development of immune checkpoint inhibitors (ICIs) in recent years. The human switch/sucrose nonfermentable (SWI/SNF) chromatin-remodeling complex has been reported to be recurrently mutated in patients with cancer, and those with SWI/SNF mutations have been reported to be sensitive to ICIs. Six reported cohorts, a total of 3416 patients, were used to analyze the mutation status of ARID1A, ARID1B, ARID2 and SMARCA4 in patients with NSCLC and the effect of mutations on prognosis after ICIs. Finally, a nomogram was established to guide the clinical use of ICIs. The results show that patients with NSCLC who have ARID1A, ARID1B, and ARID2 mutations of the SWI/SNF complex were more likely to benefit from ICI therapy.
Collapse
Affiliation(s)
- Guangsheng Zhu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ruifeng Shi
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yongwen Li
- Tianjin Lung Cancer Institute, Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Medical University General Hospital, Tianjin, China
| | - Zihe Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Songlin Xu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen Chen
- Tianjin Lung Cancer Institute, Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Medical University General Hospital, Tianjin, China
| | - Peijun Cao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongbing Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Minghui Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhenhua Pan
- Tianjin Lung Cancer Institute, Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyu Liu
- Tianjin Lung Cancer Institute, Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Medical University General Hospital, Tianjin, China.,Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Lung Cancer Institute, Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
19
|
Zhu G, Ren D, Lei X, Shi R, Zhu S, Zhou N, Zu L, Mello RAD, Chen J, XU S. Mutations Associated with No Durable Clinical Benefit to Immune Checkpoint Blockade in Non-S-Cell Lung Cancer. Cancers (Basel) 2021; 13:1397. [PMID: 33808631 PMCID: PMC8003499 DOI: 10.3390/cancers13061397] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/13/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
(1) Background: The immune checkpoint blockade (ICB) has shown promising efficacy in non-small-cell lung cancer (NSCLC) patients with significant clinical benefits and durable responses, but the overall response rate to ICBs is only 20%. The lack of responsiveness to ICBs is currently a central problem in cancer immunotherapy. (2) Methods: Four public cohorts comprising 2986 patients with NSCLC were included in the study. We screened 158 patients with NSCLC with no durable clinical benefit (NDB) to ICBs in the Rizvi cohort and identified NDB-related gene mutations in these patients using univariate and multivariate Cox regression analyses. Programmed death-ligand 1 (PD-L1) expression, tumor mutation burden (TMB), neoantigen load, tumor-infiltrating lymphocytes, and immune-related gene expression were analyzed for identifying gene mutations. A comprehensive predictive classifier model was also built to evaluate the efficacy of ICB therapy. (3) Results: Mutations in FAT1 and KEAP1 were found to correlate with NDB in patients with NSCLC to ICBs; however, the analysis suggested that only mutation in FAT1 was valuable in predicting the efficacy of ICB therapy, and that mutation in KEAP1 acted as a prognostic but not a predictive biomarker for NSCLC. Mutations in FAT1 were associated with a higher TMB and lower multiple lymphocyte infiltration, including CD8 (T-Cell Surface Glycoprotein CD8)+ T cells. We established a prognostic model according to PD-L1 expression, TMB, smoking status, treatment regimen, treatment type, and FAT1 mutation, which indicated good accuracy by receiver operating characteristic (ROC) analysis (area under the curve (AUC) for 6-months survival: 0.763; AUC for 12-months survival: 0.871). (4) Conclusions: Mutation in FAT1 may be a predictive biomarker in patients with NSCLC who exhibit NDB to ICBs. We proposed an FAT1 mutation-based model for screening more suitable NSCLC patients to receive ICBs that may contribute to individualized immunotherapy.
Collapse
Affiliation(s)
- Guangsheng Zhu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300050, China; (G.Z.); (D.R.); (X.L.); (R.S.); (S.Z.); (N.Z.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300050, China
| | - Dian Ren
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300050, China; (G.Z.); (D.R.); (X.L.); (R.S.); (S.Z.); (N.Z.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300050, China
| | - Xi Lei
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300050, China; (G.Z.); (D.R.); (X.L.); (R.S.); (S.Z.); (N.Z.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300050, China
| | - Ruifeng Shi
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300050, China; (G.Z.); (D.R.); (X.L.); (R.S.); (S.Z.); (N.Z.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300050, China
| | - Shuai Zhu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300050, China; (G.Z.); (D.R.); (X.L.); (R.S.); (S.Z.); (N.Z.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300050, China
| | - Ning Zhou
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300050, China; (G.Z.); (D.R.); (X.L.); (R.S.); (S.Z.); (N.Z.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300050, China
| | - Lingling Zu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300050, China; (G.Z.); (D.R.); (X.L.); (R.S.); (S.Z.); (N.Z.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300050, China
| | - Ramon Andrade De Mello
- Escola Paulista de Medicina, Federal University of Sao Paulo, Sao Paulo 04037-004, Brazil
- Division of Oncology, Algarve Biomedical Centre, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300050, China; (G.Z.); (D.R.); (X.L.); (R.S.); (S.Z.); (N.Z.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300050, China
| | - Song XU
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300050, China; (G.Z.); (D.R.); (X.L.); (R.S.); (S.Z.); (N.Z.); (L.Z.)
