1
|
Li D, Shi Y, Yu S, Zhang B, Huang Z, Ling F, Mao X, Deng Y, Cai M, Luo W. NK cellular derived nanovesicles in tumor immunity. Mol Immunol 2025; 182:54-61. [PMID: 40174421 DOI: 10.1016/j.molimm.2025.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/20/2025] [Accepted: 03/30/2025] [Indexed: 04/04/2025]
Abstract
Natural Killer (NK) cells are a vital element of the innate immune system, and NK cell-based therapies have demonstrated efficacy against various malignancies. However, targeting solid tumors has been challenging due to the low infiltration of NK cells into tumors and the effective evasion strategies employed by tumors. Recent studies have shown that NK cell derived nanovesicles (NK-NV) can not only replicate the functions of NK cells but also offer more advantages in clinical applications. They are capable of transporting various cellular components such as proteins, nucleic acids, and lipids across distances, thereby facilitating intercellular communication among various cells within the tumor microenvironment (TME). With the progress in nanomedical technology, these vesicles can be engineered to carry a range of functional elements and therapeutic agents to enhance their antitumoral capabilities. In this review, we summarize the current available literature on NK-NVs, discuss their potential biological functions and the role of non-coding RNAs (ncRNAs), and explore their application in the treatment of solid tumors.
Collapse
Affiliation(s)
- Dingru Li
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Yixin Shi
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China; Liaoning Laboratory of Cancer Genomics and Epigenomics, Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Sifei Yu
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China
| | - Beiying Zhang
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China
| | - Ziyi Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Fei Ling
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiaofan Mao
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China
| | - Yuhua Deng
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China
| | - Mengyun Cai
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China.
| | - Wei Luo
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China.
| |
Collapse
|
2
|
Zhang W, Li K, Jian A, Zhang G, Zhang X. Prospects for potential therapy targeting immune‑associated factors in endometriosis (Review). Mol Med Rep 2025; 31:57. [PMID: 39717957 PMCID: PMC11715623 DOI: 10.3892/mmr.2024.13422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/03/2024] [Indexed: 12/25/2024] Open
Abstract
Endometriosis (EM) is a chronic inflammatory disease that is one of the most common causes of gynecological systemic lesions in women before menopause. The most representative histological feature of EM is that the endometrium appears outside of the uterine cavity, often in the ovary. Although it is generally accepted that the epithelial and stromal cells of the ectopic endometrium are not malignant, they still have numerous similarities to malignant tumors, including considerable changes to the immune microenvironment (immune monitoring disorder), the creation of a specific hormone environment, high levels of oxidative stress, chronic inflammation and abnormal immune cell regulation. The pathogenesis of EM is not fully understood, which makes it difficult to identify specific biomarkers and potential therapeutic targets for early disease diagnosis and effective treatment. However, considerable progress has been made in this field over the past few decades. The purpose of the present review is to summarize the confirmed and potential biomarkers for EM, and to identify potential therapeutic targets based on changes in immunological factors (including natural killer cells, macrophages, the complement system, miRNA and P‑selectin) in the ectopic endometrial tissue. It is hoped that this work can be used as the basis for identifying accurate diagnostic markers for EM and developing personalized immune‑targeted therapy.
Collapse
Affiliation(s)
- Wenwen Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Kang Li
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Aiwen Jian
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Guanran Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiaoli Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
3
|
Patel SP, Alonso-Gordoa T, Banerjee S, Wang D, Naidoo J, Standifer NE, Palmer DC, Cheng LY, Kourtesis P, Ascierto ML, Das M, Diamond JR, Hellmann MD, Carneiro BA. Phase 1/2 study of monalizumab plus durvalumab in patients with advanced solid tumors. J Immunother Cancer 2024; 12:e007340. [PMID: 38309722 PMCID: PMC10840023 DOI: 10.1136/jitc-2023-007340] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND The combination of monalizumab (anti-NKG2A/CD94) and durvalumab (anti-programmed death ligand-1) may promote antitumor immunity by targeting innate and adaptive immunity. This phase 1/2 study of monalizumab and durvalumab evaluated safety, antitumor activity, and pharmacodynamics in patients with advanced solid tumors. MAIN BODY Immunotherapy-naïve patients aged ≥18 years with advanced disease, Eastern Cooperative Oncology Group performance status of 0-1, and 1-3 prior lines of systemic therapy in the recurrent/metastatic setting were enrolled. In part 1 (dose escalation), patients received durvalumab 1500 mg every 4 weeks (Q4W) with increasing doses of monalizumab Q2W/Q4W (n=15). Dose expansion in part 1 included patients with cervical cancer (n=15; durvalumab 1500 mg Q4W and monalizumab 750 mg Q2W) or metastatic microsatellite stable (MSS)-colorectal cancer (CRC) (n=15; durvalumab 1500 mg Q4W and monalizumab 750 mg Q4W). In part 2 (dose expansion), patients with MSS-CRC (n=40), non-small cell lung cancer (NSCLC; n=20), MSS-endometrial cancer (n=40), or ovarian cancer (n=40) received durvalumab 1500 mg Q4W and monalizumab 750 mg Q2W. The primary endpoint was safety. Secondary endpoints included antitumor activity per Response Evaluation Criteria In Solid Tumors version 1.1 (RECIST v1.1). Exploratory analyses included assessment of T-cell and natural killer (NK) cell activation and proliferation in peripheral blood and the tumor microenvironment (TME). The study enrolled 185 patients (part 1, 45; part 2, 140). No dose-limiting toxicities were observed and the maximum tolerated dose was not reached. In part 2, the most common treatment-related adverse events were fatigue (12.1%), asthenia (9.3%), diarrhea (9.3%), pruritus (7.9%), and pyrexia (7.1%). In the expansion cohorts, response rates were 0% (cervical), 7.7% (MSS-CRC), 10% (NSCLC), 5.4% (ovarian), and 0% (MSS-endometrial). Sustained NK cell activation, CD8+ T-cell proliferation, increased serum levels of CXCL10 (C-X-C motif chemokine ligand 10) and CXCL11, and increased tumor infiltration of CD8+ and granzyme B+ cells were observed. CONCLUSIONS Although efficacy was modest, monalizumab plus durvalumab was well tolerated and encouraging immune activation was observed in the peripheral blood and TME. TRIAL REGISTRATION NUMBER NCT02671435.
Collapse
Affiliation(s)
- Sandip P Patel
- University of California San Diego, Moores Cancer Center, San Diego, California, USA
| | | | - Susana Banerjee
- Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Ding Wang
- Henry Ford Health System, Detroit, Michigan, USA
| | - Jarushka Naidoo
- Johns Hopkins Medicine, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
- Johns Hopkins Medicine The Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, Maryland, USA
| | - Nathan E Standifer
- BioPharmaceuticals Research and Development, AstraZeneca, South San Francisco, California, USA
| | - Doug C Palmer
- Oncology Research and Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Lin-Yang Cheng
- Oncology Research and Development, AstraZeneca, Gaithersburg, Maryland, USA
| | | | - Maria L Ascierto
- Oncology Research and Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Mayukh Das
- Oncology Research and Development, AstraZeneca, Gaithersburg, Maryland, USA
| | | | | | - Benedito A Carneiro
- Legorreta Cancer Center at Brown University, Lifespan Cancer Institute, Providence, Rhode Island, USA
| |
Collapse
|
4
|
Wang Z, Zhang R, Cao Y, Chen Y, Huang S, Luo Y. Investigation of the efficacy and feasibility of combined therapy of PD-L1-enhanced exogenous peripatetic adoptive natural killer (NK) cells in combination with antiangiogenic targeted therapy in the treatment of extensive-stage small cell lung cancer. Thorac Cancer 2023; 14:2877-2885. [PMID: 37596831 PMCID: PMC10542463 DOI: 10.1111/1759-7714.15040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 08/20/2023] Open
Abstract
A 67-year-old male patient presented with extensive-stage small cell lung cancer with the primary lesion located in the right upper lung, accompanied by multiple metastases to the pleura and abdominal cavity with enlarged mediastinal lymph nodes. A combination therapy approach was used to target the patient's multiple systemic metastases after localized radiotherapy. The approach involved adoptive transfer of programmed death ligand 1 (PD-L1) enhanced exogenous natural killer (NK) cells, along with antiangiogenic treatment. Allogeneic cord blood NK cells were infused back into the patient over two consecutive days. On the first day, the treatment was followed by a dose of 1200 mg of atezolizumab. Subsequently, the patient received a daily dose of 10 mg of anlotinib administered orally for 14 days. This was followed by a 7-day break, and each cycle lasted 21 days. After delivering localized radiation to the primary lesion in the right lung and metastatic mediastinal lymph nodes, complete remission was achieved in the local lesion, effectively avoiding the risk of superior vena cava syndrome. Following six cycles of combined therapy, most of the metastatic lesions had disappeared, and the remaining metastatic lesions had significantly reduced in size. The recent therapeutic effect resulted in partial remission. The combination therapy of immune checkpoint inhibitor PD-L1-enhanced exogenous adoptive transfer NK cells, along with antiangiogenic targeted treatment, demonstrated a satisfactory short-term effect, with disappearance of most of the metastases and noticeable shrinkage in the remaining metastatic lesions.
