251
|
Lipid Nanoparticles for mRNA Delivery to Enhance Cancer Immunotherapy. Molecules 2022; 27:molecules27175607. [PMID: 36080373 PMCID: PMC9458026 DOI: 10.3390/molecules27175607] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 12/24/2022] Open
Abstract
Messenger RNA (mRNA) is being developed by researchers as a novel drug for the treatment or prevention of many diseases. However, to enable mRNA to fully exploit its effects in vivo, researchers need to develop safer and more effective mRNA delivery systems that improve mRNA stability and enhance the ability of cells to take up and release mRNA. To date, lipid nanoparticles are promising nanodrug carriers for tumor therapy, which can significantly improve the immunotherapeutic effects of conventional drugs by modulating mRNA delivery, and have attracted widespread interest in the biomedical field. This review focuses on the delivery of mRNA by lipid nanoparticles for cancer treatment. We summarize some common tumor immunotherapy and mRNA delivery strategies, describe the clinical advantages of lipid nanoparticles for mRNA delivery, and provide an outlook on the current challenges and future developments of this technology.
Collapse
|
252
|
Yu L, Gong C. Pancancer analysis of a potential gene mutation model in the prediction of immunotherapy outcomes. Front Genet 2022; 13:917118. [PMID: 36092890 PMCID: PMC9459043 DOI: 10.3389/fgene.2022.917118] [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: 05/08/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Immune checkpoint blockade (ICB) represents a promising treatment for cancer, but predictive biomarkers are needed. We aimed to develop a cost-effective signature to predict immunotherapy benefits across cancers.Methods: We proposed a study framework to construct the signature. Specifically, we built a multivariate Cox proportional hazards regression model with LASSO using 80% of an ICB-treated cohort (n = 1661) from MSKCC. The desired signature named SIGP was the risk score of the model and was validated in the remaining 20% of patients and an external ICB-treated cohort (n = 249) from DFCI.Results: SIGP was based on 18 candidate genes (NOTCH3, CREBBP, RNF43, PTPRD, FAM46C, SETD2, PTPRT, TERT, TET1, ROS1, NTRK3, PAK7, BRAF, LATS1, IL7R, VHL, TP53, and STK11), and we classified patients into SIGP high (SIGP-H), SIGP low (SIGP-L) and SIGP wild type (SIGP-WT) groups according to the SIGP score. A multicohort validation demonstrated that patients in SIGP-L had significantly longer overall survival (OS) in the context of ICB therapy than those in SIGP-WT and SIGP-H (44.00 months versus 13.00 months and 14.00 months, p < 0.001 in the test set). The survival of patients grouped by SIGP in non-ICB-treated cohorts was different, and SIGP-WT performed better than the other groups. In addition, SIGP-L + TMB-L (approximately 15% of patients) had similar survivals to TMB-H, and patients with both SIGP-L and TMB-H had better survival. Further analysis on tumor-infiltrating lymphocytes demonstrated that the SIGP-L group had significantly increased abundances of CD8+ T cells.Conclusion: Our proposed model of the SIGP signature based on 18-gene mutations has good predictive value for the clinical benefit of ICB in pancancer patients. Additional patients without TMB-H were identified by SIGP as potential candidates for ICB, and the combination of both signatures showed better performance than the single signature.