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300050, China
| |
Collapse
|
20
|
Lopez-Obregon B, Barreto MP, Fyfe A, McKinnon G, Webb C, Temple-Oberle C. Evaluation of Intra-Lesional Interleukin 2 for the Treatment of In-Transit Melanoma Disease: L'évaluation de l'interleukine-2 intralésionnelle pour traiter les mélanomes en transit. Plast Surg (Oakv) 2020; 29:4-9. [PMID: 33614534 DOI: 10.1177/2292550320936669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background Intra-lesional interleukin 2 (IL-2) therapy trials for the treatment of in-transit melanoma using different treatment protocols have been published reporting varied results. This study assesses the results of IL-2 therapy in our institution and to evaluate the reproducibility of our response rates when using the same treatment protocol as another Canadian centre. Methods A retrospective review was undertaken of patients with in-transit melanoma who were treated with intralesional IL-2 in a single institution from 2010 to 2016. Responses were evaluated using RECIST criteria. Demographic data, tumour characteristics, follow-up data, in-transit-free interval, and survival data were collected and analysed. Results Forty-nine patients were identified. Overall tumour response rate was 72%, including complete response in 23 patients (47%) and partial response in 12 patients (24%). Stable disease was observed in 4% of patients and progressive disease in 25%. The main side effects were minor discomfort with injections and auto-limited flu-like symptoms. The presence of tumour-infiltrating lymphocytes may be a predictor of better response. Conclusion This study confirms prior experience with intra-lesional IL-2, demonstrating it to be an effective, safe, and well-tolerated therapy for in-transit melanoma. Tumour-infiltrating lymphocytes as a predictor of better response warrant further study.
Collapse
Affiliation(s)
- Beatriz Lopez-Obregon
- Division of Plastic Surgery, University of Calgary, Alberta, Canada
- Division of Surgical Oncology, University of Calgary, Alberta, Canada
| | - Marcio P Barreto
- Division of Plastic Surgery, University of Calgary, Alberta, Canada
- Division of Surgical Oncology, University of Calgary, Alberta, Canada
| | - Allison Fyfe
- Division of Plastic Surgery, University of Calgary, Alberta, Canada
- Division of Surgical Oncology, University of Calgary, Alberta, Canada
| | - Greg McKinnon
- Division of Plastic Surgery, University of Calgary, Alberta, Canada
- Division of Surgical Oncology, University of Calgary, Alberta, Canada
| | - Carmen Webb
- Division of Plastic Surgery, University of Calgary, Alberta, Canada
- Division of Surgical Oncology, University of Calgary, Alberta, Canada
| | - Claire Temple-Oberle
- Division of Plastic Surgery, University of Calgary, Alberta, Canada
- Division of Surgical Oncology, University of Calgary, Alberta, Canada
| |
Collapse
|
21
|
Singh MP, Sethuraman SN, Ritchey J, Fiering S, Guha C, Malayer J, Ranjan A. In-situ vaccination using focused ultrasound heating and anti-CD-40 agonistic antibody enhances T-cell mediated local and abscopal effects in murine melanoma. Int J Hyperthermia 2020; 36:64-73. [PMID: 31795832 DOI: 10.1080/02656736.2019.1663280] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The success of melanoma immunotherapy is dependent on the presence of activated and functional T-cells in tumors. The objective of this study was to investigate the impact of local-focused ultrasound (FUS) heating (∼42-45 °C) and in-situ anti-CD-40 agonistic antibody in enhancing T-cell function for melanoma immunotherapy. We compared the following groups of mice with bilateral flank B16 F10 melanoma: (1) Control, (2) FUS, (3) CD-40, and (4) CD-40 + FUS (FUS40). FUS heating was applied for ∼15 min in right flank tumor, and intratumoral injections of CD-40 were performed sequentially within 4 h. A total of 3 FUS and 4 anti-CD-40 treatments were administered unilaterally 3 days apart. Mice were sacrificed 30 days post-inoculation, and the treated tumor and spleen tissues were profiled for T-cell function and macrophage polarization. Compared to all other groups, histology and flow cytometry showed that FUS40 increased the population of tumor-specific CD-4+ and CD-8+ T cells rich in Granzyme B+, interleukin-2 (IL-2) and IFN-γ production and poor in PD-1 expression. In addition, FUS40 promoted the infiltration of tumor-suppressing M1 phenotype macrophages in the treated mice. The resultant immune-enhancing effects of FUS40 suppressed B16 melanoma growth at the treated site by 2-3-folds compared to control, FUS, and CD-40, and also achieved significant abscopal effects in untreated tumors relative to CD40 alone. Additionally, the local FUS40 prevented adverse liver toxicities in the treated mice. Our study suggests that combined FUS and CD-40 can enhance T-cell and macrophage functions to aid effective melanoma immunotherapy.
Collapse
Affiliation(s)
- Mohit Pratap Singh
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| | | | - Jerry Ritchey
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| | - Steven Fiering
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Chandan Guha
- Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jerry Malayer
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| | - Ashish Ranjan
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| |
Collapse
|
22
|
Mastracci L, Fontana V, Queirolo P, Carosio R, Grillo F, Morabito A, Banelli B, Tanda E, Boutros A, Dozin B, Gualco M, Salvi S, Romani M, Spagnolo F, Poggi A, Pistillo MP. Response to ipilimumab therapy in metastatic melanoma patients: potential relevance of CTLA-4 + tumor infiltrating lymphocytes and their in situ localization. Cancer Immunol Immunother 2020; 69:653-662. [PMID: 32025849 PMCID: PMC11027859 DOI: 10.1007/s00262-020-02494-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/18/2020] [Indexed: 12/16/2022]
Abstract
Immune checkpoint inhibitors, including ipilimumab (IPI), achieve a clinical benefit in a small proportion of melanoma patients highlighting the need to investigate predictive biomarkers. In this study, we characterized tumor infiltrating lymphocytes (TILs), focusing on the CTLA-4+ subset, and evaluated their possible predictive significance. We characterized TIL density, cell type, and localization in 40 melanoma lesions from 17 patients treated with IPI. Associations of TILs with IPI timing, tissue localization, and response to IPI were estimated using a linear mixed-effects modelling approach. We found that most of TIL subsets increased in situ upon IPI therapy, with particular reference to FoxP3+ cells. TILs and TIL subsets, such as CD3+, CD45RO+, CTLA-4+, CD4+, CD8+ T cells, CD20+ B cells, and NKp46+ NK cells, showed significantly different spatial distributions in the tumor microenvironment being higher at the invasive margin (IM) as compared to the tumor center (TC) (P value < 0.001 for TIL score and P value < 0.05 for all subsets). Remarkably, high TIL score and density of CD3+, CD8+ T cells, and CTLA-4+ immune cells were significantly associated with a better response to IPI (P values = 0.002, 0.023, 0.007, and 0.001, respectively, for responders vs non-responders). In conclusion, we provide a detailed analysis of CTLA-4+ TIL distribution in melanoma tissues taking into account localization, relationship with CD3+/CD8+ TILs, and changes in response to IPI treatment. We identified that CTLA-4+ TILs may represent a marker of IPI response, alone or with CD3+/CD8+ subsets, although this requires confirmation in larger studies.