Collapse
Affiliation(s)
- Zhizhen Wang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Ruiping Zhang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Yuchan Cao
- Brown University School of Public HealthBrown UniversityProvidenceRhode IslandUSA
| | - Yang Chen
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Sheng Huang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for CancerTianjinChina
| | | |
Collapse
|
5
|
Budi HS, Farhood B. Targeting oral tumor microenvironment for effective therapy. Cancer Cell Int 2023; 23:101. [PMID: 37221555 DOI: 10.1186/s12935-023-02943-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023] Open
Abstract
Oral cancers are among the common head and neck malignancies. Different anticancer therapy modalities such as chemotherapy, immunotherapy, radiation therapy, and also targeted molecular therapy may be prescribed for targeting oral malignancies. Traditionally, it has been assumed that targeting malignant cells alone by anticancer modalities such as chemotherapy and radiotherapy suppresses tumor growth. In the last decade, a large number of experiments have confirmed the pivotal role of other cells and secreted molecules in the tumor microenvironment (TME) on tumor progression. Extracellular matrix and immunosuppressive cells such as tumor-associated macrophages, myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), and regulatory T cells (Tregs) play key roles in the progression of tumors like oral cancers and resistance to therapy. On the other hand, infiltrated CD4 + and CD8 + T lymphocytes, and natural killer (NK) cells are key anti-tumor cells that suppress the proliferation of malignant cells. Modulation of extracellular matrix and immunosuppressive cells, and also stimulation of anticancer immunity have been suggested to treat oral malignancies more effectively. Furthermore, the administration of some adjuvants or combination therapy modalities may suppress oral malignancies more effectively. In this review, we discuss various interactions between oral cancer cells and TME. Furthermore, we also review the basic mechanisms within oral TME that may cause resistance to therapy. Potential targets and approaches for overcoming the resistance of oral cancers to various anticancer modalities will also be reviewed. The findings for targeting cells and potential therapeutic targets in clinical studies will also be reviewed.
Collapse
Affiliation(s)
- Hendrik Setia Budi
- Department of Oral Biology, Dental Pharmacology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
6
|
Li Y, Xie S, Chen M, Li H, Wang Y, Fan Y, An K, Wu Y, Xiao W. Development of an antibody-ligand fusion protein scFvCD16A -sc4-1BBL in Komagataella phaffii with stimulatory activity for Natural Killer cells. Microb Cell Fact 2023; 22:67. [PMID: 37041591 PMCID: PMC10091686 DOI: 10.1186/s12934-023-02082-6] [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: 12/12/2022] [Accepted: 04/04/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Natural killer (NK) cell-based immunotherapies have demonstrated substantial potential for the treatment of hematologic malignancies. However, its application is limited due to the difficulty in the production of a large number of NK cells in vitro and the insufficient therapeutic efficacy against solid tumors in vivo. Engineered antibodies or fusion proteins targeting activating receptors and costimulatory molecules of NK cells have been developed to encounter these problems. They are mostly produced in mammalian cells with high cost and long processing times. Yeast systems, such as Komagataella phaffii, present a convenient manipulation of microbial systems with the key advantages of improved folding machinery and low cost. RESULTS In this study, we designed an antibody fusion protein scFvCD16A-sc4-1BBL, composed of the single chain variant fragment (scFv) of anti-CD16A antibody and the three extracellular domains (ECDs) of human 4-1BBL in a single-chain format (sc) with the GS linker, aiming to boost NK cell proliferation and activation. This protein complex was produced in the K. phaffii X33 system and purified by affinity chromatography and size exclusion chromatography. The scFvCD16A-sc4-1BBL complex showed comparable binding abilities to its two targets human CD16A and 4-1BB as its two parental moieties (scFvCD16A and monomer ECD (mn)4-1BBL). scFvCD16A-sc4-1BBL specifically stimulated the expansion of peripheral blood mononuclear cell (PBMC)-derived NK cells in vitro. Furthermore, in the ovarian cancer xenograft mouse model, adoptive NK cell infusion combined with intraperitoneal (i.p) injection of scFvCD16A-sc4-1BBL further reduced the tumor burden and prolonged the survival time of mice. CONCLUSION Our studies demonstrate the feasibility of the expression of the antibody fusion protein scFvCD16A-sc4-1BBL in K. phaffii with favourable properties. scFvCD16A-sc4-1BBL stimulates PBMC-derived NK cell expansion in vitro and improves the antitumor activity of adoptively transferred NK cells in a murine model of ovarian cancer and may serve as a synergistic drug for NK immunotherapy in future research and applications.
Collapse
Affiliation(s)
- Yangyang Li
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Siqi Xie
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Minhua Chen
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Hao Li
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Yehai Wang
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Yan Fan
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Kang An
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Yu Wu
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Weihua Xiao
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| |
Collapse
|
7
|
Yu H, Wu M, Chen S, Song M, Yue Y. Biomimetic nanoparticles for tumor immunotherapy. Front Bioeng Biotechnol 2022; 10:989881. [PMID: 36440446 PMCID: PMC9682960 DOI: 10.3389/fbioe.2022.989881] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/26/2022] [Indexed: 12/11/2023] Open
Abstract
Currently, tumor treatment research still focuses on the cancer cells themselves, but the fact that the immune system plays an important role in inhibiting tumor development cannot be ignored. The activation of the immune system depends on the difference between self and non-self. Unfortunately, cancer is characterized by genetic changes in the host cells that lead to uncontrolled cell proliferation and evade immune surveillance. Cancer immunotherapy aims to coordinate a patient's immune system to target, fight, and destroy cancer cells without destroying the normal cells. Nevertheless, antitumor immunity driven by the autoimmune system alone may be inadequate for treatment. The development of drug delivery systems (DDS) based on nanoparticles can not only promote immunotherapy but also improve the immunosuppressive tumor microenvironment (ITM), which provides promising strategies for cancer treatment. However, conventional nano drug delivery systems (NDDS) are subject to several limitations in clinical transformation, such as immunogenicity and the potential toxicity risks of the carrier materials, premature drug leakage at off-target sites during circulation and drug load content. In order to address these limitations, this paper reviews the trends and progress of biomimetic NDDS and discusses the applications of each biomimetic system in tumor immunotherapy. Furthermore, we review the various combination immunotherapies based on biomimetic NDDS and key considerations for clinical transformation.
Collapse
Affiliation(s)
- Hanqing Yu
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Meng Wu
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Siyu Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Mingming Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yulin Yue
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Feng H, Zhuo Y, Zhang X, Li Y, Li Y, Duan X, Shi J, Xu C, Gao Y, Yu Z. Tumor Microenvironment in Hepatocellular Carcinoma: Key Players for Immunotherapy. J Hepatocell Carcinoma 2022; 9:1109-1125. [PMID: 36320666 PMCID: PMC9618253 DOI: 10.2147/jhc.s381764] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Hepatocellular carcinoma (HCC) remains a serious medical therapeutic challenge as conventional curative avenues such as surgery and chemotherapy only benefit for few patients with limited tumor burden. Immunotherapy achieves clinical progress in the treatment of this prevalent malignant disease by virtue of the development of tumor immunology; however, most patients have experienced minimal or no clinical benefit in terms of overall survival. The complexity and diversity of tumor microenvironment (TME) built by immune and stromal cell subsets has been considered to be responsible for the insufficiency of immunotherapy. The advance of bioanalytical technology boosts the exploration of the composition and differentiation of these infiltrated cells, which reflect the immune state of the TME and impact the efficacy of the antitumor immune response. Targeting these cells to remodel the TME is one of the important immunotherapeutic approaches to improve HCC treatment. In this review, we focused on the role of these non-cancerous cells in the tumor progression, and elaborated their function on cancer immunotherapy when manipulating them as potential targets.