Collapse
Affiliation(s)
- Lishan Yu
- Yanqi Lake Beijing Institute Mathematical Sciences and Applications, Beijing, China
- Yau Mathematical Sciences Center, Tsinghua University, Beijing, China
| | - Caifeng Gong
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Caifeng Gong,
| |
Collapse
|
253
|
Zhou Y, Wu B, Li T, Zhang Y, Xu T, Chang N, Zhang J. Correlation between the Immune Checkpoint Inhibitors Prognostic Index and Outcomes in Nonsmall Cell Lung Cancer: A Multicentre Analysis. JOURNAL OF ONCOLOGY 2022; 2022:7050817. [PMID: 36059799 PMCID: PMC9439892 DOI: 10.1155/2022/7050817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/19/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To evaluate the prognostic value of the immune checkpoint inhibitor prognostic index (ICPI), based on the albumin (ALB) and derived neutrophil-to-lymphocyte ratio (dNLR), for nonsmall cell lung cancer (NSCLC) patients receiving immune checkpoint inhibitors (ICIs). METHODS We conducted a multicentre retrospective study with an ICIs cohort (n = 143) and a chemotherapy control cohort (n = 84). A Cox proportional hazards regression and logistic regression model were used to find the independent risk factor for progression-free survival (PFS) and overall survival (OS) and disease control rate (DCR) in NSCLC patients. The Kaplan-Meier was used to evaluating the PFS and OS. RESULTS The ALB <35 g/L and dNLR >3 were correlated with worse PFS and OS for NSCLC patients receiving ICIs, respectively. The moderately high-risk ICPI had a significantly increased risk of progression (hazard ratio (HR) 1.83, 95% confidence interval (CI) 1.14-2.91; P=0.012) and of death (HR 2.33, 95% CI 1.12-4.87; P=0.024) and of nondisease control (odds ratio (OR) 3.05, 95% CI 1.19-7.83; P=0.021) and was correlated with worse PFS and 1-year survival rates (4.0 months vs. 7.2 months; P=0.001; 44.3% vs. 76.1%; P=0.001) compared with low-risk ICPI when it was characterized two groups. When ICPI was further divided into three groups, the results showed that the high-risk ICPI was correlated with worse PFS and 1-year survival rates. However, there was no difference in the chemotherapy cohort. CONCLUSION The ICPI was correlated with worse outcomes for NSCLC patients receiving ICIs but not for patients with chemotherapy.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shannxi, China
| | - Bin Wu
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shannxi, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, Shannxi, China
| | - Yong Zhang
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shannxi, China
| | - Tianqi Xu
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shannxi, China
| | - Ning Chang
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shannxi, China
| | - Jian Zhang
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shannxi, China
| |
Collapse
|
254
|
Han J, Bhatta R, Liu Y, Bo Y, Wang H. In Situ Dendritic Cell Recruitment and T Cell Activation for Cancer Immunotherapy. Front Pharmacol 2022; 13:954955. [PMID: 36081933 PMCID: PMC9445184 DOI: 10.3389/fphar.2022.954955] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/13/2022] [Indexed: 12/18/2022] Open
Abstract
Cancer immunotherapy has shifted the paradigm for cancer treatment in the past decade, but new immunotherapies enabling the effective treatment of solid tumors are still greatly demanded. Here we report a pore-forming hydrogel-based immunotherapy that enables simultaneous recruitment of dendritic cells and in situ activation of T cells, for reshaping the immunosuppressive tumor microenvironment and amplifying cytotoxic T lymphocyte response. The injectable pore-forming hydrogel composed of porogen-dispersed alginate network can form a macroporous structure upon injection into mice, and enables controlled release of granulocyte-macrophage colony-stimulating factor (GM-CSF), a chemoattractant for recruiting dendritic cells, and epacadostat, an inhibitor of indoleamine 2, 3-dioxygenase for activating T cells. We show that gels loaded with GM-CSF and epacadostat, after peritumoral injection, can recruit massive dendritic cells in situ and activate effector T cells in the tumor tissues, resulting in enhanced frequency and activation status of dendritic cells, reduced numbers of regulatory T (Treg) cells, and increased CD8+/Treg ratios in the tumor microenvironment. This hydrogel-based immunotherapy holds great promise for treating poorly-immunogenic solid tumors.
Collapse
Affiliation(s)
- Joonsu Han
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Rimsha Bhatta
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Yusheng Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Yang Bo
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Cancer Center at Illinois (CCIL), Urbana, IL, United States
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- *Correspondence: Hua Wang,
| |
Collapse
|
255
|
Xu R, Liu K, Wang X, Zhang C, Zhang Y, Yang J. In situ release of IL-2/IL-12 from SiO 2-engineered dendritic cells for synergistic immunotherapy. NANOSCALE 2022; 14:11235-11251. [PMID: 35876611 DOI: 10.1039/d2nr02012b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Herein, a synergistic therapy strategy of cytokine and dendritic cell (DC) vaccine was developed via the chemical conjugation of cytokine-loaded SiO2 directly on the plasma membrane of DCs. Firstly, IL-2/IL-12-loaded SiO2 was prepared and modified with MAL-PEG-NHS, and then coupled on the membrane of mature DCs through the coupling of -MAL and -SH groups. The large surface area and bimodal pores of SiO2 endowed it with high cytokine loading capacity and entrapment efficiency (EE%), with EEIL-2% of 95.8% and EEIL-12% of 86.4%. SiO2 was stably attached to the surface of DCs, and thus not internalized by mature DCs, and the SiO2 conjugation blocked only 4.37% of the total available cell surface thiol groups. After SiO2 attachment, the cell viability, membrane integrity and intracellular reactive oxygen species (ROS) of DCs were not affected. Furthermore, this strategy avoids the systemic toxicity of cytokines and improves the ability of DCs to target lymph nodes. IL-2 and IL-12 were only released locally around DCs, enabling the pseudo-autocrine stimulation of the transferred DCs in vivo. Moreover, the long-term anti-tumor protection in a B16 tumor model was demonstrated. This strategy is a facile and generalizable dendritic cell-based cancer immunotherapy strategy to augment bioavailability, while minimizing the side effects of cytokines.