Collapse
Affiliation(s)
- Luca Mastracci
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Genoa, Italy
- Anatomic Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Vincenzo Fontana
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Queirolo
- Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Roberta Carosio
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Federica Grillo
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Genoa, Italy
- Anatomic Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Anna Morabito
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Barbara Banelli
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Enrica Tanda
- Skin Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Andrea Boutros
- Skin Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Beatrice Dozin
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Marina Gualco
- Anatomic Pathology Unit, Ospedale Villa Scassi, ASL3, Genoa, Italy
| | - Sandra Salvi
- Anatomic Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Massimo Romani
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | | | - Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maria Pia Pistillo
- Tumor Epigenetics Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genoa, Italy.
| |
Collapse
|
23
|
Peng J, Zou D, Gong W, Kang S, Han L. Deep neural network classification based on somatic mutations potentially predicts clinical benefit of immune checkpoint blockade in lung adenocarcinoma. Oncoimmunology 2020; 9:1734156. [PMID: 32158626 PMCID: PMC7051190 DOI: 10.1080/2162402x.2020.1734156] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/03/2020] [Accepted: 01/18/2020] [Indexed: 12/26/2022] Open
Abstract
Although several biomarkers have been proposed to predict the response of patients with lung adenocarcinoma (LUAD) to immune checkpoint blockade (ICB) therapy, existing challenges such as test platform uniformity, cutoff value definition, and low frequencies restrict their effective clinical application. Here, we attempted to use deep neural networks (DNNs) based on somatic mutations to predict the clinical benefit of ICB to LUAD patients undergoing immunotherapy. We used DNNs to train and validate the predictive model in three cohorts. Kaplan-Meier estimates determined the overall survival (OS) and progression-free survival (PFS) between specific subgroups. Then, we performed a relevant analysis on the multiple-dimension data types including immune cell infiltration, programmed death receptor 1 ligand (PD-L1) expression, and tumor mutational burden (TMB) from cohorts of LUAD public database and immunotherapeutic patients. Two classification groups (C1 and C2) in the training and two validation sets were identified for the efficacy of ICB via the DNN algorithm. Patients in C1 showed remarkably long OS and PFS to programmed death 1 (PD-1) inhibitors. The C1 group was significantly associated with increased expression of immune cell infiltration, immune checkpoints, activated T-effectors, and interferon gamma signature. C1 group also exhibited significantly higher TMB, neoantigens, transversion, or transition than the C2 group. This work provides novel insights that classification of DNNs using somatic mutations in LUAD could serve as a potentially predictive approach in developing a strategy for anti-PD-1/PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Jie Peng
- Department of Medical Oncology, The Second Affiliated Hospital, Guizhou Medical University, Kaili, P.R. China
| | - Dan Zou
- Department of Medical Oncology, The Second Affiliated Hospital, Guizhou Medical University, Kaili, P.R. China
| | - Wuxing Gong
- Department of Oncology, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, P. R. China
| | - Shuai Kang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Lijie Han
- Department of Hematology and Oncology, The First Affiliated Hospital of Zheng Zhou University, Zheng Zhou, P. R. China
| |
Collapse
|
24
|
Cheng J, Ding X, Xu S, Zhu B, Jia Q. Gene expression profiling identified TP53 MutPIK3CA Wild as a potential biomarker for patients with triple-negative breast cancer treated with immune checkpoint inhibitors. Oncol Lett 2020; 19:2817-2824. [PMID: 32218835 PMCID: PMC7068237 DOI: 10.3892/ol.2020.11381] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 01/23/2020] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for 15-30% of all breast cancer cases and is clinically difficult to treat due to the lack of hormone or human epidermal growth factor receptor 2 receptors, which are usually targeted by the most successful therapeutic approaches. Immune checkpoint inhibitors (ICIs) have offered long-term survival benefits in several types of solid tumors, however with low response rates. Thus, there is an urgent need to develop feasible biomarkers for identifying patients with TNBC, who are responsive. The present study demonstrated that the immune microenvironment of TNBC has the highest expression of immunoregulatory molecules among all pathologic types. The tumor mutation burden (TMB) of TNBC was not strongly correlated with cytolytic activity and showed no significant associations with different degrees of immune cell infiltration and TMB. The machine learning method divided patients with TNBC into two groups characterized by 'hot' and 'cold' tumors, according to whether immune-associated genes were highly expressed, and different responses to immunotherapy were seen between these two groups. Furthermore, patients with a TP53MutPIK3CAWild genotype demonstrated favorable immunotherapy-responsive signatures and may have improved outcomes with ICIs. In conclusion, the present study revealed that TP53 and PIK3CA may be appropriate biomarkers to screen for patients who would benefit most from ICIs, which could guide precise immunotherapy for patients with TNBC.
