1
|
Bayona C, Olaizola-Rodrigo C, Sharko V, Ashrafi M, Barrio JD, Doblaré M, Monge R, Ochoa I, Oliván S. A Novel Multicompartment Barrier-Free Microfluidic Device Reveals the Impact of Extracellular Matrix Stiffening and Temozolomide on Immune-Tumor Interactions in Glioblastoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409229. [PMID: 39901479 DOI: 10.1002/smll.202409229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/17/2025] [Indexed: 02/05/2025]
Abstract
The immune system plays a crucial role in shaping the glioblastoma tumor microenvironment, characterized by its complexity and dynamic interactions. Understanding the tumor-immune crosstalk is essential for advancing cancer research and therapeutic development. Here, a novel multicompartment, barrier-free microfluidic device is presented that overcomes the limitations of existing models by enabling direct tumor-immune interactions without physical barriers, preserving natural immune cell infiltration. This platform supports the independent and simultaneous culture of tumor and immune cells, replicating the healthy-tumoral stroma interface, and allows investigating the effect of matrix stiffness and chemotherapy on both populations. The findings reveal that increased collagen concentration promotes tumor invasiveness while impairing immune cell infiltration. Additionally, temozolomide treatment reduces immune cell motility but enhances anti-tumor immune responses. These insights highlight the critical roles of extracellular matrix mechanics and chemotherapy in tumor progression and immune modulation, establishing this device as a powerful tool for studying glioblastoma-immune dynamics and evaluating therapeutic strategies.
Collapse
Affiliation(s)
- Clara Bayona
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
| | - Claudia Olaizola-Rodrigo
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- BEOnChip S.L., Zaragoza, 50018, Spain
| | - Vira Sharko
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
| | - Mehran Ashrafi
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
| | - Jesús Del Barrio
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| | - Manuel Doblaré
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, Department of Organic Chemistry, Zaragoza, 50009, Spain
| | | | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, Department of Organic Chemistry, Zaragoza, 50009, Spain
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
| |
Collapse
|
2
|
Rahimi A, Malakoutikhah Z, Rahimmanesh I, Ferns GA, Nedaeinia R, Ishaghi SMM, Dana N, Haghjooy Javanmard S. The nexus of natural killer cells and melanoma tumor microenvironment: crosstalk, chemotherapeutic potential, and innovative NK cell-based therapeutic strategies. Cancer Cell Int 2023; 23:312. [PMID: 38057843 DOI: 10.1186/s12935-023-03134-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/09/2023] [Indexed: 12/08/2023] Open
Abstract
The metastasis of melanoma cells to regional lymph nodes and distant sites is an important contributor to cancer-related morbidity and mortality among patients with melanoma. This intricate process entails dynamic interactions involving tumor cells, cellular constituents, and non-cellular elements within the microenvironment. Moreover, both microenvironmental and systemic factors regulate the metastatic progression. Central to immunosurveillance for tumor cells are natural killer (NK) cells, prominent effectors of the innate immune system with potent antitumor and antimetastatic capabilities. Recognizing their pivotal role, contemporary immunotherapeutic strategies are actively integrating NK cells to combat metastatic tumors. Thus, a meticulous exploration of the interplay between metastatic melanoma and NK cells along the metastatic cascade is important. Given the critical involvement of NK cells within the melanoma tumor microenvironment, this comprehensive review illuminates the intricate relationship between components of the melanoma tumor microenvironment and NK cells, delineating their multifaceted roles. By shedding light on these critical aspects, this review advocates for a deeper understanding of NK cell dynamics within the melanoma context, driving forward transformative strategies to combat this cancer.
Collapse
Affiliation(s)
- Azadeh Rahimi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Malakoutikhah
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Nasim Dana
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
3
|
Schendel DJ. Evolution by innovation as a driving force to improve TCR-T therapies. Front Oncol 2023; 13:1216829. [PMID: 37810959 PMCID: PMC10552759 DOI: 10.3389/fonc.2023.1216829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/16/2023] [Indexed: 10/10/2023] Open
Abstract
Adoptive cell therapies continually evolve through science-based innovation. Specialized innovations for TCR-T therapies are described here that are embedded in an End-to-End Platform for TCR-T Therapy Development which aims to provide solutions for key unmet patient needs by addressing challenges of TCR-T therapy, including selection of target antigens and suitable T cell receptors, generation of TCR-T therapies that provide long term, durable efficacy and safety and development of efficient and scalable production of patient-specific (personalized) TCR-T therapy for solid tumors. Multiple, combinable, innovative technologies are used in a systematic and sequential manner in the development of TCR-T therapies. One group of technologies encompasses product enhancements that enable TCR-T therapies to be safer, more specific and more effective. The second group of technologies addresses development optimization that supports discovery and development processes for TCR-T therapies to be performed more quickly, with higher quality and greater efficiency. Each module incorporates innovations layered onto basic technologies common to the field of immunology. An active approach of "evolution by innovation" supports the overall goal to develop best-in-class TCR-T therapies for treatment of patients with solid cancer.
Collapse
Affiliation(s)
- Dolores J. Schendel
- Medigene Immunotherapies GmbH, Planegg, Germany
- Medigene AG, Planegg, Germany
| |
Collapse
|
4
|
Mao L, Lian B, Li C, Bai X, Zhou L, Cui C, Chi Z, Sheng X, Wang X, Tang B, Yan X, Li S, Kong Y, Dai J, Wei X, Li J, Duan R, Xu H, Wu X, Yang Y, Cheng F, Zhang C, Xia F, Pang Z, Guo J, Si L. Camrelizumab Plus Apatinib and Temozolomide as First-Line Treatment in Patients With Advanced Acral Melanoma: The CAP 03 Phase 2 Nonrandomized Clinical Trial. JAMA Oncol 2023; 9:1099-1107. [PMID: 37261804 PMCID: PMC10236335 DOI: 10.1001/jamaoncol.2023.1363] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/13/2023] [Indexed: 06/02/2023]
Abstract
Importance Acral melanoma, known for low tumor mutation burden, responds poorly to immunotherapy. A standard therapy is still lacking. Objective To investigate the activity and safety of camrelizumab (an anti-programmed cell death-1 antibody) plus apatinib (a vascular endothelial growth factor receptor 2 inhibitor) and temozolomide as first-line treatment in patients with advanced acral melanoma. Design, Setting, and Participants In this single-arm, single-center, phase 2 nonrandomized clinical trial, patients with treatment-naive unresectable stage III or IV acral melanoma were enrolled at Peking University Cancer Hospital and Institute between June 4, 2020, and August 24, 2021. The data cutoff date was April 10, 2022. Interventions Patients received 4-week cycles of intravenous camrelizumab, 200 mg, every 2 weeks; oral apatinib 250 mg, once daily; and intravenous temozolomide, 200 mg/m2, once daily on days 1 to 5 until disease progression or unacceptable toxic effects. Main Outcomes and Measures The primary end point was objective response rate as assessed by investigators according to the Response Evaluation Criteria In Solid Tumors (version 1.1). Secondary end points included progression-free survival, time to response, duration of response, disease control rate, overall survival, and safety. Results A total of 50 patients (32 men [64%]; median age, 57 years [IQR, 52-62 years]) were enrolled and received treatment. The median follow-up duration was 13.4 months (IQR, 9.6-16.2 months). The objective response rate was 64.0% (32 of 50; 95% CI, 49.2%-77.1%). The median time to response and duration of response were 2.7 months (IQR, 0.9-2.9 months) and 17.5 months (95% CI, 12.0 to not reached), respectively. The disease control rate was 88.0% (44 of 50; 95% CI, 75.7%-95.5%). The estimated median progression-free survival was 18.4 months (95% CI, 10.6 to not reached). The median overall survival was not reached. The most common grade 3 or 4 treatment-related adverse events were increased gamma-glutamyltransferase levels (15 [30%]), decreased neutrophil count (11 [22%]), increased conjugated bilirubin levels (10 [20%]), and increased aspartate aminotransferase levels (10 [20%]). No treatment-related deaths occurred. Conclusions and Relevance The findings of this nonrandomized clinical trial suggest that camrelizumab plus apatinib and temozolomide may be a potential first-line treatment option for patients with advanced acral melanoma, which warrants further validation in a randomized clinical trial. Trial Registration ClinicalTrials.gov Identifier: NCT04397770.