Collapse
Affiliation(s)
- Hai Feng
- Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yunhui Zhuo
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xuemei Zhang
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yuyao Li
- Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yue Li
- Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xiangjuan Duan
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jia Shi
- Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Chengbin Xu
- Department of Informatics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yueqiu Gao
- Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China,Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China,Correspondence: Yueqiu Gao, Institute of Infectious Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China, Tel +86 21 20256507, Fax +86 21 20256699, Email
| | - Zhuo Yu
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China,Zhuo Yu, Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China, Tel +86 21 20256507, Fax +86 21 20256699, Email
| |
Collapse
|
9
|
Aarsund M, Segers FM, Wu Y, Inngjerdingen M. Comparison of characteristics and tumor targeting properties of extracellular vesicles derived from primary NK cells or NK-cell lines stimulated with IL-15 or IL-12/15/18. Cancer Immunol Immunother 2022; 71:2227-2238. [PMID: 35119498 PMCID: PMC9374793 DOI: 10.1007/s00262-022-03161-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/19/2022] [Indexed: 12/17/2022]
Abstract
NK cell-based therapies have shown promise for hematological cancer forms, but their use against solid tumors is hampered by their poor ability to infiltrate the tumor. NK cells release extracellular vesicles (EVs) containing cytolytic proteins, indicating that NK-cell derived EVs may have therapeutic potential. In this study, we compared the tumor-targeting potential of EVs derived from either primary NK cells or the NK cell lines NK-92 and KHYG-1 cultured in IL-15 alone or in combination with IL-12 and IL-18. Primary NK cells were also stimulated through the activating receptor CD16. Tumor cell apoptosis was measured using a panel of human colon, melanoma, glioblastoma, prostate, breast, and ovarian tumor cell line spheroids. NK cells or NK-92 cells stimulated with IL-12, IL-15, and IL-18 generated EVs with higher efficiency than EVs from resting cells, although similar amounts of EVs were produced under both conditions. Proteomic analysis indicated similar distribution of cytolytic proteins in EVs from primary NK cells and NK-92, but lower levels in KHYG-1 EVs that translated into poor capacity for KHYG-1 EVs at targeting tumor cell lines. Further, we show that CD16-stimulated NK cells release low amounts of EVs devoid of cytolytic proteins. Importantly, EVs from cytokine-stimulated NK cells penetrate into the spheroid core, and tumor spheroid susceptibility to NK-cell derived EVs was linked to differential expression of the NKG2D ligands MICA/B, which was blocked with an anti-NKG2D antibody. We conclude that EVs from activated primary NK cells or NK-92 cells has the best potential to infiltrate and target solid tumors.
Collapse
Affiliation(s)
- Miriam Aarsund
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Filip M Segers
- Department of Pharmacology, Clinic of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
| | - Yunjie Wu
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Marit Inngjerdingen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
10
|
Della Chiesa M, Setti C, Giordano C, Obino V, Greppi M, Pesce S, Marcenaro E, Rutigliani M, Provinciali N, Paleari L, DeCensi A, Sivori S, Carlomagno S. NK Cell-Based Immunotherapy in Colorectal Cancer. Vaccines (Basel) 2022; 10:1033. [PMID: 35891197 PMCID: PMC9323201 DOI: 10.3390/vaccines10071033] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 02/01/2023] Open
Abstract
Human Natural Killer (NK) cells are all round players in immunity thanks to their powerful and immediate response against transformed cells and the ability to modulate the subsequent adaptive immune response. The potential of immunotherapies based on NK cell involvement has been initially revealed in the hematological setting but has inspired the design of different immune tools to also be applied against solid tumors, including colorectal cancer (CRC). Indeed, despite cancer prevention screening plans, surgery, and chemotherapy strategies, CRC is one of the most widespread cancers and with the highest mortality rate. Therefore, further efficient and complementary immune-based therapies are in urgent need. In this review, we gathered the most recent advances in NK cell-based immunotherapies aimed at fighting CRC, in particular, the use of monoclonal antibodies targeting tumor-associated antigens (TAAs), immune checkpoint blockade, and adoptive NK cell therapy, including NK cells modified with chimeric antigen receptor (CAR-NK).
Collapse
Affiliation(s)
- Mariella Della Chiesa
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Chiara Setti
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Chiara Giordano
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Valentina Obino
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Marco Greppi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Silvia Pesce
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Emanuela Marcenaro
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | | | | | - Laura Paleari
- A.Li.Sa., Liguria Region Health Authority, 16121 Genoa, Italy;
| | - Andrea DeCensi
- Medical Oncology, Galliera Hospital, 16128 Genoa, Italy; (N.P.); (A.D.)
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Simona Carlomagno
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| |
Collapse
|
11
|
Poggi A, Zocchi MR. Natural killer cells and immune-checkpoint inhibitor therapy: Current knowledge and new challenges. Mol Ther Oncolytics 2022; 24:26-42. [PMID: 34977340 PMCID: PMC8693432 DOI: 10.1016/j.omto.2021.11.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The discovery of immune checkpoints (ICs) and the development of specific blockers to relieve immune effector cells from this inhibiting mechanism has changed the view of anti-cancer therapy. In addition to cytotoxic T lymphocyte antigen 4 (CTLA4) and programmed death 1 (PD1), classical ICs of T lymphocytes and recently described also on a fraction of natural killer (NK) cells, several NK cell receptors, including killer immunoglobulin-like inhibitory receptors (KIRs) and NGK2A, have been recognized as checkpoint members typical of the NK cell population. This offers the opportunity of a dual-checkpoint inhibition approach, targeting classical and non-classical ICs and leading to a synergistic therapeutic effect. In this review, we will overview and discuss this new perspective, focusing on the most relevant candidates for this role among the variety of potential NK ICs. Beside listing and defining classical ICs expressed also by NK cells, or non-classical ICs either on T or on NK cells, we will address their role in NK cell survival, chronic stimulation or functional exhaustion, and the potential relevance of this phenomenon on anti-tumor immune response. Furthermore, NK ICs will be proposed as possible new targets for the development of efficient combined immunotherapy, not forgetting the relevant concerns that may be raised on NK IC blockade. Finally, the impact of epigenetic drugs in such a complex therapeutic picture will be briefly addressed.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Building 90 Tower C, 4th Floor, 16132 Genoa, Italy
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
12
|
Tarannum M, Romee R. Cytokine-induced memory-like natural killer cells for cancer immunotherapy. Stem Cell Res Ther 2021; 12:592. [PMID: 34863287 PMCID: PMC8642969 DOI: 10.1186/s13287-021-02655-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022] Open
Abstract
Natural killer cells are an important part of the innate immune system mediating robust responses to virus-infected and malignant cells without needing prior antigen priming. NK cells have always been thought to be short-lived and with no antigen specificity; however, recent data support the presence of NK cell memory including in the hapten-specific contact hypersensitivity model and in certain viral infections. The memory-like features can also be generated by short-term activation of both murine and human NK cells with cytokine combination of IL-12, IL-15 and IL-18, imparting increased longevity and enhanced anticancer functionality. Preclinical studies and very early clinical trials demonstrate safety and very promising clinical activity of these cytokine-induced memory-like (CIML) NK cells, making them an attractive cell type for developing novel adoptive cellular immunotherapy strategies. Furthermore, efforts are on to arm them with novel gene constructs for enhanced tumor targeting and function.