Collapse
Affiliation(s)
- Jia'Nan Cheng
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing Key Laboratory of Tumor Immunotherapy, Chongqing 400037, P.R. China
| | - Xiaofang Ding
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing Key Laboratory of Tumor Immunotherapy, Chongqing 400037, P.R. China
| | - Shouxia Xu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing Key Laboratory of Tumor Immunotherapy, Chongqing 400037, P.R. China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing Key Laboratory of Tumor Immunotherapy, Chongqing 400037, P.R. China
| | - Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing Key Laboratory of Tumor Immunotherapy, Chongqing 400037, P.R. China
| |
Collapse
|
25
|
Han J, Duan J, Bai H, Wang Y, Wan R, Wang X, Chen S, Tian Y, Wang D, Fei K, Yao Z, Wang S, Lu Z, Wang Z, Wang J. TCR Repertoire Diversity of Peripheral PD-1 +CD8 + T Cells Predicts Clinical Outcomes after Immunotherapy in Patients with Non-Small Cell Lung Cancer. Cancer Immunol Res 2020; 8:146-154. [PMID: 31719056 DOI: 10.1158/2326-6066.cir-19-0398] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/20/2019] [Accepted: 11/08/2019] [Indexed: 11/16/2022]
Abstract
T-cell receptor (TCR)-based biomarkers might predict patient response to immune checkpoint blockade (ICB) but need further exploration and validation for that use. We sequenced complementarity-determining region 3 of TCRβ chains isolated from PD-1+ CD8+ T cells to investigate its value for predicting the response to anti-programmed cell death 1 (PD-1)/PD-ligand 1 (PD-L1) therapy in patients with non-small cell lung cancer (NSCLC). Two independent patient cohorts (cohort A, n = 25; cohort B, n = 15) were used as discovery and validation sets, respectively. Pre- and post-ICB peripheral blood samples were collected. In cohort A, patients with high PD-1+ CD8+ TCR diversity before ICB treatment showed better response to ICB and progression-free survival (PFS) compared with patients with low diversity [6.4 months vs. 2.5 months, HR, 0.39; 95% confidence interval (CI), 0.17-0.94; P = 0.021]. The results were validated in cohort B. Pre-ICB PD-1+ CD8+ TCR diversity achieved an optimal Youden's index of 0.81 (sensitivity = 0.87 and specificity = 0.94) for differentiating the ICB response in the merged dataset (cohort A plus cohort B). Patients with increased PD-1+ CD8+ TCR clonality after ICB treatment had longer PFS (7.3 months vs. 2.6 months, HR, 0.26; 95% CI, 0.08-0.86; P = 0.002) than those with decreased clonality. Thus, TCR diversity and clonality in peripheral blood PD-1+ CD8+ T cells may serve as noninvasive predictors of patient response to ICB and survival outcomes in NSCLC.
Collapse
MESH Headings
- Aged
- Antineoplastic Agents, Immunological/therapeutic use
- B7-H1 Antigen/antagonists & inhibitors
- Biomarkers, Tumor/analysis
- CD8-Positive T-Lymphocytes/immunology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/mortality
- Cohort Studies
- Female
- Humans
- Immunotherapy/mortality
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/mortality
- Male
- Middle Aged
- Prognosis
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Receptors, Antigen, T-Cell/classification
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Survival Rate
- Treatment Outcome
Collapse
Affiliation(s)
- Jiefei Han
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianchun Duan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua Bai
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuqi Wang
- Geneplus-Beijing Institute, Beijing, China
| | - Rui Wan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Si Chen
- Geneplus-Beijing Institute, Beijing, China
| | - Yanhua Tian
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Di Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kailun Fei
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhuoran Yao
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuhang Wang
- GCP Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhimin Lu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
26
|
Lin S, Lv Y, Xu J, Mao X, Chen Z, Lu W. Over-expression of Nav1.6 channels is associated with lymph node metastases in colorectal cancer. World J Surg Oncol 2019; 17:175. [PMID: 31672162 PMCID: PMC6824047 DOI: 10.1186/s12957-019-1715-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/27/2019] [Indexed: 11/15/2022] Open
Abstract
Background and objectives Lymph node metastasis is a key factor in predicting and determining the prognosis of patients with colorectal cancer (CRC). Sodium channels are highly expressed in a variety of tumors and are closely related to tumor development, metastasis, and invasion. We investigated the relationship between the expressions of different subtypes of Nav channels and lymph node metastasis of CRC. Methods Real-time PCR (RT-qPCR) was carried out to measure the expressions of different sodium channel subtypes, chemokine receptors (CCR2, CCR4, CCR7), and lymphocyte infiltration-related biomarkers (CD3e, CD8a, IL-2RA) in CRC tissues from 97 patients. The expressions of Nav1.5 and Nav1.6 in surgically isolated lymph nodes were detected by immunohistochemistry. Correlation analysis between expressions of different genes and lymph node metastasis was performed by two-tailed t test. Results Nav1.1 and Nav1.6 were highly expressed in CRC tissues and positively correlated with CRC lymph node metastasis. Nav1.6 was also highly expressed in metastatic lymph nodes. Further analysis showed that the high expression of Nav1.6 was closely related to the one of CCR2\CCR4 in tumor lymph node metastasis. Conclusions These results suggested that Nav1.6 might be a novel marker for CRC lymph node metastasis.
