1
|
Fu Q, Luo Y, Li J, Zhang P, Tang S, Song X, Fu J, Liu M, Mo R, Wei M, Li H, Liu X, Wang T, Ni G. Improving the efficacy of cancer immunotherapy by host-defence caerin 1.1 and 1.9 peptides. Hum Vaccin Immunother 2024; 20:2385654. [PMID: 39193797 PMCID: PMC11364082 DOI: 10.1080/21645515.2024.2385654] [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: 03/05/2024] [Revised: 07/07/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Cancer remains a major global health challenge. Immunotherapy has revolutionized the management of cancer, yet only a limited number of patients respond to such treatments. This is largely attributed to the immunosuppressive tumor microenvironment, which diminishes the effectiveness of immunotherapy. Recent studies have underscored the potential of naturally derived caerin 1 peptides, particularly caerin 1.1 and caerin 1.9, which exhibit strong antitumor effects and enhance the efficacy of immunotherapies in animal models. This review encapsulates the current research aimed at augmenting the effectiveness of immunotherapy, focusing on the role of caerin 1.1 and caerin 1.9 in boosting immunotherapeutic outcomes, elucidating possible mechanisms, and discussing their limitations and challenges.
Collapse
Affiliation(s)
- Quanlan Fu
- Medical School of Guizhou University, Guiyang, Guizhou, China
| | - Yuandong Luo
- Medical School of Guizhou University, Guiyang, Guizhou, China
| | - Junjie Li
- R&D Department, Zhongao Bio-pharmaceutical Technology Co., Ltd., Zhongshan, Guangdong Province, China
| | - Pingping Zhang
- Cancer Research Institute, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Shuxian Tang
- Cancer Research Institute, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Xinyi Song
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jiawei Fu
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Mengqi Liu
- Medical School of Guizhou University, Guiyang, Guizhou, China
| | - Rongmi Mo
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Ming Wei
- School of Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Hejie Li
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, Australia
| | - Xiaosong Liu
- R&D Department, Zhongao Bio-pharmaceutical Technology Co., Ltd., Zhongshan, Guangdong Province, China
- Cancer Research Institute, The First People’s Hospital of Foshan, Foshan, Guangdong, China
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Tianfang Wang
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, Australia
| | - Guoying Ni
- R&D Department, Zhongao Bio-pharmaceutical Technology Co., Ltd., Zhongshan, Guangdong Province, China
- Cancer Research Institute, The First People’s Hospital of Foshan, Foshan, Guangdong, China
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Li C, Tian Y, Zheng Y, Yuan F, Shi Z, Yang L, Chen H, Jiang L, Wang X, Zhao P, Zhang B, Wang Z, Zhao Q, Dong J, Lian C, Xu S, Zhang A, Zheng Z, Wang K, Dang C, Wu D, Chen J, Xue Y, Liang B, Cheng X, Wang Q, Chen L, Xia T, Liu H, Xu D, Zhuang J, Wu T, Zhao X, Wu W, Wang H, Peng J, Hou Z, Zheng R, Chen Y, Yin K, Zhu Z. Pathologic Response of Phase III Study: Perioperative Camrelizumab Plus Rivoceranib and Chemotherapy Versus Chemotherapy for Locally Advanced Gastric Cancer (DRAGON IV/CAP 05). J Clin Oncol 2024:JCO2400795. [PMID: 39383487 DOI: 10.1200/jco.24.00795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/01/2024] [Accepted: 09/05/2024] [Indexed: 10/11/2024] Open
Abstract
PURPOSE This multicenter, randomized phase III trial evaluated the efficacy and safety of perioperative camrelizumab (an anti-PD-1 antibody) plus low-dose rivoceranib (a VEGFR-2 inhibitor) and S-1 and oxaliplatin (SOX) (SOXRC), high-dose rivoceranib plus SOX (SOXR), and SOX alone (SOX) for locally advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma. METHODS Patients with T3-4aN + M0 G/GEJ adenocarcinoma were randomly assigned (1:1:1) to receive perioperative treatment with SOXRC, SOXR, or SOX. The primary end points were pathologic complete response (pCR) and event-free survival. The Independent Data Monitoring Committee recommended stopping enrollment in the SOXR group on the basis of the safety data of the first 103 randomly assigned patients in the three groups. The patients were then randomly assigned 1:1 to the SOXRC or SOX groups. This report presents the pCR results obtained per protocol for the first 360 randomly assigned patients who had the opportunity for surgery in the SOXRC and SOX groups. RESULTS In the SOXRC and SOX groups, of the 180 patients in each group, 99% and 98% of patients received neoadjuvant therapy, 91% and 94% completed planned neoadjuvant therapy, and 86% and 87% underwent surgery, respectively. The pCR was significantly higher in the SOXRC group at 18.3% (95% CI, 13.0 to 24.8) compared with 5.0% (95% CI, 2.3 to 9.3) in the SOX group (difference of 13.7%; 95% CI, 7.2 to 20.1; odds ratio of 4.5 [95% CI, 2.1 to 9.9]). The one-sided P value was <.0001, crossing the prespecified statistical significance threshold of P = .005. Surgical complications and grade ≥3 neoadjuvant treatment-related adverse events were 27% versus 33% and 34% versus 17% for SOXRC and SOX, respectively. CONCLUSION The SOXRC regimen significantly improved pCR compared with SOX alone in patients with G/GEJ adenocarcinoma with a tolerable safety profile.
Collapse
Affiliation(s)
- Chen Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yantao Tian
- Pancreatogastric Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Yanan Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zheng Shi
- Gastrointestinal Surgery, Shanghai Changhai Hospital, Shanghai, China
| | - Lin Yang
- Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Hao Chen
- Oncological Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Lixin Jiang
- Gastrointestinal Surgery, Yantai Yuhuangding Hospital, Yantai, China
| | - Xixun Wang
- Gastrointestinal Surgery, Yantai Yuhuangding Hospital, Yantai, China
| | - Ping Zhao
- Gastrointestinal Surgery, Sichuan Provincial Cancer Hospital, Chengdu, China
| | - Benyan Zhang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zhenqiang Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Qun Zhao
- Gastrointestinal Oncology Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianhong Dong
- Minimally Invasive Surgery of Digestive Endoscopy, Shanxi Provincial Cancer Hospital, Taiyuan, China
| | - Changhong Lian
- Gastrointestinal Surgery, HePing Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Sanrong Xu
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Aimin Zhang
- Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Zhichao Zheng
- Gastric Cancer Surgery, Liaoning Cancer Hospital & Institution, Shenyang, China
| | - Kang Wang
- Gastrointestinal Surgery, Sichuan Provincial People's Hospital, Chengdu, China
| | - Chengxue Dang
- Surgical Oncology, The First Affiliated Hospital of XI'AN Jiaotong University, Xi'an, China
| | - Dan Wu
- Gastrointestinal Surgery, The Second Affiliated Hospital Zhejiang University, School of Medicine, Hangzhou, China
| | - Jian Chen
- Gastrointestinal Surgery, The Second Affiliated Hospital Zhejiang University, School of Medicine, Hangzhou, China
| | - Yingwei Xue
- Gastrointestinal Surgery, Harbin Medical University Affiliated Cancer Hospital, Harbin, China
| | - Bo Liang
- General Surgery, Jilin Guowen Hospital, Changchun, China
| | | | - Qian Wang
- Gastrointestinal Surgery, The Affiliated Hospital of GuiZhou Medical University, Guiyang, China
| | - Luchuan Chen
- Gastrointestinal Cancer Surgery Department, Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Tao Xia
- Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Heli Liu
- Gastrointestinal Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Dazhi Xu
- Gastrosurgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jing Zhuang
- General Surgery, Henan Cancer Hospital, Zhengzhou, China
| | - Tao Wu
- General Surgery, Tangdu Hospital of the Fourth Military Medical, Xi'an, China
| | - Xi Zhao
- Colorectal Surgery, Hainan Cancer Hospital, Haikou, China
| | - Wei Wu
- Geriatric Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Hongzhi Wang
- Center of Gastrointestinal Tumor, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Junsheng Peng
- Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiguo Hou
- Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | | | - Yuting Chen
- Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | - Kai Yin
- Gastrointestinal Surgery, Shanghai Changhai Hospital, Shanghai, China
| | - Zhenggang Zhu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
4
|
Wu Z, Song Q, Liu M, Hu Y, Peng X, Zhang Z, Yao X, Peng Q. Deciphering the role of HLF in idiopathic orbital inflammation: integrative analysis via bioinformatics and machine learning techniques. Sci Rep 2024; 14:19346. [PMID: 39164324 PMCID: PMC11336107 DOI: 10.1038/s41598-024-68890-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 07/29/2024] [Indexed: 08/22/2024] Open
Abstract
Idiopathic orbital inflammation, formerly known as NSOI (nonspecific orbital inflammation), is characterized as a spectrum disorder distinguished by the polymorphic infiltration of lymphoid tissue, presenting a complex and poorly understood etiology. Recent advancements have shed light on the HLF (Human lactoferrin), proposing its critical involvement in the regulation of hematopoiesis and the maintenance of innate mucosal immunity. This revelation has generated significant interest in exploring HLF's utility as a biomarker for NSOI, despite the existing gaps in our understanding of its biosynthetic pathways and operational mechanisms. Intersecting multi-omic datasets-specifically, common differentially expressed genes between GSE58331 and GSE105149 from the Gene Expression Omnibus and immune-related gene compendiums from the ImmPort database-we employed sophisticated analytical methodologies, including Lasso regression and support vector machine-recursive feature elimination, to identify HLF. Gene set enrichment analysis and gene set variation analysis disclosed significant immune pathway enrichment within gene sets linked to HLF. The intricate relationship between HLF expression and immunological processes was further dissected through the utilization of CIBERSORT and ESTIMATE algorithms, which assess characteristics of the immune microenvironment, highlighting a noteworthy association between increased HLF expression and enhanced immune cell infiltration. The expression levels of HLF were corroborated using data from the GSE58331 dataset, reinforcing the validity of our findings. Analysis of 218 HLF-related differentially expressed genes revealed statistically significant discrepancies. Fifteen hub genes were distilled using LASSO and SVM-RFE algorithms. Biological functions connected with HLF, such as leukocyte migration, ossification, and the negative regulation of immune processes, were illuminated. Immune cell analysis depicted a positive correlation between HLF and various cells, including resting mast cells, activated NK cells, plasma cells, and CD8 T cells. Conversely, a negative association was observed with gamma delta T cells, naive B cells, M0 and M1 macrophages, and activated mast cells. Diagnostic assessments of HLF in distinguishing NSOI showed promising accuracy. Our investigation delineates HLF as intricately associated with NSOI, casting light on novel biomarkers for diagnosis and progression monitoring of this perplexing condition.
Collapse
Affiliation(s)
- Zixuan Wu
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan, China
| | - Qiujie Song
- Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, Shandong, 257091, People's Republic of China
| | - Meiling Liu
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yi Hu
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan, China
| | - Xin Peng
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan, China
| | - Zheyuan Zhang
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan, China
| | - Xiaolei Yao
- Department of Ophthalmology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, 410007, Hunan, China.
| | - Qinghua Peng
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan, China.
- Department of Ophthalmology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, 410007, Hunan, China.
| |
Collapse
|
5
|
Li S, Sun Q, Bai R, Wang Y, Wang H, Chen H, Dong Y. Real-world efficacy, safety data and predictive clinical parameters for treatment outcomes in advanced soft tissue sarcoma treated with combined immunotherapy and antiangiogenic therapy. BMC Cancer 2024; 24:1028. [PMID: 39164643 PMCID: PMC11337792 DOI: 10.1186/s12885-024-12810-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND The combination of immunotherapy and antiangiogenic therapy has shown potential in the treatment of numerous malignant tumors, but limited evidence was available for soft tissue sarcomas (STS). Therefore, the aim of the present study is to assess the efficacy and safety of immunotherapy in conjunction with antiangiogenic therapy in patients diagnosed with advanced STS (aSTS). METHODS The study enrolled patients with aSTS from January 2014 to October 2022. Eligible participants had previously received anthracycline-based chemotherapy, presented with an anthracycline-resistant sarcoma subtype, or were ineligible for anthracycline treatment due to medical conditions. Following enrollment, these patients received a combination of immunotherapy and antiangiogenic therapy. The primary endpoints were the objective response rate (ORR) and progression-free survival (PFS), while the secondary endpoints included the disease control rate (DCR), overall survival (OS), and the incidence of adverse events. RESULTS Fifty-one patients were included in this cohort study. The median duration of follow-up was 15.8 months. The ORR and DCR were 17.6%, and 76.5%, respectively. The median PFS (mPFS) was 5.8 months (95% CI: 4.8-6.8) for all patients, and the median OS had not been reached as of the date cutoff. Multivariate analysis indicated that Eastern Cooperative Oncology Group performance status of 0-1 and ≤ second-line treatment were positive predictors for both PFS and OS. Patients with alveolar soft part sarcoma or clear cell sarcoma had longer mPFS (16.2 months, 95% CI: 7.8-25.6) when compared to those with other subtypes of STS (4.4 months, 95% CI: 1.4-7.5, P < 0.001). Among the observed adverse events, hypertension (23.5%), diarrhea (17.6%), and proteinuria (17.6%) were the most common, with no treatment-related deaths reported. CONCLUSION The combination of immunotherapy and antiangiogenic agents showed promising efficacy and acceptable toxicity in patients with aSTS, especially those with alveolar soft part sarcoma or clear cell sarcoma.