Collapse
Affiliation(s)
- Mubin Tarannum
- Division of Cellular Therapy and Stem Cell Transplantation, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Rizwan Romee
- Division of Cellular Therapy and Stem Cell Transplantation, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA, 02215, USA.
| |
Collapse
|
13
|
Natural Killer Cells and Type 1 Innate Lymphoid Cells in Hepatocellular Carcinoma: Current Knowledge and Future Perspectives. Int J Mol Sci 2021; 22:ijms22169044. [PMID: 34445750 PMCID: PMC8396475 DOI: 10.3390/ijms22169044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 02/07/2023] Open
Abstract
Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1) are specific innate lymphoid cell subsets that are key for the detection and elimination of pathogens and cancer cells. In liver, while they share a number of characteristics, they differ in many features. These include their developmental pathways, tissue distribution, phenotype and functions. NK cells and ILC1 contribute to organ homeostasis through the production of key cytokines and chemokines and the elimination of potential harmful bacteria and viruses. In addition, they are equipped with a wide range of receptors, allowing them to detect “stressed cells’ such as cancer cells. Our understanding of the role of innate lymphoid cells in hepatocellular carcinoma (HCC) is growing owing to the development of mouse models, the progress in immunotherapeutic treatment and the recent use of scRNA sequencing analyses. In this review, we summarize the current understanding of NK cells and ILC1 in hepatocellular carcinoma and discuss future strategies to take advantage of these innate immune cells in anti-tumor immunity. Immunotherapies hold great promise in HCC, and a better understanding of the role and function of NK cells and ILC1 in liver cancer could pave the way for new NK cell and/or ILC1-targeted treatment.
Collapse
|
14
|
Natural Killer Cells in Cancer and Cancer Immunotherapy. Cancer Lett 2021; 520:233-242. [PMID: 34302920 DOI: 10.1016/j.canlet.2021.07.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/17/2021] [Accepted: 07/19/2021] [Indexed: 12/20/2022]
Abstract
The detection and killing of neoplastic cells require coordination of a variety of antitumor effector cells. Natural killer (NK) cells of the innate immune system are at the forefront of the body's defense systems and evidence suggests that the infiltration and cytotoxicity of NK cells in the cancer tissue influence treatment efficacy and survival. As powerful effectors in the anticancer immune response, NK cells rapidly recognize and kill transformed cells with little reactivity against healthy self-tissues, which highlights their potential role in cancer immunotherapy. Modern immunotherapeutic approaches include immune checkpoint inhibitors to revitalize dysfunctional T cells and adoptive cell transfer using CD8+ T cells with chimeric antigen receptors to enhance their functionality. However, treatment responses may be short-lived and risk of discontinuation due to adverse effects necessitates the development of safer immuno-oncologic therapies with improved outcomes. To this end, novel combinatorial interventions using T cells and NK cells and strategies for overcoming associated challenges are currently being investigated. This review summarizes the advances in the research on NK cells in cancer and cancer immunotherapy and discusses the possible implications for future cancer treatment.
Collapse
|
15
|
Hamilton G, Plangger A. The Impact of NK Cell-Based Therapeutics for the Treatment of Lung Cancer for Biologics: Targets and Therapy. Biologics 2021; 15:265-277. [PMID: 34262255 PMCID: PMC8273903 DOI: 10.2147/btt.s290305] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022]
Abstract
Lung cancer has a dismal prognosis and novel targeted therapies leave still room for major improvements and better outcomes. Immunotherapy targeting immune checkpoint (IC) proteins, either as single agents or in combination with chemotherapy, is active but responders constitute only approximately 10-15% of non-small cell lung cancer (NSCLC) patients. Other effector immune cells such as CAR-T cells or NK cells may help to overcome the limitations of the IC inhibitor therapies for lung cancer. NK cells can kill tumor cells without previous priming and are present in the circulatory system and lymphoid organs. Tissue-residing NK cells differ from peripheral effector cells and, in case of the lung, comprise CD56bright CD16-negative populations showing high cytokine release but low cytotoxicity in contrast to the circulating CD56dim CD16-positive NK cells exhibiting high cytotoxic efficacy. This local attenuation of NK cell killing potency seems due to a specific stage of NK differentiation, immunosuppressive factors as well as presence of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (TREGs). Improved NK cell-based immunotherapies involve IL-2-stimulated effector cells, NK cells expanded with the help of cytokines, permanent NK cell lines, induced pluripotent stem cell-derived NK cells and NK cells armed with chimeric antigen receptors. Compared to CAR T cell therapy, NK cells administration is devoid of graft-versus-host disease (GvHD) and cytokine-release syndrome. Although NK cells are clearly active against lung cancer cells, the low-cytotoxicity differentiation state in lung tumors, the presence of immunosuppressive leucocyte populations, limited infiltration and adverse conditions of the microenvironment need to be overcome. This goal may be achieved in the future using large numbers of activated and armed NK cells as provided by novel methods in NK cell isolation, expansion and stimulation of cytotoxic activity, including combinations with monoclonal antibodies in antibody-dependent cytotoxicity (ADCC). This review discusses the basic characteristics of NK cells and the potential of NK cell preparations in cancer therapy.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Adelina Plangger
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Jhunjhunwala S, Hammer C, Delamarre L. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat Rev Cancer 2021; 21:298-312. [PMID: 33750922 DOI: 10.1038/s41568-021-00339-z] [Citation(s) in RCA: 783] [Impact Index Per Article: 195.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/01/2021] [Indexed: 01/31/2023]
Abstract
Immune checkpoint blockade, which blocks inhibitory signals of T cell activation, has shown tremendous success in treating cancer, although success still remains limited to a fraction of patients. To date, clinically effective CD8+ T cell responses appear to target predominantly antigens derived from tumour-specific mutations that accumulate in cancer, also called neoantigens. Tumour antigens are displayed on the surface of cells by class I human leukocyte antigens (HLA-I). To elicit an effective antitumour response, antigen presentation has to be successful at two distinct events: first, cancer antigens have to be taken up by dendritic cells (DCs) and cross-presented for CD8+ T cell priming. Second, the antigens have to be directly presented by the tumour for recognition by primed CD8+ T cells and killing. Tumours exploit multiple escape mechanisms to evade immune recognition at both of these steps. Here, we review the tumour-derived factors modulating DC function, and we summarize evidence of immune evasion by means of quantitative modulation or qualitative alteration of the antigen repertoire presented on tumours. These mechanisms include modulation of antigen expression, HLA-I surface levels, alterations in the antigen processing and presentation machinery in tumour cells. Lastly, as complete abrogation of antigen presentation can lead to natural killer (NK) cell-mediated tumour killing, we also discuss how tumours can harbour antigen presentation defects and still evade NK cell recognition.
Collapse
|
17
|
Islam R, Pupovac A, Evtimov V, Boyd N, Shu R, Boyd R, Trounson A. Enhancing a Natural Killer: Modification of NK Cells for Cancer Immunotherapy. Cells 2021; 10:cells10051058. [PMID: 33946954 PMCID: PMC8146003 DOI: 10.3390/cells10051058] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells are potent innate immune system effector lymphocytes armed with multiple mechanisms for killing cancer cells. Given the dynamic roles of NK cells in tumor surveillance, they are fast becoming a next-generation tool for adoptive immunotherapy. Many strategies are being employed to increase their number and improve their ability to overcome cancer resistance and the immunosuppressive tumor microenvironment. These include the use of cytokines and synthetic compounds to bolster propagation and killing capacity, targeting immune-function checkpoints, addition of chimeric antigen receptors (CARs) to provide cancer specificity and genetic ablation of inhibitory molecules. The next generation of NK cell products will ideally be readily available as an “off-the-shelf” product and stem cell derived to enable potentially unlimited supply. However, several considerations regarding NK cell source, genetic modification and scale up first need addressing. Understanding NK cell biology and interaction within specific tumor contexts will help identify necessary NK cell modifications and relevant choice of NK cell source. Further enhancement of manufacturing processes will allow for off-the-shelf NK cell immunotherapies to become key components of multifaceted therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Rasa Islam
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| | - Aleta Pupovac
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
| | - Vera Evtimov
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
| | - Nicholas Boyd
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
| | - Runzhe Shu
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
| | - Richard Boyd
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
| | - Alan Trounson
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
- Correspondence:
| |
Collapse
|
18
|
Intratumor Regulatory Noncytotoxic NK Cells in Patients with Hepatocellular Carcinoma. Cells 2021; 10:cells10030614. [PMID: 33802077 PMCID: PMC7999652 DOI: 10.3390/cells10030614] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 12/16/2022] Open
Abstract
Previous studies support the role of natural killer (NK) cells in controlling hepatocellular carcinoma (HCC) progression. However, ambiguity remains about the multiplicity and the role of different NK cell subsets, as a pro-oncogenic function has been suggested. We performed phenotypic and functional characterization of NK cells infiltrating HCC, with the corresponding nontumorous tissue and liver from patients undergoing liver resection for colorectal liver metastasis used as controls. We identified a reduced number of NK cells in tumors with higher frequency of CD56BRIGHTCD16- NK cells associated with higher expression of NKG2A, NKp44, and NKp30 and downregulation of NKG2D. Liver-resident (CXCR6+) NK cells were reduced in the tumors where T-bethiEomeslo expression was predominant. HCCs showed higher expression of CD49a with particular enrichment in CD49a+Eomes+ NK cells, a subset typically represented in the decidua and playing a proangiogenic function. Functional analysis showed reduced TNF-α production along with impaired cytotoxic capacity that was inversely related to CXCR6-, T-bethiEomeslo, and CD49a+Eomes+ NK cells. In conclusion, we identified a subset of NK cells infiltrating HCC, including non-liver-resident cells that coexpressed CD49a and Eomes and showed reduced cytotoxic potential. This NK cell subset likely plays a regulatory role in proangiogenic function.
