1
|
Li Y, Ma P, Li J, Wu F, Guo M, Zhou E, Song S, Wang S, Zhang S, Jin Y. Dihydroartemisinin restores the immunogenicity and enhances the anticancer immunosurveillance of cisplatin by activating the PERK/eIF2α pathway. Cell Biosci 2024; 14:100. [PMID: 39090653 PMCID: PMC11295430 DOI: 10.1186/s13578-024-01254-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/24/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Immunosurveillance is pivotal in the effectiveness of anticancer therapies and tumor control. The ineffectiveness of cisplatin in activating the immunosurveillance is attributed to its lack of adjuvanticity resulting from its inability to stimulate endoplasmic reticulum stress. Dihydroartemisinin demonstrates the anti-tumor effects through various mechanisms, including the activation of the endoplasmic reticulum stress. This study aimed to develop a novel strategy to enhance the immunogenicity of dying tumor cells by combining cisplatin with dihydroartemisinin, thereby triggering effective anti-tumor immunosurveillance and improving the efficacy of cisplatin in clinical practice. METHODS Lewis lung carcinoma (LLC) and CT26 colon cancer cell lines and subcutaneous tumor models were used in this study. The importance of immunosurveillance was validated in both immunocompetent and immunodeficient mouse models. The ability of dihydroartemisinin and cisplatin therapy to induce immunogenic cell death and tumor growth control in vivo was validated by prophylactic tumor vaccination and therapeutic tumor models. The underlying mechanism was elucidated through the pharmaceutical or genetic intervention of the PERK/eIF2α pathway in vitro and in vivo. RESULTS Dihydroartemisinin enhanced the generation of reactive oxygen species in cisplatin-treated LLC and CT26 cancer cells. The combination treatment of dihydroartemisinin with cisplatin promoted cell death and ensured an optimal release of damage-associated molecular patterns from dying cancer cells, promoting the phagocytosis of dendritic cells. In the tumor vaccination model, we confirmed that dihydroartemisinin plus cisplatin treatment induced immunogenic cell death. Utilizing immunocompetent and immunodeficient mouse models, we further demonstrated that the combination treatment suppressed the tumor growth of CT26 colon cancer and LLC lung cancer, leading to an improved prognosis through the restoration of cytotoxic T lymphocyte responses and reinstatement of anti-cancer immunosurveillance in vivo. Mechanistically, dihydroartemisinin restored the immunogenicity of cisplatin by activating the adjuvanticity of damage-associated molecular patterns, such as calreticulin exposure, through the PERK/eIF2α pathway. Additionally, the inhibition of eIF2α phosphorylation attenuated the anti-tumor efficiency of C + D in vivo. CONCLUSIONS We highlighted that dihydroartemisinin acts as an immunogenic cell death rescuer for cisplatin, activating anticancer immunosurveillance in a PERK/eIF2α-dependent manner and offering a strategy to enhance the anti-tumor efficacy of cisplatin in clinical practice.
Collapse
Affiliation(s)
- Yumei Li
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Pulmonary Diseases of National Health Commission, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- The Ministry of Education Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Ma
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Pulmonary Diseases of National Health Commission, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jingxia Li
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Pulmonary Diseases of National Health Commission, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- The Ministry of Education Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wu
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Pulmonary Diseases of National Health Commission, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- The Ministry of Education Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Pulmonary Diseases of National Health Commission, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- The Ministry of Education Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - E Zhou
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Pulmonary Diseases of National Health Commission, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- The Ministry of Education Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Siwei Song
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Pulmonary Diseases of National Health Commission, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- The Ministry of Education Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sufei Wang
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Pulmonary Diseases of National Health Commission, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- The Ministry of Education Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Zhang
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Pulmonary Diseases of National Health Commission, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
- The Ministry of Education Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, Key Laboratory of Pulmonary Diseases of National Health Commission, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
- The Ministry of Education Key Laboratory of Biological Targeted Therapy, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Beltrán-Visiedo M, Serrano-Del Valle A, Jiménez-Aldúan N, Soler-Agesta R, Naval J, Galluzzi L, Marzo I. Cytofluorometric assessment of calreticulin exposure on CD38 + plasma cells from the human bone marrow. Methods Cell Biol 2024; 189:189-206. [PMID: 39393883 DOI: 10.1016/bs.mcb.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Exposure of the endoplasmic reticulum chaperone calreticulin (CALR) on the surface of stressed and dying cells is paramount for their effective engulfment by professional antigen-presenting cells such as dendritic cells (DCs). Importantly, this is required (but not sufficient) for DCs to initiate an adaptive immune response that culminates with an effector phase as well as with the establishment of immunological memory. Conversely, the early exposure of phosphatidylserine (PS) on the outer layer of the plasma membrane is generally associated with the rapid engulfment of stressed and dying cells by tolerogenic macrophages. Supporting the clinical relevance of the CALR exposure pathway, the spontaneous or therapy-driven translocation of CALR to the surface of malignant cells, as well as intracellular biomarkers thereof, have been associated with improved disease outcome in patients affected by a variety of neoplasms, with the notable exception of multiple myeloma (MM). Here, we describe an optimized protocol for the flow cytometry-assisted quantification of surface-exposed CALR and PS on CD38+ plasma cells from the bone marrow of patients with MM. With some variations, we expect this method to be straightforwardly adaptable to the detection of CALR and PS on the surface of cancer cells isolated from patients with neoplasms other than MM.
Collapse
Affiliation(s)
- Manuel Beltrán-Visiedo
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | | | - Nelia Jiménez-Aldúan
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain
| | - Ruth Soler-Agesta
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain
| | - Javier Naval
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| | - Isabel Marzo
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain.
| |
Collapse
|
3
|
Hijazi A, Galon J. Principles of risk assessment in colon cancer: immunity is key. Oncoimmunology 2024; 13:2347441. [PMID: 38694625 PMCID: PMC11062361 DOI: 10.1080/2162402x.2024.2347441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/16/2024] [Indexed: 05/04/2024] Open
Abstract
In clinical practice, the administration of adjuvant chemotherapy (ACT) following tumor surgical resection raises a critical dilemma for stage II colon cancer (CC) patients. The prognostic features used to identify high-risk CC patients rely on the pathological assessment of tumor cells. Currently, these factors are considered for stratifying patients who may benefit from ACT at early CC stages. However, the extent to which these factors predict clinical outcomes (i.e. recurrence, survival) remains highly controversial, also uncertainty persists regarding patients' response to treatment, necessitating further investigation. Therefore, an imperious need is to explore novel biomarkers that can reliably stratify patients at risk, to optimize adjuvant treatment decisions. Recently, we evaluated the prognostic and predictive value of Immunoscore (IS), an immune digital-pathology assay, in stage II CC patients. IS emerged as the sole significant parameter for predicting disease-free survival (DFS) in high-risk patients. Moreover, IS effectively stratified patients who would benefit most from ACT based on their risk of recurrence, thus predicting their outcomes. Notably, our findings revealed that digital IS outperformed the visual quantitative assessment of the immune response conducted by expert pathologists. The latest edition of the WHO classification for digestive tumor has introduced the evaluation of the immune response, as assessed by IS, as desirable and essential diagnostic criterion. This supports the revision of current cancer guidelines and strongly recommends the implementation of IS into clinical practice as a patient stratification tool, to guide CC treatment decisions. This approach may provide appropriate personalized therapeutic decisions that could critically impact early-stage CC patient care.
Collapse
Affiliation(s)
- Assia Hijazi
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
- Veracyte, Marseille, France
| |
Collapse
|
4
|
Hijazi A, Bifulco C, Baldin P, Galon J. Digital Pathology for Better Clinical Practice. Cancers (Basel) 2024; 16:1686. [PMID: 38730638 PMCID: PMC11083211 DOI: 10.3390/cancers16091686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
(1) Background: Digital pathology (DP) is transforming the landscape of clinical practice, offering a revolutionary approach to traditional pathology analysis and diagnosis. (2) Methods: This innovative technology involves the digitization of traditional glass slides which enables pathologists to access, analyze, and share high-resolution whole-slide images (WSI) of tissue specimens in a digital format. By integrating cutting-edge imaging technology with advanced software, DP promises to enhance clinical practice in numerous ways. DP not only improves quality assurance and standardization but also allows remote collaboration among experts for a more accurate diagnosis. Artificial intelligence (AI) in pathology significantly improves cancer diagnosis, classification, and prognosis by automating various tasks. It also enhances the spatial analysis of tumor microenvironment (TME) and enables the discovery of new biomarkers, advancing their translation for therapeutic applications. (3) Results: The AI-driven immune assays, Immunoscore (IS) and Immunoscore-Immune Checkpoint (IS-IC), have emerged as powerful tools for improving cancer diagnosis, prognosis, and treatment selection by assessing the tumor immune contexture in cancer patients. Digital IS quantitative assessment performed on hematoxylin-eosin (H&E) and CD3+/CD8+ stained slides from colon cancer patients has proven to be more reproducible, concordant, and reliable than expert pathologists' evaluation of immune response. Outperforming traditional staging systems, IS demonstrated robust potential to enhance treatment efficiency in clinical practice, ultimately advancing cancer patient care. Certainly, addressing the challenges DP has encountered is essential to ensure its successful integration into clinical guidelines and its implementation into clinical use. (4) Conclusion: The ongoing progress in DP holds the potential to revolutionize pathology practices, emphasizing the need to incorporate powerful AI technologies, including IS, into clinical settings to enhance personalized cancer therapy.
Collapse
Affiliation(s)
- Assia Hijazi
- The French National Institute of Health & Medical Research (INSERM), Laboratory of Integrative Cancer Immunology, F-75006 Paris, France;
- Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, F-75006 Paris, France
| | - Carlo Bifulco
- Providence Genomics, Portland, OR 02912, USA;
- Earle A Chiles Research Institute, Portland, OR 97213, USA
| | - Pamela Baldin
- Department of Pathology, Cliniques Universitaires Saint Luc, UCLouvain, 1200 Brussels, Belgium;
| | - Jérôme Galon
- The French National Institute of Health & Medical Research (INSERM), Laboratory of Integrative Cancer Immunology, F-75006 Paris, France;
- Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, F-75006 Paris, France
- Veracyte, 13009 Marseille, France
| |
Collapse
|
5
|
Yang Z, Teng Y, Lin M, Peng Y, Du Y, Sun Q, Gao D, Yuan Q, Zhou Y, Yang Y, Li J, Zhou Y, Li X, Qi X. Reinforced Immunogenic Endoplasmic Reticulum Stress and Oxidative Stress via an Orchestrated Nanophotoinducer to Boost Cancer Photoimmunotherapy. ACS NANO 2024; 18:7267-7286. [PMID: 38382065 DOI: 10.1021/acsnano.3c13143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Cancer progression and treatment-associated cellular stress impairs therapeutic outcome by inducing resistance. Endoplasmic reticulum (ER) stress is responsible for core events. Aberrant activation of stress sensors and their downstream components to disrupt homeostasis have emerged as vital regulators of tumor progression as well as response to cancer therapy. Here, an orchestrated nanophotoinducer (ERsNP) results in specific tumor ER-homing, induces hyperthermia and mounting oxidative stress associated reactive oxygen species (ROS), and provokes intense and lethal ER stress upon near-infrared laser irradiation. The strengthened "dying" of ER stress and ROS subsequently induce apoptosis for both primary and abscopal B16F10 and GL261 tumors, and promote damage-associated molecular patterns to evoke stress-dependent immunogenic cell death effects and release "self-antigens". Thus, there is a cascade to activate maturation of dendritic cells, reprogram myeloid-derived suppressor cells to manipulate immunosuppression, and recruit cytotoxic T lymphocytes and effective antitumor response. The long-term protection against tumor recurrence is realized through cascaded combinatorial preoperative and postoperative photoimmunotherapy including the chemokine (C-C motif) receptor 2 antagonist, ERsNP upon laser irradiation, and an immune checkpoint inhibitor. The results highlight great promise of the orchestrated nanophotoinducer to exert potent immunogenic cell stress and death by reinforcing ER stress and oxidative stress to boost cancer photoimmunotherapy.
Collapse
Affiliation(s)
- Zhenzhen Yang
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
- Drug Clinical Trial Center, Institute of Medical Innovation and Research, Peking University Third Hospital, Peking University, Beijing 100191, P.R. China
| | - Yulu Teng
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Meng Lin
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Yiwei Peng
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Yitian Du
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Qi Sun
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Datong Gao
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Quan Yuan
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Yu Zhou
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Yiliang Yang
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Jiajia Li
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Yanxia Zhou
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Xinru Li
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| | - Xianrong Qi
- Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P.R. China
| |
Collapse
|
6
|
Song G, Li M, Fan S, Qin M, Shao B, Dai W, Zhang H, Wang X, He B, Zhang Q. Boosting synergism of chemo- and immuno-therapies via switching paclitaxel-induced apoptosis to mevalonate metabolism-triggered ferroptosis by bisphosphonate coordination lipid nanogranules. Acta Pharm Sin B 2024; 14:836-853. [PMID: 38322346 PMCID: PMC10840482 DOI: 10.1016/j.apsb.2023.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 07/13/2023] [Accepted: 07/25/2023] [Indexed: 02/08/2024] Open
Abstract
Conventional chemotherapy based on cytotoxic drugs is facing tough challenges recently following the advances of monoclonal antibodies and molecularly targeted drugs. It is critical to inspire new potential to remodel the value of this classical therapeutic strategy. Here, we fabricate bisphosphonate coordination lipid nanogranules (BC-LNPs) and load paclitaxel (PTX) to boost the chemo- and immuno-therapeutic synergism of cytotoxic drugs. Alendronate in BC-LNPs@PTX, a bisphosphonate to block mevalonate metabolism, works as both the structure and drug constituent in nanogranules, where alendronate coordinated with calcium ions to form the particle core. The synergy of alendronate enhances the efficacy of paclitaxel, suppresses tumor metastasis, and alters the cytotoxic mechanism. Differing from the paclitaxel-induced apoptosis, the involvement of alendronate inhibits the mevalonate metabolism, changes the mitochondrial morphology, disturbs the redox homeostasis, and causes the accumulation of mitochondrial ROS and lethal lipid peroxides (LPO). These factors finally trigger the ferroptosis of tumor cells, an immunogenic cell death mode, which remodels the suppressive tumor immune microenvironment and synergizes with immunotherapy. Therefore, by switching paclitaxel-induced apoptosis to mevalonate metabolism-triggered ferroptosis, BC-LNPs@PTX provides new insight into the development of cytotoxic drugs and highlights the potential of metabolism regulation in cancer therapy.