Collapse
Affiliation(s)
- Shuiquan Lin
- Department of Anorectal Surgery, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, People's Republic of China
| | - Yangbo Lv
- Department of Anorectal Surgery, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, People's Republic of China
| | - Jianguang Xu
- Department of Digestive System, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, People's Republic of China
| | - Xinglong Mao
- Department of Gastrointestinal Surgery, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, People's Republic of China
| | - Zhenhong Chen
- Department of Anorectal Surgery, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, People's Republic of China.
| | - Wuguang Lu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, People's Republic of China.
| |
Collapse
|
27
|
Fang C, Zhang C, Zhao WQ, Hu WW, Wu J, Ji M. Co-mutations of TP53 and KRAS serve as potential biomarkers for immune checkpoint blockade in squamous-cell non-small cell lung cancer: a case report. BMC Med Genomics 2019; 12:136. [PMID: 31619231 PMCID: PMC6794845 DOI: 10.1186/s12920-019-0592-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/24/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Unprecedented durable responses are identified in clinical studies to target the signaling of programmed cell death protein-1 (PD-1) as well as its ligand (PD-L1) in patients with squamous-cell non-small cell lung cancer (NSCLC). However, factors predicting the patient subtypes that are responsive to PD-1/PD-L1inhibitors have not been fully understood yet. Biomarkers, like PD-L1 expression, tumor mutational burden(TMB), DNA mismatch repair deficiency (dMMR), and tumor-infiltrating lymphocytes (TILs), have been utilized to select patients responsive to PD-1/PD-L1inhibitors in the clinic, but each of them has limited use. Lung adenocarcinoma patients with a mutation of TP53 or KRAS, particularly those with co-mutations of TP53 and KRAS, can benefit from anti-PD-1 treatment. CASE PRESENTATION In this study, the co-mutations of TP53 and KRAS in a 64-year-old non-smoking man with squamous-cell NSCLC patient was described using the next-generation sequencing (NGS) technology. The patient was treated with the pembrolizumab combined with gemcitabine as the salvage therapy, and a marked partial response could be attained, which had persisted for over 7 months. CONCLUSION In addition to testing common driving genes, like EGFR, ALK, ROS1 and BRAF, both TP53, and KRAS should also be considered in advanced or metastatic squamous-cell NSCLC.TP53 and KRAS co-mutations in squamous-cell NSCLC can be a potential factor to assess possible response to PD-1 blockade immunotherapy, but further studies with more cases are needed to confirm the prediction power.
Collapse
Affiliation(s)
- Cheng Fang
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213003 China
| | - Chu Zhang
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213003 China
| | - Wei-Qing Zhao
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213003 China
| | - Wen-Wei Hu
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213003 China
| | - Jun Wu
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213003 China
| | - Mei Ji
- Departments of Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213003 China
| |
Collapse
|
28
|
Shah N, Jacob J, Househ Z, Shiner E, Baird L, Soudy H. Unchecked immunity: a unique case of sequential immune-related adverse events with Pembrolizumab. J Immunother Cancer 2019; 7:247. [PMID: 31511075 PMCID: PMC6740018 DOI: 10.1186/s40425-019-0727-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibition has dramatically transformed the treatment of malignant melanoma. With increasing use, their unique spectrum of immune-mediated toxicity has become apparent. CASE PRESENTATION We describe a case of sequential immune-related adverse events (irAEs) in a patient with metastatic melanoma treated with single-agent anti-programmed cell death-1 (PD-1) therapy, pembrolizumab. Although numerous cases of irAEs have been reported, sequential multi-organ involvement, including progressive atopic dermatitis, vitiligo, autoimmune nephritis, autoimmune hepatitis, and autoimmune encephalitis after cessation of therapy, has not been previously documented. CONCLUSIONS Immunosuppression resulted in clinical remission of each irAE, highlighting the importance of vigilance for autoimmune complications in patients treated with checkpoint inhibition, even after immunotherapy cessation.
Collapse
MESH Headings
- Aged
- Antibodies, Monoclonal, Humanized/adverse effects
- Antineoplastic Agents, Immunological/adverse effects
- Dermatitis, Atopic/chemically induced
- Dermatitis, Atopic/drug therapy
- Dermatitis, Atopic/pathology
- Encephalitis/chemically induced
- Encephalitis/drug therapy
- Encephalitis/pathology
- Hepatitis, Autoimmune/drug therapy
- Hepatitis, Autoimmune/etiology
- Hepatitis, Autoimmune/pathology
- Humans
- Immunotherapy/adverse effects
- Male
- Melanoma/drug therapy
- Melanoma/immunology
- Melanoma/secondary
- Nephritis, Interstitial/chemically induced
- Nephritis, Interstitial/drug therapy
- Nephritis, Interstitial/pathology
- Prognosis
- Vitiligo/chemically induced
- Vitiligo/drug therapy
- Vitiligo/pathology
Collapse
Affiliation(s)
- N Shah
- St George-Sutherland Basic Physician Training Network, Kogarah, New South Wales, Australia.