Collapse
Affiliation(s)
- Lili Mao
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Bin Lian
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Caili Li
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xue Bai
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Li Zhou
- Department of Genitourinary Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Chuanliang Cui
- Department of Genitourinary Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhihong Chi
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xinan Sheng
- Department of Genitourinary Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xuan Wang
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Bixia Tang
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xieqiao Yan
- Department of Genitourinary Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Siming Li
- Department of Genitourinary Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yan Kong
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jie Dai
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaoting Wei
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Juan Li
- Department of Genitourinary Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Rong Duan
- Department of Genitourinary Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Huayan Xu
- Department of Genitourinary Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaowen Wu
- Department of Genitourinary Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yue Yang
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Fengzhuo Cheng
- Department of Medical Affairs, Jiangsu Hengrui Pharmaceuticals Co, Ltd, Shanghai, China
| | - Cheng Zhang
- Department of Medical Affairs, Jiangsu Hengrui Pharmaceuticals Co, Ltd, Shanghai, China
| | - Fangzhou Xia
- Department of Medical Affairs, Jiangsu Hengrui Pharmaceuticals Co, Ltd, Shanghai, China
| | - Zheng Pang
- Department of Medical Affairs, Jiangsu Hengrui Pharmaceuticals Co, Ltd, Shanghai, China
| | - Jun Guo
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lu Si
- Department of Melanoma and Sarcoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
5
|
Yin L, Wan Z, Sun P, Shuai P, Liu Y. Time to abandon CAR-T monotherapy for solid tumors. Biochim Biophys Acta Rev Cancer 2023; 1878:188930. [PMID: 37286147 DOI: 10.1016/j.bbcan.2023.188930] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023]
Abstract
In recent decades, chimeric antigen receptor T (CAR-T) cell therapy has achieved dramatic success in patients with hematological malignancies. However, CAR-T cell therapy failed to effectively treat solid tumors as a monotherapy. By summarizing the challenges of CAR-T cell monotherapy for solid tumors and analyzing the underlying mechanisms of combinatorial strategies to counteract these hurdles, we found that complementary therapeutics are needed to improve the scant and transient responses of CAR-T cell monotherapy in solid tumors. Further data, especially data from multicenter clinical trials regarding efficacy, toxicity, and predictive biomarkers are required before the CAR-T combination therapy can be translated into clinical settings.
Collapse
Affiliation(s)
- Limei Yin
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Zhengwei Wan
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Ping Sun
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Ping Shuai
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.
| | - Yuping Liu
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.
| |
Collapse
|
6
|
Wang Q, Shen X, An R, Bai J, Dong J, Cai H, Zhu H, Zhong W, Chen W, Liu A, Du J. Peritumoral tertiary lymphoid structure and tumor stroma percentage predict the prognosis of patients with non-metastatic colorectal cancer. Front Immunol 2022; 13:962056. [PMID: 36189233 PMCID: PMC9524924 DOI: 10.3389/fimmu.2022.962056] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTertiary lymphoid structures (TLSs) are crucial in promoting and maintaining positive anti-tumor immune responses. The tumor stroma has a powerful immunosuppressive function that could exclude tumor-infiltrating lymphocytes from the tumor beds and lead to a “cold” phenotype. TLSs and tumor stroma percentage (TSP) are significantly associated with the prognosis of patients with certain cancers. However, the exact roles of TLSs and TSP and their intrinsic relationship are still largely unknown in colorectal cancer (CRC).MethodsTLSs and TSP were assessed using hematoxylin-eosin (H&E) and/or immunohistochemistry (IHC) staining from 114 CRC patients in the training set and 60 CRC patients in the external validation set. The correlation between TILs, TLS and clinicopathological characteristics and their prognostic values were assessed. Finally, we plotted a Nomogram including the TLS, TSP and tumor-node-metastasis (TNM) stage to predict the probability of recurrence-free survival (RFS) at 2- and 5-years in non-metastatic colorectal cancer (nmCRC) patients.ResultsPeritumoral TLS (P-TLS), intratumoral TLS (In-TLS) and high TSP (H-TSP, >50%) were present in 99.1%, 26.3% and 41.2% patients, respectively. H-TSP tumor tends to be associated with lower P-TLS density (P =0.0205). The low P-TLS density (< 0.098/mm2) was significantly associated with reduced RFS (HR=6.597 95% CI: 2.882-15.103, P <0.001) and reduced overall survival (OS) (HR=6.628 95% CI: 2.893-15.183, P < 0.001) of nmCRC patients. In-TLS was not of significance in evaluating the clinical outcomes of nmCRC patients. H-TSP was significantly associated with reduced RFS (HR=0.126 95% CI: 0.048-0.333, P <0.001) and reduced OS (HR=0.125 95% CI: 0.047-0.332, P <0.001) of nmCRC patients. The 5-year RFS of the high P-TLS, low-TLS, H-TSP, and L-TSP groups were 89.7%, 47.2%, 53.2%, and 92.5%, respectively. The P-TLS density, TSP and TNM stage were independent prognosis factors of nmCRC patients. The Nomogram, including the P-TLS density, TSP and TNM stage, outperformed the TNM stage.ConclusionsHigh P-TLS density and low TSP (L-TSP) were independent and favorable prognostic factors of nmCRC patients, which might provide new directions for targeted therapy in the CRC tumor microenvironment, especially the tumor immune microenvironment.
Collapse
Affiliation(s)
- Qianyu Wang
- The 2nd School of Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Xiaofei Shen
- Department of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Ran An
- Department of Pathology, The 7th Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Junchao Bai
- Department of General Surgery, The 7th Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Junhua Dong
- Department of General Surgery, The 7th Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Huiyun Cai
- Department of General Surgery, The 7th Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Hongyan Zhu
- Department of Pathology, The 7th Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wentao Zhong
- Department of General Surgery, The 7th Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- The 2nd School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wenliang Chen
- The 2nd School of Clinical Medicine, Shanxi Medical University, Taiyuan, China
- Department of General Surgery, The 2nd Affiliated Hospital of Shanxi Medical University, Taiyuan, China
- *Correspondence: Junfeng Du, ; Aijun Liu, ; Wenliang Chen,
| | - Aijun Liu
- Department of Pathology, The 7th Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Junfeng Du, ; Aijun Liu, ; Wenliang Chen,
| | - Junfeng Du
- Department of General Surgery, The 7th Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- The 2nd School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Medical Department of General Surgery, The 1st Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Junfeng Du, ; Aijun Liu, ; Wenliang Chen,
| |
Collapse
|
7
|
Harnessing Antitumor CD4 + T Cells for Cancer Immunotherapy. Cancers (Basel) 2022; 14:cancers14010260. [PMID: 35008422 PMCID: PMC8750687 DOI: 10.3390/cancers14010260] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/22/2021] [Accepted: 12/31/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary Diverse evidence revealed that CD4+ T cells play an important role in antitumor immunity by promoting or suppressing cytotoxic T cell responses. This review outlines the role of CD4+ T subsets within the tumor microenvironment and summarizes the latest progress regarding their potentials in cancer immunotherapy and methods for improving outcomes in cancer strategies by modulating CD4+ T responses. Abstract Over the past decades, CD4+ T cells have been considered as a supporting actor in the fields of cancer immunotherapy. Until recently, accumulating evidence has demonstrated the critical role of CD4+ T cells during antitumor immunity. CD4+ T cells can either suppress or promote the antitumor cytotoxic CD8+ T cell responses, either in secondary lymphoid organs or in the tumor. In this review, we provide an overview of the multifaceted role of different CD4+ T cell subsets in cancer immune response and their contribution during cancer therapies. Specifically, we focus on the latest progress regarding the impact of CD4+ T cell modulation on immunotherapies and other cancer therapies and discuss the prospect for harnessing CD4+ T cells to control tumor progression and prevent recurrence in patients.