Collapse
Affiliation(s)
- Shaoli Li
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Qunan Sun
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Rui Bai
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Youping Wang
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310009, China
| | - Hui Wang
- Department of Pathology, Institute of Basic Medicine and Cancer (IBMC), the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Haifeng Chen
- Department of Medical Oncology, Shaoxing Second Hospital, Shaoxing, 312000, China
| | - Ying Dong
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Cancer Center, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
6
|
Datta M, Via LE, Dartois V, Xu L, Barry CE, Jain RK. Leveraging insights from cancer to improve tuberculosis therapy. Trends Mol Med 2024:S1471-4914(24)00205-3. [PMID: 39142973 DOI: 10.1016/j.molmed.2024.07.011] [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: 06/18/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 08/16/2024]
Abstract
Exploring and exploiting the microenvironmental similarities between pulmonary tuberculosis (TB) granulomas and malignant tumors has revealed new strategies for more efficacious host-directed therapies (HDTs). This opinion article discusses a paradigm shift in TB therapeutic development, drawing on critical insights from oncology. We summarize recent efforts to characterize and overcome key shared features between tumors and granulomas, including excessive fibrosis, abnormal angiogenesis, hypoxia and necrosis, and immunosuppression. We provide specific examples of cancer therapy application to TB to overcome these microenvironmental abnormalities, including matrix-targeting therapies, antiangiogenic agents, and immune-stimulatory drugs. Finally, we propose a new framework for combining HDTs with anti-TB agents to maximize therapeutic delivery and efficacy while reducing treatment dosages, duration, and harmful side effects to benefit TB patients.
Collapse
Affiliation(s)
- Meenal Datta
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Laura E Via
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA; Hackensack Meridian School of Medicine, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Lei Xu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
7
|
Yuan Y, Mishra F, Li B, Peng G, Chan P, Yang J, Liu Z. Modulating Tumor Immunity by Targeting Tumor Fibrotic Stroma and Angiogenic Vessels for Lung Cancer Treatment. Cancers (Basel) 2024; 16:2483. [PMID: 39001545 PMCID: PMC11240634 DOI: 10.3390/cancers16132483] [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: 06/02/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Fibrotic stroma and angiogenic tumor vessels play an important role in modulating tumor immunity. We previously reported a rationally designed protein (ProAgio) that targets integrin αvβ3 at a novel site. ProAgio induces the apoptosis of cells that express high levels of the integrin. Both activated cancer-associated fibroblasts (CAFs) and angiogenic endothelial cells (aECs) in tumors express high levels of integrin αvβ3. ProAgio simultaneously and specifically induces apoptosis in CAFs and aECs in tumors. We provide evidence here that the depletion of CAFs and the elimination of leaky tumor angiogenic vessels by ProAgio alter tumor immunity. ProAgio reduces CD4+ Treg and Myeloid-derived suppressor cells (MDSCs), increases CD8+ T-cells, and increases the M1/M2 macrophage ratio in the tumor. The depletion of dense fibrotic stroma (CAFs) by ProAgio decreases the Programmed Death Ligand 1 (PDL-1) levels in the stroma areas surrounding the tumors, and thus strongly increases the delivery of anti-PDL-1 antibody to the target cancer cells. The impact of ProAgio on tumor immunity provides strong synergistical effects of checkpoint inhibitors on lung cancer treatment.
Collapse
Affiliation(s)
- Yi Yuan
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Falguni Mishra
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Bin Li
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Guangda Peng
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Payton Chan
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Jenny Yang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA;
| | - Zhiren Liu
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| |
Collapse
|
8
|
Wu Z, Xu J, Hu Y, Peng X, Zhang Z, Yao X, Peng Q. The roles of IRF8 in nonspecific orbital inflammation: an integrated analysis by bioinformatics and machine learning. J Ophthalmic Inflamm Infect 2024; 14:29. [PMID: 38900395 PMCID: PMC11190126 DOI: 10.1186/s12348-024-00410-4] [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: 12/09/2023] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Nonspecific Orbital Inflammation (NSOI) represents a persistent and idiopathic proliferative inflammatory disorder, characterized by polymorphous lymphoid infiltration within the orbit. The transcription factor Interferon Regulatory Factor 8 (IRF8), integral to the IRF protein family, was initially identified as a pivotal element for the commitment and differentiation of myeloid cell lineage. Serving as a central regulator of innate immune receptor signaling, IRF8 orchestrates a myriad of functions in hematopoietic cell development. However, the intricate mechanisms underlying IRF8 production remain to be elucidated, and its potential role as a biomarker for NSOI is yet to be resolved. METHODS IRF8 was extracted from the intersection analysis of common DEGs of GSE58331 and GSE105149 from the GEO and immune- related gene lists in the ImmPort database using The Lasso regression and SVM-RFE analysis. We performed GSEA and GSVA with gene sets coexpressed with IRF8, and observed that gene sets positively related to IRF8 were enriched in immune-related pathways. To further explore the correlation between IRF8 and immune-related biological process, the CIBERSORT algorithm and ESTIMATE method were employed to evaluate TME characteristics of each sample and confirmed that high IRF8 expression might give rise to high immune cell infiltration. Finally, the GSE58331 was utilized to confirm the levels of expression of IRF8. RESULTS Among the 314 differentially expressed genes (DEGs), some DEGs were found to be significantly different. With LASSO and SVM-RFE algorithms, we obtained 15 hub genes. For biological function analysis in IRF8, leukocyte mediated immunity, leukocyte cell-cell adhesion, negative regulation of immune system process were emphasized. B cells naive, Macrophages M0, Macrophages M1, T cells CD4 memory activated, T cells CD4 memory resting, T cells CD4 naive, and T cells gamma delta were shown to be positively associated with IRF8. While, Mast cells resting, Monocytes, NK cells activated, Plasma cells, T cells CD8, and T cells regulatory (Tregs) were shown to be negatively linked with IRF8. The diagnostic ability of the IRF8 in differentiating NSOI exhibited a good value. CONCLUSIONS This study discovered IRF8 that are linked to NSOI. IRF8 shed light on potential new biomarkers for NSOI and tracking its progression.
Collapse
Affiliation(s)
- Zixuan Wu
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Jinfeng Xu
- Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, Shandong, 257091, PR China
| | - Yi Hu
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Xin Peng
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Zheyuan Zhang
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Xiaolei Yao
- Department of Ophthalmology, the First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, 410007, Hunan Province, China
- Ophthalmology Department, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410011, China
| | - Qinghua Peng
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, Hunan Province, China.
- Department of Ophthalmology, the First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, 410007, Hunan Province, China.
| |
Collapse
|
9
|
Li C, Li Z, Sun Q, Xiang Y, Liu A. Severe cutaneous adverse reactions associated with immune checkpoint inhibitors therapy and anti-VEGF combination therapy: a real-world study of the FDA adverse event reporting system. Expert Opin Drug Saf 2024; 23:777-784. [PMID: 37622438 DOI: 10.1080/14740338.2023.2251381] [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: 05/06/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) therapy combined with anti-vascular endothelial growth factor (anti-VEGF) regimens showed new hope for cancer patients and considered as future pillar of cancer therapy. However, severe cutaneous adverse reactions (SCARs) in patients with ICIs and anti-VEGF combined therapy raise a serious concern and remain thoroughly assessed in clinics. RESEARCH DESIGN AND METHODS Data retrieved from the first quarter of 2004 to the third quarter of 2022 in FAERS database underwent disproportionality analysis and Bayesian analysis were utilized to detect and assess the SCAR signals of ICIs and ICIs and anti-VEGF combined therapy for comparison. RESULTS In total, 854 (1.10%) and 80 (1.06%) reports on SCARs associated with ICIs and a combination of ICIs and anti-VEGF therapy, respectively, were analyzed. Most of SCARs reports were associated with the use of pembrolizumab (36.01%), nivolumab (23.97%) and a combination of ipilimumab and nivolumab (19.71%). A use of atezolizumab and bevacizumab combined therapy (60.00%) caused the most SCARs records out of ICIs and anti-VEGF combined therapies. CONCLUSIONS Treatment with joint therapy of ICIs and anti-VEGF agents may cause severe cutaneous adverse events. It is vital to identify ICI-related SCARs early, and to manage them appropriately.
Collapse
Affiliation(s)
- Chunlei Li
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, China
| | - Zhengjun Li
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
| | - Qing Sun
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
| | - Yanxiao Xiang
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, China
| | - Anchang Liu
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, China
- Department of Pharmacy, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
10
|
Datta M, Via LE, Dartois V, Weiner DM, Zimmerman M, Kaya F, Walker AM, Fleegle JD, Raplee ID, McNinch C, Zarodniuk M, Kamoun WS, Yue C, Kumar AS, Subudhi S, Xu L, Barry CE, Jain RK. Normalizing granuloma vasculature and matrix improves drug delivery and reduces bacterial burden in tuberculosis-infected rabbits. Proc Natl Acad Sci U S A 2024; 121:e2321336121. [PMID: 38530888 PMCID: PMC10998582 DOI: 10.1073/pnas.2321336121] [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: 12/04/2023] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
Host-directed therapies (HDTs) represent an emerging approach for bacterial clearance during tuberculosis (TB) infection. While most HDTs are designed and implemented for immuno-modulation, other host targets-such as nonimmune stromal components found in pulmonary granulomas-may prove equally viable. Building on our previous work characterizing and normalizing the aberrant granuloma-associated vasculature, here we demonstrate that FDA-approved therapies (bevacizumab and losartan, respectively) can be repurposed as HDTs to normalize blood vessels and extracellular matrix (ECM), improve drug delivery, and reduce bacterial loads in TB granulomas. Granulomas feature an overabundance of ECM and compressed blood vessels, both of which are effectively reduced by losartan treatment in the rabbit model of TB. Combining both HDTs promotes secretion of proinflammatory cytokines and improves anti-TB drug delivery. Finally, alone and in combination with second-line antitubercular agents (moxifloxacin or bedaquiline), these HDTs significantly reduce bacterial burden. RNA sequencing analysis of HDT-treated lung and granuloma tissues implicates up-regulated antimicrobial peptide and proinflammatory gene expression by ciliated epithelial airway cells as a putative mechanism of the observed antitubercular benefits in the absence of chemotherapy. These findings demonstrate that bevacizumab and losartan are well-tolerated stroma-targeting HDTs, normalize the granuloma microenvironment, and improve TB outcomes, providing the rationale to clinically test this combination in TB patients.
Collapse
Affiliation(s)
- Meenal Datta
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN46556
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Laura E. Via
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD20892
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
- Hackensack Meridian School of Medicine, Hackensack Meridian Health, Nutley, NJ07110
| | - Danielle M. Weiner
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD20892
| | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | - Firat Kaya
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | - April M. Walker
- Tuberculosis Imaging Program, Division of Intramural Research, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD20892
| | - Joel D. Fleegle
- Tuberculosis Imaging Program, Division of Intramural Research, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD20892
| | - Isaac D. Raplee
- Bioinformatics and Computational Bioscience Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Colton McNinch
- Bioinformatics and Computational Bioscience Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Maksym Zarodniuk
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN46556
| | - Walid S. Kamoun
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Changli Yue
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN46556
| | - Ashwin S. Kumar
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Sonu Subudhi
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Lei Xu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Clifton E. Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD20892
| | - Rakesh K. Jain
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| |
Collapse
|
11
|
Lee IH, Wang HY, Chen YY, Chen CY, Liao HF. Synergistic B and T lymphocyte interaction: prognostic implications in non-small cell lung cancer. Am J Cancer Res 2024; 14:1227-1242. [PMID: 38590414 PMCID: PMC10998741 DOI: 10.62347/tdiv2436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
While T-cell-mediated immune responses in solid tumors have been well-established and have driven major therapeutic advances, our understanding of B-cell biology in cancer is comparatively less developed. A total of 60 lung cancer patients were included, of which 53% were diagnosed at an early stage while 47% were diagnosed at an advanced stage. Flow cytometry was used to analyze the proportion of T and B cells in all blood samples, and the levels of human serum cytokines were also assessed. Compared to the control group, cancer patients showed lower frequencies of IgD+CD27+ marginal B cells and CD32+ B cells, and higher frequencies of T cells with lower CD8+ T cells and higher central memory and naïve CD4+ T cells. Additionally, advanced-stage cancer patients exhibited higher levels of cytokines, a higher proportion of effector memory CD8+ T cells, and a lower frequency of CD27+CD28+CD4+/CD8+ T cells. Linear regression analysis revealed significant correlations between cancer stage and the frequency of B and T cell subsets, leukocyte count, and cytokine levels. Survival analysis demonstrated that patients with higher frequency of class-switched B cells had a worse prognosis, while patients with higher frequency of CD8+ effector T cells and lower frequency of CD4+57+ T cells appeared to have a better survival rate. These findings provide valuable insight into the immunological changes that occur during lung cancer progression and have the potential to inform the development of new immunotherapeutic strategies.