| | - J Jacob
- St George-Sutherland Basic Physician Training Network, Kogarah, New South Wales, Australia
| | - Z Househ
- Department of Anatomical Pathology, SEALS, St George Hospital, Kogarah, New South Wales, Australia
| | - E Shiner
- Department of Neurology, St George Hospital, Kogarah, New South Wales, Australia
| | - L Baird
- Department of Aged Care, St George Hospital, Kogarah, New South Wales, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - H Soudy
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
- Department of Medical Oncology, St George Hospital, Kogarah, New South Wales, Australia
- Department of Medical Oncology, The Sutherland Hospital, Caringbah, New South Wales, Australia
| |
Collapse
|
29
|
Melanocyte Hyaluronan Coat Fragmentation Enhances the UVB-Induced TLR-4 Receptor Signaling and Expression of Proinflammatory Mediators IL6, IL8, CXCL1, and CXCL10 via NF-κB Activation. J Invest Dermatol 2019; 139:1993-2003.e4. [PMID: 30935974 DOI: 10.1016/j.jid.2019.03.1135] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 02/20/2019] [Accepted: 03/05/2019] [Indexed: 12/18/2022]
|
30
|
Incorvaia L, Badalamenti G, Rinaldi G, Iovanna JL, Olive D, Swayden M, Terruso L, Vincenzi B, Fulfaro F, Bazan V, Russo A, Fanale D. Can the plasma PD-1 levels predict the presence and efficiency of tumor-infiltrating lymphocytes in patients with metastatic melanoma? Ther Adv Med Oncol 2019; 11:1758835919848872. [PMID: 31205506 PMCID: PMC6535916 DOI: 10.1177/1758835919848872] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/13/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The immune response in melanoma patients is locally affected by presence of tumor-infiltrating lymphocytes (TILs), generally divided into brisk, nonbrisk, and absent. Several studies have shown that a greater presence of TILs, especially brisk, in primary melanoma is associated with a better prognosis and higher survival rate. PATIENTS AND METHODS We investigated by enzyme-linked immunosorbent assay (ELISA) the correlation between PD-1 levels in plasma and the presence/absence of TILs in 28 patients with metastatic melanoma. RESULTS Low plasma PD-1 levels were correlated with brisk TILs in primary melanoma, whereas intermediate values correlated with the nonbrisk TILs, and high PD-1 levels with absent TILs. Although the low number of samples did not allow us to obtain a statistically significant correlation between the plasma PD-1 levels and the patients' overall survival depending on the absence/presence of TILs, the median survival of patients having brisk type TILs was 5 months higher than that of patients with absent and nonbrisk TILs. CONCLUSIONS This work highlights the ability of measuring the plasma PD-1 levels in order to predict the prognosis of patients with untreated metastatic melanoma without a BRAF mutation at the time of diagnosis.
Collapse
Affiliation(s)
- Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Gaetana Rinaldi
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Juan Lucio Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 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
| | - Mirna Swayden
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Lidia Terruso
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Bruno Vincenzi
- Medical Oncology Department, University Campus Bio-Medico, Rome, Italy
| | - Fabio Fulfaro
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Viviana Bazan
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| |
Collapse
|
31
|
Antibody–Drug Conjugates: Future Directions in Clinical and Translational Strategies to Improve the Therapeutic Index. Clin Cancer Res 2019; 25:5441-5448. [DOI: 10.1158/1078-0432.ccr-19-0272] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/15/2019] [Accepted: 04/08/2019] [Indexed: 11/16/2022]
|
32
|
Koelzer VH, Sirinukunwattana K, Rittscher J, Mertz KD. Precision immunoprofiling by image analysis and artificial intelligence. Virchows Arch 2019; 474:511-522. [PMID: 30470933 PMCID: PMC6447694 DOI: 10.1007/s00428-018-2485-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 11/06/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023]
Abstract
Clinical success of immunotherapy is driving the need for new prognostic and predictive assays to inform patient selection and stratification. This requirement can be met by a combination of computational pathology and artificial intelligence. Here, we critically assess computational approaches supporting the development of a standardized methodology in the assessment of immune-oncology biomarkers, such as PD-L1 and immune cell infiltrates. We examine immunoprofiling through spatial analysis of tumor-immune cell interactions and multiplexing technologies as a predictor of patient response to cancer treatment. Further, we discuss how integrated bioinformatics can enable the amalgamation of complex morphological phenotypes with the multiomics datasets that drive precision medicine. We provide an outline to machine learning (ML) and artificial intelligence tools and illustrate fields of application in immune-oncology, such as pattern-recognition in large and complex datasets and deep learning approaches for survival analysis. Synergies of surgical pathology and computational analyses are expected to improve patient stratification in immuno-oncology. We propose that future clinical demands will be best met by (1) dedicated research at the interface of pathology and bioinformatics, supported by professional societies, and (2) the integration of data sciences and digital image analysis in the professional education of pathologists.
Collapse
Affiliation(s)
- Viktor H Koelzer
- Institute of Cancer and Genomic Science, University of Birmingham, 6 Mindelsohn Way, Birmingham, B15 2SY, UK.
- Molecular and Population Genetics Laboratory, Wellcome Centre for Human Genetics, University of Oxford, Headington, Oxford, OX3 7BN, UK.
| | - Korsuk Sirinukunwattana
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Headington, Oxford, OX3 7DQ, UK
| | - Jens Rittscher
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Headington, Oxford, OX3 7DQ, UK
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
- Target Discovery Institute, NDM Research Building, University of Oxford, Old Road Campus, Headington, OX3 7FZ, UK
| | - Kirsten D Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Mühlemattstrasse 11, CH-4410, Liestal, Switzerland
| |
Collapse
|
33
|
Szostak B, Machaj F, Rosik J, Pawlik A. CTLA4 antagonists in phase I and phase II clinical trials, current status and future perspectives for cancer therapy. Expert Opin Investig Drugs 2018; 28:149-159. [PMID: 30577709 DOI: 10.1080/13543784.2019.1559297] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION In cancer, the immune response to tumor antigens is often suppressed by inhibitors and ligands. Checkpoint blockade, considered one of the most promising frontiers for anti-cancer therapy, aims to stimulate the immune anti-cancer response. Agents such as cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) inhibitors offer prolonged survival with manageable side effects. AREAS COVERED We summarize the recent clinical successes of CTLA-4 inhibitors and place a strong emphasis on those in early phase clinical trials, often in combination with other immune check-point inhibitors, i.e., programmed cell death protein 1 (PD-1) and BRAF/mitogen-activated protein kinase inhibitors. EXPERT OPINION Recent phase I and phase II clinical trials confirm the efficacy of anti-CTLA-4 therapy for treatment of cancers such as renal cell carcinoma. These studies also indicated increased efficacy with combined immune checkpoint blockade with PD-1 or Ras/Raf/mitogen-activated protein kinase/ERK kinase (MEK)/extracellular-signal-regulated kinase (ERK) inhibitors. Researchers must search for new immune targets that may enable more effective and safe immune checkpoint blockade and cancer therapy. This goal may be achieved by next-generation combination therapies to overcome immune checkpoint therapy resistance.