Collapse
|
8
|
Garofalo C, De Marco C, Cristiani CM. NK Cells in the Tumor Microenvironment as New Potential Players Mediating Chemotherapy Effects in Metastatic Melanoma. Front Oncol 2021; 11:754541. [PMID: 34712615 PMCID: PMC8547654 DOI: 10.3389/fonc.2021.754541] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
Until the last decade, chemotherapy was the standard treatment for metastatic cutaneous melanoma, even with poor results. The introduction of immune checkpoints inhibitors (ICIs) radically changed the outcome, increasing 5-year survival from 5% to 60%. However, there is still a large portion of unresponsive patients that would need further therapies. NK cells are skin-resident innate cytotoxic lymphocytes that recognize and kill virus-infected as well as cancer cells thanks to a balance between inhibitory and activating signals delivered by surface molecules expressed by the target. Since NK cells are equipped with cytotoxic machinery but lack of antigen restriction and needing to be primed, they are nowadays gaining attention as an alternative to T cells to be exploited in immunotherapy. However, their usage suffers of the same limitations reported for T cells, that is the loss of immunogenicity by target cells and the difficulty to penetrate and be activated in the suppressive tumor microenvironment (TME). Several evidence showed that chemotherapy used in metastatic melanoma therapy possess immunomodulatory properties that may restore NK cells functions within TME. Here, we will discuss the capability of such chemotherapeutics to: i) up-regulate melanoma cells susceptibility to NK cell-mediated killing, ii) promote NK cells infiltration within TME, iii) target other immune cell subsets that affect NK cells activities. Alongside traditional systemic melanoma chemotherapy, a new pharmacological strategy based on nanocarriers loaded with chemotherapeutics is developing. The use of nanotechnologies represents a very promising approach to improve drug tolerability and effectiveness thanks to the targeted delivery of the therapeutic molecules. Here, we will also discuss the recent developments in using nanocarriers to deliver anti-cancer drugs within the melanoma microenvironment in order to improve chemotherapeutics effects. Overall, we highlight the possibility to use standard chemotherapeutics, possibly delivered by nanosystems, to enhance NK cells anti-tumor cytotoxicity. Combined with immunotherapies targeting NK cells, this may represent a valuable alternative approach to treat those patients that do not respond to current ICIs.
Collapse
Affiliation(s)
- Cinzia Garofalo
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Carmela De Marco
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Costanza Maria Cristiani
- Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
9
|
Preclinical Evaluation of LVR01 Attenuated Salmonella as Neoadjuvant Intralesional Therapy in Combination with Chemotherapy for Melanoma Treatment. J Invest Dermatol 2021; 142:1435-1443.e2. [PMID: 34662563 DOI: 10.1016/j.jid.2021.08.442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/16/2021] [Accepted: 08/27/2021] [Indexed: 11/22/2022]
Abstract
Treatment of malignant melanoma has improved in the last few years owing to early detection and new therapeutic options. Still, management of advanced disease remains a challenge because it requires systemic treatment. In such cases, dacarbazine-based chemotherapy has been widely used, despite low efficacy. Neoadjuvant therapies emerge as alternative options that could help chemotherapy to achieve increased benefit. In this work, we evaluate LVR01, an attenuated Salmonella enterica serovar typhimurium, as neoadjuvant intralesional therapy in combination with dacarbazine in a preclinical melanoma model. B16F1 melanoma‒bearing mice received intraperitoneal administration of dacarbazine for 3 consecutive days. LVR01 treatment, consisting of one single intratumoral injection, was applied 1 day before chemotherapy began. This therapeutic approach retarded tumor growth and prolonged overall survival, revealing a strong synergistic antitumor effect. Dacarbazine induced a drastic reduction of secondary lymphoid organ cellularity, which was partially restored by Salmonella, particularly potentiating activated cytotoxic cell compartments. Systemic immune reactivation could be a consequence of the intense inflammatory tumor microenvironment induced by LVR01. We propose that the use of LVR01 as neoadjuvant intralesional therapy could be considered as an interesting strategy with close clinical application to boost chemotherapy effect in patients with melanoma.
Collapse
|
10
|
Soltantoyeh T, Akbari B, Karimi A, Mahmoodi Chalbatani G, Ghahri-Saremi N, Hadjati J, Hamblin MR, Mirzaei HR. Chimeric Antigen Receptor (CAR) T Cell Therapy for Metastatic Melanoma: Challenges and Road Ahead. Cells 2021; 10:cells10061450. [PMID: 34207884 PMCID: PMC8230324 DOI: 10.3390/cells10061450] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 12/11/2022] Open
Abstract
Metastatic melanoma is the most aggressive and difficult to treat type of skin cancer, with a survival rate of less than 10%. Metastatic melanoma has conventionally been considered very difficult to treat; however, recent progress in understanding the cellular and molecular mechanisms involved in the tumorigenesis, metastasis and immune escape have led to the introduction of new therapies. These include targeted molecular therapy and novel immune-based approaches such as immune checkpoint blockade (ICB), tumor-infiltrating lymphocytes (TILs), and genetically engineered T-lymphocytes such as chimeric antigen receptor (CAR) T cells. Among these, CAR T cell therapy has recently made promising strides towards the treatment of advanced hematological and solid cancers. Although CAR T cell therapy might offer new hope for melanoma patients, it is not without its shortcomings, which include off-target toxicity, and the emergence of resistance to therapy (e.g., due to antigen loss), leading to eventual relapse. The present review will not only describe the basic steps of melanoma metastasis, but also discuss how CAR T cells could treat metastatic melanoma. We will outline specific strategies including combination approaches that could be used to overcome some limitations of CAR T cell therapy for metastatic melanoma.
Collapse
Affiliation(s)
- Tahereh Soltantoyeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
| | - Behnia Akbari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
| | - Amirali Karimi
- School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran;
| | - Ghanbar Mahmoodi Chalbatani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
| | - Navid Ghahri-Saremi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
| | - Jamshid Hadjati
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa;
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran; (T.S.); (B.A.); (G.M.C.); (N.G.-S.); (J.H.)