Collapse
Affiliation(s)
- Ge Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Minghui Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Shumin Fan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Mengmeng Qin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bin Shao
- Department of Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing 100142, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
7
|
Nakayama Y, Ando T, Takahashi N, Tsukada K, Takagi H, Goto Y, Nakaya A, Nakada N, Yoshita H, Motoo I, Ueda A, Ueda Y, Sakumura M, Kajiura S, Ogawa K, Hosokawa A, Yasuda I. The Efficacy and Safety of Nivolumab Plus mFOLFOX6 in Gastric Cancer with Severe Peritoneal Metastasis. J Clin Med 2024; 13:834. [PMID: 38337528 PMCID: PMC10856034 DOI: 10.3390/jcm13030834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
(1) Background: Nivolumab plus chemotherapy is established as a first-line treatment for advanced gastric cancer (AGC). While mFOLFOX6 is commonly used for AGC with severe peritoneal metastasis, the efficacy of nivolumab combined with it remains uncertain. We evaluated the outcomes of nivolumab plus mFOLFOX6 for AGC with severe peritoneal metastasis in clinical practice. (2) Methods: This multicenter retrospective study was conducted between December 2021 and June 2023. We investigated AGC patients with massive ascites or inadequate oral intake due to severe peritoneal metastasis and who received nivolumab plus mFOLFOX6. (3) Results: Among 106 patients treated with nivolumab plus chemotherapy, 21 (19.8%) had severe peritoneal metastasis, with 14 receiving nivolumab plus mFOLFOX6. The median progression-free survival was 7.4 months (95%CI 1.9-10.1), and the median overall survival was 10.7 months (95%CI 5.3-NA), with four patients (28.5%) surviving more than 12 months. Improved ascites and oral intake were observed in 6/14 patients (42.8%) and 10/11 patients (90.9%), respectively. The major grade 3 or more adverse events included leukopenia (28.5%) and neutropenia (21.4%), with no severe immune-related adverse events reported. (4) Conclusions: The safety and moderate efficacy of nivolumab plus mFOLFOX6 were suggested even in AGC patients with severe peritoneal metastasis.
Collapse
Affiliation(s)
- Yurika Nakayama
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (I.M.); (A.U.); (Y.U.); (M.S.); (S.K.); (I.Y.)
| | - Takayuki Ando
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (I.M.); (A.U.); (Y.U.); (M.S.); (S.K.); (I.Y.)
| | - Naoki Takahashi
- Department of Gastroenterology, Kouseiren Takaoka Hospital, 5-10 Eirakumachi, Takaoka-shi 933-8555, Japan; (N.T.); (K.T.)
| | - Kenichiro Tsukada
- Department of Gastroenterology, Kouseiren Takaoka Hospital, 5-10 Eirakumachi, Takaoka-shi 933-8555, Japan; (N.T.); (K.T.)
| | - Hiroaki Takagi
- Department of Medical Oncology, Toyama Prefectural Central Hospital, 2-2-78 Nishinagae, Toyama-shi 930-8550, Japan; (H.T.); (K.O.)
| | - Yuno Goto
- Department of Gastroenterology, Takaoka City Hospital, 4-1 Takaramachi, Takaoka-shi 933-8550, Japan; (Y.G.); (A.N.)
| | - Atsuko Nakaya
- Department of Gastroenterology, Takaoka City Hospital, 4-1 Takaramachi, Takaoka-shi 933-8550, Japan; (Y.G.); (A.N.)
| | - Naokatsu Nakada
- Department of Gastroenterology, Itoigawa General Hospital, 457-1 Takegahana, Itoigawa-shi 941-8502, Japan;
| | - Hiroki Yoshita
- Department of Gastroenterology, Toyama Nishi General Hospital, 1019 Fuchumachi Shimokutsuwada, Toyama-shi 939-2716, Japan;
| | - Iori Motoo
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (I.M.); (A.U.); (Y.U.); (M.S.); (S.K.); (I.Y.)
| | - Akira Ueda
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (I.M.); (A.U.); (Y.U.); (M.S.); (S.K.); (I.Y.)
| | - Yuko Ueda
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (I.M.); (A.U.); (Y.U.); (M.S.); (S.K.); (I.Y.)
| | - Miho Sakumura
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (I.M.); (A.U.); (Y.U.); (M.S.); (S.K.); (I.Y.)
| | - Shinya Kajiura
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (I.M.); (A.U.); (Y.U.); (M.S.); (S.K.); (I.Y.)
| | - Kohei Ogawa
- Department of Medical Oncology, Toyama Prefectural Central Hospital, 2-2-78 Nishinagae, Toyama-shi 930-8550, Japan; (H.T.); (K.O.)
| | - Ayumu Hosokawa
- Department of Clinical Oncology, University of Miyazaki Hospital, Kihara-5200 Kiyotakecho, Miyazaki-shi 889-1692, Japan;
| | - Ichiro Yasuda
- Third Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (I.M.); (A.U.); (Y.U.); (M.S.); (S.K.); (I.Y.)
| |
Collapse
|
8
|
Eljilany I, Saghand PG, Chen J, Ratan A, McCarter M, Carpten J, Colman H, Ikeguchi AP, Puzanov I, Arnold S, Churchman M, Hwu P, Conejo-Garcia J, Dalton WS, Weiner GJ, El Naqa IM, Tarhini AA. The T Cell Immunoscore as a Reference for Biomarker Development Utilizing Real-World Data from Patients with Advanced Malignancies Treated with Immune Checkpoint Inhibitors. Cancers (Basel) 2023; 15:4913. [PMID: 37894280 PMCID: PMC10605389 DOI: 10.3390/cancers15204913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/14/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND We aimed to determine the prognostic value of an immunoscore reflecting CD3+ and CD8+ T cell density estimated from real-world transcriptomic data of a patient cohort with advanced malignancies treated with immune checkpoint inhibitors (ICIs) in an effort to validate a reference for future machine learning-based biomarker development. METHODS Transcriptomic data was collected under the Total Cancer Care Protocol (NCT03977402) Avatar® project. The real-world immunoscore for each patient was calculated based on the estimated densities of tumor CD3+ and CD8+ T cells utilizing CIBERSORTx and the LM22 gene signature matrix. Then, the immunoscore association with overall survival (OS) was estimated using Cox regression and analyzed using Kaplan-Meier curves. The OS predictions were assessed using Harrell's concordance index (C-index). The Youden index was used to identify the optimal cut-off point. Statistical significance was assessed using the log-rank test. RESULTS Our study encompassed 522 patients with four cancer types. The median duration to death was 10.5 months for the 275 participants who encountered an event. For the entire cohort, the results demonstrated that transcriptomics-based immunoscore could significantly predict patients at risk of death (p-value < 0.001). Notably, patients with an intermediate-high immunoscore achieved better OS than those with a low immunoscore. In subgroup analysis, the prediction of OS was significant for melanoma and head and neck cancer patients but did not reach significance in the non-small cell lung cancer or renal cell carcinoma cohorts. CONCLUSIONS Calculating CD3+ and CD8+ T cell immunoscore using real-world transcriptomic data represents a promising signature for estimating OS with ICIs and can be used as a reference for future machine learning-based biomarker development.
Collapse
Affiliation(s)
- Islam Eljilany
- Departments of Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Payman Ghasemi Saghand
- Department of Machine Learning, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - James Chen
- Department of Internal Medicine, Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Aakrosh Ratan
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Martin McCarter
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - John Carpten
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Howard Colman
- Department of Neurosurgery, School of Medicine, University of Utah, Salt Lake City, UT 84132, USA
- Huntsman Cancer Institute, Salt Lake City, UT 84132, USA
| | | | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Susanne Arnold
- University of Kentucky Markey Cancer Center, Lexington, KY 40536, USA
| | - Michelle Churchman
- Clinical & Life Sciences Department, Aster Insights, Hudson, FL 34667, USA
| | - Patrick Hwu
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Jose Conejo-Garcia
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | | | - George J. Weiner
- Department of Internal Medicine, Carver College of Medicine, University of Iowa Health Care, Iowa City, IA 52242, USA
| | - Issam M. El Naqa
- Department of Machine Learning, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Ahmad A. Tarhini
- Departments of Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
9
|
Willis J, Anders RA, Torigoe T, Hirohashi Y, Bifulco C, Zlobec I, Mlecnik B, Demaria S, Choi WT, Dundr P, Tatangelo F, Di Mauro A, Baldin P, Bindea G, Marliot F, Haicheur N, Fredriksen T, Kirilovsky A, Buttard B, Vasaturo A, Lafontaine L, Maby P, El Sissy C, Hijazi A, Majdi A, Lagorce C, Berger A, Van den Eynde M, Pagès F, Lugli A, Galon J. Multi-Institutional Evaluation of Pathologists' Assessment Compared to Immunoscore. Cancers (Basel) 2023; 15:4045. [PMID: 37627073 PMCID: PMC10452341 DOI: 10.3390/cancers15164045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/31/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND The Immunoscore (IS) is a quantitative digital pathology assay that evaluates the immune response in cancer patients. This study reports on the reproducibility of pathologists' visual assessment of CD3+- and CD8+-stained colon tumors, compared to IS quantification. METHODS An international group of expert pathologists evaluated 540 images from 270 randomly selected colon cancer (CC) cases. Concordance between pathologists' T-score, corresponding hematoxylin-eosin (H&E) slides, and the digital IS was evaluated for two- and three-category IS. RESULTS Non-concordant T-scores were reported in more than 92% of cases. Disagreement between semi-quantitative visual assessment of T-score and the reference IS was observed in 91% and 96% of cases before and after training, respectively. Statistical analyses showed that the concordance index between pathologists and the digital IS was weak in two- and three-category IS, respectively. After training, 42% of cases had a change in T-score, but no improvement was observed with a Kappa of 0.465 and 0.374. For the 20% of patients around the cut points, no concordance was observed between pathologists and digital pathology analysis in both two- and three-category IS, before or after training (all Kappa < 0.12). CONCLUSIONS The standardized IS assay outperformed expert pathologists' T-score evaluation in the clinical setting. This study demonstrates that digital pathology, in particular digital IS, represents a novel generation of immune pathology tools for reproducible and quantitative assessment of tumor-infiltrated immune cell subtypes.
Collapse
Affiliation(s)
- Joseph Willis
- Department of Pathology, UH Cleveland Medical Center, Cleveland, OH 44106, USA;
| | | | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (T.T.); (Y.H.)
| | - Yoshihiko Hirohashi
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; (T.T.); (Y.H.)
| | - Carlo Bifulco
- Department of Pathology and Molecular Genomics, Providence Portland Medical Center, Portland, OR 97213, USA;
| | - Inti Zlobec
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (I.Z.); (A.L.)
| | - Bernhard Mlecnik
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Inovarion, 75005 Paris, France
| | - Sandra Demaria
- Department of Pathology, Weill Cornell Medicine, New York, NY 10021, USA;
| | - Won-Tak Choi
- Department of Pathology, University of California, San Francisco, CA 94143, USA;
| | - Pavel Dundr
- Institute of Pathology, First Faculty of Medicine, Charles University, General University Hospital in Prague, 12808 Prague, Czech Republic;
| | - Fabiana Tatangelo
- Department of Pathology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy; (F.T.); (A.D.M.)
| | - Annabella Di Mauro
- Department of Pathology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy; (F.T.); (A.D.M.)
| | - Pamela Baldin
- Department of Pathology, Cliniques Universitaires St-Luc, Institut de Recherche Clinique et Experimentale (Pole GAEN), Université Catholique de Louvain, 1348 Brussels, Belgium;
| | - Gabriela Bindea
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Florence Marliot
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Nacilla Haicheur
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Tessa Fredriksen
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Amos Kirilovsky
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Bénédicte Buttard
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Angela Vasaturo
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Lucie Lafontaine
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Pauline Maby
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Carine El Sissy
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Assia Hijazi
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Amine Majdi
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Christine Lagorce
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Department of Pathology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Anne Berger
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Digestive Surgery Department, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Marc Van den Eynde
- Institut Roi Albert II, Department of Medical Oncology, Cliniques Universitaires St-Luc, Institut de Recherche Clinique et Experimentale (Pole MIRO), Université Catholique de Louvain, 1030 Brussels, Belgium;
| | - Franck Pagès
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Alessandro Lugli
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (I.Z.); (A.L.)
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France; (B.M.); (G.B.); (F.M.); (N.H.); (T.F.); (A.K.); (B.B.); (A.V.); (L.L.); (P.M.); (C.E.S.); (A.H.); (A.M.); (C.L.); (A.B.); (F.P.)
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| |
Collapse
|
10
|
Hijazi A, Antoniotti C, Cremolini C, Galon J. Light on life: immunoscore immune-checkpoint, a predictor of immunotherapy response. Oncoimmunology 2023; 12:2243169. [PMID: 37554310 PMCID: PMC10405746 DOI: 10.1080/2162402x.2023.2243169] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
In the last decade, a plethora of immunotherapeutic strategies have been designed to modulate the tumor immune microenvironment. In particular, immune checkpoint (IC) blockade therapies present the most promising advances made in cancer treatment in recent years. In non-small cell lung cancer (NSCLC), biomarkers predicting response to IC treatments are currently lacking. We have recently identified Immunoscore-IC, a powerful biomarker that predicts the efficiency of immune-checkpoint inhibitors (ICIs) in NSCLC patients. Immunoscore-IC is an in vitro diagnostic assay that quantifies densities of PD-L1+, CD8+ cells, and distances between CD8+ and PD-L1+ cells in the tumor microenvironment. Immunoscore-IC can classify responder vs non-responder NSCLC patients for ICIs therapy and is revealed as a promising predictive marker of response to anti-PD-1/PD-L1 immunotherapy in these patients. Immunoscore-IC has also shown a significant predictive value, superior to the currently used PD-L1 marker. In colorectal cancer (CRC), the addition of atezolizumab to first-line FOLFOXIRI plus bevacizumab improved progression-free survival (PFS) in patients with previously untreated metastatic CRC. In the AtezoTRIBE trial, Immunoscore-IC emerged as the first biomarker with robust predictive value in stratifying pMMR metastatic CRC patients who critically benefit from checkpoint inhibitors. Thus, Immunoscore-IC could be a universal biomarker to predict response to PD-1/PD-L1 checkpoint inhibitor immunotherapy across multiple cancer indications. Therefore, cancer patient stratification (by Immunoscore-IC), based on the presence of T lymphocytes and PD-L1 potentially provides support for clinicians to guide them through combination cancer treatment decisions.