Collapse
Affiliation(s)
- I-Hsien Lee
- Department of Internal Medicine, Fu Jen Catholic University HospitalNew Taipei, Taiwan
| | - Hsin-Yi Wang
- Department of Internal Medicine, National Taiwan University Hospital Yunlin BranchYunlin, Taiwan
- College of Medicine, National Taiwan UniversityTaipei, Taiwan
| | - Ying-Yin Chen
- Department of Internal Medicine, National Taiwan University Hospital Yunlin BranchYunlin, Taiwan
- College of Medicine, National Taiwan UniversityTaipei, Taiwan
| | - Chung-Yu Chen
- Department of Internal Medicine, National Taiwan University Hospital Yunlin BranchYunlin, Taiwan
- College of Medicine, National Taiwan UniversityTaipei, Taiwan
| | - Hui-Fen Liao
- Department of Biochemical Science and Technology, National Chiayi UniversityChiayi, Taiwan
| |
Collapse
|
12
|
Yang F, Akhtar MN, Zhang D, El-Mayta R, Shin J, Dorsey JF, Zhang L, Xu X, Guo W, Bagley SJ, Fuchs SY, Koumenis C, Lathia JD, Mitchell MJ, Gong Y, Fan Y. An immunosuppressive vascular niche drives macrophage polarization and immunotherapy resistance in glioblastoma. SCIENCE ADVANCES 2024; 10:eadj4678. [PMID: 38416830 PMCID: PMC10901371 DOI: 10.1126/sciadv.adj4678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/25/2024] [Indexed: 03/01/2024]
Abstract
Cancer immunity is subjected to spatiotemporal regulation by leukocyte interaction with neoplastic and stromal cells, contributing to immune evasion and immunotherapy resistance. Here, we identify a distinct mesenchymal-like population of endothelial cells (ECs) that form an immunosuppressive vascular niche in glioblastoma (GBM). We reveal a spatially restricted, Twist1/SATB1-mediated sequential transcriptional activation mechanism, through which tumor ECs produce osteopontin to promote immunosuppressive macrophage (Mφ) phenotypes. Genetic or pharmacological ablation of Twist1 reverses Mφ-mediated immunosuppression and enhances T cell infiltration and activation, leading to reduced GBM growth and extended mouse survival, and sensitizing tumor to chimeric antigen receptor T immunotherapy. Thus, these findings uncover a spatially restricted mechanism controlling tumor immunity and suggest that targeting endothelial Twist1 may offer attractive opportunities for optimizing cancer immunotherapy.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Md Naushad Akhtar
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, PA 19104, USA
| | - Junyoung Shin
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jay F. Dorsey
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen J. Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justin D. Lathia
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, PA 19104, USA
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Clark GC, Lai A, Agarwal A, Liu Z, Wang XY. Biopterin metabolism and nitric oxide recoupling in cancer. Front Oncol 2024; 13:1321326. [PMID: 38469569 PMCID: PMC10925643 DOI: 10.3389/fonc.2023.1321326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/26/2023] [Indexed: 03/13/2024] Open
Abstract
Tetrahydrobiopterin is a cofactor necessary for the activity of several enzymes, the most studied of which is nitric oxide synthase. The role of this cofactor-enzyme relationship in vascular biology is well established. Recently, tetrahydrobiopterin metabolism has received increasing attention in the field of cancer immunology and immunotherapy due to its involvement in the cytotoxic T cell response. Past research has demonstrated that when the availability of BH4 is low, as it is in chronic inflammatory conditions and tumors, electron transfer in the active site of nitric oxide synthase becomes uncoupled from the oxidation of arginine. This results in the production of radical species that are capable of a direct attack on tetrahydrobiopterin, further depleting its local availability. This feedforward loop may act like a molecular switch, reinforcing low tetrahydrobiopterin levels leading to altered NO signaling, restrained immune effector activity, and perpetual vascular inflammation within the tumor microenvironment. In this review, we discuss the evidence for this underappreciated mechanism in different aspects of tumor progression and therapeutic responses. Furthermore, we discuss the preclinical evidence supporting a clinical role for tetrahydrobiopterin supplementation to enhance immunotherapy and radiotherapy for solid tumors and the potential safety concerns.
Collapse
Affiliation(s)
- Gene Chatman Clark
- Department of Biochemistry, Virginia Commonwealth University, Richmond, VA, United States
- School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Alan Lai
- School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | | | - Zheng Liu
- Department of Human Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Xiang-Yang Wang
- Department of Human Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
14
|
Burchett A, Siri S, Li J, Lu X, Datta M. Novel 3-D macrophage spheroid model reveals reciprocal regulation of immunomechanical stress and mechano-immunological response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580327. [PMID: 38405787 PMCID: PMC10888788 DOI: 10.1101/2024.02.14.580327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Purpose In many diseases, an overabundance of macrophages contributes to adverse outcomes. While numerous studies have compared macrophage phenotype after mechanical stimulation or with varying local stiffness, it is unclear if and how macrophages themselves contribute to mechanical forces in their microenvironment. Methods Raw 264.7 murine macrophages were embedded in a confining agarose gel, where they proliferated to form spheroids over time. Gels were synthesized at various concentrations to tune the stiffness and treated with various growth supplements to promote macrophage polarization. The spheroids were then analyzed by immunofluorescent staining and qPCR for markers of proliferation, mechanosensory channels, and polarization. Finally, spheroid geometries were used to computationally model the strain generated in the agarose by macrophage spheroid growth. Results Macrophages form spheroids and generate growth-induced mechanical forces (i.e., solid stress) within confining agarose gels, which can be maintained for at least 16 days in culture. Increasing agarose concentration restricts spheroid expansion, promotes discoid geometries, limits gel deformation, and induces an increase in iNOS expression. LPS stimulation increases spheroid growth, though this effect is reversed with the addition of IFN-γ. Ki67 expression decreases with increasing agarose concentration, in line with the growth measurements. Conclusions Macrophages alone both respond to and generate solid stress. Understanding how macrophage generation of growth-induced solid stress responds to different environmental conditions will help to inform treatment strategies for the plethora of diseases that involve macrophage accumulation.
Collapse
Affiliation(s)
- Alice Burchett
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, IN, USA
| | - Saeed Siri
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, IN, USA
| | - Jun Li
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, IN, USA
| | - Xin Lu
- Department of Biological Sciences, University of Notre Dame, IN, USA
| | - Meenal Datta
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, IN, USA
| |
Collapse
|
15
|
Jiang C, Sun L, Wen S, Tian Y, Xu C, Xu Q, Xue H. BRIX1 promotes ribosome synthesis and enhances glycolysis by selected translation of GLUT1 in colorectal cancer. J Gene Med 2024; 26:e3632. [PMID: 38282151 DOI: 10.1002/jgm.3632] [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: 04/21/2023] [Revised: 09/25/2023] [Accepted: 10/28/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Ribosome biogenesis protein BRX1 homolog (BRIX1) is critically required for the synthesis of the 60S ribosome subunit. However, the role and mechanism of BRIX1 in colorectal cancer (CRC) remain unclear. METHODS Kyoto Encyclopedia of Gene and Genome pathway and Gene Ontology analyses were used for bioinformatics analysis. The rRNA levels were detected in CRC tissues and cells. Nascent RNA synthesis was detected via cellular immunofluorescence. The correlation was analyzed between patient Positron Emission Tomography-Computed Tomography (PET-CT) values and their BRIX1 expression. The extracellular acidification rate (ECAR) and oxygen consumption rate were determined via live metabolic analyses. Polysome fractions were collected for BRIX1 mRNA used in translation. The orthotopic model and Cell Counting Kit-8 (CCK8) assay were used to assess BRIX1 function in CRC. RESULTS BRIX1 is a core protein involved in ribosome-related pathway changes in CRC. Gene Ontology analysis showed that BRIX1 was primarily enriched in ribosome assembly and ribosome biogenesis pathways. In fresh CRC tissue, rRNA levels (5S, 5.8S, 18S and 28S) were higher in the BRIX1 high-expression group than in the BRIX1 low-expression group. Similarly, BRIX1 knockdown significantly decreased rRNA levels for 5S, 5.8S, 18S and 28S in CRC cells, whereas overexpression of BRIX1 significantly increased these levels. In addition, BRIX1 knockdown inhibited nascent RNA synthesis in CRC cells. In clinical data analysis, BRIX1 expression was related to the glucose uptake in PET-CT. BRIX1 knockdown significantly decreased the ECAR value, glucose uptake and lactic acid production in CRC cells, whereas BRIX1 overexpression significantly increased these. Furthermore, BRIX1 knockdown significantly decreased the protein expression of GLUT1, whereas BRIX1 overexpression significantly increased this; however, expression of BRIX1 mRNA was unaffected in either case. Blocking glycolysis by si-GLUT1 or galactose reversed BRIX1 promotion of glycolysis and cell proliferation in CRC cells.
Collapse
Affiliation(s)
- Chunhui Jiang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Longci Sun
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Siyuan Wen
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Tian
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chunjie Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hanbing Xue
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
16
|
Savchenko IV, Zlotnikov ID, Kudryashova EV. Biomimetic Systems Involving Macrophages and Their Potential for Targeted Drug Delivery. Biomimetics (Basel) 2023; 8:543. [PMID: 37999184 PMCID: PMC10669405 DOI: 10.3390/biomimetics8070543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/10/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
The concept of targeted drug delivery can be described in terms of the drug systems' ability to mimic the biological objects' property to localize to target cells or tissues. For example, drug delivery systems based on red blood cells or mimicking some of their useful features, such as long circulation in stealth mode, have been known for decades. On the contrary, therapeutic strategies based on macrophages have gained very limited attention until recently. Here, we review two biomimetic strategies associated with macrophages that can be used to develop new therapeutic modalities: first, the mimicry of certain types of macrophages (i.e., the use of macrophages, including tumor-associated or macrophage-derived particles as a carrier for the targeted delivery of therapeutic agents); second, the mimicry of ligands, naturally absorbed by macrophages (i.e., the use of therapeutic agents specifically targeted at macrophages). We discuss the potential applications of biomimetic systems involving macrophages for new advancements in the treatment of infections, inflammatory diseases, and cancer.
Collapse
Affiliation(s)
| | | | - Elena V. Kudryashova
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia (I.D.Z.)
| |
Collapse
|
17
|
Gu X, Zhu Y, Zhao C, Cao Y, Wang J, Zhang Q, Li L. TNFSF15 facilitates the differentiation of CD11b + myeloid cells into vascular pericytes in tumors. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0245. [PMID: 37921408 PMCID: PMC10690882 DOI: 10.20892/j.issn.2095-3941.2023.0245] [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: 07/11/2023] [Accepted: 09/13/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVE Immature vasculature lacking pericyte coverage substantially contributes to tumor growth, drug resistance, and cancer cell dissemination. We previously demonstrated that tumor necrosis factor superfamily 15 (TNFSF15) is a cytokine with important roles in modulating hematopoiesis and vascular homeostasis. The main purpose of this study was to explore whether TNFSF15 might promote freshly isolated myeloid cells to differentiate into CD11b+ cells and further into pericytes. METHODS A model of Lewis lung cancer was established in mice with red fluorescent bone marrow. After TNFSF15 treatment, CD11b+ myeloid cells and vascular pericytes in the tumors, and the co-localization of pericytes and vascular endothelial cells, were assessed. Additionally, CD11b+ cells were isolated from wild-type mice and treated with TNFSF15 to determine the effects on the differentiation of these cells. RESULTS We observed elevated percentages of bone marrow-derived CD11b+ myeloid cells and vascular pericytes in TNFSF15-treated tumors, and the latter cells co-localized with vascular endothelial cells. TNFSF15 protected against CD11b+ cell apoptosis and facilitated the differentiation of these cells into pericytes by down-regulating Wnt3a-VEGFR1 and up-regulating CD49e-FN signaling pathways. CONCLUSIONS TNFSF15 facilitates the production of CD11b+ cells in the bone marrow and promotes the differentiation of these cells into pericytes, which may stabilize the tumor neovasculature.
Collapse
Affiliation(s)
- Xiangxiang Gu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Yipan Zhu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Cancan Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Yixin Cao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Jingying Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Qiangzhe Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Luyuan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| |
Collapse
|
18
|
Desai S. Influence of pathogens on host genome and epigenome in development of head and neck cancer. Cancer Rep (Hoboken) 2023; 6:e1846. [PMID: 37322598 PMCID: PMC10644332 DOI: 10.1002/cnr2.1846] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/11/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Head and neck cancer (HNSCC) is a heterogeneous group of cancers, affecting multiple regions such as oral cavity, pharynx, larynx, and nasal region, each showing a distinct molecular profile. HNSCC accounts for more than 6 million cases worldwide, soaring mainly in the developing countries. RECENT FINDINGS The aetiology of HNSCC is complex and multifactorial, involving both genetic and environmental factors. The critical role of microbiome, which includes bacteria, viruses, and fungi, is under spotlight due to the recent reports on their contribution in the development and progression of HNSCC. This review focuses on the effect of opportunistic pathogens on the host genome and epigenome, which contributes to the disease progression. Drawing parallels from the host-pathogen interactions observed in other tumour types arising from the epithelial tissue such as colorectal cancer, the review also calls attention to the potential explorations of the role of pathogens in HNSCC biology and discusses the clinical implications of microbiome research in detection and treatment of HNSCC. CONCLUSION Our understanding of the genomic effects of the microbes on the disease progression and the mechanistic insights of the host-pathogen interaction will pave way to novel treatment and preventive approaches in HNSCC.
Collapse
|
19
|
Wang L, Li Z, Li Z, Ren Y, Qian L, Yu Y, Shi W, Xiong Y. Identification of A Novel Gene Signature Combining Ferroptosis- and Immunity-Related Genes for Prognostic Prediction, Immunotherapy and Potential Therapeutic Targets in Gastric Cancer. J Cancer 2023; 14:3457-3476. [PMID: 38021154 PMCID: PMC10647194 DOI: 10.7150/jca.87223] [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: 06/16/2023] [Accepted: 09/17/2023] [Indexed: 12/01/2023] Open
Abstract
Gastric cancer (GC) is one of the most prevalent cancers worldwide. Ferroptosis and the immune status of tumor tissue play vital roles in the initiation and progression of GC. However, the role and functional mechanisms of ferroptosis- and immunity-related genes (FIRGs) in GC pathogenesis and their correlations with GC prognosis have not been elucidated. We aim to establish a prognostic prediction model based on the FIRGs signature for GC patients. Differentially expressed genes were screened from the Cancer Genome Atlas (TCGA) GC cohorts. The least absolute shrinkage and selection operator (LASSO) regression was performed to establish a FIRGs-based risk model. This gene signature with 7 FIRGs was identified as an independent prognostic factor. A nomogram incorporating clinical parameters and the FIRG signature was constructed to individualize outcome predictions. Finally, we provided in vivo and in vitro evidence to verify the reliability of FIRG signature for GC prognosis, and validate the expression and function of FIRGs contributing to the development and progression of GC. Herein, our work represents great therapeutic and prognostic potentials for GC.