- Correspondence: ; Tel.: +98-21-64053268; Fax: +98-21-66419536
| |
Collapse
|
11
|
Korbecki J, Kojder K, Kapczuk P, Kupnicka P, Gawrońska-Szklarz B, Gutowska I, Chlubek D, Baranowska-Bosiacka I. The Effect of Hypoxia on the Expression of CXC Chemokines and CXC Chemokine Receptors-A Review of Literature. Int J Mol Sci 2021; 22:ijms22020843. [PMID: 33467722 PMCID: PMC7830156 DOI: 10.3390/ijms22020843] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/26/2022] Open
Abstract
Hypoxia is an integral component of the tumor microenvironment. Either as chronic or cycling hypoxia, it exerts a similar effect on cancer processes by activating hypoxia-inducible factor-1 (HIF-1) and nuclear factor (NF-κB), with cycling hypoxia showing a stronger proinflammatory influence. One of the systems affected by hypoxia is the CXC chemokine system. This paper reviews all available information on hypoxia-induced changes in the expression of all CXC chemokines (CXCL1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8 (IL-8), CXCL9, CXCL10, CXCL11, CXCL12 (SDF-1), CXCL13, CXCL14, CXCL15, CXCL16, CXCL17) as well as CXC chemokine receptors—CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, CXCR7 and CXCR8. First, we present basic information on the effect of these chemoattractant cytokines on cancer processes. We then discuss the effect of hypoxia-induced changes on CXC chemokine expression on the angiogenesis, lymphangiogenesis and recruitment of various cells to the tumor niche, including myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), regulatory T cells (Tregs) and tumor-infiltrating lymphocytes (TILs). Finally, the review summarizes data on the use of drugs targeting the CXC chemokine system in cancer therapies.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-281 Szczecin, Poland;
| | - Patrycja Kapczuk
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Barbara Gawrońska-Szklarz
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland;
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
- Correspondence: ; Tel.: +48-914661515
| |
Collapse
|
12
|
Wu B, Wang J, Wang X, Zhu M, Chen F, Shen Y, Zhong Z. CXCL5 expression in tumor tissues is associated with poor prognosis in patients with pancreatic cancer. Oncol Lett 2020; 20:257. [PMID: 32994820 PMCID: PMC7509746 DOI: 10.3892/ol.2020.12120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/25/2020] [Indexed: 02/02/2023] Open
Abstract
Immunotherapy based on the tumor microenvironment is a feasible method for treating cancer; therefore, it is necessary to investigate the immune microenvironment of pancreatic cancer and the influencing factors of the immune microenvironment. Chemokines are an important factor affecting the tumor immune microenvironment. In the present study, chemokines or chemokine receptors were screened to identify those differentially expressed in pancreatic cancer compared with normal controls and associated with patient prognosis. Chemokines or chemokine receptors that are differentially expressed in pancreatic cancer tumor tissues were initially screened using the Gene Expression Omnibus database. Next, survival analysis was performed using GEPIA, a website based on The Cancer Genome Atlas (TCGA) database. Immunohistochemical staining of CXCL5 was performed in tissue microarrays (TMAs) containing 119 cases of pancreatic cancer. Histochemistry score (H-SCORE) was used to evaluate the expression of CXCL5. Next, association analysis of the H-SCORE of CXCL5 and the clinical characteristics of patients was performed, as well as Kaplan-Meier survival and Cox multivariate regression analyses. The results of the bioinformatics analysis demonstrated that CXCL5 was highly expressed in pancreatic cancer tissues. High expression of CXCL5 in pancreatic cancer tissues was associated with a poor prognosis in patients in TCGA cohort. The expression level of CXCL5 in tumor tissues was significantly higher compared with that in adjacent peritumoral normal tissues in the immunohistochemical analysis. There was no significant association between CXCL5 expression in pancreatic cancer tumor tissues and clinicopathological factors. Patients with pancreatic cancer with high CXCL5 expression had a poor prognosis, as determined by Kaplan-Meier survival analysis based on the TMA dataset. The results of Cox multivariate regression analysis showed that CXCL5 was an independent factor for a poor prognosis in patients with pancreatic cancer. In conclusion, the results of the present study revealed that the chemokine CXCL5 was highly expressed in pancreatic cancer tissues; high CXCL5 expression was associated with a poor prognosis in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Bin Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Jing Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Xiaoguang Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Mingyuan Zhu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Fei Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Yiyu Shen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Zhengxiang Zhong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| |
Collapse
|
13
|
Mempel TR, Marangoni F. Guidance factors orchestrating regulatory T cell positioning in tissues during development, homeostasis, and response. Immunol Rev 2020; 289:129-141. [PMID: 30977195 DOI: 10.1111/imr.12761] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 12/29/2022]
Abstract
Over their lifetime, regulatory T cells (Treg) recalibrate their expression of trafficking receptors multiple times as they progress through development, respond to immune challenges, or adapt to the requirements of functioning in various non-lymphoid tissue environments. These trafficking receptors, which include chemokine receptors and other G-protein coupled receptors, integrins, as well as selectins and their ligands, enable Treg not only to enter appropriate tissues from the bloodstream via post-capillary venules, but also to navigate these tissues to locally execute their immune-regulatory functions, and finally to seek out the right antigen-presenting cells and interact with these, in part in order to receive the signals that sustain their survival, proliferation, and functional activity, in part in order to execute their immuno-regulatory function by altering antigen presenting cell function. Here, we will review our current knowledge of when and in what ways Treg alter their trafficking properties. We will focus on the chemokine system and try to identify specialized, non-redundant roles of individual receptors as well as similarities and differences to the conventional T cell compartment.
Collapse
Affiliation(s)
- Thorsten R Mempel
- The Center for Immunology and Inflammatory Diseases at Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Francesco Marangoni
- The Center for Immunology and Inflammatory Diseases at Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
14
|
Nicolas-Boluda A, Donnadieu E. Obstacles to T cell migration in the tumor microenvironment. Comp Immunol Microbiol Infect Dis 2018; 63:22-30. [PMID: 30961814 DOI: 10.1016/j.cimid.2018.12.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 11/27/2018] [Accepted: 12/20/2018] [Indexed: 12/21/2022]
Abstract
These last years, significant progress has been made in the design of strategies empowering T cells with efficient anti-tumor activities. Hence, adoptive T cell therapy and the use of monoclonal antibodies against the immunosuppressive surface molecules CTLA-4 and PD-1 appear as the most promising immunotherapies against cancer. One of the challenges ahead is to render these therapeutic interventions even more effective as a still elevated fraction of cancer patients is refractory to these treatments. A frequently overlooked determinant of the success of T cell-based immunotherapy relates to the ability of effector T cells to migrate into and within tumors, as well as to have access to tumor antigens. Here, we will focus on recent advances in understanding T cell trafficking into and within tumors. Both chemoattractant molecules and structural determinants are essential for regulating T cell motile behavior along with cellular interactions-mediated antigen recognition. In addition, we will review evidence that the microenvironment of advanced tumors creates multiple obstacles limiting T cells from migrating and making contact with their malignant targets. We will particularly focus on the extracellular matrix and tumor-associated macrophages that make tumors a hostile environment for T cell ability to contact and kill malignant cells. Finally, we will discuss possible strategies to restore a tumor microenvironment more favorable to T cell migration and functions with a special emphasis on approaches targeting the dysregulated extracellular matrix of growing tumors.
Collapse
Affiliation(s)
- Alba Nicolas-Boluda
- Inserm, U1016, Institut Cochin, Paris, France; Cnrs, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Emmanuel Donnadieu
- Inserm, U1016, Institut Cochin, Paris, France; Cnrs, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
15
|
Susek KH, Karvouni M, Alici E, Lundqvist A. The Role of CXC Chemokine Receptors 1-4 on Immune Cells in the Tumor Microenvironment. Front Immunol 2018; 9:2159. [PMID: 30319622 PMCID: PMC6167945 DOI: 10.3389/fimmu.2018.02159] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/31/2018] [Indexed: 12/14/2022] Open
Abstract
Chemokines govern leukocyte migration by attracting cells that express their cognate ligands. Many cancer types show altered chemokine secretion profiles, favoring the recruitment of pro-tumorigenic immune cells and preventing the accumulation of anti-tumorigenic effector cells. This can ultimately result in cancer immune evasion. The manipulation of chemokine and chemokine-receptor signaling can reshape the immunological phenotypes within the tumor microenvironment in order to increase the therapeutic efficacy of cancer immunotherapy. Here we discuss the three chemokine-chemokine receptor axes, CXCR1/2–CXCL1-3/5-8, CXCR3–CXCL9/10/11, and CXCR4-CXCL12 and their role on pro-tumorigenic immune cells and anti-tumorigenic effector cells in solid tumors. In particular, we summarize current strategies to target these axes and discuss their potential use in treatment approaches.