Collapse
Affiliation(s)
- Assia Hijazi
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
| | - Carlotta Antoniotti
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France
- Veracyte, Marseille, France
| |
Collapse
|
11
|
Bausart M, Rodella G, Dumont M, Ucakar B, Vanvarenberg K, Malfanti A, Préat V. Combination of local immunogenic cell death-inducing chemotherapy and DNA vaccine increases the survival of glioblastoma-bearing mice. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 50:102681. [PMID: 37105343 DOI: 10.1016/j.nano.2023.102681] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/22/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023]
Abstract
Immunotherapy efficacy as monotherapy is negligible for glioblastoma (GBM). We hypothesized that combining therapeutic vaccination using a plasmid encoding an epitope derived from GBM-associated antigen (pTOP) with local delivery of immunogenic chemotherapy using mitoxantrone-loaded PEGylated PLGA-based nanoparticles (NP-MTX) would improve the survival of GBM-bearing mice by stimulating an antitumor immune response. We first proved that MTX retained its ability to induce cytotoxicity and immunogenic cell death of GBM cells after encapsulation. Intratumoral delivery of MTX or NP-MTX increased the frequency of IFN-γ-secreting CD8 T cells. NP-MTX mixed with free MTX in combination with pTOP DNA vaccine increased the median survival of GL261-bearing mice and increased M1-like macrophages in the brain. The addition of CpG to this combination abolished the survival benefit but led to increased M1 to M2 macrophage ratio and IFN-γ-secreting CD4 T cell frequency. These results highlight the benefits of combination strategies to potentiate immunotherapy and improve GBM outcome.
Collapse
Affiliation(s)
- Mathilde Bausart
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Giulia Rodella
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Mathilde Dumont
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Bernard Ucakar
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Kevin Vanvarenberg
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium.
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium.
| |
Collapse
|
12
|
Wang F, Lu S, Cao D, Qian J, Li C, Zhang R, Wang F, Wu M, Liu Y, Pan Z, Wu X, Lu Z, Ding P, Li L, Lin J, Catteau A, Galon J, Chen G. Prognostic and predictive value of Immunoscore and its correlation with ctDNA in stage II colorectal cancer. Oncoimmunology 2023; 12:2161167. [PMID: 36632564 PMCID: PMC9828598 DOI: 10.1080/2162402x.2022.2161167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
This study aimed to validate the prognostic value of Immunoscore (IS) in stage II colorectal cancer (CRC), and explore the roles of IS and circulating tumor DNA (ctDNA) in the adjuvant treatment for early-stage CRC. Resected tumor samples from stage II CRC patients were collected from the Sun Yat-sen University Cancer Center. The densities of CD3+ and CD8+ lymphocytes were quantified and converted to IS and classified into Low, Intermediate (Int), and High groups according to predefined cutoffs. A total of 113 patients were included in the study. Patients with IS-High, Int, and Low were 43 (38%), 62 (55%), and 8 (7%), respectively. Patients with IS-High had an excellent clinical outcome, with none recurring during a median follow-up of 3 years, including 15 (35%) clinical high-risk patients. The 3-year disease-free survival (DFS) was 100% for IS-High, 76% for IS-Int, and 47% for IS-Low (P < .001). In the multivariate Cox analysis, IS was the only significant parameter associated with DFS. IS-Int and IS-Low patients with adjuvant chemotherapy had improved DFS compared to those who did not receive adjuvant chemotherapy (HR = 0.3; 95% CI 0.1-0.92; P = .026). Among the 49 patients with postoperative ctDNA data, IS-High patients had the lowest ctDNA positivity rate, suggesting that they were most eligible for chemotherapy-free treatment. IS had a strong prognostic value in Chinese patients with stage II CRC and demonstrates its clinical utility. IS and ctDNA will jointly optimize the adjuvant treatment strategies for early-stage CRC.
Collapse
Affiliation(s)
- Fulong Wang
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Shixun Lu
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Di Cao
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Juanjuan Qian
- Department of Medicine, Genecast Biotechnology Co., Ltd, Beijing, China
| | - Cong Li
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Rongxin Zhang
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Feng Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Miaoqing Wu
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Yifan Liu
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Zhizhong Pan
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Xiaojun Wu
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Zhenhai Lu
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Peirong Ding
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Liren Li
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | - Junzhong Lin
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| | | | - Jérôme Galon
- Veracyte, Marseille, France,INSERM, Laboratory of Integrative Cancer Immunology, Paris, France,Equipe Labellisée Ligue Contre le Cancer, Paris, France,Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris, France,CONTACT Jérôme Galon INSERM, Laboratory of Integrative Cancer Immunology. Cordeliers Research Centre. 15 rue de l’école de médecine, Paris75006, France
| | - Gong Chen
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Hong Kong, China
| |
Collapse
|
13
|
Mlecnik B, Lugli A, Bindea G, Marliot F, Bifulco C, Lee JKJ, Zlobec I, Rau TT, Berger MD, Nagtegaal ID, Vink-Börger E, Hartmann A, Geppert CI, Kolwelter J, Merkel S, Grützmann R, Van den Eynde M, Jouret-Mourin A, Kartheuser A, Léonard D, Remue C, Wang J, Bavi P, Roehrl MHA, Ohashi PS, Nguyen LT, Han S, MacGregor HL, Hafezi-Bakhtiari S, Wouters BG, Masucci GV, Andersson EK, Zavadova E, Vocka M, Spacek J, Petruzelka L, Konopasek B, Dundr P, Skalova H, Nemejcova K, Botti G, Tatangelo F, Delrio P, Ciliberto G, Maio M, Laghi L, Grizzi F, Fredriksen T, Buttard B, Lafontaine L, Maby P, Majdi A, Hijazi A, El Sissy C, Kirilovsky A, Berger A, Lagorce C, Paustian C, Ballesteros-Merino C, Dijkstra J, van de Water C, Vliet SVLV, Knijn N, Mușină AM, Scripcariu DV, Popivanova B, Xu M, Fujita T, Hazama S, Suzuki N, Nagano H, Okuno K, Torigoe T, Sato N, Furuhata T, Takemasa I, Patel P, Vora HH, Shah B, Patel JB, Rajvik KN, Pandya SJ, Shukla SN, Wang Y, Zhang G, Kawakami Y, Marincola FM, Ascierto PA, Fox BA, Pagès F, Galon J. Multicenter International Study of the Consensus Immunoscore for the Prediction of Relapse and Survival in Early-Stage Colon Cancer. Cancers (Basel) 2023; 15:418. [PMID: 36672367 PMCID: PMC9856473 DOI: 10.3390/cancers15020418] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/23/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Background: The prognostic value of Immunoscore was evaluated in Stage II/III colon cancer (CC) patients, but it remains unclear in Stage I/II, and in early-stage subgroups at risk. An international Society for Immunotherapy of Cancer (SITC) study evaluated the pre-defined consensus Immunoscore in tumors from 1885 AJCC/UICC-TNM Stage I/II CC patients from Canada/USA (Cohort 1) and Europe/Asia (Cohort 2). METHODS: Digital-pathology is used to quantify the densities of CD3+ and CD8+ T-lymphocyte in the center of tumor (CT) and the invasive margin (IM). The time to recurrence (TTR) was the primary endpoint. Secondary endpoints were disease-free survival (DFS), overall survival (OS), prognosis in Stage I, Stage II, Stage II-high-risk, and microsatellite-stable (MSS) patients. RESULTS: High-Immunoscore presented with the lowest risk of recurrence in both cohorts. In Stage I/II, recurrence-free rates at 5 years were 78.4% (95%-CI, 74.4−82.6), 88.1% (95%-CI, 85.7−90.4), 93.4% (95%-CI, 91.1−95.8) in low, intermediate and high Immunoscore, respectively (HR (Hi vs. Lo) = 0.27 (95%-CI, 0.18−0.41); p < 0.0001). In Cox multivariable analysis, the association of Immunoscore to outcome was independent (TTR: HR (Hi vs. Lo) = 0.29, (95%-CI, 0.17−0.50); p < 0.0001) of the patient’s gender, T-stage, sidedness, and microsatellite instability-status (MSI). A significant association of Immunoscore with survival was found for Stage II, high-risk Stage II, T4N0 and MSS patients. The Immunoscore also showed significant association with TTR in Stage-I (HR (Hi vs. Lo) = 0.07 (95%-CI, 0.01−0.61); P = 0.016). The Immunoscore had the strongest (69.5%) contribution χ2 for influencing survival. Patients with a high Immunoscore had prolonged TTR in T4N0 tumors even for patients not receiving chemotherapy, and the Immunoscore remained the only significant parameter in multivariable analysis. CONCLUSION: In early CC, low Immunoscore reliably identifies patients at risk of relapse for whom a more intensive surveillance program or adjuvant treatment should be considered.
Collapse
Affiliation(s)
- Bernhard Mlecnik
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Inovarion, 75005 Paris, France
| | - Alessandro Lugli
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Gabriela Bindea
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Florence Marliot
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Carlo Bifulco
- Department of Pathology, Providence Portland Medical Center, Portland, OR 97213, USA
| | - Jiun-Kae Jack Lee
- Department of Biostatistics, M.D. Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Inti Zlobec
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Tilman T. Rau
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Martin D. Berger
- Department of Medical Oncology, University Hospital of Bern, 3010 Bern, Switzerland
| | - Iris D. Nagtegaal
- Pathology Department, Radboud University, 6500 HC Nijmegen, The Netherlands
| | - Elisa Vink-Börger
- Pathology Department, Radboud University, 6500 HC Nijmegen, The Netherlands
| | - Arndt Hartmann
- Department of Pathology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Carol I. Geppert
- Department of Pathology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Julie Kolwelter
- Department of Pathology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Susanne Merkel
- Department of Surgery, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Robert Grützmann
- Department of Surgery, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Marc Van den Eynde
- Institut Roi Albert II, Department of Medical Oncology, Cliniques Universitaires St-Luc, 1200 Brussels, Belgium
- Institut de Recherche Clinique et Experimentale (Pole MIRO), Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Anne Jouret-Mourin
- Department of Pathology, Cliniques Universitaires St-Luc, 1200 Brussels, Belgium
- Institut de Recherche Clinique et Experimentale (Pole GAEN), Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Alex Kartheuser
- Institut Roi Albert II, Department of Digestive Surgery, Cliniques Universitaires St-Luc Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Daniel Léonard
- Institut Roi Albert II, Department of Digestive Surgery, Cliniques Universitaires St-Luc Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Christophe Remue
- Institut Roi Albert II, Department of Digestive Surgery, Cliniques Universitaires St-Luc Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Julia Wang
- Curandis, New York, NY 10583, USA
- Department of Pathology, Laboratory Medicine Program, University Health Network, 11-E444, Toronto, ON M5G 2C4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Prashant Bavi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael H. A. Roehrl
- Department of Pathology, Laboratory Medicine Program, University Health Network, 11-E444, Toronto, ON M5G 2C4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Linh T. Nguyen
- Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| | - SeongJun Han
- Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| | | | - Sara Hafezi-Bakhtiari
- Department of Pathology, Laboratory Medicine Program, University Health Network, 11-E444, Toronto, ON M5G 2C4, Canada
| | | | - Giuseppe V. Masucci
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University, 17177 Stockholm, Sweden
| | - Emilia K. Andersson
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University, 17177 Stockholm, Sweden
| | - Eva Zavadova
- Department of Oncology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Michal Vocka
- Department of Oncology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Jan Spacek
- Department of Oncology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Lubos Petruzelka
- Department of Oncology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Bohuslav Konopasek
- Department of Oncology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Pavel Dundr
- Institute of Pathology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Helena Skalova
- Institute of Pathology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Kristyna Nemejcova
- Institute of Pathology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Gerardo Botti
- Department of Pathology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy
| | - Fabiana Tatangelo
- Department of Pathology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy
| | - Paolo Delrio
- Colorectal Surgery Department, Instituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy
| | | | - Michele Maio
- Center for Immuno-Oncology, University Hospital, 53100 Siena, Italy
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, 20090 Milan, Italy
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Fabio Grizzi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, 20090 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Tessa Fredriksen
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Bénédicte Buttard
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Lucie Lafontaine
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Pauline Maby
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Amine Majdi
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Assia Hijazi
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Carine El Sissy
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Amos Kirilovsky
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Anne Berger
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Digestive Surgery Department, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Christine Lagorce
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Department of Pathology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Christopher Paustian
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Carmen Ballesteros-Merino
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jeroen Dijkstra
- Pathology Department, Radboud University, 6500 HC Nijmegen, The Netherlands
| | | | | | - Nikki Knijn
- Pathology Department, Radboud University, 6500 HC Nijmegen, The Netherlands
| | - Ana-Maria Mușină
- Department of Surgical Oncology, Regional Institute of Oncology, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iaşi, Romania
| | - Dragos-Viorel Scripcariu
- Department of Surgical Oncology, Regional Institute of Oncology, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iaşi, Romania
| | - Boryana Popivanova
- Division of Cellular Signaling, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Mingli Xu
- Division of Cellular Signaling, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Tomonobu Fujita
- Division of Cellular Signaling, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Shoichi Hazama
- Department of Translational Research and Developmental Therapeutics against Cancer, Yamaguchi University School of Medicine, Yamaguchi 755-8505, Japan
| | - Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi 753-8511, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi 753-8511, Japan
| | - Kiyotaka Okuno
- Department of Surgery, School of Medicine, Kindai University, Osaka-sayama 589-0014, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Noriyuki Sato
- Department of Pathology, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Tomohisa Furuhata
- Department of Surgery, Surgical Oncology, and Science, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Ichiro Takemasa
- Department of Surgery, Surgical Oncology, and Science, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Prabhu Patel
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | - Hemangini H. Vora
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | - Birva Shah
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | | | - Kruti N. Rajvik
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | - Shashank J. Pandya
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | - Shilin N. Shukla
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | - Yili Wang
- Institute for Cancer Research, School of Basic Medical Science, Xi’an 710061, China
- Health Science Center of Xi’an Jiaotong University, Xi’an 710061, China
| | - Guanjun Zhang
- Institute for Cancer Research, School of Basic Medical Science, Xi’an 710061, China
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | | | - Paolo A. Ascierto
- Melanoma Cancer Immunotherapy and Innovative Therapies Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, 80131 Napoli, Italy
| | - Bernard A. Fox
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
- Laboratory of Molecular and Tumor Immunology, Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213, USA
| | - Franck Pagès
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| |
Collapse
|
14
|
Goto W, Kashiwagi S, Takada K, Asano Y, Ogisawa K, Morisaki T, Shibutani M, Tanaka H, Maeda K. Clinical verification of the relationship between serum lipid metabolism and immune activity in breast cancer patients treated with neoadjuvant chemotherapy. Eur J Med Res 2023; 28:2. [PMID: 36593486 PMCID: PMC9806883 DOI: 10.1186/s40001-022-00964-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Lipid metabolism has been recently reported to affect the prognosis and tumor immune activity in cancer patients. However, the effect of lipid metabolism on chemosensitivity in patients with breast cancer treated with neoadjuvant chemotherapy (NAC) remains unclear. METHODS We examined 327 patients with breast cancer who were treated with NAC followed by curative surgery. The correlations between the serum levels of total cholesterol (TC) and triglyceride (TG) and the clinicopathological features, including the efficacy of NAC, neutrophil-to-lymphocyte ratio (NLR), and absolute lymphocyte count (ALC), were evaluated retrospectively. RESULTS Serum TG levels were increased after NAC in all the subtypes, and the rate of change was the highest, especially in triple-negative breast cancer (TNBC) (21.0% → 48.1%). In addition, only TNBC patients with an objective response (OR) had significantly higher TG levels after NAC than those without (P = 0.049). Patients with a high ALC before NAC had significantly higher TG levels after NAC than patients with all breast cancer (P = 0.001), HER2-enriched breast cancer (P = 0.021), and TNBC (P = 0.008). Patients with a low NLR before NAC had significantly higher TG levels after NAC only among patients with TNBC (P = 0.025). In patients with human epidermal growth factor receptor 2-enriched breast cancer, the group with normal TC levels before NAC had significantly better OS than those with high TC levels (P = 0.013, log-rank test), and in patients with TNBC, the group with high TC levels after NAC had significantly better OS than those with normal TC levels (P = 0.014, log-rank test). CONCLUSIONS Good systemic immune activity and chemosensitivity may be associated with lipid metabolism regulated by NAC in TNBC patients.