Collapse
|
19
|
Expansions of adaptive-like NK cells with a tissue-resident phenotype in human lung and blood. Proc Natl Acad Sci U S A 2021; 118:2016580118. [PMID: 33836578 PMCID: PMC7980282 DOI: 10.1073/pnas.2016580118] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Respiratory diseases are leading causes of death worldwide. However, the local immune cell composition in the human lung and individual outliers within the population still remain largely undescribed. We here identify adaptive-like NK cell expansions with tissue-resident traits in lung and blood in approximately 20% of individuals. This particular NK cell subset, which differed from adaptive-like CD16+ blood NK cells, was hyperresponsive to target cell stimulation. Individuals with such in vivo-primed, expanded NK cells will likely experience a different course of acute lung disease such as viral infections. Furthermore, we believe that target cell-hyperresponsive tissue-resident NK cells represent a future tool in the treatment of lung cancer. Human adaptive-like “memory” CD56dimCD16+ natural killer (NK) cells in peripheral blood from cytomegalovirus-seropositive individuals have been extensively investigated in recent years and are currently explored as a treatment strategy for hematological cancers. However, treatment of solid tumors remains limited due to insufficient NK cell tumor infiltration, and it is unknown whether large expansions of adaptive-like NK cells that are equipped for tissue residency and tumor homing exist in peripheral tissues. Here, we show that human lung and blood contains adaptive-like CD56brightCD16− NK cells with hallmarks of tissue residency, including expression of CD49a. Expansions of adaptive-like lung tissue-resident NK (trNK) cells were found to be present independently of adaptive-like CD56dimCD16+ NK cells and to be hyperresponsive toward target cells. Together, our data demonstrate that phenotypically, functionally, and developmentally distinct subsets of adaptive-like NK cells exist in human lung and blood. Given their tissue-related character and hyperresponsiveness, human lung adaptive-like trNK cells might represent a suitable alternative for therapies targeting solid tumors.
Collapse
|
20
|
Vancsik T, Máthé D, Horváth I, Várallyaly AA, Benedek A, Bergmann R, Krenács T, Benyó Z, Balogh A. Modulated Electro-Hyperthermia Facilitates NK-Cell Infiltration and Growth Arrest of Human A2058 Melanoma in a Xenograft Model. Front Oncol 2021; 11:590764. [PMID: 33732640 PMCID: PMC7959784 DOI: 10.3389/fonc.2021.590764] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 01/13/2021] [Indexed: 11/13/2022] Open
Abstract
Modulated electro-hyperthermia (mEHT), induced by 13.56 MHz radiofrequency, has been demonstrated both in preclinical and clinical studies to efficiently induce tumor damage and complement other treatment modalities. Here, we used a mouse xenograft model of human melanoma (A2058) to test mEHT (~42°C) both alone and combined with NK-cell immunotherapy. A single 30 min shot of mEHT resulted in significant tumor damage due to induced stress, marked by high hsp70 expression followed by significant upregulation of cleaved/activated caspase-3 and p53. When mEHT was combined with either primary human NK cells or the IL-2 independent NK-92MI cell line injected subcutaneously, the accumulation of NK cells was observed at the mEHT pretreated melanoma nodules but not at the untreated controls. mEHT induced the upregulation of the chemoattractant CXCL11 and increased the expression of the matrix metalloproteinase MMP2 which could account for the NK-cell attraction into the treated melanoma. In conclusion, mEHT monotherapy of melanoma xenograft tumors induced irreversible heat and cell stress leading to caspase dependent apoptosis to be driven by p53. mEHT could support the intratumoral attraction of distantly injected NK-cells, contributed by CXCL11 and MMP2 upregulation, resulting in an additive tumor destruction and growth inhibition. Therefore, mEHT may offer itself as a good partner for immunotherapy.
Collapse
Affiliation(s)
- Tamás Vancsik
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Ildikó Horváth
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | | | - Anett Benedek
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Ralf Bergmann
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Tibor Krenács
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Andrea Balogh
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
21
|
Yao X, Matosevic S. Chemokine networks modulating natural killer cell trafficking to solid tumors. Cytokine Growth Factor Rev 2021; 59:36-45. [PMID: 33495094 DOI: 10.1016/j.cytogfr.2020.12.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/17/2020] [Accepted: 12/24/2020] [Indexed: 01/02/2023]
Abstract
Natural killer (NK) cell-based cell therapy has been emerging as a powerful weapon in the treatment of multiple malignancies. However, the inadequate infiltration of the therapeutic NK cells into solid tumors remains a big challenge to their clinical utility. Chemokine networks, which play essential roles in the migration of lymphocytes, have been recognized as critical in driving the intratumoral infiltration of NK cells via interactions between soluble chemokines and their receptors. Often, such interactions are complex and disease-specific. In the context of NK cells, chemokine receptors of note have included CCR2, CCR5, CCR7, CXCR3, and CX3CR1. The immunobiology of chemokine-receptor interactions has fueled the development of approaches that hope to improve the infiltration of NK cells into the microenvironment of solid tumors. Stimulation of NK cells ex vivo in the presence of various cytokines (such as IL-2, IL-15, and IL-21) and genetic engineering of NK cells have been utilized to alter the chemokine receptor profile and generate NK cells with higher infiltrating capacity. Additionally, the immune-suppressive tumor microenvironment has also been targeted, by introducing, either directly or indirectly, chemokine ligands which NK cells are able to respond to, ultimately creating a more hospitable niche for NK cell trafficking. Such strategies have promoted the infiltration and activity of infused NK cells into multiple solid tumors. In this review, we discuss how chemokine receptors and their ligands coordinate and how they can be manipulated to regulate the trafficking, distribution, and residence of NK cells in solid tumors.
Collapse
Affiliation(s)
- Xue Yao
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| | - Sandro Matosevic
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, 47907, USA; Center for Cancer Research, Purdue University, West Lafayette, IN, 47907 USA.
| |
Collapse
|
22
|
EZH1/2 Inhibitors Favor ILC3 Development from Human HSPC-CD34 + Cells. Cancers (Basel) 2021; 13:cancers13020319. [PMID: 33467134 PMCID: PMC7830003 DOI: 10.3390/cancers13020319] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 01/19/2023] Open
Abstract
Simple Summary It has been well-demonstrated that EZH1/2 enzymes are involved not only in tumor development and progression, but also in the regulation of normal hematopoiesis from CD34+-HSPC. Given the crucial role of NK cells in tumor immune surveillance, in this study, we investigated whether EZH1/2 inhibitors can interfere with NK cell differentiation and functional maturation. Our results suggest that EZH1/2 inhibitors push CD56+ precursor proliferation, skewing precursor cell lineage commitment towards ILC3. In recent years, several clinical trials on the use of EZH1/2 inhibitors against solid tumors have been carried out. Since these in vitro observations revealed possible epigenetic mechanisms involved in NK/ILC development, it is important to evaluate patient monitoring of competent NK cells repertoire in order to design appropriate therapeutic protocols. Abstract The dysregulation of epigenetic modifications has a well-established role in the development and progression of hematological malignancies and of solid tumors. In this context, EZH1/2 inhibitors have been designed to interfere with EZH1/2 enzymes involved in histone methylation (e.g., H3K27me3), leading to tumor growth arrest or the restoration of tumor suppressor gene transcription. However, these compounds also affect normal hematopoiesis, interfering with self-renewal and differentiation of CD34+-Hematopoietic Stem/Progenitor Cells (HSPC), and, in turn, could modulate the generation of potential anti-tumor effector lymphocytes. Given the important role of NK cells in the immune surveillance of tumors, it would be useful to understand whether epigenetic drugs can modulate NK cell differentiation and functional maturation. CD34+-HSPC were cultured in the absence or in the presence of the EZH1/2 inhibitor UNC1999 and EZH2 inhibitor GSK126. Our results show that UNC1999 and GSK126 increased CD56+ cell proliferation compared to the control condition. However, UNC1999 and GSK 126 favored the proliferation of no-cytotoxic CD56+ILC3, according to the early expression of the AHR and ROR-γt transcription factors. Our results describe novel epigenetic mechanisms involved in the modulation of NK cell maturation that may provide new tools for designing NK cell-based immunotherapy.