Collapse
Affiliation(s)
| | - Maria Karvouni
- Department of Medicine, Karolinska Institutet (KI), Solna, Sweden
| | - Evren Alici
- Department of Medicine, Karolinska Institutet (KI), Solna, Sweden.,Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Andreas Lundqvist
- Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL, United States.,Department of Oncology-Pathology, Karolinska Institutet (KI), Solna, Sweden
| |
Collapse
|
16
|
Hu J, Sun C, Bernatchez C, Xia X, Hwu P, Dotti G, Li S. T-cell Homing Therapy for Reducing Regulatory T Cells and Preserving Effector T-cell Function in Large Solid Tumors. Clin Cancer Res 2018; 24:2920-2934. [PMID: 29391351 PMCID: PMC6004229 DOI: 10.1158/1078-0432.ccr-17-1365] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 11/20/2017] [Accepted: 01/25/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Infused autologous tumor-infiltrating lymphocytes (TIL) and tumor-targeted chimeric antigen receptor (CAR) T cells typically surround malignant lesions or penetrate small tumor nodules but fail to penetrate large solid tumors, significantly compromising their antitumor impact. Strategies to overcome this primary challenge are largely required.Experimental Design: We tested the effects of IL12 plus doxorubicin on T-cell penetration and efficacy in solid tumors in a murine lung cancer model, a murine breast carcinoma lung metastasis model, and two human xenograft tumor models bearing large tumors (>10 mm).Results: Intriguingly, this simple approach increased the numbers, the distribution, and the depth of penetration of infused CD8+ T cells in these tumors, including both TILs and CAR T cells. This combined treatment halted tumor progression and significantly extended survival time. Studies of the underlying mechanism revealed multiple effects. First, the combined treatment maintained the high ratios of immune-stimulatory receptors to immune-inhibitory receptors on infiltrated CD8+ T cells, reduced the accumulation of immunosuppressive regulatory T cells, and enhanced the numbers of T-bet+ effector T cells in the tumors. Second, doxorubicin induced chemokines CXCL9 and CXCL10, which may attract NKG2D+CD8+ T cells to tumors, and this effect was boosted by IL12-induced IFNγ accumulation in tumors, promoting the penetration of NKG2D+CD8+ T cells.Conclusions: The deep penetration of infused T cells associated with combined IL12 plus doxorubicin yielded striking therapeutic effects in murine and human xenograft solid tumors. This approach might broaden the application of T-cell therapy to a wider range of solid tumors. Clin Cancer Res; 24(12); 2920-34. ©2018 AACRSee related commentary by Berraondo et al., p. 2716.
Collapse
Affiliation(s)
- Jiemiao Hu
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chuang Sun
- Department of Microbiology and Immunology, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| | - Chantale Bernatchez
- Department of Melanoma Medical Oncology, Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xueqing Xia
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gianpietro Dotti
- Department of Microbiology and Immunology, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| | - Shulin Li
- Department of Pediatrics-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
17
|
Xu L, Liang Z, Li S, Ma J. Signaling via the CXCR5/ERK pathway is mediated by CXCL13 in mice with breast cancer. Oncol Lett 2018; 15:9293-9298. [PMID: 29844827 DOI: 10.3892/ol.2018.8510] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/10/2018] [Indexed: 01/01/2023] Open
Abstract
Breast cancer is the most common cause of cancer-associated mortality and the most frequently diagnosed type of cancer in women worldwide. It has been revealed that the chemokine C-X-C motif chemokine ligand 13 (CXCL13) serves a pivotal role in breast cancer growth and is associated with lymph node metastasis. However, to the best of our knowledge, the mechanism by which CXCL13 mediates breast cancer growth remains uncharacterized. Female BALB/c mice were used in this study. Tumor volume was calculated and changes of gross tumor morphology were observed by hematoxylin and eosin staining. The expression of CXCL13, C-X-C motif chemokine receptor 5 (CXCR5) and extracellular signaling-related kinase (ERK) mRNA and protein expression were detected by reverse transcriptase quantitative-polymerase chain reaction and western blot analysis. Simultaneously, the production of cytokines [interleukin-1β (IL-1β), tumor necrosis factor (TNF) and tumor growth factor β1 (TGF-β1)] was detected by an ELISA. The CXCL13 inhibitor reduced tumor volume and growth, and reduced the mRNA and protein expression levels of key members of the CXCR5/ERK signaling pathway: CXCL13, CXCR5 and ERK. Furthermore, the detectable concentration of the cytokines IL-1β and TNF decreased following CXCL13 inhibition, whereas the concentration of TGF-β1 was increased. The attenuation of tumor growth resulting from CXCL13 inhibition may be associated with the CXCR5/ERK signaling pathway. This study provides a theoretical basis for treating breast cancer through CXCL13 inhibition in clinical trials.
Collapse
Affiliation(s)
- Licheng Xu
- Department of Breast Surgery, Yantaishan Hospital, Yantai, Shandong 264002, P.R. China
| | - Zhi Liang
- Department of Breast Surgery, Yantaishan Hospital, Yantai, Shandong 264002, P.R. China
| | - Shuyan Li
- Department of Breast Surgery, Yantaishan Hospital, Yantai, Shandong 264002, P.R. China
| | - Jianjun Ma
- Department of Medical Oncology, The People's Liberation Army 107th Hospital, Yantai, Shandong 264002, P.R. China
| |
Collapse
|
18
|
Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti-PD-1 treatment. Proc Natl Acad Sci U S A 2018; 115:E4041-E4050. [PMID: 29632196 PMCID: PMC5924916 DOI: 10.1073/pnas.1720948115] [Citation(s) in RCA: 613] [Impact Index Per Article: 87.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapy is a promising therapeutic intervention. However, complete and durable responses are seen in only a fraction of cancer patients. A key factor that limits therapeutic success is the lack of T cells in tumor cell regions, a profile termed “immune-excluded.” Here, we provide evidence that tumor-associated macrophages (TAMs) are an important determinant of the establishment of a T cell-excluded tumor phenotype. In human and murine tumors, we found that CD8 T cells poorly migrate and invade tumor nests due to long-lasting interactions with TAMs. The depletion of TAMs restores T cell migration and infiltration into tumor islets and improves the efficacy of anti–PD-1 immunotherapy. This study highlights the rationale of combining approaches targeting TAM and immune checkpoint proteins. In a large proportion of cancer patients, CD8 T cells are excluded from the vicinity of cancer cells. The inability of CD8 T cells to reach tumor cells is considered an important mechanism of resistance to cancer immunotherapy. We show that, in human lung squamous-cell carcinomas, exclusion of CD8 T cells from tumor islets is correlated with a poor clinical outcome and with a low lymphocyte motility, as assessed by dynamic imaging on fresh tumor slices. In the tumor stroma, macrophages mediate lymphocyte trapping by forming long-lasting interactions with CD8 T cells. Using a mouse tumor model with well-defined stromal and tumor cell areas, macrophages were depleted with PLX3397, an inhibitor of colony-stimulating factor-1 receptor (CSF-1R). Our results reveal that a CSF-1R blockade enhances CD8 T cell migration and infiltration into tumor islets. Although this treatment alone has minor effects on tumor growth, its combination with anti–PD-1 therapy further increases the accumulation of CD8 T cells in close contact with malignant cells and delays tumor progression. These data suggest that the reduction of macrophage-mediated T cell exclusion increases tumor surveillance by CD8 T cells and renders tumors more responsive to anti–PD-1 treatment.