Collapse
Affiliation(s)
- Liwei Wang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Zhuozhuo Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Zi Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Yuanyuan Ren
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Department of Endocrinology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Wenzhen Shi
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Medical Research Center, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
| | - Yuyan Xiong
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| |
Collapse
|
20
|
Qin Z, Huang Y, Li Z, Pan G, Zheng L, Xiao X, Wang F, Chen J, Chen X, Lin X, Li K, Yan G, Zhang H, Xing F. Glioblastoma Vascular Plasticity Limits Effector T-cell Infiltration and Is Blocked by cAMP Activation. Cancer Immunol Res 2023; 11:1351-1366. [PMID: 37540804 DOI: 10.1158/2326-6066.cir-22-0872] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/20/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023]
Abstract
Glioblastoma (GBM) is the deadliest form of brain cancer. It is a highly angiogenic and immunosuppressive malignancy. Although immune checkpoint blockade therapies have revolutionized treatment for many types of cancer, their therapeutic efficacy in GBM has been far less than expected or even ineffective. In this study, we found that the genomic signature of glioma-derived endothelial cells (GdEC) correlates with an immunosuppressive state and poor prognosis of patients with glioma. We established an in vitro model of GdEC differentiation for drug screening and used this to determine that cyclic adenosine monophosphate (cAMP) activators could effectively block GdEC formation by inducing oxidative stress. Furthermore, cAMP activators impaired GdEC differentiation in vivo, normalized the tumor vessels, and altered the tumor immune profile, especially increasing the influx and function of CD8+ effector T cells. Dual blockade of GdECs and PD-1 induced tumor regression and established antitumor immune memory. Thus, our study reveals that endothelial transdifferentiation of GBM shapes an endothelial immune cell barrier and supports the clinical development of combining GdEC blockade and immunotherapy for GBM. See related Spotlight by Lee et al., p. 1300.
Collapse
Affiliation(s)
- Zixi Qin
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Youwei Huang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, P.R. China
| | - Zeying Li
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Guopeng Pan
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Liangying Zheng
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, P.R. China
| | - Xiao Xiao
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, P.R. China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Fang Wang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Jiahong Chen
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Xueqin Chen
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Xi Lin
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Kai Li
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Guangmei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, P.R. China
| | - Haipeng Zhang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Fan Xing
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, P.R. China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
21
|
Konstantinopoulos PA, Matulonis UA. Clinical and translational advances in ovarian cancer therapy. NATURE CANCER 2023; 4:1239-1257. [PMID: 37653142 DOI: 10.1038/s43018-023-00617-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/17/2023] [Indexed: 09/02/2023]
Abstract
Ovarian cancer is an aggressive disease that is frequently detected at advanced stages and is initially very responsive to platinum-based chemotherapy. However, the majority of patients relapse following initial surgery and chemotherapy, highlighting the urgent need to develop new therapeutic strategies. In this Review, we outline the main therapeutic principles behind the management of newly diagnosed and recurrent epithelial ovarian cancer and discuss the current landscape of targeted and immune-based approaches.
Collapse
|
22
|
Sun XX, Nosrati Z, Ko J, Lee CM, Bennewith KL, Bally MB. Induced Vascular Normalization-Can One Force Tumors to Surrender to a Better Microenvironment? Pharmaceutics 2023; 15:2022. [PMID: 37631236 PMCID: PMC10458586 DOI: 10.3390/pharmaceutics15082022] [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: 05/03/2023] [Revised: 06/19/2023] [Accepted: 07/14/2023] [Indexed: 08/27/2023] Open
Abstract
Immunotherapy has changed the way many cancers are being treated. Researchers in the field of immunotherapy and tumor immunology are investigating similar questions: How can the positive benefits achieved with immunotherapies be enhanced? Can this be achieved through combinations with other agents and if so, which ones? In our view, there is an urgent need to improve immunotherapy to make further gains in the overall survival for those patients that should benefit from immunotherapy. While numerous different approaches are being considered, our team believes that drug delivery methods along with appropriately selected small-molecule drugs and drug candidates could help reach the goal of doubling the overall survival rate that is seen in some patients that are given immunotherapeutics. This review article is prepared to address how immunotherapies should be combined with a second treatment using an approach that could realize therapeutic gains 10 years from now. For context, an overview of immunotherapy and cancer angiogenesis is provided. The major targets in angiogenesis that have modulatory effects on the tumor microenvironment and immune cells are highlighted. A combination approach that, for us, has the greatest potential for success involves treatments that will normalize the tumor's blood vessel structure and alter the immune microenvironment to support the action of immunotherapeutics. So, this is reviewed as well. Our focus is to provide an insight into some strategies that will engender vascular normalization that may be better than previously described approaches. The potential for drug delivery systems to promote tumor blood vessel normalization is considered.
Collapse
Affiliation(s)
- Xu Xin Sun
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- NanoMedicines Innovation Network, Vancouver, BC V6T 1Z3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
| | - Zeynab Nosrati
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
| | - Janell Ko
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
| | - Che-Min Lee
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Kevin L. Bennewith
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marcel B. Bally
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- NanoMedicines Innovation Network, Vancouver, BC V6T 1Z3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
23
|
Hu G, Xiao Y, Ma C, Wang J, Qian X, Wu X, Zhu F, Sun S, Qian J. Lumican is a potential predictor on the efficacy of concurrent chemoradiotherapy in cervical squamous cell carcinoma. Heliyon 2023; 9:e18011. [PMID: 37483824 PMCID: PMC10362307 DOI: 10.1016/j.heliyon.2023.e18011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/25/2023] [Accepted: 07/05/2023] [Indexed: 07/25/2023] Open
Abstract
Purpose To identify new novel biomarkers for predicting the efficacy of concurrent chemoradiotherapy(CCRT) in cervical squamous cell carcinoma(CESC). Methods Gene expression datasets GSE56363, GSE5787, and GSE168009 were analyzed to identify candidate genes to predict the efficacy of CCRT in CESC. Single-cell RNA sequencing (scRNA-seq) data from GSE168652 and CESC patients in The Cancer Genome Atlas(TCGA) were systematically analyzed to explore possible molecular mechanisms. Kaplan-Meier evaluated the correlation between LUM (Lumican) and prognostic significance. The expression of LUM protein in biopsy tissues before CCRT was detected by immunohistochemistry in 15 CESC patients. Results LUM mRNA levels were significantly upregulated in nonresponders of CESC.patients receiving CCRT and positively correlated with poor therapeutic effect. Furthermore, high expression of LUM influenced the immune microenvironment in CESC patient-derived organoids treated with CCRT. LUM overexpression in CESC cells induced resistance to CCRT, potentially via immune landscape modulation. Gene Set Enrichment Analysis (GSEA) revealed that possible mechanisms underlying resistance to CCRT might involve the PARs and IL1 signaling pathway affecting the immune landscape. Conclusions High LUM expression is correlated with poor efficacy in CESC patients receiving CCRT, possibly through the PARs and IL1 signaling pathway affecting the immune landscape.
Collapse
Affiliation(s)
- Ge Hu
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei,230031, PR China
| | - Ying Xiao
- The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, PR China
| | - Chanchan Ma
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230031, PR China
| | - Jinyun Wang
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei,230031, PR China
| | - Xiaotao Qian
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei,230031, PR China
| | - Xiaowei Wu
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei,230031, PR China
| | - Fengqin Zhu
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei,230031, PR China
| | - Shiying Sun
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230031, PR China
| | - Junchao Qian
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei,230031, PR China
| |
Collapse
|
24
|
Suzuki T, Takei J, Fukasawa N, Suzuki K, Ogawa D, Yamamoto Y, Akasaki Y, Murayama Y, Shimoda M, Miyake K, Tanaka T. 18F-Fluoromisonidazole-Positron Emission Tomography and Immunohistochemistry Verified Tumor Oxygenation, Stemness, and Immunosupportive Microenvironment After Preoperative Neoadjuvant Bevacizumab for Newly Diagnosed Glioblastoma. World Neurosurg 2023; 175:e1364-e1374. [PMID: 37187346 DOI: 10.1016/j.wneu.2023.05.030] [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: 03/14/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND Cancer stemness and immunosuppressive tumor microenvironment (TME) in accordance with tumor oxygenation are variable during bevacizumab (Bev) therapy for glioblastoma (GBM). Positron emission tomography (PET) using 18F-fluoromisonidazole (FMISO) reflects hypoxic TME. The aim of this study was to compare FMISO-PET and immunohistochemical findings of tumor oxygenation in the TME of GBM during Bev treatment. METHODS Seven patients with newly diagnosed IDH-wildtype GBM underwent FMISO-PET during follow-up. Three patients received preoperative neoadjuvant Bev (neo-Bev) and subsequently underwent surgical resection. Reoperation was performed at the recurrence. FMISO-PET was performed before and after neo-Bev. Four patients who underwent tumor resection without neo-Bev were included as the control group. Expressions of hypoxic markers (carbonic anhydrase; CA9), stem cell markers (nestin, FOXM1), and immunoregulatory molecules (CD163, FOXP3, PD-L1) in tumor tissues were analyzed by immunohistochemistry (IHC). RESULTS All 3 patients treated with neo-Bev showed decrease in FMISO accumulation in accordance with expressions of CA9 and FOXM1 compared with the control group. Two of these 3 patients at the recurrence showed increase in FMISO accumulation. IHC showed increased CA9-and FOXM1-positive cells in recurrent tumors. Expression of PD-L1 tended to be lower after neo-Bev compared with the control group. CONCLUSIONS FMISO-PET effectively visualized TME oxygenation after neo-Bev. Increased FMISO accumulation at the time of recurrence, even under Bev treatment, suggests that FMISO-PET might be useful for monitoring the duration of Bev efficacy by reflecting tumor oxygenation.
Collapse
Affiliation(s)
- Tomoya Suzuki
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| | - Jun Takei
- Department of Neurosurgery, Jikei University School of Medicine, Katsushika Medical Center, Tokyo, Japan
| | - Nei Fukasawa
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Kenta Suzuki
- Department of Neurosurgery, Kagawa Graduate University School of Medicine, Kagawa, Japan
| | - Daisuke Ogawa
- Department of Neurosurgery, Kagawa Graduate University School of Medicine, Kagawa, Japan
| | - Yohei Yamamoto
- Department of Neurosurgery, Jikei University School of Medicine, Daisan Hospital, Tokyo, Japan
| | - Yasuharu Akasaki
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| | - Yuichi Murayama
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Keisuke Miyake
- Department of Neurosurgery, Kagawa Graduate University School of Medicine, Kagawa, Japan
| | - Toshihide Tanaka
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
25
|
He K, Berz D, Gadgeel SM, Iams WT, Bruno DS, Blakely CM, Spira AI, Patel MR, Waterhouse DM, Richards DA, Pham A, Jotte R, Hong DS, Garon EB, Traynor A, Olson P, Latven L, Yan X, Shazer R, Leal TA. MRTX-500 Phase 2 Trial: Sitravatinib With Nivolumab in Patients With Nonsquamous NSCLC Progressing On or After Checkpoint Inhibitor Therapy or Chemotherapy. J Thorac Oncol 2023; 18:907-921. [PMID: 36842467 PMCID: PMC10330304 DOI: 10.1016/j.jtho.2023.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 02/28/2023]
Abstract
INTRODUCTION Sitravatinib, a receptor tyrosine kinase inhibitor targeting TYRO3, AXL, MERTK receptors, and vascular epithelial growth factor receptor 2, can shift the tumor microenvironment toward an immunostimulatory state. Combining sitravatinib with checkpoint inhibitors (CPIs) may augment antitumor activity. METHODS The phase 2 MRTX-500 study evaluated sitravatinib (120 mg daily) with nivolumab (every 2 or 4 wk) in patients with advanced nonsquamous NSCLC who progressed on or after previous CPI (CPI-experienced) or chemotherapy (CPI-naive). CPI-experienced patients had a previous clinical benefit (PCB) (complete response, partial response, or stable disease for at least 12 weeks then disease progression) or no PCB (NPCB) from CPI. The primary end point was objective response rate (ORR); secondary objectives included safety and secondary efficacy end points. RESULTS Overall, 124 CPI-experienced (NPCB, n = 35; PCB, n = 89) and 32 CPI-naive patients were treated. Investigator-assessed ORR was 11.4% in patients with NPCB, 16.9% with PCB, and 25.0% in CPI-naive. The median progression-free survival was 3.7, 5.6, and 7.1 months with NPCB, PCB, and CPI-naive, respectively; the median overall survival was 7.9 and 13.6 months with NPCB and PCB, respectively (not reached in CPI-naive patients; median follow-up 20.4 mo). Overall, (N = 156), any grade treatment-related adverse events (TRAEs) occurred in 93.6%; grade 3/4 in 58.3%. One grade 5 TRAE occurred in a CPI-naive patient. TRAEs led to treatment discontinuation in 14.1% and dose reduction or interruption in 42.9%. Biomarker analyses supported an immunostimulatory mechanism of action. CONCLUSIONS Sitravatinib with nivolumab had a manageable safety profile. Although ORR was not met, this combination exhibited antitumor activity and encouraged survival in CPI-experienced patients with nonsquamous NSCLC.