Collapse
Affiliation(s)
- Wataru Goto
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Shinichiro Kashiwagi
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Koji Takada
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Yuka Asano
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Kana Ogisawa
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Tamami Morisaki
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Masatsune Shibutani
- grid.258799.80000 0004 0372 2033Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Hiroaki Tanaka
- grid.258799.80000 0004 0372 2033Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Kiyoshi Maeda
- grid.258799.80000 0004 0372 2033Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| |
Collapse
|
15
|
Tsao SY. Perspectives of traditional Chinese medicine to patch up immune checkpoint blockers. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:676-693. [PMCID: PMC9630551 DOI: 10.37349/etat.2022.00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/29/2022] [Indexed: 11/06/2022] Open
Abstract
In this era of cancer immunotherapy, the response rates of immune checkpoint blockers (ICBs) are still too low and the adverse events may also be significant. Of the ways of patching up such deficits, chemotherapy (ChT), especially if metronomic, seems promising, especially as immunity induced by immunogenic cell death (ICD) may be preserved. However, side effects, e.g., lymphocytopenia and interstitial pneumonitis cannot be ignored; eventually, resistance may also ensue. Vascular endothelial growth factors (VEGFs), being potent angiogenic factors, promote cancer cells’ purposeful angiogenesis rendering an extremely resistant tumor microenvironment (TME). This highly evasive and extremely resilient TME actually demands multi-agent, multi-target agents as currently in use through traditional Chinese medicine (TCM). With a good track record of 3,000 years, TCM is favored by mainland Chinese cancer patients. Although TCM had been criticized as unscientific and imprecise, recently, artificial intelligence (AI) technologies serve to elucidate the sound scientific basis and validity of TCM. Several TCM preparations having anti-VEGF actions are found; others suppress immune checkpoints. Especially, these herbs’ multi-prong approach appears to be more effective than Western medicine’s primarily monotherapy approach if one wishes to eradicate the very resistant TME. A “bonus” point is that some autoimmune-related adverse side effects of ICBs may also be reduced by TCM. Nevertheless, as the TCM experience is mostly anecdotal, robust clinical trials are mandatory. Moreover, other TCM problems, e.g., herbal batch variations and consistency and uniformity of herbal prescriptions are outstanding. Invariably, TCM prescriptions have daily variations as the practice of “syndrome differentiation” is hailed. Despite experienced TCM practitioners would refuse to give up their time-honored traditional practice, the multi-prong approach is still very attractive for the undue resilience of TME, let alone its good safety profile, ready availability, and eminent affordability. Although the passage is dark, light is now appearing at the end of the tunnel.
Collapse
Affiliation(s)
- Shiu Ying Tsao
- Department of Clinical Research, Hong Kong SAR Oncology Centre, Hong Kong SAR 999077, China
| |
Collapse
|
16
|
Liu P, Chen J, Zhao L, Hollebecque A, Kepp O, Zitvogel L, Kroemer G. PD-1 blockade synergizes with oxaliplatin-based, but not cisplatin-based, chemotherapy of gastric cancer. Oncoimmunology 2022; 11:2093518. [PMID: 35769948 PMCID: PMC9235886 DOI: 10.1080/2162402x.2022.2093518] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Preclinical experimentation revealed that established cancers treated with the immunogenic cell death (ICD) inducer oxaliplatin are sensitized to immune checkpoint inhibitors targeting PD-1. In contrast, no such sensitizing effect is observed when cisplatin, a non-immunogenic cell death inducer is used. Two randomized phase III clinical trials targeting unresectable gastric and gastro-esophageal junction carcinomas apparently validate this observation. Thus, oxaliplatin-based chemotherapy (together with capecitabine or 5-fluorouracil plus leucovorin) favorably interacted with nivolumab, yielding improved outcome. In contrast, the outcome of cisplatin-based chemotherapy (together with capecitabine or 5-fluorouracil) failed to be improved by concomitant treatment with pembrolizumab. These clinical findings underscore the importance of choosing appropriate ICD-inducing cytotoxicants for the development of chemoimmunotherapeutic regimens. Unfortunately, the FDA and EMA have approved PD-1 blockade in combination with “platinum-based chemotherapy” without specifying the precise nature of the platinum-containing drug. This is a non sequitur. Based on the available clinical data, such approvals should be restricted to the use of oxaliplatin.
Collapse
Affiliation(s)
- Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Jianzhou Chen
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
| | - Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Antoine Hollebecque
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France.,Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France.,Gustave Roussy, ClinicObiome, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France.,Institut du Cancer Paris CARPEM, Department of Biology, APHP, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
17
|
Orhan A, Khesrawi F, Tvilling Madsen M, Peuliche Vogelsang R, Dohrn N, Kanstrup Fiehn AM, Gögenur I. Tumor-Infiltrating Lymphocytes as Biomarkers of Treatment Response and Long-Term Survival in Patients with Rectal Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2022; 14:cancers14030636. [PMID: 35158905 PMCID: PMC8833320 DOI: 10.3390/cancers14030636] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary This study investigated tumor-infiltrating lymphocytes (TILs) in pretherapeutic biopsies as biomarkers of treatment response and long-term prognosis in patients with locally advanced rectal cancer undergoing neoadjuvant chemoradiotherapy. A systematic review and meta-analysis was performed in accordance with the PRISMA guidelines. The results indicate that it is possible to identify a sub-group of patients with improved treatment response and long-term prognosis by assessing the density of CD8+ TILs at the time of diagnosis. Abstract Neoadjuvant chemoradiotherapy (NCRT) is indicated in locally advanced rectal cancer (LARC) to downstage tumors before surgery. Watchful waiting may be a treatment option to avoid surgery in patients, obtaining a complete clinical response. However, biomarkers predictive of treatment response and long-term prognosis are lacking. Here we investigated tumor-infiltrating lymphocytes (TILs) in pretherapeutic biopsies as predictive and prognostic biomarkers. A systematic review and meta-analysis was performed in accordance with the PRISMA guidelines. In total, 429 articles were identified, of which 19 studies were included in the systematic review and 14 studies in the meta-analysis. Patients with high pretherapeutic CD8+ TILs density had an increased likelihood of achieving a pathological complete response (RR = 2.71; 95% CI: 1.58–4.66) or a complete or near-complete pathological treatment response (RR = 1.86; 95% CI: 1.50–2.29). Furthermore, high CD8+ TILs density was a favorable prognostic factor for disease-free survival (HR = 0.57; 95% CI: 0.38–0.86) and overall survival (HR = 0.43; 95% CI: 0.27–0.69). CD3+, CD4+, and FOXP3+ TILs were not identified as predictive or prognostic biomarkers. Thus, assessing pretherapeutic CD8+ TILs density may assist in identifying patients with increased sensitivity to NCRT and favorable long-term prognosis.
Collapse
Affiliation(s)
- Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Lykkebaekvej 1, DK-4600 Køge, Denmark; (F.K.); (M.T.M.); (R.P.V.); (N.D.); (A.-M.K.F.); (I.G.)
- Correspondence:
| | - Faisal Khesrawi
- Center for Surgical Science, Zealand University Hospital, Lykkebaekvej 1, DK-4600 Køge, Denmark; (F.K.); (M.T.M.); (R.P.V.); (N.D.); (A.-M.K.F.); (I.G.)
| | - Michael Tvilling Madsen
- Center for Surgical Science, Zealand University Hospital, Lykkebaekvej 1, DK-4600 Køge, Denmark; (F.K.); (M.T.M.); (R.P.V.); (N.D.); (A.-M.K.F.); (I.G.)
| | - Rasmus Peuliche Vogelsang
- Center for Surgical Science, Zealand University Hospital, Lykkebaekvej 1, DK-4600 Køge, Denmark; (F.K.); (M.T.M.); (R.P.V.); (N.D.); (A.-M.K.F.); (I.G.)
| | - Niclas Dohrn
- Center for Surgical Science, Zealand University Hospital, Lykkebaekvej 1, DK-4600 Køge, Denmark; (F.K.); (M.T.M.); (R.P.V.); (N.D.); (A.-M.K.F.); (I.G.)
- Department of Surgery, Copenhagen University Hospital, Herlev & Gentofte, Borgmester Ib Juuls Vej 1, DK-2730 Herlev, Denmark
| | - Anne-Marie Kanstrup Fiehn
- Center for Surgical Science, Zealand University Hospital, Lykkebaekvej 1, DK-4600 Køge, Denmark; (F.K.); (M.T.M.); (R.P.V.); (N.D.); (A.-M.K.F.); (I.G.)
- Department of Pathology, Zealand University Hospital, Sygehusvej 10, DK-4000 Roskilde, Denmark
- Institute for Clinical Medicine, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Lykkebaekvej 1, DK-4600 Køge, Denmark; (F.K.); (M.T.M.); (R.P.V.); (N.D.); (A.-M.K.F.); (I.G.)
- Institute for Clinical Medicine, University of Copenhagen, Blegdamsvej 3b, DK-2200 Copenhagen, Denmark
| |
Collapse
|
18
|
Zaborowski AM, Winter DC, Lynch L. The therapeutic and prognostic implications of immunobiology in colorectal cancer: a review. Br J Cancer 2021; 125:1341-1349. [PMID: 34302062 PMCID: PMC8575924 DOI: 10.1038/s41416-021-01475-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/13/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer represents the second leading cause of cancer-related death worldwide. The therapeutic field of immuno-oncology has rapidly gained momentum, with strikingly promising results observed in clinical practice. Increasing emphasis has been placed on the role of the immune response in tumorigenesis, therapy and predicting prognosis. Enhanced understanding of the dynamic and complex tumour-immune microenvironment has enabled the development of molecularly directed, individualised treatment. Analysis of intra-tumoural lymphocyte infiltration and the dichotomisation of colorectal cancer into microsatellite stable and unstable disease has important therapeutic and prognostic implications, with potential to capitalise further on this data. This review discusses the latest evidence surrounding the tumour biology and immune landscape of colorectal cancer, novel immunotherapies and the interaction of the immune system with each apex of the tripartite of cancer management (oncotherapeutics, radiotherapy and surgery). By utilising the synergy of chemotherapeutic agents and immunotherapies, and identifying prognostic and predictive immunological biomarkers, we may enter an era of unprecedented disease control, survivorship and cure rates.
Collapse
Affiliation(s)
- Alexandra M. Zaborowski
- grid.412751.40000 0001 0315 8143Centre for Colorectal Disease, St. Vincent’s University Hospital, Dublin 4, Ireland ,grid.8217.c0000 0004 1936 9705School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Des C. Winter
- grid.412751.40000 0001 0315 8143Centre for Colorectal Disease, St. Vincent’s University Hospital, Dublin 4, Ireland ,grid.7886.10000 0001 0768 2743School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Lydia Lynch
- grid.8217.c0000 0004 1936 9705School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland ,grid.38142.3c000000041936754XHarvard Institutes of Medicine, Harvard Medical School, Boston, MA USA
| |
Collapse
|
19
|
Vito A, Salem O, El-Sayes N, MacFawn IP, Portillo AL, Milne K, Harrington D, Ashkar AA, Wan Y, Workenhe ST, Nelson BH, Bruno TC, Mossman KL. Immune checkpoint blockade in triple negative breast cancer influenced by B cells through myeloid-derived suppressor cells. Commun Biol 2021; 4:859. [PMID: 34253827 PMCID: PMC8275624 DOI: 10.1038/s42003-021-02375-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022] Open
Abstract
Triple negative breast cancer holds a dismal clinical outcome and as such, patients routinely undergo aggressive, highly toxic treatment regimens. Clinical trials for TNBC employing immune checkpoint blockade in combination with chemotherapy show modest prognostic benefit, but the percentage of patients that respond to treatment is low, and patients often succumb to relapsed disease. Here, we show that a combination immunotherapy platform utilizing low dose chemotherapy (FEC) combined with oncolytic virotherapy (oHSV-1) increases tumor-infiltrating lymphocytes, in otherwise immune-bare tumors, allowing 60% of mice to achieve durable tumor regression when treated with immune checkpoint blockade. Whole-tumor RNA sequencing of mice treated with FEC + oHSV-1 shows an upregulation of B cell receptor signaling pathways and depletion of B cells prior to the start of treatment in mice results in complete loss of therapeutic efficacy and expansion of myeloid-derived suppressor cells. Additionally, RNA sequencing data shows that FEC + oHSV-1 suppresses genes associated with myeloid-derived suppressor cells, a key population of cells that drive immune escape and mediate therapeutic resistance. These findings highlight the importance of tumor-infiltrating B cells as drivers of antitumor immunity and their potential role in the regulation of myeloid-derived suppressor cells.