Collapse
|
19
|
Ferrer VP, Moura Neto V, Mentlein R. Glioma infiltration and extracellular matrix: key players and modulators. Glia 2018; 66:1542-1565. [DOI: 10.1002/glia.23309] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/18/2018] [Accepted: 01/29/2018] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Rolf Mentlein
- Department of Anatomy; University of Kiel; Kiel Germany
| |
Collapse
|
20
|
Kong Z, Wang Y, Ma W. Vaccination in the immunotherapy of glioblastoma. Hum Vaccin Immunother 2018; 14:255-268. [PMID: 29087782 PMCID: PMC5806656 DOI: 10.1080/21645515.2017.1388481] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/10/2017] [Accepted: 10/02/2017] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma remains one of the most common central nervous system tumors with an extremely poor prognosis. Recently, rapid progress in immunotherapy has provided new options for the treatment of glioblastoma. Vaccination, the primary method of immunotherapy, stimulates the body's tumor-specific immune response by the injection of foreign antigens. Peptide vaccines involve the injection of tumor-specific antigens, such as EGFRvIII or heat-shock proteins. Cell-based vaccines, which primarily include dendritic cell vaccines and tumor cell vaccines, involve injections of ex vivo-modified cells. Despite the encouraging results of phase I/II clinical trials, no successful phase III clinical trials involving glioblastoma immunotherapy, including glioblastoma vaccinations, have been reported to date. In this review, the authors summarize the published outcomes of glioblastoma vaccine therapy, explore its future prospects based on ongoing clinical trials, and discuss combined therapy as a future direction for glioblastoma treatment.
Collapse
Affiliation(s)
- Ziren Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
Fujimura T, Kakizaki A, Kambayashi Y, Sato Y, Tanita K, Lyu C, Furudate S, Aiba S. Cytotoxic antimelanoma drugs suppress the activation of M2 macrophages. Exp Dermatol 2018; 27:64-70. [PMID: 28833504 DOI: 10.1111/exd.13417] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2017] [Indexed: 12/18/2022]
Abstract
Together with regulatory T cells (Tregs), tumor-associated macrophages (TAMs) play roles in maintaining the tumor microenvironment. Although cytotoxic antimelanoma drugs such as dacarbazine (DTIC), nimustine hydrochloride (ACNU) and vincristine (VCR) have been used for the treatment of malignant melanoma as adjuvant therapy in Japan, the detailed mechanisms of their immunomodulatory effects are not fully understood. As the majority of TAMs are alternatively activated M2 macrophages that favour tumor development, the aim of this study was to elucidate the immunomodulatory effects of these reagents on human monocyte-derived M2 macrophages. First, mRNA expressions and protein production of immune checkpoint molecules, PD-L1 and chemokines by CD163+ CD206+ M2 macrophages derived from peripheral blood mononuclear cells were investigated to determine the immunomodulatory effects of DTIC, ACNU, and VCR. DTIC and VCR significantly decreased PD-L1 mRNA expression, which was confirmed by flow cytometry. Moreover, the mRNA expression and production of CCL22 were significantly decreased by DTIC, which suggested that DTIC might suppress the recruitment of Tregs in the tumor site. Furthermore, the decreased expression of PD-L1 and production of CCL22 were validated in vivo, using the B16F10 mouse melanoma model, leading to abrogation of the suppressive function of T-cell proliferation. The present report suggests one of the possible antimelanoma mechanisms of DAV combination chemotherapy for melanoma patients.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antineoplastic Agents/pharmacology
- B7-H1 Antigen/metabolism
- Dacarbazine/pharmacology
- Female
- Japan
- Lectins, C-Type/metabolism
- Leukocytes, Mononuclear/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Mannose Receptor
- Mannose-Binding Lectins/metabolism
- Melanoma/drug therapy
- Melanoma, Experimental
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Monocytes/metabolism
- Nimustine/pharmacology
- RNA, Messenger/metabolism
- Receptors, Cell Surface/metabolism
- Skin Neoplasms/drug therapy
- Vincristine/pharmacology
- Melanoma, Cutaneous Malignant
Collapse
Affiliation(s)
- Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Aya Kakizaki
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yota Sato
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kayo Tanita
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chunbing Lyu
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sadanori Furudate
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
22
|
Gordon-Alonso M, Hirsch T, Wildmann C, van der Bruggen P. Galectin-3 captures interferon-gamma in the tumor matrix reducing chemokine gradient production and T-cell tumor infiltration. Nat Commun 2017; 8:793. [PMID: 28986561 PMCID: PMC5630615 DOI: 10.1038/s41467-017-00925-6] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 08/07/2017] [Indexed: 01/15/2023] Open
Abstract
The presence of T cells in tumors predicts overall survival for cancer patients. However, why most tumors are poorly infiltrated by T cells is barely understood. T-cell recruitment towards the tumor requires a chemokine gradient of the critical IFNγ-induced chemokines CXCL9/10/11. Here, we describe how tumors can abolish IFNγ-induced chemokines, thereby reducing T-cell attraction. This mechanism requires extracellular galectin-3, a lectin secreted by tumors. Galectins bind the glycans of glycoproteins and form lattices by oligomerization. We demonstrate that galectin-3 binds the glycans of the extracellular matrix and those decorating IFNγ. In mice bearing human tumors, galectin-3 reduces IFNγ diffusion through the tumor matrix. Galectin antagonists increase intratumoral IFNγ diffusion, CXCL9 gradient and tumor recruitment of adoptively transferred human CD8+ T cells specific for a tumor antigen. Transfer of T cells reduces tumor growth only if galectin antagonists are injected. Considering that most human cytokines are glycosylated, galectin secretion could be a general strategy for tumor immune evasion. Most tumours are poorly infiltrated by T cells. Here the authors show that galectin-3 secreted by tumours binds both glycosylated IFNγ and glycoproteins of the tumour extracellular matrix, thus avoiding IFNγ diffusion and the formation of an IFNγ-induced chemokine gradient required for T cell infiltration.
Collapse
Affiliation(s)
- Monica Gordon-Alonso
- Ludwig Institute for Cancer Research, de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 74, 1200, Brussels, Belgium. .,WELBIO, Avenue Hippocrate 74, 1200, Brussels, Belgium.
| | - Thibault Hirsch
- Ludwig Institute for Cancer Research, de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 74, 1200, Brussels, Belgium
| | - Claude Wildmann
- Ludwig Institute for Cancer Research, de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 74, 1200, Brussels, Belgium.,WELBIO, Avenue Hippocrate 74, 1200, Brussels, Belgium
| | - Pierre van der Bruggen
- Ludwig Institute for Cancer Research, de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 74, 1200, Brussels, Belgium. .,WELBIO, Avenue Hippocrate 74, 1200, Brussels, Belgium.
| |
Collapse
|
23
|
Vanpouille-Box C, Lhuillier C, Bezu L, Aranda F, Yamazaki T, Kepp O, Fucikova J, Spisek R, Demaria S, Formenti SC, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Immune checkpoint blockers for cancer therapy. Oncoimmunology 2017; 6:e1373237. [PMID: 29147629 DOI: 10.1080/2162402x.2017.1373237] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 08/25/2017] [Indexed: 02/08/2023] Open
Abstract
Immune checkpoint blockers (ICBs) are literally revolutionizing the clinical management of an ever more diversified panel of oncological indications. Although considerable attention persists around the inhibition of cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1, best known as PD-1) signaling, several other co-inhibitory T-cell receptors are being evaluated as potential targets for the development of novel ICBs. Moreover, substantial efforts are being devoted to the identification of biomarkers that reliably predict the likelihood of each patient to obtain clinical benefits from ICBs in the absence of severe toxicity. Tailoring the delivery of specific ICBs or combinations thereof to selected patient populations in the context of precision medicine programs constitutes indeed a major objective of the future of ICB-based immunotherapy. Here, we discuss recent preclinical and clinical advances on the development of ICBs for oncological indications.