Collapse
Affiliation(s)
- Kai He
- Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, Ohio.
| | - David Berz
- Department of Cellular Therapeutics, Beverly Hills Cancer Center, Beverly Hills, California; Current Affiliation: Valkyrie Clinical Trials, Los Angeles, California
| | - Shirish M Gadgeel
- Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan
| | - Wade T Iams
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee
| | - Debora S Bruno
- University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Collin M Blakely
- Department of Medicine, University of California San Francisco, San Francisco, California; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Alexander I Spira
- Virginia Cancer Specialists, Fairfax, Virginia; US Oncology Network, The Woodlands, Texas
| | - Manish R Patel
- Division Of Hematology, Oncology and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota
| | - David M Waterhouse
- US Oncology Network, The Woodlands, Texas; Department of Clinical Research, Oncology Hematology Care, Cincinnati, Ohio; Current affiliation: Dana-Farber/Brigham and Women's Cancer Center at Milford Regional Medical Center, Milford, Massachusetts
| | - Donald A Richards
- US Oncology Network, The Woodlands, Texas; Texas Oncology, Tyler, Texas
| | | | - Robert Jotte
- US Oncology Network, The Woodlands, Texas; Rocky Mountain Cancer Centers, Denver, Colorado
| | - David S Hong
- MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Edward B Garon
- Department Of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Anne Traynor
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Peter Olson
- Mirati Therapeutics, Inc., San Diego, California
| | - Lisa Latven
- Mirati Therapeutics, Inc., San Diego, California
| | - Xiaohong Yan
- Mirati Therapeutics, Inc., San Diego, California
| | | | - Ticiana A Leal
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin; Current Affiliation: Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
26
|
Roy S, Fitzgerald K, Lalani A, Lai CW, Kim A, Kim J, Ou P, Mirsoian A, Liu X, Ramrakhiani A, Zhao H, Zhou H, Xu H, Meisen H, Li CM, Lugt BV, Thibault S, Tinberg CE, DeVoss J, Egen J, Wu LC, Noubade R. Autonomous IL-36R signaling in neutrophils activates potent antitumor effector functions. J Clin Invest 2023; 133:e162088. [PMID: 37317970 DOI: 10.1172/jci162088] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 04/19/2023] [Indexed: 06/16/2023] Open
Abstract
While the rapid advancement of immunotherapies has revolutionized cancer treatment, only a small fraction of patients derive clinical benefit. Eradication of large, established tumors appears to depend on engaging and activating both innate and adaptive immune system components to mount a rigorous and comprehensive immune response. Identifying such agents is a high unmet medical need, because they are sparse in the therapeutic landscape of cancer treatment. Here, we report that IL-36 cytokine can engage both innate and adaptive immunity to remodel an immune-suppressive tumor microenvironment (TME) and mediate potent antitumor immune responses via signaling in host hematopoietic cells. Mechanistically, IL-36 signaling modulates neutrophils in a cell-intrinsic manner to greatly enhance not only their ability to directly kill tumor cells but also promote T and NK cell responses. Thus, while poor prognostic outcomes are typically associated with neutrophil enrichment in the TME, our results highlight the pleiotropic effects of IL-36 and its therapeutic potential to modify tumor-infiltrating neutrophils into potent effector cells and engage both the innate and adaptive immune system to achieve durable antitumor responses in solid tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Haoda Xu
- Therapeutic Discovery, Amgen, South San Francisco, California, USA
| | | | | | | | - Steve Thibault
- Therapeutic Discovery, Amgen, South San Francisco, California, USA
| | | | | | | | | | | |
Collapse
|
27
|
Bruni S, Mercogliano MF, Mauro FL, Cordo Russo RI, Schillaci R. Cancer immune exclusion: breaking the barricade for a successful immunotherapy. Front Oncol 2023; 13:1135456. [PMID: 37284199 PMCID: PMC10239871 DOI: 10.3389/fonc.2023.1135456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/10/2023] [Indexed: 06/08/2023] Open
Abstract
Immunotherapy has changed the course of cancer treatment. The initial steps were made through tumor-specific antibodies that guided the setup of an antitumor immune response. A new and successful generation of antibodies are designed to target immune checkpoint molecules aimed to reinvigorate the antitumor immune response. The cellular counterpart is the adoptive cell therapy, where specific immune cells are expanded or engineered to target cancer cells. In all cases, the key for achieving positive clinical resolutions rests upon the access of immune cells to the tumor. In this review, we focus on how the tumor microenvironment architecture, including stromal cells, immunosuppressive cells and extracellular matrix, protects tumor cells from an immune attack leading to immunotherapy resistance, and on the available strategies to tackle immune evasion.
Collapse
|
28
|
Shi X, Shu L, Wang M, Yao J, Yao Q, Bian S, Chen X, Wan J, Zhang F, Zheng S, Wang H. Triple-Combination Immunogenic Nanovesicles Reshape the Tumor Microenvironment to Potentiate Chemo-Immunotherapy in Preclinical Cancer Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204890. [PMID: 37017572 PMCID: PMC10214259 DOI: 10.1002/advs.202204890] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 02/22/2023] [Indexed: 05/27/2023]
Abstract
Immune checkpoint blockade (ICB) therapies have had a tremendous impact on cancer therapy. However, most patients harbor a poorly immunogenic tumor microenvironment (TME), presenting overwhelming de novo refractoriness to ICB inhibitors. To address these challenges, combinatorial regimens that employ chemotherapies and immunostimulatory agents are urgently needed. Here, a combination chemoimmunotherapeutic nanosystem consisting of a polymeric monoconjugated gemcitabine (GEM) prodrug nanoparticle decorated with an anti-programmed cell death-ligand 1 (PD-L1) antibody (αPD-L1) on the surface and a stimulator of interferon genes (STING) agonist encapsulated inside is developed. Treatment with GEM nanoparticles upregulates PD-L1 expression in ICB-refractory tumors, resulting in augmented intratumor drug delivery in vivo and synergistic antitumor efficacy via activation of intratumor CD8+ T cell responses. Integration of a STING agonist into the αPD-L1-decorated GEM nanoparticles further improves response rates by transforming low-immunogenic tumors into inflamed tumors. Systemically administered triple-combination nanovesicles induce robust antitumor immunity, resulting in durable regression of established large tumors and a reduction in the metastatic burden, coincident with immunological memory against tumor rechallenge in multiple murine tumor models. These findings provide a design rationale for synchronizing STING agonists, PD-L1 antibodies, and chemotherapeutic prodrugs to generate a chemoimmunotherapeutic effect in treating ICB-nonresponsive tumors.
Collapse
Affiliation(s)
- Xiaowei Shi
- The First Affiliated HospitalNational Health Commission (NHC) Key Laboratory of Combined Multi‐Organ TransplantationZhejiang University School of MedicineHangzhouZhejiang Province310003P. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandong Province250117P. R. China
| | - Liwei Shu
- Department of Medical OncologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiang Province310016P. R. China
| | - Minwen Wang
- Department of Medical OncologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiang Province310016P. R. China
| | - Jie Yao
- The First Affiliated HospitalNational Health Commission (NHC) Key Laboratory of Combined Multi‐Organ TransplantationZhejiang University School of MedicineHangzhouZhejiang Province310003P. R. China
- Department of Chemical EngineeringZhejiang UniversityHangzhou310027P. R. China
| | - Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003P. R. China
- National Clinical Research Center for Infectious DiseasesHangzhou310003P. R. China
| | - Suchen Bian
- The First Affiliated HospitalNational Health Commission (NHC) Key Laboratory of Combined Multi‐Organ TransplantationZhejiang University School of MedicineHangzhouZhejiang Province310003P. R. China
| | - Xiaona Chen
- The First Affiliated HospitalNational Health Commission (NHC) Key Laboratory of Combined Multi‐Organ TransplantationZhejiang University School of MedicineHangzhouZhejiang Province310003P. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandong Province250117P. R. China
| | - Jianqin Wan
- The First Affiliated HospitalNational Health Commission (NHC) Key Laboratory of Combined Multi‐Organ TransplantationZhejiang University School of MedicineHangzhouZhejiang Province310003P. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandong Province250117P. R. China
| | - Fu Zhang
- The First Affiliated HospitalNational Health Commission (NHC) Key Laboratory of Combined Multi‐Organ TransplantationZhejiang University School of MedicineHangzhouZhejiang Province310003P. R. China
| | - Shusen Zheng
- The First Affiliated HospitalNational Health Commission (NHC) Key Laboratory of Combined Multi‐Organ TransplantationZhejiang University School of MedicineHangzhouZhejiang Province310003P. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandong Province250117P. R. China
| | - Hangxiang Wang
- The First Affiliated HospitalNational Health Commission (NHC) Key Laboratory of Combined Multi‐Organ TransplantationZhejiang University School of MedicineHangzhouZhejiang Province310003P. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandong Province250117P. R. China
| |
Collapse
|
29
|
Li L, Liang Y, Yu M, Zhao L, Mei Q, Yu Y, Wang N, Zhang D, Wang Z, Jia Y, Kong F. Advances in immune checkpoint inhibitors therapy for small cell lung cancer. Cancer Med 2023; 12:11097-11106. [PMID: 36880420 PMCID: PMC10242320 DOI: 10.1002/cam4.5659] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/09/2023] [Accepted: 01/17/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND As one of the most aggressive neuroendocrine tumors, small cell lung cancer (SCLC) has the most disappointing prognosis of all lung cancers. Although SCLC responds well to initial chemotherapy, the majority of patients experience disease recurrence within one year, and patient survival is poor. It is still necessary to explore the application of ICIs in SCLC since the beginning of the road to immunotherapy, which broke the 30-year treatment deadlock of SCLC. METHODS We searched PubMed, Web of Science, and Embase with search terms such as "SCLC", "ES-SCLC", "ICIs", and "ICBs", and categorized and summarized the relevant literature obtained, and we compiled the latest progress about the application of ICIs in SCLC. RESULTS We listed 14 clinical trials on ICIs, including 8 clinical trials on first-line SCLC treatment, 2 clinical trials on second-line SCLC treatment, 3 clinical trials on third-line SCLC treatment, and 1 clinical trial on SCLC maintenance treatment. CONCLUSION ICIs in combination with chemotherapy can improve OS in SCLC patients, but the extent to which SCLC patients can benefit from ICIs is limited, and ICIs' combination treatment strategies still need to be continuously explored.
Collapse
Affiliation(s)
- Longhui Li
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Yangyueying Liang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Minghui Yu
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Lu Zhao
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Qingyun Mei
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Yongchao Yu
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Na Wang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Dou Zhang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Ziwei Wang
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Yingjie Jia
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Fanming Kong
- Department of OncologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| |
Collapse
|
30
|
Konishi K, Kuwahara H, Morita D, Imai S, Nagata K. Prolonged Survival in a Patient With Extensive-Stage Small Cell Lung Cancer in Spite of Discontinued Immunotherapy With Atezolizumab. Cureus 2023; 15:e37757. [PMID: 37213944 PMCID: PMC10198584 DOI: 10.7759/cureus.37757] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2023] [Indexed: 05/23/2023] Open
Abstract
A 64-year-old man was referred from a local clinic with a chief complaint of cough. Computed tomography (CT) revealed a mass comprising a tumor in the right lower lobe and enlarged mediastinal lymph nodes, and a whole-body workup with positron emission tomography-CT showed bilateral lymph node enlargement and cancerous pericarditis. Biopsy with bronchoscopy of the right lower lobe tumor and mediastinal lymph node confirmed the histological findings of small cell lung carcinoma. The clinical diagnosis of extensive-stage small cell lung cancer (ES-SCLC) was confirmed, and first-line treatment with carboplatin, etoposide, and atezolizumab was initiated, followed by tri-weekly atezolizumab thrice. The patient experienced worsening pleural effusion treated with thoracentesis, pleural drainage, and pleurodesis. He also experienced several recurrences, which were managed with second and third-line chemotherapy with nogitecan and amrubicin. He has been receiving third-line therapy for over 30 months since his initial visit and remains stable as of today. The patient experienced an exceptional treatment outcome considering that the prognosis of ES-SCLC remains poor, with a median survival of approximately 10 months with conventional chemotherapies using cytotoxic agents. The use of immune checkpoint inhibitors (ICI) for ES-SCLC as first-line treatment may demonstrate a persistent antitumor effect, and result in improved survival following discontinuation. In conclusion, therapy including ICI for patients with ES-SCLC is a treatment option that shows possibilities in improving survival even after discontinuation.