Collapse
|
23
|
Agostinis C, Balduit A, Mangogna A, Zito G, Romano F, Ricci G, Kishore U, Bulla R. Immunological Basis of the Endometriosis: The Complement System as a Potential Therapeutic Target. Front Immunol 2021; 11:599117. [PMID: 33505394 PMCID: PMC7829336 DOI: 10.3389/fimmu.2020.599117] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Endometriosis (EM) is a chronic disease characterized by the presence and proliferation of functional endometrial glands and stroma outside the uterine cavity. Ovaries and pelvic peritoneum are the most common locations for endometrial ectopic tissue, followed by deep infiltrating EM sites. The cyclic and recurrent bleeding, the progressive fibrosis and the peritoneal adhesions of ectopic endometrial glands, may cause different symptoms depending on the origin involved. EM is a frequent clinical condition affecting around 10% of women of mainly reproductive age, as well as in post-menopausal women and adolescents, especially with uterine anomalies. The risk of developing EM depends on a complex interaction between genetic, immunological, hormonal, and environmental factors. It is largely considered to arise due to a dysfunction of immunological surveillance. In fact, women with EM exhibit altered functions of peritoneal macrophages, lymphocytes and natural killer cells, as well as levels of inflammatory mediators and growth factors in the peritoneal fluid. In EM patients, peritoneal macrophages are preponderant and highly active compared to healthy women. Peritoneal macrophages are able to regulate the events that determine the production of cytokines, prostaglandins, growth factors and complement components. Several studies have shown alteration in the regulation of the complement activation, leading to chronic inflammation characteristic of EM. Aberrant regulation/activation of the complement system has been observed in the peritoneal cavity of women affected by EM. Thus, complement inhibition may represent a new approach for the treatment of EM, given that a number of complement inhibitors are under pre-clinical and clinical development. Such an intervention may provide a broader therapeutic control of complement-mediated inflammatory damage in EM patients. This review will focus on our current understanding of the role of complement activation in EM and possible modalities available for complement-based therapy.
Collapse
Affiliation(s)
- Chiara Agostinis
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Andrea Balduit
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Gabriella Zito
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Federico Romano
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo", Trieste, Italy.,Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
24
|
Current Immunotherapy Approaches in Non-Hodgkin Lymphomas. Vaccines (Basel) 2020; 8:vaccines8040708. [PMID: 33260966 PMCID: PMC7768428 DOI: 10.3390/vaccines8040708] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
Non-Hodgkin lymphomas (NHLs) are lymphoid malignancies of B- or T-cell origin. Despite great advances in treatment options and significant improvement of survival parameters, a large part of NHL patients either present with a chemotherapy-refractory disease or experience lymphoma relapse. Chemotherapy-based salvage therapy of relapsed/refractory NHL is, however, capable of re-inducing long-term remissions only in a minority of patients. Immunotherapy-based approaches, including bispecific antibodies, immune checkpoint inhibitors and genetically engineered T-cells carrying chimeric antigen receptors, single-agent or in combination with therapeutic monoclonal antibodies, immunomodulatory agents, chemotherapy or targeted agents demonstrated unprecedented clinical activity in heavily-pretreated patients with NHL, including chemotherapy-refractory cases with complex karyotype changes and other adverse prognostic factors. In this review, we recapitulate currently used immunotherapy modalities in NHL and discuss future perspectives of combinatorial immunotherapy strategies, including patient-tailored approaches.
Collapse
|
25
|
Farcas M, Inngjerdingen M. Natural killer cell–derived extracellular vesicles in cancer therapy. Scand J Immunol 2020; 92:e12938. [DOI: 10.1111/sji.12938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/25/2020] [Accepted: 07/17/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Marius Farcas
- Department of Pharmacology Institute of Clinical Medicine University of Oslo and Oslo University Hospital Oslo Norway
| | - Marit Inngjerdingen
- Department of Pharmacology Institute of Clinical Medicine University of Oslo and Oslo University Hospital Oslo Norway
| |
Collapse
|
26
|
Isolation of Two Novel Human Anti-CTLA-4 mAbs with Intriguing Biological Properties on Tumor and NK Cells. Cancers (Basel) 2020; 12:cancers12082204. [PMID: 32781690 PMCID: PMC7464132 DOI: 10.3390/cancers12082204] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/01/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
The cytotoxic T lymphocyte-antigen 4 (CTLA-4) has been considered an IC exclusively expressed on T cells, where it counteracts the co-stimulatory CD28 receptor, by competing for its binding to CD-80 and CD-86. We recently found that it is expressed also on tumor and NK cells, suggesting other possible unknown roles of CTLA-4. To shed light on these novel aspects of CTLA-4, we used Ipilimumab, the first FDA approved human antibody targeting CTLA-4, in parallel studies with two novel human mAbs we isolated by using an efficient phage display selection strategy on live activated lymphocytes and purified mouse and human CTLA-4. The selection for cross-reactive mAbs was guaranteed by a high throughput sequencing to identify the sequences commonly enriched by two parallel pannings on human and mouse CTLA-4. Two isolated antibodies were found to bind with high affinity to both human and mouse CTLA-4 and lymphocytes, showing nanomolar or sub-nanomolar Kd values. They were able to kill Treg cells by ADCC, and to activate both human and mouse PBMCs, by strongly increasing cytokines secretion. Interestingly, they activated NK cells, exhibited cytotoxicity against cancer cells by inducing ADCC and inhibited tumor cell growth by affecting CTLA-4 downstream pathways in a similar fashion to CD-80 and CD-86 ligands and differently from Ipilimumab. Moreover, the novel mAbs showed a reduced ability to interfere in the binding of CD-80 ligands to CTLA-4 on T cells with respect to Ipilimumab, suggesting that they could allow for anti-tumor effects without the irAEs associated with the potent antagonistic activity of Ipilimumab.
Collapse
|
27
|
Bald T, Krummel MF, Smyth MJ, Barry KC. The NK cell-cancer cycle: advances and new challenges in NK cell-based immunotherapies. Nat Immunol 2020; 21:835-847. [PMID: 32690952 DOI: 10.1038/s41590-020-0728-z] [Citation(s) in RCA: 310] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022]
Abstract
Natural killer (NK) cells belong to the innate immune system and contribute to protecting the host through killing of infected, foreign, stressed or transformed cells. Additionally, via cellular cross-talk, NK cells orchestrate antitumor immune responses. Hence, significant efforts have been undertaken to exploit the therapeutic properties of NK cells in cancer. Current strategies in preclinical and clinical development include adoptive transfer therapies, direct stimulation, recruitment of NK cells into the tumor microenvironment (TME), blockade of inhibitory receptors that limit NK cell functions, and therapeutic modulation of the TME to enhance antitumor NK cell function. In this Review, we introduce the NK cell-cancer cycle to highlight recent advances in NK cell biology and to discuss the progress and problems of NK cell-based cancer immunotherapies.