Collapse
Affiliation(s)
| | - Claire Lhuillier
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lucillia Bezu
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Fernando Aranda
- Immunoreceptors of the Innate and Adaptive System Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Oliver Kepp
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Université Paris Descartes/Paris V, Paris, France.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
24
|
Koeck S, Kern J, Zwierzina M, Gamerith G, Lorenz E, Sopper S, Zwierzina H, Amann A. The influence of stromal cells and tumor-microenvironment-derived cytokines and chemokines on CD3 +CD8 + tumor infiltrating lymphocyte subpopulations. Oncoimmunology 2017; 6:e1323617. [PMID: 28680763 DOI: 10.1080/2162402x.2017.1323617] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 04/08/2017] [Accepted: 04/21/2017] [Indexed: 12/21/2022] Open
Abstract
The tumor microenvironment has been identified as a major mediator of immunological processes in solid tumors. In particular, tumor-associated fibroblasts are known to interact with tumor infiltrating immune cells. We describe the influence of fibroblasts and tumor-microenvironment-derived cytokines on the infiltration capacity of CD3+CD8+ cytotoxic T lymphocyte subpopulations using a multicellular 3D co-culture system. 3D tumor microtissues were cultivated using a hanging drop system. Human A549 and Calu-6 cancer cell lines were incubated alone or together with the human fibroblast cell line SV80 for 10 d to form microtissues. On day 10, peripheral blood mononuclear cells (PBMC) were added with or without cytokine stimulation for 24 h. Infiltrating PBMC subpopulations were investigated by flow cytometry. Aggregation of the microtissues and the infiltration of the PBMCs were analyzed by immunohistochemistry, and endogenous cytokine and chemokine expression was analyzed with a multi-cytokine immunoassay. Secretion of chemokines is increased in microtissues consisting of cancer cells and fibroblasts. PBMC infiltrate the whole spheroid in cancer cell monocultures, whereas in co-cultures of cancer cells and fibroblasts, PBMCs are rather localized at the margin. Activated CD69+ and CD49d+ T lymphocytes show an increased microtissue infiltration in the presence of fibroblasts. We demonstrate that the stromal component of cancer microtissues significantly influences immune cell infiltration. The presence of fibroblasts in cancer microtissues induces a shift of T lymphocyte infiltration toward activated T lymphocytes.
Collapse
Affiliation(s)
- Stefan Koeck
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Tyrol, Austria
| | - Johan Kern
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Tyrol, Austria
| | - Marit Zwierzina
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Gabriele Gamerith
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Tyrol, Austria
| | - Edith Lorenz
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Tyrol, Austria
| | - Sieghart Sopper
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Tyrol, Austria
| | - Heinz Zwierzina
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Tyrol, Austria
| | - Arno Amann
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Tyrol, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Tyrol, Austria
| |
Collapse
|
25
|
Jazayeri SD, Kuo PT, Leggatt GR, Frazer IH. HPV16-E7-Specific Activated CD8 T Cells in E7 Transgenic Skin and Skin Grafts. Front Immunol 2017; 8:524. [PMID: 28523003 PMCID: PMC5415560 DOI: 10.3389/fimmu.2017.00524] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/19/2017] [Indexed: 11/13/2022] Open
Abstract
Human papillomavirus (HPV) 16 E7 (E7) protein expression in skin promotes epithelial hyperproliferation and transformation to malignancy. Grafts of murine skin expressing E7 protein as a transgene in keratinocytes are not rejected from immunocompetent recipients, whereas grafts expressing ovalbumin (OVA), with or without coexpression of E7 protein, are promptly rejected, demonstrating that E7-associated non-antigen-specific local immunosuppression is not a major determinant of lack of rejection of E7 transgenic skin. To determine whether failure of rejection of E7 skin grafts is due to failure to attract E7-specific effector T cells, E7- and OVA-specific effector CD8+ T cells, activated in vitro, were transferred to animals bearing E7 transgenic skin grafts. Three days after T cell transfer, E7-specific T cells were present in significantly greater numbers than OVA-specific T cells in the grafted skin on animals bearing recently placed or healed E7 grafts, without graft rejection, and also in the ear skin of E7 transgenic animals, without obvious pathology. E7 and OVA-specific T cells were present in lesser numbers in healed E7 grafts than in recently placed grafts and in lesser numbers in recently placed E7 transgenic epidermal grafts without E7-associated hyperproliferation, derived from E7 transgenic mice with a mutated retinoblastoma gene. These data demonstrate that effector T cells are to some extent attracted to E7 transgenic skin specifically by E7 expression, but in large measure non-specifically by the epithelial proliferation associated with E7 expression, and by the local inflammation produced by grafting. Failure of E7 graft rejection was observed despite trafficking of E7-specific effector T cells to E7-expressing epithelium, a finding of consequence for immunotherapy of HPV 16 E7-associated human cancers.
Collapse
Affiliation(s)
| | - Paula T Kuo
- Diamantina Institute, University of Queensland, Brisbane, QLD, Australia
| | | | - Ian H Frazer
- Diamantina Institute, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
26
|
Berghoff AS, Ricken G, Wilhelm D, Rajky O, Widhalm G, Dieckmann K, Birner P, Bartsch R, Preusser M. Tumor infiltrating lymphocytes and PD-L1 expression in brain metastases of small cell lung cancer (SCLC). J Neurooncol 2016; 130:19-29. [DOI: 10.1007/s11060-016-2216-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 07/09/2016] [Indexed: 12/31/2022]
|
27
|
Ret mouse very large tumors (VLTs) display altered ratios of infiltrating memory to naive T cells: Roles in tumor expansion. ACTA ACUST UNITED AC 2016; 23:211-20. [PMID: 27397900 DOI: 10.1016/j.pathophys.2016.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 06/24/2016] [Accepted: 06/26/2016] [Indexed: 01/12/2023]
Abstract
Melanoma is an aggressive skin cancer, however it is immunogenic. The size of the primary tumor is associated with the nodal metastases. Our goals were to characterize melanoma-associated antigens (MAAs) and tumor-infiltrating T-lymphocytes (TILs) subsets in the few very large tumors (VLTs) developing in ret transgenic mice of melanoma. Tumors >700mg (VLTs) were investigated for MAAs and subsets of TILs. Immunohistochemistry and flow cytometry-based studies were performed to determine the infiltration patterns of T-lymphocytes in VLTs. It was observed that zinc fixative restores the antigenicity of the cell-surface markers of lymphocyte subpopulations without the need of antigen retrieval, whereas formalin-based fixative fails to restore the antigenicity in the presence of antigen retrieval in the immunohistochemistry. VLTs from ret mice express MAAs, such as Tyrosinase, TRP-1, TRP-2 and gp-100. The mean±standard deviation (S.D.) T-cell infiltration per 400 times-high power field in VLTs; CD4(+) (2.33±1.3), CD8(+) (2.00±1.0), and CD4(+) Foxp3(+) (2.5±0.5) regulatory T cells infiltration was exclusively restricted to the tumor stroma. Moreover, our flow cytometry-based data reveal that % mean±S.D. naive CD3(+) CD4(+) T cell infiltration (32.8±4.0%) was significantly larger than effector (25.8±2.8%, p<0.01) and central memory cells (16.1±3.7%, p<0.001) in VLTs. Similarly, between CD3(+) CD8(+) T cells, naive cells infiltrate (57.7±2.3%) in a significantly larger frequency than effector (5.0±0.4%, p<0.0001) and central memory cell (4.8±1.7%, p<0.0001) subsets. These results suggest that the VLTs from ret mice display lowered infiltration ratios between memory and naive T cells, which could be associated with the relatively large growth of VLTs.