Collapse
Affiliation(s)
- Bartosz Szostak
- a Department of Physiology , Pomeranian Medical University , Szczecin , Poland
| | - Filip Machaj
- a Department of Physiology , Pomeranian Medical University , Szczecin , Poland
| | - Jakub Rosik
- a Department of Physiology , Pomeranian Medical University , Szczecin , Poland
| | - Andrzej Pawlik
- a Department of Physiology , Pomeranian Medical University , Szczecin , Poland
| |
Collapse
|
34
|
Schmid S, Diem S, Li Q, Krapf M, Flatz L, Leschka S, Desbiolles L, Klingbiel D, Jochum W, Früh M. Organ-specific response to nivolumab in patients with non-small cell lung cancer (NSCLC). Cancer Immunol Immunother 2018; 67:1825-1832. [PMID: 30171269 PMCID: PMC11028265 DOI: 10.1007/s00262-018-2239-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/27/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Response to immune checkpoint inhibitors depends on tumor intrinsic properties and also on host factors in the tumour microenvironment including the presence of immune cells (IC). We hypothesized that nivolumab efficacy varies across different metastatic sites. METHODS We retrospectively analyzed computed tomography scans of patients with metastatic non-small cell lung carcinoma (NSCLC) receiving nivolumab. RECIST 1.1 criteria were applied to assess the overall response rate (ORR) and organ-specific response rate (OSRR). RESULTS We analyzed 52 patients including 44% females, 58% adenocarcinoma and 8% never smokers. Involved organs had target-lesions in the lung (42%), liver (25%), lymph nodes (56%) and soft tissue (13%) and non-target lesions in the bones (23%). ORR and disease control rate (DCR) were 20% and 45%, respectively. Median overall survival, progression-free survival and duration of response were 11.9, 2.3 and 10.3 months. OSRR and organ-specific DCR (OSDCR) were 28% and 90% in lymph nodes, 8% and 54 in the liver, and 9% and 55% in lung metastases. Nine out of 12 patients with bone metastases had progressive lesions. The cumulative incidence probability of organ-specific progression at 6 months was 14% in lymph nodes, 42% in the liver, 36% in lung metastases and 26% in the primary tumor, 29% in soft tissue and 33% in adrenal metastases. CONCLUSION In conclusion, the efficacy of immunotherapy is dependent on the metastatic location. Treatment appears more active in lymph nodes compared to other organ sites such as liver, adrenals and bone. Future strategies may include additional local treatment in case of oligoprogression in these organs in patients with otherwise sustained treatment benefit.
Collapse
Affiliation(s)
- Sabine Schmid
- Department of Oncology and Haematology, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland.
- University of Bern, Bern, Switzerland.
| | - Stefan Diem
- Department of Oncology and Haematology, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland
- Department of Immunbiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
- Department of Oncology/Haematology, Spital Grabs, Grabs, Switzerland
| | - Qiyu Li
- SAKK, Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | | | - Lukas Flatz
- Department of Immunbiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Sebastian Leschka
- Department of Radiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Lotus Desbiolles
- Department of Radiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Dirk Klingbiel
- SAKK, Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Wolfram Jochum
- Institute of Pathology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Martin Früh
- Department of Oncology and Haematology, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland
- University of Bern, Bern, Switzerland
| |
Collapse
|
35
|
Wang Z, Zhao J, Wang G, Zhang F, Zhang Z, Zhang F, Zhang Y, Dong H, Zhao X, Duan J, Bai H, Tian Y, Wan R, Han M, Cao Y, Xiong L, Liu L, Wang S, Cai S, Mok TSK, Wang J. Comutations in DNA Damage Response Pathways Serve as Potential Biomarkers for Immune Checkpoint Blockade. Cancer Res 2018; 78:6486-6496. [PMID: 30171052 DOI: 10.1158/0008-5472.can-18-1814] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/25/2018] [Accepted: 08/29/2018] [Indexed: 11/16/2022]
Abstract
Biomarkers such as programmed death receptor 1 ligand (PD-L1) expression, tumor mutational burden (TMB), and high microsatellite instability are potentially applicable to predict the efficacy of immune checkpoint blockade (ICB). However, several challenges such as defining the cut-off value, test platform uniformity, and low frequencies limit their broad clinical application. Here we identify comutations in the DNA damage response (DDR) pathways of homologous recombination repair and mismatch repair (HRR-MMR) or HRR and base excision repair (HRR-BER; defined as co-mut+) that are associated with increased TMB and neoantigen load and increased levels of immune gene expression signatures. In four public clinical cohorts, co-mut+ patients presented a higher objective response rate and a longer progression-free survival or overall survival than co-mut- patients. Overall, identification of DDR comutations in HRR-MMR or HRR-BER as predictors of response to ICB provides a potentially convenient approach for future clinical practice.Significance: Identification of comutations in specific DDR pathways as predictors of superior survival outcomes in response to immune checkpoint blockade provide a clinically convenient approach for estimation of tumor mutational burden and delivery of ICB therapy. Cancer Res; 78(22); 6486-96. ©2018 AACR.