Collapse
Affiliation(s)
- Tobias Bald
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Matthew F Krummel
- Department of Pathology, ImmunoX Initiative, and Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA
| | - Mark J Smyth
- Immunology of Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.
| | - Kevin C Barry
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| |
Collapse
|
28
|
Damele L, Ottonello S, Mingari MC, Pietra G, Vitale C. Targeted Therapies: Friends or Foes for Patient's NK Cell-Mediated Tumor Immune-Surveillance? Cancers (Basel) 2020; 12:cancers12040774. [PMID: 32218226 PMCID: PMC7226262 DOI: 10.3390/cancers12040774] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/14/2020] [Accepted: 03/23/2020] [Indexed: 12/30/2022] Open
Abstract
In the last 20 years there has been a huge increase in the number of novel drugs for cancer treatment. Most of them exploit their ability to target specific oncogenic mutations in the tumors (targeted therapies–TT), while others target the immune-checkpoint inhibitor molecules (ICI) or the epigenetic DNA modifications. Among them, TT are the longest established drugs exploited against a wide spectrum of both solid and hematological tumors, often with reasonable costs and good efficacy as compared to other innovative therapies (i.e., ICI). Although they have greatly improved the treatment of cancer patients and their survival, patients often relapse or develop drug-resistance, leading to the impossibility to eradicate the disease. The outcome of TT has been often correlated with their ability to affect not only tumor cells, but also the repertoire of immune cells and their ability to interact with cancer cells. Thus, the possibility to create novel synergies among drugs an immunotherapy prompted scientists and physicians to deeply characterize the effects of TT on immune cells both by in-vitro and by ex-vivo analyses. In this context, NK cells may represent a key issue, since they have been shown to exert a potent anti-tumor activity, both against hematological malignancies and solid tumors. In the present review we will discuss most recent ex-vivo analyses that clarify the effect of TT treatment on patient’s NK cells comparing them with clinical outcome and previous in-vitro data.
Collapse
Affiliation(s)
- Laura Damele
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.D.); (S.O.); (M.C.M.); (G.P.)
| | - Selene Ottonello
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.D.); (S.O.); (M.C.M.); (G.P.)
| | - Maria Cristina Mingari
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.D.); (S.O.); (M.C.M.); (G.P.)
- Dipartimento Medicina Sperimentale (DIMES), Università degli Studi di Genova, 16132 Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), Università degli Studi di Genova, 16132 Genoa, Italy
| | - Gabriella Pietra
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.D.); (S.O.); (M.C.M.); (G.P.)
- Dipartimento Medicina Sperimentale (DIMES), Università degli Studi di Genova, 16132 Genoa, Italy
| | - Chiara Vitale
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (L.D.); (S.O.); (M.C.M.); (G.P.)
- Dipartimento Medicina Sperimentale (DIMES), Università degli Studi di Genova, 16132 Genoa, Italy
- Correspondence:
| |
Collapse
|
29
|
Passariello M, Camorani S, Vetrei C, Ricci S, Cerchia L, De Lorenzo C. Ipilimumab and Its Derived EGFR Aptamer-Based Conjugate Induce Efficient NK Cell Activation against Cancer Cells. Cancers (Basel) 2020; 12:E331. [PMID: 32024070 PMCID: PMC7072174 DOI: 10.3390/cancers12020331] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/21/2020] [Accepted: 01/30/2020] [Indexed: 01/05/2023] Open
Abstract
The immune checkpoint CTLA-4 (cytotoxic T-lymphocyte-antigen 4), which inhibits the co-stimulatory CD28 signal on T cells, has been recently found expressed on other cell populations, such as tumor and natural killer (NK) cells. We tested for the first time the effects of ipilimumab, the human anti-CTLA4 mAb in clinical use, on these cells and found that it inhibits the growth of tumor cells expressing CTLA-4 also in the absence of lymphocytes, and efficiently activates NK cells, thus suggesting an important unexplored role of NK cells in ipilimumab-modulated immune responses. Interestingly, the epidermal growth factor receptor (EGFR) has been shown to play a key role in tumor cell escape from immune surveillance, and in cytotoxic T lymphocyte inhibition. Thus, we tested combinatorial treatments of ipilimumab with an anti-EGFR aptamer endowed with anti-tumor activity, and constructed for the first time a novel bispecific immunoconjugate, made up of these two compounds. The novel immunoconjugate binds to the target cells, induces the activation of lymphocytes, including NK cells, and inhibits the growth of tumor target cells more efficiently than the parental compounds, by strongly enhancing the cytotoxic activity of both human peripheral blood mononuclear cells and NK cells against tumor cells.
Collapse
Affiliation(s)
- Margherita Passariello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (M.P.); (C.V.); (S.R.)
- Ceinge – Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Simona Camorani
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS), CNR, Via S. Pansini 5, 80131 Naples, Italy
| | - Cinzia Vetrei
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (M.P.); (C.V.); (S.R.)
- Ceinge – Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Stefania Ricci
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (M.P.); (C.V.); (S.R.)
- Ceinge – Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS), CNR, Via S. Pansini 5, 80131 Naples, Italy
| | - Claudia De Lorenzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (M.P.); (C.V.); (S.R.)
- Ceinge – Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145 Naples, Italy
| |
Collapse
|
30
|
Roderburg C, Wree A, Demir M, Schmelzle M, Tacke F. The role of the innate immune system in the development and treatment of hepatocellular carcinoma. Hepat Oncol 2020; 7:HEP17. [PMID: 32273975 PMCID: PMC7137177 DOI: 10.2217/hep-2019-0007] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer. Most patients present with advanced or metastatic HCC at diagnosis and face a dismal prognosis. Tyrosine kinases are the gold standard treatment for this disease but yield limited survival benefits. Immune checkpoint inhibitors that augment adaptive immunity have been tested in HCC. Complex interactions between tumor cells, lymphocytes and the tumor environment determine the efficacy of such immunotherapies. Innate immune mechanisms – known drivers of liver disease progression in pre-HCC conditions such as fibrosis or cirrhosis – may either support or counteract tumor-related immune activation. In this review, we will highlight current concepts of the role of the innate immune system in hepatocarcinogenesis and discuss their relevance for translation into clinics.
Collapse
Affiliation(s)
- Christoph Roderburg
- Department of Hepatology & Gastroenterology, Charité, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Campus Charité Mitte, Campus Virchow-Klinikum, Berlin Institute of Health, Augustenburger, Platz 1 13353, Berlin
| | - Alexander Wree
- Department of Hepatology & Gastroenterology, Charité, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Campus Charité Mitte, Campus Virchow-Klinikum, Berlin Institute of Health, Augustenburger, Platz 1 13353, Berlin
| | - Münevver Demir
- Department of Hepatology & Gastroenterology, Charité, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Campus Charité Mitte, Campus Virchow-Klinikum, Berlin Institute of Health, Augustenburger, Platz 1 13353, Berlin
| | - Moritz Schmelzle
- Department of Surgery, Charité, Universitätsmedizin Berlin,Humboldt-Universität zu Berlin, Campus Charité Mitte, Campus Virchow-Klinikum, Berlin Institute of Health, Augustenburger, Platz 1 13353, Berlin
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Campus Charité Mitte, Campus Virchow-Klinikum, Berlin Institute of Health, Augustenburger, Platz 1 13353, Berlin
| |
Collapse
|
31
|
Machado FC, Girola N, Maia VSC, Bergami-Santos PC, Morais AS, Azevedo RA, Figueiredo CR, Barbuto JAM, Travassos LR. Immunomodulatory Protective Effects of Rb9 Cyclic-Peptide in a Metastatic Melanoma Setting and the Involvement of Dendritic Cells. Front Immunol 2020; 10:3122. [PMID: 32010152 PMCID: PMC6974543 DOI: 10.3389/fimmu.2019.03122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 12/23/2019] [Indexed: 11/30/2022] Open
Abstract
The cyclic VHCDR3-derived peptide (Rb9) from RebMab200 antibody, directed to a NaPi2B phosphate-transport protein, displayed anti-metastatic melanoma activity at 50–300 μg intraperitoneally injected in syngeneic mice. Immune deficient mice failed to respond to the peptide protective effect. Rb9 induced increased CD8+ T and low Foxp3+ T cell infiltration in lung metastases and high IFN-γ and low TGF-β in lymphoid organs. The peptide co-localized with F-actin and a nuclear site in dendritic cells and specifically bound to MIF and CD74 in a dot-blot setting. Murine bone-marrow dendritic cells preincubated with Rb9 for 6 h were treated with MIF for short time periods. The modulated responses showed stimulation of CD74 and inhibition of pPI3K, pERK, and pNF-κB as compared to MIF alone. Rb9 in a melanoma-conditioned medium, stimulated the M1 type conversion in bone marrow-macrophages. Functional aspects of Rb9 in vivo were studied in therapeutic and prophylactic protocols using a melanoma metastatic model. In both protocols Rb9 exhibited a marked anti-melanoma protection. Human dendritic cells were also investigated showing increased expression of surface markers in response to Rb9 incubation. Rb9 either stimulated or slightly inhibited moDCs submitted to inhibitory (TGF-β and IL-10) or activating (LPS) conditions, respectively. Lymphocyte proliferation was obtained with moDCs stimulated by Rb9 and tumor cell lysate. In moDCs from cancer patients Rb9 exerted immunomodulatory activities depending on their functional status. The peptide may inhibit over-stimulated cells, stimulate poorly activated and suppressed cells, or cause instead, little phenotypic and functional alterations. Recently, the interaction MIF-CD74 has been associated to PD-L1 expression and IFN-γ, suggesting a target for melanoma treatment. The effects described for Rb9 and the protection against metastatic melanoma may suggest the possibility of a peptide reagent that could be relevant when associated to modern immunotherapeutic procedures.