Collapse
Affiliation(s)
- Kazuhisa Konishi
- Department of Respiratory Medicine, Koseikai Takeda Hospital, Kyoto, JPN
| | - Hiroomi Kuwahara
- Department of Respiratory Medicine, Koseikai Takeda Hospital, Kyoto, JPN
| | - Daiki Morita
- Department of Respiratory Medicine, Koseikai Takeda Hospital, Kyoto, JPN
| | - Shunsuke Imai
- Department of Pathology, Koseikai Takeda Hospital, Kyoto, JPN
| | - Kazuhiro Nagata
- Department of Respiratory Medicine, Koseikai Takeda Hospital, Kyoto, JPN
| |
Collapse
|
31
|
Zhang D, Li AM, Hu G, Huang M, Yang F, Zhang L, Wellen KE, Xu X, Conn CS, Zou W, Kahn M, Rhoades SD, Weljie AM, Fuchs SY, Amankulor N, Yoshor D, Ye J, Koumenis C, Gong Y, Fan Y. PHGDH-mediated endothelial metabolism drives glioblastoma resistance to chimeric antigen receptor T cell immunotherapy. Cell Metab 2023; 35:517-534.e8. [PMID: 36804058 PMCID: PMC10088869 DOI: 10.1016/j.cmet.2023.01.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 10/24/2022] [Accepted: 01/26/2023] [Indexed: 02/19/2023]
Abstract
The efficacy of immunotherapy is limited by the paucity of T cells delivered and infiltrated into the tumors through aberrant tumor vasculature. Here, we report that phosphoglycerate dehydrogenase (PHGDH)-mediated endothelial cell (EC) metabolism fuels the formation of a hypoxic and immune-hostile vascular microenvironment, driving glioblastoma (GBM) resistance to chimeric antigen receptor (CAR)-T cell immunotherapy. Our metabolome and transcriptome analyses of human and mouse GBM tumors identify that PHGDH expression and serine metabolism are preferentially altered in tumor ECs. Tumor microenvironmental cues induce ATF4-mediated PHGDH expression in ECs, triggering a redox-dependent mechanism that regulates endothelial glycolysis and leads to EC overgrowth. Genetic PHGDH ablation in ECs prunes over-sprouting vasculature, abrogates intratumoral hypoxia, and improves T cell infiltration into the tumors. PHGDH inhibition activates anti-tumor T cell immunity and sensitizes GBM to CAR T therapy. Thus, reprogramming endothelial metabolism by targeting PHGDH may offer a unique opportunity to improve T cell-based immunotherapy.
Collapse
Affiliation(s)
- Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Albert M Li
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Guanghui Hu
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Menggui Huang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Crystal S Conn
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Zou
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark Kahn
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seth D Rhoades
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nduka Amankulor
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Yoshor
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Cheema AK, Li Y, Ventimiglia M, Kowalczyk K, Hankins R, Bandi G, Janowski EM, Grindrod S, Villagra A, Dritschilo A. Radiotherapy Induces Innate Immune Responses in Patients Treated for Prostate Cancers. Clin Cancer Res 2023; 29:921-929. [PMID: 36508164 PMCID: PMC9975665 DOI: 10.1158/1078-0432.ccr-22-2340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/03/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE Radiotherapy is a curative therapeutic modality used to treat cancers as a single agent or in combination with surgery and chemotherapy. Advanced radiotherapy technologies enable treatment with large fractions and highly conformal radiation doses to effect free-radical damage to cellular DNA leading to cell-cycle arrest, cell death, and innate immune response (IIR) stimulation. EXPERIMENTAL DESIGN To understand systemic clinical responses after radiation exposure, proteomic and metabolomic analyses were performed on plasma obtained from patients with cancer at intervals after prostate stereotactic body radiotherapy. Pathway and multivariate analyses were used to delineate molecular alterations following radiotherapy and its correlation with clinical outcomes. RESULTS DNA damage response increased within the first hour after treatment and returned to baseline by 1 month. IIR signaling also increased within 1 hour of treatment but persisted for up to 3 months thereafter. Furthermore, robust IIR and metabolite elevations, consistent with an early proinflammatory M1-mediated innate immune activation, were observed in patients in remission, whereas patients experiencing prostate serum antigen-determined disease progression demonstrated less robust immune responses and M2-mediated metabolite elevations. CONCLUSIONS To our knowledge, these data are the first report of longitudinal proteomic and metabolomic molecular responses in patients after radiotherapy for cancers. The data supports innate immune activation as a critical clinical response of patients receiving radiotherapy for prostate cancer. Furthermore, we propose that the observed IIR may be generalized to the treatment of other cancer types, potentially informing multidisciplinary therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Amrita K. Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington DC
- Corresponding Author: Amrita K. Cheema, GC2, Pre-clinical Science Building, 3900 Reservoir Road NW, Washington DC 20007. Phone: 202-687-2756; E-mail:
| | - Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC
| | - Mary Ventimiglia
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC
| | - Keith Kowalczyk
- Department of Radiation Medicine, LL Bles, MedStar-Georgetown University Hospital, Washington DC
| | - Ryan Hankins
- Department of Radiation Medicine, LL Bles, MedStar-Georgetown University Hospital, Washington DC
| | - Gaurav Bandi
- Department of Radiation Medicine, LL Bles, MedStar-Georgetown University Hospital, Washington DC
| | - Einsley-Marie Janowski
- Department of Radiation Oncology, University of Virginia School of Medicine, Charlottesville, Virginia
| | | | - Alejandro Villagra
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC
| | - Anatoly Dritschilo
- Department of Radiation Medicine, LL Bles, MedStar-Georgetown University Hospital, Washington DC
| |
Collapse
|
33
|
Wan J, Ren L, Li X, He S, Fu Y, Xu P, Meng F, Xian S, Pu K, Wang H. Photoactivatable nanoagonists chemically programmed for pharmacokinetic tuning and in situ cancer vaccination. Proc Natl Acad Sci U S A 2023; 120:e2210385120. [PMID: 36787350 PMCID: PMC9974508 DOI: 10.1073/pnas.2210385120] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 01/17/2023] [Indexed: 02/15/2023] Open
Abstract
Immunotherapy holds great promise for the treatment of aggressive and metastatic cancers; however, currently available immunotherapeutics, such as immune checkpoint blockade, benefit only a small subset of patients. A photoactivatable toll-like receptor 7/8 (TLR7/8) nanoagonist (PNA) system that imparts near-infrared (NIR) light-induced immunogenic cell death (ICD) in dying tumor cells in synchrony with the spontaneous release of a potent immunoadjuvant is developed here. The PNA consists of polymer-derived proimmunoadjuvants ligated via a reactive oxygen species (ROS)-cleavable linker and polymer-derived photosensitizers, which are further encapsulated in amphiphilic matrices for systemic injection. In particular, conjugation of the TLR7/8 agonist resiquimod to biodegradable macromolecular moieties with different molecular weights enabled pharmacokinetic tuning of small-molecule agonists and optimized delivery efficiency in mice. Upon NIR photoirradiation, PNA effectively generated ROS not only to ablate tumors and induce the ICD cascade but also to trigger the on-demand release of TLR agonists. In several preclinical cancer models, intravenous PNA administration followed by NIR tumor irradiation resulted in remarkable tumor regression and suppressed postsurgical tumor recurrence and metastasis. Furthermore, this treatment profoundly shifted the tumor immune landscape to a tumoricidal one, eliciting robust tumor-specific T cell priming in vivo. This work highlights a simple and cost-effective approach to generate in situ cancer vaccines for synergistic photodynamic immunotherapy of metastatic cancers.
Collapse
Affiliation(s)
- Jianqin Wan
- The First Affiliated Hospital, National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, Zhejiang University School of Medicine, Hangzhou310003, P. R. China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan250117, P. R. China
| | - Lulu Ren
- The First Affiliated Hospital, National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, Zhejiang University School of Medicine, Hangzhou310003, P. R. China
| | - Xiaoyan Li
- The First Affiliated Hospital, National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, Zhejiang University School of Medicine, Hangzhou310003, P. R. China
- Department of Chemical Engineering, Zhejiang University, Hangzhou310027, P. R. China
| | - Shasha He
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore637457, Singapore
| | - Yang Fu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou310016, P. R. China
| | - Peirong Xu
- The First Affiliated Hospital, National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, Zhejiang University School of Medicine, Hangzhou310003, P. R. China
- Department of Chemical Engineering, Zhejiang University, Hangzhou310027, P. R. China
| | - Fanchao Meng
- The First Affiliated Hospital, National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, Zhejiang University School of Medicine, Hangzhou310003, P. R. China
| | - Shiyun Xian
- The First Affiliated Hospital, National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, Zhejiang University School of Medicine, Hangzhou310003, P. R. China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore637457, Singapore
| | - Hangxiang Wang
- The First Affiliated Hospital, National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, Zhejiang University School of Medicine, Hangzhou310003, P. R. China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan250117, P. R. China
| |
Collapse
|
34
|
Yang F, Zhang D, Jiang H, Ye J, Zhang L, Bagley SJ, Winkler J, Gong Y, Fan Y. Small-molecule toosendanin reverses macrophage-mediated immunosuppression to overcome glioblastoma resistance to immunotherapy. Sci Transl Med 2023; 15:eabq3558. [PMID: 36791206 PMCID: PMC10394757 DOI: 10.1126/scitranslmed.abq3558] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 01/17/2023] [Indexed: 02/17/2023]
Abstract
T cell-based immunotherapy holds promise for treating solid tumors, but its therapeutic efficacy is limited by intratumoral immune suppression. This immune suppressive tumor microenvironment is largely driven by tumor-associated myeloid cells, including macrophages. Here, we report that toosendanin (TSN), a small-molecule compound, reprograms macrophages to enforce antitumor immunity in glioblastoma (GBM) in mouse models. Our functional screen of genetically probed macrophages with a chemical library identifies that TSN reverses macrophage-mediated tumor immunosuppression, leading to enhanced T cell infiltration, activation, and reduced exhaustion. Chemoproteomic and structural analyses revealed that TSN interacts with Hck and Lyn to abrogate suppressive macrophage immunity. In addition, a combination of immune checkpoint blockade and TSN therapy induced regression of syngeneic GBM tumors in mice. Furthermore, TSN treatment sensitized GBM to Egfrviii chimeric antigen receptor (CAR) T cell therapy. These findings suggest that TSN may serve as a therapeutic compound that blocks tumor immunosuppression and circumvents tumor resistance to T cell-based immunotherapy in GBM and other solid tumors that warrants further investigation.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Haowen Jiang
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen J. Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeffery Winkler
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
35
|
Kim M, Lee NK, Wang CPJ, Lim J, Byun MJ, Kim TH, Park W, Park DH, Kim SN, Park CG. Reprogramming the tumor microenvironment with biotechnology. Biomater Res 2023; 27:5. [PMID: 36721212 PMCID: PMC9890796 DOI: 10.1186/s40824-023-00343-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/22/2023] [Indexed: 02/02/2023] Open
Abstract
The tumor microenvironment (TME) is a unique environment that is developed by the tumor and controlled by tumor-induced interactions with host cells during tumor progression. The TME includes immune cells, which can be classified into two types: tumor- antagonizing and tumor-promoting immune cells. Increasing the tumor treatment responses is associated with the tumor immune microenvironment. Targeting the TME has become a popular topic in research, which includes polarizing macrophage phenotype 2 into macrophage phenotype 1 using Toll-like receptor agonists with cytokines, anti-CD47, and anti-SIPRα. Moreover, inhibiting regulatory T cells through blockades and depletion restricts immunosuppressive cells in the TME. Reprogramming T cell infiltration and T cell exhaustion improves tumor infiltrating lymphocytes, such as CD8+ or CD4+ T cells. Targeting metabolic pathways, including glucose, lipid, and amino acid metabolisms, can suppress tumor growth by restricting the absorption of nutrients and adenosine triphosphate energy into tumor cells. In conclusion, these TME reprogramming strategies exhibit more effective responses using combination treatments, biomaterials, and nanoparticles. This review highlights how biomaterials and immunotherapy can reprogram TME and improve the immune activity.
Collapse
Affiliation(s)
- Minjeong Kim
- grid.264381.a0000 0001 2181 989XDepartment of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea ,grid.264381.a0000 0001 2181 989XDepartment of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
| | - Na Kyeong Lee
- grid.264381.a0000 0001 2181 989XDepartment of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea ,grid.264381.a0000 0001 2181 989XDepartment of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
| | - Chi-Pin James Wang
- grid.264381.a0000 0001 2181 989XDepartment of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea ,grid.264381.a0000 0001 2181 989XDepartment of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
| | - Jaesung Lim
- grid.264381.a0000 0001 2181 989XDepartment of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea ,grid.264381.a0000 0001 2181 989XDepartment of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
| | - Min Ji Byun
- grid.264381.a0000 0001 2181 989XDepartment of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea ,grid.264381.a0000 0001 2181 989XDepartment of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
| | - Tae-Hyung Kim
- grid.254224.70000 0001 0789 9563School of Integrative Engineering, Chung-Ang University, 84 Heukseok-Ro, Dongjak-Gu, Seoul, 06974 Republic of Korea
| | - Wooram Park
- grid.264381.a0000 0001 2181 989XDepartment of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
| | - Dae-Hwan Park
- grid.254229.a0000 0000 9611 0917Department of Engineering Chemistry, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea ,grid.254229.a0000 0000 9611 0917Department of Industrial Cosmetic Science, College of Bio-Health University System, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea ,grid.254229.a0000 0000 9611 0917Department of Synchrotron Radiation Science and Technology, College of Bio-Health University System, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea ,grid.254229.a0000 0000 9611 0917LANG SCIENCE Inc., Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Se-Na Kim
- Research and Development Center, MediArk Inc., Cheongju, Chungbuk 28644 Republic of Korea
| | - Chun Gwon Park
- grid.264381.a0000 0001 2181 989XDepartment of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea ,grid.264381.a0000 0001 2181 989XDepartment of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea ,Research and Development Center, MediArk Inc., Cheongju, Chungbuk 28644 Republic of Korea ,grid.264381.a0000 0001 2181 989XBiomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Gyeonggi 16419 Republic of Korea ,grid.410720.00000 0004 1784 4496Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Gyeonggi 16419 Republic of Korea
| |
Collapse
|
36
|
Abstract
Cholangiocarcinoma is the second most common primary liver cancer. Its incidence is low in the Western world but is rising globally. Surgery, chemotherapy and radiation therapy have been the only treatment options for decades. Progress in our molecular understanding of the disease and the identification of druggable targets, such as IDH1 mutations and FGFR2 fusions, has provided new treatment options. Immunotherapy has emerged as a potent strategy for many different types of cancer and has shown efficacy in combination with chemotherapy for cholangiocarcinoma. In this Review, we discuss findings related to key immunological aspects of cholangiocarcinoma, including the heterogeneous landscape of immune cells within the tumour microenvironment, the immunomodulatory effect of the microbiota and IDH1 mutations, and the association of immune-related signatures and patient outcomes. We introduce findings from preclinical immunotherapy studies, discuss future immune-mediated treatment options, and provide a summary of results from clinical trials testing immune-based approaches in patients with cholangiocarcinoma. This Review provides a thorough survey of our knowledge on immune signatures and immunotherapy in cholangiocarcinoma.