Collapse
Affiliation(s)
- Alyssa Vito
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Omar Salem
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Nader El-Sayes
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Ian P MacFawn
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ana L Portillo
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Katy Milne
- Deeley Research Centre, BC Cancer, Victoria, BC, Canada
| | | | - Ali A Ashkar
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Yonghong Wan
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Samuel T Workenhe
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer, Victoria, BC, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Karen L Mossman
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
20
|
Li C, Qiu Q, Gao X, Yan X, Fan C, Luo X, Liu X, Wang S, Lai X, Song Y, Deng Y. Sialic acid conjugate-modified liposomal platform modulates immunosuppressive tumor microenvironment in multiple ways for improved immune checkpoint blockade therapy. J Control Release 2021; 337:393-406. [PMID: 34171446 DOI: 10.1016/j.jconrel.2021.06.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/08/2021] [Accepted: 06/20/2021] [Indexed: 12/22/2022]
Abstract
Immune checkpoint blockade (ICB) treatment is promising for the clinical therapy of numerous malignancies. However, most cancer patients rarely benefit from such single-agent immunotherapies because of the complexity of both the tumor and tumor microenvironment. A tumor-specific liposomal vehicle (DOX-SAL) modified with a sialic acid-cholesterol conjugate (SA-CH) and remotely loaded with doxorubicin (DOX) is herein reported for improving chemoimmunotherapy. The intravenous administration of DOX-SAL dramatically downregulates tumor-associated macrophage (TAM)-mediated immunosuppression, inhibits immunoregulatory functions, and promotes intratumoral infiltration of CD8+ T cells. Compared to conventional liposomes, DOX-SAL-mediated combination therapy with a PD-1-blocking monoclonal antibody (aPD-1 mAb) almost completely eliminates B16F10 tumors and efficiently inhibits 4T1 tumors. Moreover, cancer stem cells exhibit efficient tumor-initiating, tumor-propagating, and immunosuppressive tumor microenvironment-shaping capabilities. To further improve the treatment efficacy of an immunologically "cold" tumor, metformin (MET), which selectively eradicates breast cancer tumor stem cells, is co-encapsulated with DOX into liposomes to develop DOX/MET-SAL. The combination therapy with DOX/MET-SAL and aPD-1 mAb in a 4T1 orthotopic mouse model indicates their synergetic benefit on primary tumor inhibition, metastasis suppression, and survival rate improvement. Thus, the multifunctional liposomal platform has potential value for ICB combination immunotherapy.
Collapse
Affiliation(s)
- Cong Li
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qiujun Qiu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xin Gao
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xinyang Yan
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Chuizhong Fan
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xiang Luo
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shuo Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xiaoxue Lai
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
21
|
Maby P, Bindea G, Mlecnik B, Galon J. License to kill: microsatellite instability and immune contexture. Oncoimmunology 2021; 10:1905935. [PMID: 33868790 PMCID: PMC8023238 DOI: 10.1080/2162402x.2021.1905935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancers (CRCs) with microsatellite instability (MSI) are due to a defect in the DNA mismatch repair (MMR) system resulting in an accumulation of frame-shift mutations. They are characterized by a tumor microenvironment richer in cytotoxic CD8 T-cells (CTLs) and a better prognosis compared to microsatellite stable (MSS) CRCs. The mechanisms by which defective MMR system may influence tumor-infiltrating immune cells and their impact on patient survival were still unclear. Thus, we performed a comprehensive analysis of MSI colorectal tumors. We found that the numbers of frame-shift mutations potentially resulting in neo-epitopes were positively correlated to the density of tumor infiltrating CD8 T-cells but were lower than expected at random. We also evidenced that MSI patients could naturally harbor CTLs targeting frame-shift mutation-derived antigens. This favors the hypothesis of an active immunosurveillance in MSI colorectal tumors leading to the genetic evidence of an immunoediting. To evaluate the link between MSI tumor immune contexture and prognosis, we took advantage of a standardized assay that we developed to quantify tumor-infiltrating T-cells, the Immunoscore. Multivariate analyses revealed an advantage of Immunoscore over MSI in predicting recurrence and survival. Our data suggests that the prognostic value of MSI could be attributed to major underlying differences of infiltrating immune cells. Immunotherapeutic treatments, that are more efficient in patients with a preexisting anti-tumor immunity, were approved in MSI patients following successful clinical trials. We suggest that the Immunoscore could be used not only for colorectal tumor prognosis but also for predicting responses to immunotherapies.
Collapse
Affiliation(s)
- Pauline Maby
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre Le Cancer, Paris, France.,Centre De Recherche Des Cordeliers, Sorbonne Université, Université De Paris, Paris, France
| | - Gabriela Bindea
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre Le Cancer, Paris, France.,Centre De Recherche Des Cordeliers, Sorbonne Université, Université De Paris, Paris, France
| | - Bernhard Mlecnik
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre Le Cancer, Paris, France.,Centre De Recherche Des Cordeliers, Sorbonne Université, Université De Paris, Paris, France.,Inovarion, Paris, France
| | - Jérôme Galon
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre Le Cancer, Paris, France.,Centre De Recherche Des Cordeliers, Sorbonne Université, Université De Paris, Paris, France
| |
Collapse
|
22
|
Cytotoxic chemotherapeutic agents and the EGFR-TKI osimertinib induce calreticulin exposure in non-small cell lung cancer. Lung Cancer 2021; 155:144-150. [PMID: 33819860 DOI: 10.1016/j.lungcan.2021.03.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/27/2021] [Accepted: 03/21/2021] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Synergistic anticancer efficacy of combination treatment with immune checkpoint inhibitors (ICIs) and platinum-based chemotherapy in patients with advanced non-small cell lung cancer (NSCLC) may be attributable in part to the phenomenon of immunogenic cell death (ICD), which is characterized by the release of damage-associated molecular patterns (DAMPs) from dying tumor cells. The ability of cytotoxic chemotherapeutic agents and molecularly targeted drugs such as epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) to induce DAMPs during the treatment of NSCLC has remained unclear, however. MATERIALS AND METHODS We investigated the ability of seven cytotoxic chemotherapeutic agents and the third-generation EGFR-TKI osimertinib to induce translocation of the DAMP calreticulin to the cell surface in multiple NSCLC cell lines. The plasma concentration of soluble CRT in advanced NSCLC patients treated with cytotoxic chemotherapy or osimertinib was measured. RESULTS Antimetabolites and microtubule inhibitors induced expression of CRT at the cell surface (ecto-CRT) to a greater extent than did platinum agents in six NSCLC cell lines, exhibiting higher up-regulation of phosphorylation of eukaryotic initiation factor-2α (eIF2α). Ecto-CRT expression was positively correlated with apoptosis induction in NSCLC cells treated with these various chemotherapeutic agents. The drug-induced up-regulation of ecto-CRT in NSCLC cells was attenuated by the pan-caspase inhibitor Z-VAD-FMK. Osimertinib similarly increased ecto-CRT expression in association with apoptosis induction in five EGFR-mutated NSCLC cell lines. Furthermore, the plasma concentration of soluble CRT in 16 NSCLC patients treated with single-agent pemetrexed or docetaxel and in nine EGFR-mutated NSCLC patients treated with osimertinib was increased after treatment onset. CONCLUSION Our findings indicate that antimetabolites, microtubule inhibitors, and osimertinib are effective inducers both of CRT exposure in NSCLC cell lines and of soluble CRT release in patients with advanced NSCLC, suggesting that these agents might prove effective for promotion of antitumor immunity in combination immunotherapy.
Collapse
|
23
|
Nassif EF, Thibault C, Oudard S, Galon J. Precision immunity: Immunoscore and neoadjuvant treatment in bladder cancer. Oncoimmunology 2021; 10:1888488. [PMID: 33659099 PMCID: PMC7899086 DOI: 10.1080/2162402x.2021.1888488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This review details the clinical utility of Immunoscore, measuring the immune response to cancer within the tumor microenvironment, in bladder cancer. Immunoscore was recently introduced into ESMO Clinical Practice Guidelines for gastrointestinal cancer and into the WHO classification of the Digestive System Tumors. In muscle-invasive bladder cancer (MIBC), the standard-of-care treatment is neo-adjuvant chemotherapy and cystectomy. However, only 50% of the patients are still alive at 5 years. The degree of histologic response positively correlated with Immunoscore and patients at lower risk of relapse or death were associated with a high-Immunoscore. Immunoscore is also predicting response to neoadjuvant chemotherapy-based treatment in several indications. This paves the way for the use of Immunoscore in clinical practice not only in gastrointestinal tumors but also in bladder cancer, and beyond.
Collapse
Affiliation(s)
- Elise F Nassif
- Oncology Department, Hopital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, University of Paris, Paris, France
| | - Constance Thibault
- Oncology Department, Hopital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, University of Paris, Paris, France.,Université de Paris, Paris, France
| | - Stéphane Oudard
- Oncology Department, Hopital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, University of Paris, Paris, France.,Université de Paris, Paris, France
| | - Jérôme Galon
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
24
|
Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology 2021; 1:894-907. [PMID: 23162757 PMCID: PMC3489745 DOI: 10.4161/onci.20931] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptors (TLRs) have first been characterized for their capacity to detect conserved microbial components like lipopolysaccharide (LPS) and double-stranded RNA, resulting in the elicitation of potent (innate) immune responses against invading pathogens. More recently, TLRs have also been shown to promote the activation of the cognate immune system against cancer cells. Today, only three TLR agonists are approved by FDA for use in humans: the bacillus Calmette-Guérin (BCG), monophosphoryl lipid A (MPL) and imiquimod. BCG (an attenuated strain of Mycobacterium bovis) is mainly used as a vaccine against tuberculosis, but also for the immunotherapy of in situ bladder carcinoma. MPL (derived from the LPS of Salmonella minnesota) is included in the formulation of Cervarix®, a vaccine against human papillomavirus-16 and -18. Imiquimod (a synthetic imidazoquinoline) is routinely employed for actinic keratosis, superficial basal cell carcinoma, and external genital warts (condylomata acuminata). In this Trial Watch, we will summarize the results of recently completed clinical trials and discuss the progress of ongoing studies that have evaluated/are evaluating FDA-approved TLR agonists as off-label medications for cancer therapy.
Collapse
Affiliation(s)
- Erika Vacchelli
- INSERM, U848; Villejuif, France ; Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Vacchelli E, Aranda F, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Tumor-targeting monoclonal antibodies in cancer therapy. Oncoimmunology 2021; 3:e27048. [PMID: 24605265 PMCID: PMC3937194 DOI: 10.4161/onci.27048] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 11/01/2013] [Indexed: 02/06/2023] Open
Abstract
In 1997, for the first time in history, a monoclonal antibody (mAb), i.e., the chimeric anti-CD20 molecule rituximab, was approved by the US Food and Drug Administration for use in cancer patients. Since then, the panel of mAbs that are approved by international regulatory agencies for the treatment of hematopoietic and solid malignancies has not stopped to expand, nowadays encompassing a stunning amount of 15 distinct molecules. This therapeutic armamentarium includes mAbs that target tumor-associated antigens, as well as molecules that interfere with tumor-stroma interactions or exert direct immunostimulatory effects. These three classes of mAbs exert antineoplastic activity via distinct mechanisms, which may or may not involve immune effectors other than the mAbs themselves. In previous issues of OncoImmunology, we provided a brief scientific background to the use of mAbs, all types confounded, in cancer therapy, and discussed the results of recent clinical trials investigating the safety and efficacy of this approach. Here, we focus on mAbs that primarily target malignant cells or their interactions with stromal components, as opposed to mAbs that mediate antineoplastic effects by activating the immune system. In particular, we discuss relevant clinical findings that have been published during the last 13 months as well as clinical trials that have been launched in the same period to investigate the therapeutic profile of hitherto investigational tumor-targeting mAbs.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Fernando Aranda
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| | | | - Jérôme Galon
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, U872; Paris, France ; Equipe 15, Centre de Recherche des Cordeliers; Paris, France
| | - Catherine Sautès-Fridman
- Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, U872; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| |
Collapse
|
26
|
Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology 2021; 1:1557-1576. [PMID: 23264902 PMCID: PMC3525611 DOI: 10.4161/onci.22428] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Prophylactic vaccination constitutes one of the most prominent medical achievements of history. This concept was first demonstrated by the pioneer work of Edward Jenner, dating back to the late 1790s, after which an array of preparations that confer life-long protective immunity against several infectious agents has been developed. The ensuing implementation of nation-wide vaccination programs has de facto abated the incidence of dreadful diseases including rabies, typhoid, cholera and many others. Among all, the most impressive result of vaccination campaigns is surely represented by the eradication of natural smallpox infection, which was definitively certified by the WHO in 1980. The idea of employing vaccines as anticancer interventions was first theorized in the 1890s by Paul Ehrlich and William Coley. However, it soon became clear that while vaccination could be efficiently employed as a preventive measure against infectious agents, anticancer vaccines would have to (1) operate as therapeutic, rather than preventive, interventions (at least in the vast majority of settings), and (2) circumvent the fact that tumor cells often fail to elicit immune responses. During the past 30 y, along with the recognition that the immune system is not irresponsive to tumors (as it was initially thought) and that malignant cells express tumor-associated antigens whereby they can be discriminated from normal cells, considerable efforts have been dedicated to the development of anticancer vaccines. Some of these approaches, encompassing cell-based, DNA-based and purified component-based preparations, have already been shown to exert conspicuous anticancer effects in cohorts of patients affected by both hematological and solid malignancies. In this Trial Watch, we will summarize the results of recent clinical trials that have evaluated/are evaluating purified peptides or full-length proteins as therapeutic interventions against cancer.
Collapse
Affiliation(s)
- Erika Vacchelli
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kepp O, Menger L, Vacchelli E, Adjemian S, Martins I, Ma Y, Sukkurwala AQ, Michaud M, Galluzzi L, Zitvogel L, Kroemer G. Anticancer activity of cardiac glycosides: At the frontier between cell-autonomous and immunological effects. Oncoimmunology 2021; 1:1640-1642. [PMID: 23264921 PMCID: PMC3525630 DOI: 10.4161/onci.21684] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Retrospective clinical data indicate that cardiac glycosides (CGs), notably digoxin, prolong the survival of carcinoma patients treated with conventional chemotherapy. CGs are known to influence the immune response at multiple levels. In addition, recent results suggest that CGs trigger the immunogenic demise of cancer cells, an effect that most likely contributes to their clinical anticancer activity.
Collapse
Affiliation(s)
- Oliver Kepp
- INSERM; U848; Villejuif, France ; Institut Gustave Roussy; Villejuif, France ; Université Paris Sud/Paris XI; Le Kremlin Bicêtre, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, Sautès-Fridman C, Ma Y, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology 2021; 1:1323-1343. [PMID: 23243596 PMCID: PMC3518505 DOI: 10.4161/onci.22009] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Solid tumors are constituted of a variety of cellular components, including bona fide malignant cells as well as endothelial, structural and immune cells. On one hand, the tumor stroma exerts major pro-tumorigenic and immunosuppressive functions, reflecting the capacity of cancer cells to shape the microenvironment to satisfy their own metabolic and immunological needs. On the other hand, there is a component of tumor-infiltrating leucocytes (TILs) that has been specifically recruited in the attempt to control tumor growth. Along with the recognition of the critical role played by the immune system in oncogenesis, tumor progression and response to therapy, increasing attention has been attracted by the potential prognostic and/or predictive role of the immune infiltrate in this setting. Data from large clinical studies demonstrate indeed that a robust infiltration of neoplastic lesions by specific immune cell populations, including (but not limited to) CD8+ cytotoxic T lymphocytes, Th1 and Th17 CD4+ T cells, natural killer cells, dendritic cells, and M1 macrophages constitutes an independent prognostic indicator in several types of cancer. Conversely, high levels of intratumoral CD4+CD25+FOXP3+ regulatory T cells, Th2 CD4+ T cells, myeloid-derived suppressor cells, M2 macrophages and neutrophils have frequently been associated with dismal prognosis. So far, only a few studies have addressed the true predictive potential of TILs in cancer patients, generally comforting the notion that—at least in some clinical settings—the immune infiltrate can reliably predict if a specific patient will respond to therapy or not. In this Trial Watch, we will summarize the results of clinical trials that have evaluated/are evaluating the prognostic and predictive value of the immune infiltrate in the context of solid malignancies.