Collapse
Affiliation(s)
- Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China.
| | - Jing Zhao
- The Medical Department, 3D Medicines Inc., Shanghai, P.R. China
| | - Guoqiang Wang
- The Medical Department, 3D Medicines Inc., Shanghai, P.R. China
| | - Fan Zhang
- Department of Oncology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Zemin Zhang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, P.R. China
| | - Fan Zhang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, P.R. China
| | - Yuzi Zhang
- The Medical Department, 3D Medicines Inc., Shanghai, P.R. China
| | - Hua Dong
- The Bioinformatics Department, R&D Center of Precision Medicine, 3D Medicines Inc., Shanghai, P.R. China
| | - Xiaochen Zhao
- The Medical Department, 3D Medicines Inc., Shanghai, P.R. China
| | - Jianchun Duan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Hua Bai
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Yanhua Tian
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Rui Wan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Miao Han
- The Bioinformatics Department, R&D Center of Precision Medicine, 3D Medicines Inc., Shanghai, P.R. China
| | - Yan Cao
- The Bioinformatics Department, R&D Center of Precision Medicine, 3D Medicines Inc., Shanghai, P.R. China
| | - Lei Xiong
- The Medical Department, 3D Medicines Inc., Shanghai, P.R. China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Shuhang Wang
- Clinical Trial Center of National Cancer Center, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Shangli Cai
- The Medical Department, 3D Medicines Inc., Shanghai, P.R. China
| | - Tony S K Mok
- Department of Clinical Oncology, State Key Laboratory of South China, Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, P.R. China.
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| |
Collapse
|
36
|
Shi Y. Regulatory mechanisms of PD-L1 expression in cancer cells. Cancer Immunol Immunother 2018; 67:1481-1489. [PMID: 30120503 PMCID: PMC11028058 DOI: 10.1007/s00262-018-2226-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/01/2018] [Indexed: 01/05/2023]
Abstract
Immunotherapy targeting the PD-L1/PD-1 pathway using antibodies is effective in the clinical treatment of a multitude of cancers. This makes research of the regulatory mechanisms of PD-1 expression in cancer cells intriguing. PD-L1 expression can be categorized into inducible expression, attributed to extrinsic factors in the microenvironment, and constitutive expression, attributed to intrinsic cancer-driving gene alteration. The mechanisms of PD-L1 expression in cancer cells operate at multiple levels, including gene amplification, chromatin modification, transcription, posttranscription, translation and posttranslation. Moreover, some open questions in this field that need to be answered in future research are proposed. Studies of regulatory mechanisms of PD-L1 expression pave the way for the application of more effective approaches in the future of cancer immunotherapy.
Collapse
Affiliation(s)
- Yongyu Shi
- Department of Immunology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, 44# Wenhua Xi Road, Jinan, 250012, China.
| |
Collapse
|
37
|
Roufas C, Chasiotis D, Makris A, Efstathiades C, Dimopoulos C, Zaravinos A. The Expression and Prognostic Impact of Immune Cytolytic Activity-Related Markers in Human Malignancies: A Comprehensive Meta-analysis. Front Oncol 2018; 8:27. [PMID: 29515971 PMCID: PMC5826382 DOI: 10.3389/fonc.2018.00027] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/29/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Recently, immune-checkpoint blockade has shown striking clinical results in different cancer patients. However, a significant inter-individual and inter-tumor variability exists among different cancers. The expression of the toxins granzyme A (GZMA) and perforin 1 (PRF1), secreted by effector cytotoxic T cells and natural killer (NK) cells, were recently used as a denominator of the intratumoral immune cytolytic activity (CYT). These levels are significantly elevated upon CD8+ T-cell activation as well as during a productive clinical response against immune-checkpoint blockade therapies. Still, it is not completely understood how different tumors induce and adapt to immune responses. METHODS Here, we calculated the CYT across different cancer types and focused on differences between primary and metastatic tumors. Using data from 10,355, primary tumor resection samples and 2,787 normal samples that we extracted from The Cancer Genome Atlas and Genotype-Tissue Expression project databases, we screened the variation of CYT across 32 different cancer types and 28 different normal tissue types. We correlated the cytolytic levels in each cancer type with the corresponding patient group's overall survival, the expression of several immune-checkpoint molecules, as well as with the load of tumor-infiltrating lymphocytes (TILs), and tumor-associated neutrophils (TANs) in these tumors. RESULTS We found diverse levels of CYT across different cancer types, with highest levels in kidney, lung, and cervical cancers, and lowest levels in glioma, adrenocortical carcinoma (ACC), and uveal melanoma. GZMA protein was either lowly expressed or absent in at least half of these tumors; whereas PRF1 protein was not detected in almost any of the different tumor types, analyzing tissue microarrays from 20 different tumor types. CYT was significantly higher in metastatic skin melanoma and correlated significantly to the TIL load. In TCGA-ACC, skin melanoma, and bladder cancer, CYT was associated with an improved patient outcome and high levels of both GZMA and PRF1 synergistically affected patient survival in these cancers. In bladder, breast, colon, esophageal, kidney, ovarian, pancreatic, testicular, and thyroid cancers, high CYT was accompanied by upregulation of at least one immune-checkpoint molecule, indicating that similar to melanoma and prostate cancer, immune responses in cytolytic-high tumors elicit immune suppression in the tumor microenvironment. CONCLUSION Overall, our data highlight the existence of diverse levels of CYT across different cancer types and suggest that along with the existence of complicated associations among various tumor-infiltrated immune cells, it is capable to promote or inhibit the establishment of a permissive tumor microenvironment, depending on the cancer type. High levels of immunosuppression seem to exist in several tumor types.
Collapse
Affiliation(s)
- Constantinos Roufas
- Department of Life Sciences, Biomedical Sciences Program, School of Sciences, European University Cyprus, Nicosia, Cyprus
- The Center for Risk and Decision Sciences (CERIDES), Department of Computer Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Dimitrios Chasiotis
- Department of Life Sciences, Biomedical Sciences Program, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Anestis Makris
- Department of Life Sciences, Biomedical Sciences Program, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Christodoulos Efstathiades
- The Center for Risk and Decision Sciences (CERIDES), Department of Computer Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Christos Dimopoulos
- The Center for Risk and Decision Sciences (CERIDES), Department of Computer Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Apostolos Zaravinos
- Department of Life Sciences, Biomedical Sciences Program, School of Sciences, European University Cyprus, Nicosia, Cyprus
| |
Collapse
|