Collapse
|
37
|
Systemic therapy issues: Immunotherapy in nonmetastatic urothelial cancer. Urol Oncol 2023; 41:27-34. [PMID: 34756410 DOI: 10.1016/j.urolonc.2020.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/01/2020] [Accepted: 10/07/2020] [Indexed: 11/21/2022]
Abstract
Non-muscle-invasive bladder cancer is one of the most common malignancies. Patients with intermediate-risk or high-risk disease can be treated with intravesical Bacillus Calmette-Guerin, a vaccine against tuberculosis. However, many of these patients will experience tumor recurrence, despite appropriate treatment. 1 The standard of care in these patients is radical cystectomy (RC) with urinary diversion. 2 Patients diagnosed with muscle-invasive bladder cancer (MIBC) have traditionally faced 2 main treatment options: RC and urinary diversion, as in Bacillus Calmette-Guerin-unresponsive Non-muscle-invasive bladder cancer, or alternatively, trimodal therapy comprising maximal transurethral resection of bladder tumor plus chemoradiation. 3 For patients with MIBC and clinical (c)T2-T4a, neoadjuvant chemotherapy (NAC) preceding RC is supported by Level 1 evidence with a modest 5-year overall survival benefit of 5% with cisplatin-based regimens. 4-9 A number of factors preclude MIBC patients from standard treatment options. For example, patients with serious comorbidities might be unable to tolerate general anesthesia, while others might be unwilling to adapt to the lifestyle changes after RC. 10-12 Likewise, patients with extensive carcinoma in situ or poor bladder function might not be optimal candidates for trimodal therapy or be prepared for the ongoing risk that salvage RC might be ultimately required. Reasons for the underuse of NAC range from the fear of delaying potentially curative surgery in nonresponders to patient ineligibility to cisplatin-based NAC. 13,14 Despite best efforts, in both surgical and bladder-sparing approaches, the 5-year overall survival in treated patients with MIBC is only 35% to 50%. 3,15 Strategies to improve overall prognosis as well as to reduce the indications of RC are desperately needed. Trial results have demonstrated the unprecedented ability of immune-checkpoint inhibitors to induce durable remissions in some patients with metastatic urothelial carcinoma. 16-20 Furthermore, immune-checkpoint inhibitors have shown to be better tolerated than traditional chemotherapy. 16 These successful results have spearheaded the research on these agents in earlier curative settings, with the shared goal of improving overall outcomes, and potentially avoid surgery in patients who show complete response (pT0). Strategies to enhance the immune response by combining immunotherapy with immune sensitizers such as chemotherapy, immunotherapy, targeted therapy or radiation are on the rise.
Collapse
|
38
|
Zhang D, Liang J, Lv Y, Huang X, Guo W. Tislelizumab combined with anlotinib in the second-line treatment of malignant pleural mesothelioma. Medicine (Baltimore) 2022; 101:e32459. [PMID: 36596035 PMCID: PMC9803428 DOI: 10.1097/md.0000000000032459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
RATIONALE Malignant pleural mesothelioma (MPM) is a malevolent tumor originated from pleura and often leads to poor prognosis. Chemotherapy of pemetrexed and cisplatin combined with antiangiogenic therapy of bevacizumab is recommended as the first-line regimen by guidelines. However, there are few sustainable second-line anti-tumor theraies that bring distinct survival benefit after the occurrence of drug resistance as the reported mPFS (median progression-free survival) scarcely exceeds 6 months. Immune checkpoint inhibitors are extensively investigated in pan-cancer, and dual immunotherapy has been listed in the first-line recommendation of MPM in several guidelines, while MPM patients benefit modestly from immune checkpoint inhibitors combination or monotherapy in second-line practice. PATIENT CONCERNS AND DIAGNOSIS We report a 59-year-old male patient who was diagnosed with unresectable MPM in April 2021. INTERVENTIONS He received firstly pemetrexed combined with platinum and bevacizumab, which barely curbed disease progression; When the first line treatment failed, he was switched to tislelizumab combined with anlotinib. OUTCOMES Tislelizumab combined with anlotinib significantly relieved his clinical symptoms, and imaging examination further validated the improvement. Until present, the second-line treatment PFS is more than 10 months. LESSONS The case firstly demonstrated the efficacy of tislelizumab combined with anlotinib in the second-line management of MPM. Thus, immunotherapy combined with small-molecule multi-target anti-angiogenic medication may be alternative for the second-line schemes of MPM.
Collapse
Affiliation(s)
- Dandan Zhang
- Zhongshan City People’s Hostipial, Zhongshan, China
| | | | - Yanhua Lv
- Zhongshan City People’s Hostipial, Zhongshan, China
| | - Xikun Huang
- Zhongshan City People’s Hostipial, Zhongshan, China
| | - Weihong Guo
- Zhongshan City People’s Hostipial, Zhongshan, China
- *Correspondence: Weihong Guo, Department of Respiratory and Critical Care Medicine, Zhongshan City People’s Hospital, Zhongshan City, Guangdong Province, China (e-mail: )
| |
Collapse
|
39
|
Cao H, Gao S, Jogani R, Sugimura R. The Tumor Microenvironment Reprograms Immune Cells. Cell Reprogram 2022; 24:343-352. [PMID: 36301256 DOI: 10.1089/cell.2022.0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Tumor tissue comprises a highly complex network of diverse cell types. The tumor microenvironment (TME) can be mainly subdivided into cancer cells and stromal cell compartments, the latter include different types of immune cells, fibroblasts, endothelial cells, and pericytes. Tumor cells reprogram immune cells and other stromal cells in the TME to constrain their antitumor capacity by creating an immunosuppressive milieu and metabolism competition. Moreover, the reprogramming effect on immune cells is localized not only in the tumor but also at the systemic level. With wide application of single-cell sequencing technology, tumor-specific characteristics of immune cells and other stromal cells in the TME have been dissected. In this review, we mainly focus on how tumor cells reprogram immune cells both within the TME and peripheral blood. This information can further help us to improve the efficiency of current immunotherapy as well as bring up new ideas to combat cancer.
Collapse
Affiliation(s)
- Handi Cao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,Centre for Translational Stem Cell Biology, Science Park, Hong Kong
| | - Sanxing Gao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Ritika Jogani
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Ryohichi Sugimura
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong.,Centre for Translational Stem Cell Biology, Science Park, Hong Kong
| |
Collapse
|
40
|
Pinto S, Pahl J, Schottelius A, Carter PJ, Koch J. Reimagining antibody-dependent cellular cytotoxicity in cancer: the potential of natural killer cell engagers. Trends Immunol 2022; 43:932-946. [PMID: 36306739 DOI: 10.1016/j.it.2022.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 01/12/2023]
Abstract
Bi-, tri- and multispecific antibodies have enabled the development of targeted cancer immunotherapies redirecting immune effector cells to eliminate malignantly transformed cells. These antibodies allow for simultaneous binding of surface antigens on malignant cells and activating receptors on innate immune cells, such as natural killer (NK) cells, macrophages, and neutrophils. Significant progress with such antibodies has been achieved, particularly in hematological malignancies. Nevertheless, several major challenges remain, including increasing their immunotherapeutic efficacy in a greater proportion of patients, particularly in those harboring solid tumors, and overcoming dose-limiting toxicities and immunogenicity. Here, we discuss novel antibody-engineering developments designed to maximize the potential of NK cells by NK cell engagers mediating antibody-dependent cellular cytotoxicity (ADCC), thereby expanding the armamentarium for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Paul J Carter
- Genentech, Department of Antibody Engineering, San Francisco, CA, USA
| | | |
Collapse
|
41
|
Xu Z, Zhang L, Wang M, Huang Y, Zhang M, Li S, Wang L, Li K, Hou Y. A novel subtype to predict prognosis and treatment response with DNA driver methylation-transcription in ovarian cancer. Epigenomics 2022; 14:1073-1088. [PMID: 36200265 DOI: 10.2217/epi-2022-0206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: To identify a novel subtype with DNA driver methylation-transcriptomic multiomics and predict prognosis and therapy response in serous ovarian cancer (SOC). Methods: SOC cohorts with both mRNA and methylation were collected, and DNA driver methylation (DNAme) was identified with the MithSig method. A novel prognostic subtype was developed by integrating the information on DNAme and prognosis-regulated DNAme-associated mRNA by similarity network fusion. Results: 43 overlapped DNAme were identified in three independent cohorts. SOC patients were categorized into three distinct subtypes by integrated multiomics. There were differences in prognosis, tumor microenvironment and response to therapy among the subtypes. Conclusion: This study identified 43 DNAmes and proposes a novel subtype toward personalized chemotherapy and immunotherapy for SOC patients based on multiomics.
Collapse
Affiliation(s)
- Zhenyi Xu
- Department of Epidemiology & Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, China
| | - Liuchao Zhang
- Department of Epidemiology & Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, China
| | - Meng Wang
- Department of Epidemiology & Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, China
| | - Yue Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, China
| | - Min Zhang
- Department of Epidemiology & Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, China
| | - Shuang Li
- Department of Epidemiology & Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, China
| | - Liuying Wang
- Department of Epidemiology & Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, China
| | - Kang Li
- Department of Epidemiology & Biostatistics, School of Public Health, Harbin Medical University, Harbin, 150086, China
| | - Yan Hou
- Department of Biostatistics, Peking University, Beijing, 100000, China
| |
Collapse
|
42
|
Ghosh M, Lenkiewicz AM, Kaminska B. The Interplay of Tumor Vessels and Immune Cells Affects Immunotherapy of Glioblastoma. Biomedicines 2022; 10:biomedicines10092292. [PMID: 36140392 PMCID: PMC9496044 DOI: 10.3390/biomedicines10092292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Immunotherapies with immune checkpoint inhibitors or adoptive cell transfer have become powerful tools to treat cancer. These treatments act via overcoming or alleviating tumor-induced immunosuppression, thereby enabling effective tumor clearance. Glioblastoma (GBM) represents the most aggressive, primary brain tumor that remains refractory to the benefits of immunotherapy. The immunosuppressive immune tumor microenvironment (TME), genetic and cellular heterogeneity, and disorganized vasculature hinder drug delivery and block effector immune cell trafficking and activation, consequently rendering immunotherapy ineffective. Within the TME, the mutual interactions between tumor, immune and endothelial cells result in the generation of positive feedback loops, which intensify immunosuppression and support tumor progression. We focus here on the role of aberrant tumor vasculature and how it can mediate hypoxia and immunosuppression. We discuss how immune cells use immunosuppressive signaling for tumor progression and contribute to the development of resistance to immunotherapy. Finally, we assess how a positive feedback loop between vascular normalization and immune cells, including myeloid cells, could be targeted by combinatorial therapies with immune checkpoint blockers and sensitize the tumor to immunotherapy.
Collapse
|
43
|
Yin L, Li XY, Zhu LL, Chen GL, Xiang Z, Wang QQ, Bi JW, Wang Q. Clinical application status and prospect of the combined anti-tumor strategy of ablation and immunotherapy. Front Immunol 2022; 13:965120. [PMID: 36131929 PMCID: PMC9483102 DOI: 10.3389/fimmu.2022.965120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Image-guided tumor ablation eliminates tumor cells by physical or chemical stimulation, which shows less invasive and more precise in local tumor treatment. Tumor ablation provides a treatment option for medically inoperable patients. Currently, clinical ablation techniques are widely used in clinical practice, including cryoablation, radiofrequency ablation (RFA), and microwave ablation (MWA). Previous clinical studies indicated that ablation treatment activated immune responses besides killing tumor cells directly, such as short-term anti-tumor response, immunosuppression reduction, specific and non-specific immune enhancement, and the reduction or disappearance of distant tumor foci. However, tumor ablation transiently induced immune response. The combination of ablation and immunotherapy is expected to achieve better therapeutic results in clinical application. In this paper, we provided a summary of the principle, clinical application status, and immune effects of tumor ablation technologies for tumor treatment. Moreover, we discussed the clinical application of different combination of ablation techniques with immunotherapy and proposed possible solutions for the challenges encountered by combined therapy. It is hoped to provide a new idea and reference for the clinical application of combinate treatment of tumor ablation and immunotherapy.