Collapse
Affiliation(s)
- Laura Senovilla
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Orsay, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology 2021; 1:699-716. [PMID: 22934262 PMCID: PMC3429574 DOI: 10.4161/onci.20696] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are prototypic pattern recognition receptors (PRRs) best known for their ability to activate the innate immune system in response to conserved microbial components such as lipopolysaccharide and double-stranded RNA. Accumulating evidence indicates that the function of TLRs is not restricted to the elicitation of innate immune responses against invading pathogens. TLRs have indeed been shown to participate in tissue repair and injury-induced regeneration as well as in adaptive immune responses against cancer. In particular, TLR4 signaling appears to be required for the efficient processing and cross-presentation of cell-associated tumor antigens by dendritic cells, which de facto underlie optimal therapeutic responses to some anticancer drugs. Thus, TLRs constitute prominent therapeutic targets for the activation/intensification of anticancer immune responses. In line with this notion, long-used preparations such as the Coley toxin (a mixture of killed Streptococcus pyogenes and Serratia marcescens bacteria) and the bacillus Calmette-Guérin (BCG, an attenuated strain of Mycobacterium bovis originally developed as a vaccine against tuberculosis), both of which have been associated with consistent anticancer responses, potently activate TLR2 and TLR4 signaling. Today, besides BCG, only one TLR agonist is FDA-approved for therapeutic use in cancer patients: imiquimod. In this Trial Watch, we will briefly present the role of TLRs in innate and cognate immunity and discuss the progress of clinical studies evaluating the safety and efficacy of experimental TLR agonists as immunostimulatory agents for oncological indications.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Institut Gustave Roussy; Villejuif, France
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Damage-associated molecular pattern (DAMP) activation in melanoma: investigation of the immunogenic activity of 15-deoxy, Δ 12,14 prostamide J 2. Oncotarget 2020; 11:4788-4802. [PMID: 33447347 PMCID: PMC7779254 DOI: 10.18632/oncotarget.27856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/16/2020] [Indexed: 11/25/2022] Open
Abstract
Metastatic melanoma is the most deadly skin neoplasm in the United States. Outcomes for this lethal disease have improved dramatically due to the use of both targeted and immunostimulatory drugs. Immunogenic cell death (ICD) has emerged as another approach for initiating antitumor immunity. ICD is triggered by tumor cells that display damage-associated molecular patterns (DAMPs). These DAMP molecules recruit and activate dendritic cells (DCs) that present tumor-specific antigens to T cells which eliminate neoplastic cells. Interestingly, the expression of DAMP molecules occurs in an endoplasmic reticulum (ER) stress-dependent manner. We have previously shown that ER stress was required for the cytotoxic activity of the endocannabinoid metabolite, 15-deoxy, Δ12,14 prostamide J2 (15dPMJ2). As such, the current study investigates whether 15dPMJ2 induces DAMP signaling in melanoma. In B16F10 cells, 15dPMJ2 caused a significant increase in the cell surface expression of calreticulin (CRT), the release of ATP and the secretion of high-mobility group box 1 (HMGB1), three molecules that serve as surrogate markers of ICD. 15dPMJ2 also stimulated the cell surface expression of the DAMP molecules, heat shock protein 70 (Hsp70) and Hsp90. In addition, the display of CRT and ATP was increased by 15dPMJ2 to a greater extent in tumorigenic compared to non-tumorigenic melanocytes. Consistent with this finding, the activation of bone marrow-derived DCs was upregulated in co-cultures with 15dPMJ2-treated tumor compared to non-tumor melanocytes. Moreover, 15dPMJ2-mediated DAMP exposure and DC activation required the electrophilic cyclopentenone double bond within the structure of 15dPMJ2 and the ER stress pathway. These results demonstrate that 15dPMJ2 is a tumor-selective inducer of DAMP signaling in melanoma.
Collapse
|
31
|
Bruni D, Angell HK, Galon J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat Rev Cancer 2020; 20:662-680. [PMID: 32753728 DOI: 10.1038/s41568-020-0285-7] [Citation(s) in RCA: 1019] [Impact Index Per Article: 203.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
Abstract
The international American Joint Committee on Cancer/Union for International Cancer Control (AJCC/UICC) tumour-node-metastasis (TNM) staging system provides the current guidelines for the classification of cancer. However, among patients within the same stage, the clinical outcome can be very different. More recently, a novel definition of cancer has emerged, implicating at all stages a complex and dynamic interaction between tumour cells and the immune system. This has enabled the definition of the immune contexture, representing the pre-existing immune parameters associated with patient survival. Even so, the role of distinct immune cell types in modulating cancer progression is increasingly emerging. An immune-based assay named the 'Immunoscore' was defined to quantify the in situ T cell infiltrate and was demonstrated to be superior to the AJCC/UICC TNM classification for patients with colorectal cancer. This Review provides a broad overview of the main immune parameters positively or negatively shaping cancer development, including the Immunoscore, and their prognostic and predictive value. The importance of the immune system in cancer control is demonstrated by the requirement for a pre-existing intratumour adaptive immune response for effective immunotherapies, such as checkpoint inhibitors. Finally, we discuss how the combination of multiple immune parameters, rather than individual ones, might increase prognostic and/or predictive power.
Collapse
Affiliation(s)
- Daniela Bruni
- INSERM, Laboratory of Integrative Cancer Immunology; Équipe Labellisée Ligue Contre le Cancer; Sorbonne Université; Sorbonne Paris Cité; Université de Paris; Centre de Recherche des Cordeliers, Paris, France
| | - Helen K Angell
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology; Équipe Labellisée Ligue Contre le Cancer; Sorbonne Université; Sorbonne Paris Cité; Université de Paris; Centre de Recherche des Cordeliers, Paris, France.
| |
Collapse
|
32
|
Ascierto PA, Marincola FM, Fox BA, Galon J. No time to die: the consensus immunoscore for predicting survival and response to chemotherapy of locally advanced colon cancer patients in a multicenter international study. Oncoimmunology 2020; 9:1826132. [PMID: 33194317 PMCID: PMC7644246 DOI: 10.1080/2162402x.2020.1826132] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The multicenter international Society for Immunotherapy of Cancer (SITC) study of the consensus Immunoscore demonstrated the prediction of survival and response to chemotherapy in 763 Stage III colon cancer (CC) patients. Similar Immunoscore groups were found in elderly patients, and densities of immune cells and intratumoral T-cell repertoire were not decreasing with age in the tumor microenvironment. In two independent cohorts, Immunoscore significantly predicted time to recurrence (TTR), disease-free survival (DFS), and overall survival (OS), including within high-risk (T4 or N2) and low-risk (T1-3, N1) patients. In stratified Cox multivariable analysis for TTR, DFS, and OS, Immunoscore’s association to outcomes was independent of the patient’s age, sidedness, gender, T-stage, N-stage, and microsatellite instability status. Furthermore, the relative contribution to the risk test showed that Immunoscore had the highest contribution to survival. Importantly Immunoscore predicted the likelihood of response to chemotherapy. Only patients with a high-Immunoscore significantly benefited from chemotherapy. The prognostic value of Immunoscore was confirmed in two independent phase 3 clinical trials (NCCTG-N0147, n = 559; Prodige-IDEA, n = 1062). Moreover, results from IDEA phase 3 randomized trial revealed the predictive value of Immunoscore for response to adjuvant FOLFOX chemotherapy duration. The latest edition of the WHO Digestive System Tumors classification introduced the immune response as measured by Immunoscore as essential and desirable diagnostic criteria for CC, and Immunoscore was introduced into the 2020 ESMO Clinical Practice Guidelines for CC to refine the prognosis and adjust chemotherapy decision-making process in stages II and III patients. These results highlight the clinical utility of Immunoscore.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Innovative Therapies Unit, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Napoli, Italy
| | | | - Bernard A Fox
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA.,Laboratory of Molecular and Tumor Immunology, Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Jérôme Galon
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
33
|
Marliot F, Pagès F, Galon J. Usefulness and robustness of Immunoscore for personalized management of cancer patients. Oncoimmunology 2020; 9:1832324. [PMID: 33194318 PMCID: PMC7644247 DOI: 10.1080/2162402x.2020.1832324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This review details the analytical performance characteristics of the consensus Immunoscore, measuring the immune response to cancer, improving the estimation of risk of recurrence, and predicting response to treatment for patients with colon cancer. The analytical validation of Immunoscore has been documented. Immunoscore is a robust, reproducible, quantitative, and standardized immune assay, with a high prognostic performance, independent of all of the prognostic markers currently used in clinical practice. Immunoscore evaluation within the tumor microenvironment is clinically relevant, and Immunoscore was recently introduced into ESMO Clinical Practice Guidelines for colon cancer and into the WHO classification of the Digestive System Tumors. This paves the way for the use of Immunoscore in clinical practice in colorectal tumors and likely soon in many other solid tumors.
Collapse
Affiliation(s)
- Florence Marliot
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche DES Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Franck Pagès
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche DES Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche DES Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
34
|
Baldin P, Van den Eynde M, Mlecnik B, Galon J. Immunity to live: an immunopathoscore using the consensus Immunoscore to best define the risk of recurrence and death in stage IV metastatic patients. Oncoimmunology 2020; 9:1826133. [PMID: 33110705 PMCID: PMC7561331 DOI: 10.1080/2162402x.2020.1826133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The consensus Immunoscore is a routine assay quantifying the adaptive immune response within the tumor microenvironment. Its evaluation in the primary tumor of patients with stages I/II/III colorectal cancer (CRC) has prognostic value that has been confirmed in multiple studies. For metastatic patients, the evaluation of the consensus Immunoscore within resected metastases also significantly predicts the recurrence and survival of Stage IV patients. Since recurrence rates post-surgery are still very high, it is important to best evaluate risk parameters using the main patho-molecular and immune parameters. After preoperative treatment and curative resection of 582 metastases from 221 patients, clinico-pathological parameters, RAS mutation, and Immunoscore within metastases were assessed. Immunoscore and clinico-pathological parameters (number of metastases, surgical margin, histopathological growth pattern, and steatohepatitis) were associated with relapse in multivariable analysis. A Pathological Score (PS) that combines relevant clinico-pathological factors for relapse and Immunoscore was significantly (P < .0001) associated with Time to recurrence. In multivariable analysis, only Immunoscore (P < .001) and RAS mutations (P= .03) were prognostic and significantly associated with overall survival. Thus, among all parameters clinically relevant in the metastatic settings, PS and Immunoscore allow the stratification of stage IV CRC patients and identify patients with higher risk of recurrence. Immunoscore remained the major prognostic factor for overall survival (OS). In its latest edition, the WHO classification of Digestive System Tumors introduced for the first time the immune response as an essential and desirable diagnostic criterion for CRC. These novel results highlight the clinical utility of Immunoscore in Stage IV patients.
Collapse
Affiliation(s)
- Pamela Baldin
- Department of Pathology, Cliniques Universitaires Saint-Luc/Université Catholique de Louvain (Uclouvain), Brussels, Belgium
| | - Marc Van den Eynde
- Department of Medical Oncology and Hepato-gastroenterology, Cliniques Universitaires Saint-Luc, Institut Roi Albert II, Université Catholique de Louvain (Uclouvain), Brussels, Belgium
| | - Bernhard Mlecnik
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Inovarion, Paris, France
| | - Jérôme Galon
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
35
|
Van den Eynde M, Mlecnik B, Bindea G, Galon J. Multiverse of immune microenvironment in metastatic colorectal cancer. Oncoimmunology 2020; 9:1824316. [PMID: 33457100 PMCID: PMC7781760 DOI: 10.1080/2162402x.2020.1824316] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The comprehensive analysis of patients with a complete resection of all metastases reveals the heterogeneity of the colorectal metastatic disease and its clinical impact. Complex tumor immune interrelations shape the metastatic landscape, not only in terms of number and size of lesions, or mutational pattern, but also in terms of immune cell infiltrate. Significantly higher densities of T-cells and lower density of B-cells were quantified in the tumor microenvironment of metastases compared with primary tumors. A high T cell infiltration and Immunoscore measured in the least-infiltrated metastasis were associated with a significantly lower number of metastases, larger metastasis, and prolonged survival while patients with increased metastatic burden had a lower Immunoscore. Immunoscore was evaluated on a biopsy, in a random metastasis or as the mean value of all metastases significantly predicting outcome. However, the most immune-infiltrated metastasis was not significantly predicting outcome, whereas the least immune-infiltrated metastasis was best in predicting clinical outcome. A good likelihood of concordance of Immunoscore was observed between one biopsy and complete metastasis, but the overall intra-metastatic immune infiltrate might be better estimated with multiple biopsies or sampling of larger tumor areas. This intra-metastatic adaptive immune reaction increases following aneoadjuvant treatment containing anti-EGFR monoclonal antibody, an effect that is currently therapeutically evaluated in clinical trials to improve the survival of metastatic patients.