Collapse
|
28
|
Abstract
Infectious agents are not the only agressors, and the immune system is not the sole defender of the organism. In an enlarged perspective, the ‘normative self model’ postulates that a ‘natural defense system’ protects man and other complex organisms against the environmental and internal hazards of life, including infections and cancers. It involves multiple error detection and correction mechanisms that confer robustness to the body at all levels of its organization. According to the model, the self relies on a set of physiological norms, and NONself (meaning : Non Obedient to the Norms of the self) is anything ‘off-norms’. The natural defense system comprises a set of ‘civil defenses’ (to which all cells in organs and tissues contribute), and a ‘professional army ‘, made of a smaller set of mobile cells. Mobile and non mobile cells differ in their tuning abilities. Tuning extends the recognition capabilities of NONself by the mobile cells, which increase their defensive function. To prevent them to drift, which would compromise self/NONself discrimination, the more plastic mobile cells need to periodically refer to the more stable non mobile cells to keep within physiological standards.
Collapse
Affiliation(s)
- Philippe Kourilsky
- Department of Immunology, Institut Pasteur, Paris, France; Center for Interdisciplinary Research in Biology, CNRS/UMR 7241 - INSERM U1050, Collège de France, Paris, France
| |
Collapse
|
29
|
Luo C, Wei J, Han W. Spotlight on chimeric antigen receptor engineered T cell research and clinical trials in China. SCIENCE CHINA-LIFE SCIENCES 2016; 59:349-59. [PMID: 27009301 DOI: 10.1007/s11427-016-5034-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/19/2016] [Indexed: 01/22/2023]
Abstract
T cell mediated adoptive immune response has been characterized as the key to anti-tumor immunity. Scientists around the world including in China, have been trying to harness the power of T cells against tumors for decades. Recently, the biosynthetic chimeric antigen receptor engineered T cell (CAR-T) strategy was developed and exhibited encouraging clinical efficacy, especially in hematological malignancies. Chimeric antigen receptor research reports began in 2009 in China according to our PubMed search results. Clinical trials have been ongoing in China since 2013 according to the trial registrations on clinicaltrials. gov.. After years of assiduous efforts, research and clinical scientists in China have made their own achievements in the CAR-T therapy field. In this review, we aim to highlight CAR-T research and clinical trials in China, to provide an informative reference for colleagues in the field.
Collapse
Affiliation(s)
- Can Luo
- Institute of Basic Medicine/Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jianshu Wei
- Institute of Basic Medicine/Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Weidong Han
- Institute of Basic Medicine/Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
30
|
TCR-engineered T cells to treat tumors: Seeing but not touching? Semin Immunol 2016; 28:10-21. [PMID: 26997556 DOI: 10.1016/j.smim.2016.03.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 12/17/2022]
Abstract
Adoptive transfer of T cells gene-engineered with T cell receptors (TCRs) has proven its feasibility and therapeutic potential in the treatment of malignant tumors. To ensure further clinical development of TCR gene therapy, it is necessary to accurately select TCRs that demonstrate antigen-selective responses that are restricted to tumor cells and, at the same time, include strategies that restore or enhance the entry, migration and local accumulation of T cells in tumor tissues. Here, we present the current standing of TCR-engineered T cell therapy, discuss and propose procedures to select TCRs as well as strategies to sensitize the tumor to T cell trafficking, and provide a rationale for combination therapies with TCR-engineered T cells.
Collapse
|
31
|
Bougherara H, Mansuet-Lupo A, Alifano M, Ngô C, Damotte D, Le Frère-Belda MA, Donnadieu E, Peranzoni E. Real-Time Imaging of Resident T Cells in Human Lung and Ovarian Carcinomas Reveals How Different Tumor Microenvironments Control T Lymphocyte Migration. Front Immunol 2015; 6:500. [PMID: 26528284 PMCID: PMC4600956 DOI: 10.3389/fimmu.2015.00500] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/14/2015] [Indexed: 12/30/2022] Open
Abstract
T cells play a key role in the battle against cancer. To perform their antitumor activities, T cells need to adequately respond to tumor antigens by establishing contacts with either malignant cells or antigen-presenting cells. These latter functions rely on a series of migratory steps that go from entry of T cells into the tumor followed by their locomotion in the tumor stroma. Our knowledge of how T cells migrate within tumors mainly comes from experiments performed in mouse models. Whereas such systems have greatly advanced our understanding, they do not always faithfully recapitulate the disease observed in cancer patients. We previously described a technique based on tissue slices that enables to track with real-time imaging microscopy the motile behavior of fluorescent T cells plated onto fresh sections of human lung tumors. We have now refined this approach to monitor the locomotion of resident tumor-infiltrating CD8 T cells labeled with fluorescently coupled antibodies. Using this approach, our findings reveal that CD8 T cells accumulate in the stroma of ovarian and lung carcinomas but move slowly in this compartment. Conversely, even though less populated, tumors islets were found to be zones of faster migration for resident CD8 T cells. We also confirm the key role played by collagen fibers, which, by their orientation, spacing and density, control the distribution and migration of resident CD8 T cells within the tumor stroma. We have subsequently demonstrated that, under some physical tissue constraints, CD8 T cells exhibited a mode of migration characterized by alternate forward and backward movements. In sum, using an ex vivo assay to track CD8 T cells in fresh human tumor tissues, we have identified the extracellular matrix as a major stromal component in influencing T cell migration, thereby impacting the control of tumor growth. This approach will aid in the development and testing of novel immunotherapy strategies to promote T cell migration in tumors.
Collapse
Affiliation(s)
- Houcine Bougherara
- INSERM U1016, Institut Cochin , Paris , France ; CNRS, UMR8104 , Paris , France ; Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| | - Audrey Mansuet-Lupo
- Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; Department of Pathology, Paris Centre University Hospitals, Assistance Publique-Hôpitaux de Paris , Paris , France ; Cancer and Immune Escape, Cordeliers Research Center, INSERM U1138 , Paris , France ; University Pierre and Marie Curie , Paris , France
| | - Marco Alifano
- Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; Department of Thoracic Surgery, Paris Centre University Hospitals, Assistance Publique-Hôpitaux de Paris , Paris , France
| | - Charlotte Ngô
- Department of Gynaecological and Oncological Surgery, Hôpital Européen Georges Pompidou, Université Paris Descartes, Assistance Publique-Hôpitaux de Paris , Paris , France
| | - Diane Damotte
- Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; Department of Pathology, Paris Centre University Hospitals, Assistance Publique-Hôpitaux de Paris , Paris , France ; Cancer and Immune Escape, Cordeliers Research Center, INSERM U1138 , Paris , France ; University Pierre and Marie Curie , Paris , France
| | - Marie-Aude Le Frère-Belda
- Department of Pathology, Hôpital Européen Georges Pompidou, Université Paris Descartes, Assistance Publique-Hôpitaux de Paris , Paris , France
| | - Emmanuel Donnadieu
- INSERM U1016, Institut Cochin , Paris , France ; CNRS, UMR8104 , Paris , France ; Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| | - Elisa Peranzoni
- INSERM U1016, Institut Cochin , Paris , France ; CNRS, UMR8104 , Paris , France ; Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| |
Collapse
|