Collapse
Affiliation(s)
- Li Yin
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Xing-yu Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Lin-lin Zhu
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Gui-lai Chen
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Zhuo Xiang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Qing-qing Wang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Jing-wang Bi
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Qiang Wang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- *Correspondence: Qiang Wang,
| |
Collapse
|
44
|
Zhu Y, Lin X, Zhou X, Prochownik EV, Wang F, Li Y. Posttranslational control of lipogenesis in the tumor microenvironment. J Hematol Oncol 2022; 15:120. [PMID: 36038892 PMCID: PMC9422141 DOI: 10.1186/s13045-022-01340-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022] Open
Abstract
Metabolic reprogramming of cancer cells within the tumor microenvironment typically occurs in response to increased nutritional, translation and proliferative demands. Altered lipid metabolism is a marker of tumor progression that is frequently observed in aggressive tumors with poor prognosis. Underlying these abnormal metabolic behaviors are posttranslational modifications (PTMs) of lipid metabolism-related enzymes and other factors that can impact their activity and/or subcellular localization. This review focuses on the roles of these PTMs and specifically on how they permit the re-wiring of cancer lipid metabolism, particularly within the context of the tumor microenvironment.
Collapse
Affiliation(s)
- Yahui Zhu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.,School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Xingrong Lin
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Xiaojun Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Edward V Prochownik
- Division of Hematology/Oncology, Children's Hospital of Pittsburgh of UPMC, The Department of Microbiology and Molecular Genetics, The Pittsburgh Liver Research Center and The Hillman Cancer Center of UPMC, The University of Pittsburgh Medical Center, Pittsburgh, PA, 15224, USA
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430072, China.
| | - Youjun Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China. .,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
45
|
Yu X, He S, Shen J, Huang Q, Yang P, Huang L, Pu D, Wang L, Li L, Liu J, Liu Z, Zhu L. Tumor vessel normalization and immunotherapy in gastric cancer. Ther Adv Med Oncol 2022; 14:17588359221110176. [PMID: 35872968 PMCID: PMC9297465 DOI: 10.1177/17588359221110176] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 06/09/2022] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is a common malignant tumor, and patients with GC have a low survival rate due to limited effective treatment methods. Angiogenesis and immune evasion are two key processes in GC progression, and they act synergistically to promote tumor progression. Tumor vascular normalization has been shown to improve the efficacy of cancer immunotherapy, which in turn may be improved through enhanced immune stimulation. Therefore, it may be interesting to identify synergies between immunomodulatory agents and anti-angiogenic therapies in GC. This strategy aims to normalize the tumor microenvironment through the action of the anti-vascular endothelial growth factor while stimulating the immune response through immunotherapy and prolonging the survival of GC patients.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Shan He
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Jian Shen
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Qiushi Huang
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Peng Yang
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan, People's Republic of China
| | - Lin Huang
- West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Dan Pu
- West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Li Wang
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Lu Li
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Jinghua Liu
- Department of Hepatobiliary Surgery, Linyi People's Hospital, Linyi, Shandong 276000, People's Republic of China
| | - Zelong Liu
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lingling Zhu
- Lung Cancer Center, West China Hospital of Sichuan University, No. 37, Guo Xue Xiang, Wuhou District, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|
46
|
Peters S, Paz-Ares L, Herbst RS, Reck M. Addressing CPI resistance in NSCLC: targeting TAM receptors to modulate the tumor microenvironment and future prospects. J Immunother Cancer 2022; 10:jitc-2022-004863. [PMID: 35858709 PMCID: PMC9305809 DOI: 10.1136/jitc-2022-004863] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2022] [Indexed: 01/09/2023] Open
Abstract
Lung cancer remains a leading cause of cancer death worldwide, with non-small-cell lung cancer (NSCLC) accounting for the majority of cases. Immune checkpoint inhibitors (CPIs), including those targeting programmed cell death protein-1 and its ligand (PD-1/PD-L1), have revolutionized the treatment landscape for various cancers. Notably, PD-1/PD-L1 inhibitor-based regimens now form the standard first-line therapy for metastatic NSCLC, substantially improving patients' overall survival. Despite the progress made using CPI-based therapies in advanced NSCLC, most patients experience disease progression after an initial response due to resistance. Given the currently limited therapeutic options available for second-line and beyond settings in NSCLC, new treatment approaches are needed to improve long-term survival in these patients. Thus, CPI resistance is an emerging concept in cancer treatment and an active area of clinical research.Among the key mechanisms of CPI resistance is the immunosuppressive tumor microenvironment (TME). Effective CPI therapy is based on shifting immune responses against cancer cells, therefore, manipulating the immunosuppressive TME comprises an important strategy to combat CPI resistance. Several aspects of the TME can contribute to treatment resistance in NSCLC, including through the activation of Tyro3, Axl, MerTK (TAM) receptors which are essential pleiotropic regulators of immune homeostasis. Their roles include negatively modulating the immune response, therefore ectopic expression of TAM receptors in the context of cancer can contribute to the immunosuppressive, protumorigenic TME. Furthermore, TAM receptors represent important candidates to simultaneously target both tumor cells and immune cells in the TME. Clinical development of TAM receptor inhibitors (TAM RIs) is increasingly focused on their ability to rescue the antitumor immune response, thereby shifting the immunosuppressive TME to an immunostimulatory TME. There is a strong biological rationale for combining TAM RIs with a CPI to overcome resistance and improve long-term clinical responses in NSCLC. Combinatorial clinical trials of TAM RIs with CPIs are underway with encouraging preliminary results. This review outlines the key mechanisms of CPI resistance, including the role of the immunosuppressive TME, and discusses the rationale for targeting TAM receptors as a novel, promising therapeutic strategy to overcome CPI resistance in NSCLC.
Collapse
Affiliation(s)
- Solange Peters
- Medical Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Luis Paz-Ares
- Medical Oncology Department, Hospital Universitario 12 de Octubre and CNIO-H12O Lung Cancer Unit, Universidad Complutense and Ciberonc, Madrid, Spain
| | - Roy S Herbst
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, Center for Lung Research, Grosshansdorf, Germany
| |
Collapse
|
47
|
Identification and Validation of Prognosis-Related Necroptosis Genes for Prognostic Prediction in Hepatocellular Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:3172099. [PMID: 35813858 PMCID: PMC9259286 DOI: 10.1155/2022/3172099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 11/25/2022]
Abstract
Background The prediction of hepatocellular carcinoma (HCC) survival is challenging because of its rapid progression. In recent years, necroptosis was found to be involved in the progression of multiple cancer types. However, the role of necroptosis in HCC remains unclear. Methods Clinicopathological parameters and transcriptomic data of 370 HCC patients were obtained from TCGA-LIHC dataset. Prognosis-related necroptosis genes (PRNGs) were identified and utilized to construct a LASSO risk model. The GEO cohorts (GSE54236 and GSE14520) were used for external validation. We evaluated the distribution of HCC patients, the difference in prognosis, and the accuracy of the prognostic prediction of the LASSO risk model. The immune microenvironment and functional enrichment of different risk groups were further clarified. Finally, we performed a drug sensitivity analysis on the PRNGs that constructed the LASSO model and verified their mRNA expression levels in vitro. Results: A total of 48 differentially expressed genes were identified, 23 of which were PRNGs. We constructed the LASSO risk model using nine genes: SQSTM1, FLT3, HAT1, PLK1, MYCN, KLF9, HSP90AA1, TARDBP, and TNFRSF21. The outcomes of low-risk patients were considerably better than those of high-risk patients in both the training and validation cohorts. In addition, stronger bile acid metabolism, xenobiotic metabolism, and more active immune cells and immune functions were observed in low-risk patients, and high expressions of TARDBP, PLK1, and FLT3 were associated with greater drug sensitivity. With the exception of FLT3, the mRNA expression of the other eight genes was verified in Huh7 and 97H cells. Conclusions. The PRNG signature provides a novel and effective method for predicting the outcome of HCC as well as potential targets for further research.
Collapse
|
48
|
Bruschini S, Pallocca M, Sperandio E, D'Ambrosio L, Ascenzi F, De Vitis C, Salvati V, Esposito A, Di Martino S, De Nicola F, Paolini F, Fattore L, Alessandrini G, Facciolo F, Foddai ML, Bassi M, Venuta F, D'Ascanio M, Ricci A, D' Andrilli A, Napoli C, Aurisicchio L, Fanciulli M, Rendina EA, Ciliberto G, Mancini R. Deconvolution of malignant pleural effusions immune landscape unravels a novel macrophage signature associated with worse clinical outcome in lung adenocarcinoma patients. J Immunother Cancer 2022; 10:jitc-2021-004239. [PMID: 35584864 PMCID: PMC9119185 DOI: 10.1136/jitc-2021-004239] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2022] [Indexed: 12/13/2022] Open
Abstract
Background Immune checkpoint inhibitors are still unable to provide clinical benefit to the large majority of non-small cell lung cancer (NSCLC) patients. A deeper characterization of the tumor immune microenvironment (TIME) is expected to shed light on the mechanisms of cancer immune evasion and resistance to immunotherapy. Here, we exploited malignant pleural effusions (MPEs) from lung adenocarcinoma (LUAD) patients as a model system to decipher TIME in metastatic NSCLC. Methods Mononuclear cells from MPEs (PEMC) and peripheral blood (PBMC), cell free pleural fluid and/or plasma were collected from a total of 24 LUAD patients and 12 healthy donors. Bulk-RNA sequencing was performed on total RNA extracted from PEMC and matched PBMC. The DEseq2 Bioconductor package was used to perform differential expression analysis and CIBERSORTx for the regression-based immune deconvolution of bulk gene expression data. Cytokinome analysis of cell-free pleural fluid and plasma samples was performed using a 48-Plex Assay panel. THP-1 monocytic cells were used to assess macrophage polarization. Survival analyses on NSCLC patients were performed using KM Plotter (LUAD, N=672; lung squamous cell carcinoma, N=271). Results Transcriptomic analysis of immune cells and cytokinome analysis of soluble factors in the pleural fluid depicted MPEs as a metastatic niche in which all the components required for an effective antitumor response are present, but conscripted in a wound-healing, proinflammatory and tumor-supportive mode. The bioinformatic deconvolution analysis revealed an immune landscape dominated by myeloid subsets with the prevalence of monocytes, protumoral macrophages and activated mast cells. Focusing on macrophages we identified an MPEs-distinctive signature associated with worse clinical outcome in LUAD patients. Conclusions Our study reports for the first time a wide characterization of MPEs LUAD microenvironment, highlighting the importance of specific components of the myeloid compartment and opens new perspectives for the rational design of new therapies for metastatic NSCLC.
Collapse
Affiliation(s)
- Sara Bruschini
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Department Clinical and Molecular Medicine, Sant' Andrea Hospital-Sapienza University of Rome, Rome, Italy
| | - Matteo Pallocca
- Biostatistics, Bioinformatics and Clinical Trial Center, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Eleonora Sperandio
- Biostatistics, Bioinformatics and Clinical Trial Center, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Lorenzo D'Ambrosio
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Francesca Ascenzi
- Department of Clinical and Molecular Medicine, Sant' Andrea Hospital-Sapienza University of Rome, Rome, Italy
| | - Claudia De Vitis
- Department of Clinical and Molecular Medicine, Sant' Andrea Hospital-Sapienza University of Rome, Rome, Italy
| | - Valentina Salvati
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Antonella Esposito
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Simona Di Martino
- Pathology Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | | | - Francesca Paolini
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy.,HPV-Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Luigi Fattore
- SAFU Laboratory, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Francesco Facciolo
- Thoracic Surgery Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Laura Foddai
- Immunohematology and Transfusional Medicine Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Federico Venuta
- Thoracic Surgery Unit, Sapienza University of Rome, Rome, Italy
| | - Michela D'Ascanio
- Department of Clinical and Molecular Medicine, Sant' Andrea Hospital-Sapienza University of Rome, Rome, Italy
| | - Alberto Ricci
- Department of Clinical and Molecular Medicine, Sant' Andrea Hospital-Sapienza University of Rome, Rome, Italy
| | - Antonio D' Andrilli
- Thoracic Surgery Unit, Sant' Andrea Hospital-Sapienza University of Rome, Rome, Italy
| | - Christian Napoli
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Maurizio Fanciulli
- SAFU Laboratory, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Erino Angelo Rendina
- Thoracic Surgery Unit, Sant' Andrea Hospital-Sapienza University of Rome, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Directorate, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Sant' Andrea Hospital-Sapienza University of Rome, Rome, Italy
| |
Collapse
|
49
|
Zhang H, Zhuo Y, Li D, Zhang L, Gao Q, Yang L, Yuan X. Dihydroartemisinin inhibits the growth of pancreatic cells by inducing ferroptosis and activating antitumor immunity. Eur J Pharmacol 2022; 926:175028. [DOI: 10.1016/j.ejphar.2022.175028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 12/19/2022]
|
50
|
Chen XQ, Zhao YX, Zhang CL, Wang XT, Zhang X, Chen X, Yuan CW, Zhao Q, Chen XJ. Effectiveness and Safety of Anlotinib with or without PD-1 Blockades in the Treatment of Patients with Advanced Primary Hepatocellular Carcinoma: A Retrospective, Real-World Study in China. Drug Des Devel Ther 2022; 16:1483-1493. [PMID: 35607597 PMCID: PMC9123907 DOI: 10.2147/dddt.s358092] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/30/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Xiao-Qi Chen
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450000, People’s Republic of China
| | - Yun-Xia Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450000, People’s Republic of China
| | - Chuan-Lei Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450000, People’s Republic of China
| | - Xin-Ting Wang
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450000, People’s Republic of China
| | - Xin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450000, People’s Republic of China
| | - Xi Chen
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450000, People’s Republic of China
| | - Chang-Wei Yuan
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450000, People’s Republic of China
| | - Qing Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450000, People’s Republic of China
| | - Xin-Ju Chen
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, 450000, People’s Republic of China
- Correspondence: Xin-Ju Chen, Department of Gastroenterology, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, No. 19, Renmin Road, Jinshui District, Zhengzhou, Henan, 450000, People’s Republic of China, Tel +86-13700867158, Email
| |
Collapse
|