Collapse
Affiliation(s)
- Marc Van den Eynde
- Department of Medical Oncology and Hepato-gastroenterology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc/Université Catholique De Louvain (Uclouvain), Brussels, Belgium
| | - Bernhard Mlecnik
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France.,Equipe Labellisée Ligue Contre Le Cancer, Paris, France.,Centre De Recherche Des Cordeliers, Sorbonne Université, Université De Paris, Paris, France.,Inovarion, Paris, France
| | - Gabriela Bindea
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France.,Equipe Labellisée Ligue Contre Le Cancer, Paris, France.,Centre De Recherche Des Cordeliers, Sorbonne Université, Université De Paris, Paris, France
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France.,Equipe Labellisée Ligue Contre Le Cancer, Paris, France.,Centre De Recherche Des Cordeliers, Sorbonne Université, Université De Paris, Paris, France
| |
Collapse
|
36
|
Pagès F, Taieb J, Laurent-Puig P, Galon J. The consensus Immunoscore in phase 3 clinical trials; potential impact on patient management decisions. Oncoimmunology 2020; 9:1812221. [PMID: 32939329 PMCID: PMC7480815 DOI: 10.1080/2162402x.2020.1812221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The consensus Immunoscore has a prognostic value that has been confirmed in two randomized phase 3 clinical trials, and it provides a reliable estimate of the recurrence risk in colon cancer. The latest edition of the WHO classification of the Digestive System Tumors introduced for the first time the immune response as an essential and desirable diagnostic criteria for digestive cancers. Therefore, the immune response and Immunoscore evaluation within the tumor microenvironment is clinically relevant. In addition, the evaluation of the Immunoscore in stage III colon cancer patients from the IDEA France clinical trial evaluating 3 versus 6 months of oxaliplatin-based adjuvant chemotherapy demonstrated the predictive value of Immunoscore for treatment duration. Immunoscore predicted response to 6 months FOLFOX chemotherapy both in low- and high-risk Stage III patients. Low-risk patients (T1-3, N1) with High-Immunoscore had the 3-year DFS of 91.4% when treated with the 6-month FOLFOX, and only 80.8% with the 3-month regimen. The international validation of the prognostic value of the consensus Immunoscore together with its predictive value to guide treatment provides important information for the personalized management of colon cancer patients.
Collapse
Affiliation(s)
- Franck Pagès
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Julien Taieb
- Department of Gastroenterology and Gastrointestinal Oncology, Georges-Pompidou European Hospital, AP-HP, Paris, France.,Sorbonne Paris Cité, University of Paris, Paris, France
| | - Pierre Laurent-Puig
- INSERM UMR-S1147 MEPPOT, CNRS SNC5014, Centre Universitaire des Saints-Pères, Equipe Labellisée Ligue Nationale Contre le Cancer, Université Sorbonne Paris Cité, Paris, France
| | - Jérôme Galon
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
37
|
Abstract
Treatment of metastatic colorectal cancer is based upon the assumption that metastases are homogeneous within a patient. A comprehensive analysis of all metastases of each patient revealed the heterogeneity of the colorectal metastatic disease and its clinical impact. Complex tumor-immune interrelations shape the metastatic landscape. Adaptive immune cells and Immunoscore quantified in a random metastatic biopsy predict clinical outcome and their evaluation in the tumor microenvironment of the least infiltrated metastasis most accurately predict long-term survival. The adaptive immune cell infiltration was more informative than tumor regression and pathological response to predict long-term survival. These results highlight the clinical utility of Immunoscore for patient management. The immune response within the tumor microenvironment is an essential diagnostic criterion for colorectal cancer that has recently been integrated into the international WHO classification of Digestive System Tumors.
Collapse
Affiliation(s)
- Bernhard Mlecnik
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,Inovarion, Paris, France
| | - Gabriela Bindea
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| | - Marc Van den Eynde
- Department of Medical Oncology and Hepato-gastroenterology, Cliniques Universitaires Saint-Luc/Université Catholique de Louvain (Uclouvain), Brussels, Belgium
| | - Jérôme Galon
- Laboratory of Integrative Cancer Immunology, INSERM, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
38
|
Lanzi A, Sinicrope FA, Benson AB, Galon J. The consensus Immunoscore in phase 3 clinical trial (N0147) and impact on patient management decisions. Oncoimmunology 2020; 9:1796003. [PMID: 32934890 PMCID: PMC7466859 DOI: 10.1080/2162402x.2020.1796003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The consensus Immunoscore is a routine assay quantifying the adaptive immune response within the tumor microenvironment. It has a prognostic value that has been confirmed in a phase 3 clinical trial (NCCTG N0147) in stage III colon cancers. Moreover, results from another phase 3 randomized trial revealed the predictive value of Immunoscore for response to adjuvant chemotherapy duration. These results highlight the clinical utility of Immunoscore. In its latest edition, the World Health Organization classification of Digestive System Tumors introduced for the first time the immune response as an essential and desirable diagnostic criterion for colorectal cancer. Within the tumor microenvironment, the immune response provides an important estimate of the risk of recurrence and death in colon cancer. The international validation of the prognostic value of the consensus Immunoscore together with its prognostic value in the N0147 trial and its predictive utility for response to chemotherapy in stage III patients provide valuable information for patient management.
Collapse
Affiliation(s)
- Anastasia Lanzi
- INSERM, Laboratory of Integrative Cancer Immunology, Cordeliers Research Center, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université de Paris, Paris, France
| | | | - A B Benson
- Northwestern University, Chicago, IL, USA
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Cordeliers Research Center, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université de Paris, Paris, France
| |
Collapse
|
39
|
Hou L, Tian C, Yan Y, Zhang L, Zhang H, Zhang Z. Manganese-Based Nanoactivator Optimizes Cancer Immunotherapy via Enhancing Innate Immunity. ACS NANO 2020; 14:3927-3940. [PMID: 32298077 DOI: 10.1021/acsnano.9b06111] [Citation(s) in RCA: 193] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) are essential components of the innate immune sensors to cytosolic DNA and elicit type I interferon (IFN). Recent studies have revealed that manganese (Mn) can enhance cGAS and STING activation to viral infection. However, the role of Mn in antitumor immunity has not been explored. Here, we designed a nanoactivator, which can induce the presence of DNA in cytoplasm and simultaneously elevate Mn2+ accumulation within tumor cells. In detail, amorphous porous manganese phosphate (APMP) NPs that are highly responsive to tumor microenvironment were employed to construct doxorubicin (DOX)-loaded and phospholipid (PL)-coated hybrid nanoparticles (PL/APMP-DOX NPs). PL/APMP-DOX NPs were stably maintained during systemic circulation, but triggered to release DOX for inducing DNA damage and Mn2+ to augment cGAS/STING activity. We found that PL/APMP-DOX NPs with superior tumor-targeting capacity boosted dendritic cell maturation and increased cytotoxic T lymphocyte infiltration as well as natural killer cell recruitment into the tumor site. Furthermore, the NPs increased production of type I IFN and secretion of pro-inflammatory cytokines (for example, TNF-α and IL-6). Consequently, PL/APMP-DOX NPs exhibited excellent antitumor efficacy and prolonged the lifespan of the tumor-bearing mice. Collectively, we developed a PL-decorated Mn-based hybrid nanoactivator to intensify immune activation and that might provide therapeutic potential for caner immunotherapy.
Collapse
Affiliation(s)
- Lin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, and Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| | - Chunyu Tian
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
- Modern Analysis and Computer Center of Zhengzhou University, Zhengzhou 450001, China
| | - Yingshan Yan
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
- Modern Analysis and Computer Center of Zhengzhou University, Zhengzhou 450001, China
| | - Lewen Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, and Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
| | - Huijuan Zhang
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
40
|
Alhmoud JF, Woolley JF, Al Moustafa AE, Malki MI. DNA Damage/Repair Management in Cancers. Cancers (Basel) 2020; 12:E1050. [PMID: 32340362 PMCID: PMC7226105 DOI: 10.3390/cancers12041050] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
DNA damage is well recognized as a critical factor in cancer development and progression. DNA lesions create an abnormal nucleotide or nucleotide fragment, causing a break in one or both chains of the DNA strand. When DNA damage occurs, the possibility of generated mutations increases. Genomic instability is one of the most important factors that lead to cancer development. DNA repair pathways perform the essential role of correcting the DNA lesions that occur from DNA damaging agents or carcinogens, thus maintaining genomic stability. Inefficient DNA repair is a critical driving force behind cancer establishment, progression and evolution. A thorough understanding of DNA repair mechanisms in cancer will allow for better therapeutic intervention. In this review we will discuss the relationship between DNA damage/repair mechanisms and cancer, and how we can target these pathways.
Collapse
Affiliation(s)
- Jehad F. Alhmoud
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - John F. Woolley
- Department of Molecular & Clinical Pharmacology, Liverpool University, Liverpool L69 3GE, UK;
| | | | - Mohammed Imad Malki
- College of Medicine, QU Health, Qatar University, Doha P. O. Box 2713, Qatar;
| |
Collapse
|
41
|
Akin Telli T, Bregni G, Camera S, Deleporte A, Hendlisz A, Sclafani F. PD-1 and PD-L1 inhibitors in oesophago-gastric cancers. Cancer Lett 2019; 469:142-150. [PMID: 31669518 DOI: 10.1016/j.canlet.2019.10.036] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 12/25/2022]
Abstract
Oesophago-gastric cancers (OGCs) are aggressive tumours. While better peri-operative strategies, increased number of cytotoxic agents and availability of targeted therapies have improved survival, there remains an unmet need for novel treatment approaches. Immune checkpoint inhibitors (ICIs) have marked a new era in cancer management with unprecedented results in several malignancies. Although OGC lagged behind other solid tumours, evidence has increasingly accumulated supporting the contention that modulation of the anti-cancer host immune response may be beneficial for at least some patients. Many trials have been completed in Eastern and Western countries, some of these leading to the approval of ICIs in the refractory setting, and favorable opinion from regulatory agencies is expected also in treatment-naïve, advanced OGC. Furthermore, studies are evaluating ICIs in the early stage setting and exploring the potential of combination treatments. In this article we discuss the biological bases underlying the successful development of ICIs in OGC and review the available data on PD-1 and PD-L1 monoclonal antibodies in this disease. Also, we present ongoing clinical trials of these ICIs that could shape the future treatment landscape of OGC.
Collapse
Affiliation(s)
- Tugba Akin Telli
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Giacomo Bregni
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Silvia Camera
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Amelie Deleporte
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alain Hendlisz
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Francesco Sclafani
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
42
|
Ocadlikova D, Lecciso M, Isidori A, Loscocco F, Visani G, Amadori S, Cavo M, Curti A. Chemotherapy-Induced Tumor Cell Death at the Crossroads Between Immunogenicity and Immunotolerance: Focus on Acute Myeloid Leukemia. Front Oncol 2019; 9:1004. [PMID: 31649875 PMCID: PMC6794495 DOI: 10.3389/fonc.2019.01004] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/18/2019] [Indexed: 01/25/2023] Open
Abstract
In solid tumors and hematological malignancies, including acute myeloid leukemia, some chemotherapeutic agents, such as anthracyclines, have proven to activate an immune response via dendritic cell-based cross-priming of anti-tumor T lymphocytes. This process, known as immunogenic cell death, is characterized by a variety of tumor cell modifications, i.e., cell surface translocation of calreticulin, extracellular release of adenosine triphosphate and pro-inflammatory factors, such as high mobility group box 1 proteins. However, in addition to with immunogenic cell death, chemotherapy is known to induce inflammatory modifications within the tumor microenvironment, which may also elicit immunosuppressive pathways. In particular, DCs may be driven to acquire tolerogenic features, such as the overexpression of indoleamine 2,3-dioxygensase 1, which may ultimately hamper anti-tumor T-cells via the induction of T regulatory cells. The aim of this review is to summarize the current knowledge about the mechanisms and effects by which chemotherapy results in both activation and suppression of anti-tumor immune response. Indeed, a better understanding of the whole process underlying chemotherapy-induced alterations of the immunological tumor microenvironment has important clinical implications to fully exploit the immunogenic potential of anti-leukemia agents and tune their application.
Collapse
Affiliation(s)
- Darina Ocadlikova
- Department of Hematology and Oncology, University Hospital S.Orsola-Malpighi, Institute of Hematology "L. and A. Seràgnoli", Bologna, Italy
| | - Mariangela Lecciso
- Department of Hematology and Oncology, University Hospital S.Orsola-Malpighi, Institute of Hematology "L. and A. Seràgnoli", Bologna, Italy
| | - Alessandro Isidori
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Federica Loscocco
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Sergio Amadori
- Department of Medicine, Institute of Hematology, University Hospital Tor Vergata, Rome, Italy
| | - Michele Cavo
- Department of Hematology and Oncology, University Hospital S.Orsola-Malpighi, Institute of Hematology "L. and A. Seràgnoli", Bologna, Italy
| | - Antonio Curti
- Department of Hematology and Oncology, University Hospital S.Orsola-Malpighi, Institute of Hematology "L. and A. Seràgnoli", Bologna, Italy
| |
Collapse
|
43
|
Immunological consequences of chemotherapy: Single drugs, combination therapies and nanoparticle-based treatments. J Control Release 2019; 305:130-154. [DOI: 10.1016/j.jconrel.2019.04.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/09/2019] [Accepted: 04/14/2019] [Indexed: 02/07/2023]
|
44
|
Murakami T, Hiroshima Y, Matsuyama R, Homma Y, Hoffman RM, Endo I. Role of the tumor microenvironment in pancreatic cancer. Ann Gastroenterol Surg 2019; 3:130-137. [PMID: 30923782 PMCID: PMC6422798 DOI: 10.1002/ags3.12225] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/08/2018] [Accepted: 11/04/2018] [Indexed: 12/22/2022] Open
Abstract
Pancreatic cancer remains a highly recalcitrant disease despite the development of systemic chemotherapies. New treatment options are thus urgently required. Dense stromal formation, so-called "desmoplastic stroma," plays controversial roles in terms of pancreatic cancer growth, invasion, and metastasis. Cells such as cancer-associated fibroblasts, endothelial cells, and immune cells comprise the tumor microenvironment of pancreatic cancer. Pancreatic cancer is considered an immune-quiescent disease, but activation of immunological response in pancreatic cancer may contribute to favorable outcomes. Herein, we review the role of the tumor microenvironment in pancreatic cancer, with a focus on immunological aspects.