Collapse
Affiliation(s)
- Fabrício C Machado
- Recepta Bio, São Paulo, Brazil.,Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Natália Girola
- Recepta Bio, São Paulo, Brazil.,Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Vera S C Maia
- Recepta Bio, São Paulo, Brazil.,Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Patrícia C Bergami-Santos
- Recepta Bio, São Paulo, Brazil.,Tumor Immunology Laboratory, Department of Immunology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | | | - Ricardo A Azevedo
- Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Carlos R Figueiredo
- Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil.,MediCity, University of Turku, Turku, Finland
| | - José A M Barbuto
- Recepta Bio, São Paulo, Brazil.,Tumor Immunology Laboratory, Department of Immunology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Luiz R Travassos
- Recepta Bio, São Paulo, Brazil.,Experimental Oncology Unit, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
32
|
Fantini M, David JM, Annunziata CM, Morelli MP, Arlen PM, Tsang KY. The Monoclonal Antibody NEO-201 Enhances Natural Killer Cell Cytotoxicity Against Tumor Cells Through Blockade of the Inhibitory CEACAM5/CEACAM1 Immune Checkpoint Pathway. Cancer Biother Radiopharm 2020; 35:190-198. [PMID: 31928422 DOI: 10.1089/cbr.2019.3141] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background: Natural killer (NK) cells are essential to innate immunity and participate in cancer immune surveillance. Heterophilic interactions between carcinoembryonic antigen (CEA) on tumor cells and carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) on NK cells inhibit NK cell cytotoxicity against tumor cells. NEO-201 is a humanized IgG1 monoclonal antibody that recognizes members of CEACAM family, expressed specifically on a variety of human carcinoma cell lines and tumor tissues. This investigation was designed to determine whether the binding of NEO-201 with CEACAM5 on tumor cells can block the CEACAM5/CEACAM1 interaction to restore antitumor cytotoxicity of NK cells. Materials and Methods: In vitro functional assays, using various human tumor cell lines as target cells and NK-92 cells as effectors, were conducted to assess the ability of NEO-201 to block the interaction between CEACAM5 on tumor cells and CEACAM1 on NK cells to enhance the in vitro killing of tumor cells by NK-92. NK-92 cells were used as a model of direct NK killing of tumor cells because they lack antibody-dependent cellular cytotoxicity activity. Results: Expression profiling revealed that various human carcinoma cell lines expressed different levels of CEACAM5+ and NEO-201+ cells. Addition of NEO-201 significantly enhanced NK-92 cell cytotoxicity against highly CEACAM5+/NEO-201+ expressing tumor cells, suggesting that its activity is correlated with the level of CEACAM5+/NEO-201+ expression. Conclusions: These findings demonstrate that NEO-201 can block the interaction between CEACAM5 on tumor cells and CEACAM1 on NK cells to reverse CEACAM1-dependent inhibition of NK cytotoxicity.
Collapse
Affiliation(s)
| | | | - Christina M Annunziata
- Women's Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Maria Pia Morelli
- Women's Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
33
|
Minetto P, Guolo F, Pesce S, Greppi M, Obino V, Ferretti E, Sivori S, Genova C, Lemoli RM, Marcenaro E. Harnessing NK Cells for Cancer Treatment. Front Immunol 2019; 10:2836. [PMID: 31867006 PMCID: PMC6908847 DOI: 10.3389/fimmu.2019.02836] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
In the last years, natural killer (NK) cell-based immunotherapy has emerged as a promising therapeutic approach for solid tumors and hematological malignancies. NK cells are innate lymphocytes with an array of functional competences, including anti-cancer, anti-viral, and anti-graft-vs.-host disease potential. The intriguing idea of harnessing such potent innate immune system effectors for cancer treatment led to the development of clinical trials based on the adoptive therapy of NK cells or on the use of monoclonal antibodies targeting the main NK cell immune checkpoints. Indeed, checkpoint immunotherapy that targets inhibitory receptors of T cells, reversing their functional blocking, marked a breakthrough in anticancer therapy, opening new approaches for cancer immunotherapy and resulted in extensive research on immune checkpoints. However, the clinical efficacy of T cell-based immunotherapy presents a series of limitations, including the inability of T cells to recognize and kill HLA-Ineg tumor cells. For these reasons, new strategies for cancer immunotherapy are now focusing on NK cells. Blockade with NK cell checkpoint inhibitors that reverse their functional block may overcome the limitations of T cell-based immunotherapy, mainly against HLA-Ineg tumor targets. Here, we discuss recent anti-tumor approaches based on mAb-mediated blocking of immune checkpoints (either restricted to NK cells or shared with T cells), used either as a single agent or in combination with other compounds, that have demonstrated promising clinical responses in both solid tumors and hematological malignancies.
Collapse
Affiliation(s)
- Paola Minetto
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genova, Italy.,Ospedale Policlinico San Martino IRCCS, Genova, Italy
| | - Fabio Guolo
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genova, Italy.,Ospedale Policlinico San Martino IRCCS, Genova, Italy
| | - Silvia Pesce
- Department of Experimental Medicine, University of Genoa, Genova, Italy
| | - Marco Greppi
- Department of Experimental Medicine, University of Genoa, Genova, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Valentina Obino
- Department of Experimental Medicine, University of Genoa, Genova, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Elisa Ferretti
- Department of Experimental Medicine, University of Genoa, Genova, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Simona Sivori
- Department of Experimental Medicine, University of Genoa, Genova, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Carlo Genova
- Lung Cancer Unit, Ospedale Policlinico San Martino IRCCS, Genova, Italy
| | - Roberto Massimo Lemoli
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genova, Italy.,Ospedale Policlinico San Martino IRCCS, Genova, Italy
| | - Emanuela Marcenaro
- Department of Experimental Medicine, University of Genoa, Genova, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| |
Collapse
|
34
|
NK Cells as Potential Targets for Immunotherapy in Endometriosis. J Clin Med 2019; 8:jcm8091468. [PMID: 31540116 PMCID: PMC6780982 DOI: 10.3390/jcm8091468] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 12/19/2022] Open
Abstract
Endometriosis is a common gynecological disease defined by the presence of endometrial-like tissue outside the uterus, most frequently on the pelvic viscera and ovaries, which is associated with pelvic pains and infertility. It is an inflammatory disorder with some features of autoimmunity. It is accepted that ectopic endometriotic tissue originates from endometrial cells exfoliated during menstruation and disseminating into the peritoneum by retrograde menstrual blood flow. It is assumed that the survival of endometriotic cells in the peritoneal cavity may be partially due to their abrogated elimination by natural killer (NK) cells. The decrease of NK cell cytotoxic activity in endometriosis is associated with an increased expression of some inhibitory NK cell receptors. It may be also related to the expression of human leukocyte antigen G (HLA-G), a ligand for inhibitory leukocyte immunoglobulin-like receptor subfamily B member 1 (LILRB1) receptors. The downregulated cytotoxic activity of NK cells may be due to inhibitory cytokines present in the peritoneal milieu of patients with endometriosis. The role of NK cell receptors and their ligands in endometriosis is also confirmed by genetic association studies. Thus, endometriosis may be a subject of immunotherapy by blocking NK cell negative control checkpoints including inhibitory NK cell receptors. Immunotherapies with genetically modified NK cells also cannot be excluded.
Collapse
|