Collapse
Affiliation(s)
- Takashi Murakami
- Department of Gastroenterological SurgeryYokohama City University Graduate School of MedicineYokohamaJapan
| | - Yukihiko Hiroshima
- Department of Gastroenterological SurgeryYokohama City University Graduate School of MedicineYokohamaJapan
| | - Ryusei Matsuyama
- Department of Gastroenterological SurgeryYokohama City University Graduate School of MedicineYokohamaJapan
| | - Yuki Homma
- Department of Gastroenterological SurgeryYokohama City University Graduate School of MedicineYokohamaJapan
| | - Robert M. Hoffman
- AntiCancer, Inc.San DiegoCalifornia
- Department of SurgeryUniversity of CaliforniaSan DiegoCalifornia
| | - Itaru Endo
- Department of Gastroenterological SurgeryYokohama City University Graduate School of MedicineYokohamaJapan
| |
Collapse
|
45
|
Wagner S, Mullins CS, Linnebacher M. Colorectal cancer vaccines: Tumor-associated antigens vs neoantigens. World J Gastroenterol 2018; 24:5418-5432. [PMID: 30622371 PMCID: PMC6319136 DOI: 10.3748/wjg.v24.i48.5418] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/11/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023] Open
Abstract
Therapeutic options for the treatment of colorectal cancer (CRC) are diverse but still not always satisfying. Recent success of immune checkpoint inhibition treatment for the subgroup of CRC patients suffering from hyper-mutated tumors suggests a permanent role of immune therapy in the clinical management of CRC. Substantial improvement in treatment outcome could be achieved by development of efficient patient-individual CRC vaccination strategies. This mini-review summarizes the current knowledge on the two general classes of targets: tumor-associated antigens (TAAs) and tumor-specific antigens. TAAs like carcinoembryonic antigen and melanoma associated antigen are present in and shared by a subgroup of patients and a variety of clinical studies examined the efficacy of different TAA-derived peptide vaccines. Combinations of several TAAs as the next step and the development of personalized TAA-based peptide vaccines are discussed. Improvements of peptide-based vaccines achievable by adjuvants and immune-stimulatory chemotherapeutics are highlighted. Finally, we sum up clinical studies using tumor-specific antigens - in CRC almost exclusively neoantigens - which revealed promising results; particularly no severe adverse events were reported so far. Critical progress for clinical outcomes can be expected by individualizing neoantigen-based peptide vaccines and combining them with immune-stimulatory chemotherapeutics and immune checkpoint inhibitors. In light of these data and latest developments, truly personalized neoantigen-based peptide vaccines can be expected to fulfill modern precision medicine's requirements and will manifest as treatment pillar for routine clinical management of CRC.
Collapse
Affiliation(s)
- Sandra Wagner
- Section of Molecular Oncology and Immunotherapy, General Surgery, University Medical Center, Rostock D-18057, Germany
| | - Christina S Mullins
- Section of Molecular Oncology and Immunotherapy, General Surgery, University Medical Center, Rostock D-18057, Germany
| | - Michael Linnebacher
- Section of Molecular Oncology and Immunotherapy, General Surgery, University Medical Center, Rostock D-18057, Germany
| |
Collapse
|
46
|
Goto W, Kashiwagi S, Asano Y, Takada K, Takahashi K, Hatano T, Takashima T, Tomita S, Motomura H, Hirakawa K, Ohira M. Predictive value of lymphocyte-to-monocyte ratio in the preoperative setting for progression of patients with breast cancer. BMC Cancer 2018; 18:1137. [PMID: 30453914 PMCID: PMC6245848 DOI: 10.1186/s12885-018-5051-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/06/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The lymphocyte-to-monocyte ratio (LMR) has been used as a parameter reflecting systemic inflammation in several tumors, and is reportedly associated with prognosis in cancer patients. In this study, we evaluated the predictive value of LMR for progression and chemosensitivity in breast cancer patients treated with preoperative chemotherapy. METHODS LMR was evaluated in 239 patients with breast cancer treated with neoadjuvant chemotherapy (NAC) with 5-fluorouracil, epirubicin, and cyclophosphamide, followed by weekly paclitaxel with or without trastuzumab, and subsequent curative surgery. The correlations between LMR and clinicopathological features, prognosis, and pathological complete response (pCR) rate of NAC were evaluated retrospectively. We also evaluated the predictive value of neutrophil-to-lymphocyte ratio (NLR), and compared the predictive values of LMR and NLR. RESULTS We set 6.00 as the cut-off level for LMR based on the receiver operating characteristic (ROC) curve. A total of 119 patients (49.8%) were classified in the high-LMR group and 120 (50.2%) were classified in the low-LMR group. The low-LMR group had significantly worse disease-free survival rate (DFS) in all patients (p = 0.005) and in triple-negative breast cancer patients (p = 0.006). However, there was no significant correlation between LMR and pCR. Multivariate analysis showed that low LMR was an independent risk factor for DFS (p = 0.008, hazard ratio = 2.245). However, there was no significant difference in DFS (p = 0.143, log-rank) between patients in the low- and high-NLR groups. CONCLUSIONS LMR may be a useful prognostic marker in patients with breast cancer.
Collapse
Affiliation(s)
- Wataru Goto
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shinichiro Kashiwagi
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.
| | - Yuka Asano
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Koji Takada
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Katsuyuki Takahashi
- Department of Pharmacology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Takaharu Hatano
- Department of Plastic and Reconstructive Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Tsutomu Takashima
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shuhei Tomita
- Department of Pharmacology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Hisashi Motomura
- Department of Plastic and Reconstructive Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Kosei Hirakawa
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Masaichi Ohira
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| |
Collapse
|
47
|
Gilabert-Oriol R, Ryan GM, Leung AWY, Firmino NS, Bennewith KL, Bally MB. Liposomal Formulations to Modulate the Tumour Microenvironment and Antitumour Immune Response. Int J Mol Sci 2018; 19:ijms19102922. [PMID: 30261606 PMCID: PMC6213379 DOI: 10.3390/ijms19102922] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/22/2022] Open
Abstract
Tumours are complex systems of genetically diverse malignant cells that proliferate in the presence of a heterogeneous microenvironment consisting of host derived microvasculature, stromal, and immune cells. The components of the tumour microenvironment (TME) communicate with each other and with cancer cells, to regulate cellular processes that can inhibit, as well as enhance, tumour growth. Therapeutic strategies have been developed to modulate the TME and cancer-associated immune response. However, modulating compounds are often insoluble (aqueous solubility of less than 1 mg/mL) and have suboptimal pharmacokinetics that prevent therapeutically relevant drug concentrations from reaching the appropriate sites within the tumour. Nanomedicines and, in particular, liposomal formulations of relevant drug candidates, define clinically meaningful drug delivery systems that have the potential to ensure that the right drug candidate is delivered to the right area within tumours at the right time. Following encapsulation in liposomes, drug candidates often display extended plasma half-lives, higher plasma concentrations and may accumulate directly in the tumour tissue. Liposomes can normalise the tumour blood vessel structure and enhance the immunogenicity of tumour cell death; relatively unrecognised impacts associated with using liposomal formulations. This review describes liposomal formulations that affect components of the TME. A focus is placed on formulations which are approved for use in the clinic. The concept of tumour immunogenicity, and how liposomes may enhance radiation and chemotherapy-induced immunogenic cell death (ICD), is discussed. Liposomes are currently an indispensable tool in the treatment of cancer, and their contribution to cancer therapy may gain even further importance by incorporating modulators of the TME and the cancer-associated immune response.
Collapse
Affiliation(s)
- Roger Gilabert-Oriol
- Department of Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
| | - Gemma M Ryan
- Department of Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
| | - Ada W Y Leung
- Department of Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
- Cuprous Pharmaceuticals Inc., Vancouver, BC V6N 3P8, Canada.
- Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z1, Canada.
| | - Natalie S Firmino
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada.
| | - Kevin L Bennewith
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada.
| | - Marcel B Bally
- Department of Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada.
- Cuprous Pharmaceuticals Inc., Vancouver, BC V6N 3P8, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
- Centre for Drug Research and Development, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
48
|
Goto W, Kashiwagi S, Asano Y, Takada K, Takahashi K, Hatano T, Takashima T, Tomita S, Motomura H, Ohsawa M, Hirakawa K, Ohira M. Predictive value of improvement in the immune tumour microenvironment in patients with breast cancer treated with neoadjuvant chemotherapy. ESMO Open 2018; 3:e000305. [PMID: 30233820 PMCID: PMC6135412 DOI: 10.1136/esmoopen-2017-000305] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 01/07/2023] Open
Abstract
Background Tumour-infiltrating lymphocytes (TILs) can be used to monitor the immune tumour microenvironment (iTME) and predict treatment response and outcome in breast cancer. We evaluated the prognostic significance of the levels of CD8+ TILs and forkhead box protein (FOXP3)-positive TILs before and after neoadjuvant chemotherapy (NAC). Patients and methods We examined 136 patients with breast cancer treated with NAC. The number of CD8+ TILs and FOXP3+ TILs in biopsy specimens and residual tumours was evaluated by immunohistochemistry. Results Patients with a high rate of change in the CD8/FOXP3 ratio (CFR) had significantly better recurrence-free survival (RFS) (p<0.001, log-rank). In multivariate analysis, the rates of change in the CD8+ TIL levels and the CFR were independent predictors for RFS (HR=2.304, p=0.036 and HR=4.663, p<0.001). In patients with triple-negative and hormone receptor-positive breast cancer, the rate of change in the CFR was an independent predictor for RFS (HR=13.021, p=0.002 and HR=4.377, p=0.003). Conclusion Improvement in the iTME following NAC is correlated with good outcome. The rate of change in the CFR may be a useful biomarker to predict prognosis of patients treated with NAC.
Collapse
Affiliation(s)
- Wataru Goto
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shinichiro Kashiwagi
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yuka Asano
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Koji Takada
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Katsuyuki Takahashi
- Department of Pharmacology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Takaharu Hatano
- Department of Plastic and Reconstructive Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tsutomu Takashima
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shuhei Tomita
- Department of Pharmacology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hisashi Motomura
- Department of Plastic and Reconstructive Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masahiko Ohsawa
- Department of Diagnostic Pathology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaichi Ohira
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
49
|
Asano Y, Kashiwagi S, Goto W, Takada K, Takahashi K, Morisaki T, Fujita H, Takashima T, Tomita S, Ohsawa M, Hirakawa K, Ohira M. Prediction of treatment responses to neoadjuvant chemotherapy in triple-negative breast cancer by analysis of immune checkpoint protein expression. J Transl Med 2018; 16:87. [PMID: 29615063 PMCID: PMC5883348 DOI: 10.1186/s12967-018-1458-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/23/2018] [Indexed: 12/22/2022] Open
Abstract
Background “Avoiding immune destruction” has recently been established as one of the hallmarks of cancer. The programmed cell death (PD)-1/programmed cell death-ligand (PD-L) 1 pathway is an important immunosuppression mechanism that allows cancer cells to escape host immunity. The present study investigated how the expressions of these immune checkpoint proteins affected responses to neo-adjuvant chemotherapy (NAC) in breast cancer. Methods A total of 177 patients with resectable early-stage breast cancer were treated with NAC. Estrogen receptor, progesteron receptor, human epidermal growth factor receptor 2, Ki67, PD-L1, PDL-2 and PD-1 status were assessed by immunohistochemistry. Results There were 37 (20.9%) patients with high PD-1 expression, 42 (23.7%) patients had high PD-L1 expression, and 52 (29.4%) patients had high PD-L2 expression. The patients with high PD-1 and PD-L1 expressions had a significantly higher rate of triple-negative breast cancer (TNBC) (p = 0.041) (p < 0.001). In TNBC, patients with high PD-1 and PD-L1 expressions had significantly higher rates of non-pCR (p = 0.003) (p < 0.001). Univariate analysis showed that PD-1 and PD-L1 expressions also significantly shortened disease free survival in TNBC (p = 0.048, HR = 3.318) (p = 0.007, HR = 8.375). However, multivariate analysis found that only PD-L1 expression was an independent prognostic factor (p = 0.041, HR = 9.479). Conclusions PD-1 and PD-L1 expressions may be useful as biomarkers to predict treatment responses to NAC in breast cancer. Above all, PD-L1 expression may also be useful as biomarkers for more effective chemotherapy in TNBC. Electronic supplementary material The online version of this article (10.1186/s12967-018-1458-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuka Asano
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shinichiro Kashiwagi
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.
| | - Wataru Goto
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Koji Takada
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Katsuyuki Takahashi
- Department of Pharmacology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Tamami Morisaki
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Hisakazu Fujita
- Department of Scientific and Linguistic Fundamentals of Nursing, Osaka City University Graduate School of Nursing, 1-5-17 Asahi-machi, Abeno-ku, Osaka, 545-0051, Japan
| | - Tsutomu Takashima
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shuhei Tomita
- Department of Pharmacology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Masahiko Ohsawa
- Department of Diagnostic Pathology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Masaichi Ohira
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| |
Collapse
|
50
|
Tanaka M, Kataoka H, Yano S, Sawada T, Akashi H, Inoue M, Suzuki S, Inagaki Y, Hayashi N, Nishie H, Shimura T, Mizoshita T, Mori Y, Kubota E, Tanida S, Takahashi S, Joh T. Immunogenic cell death due to a new photodynamic therapy (PDT) with glycoconjugated chlorin (G-chlorin). Oncotarget 2018; 7:47242-47251. [PMID: 27363018 PMCID: PMC5216938 DOI: 10.18632/oncotarget.9725] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 05/13/2016] [Indexed: 12/22/2022] Open
Abstract
Both the pre-apoptotic exposure to calreticulin (CRT) and the post-apoptotic release of high-mobility group box 1 protein (HMGB1) are required for immunogenic cell death. Photodynamic therapy (PDT) uses non-toxic photosensitizers and visible light at a specific wavelength in combination with oxygen to produce cytotoxic reactive oxygen species that kill malignant cells by apoptosis and/or necrosis, shut down the tumor microvasculature, and stimulate the host immune system. We have previously shown that glycoconjugated chlorin (G-chlorin) has superior cancer cell selectivity and effectively suppresses the growth of xenograft tumors. In the present study, we evaluated the immunogenicity of PDT with G-chlorin treatment in colon cancer cells. PDT with G-chlorin suppressed CT26 (mouse colon cancer cells) tumor growth considerably more efficiently in immunocompetent mice (wild-type mice, allograft model) than in immune-deficient mice (nude mice, xenograft model), although control treatments were not different between the two. This treatment also induced CRT translocation and HMGB1 release in cells, as shown by western blot and immunofluorescence staining. To evaluate the use of PDT-treated cells as a tumor vaccine, we employed a syngeneic mouse tumor model (allograft model). Mice inoculated with PDT-treated CT26 cells were significantly protected against a subsequent challenge with live CT26 cells, and this protection was inhibited by siRNA for CRT or HMGB1. In conclusion, PDT with G-chlorin treatment induced immunogenic cell death in a mouse model, where the immunogenicity of this treatment was directed by CRT expression and HMGB1 release.
Collapse
Affiliation(s)
- Mamoru Tanaka
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hiromi Kataoka
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Shigenobu Yano
- Graduate School of Materials Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Takuya Sawada
- Research Institute of Natural Sciences, Okayama University of Science, Okayama 700-0005, Japan
| | - Haruo Akashi
- Research Institute of Natural Sciences, Okayama University of Science, Okayama 700-0005, Japan
| | - Masahiro Inoue
- Department of Biochemistry, Osaka Medical Centre for Cancer and Cardiovascular Diseases, Higashinari-ku, Osaka 537-8511, Japan
| | - Shugo Suzuki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Yusuke Inagaki
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Noriyuki Hayashi
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hirotada Nishie
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Takaya Shimura
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Tsutomu Mizoshita
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Yoshinori Mori
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Eiji Kubota
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Satoshi Tanida
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Takashi Joh
- Departments of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| |
Collapse
|