Collapse
Affiliation(s)
- Rong Xu
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Kaijing Liu
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Xiaoli Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Chuangnian Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yajing Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Jing Yang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
256
|
Yang M, Olaoba OT, Zhang C, Kimchi ET, Staveley-O’Carroll KF, Li G. Cancer Immunotherapy and Delivery System: An Update. Pharmaceutics 2022; 14:1630. [PMID: 36015256 PMCID: PMC9413869 DOI: 10.3390/pharmaceutics14081630] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 02/06/2023] Open
Abstract
With an understanding of immunity in the tumor microenvironment, immunotherapy turns out to be a powerful tool in the clinic to treat many cancers. The strategies applied in cancer immunotherapy mainly include blockade of immune checkpoints, adoptive transfer of engineered cells, such as T cells, natural killer cells, and macrophages, cytokine therapy, cancer vaccines, and oncolytic virotherapy. Many factors, such as product price, off-target side effects, immunosuppressive tumor microenvironment, and cancer cell heterogeneity, affect the treatment efficacy of immunotherapies against cancers. In addition, some treatments, such as chimeric antigen receptor (CAR) T cell therapy, are more effective in treating patients with lymphoma, leukemia, and multiple myeloma rather than solid tumors. To improve the efficacy of targeted immunotherapy and reduce off-target effects, delivery systems for immunotherapies have been developed in past decades using tools such as nanoparticles, hydrogel matrix, and implantable scaffolds. This review first summarizes the currently common immunotherapies and their limitations. It then synopsizes the relative delivery systems that can be applied to improve treatment efficacy and minimize side effects. The challenges, frontiers, and prospects for applying these delivery systems in cancer immunotherapy are also discussed. Finally, the application of these approaches in clinical trials is reviewed.
Collapse
Affiliation(s)
- Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
| | - Olamide Tosin Olaoba
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
| | - Chunye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65212, USA
| | - Eric T. Kimchi
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Kevin F. Staveley-O’Carroll
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Guangfu Li
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
257
|
Wang S, Wang Y, Jin K, Zhang B, Peng S, Nayak AK, Pang Z. Recent advances in erythrocyte membrane-camouflaged nanoparticles for the delivery of anti-cancer therapeutics. Expert Opin Drug Deliv 2022; 19:965-984. [PMID: 35917435 DOI: 10.1080/17425247.2022.2108786] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Red blood cell (or erythrocyte) membrane-camouflaged nanoparticles (RBC-NPs) not only have a superior circulation life and do not induce accelerated blood clearance, but also possess special functions, which offers great potential in cancer therapy. AREAS COVERED This review focuses on the recent advances of RBC-NPs for delivering various agents to treat cancers in light of their vital role in improving drug delivery. Meanwhile, the construction and in vivo behavior of RBC-NPs are discussed to provide an in-depth understanding of the basis of RBC-NPs for improved cancer drug delivery. EXPERT OPINION Although RBC-NPs are quite prospective in delivering anti-cancer therapeutics, they are still in their infancy stage and many challenges need to be overcome for successful translation into the clinic. The preparation and modification of RBC membranes, the optimization of coating methods, the scale-up production and the quality control of RBC-NPs, and the drug loading and release should be carefully considered in the clinical translation of RBC-NPs for cancer therapy.
Collapse
Affiliation(s)
- Siyu Wang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, China
| | - Yiwei Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Kai Jin
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, China
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Shaojun Peng
- Zhuhai Institute of Translational Medicine, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong 519000, China
| | - Amit Kumar Nayak
- Department of Pharmaceutics, Seemanta Institute of Pharmaceutical Sciences, Mayurbhanj-757086, Odisha, India
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, China
| |
Collapse
|
258
|
Liu J, Wang Y, Mu C, Li M, Li K, Li S, Wu W, Du L, Zhang X, Li C, Peng W, Shen J, Liu Y, Yang D, Zhang K, Ning Q, Fu X, Zeng Y, Ni Y, Zhou Z, Liu Y, Hu Y, Zheng X, Wen T, Li Z, Liu Y. Pancreatic tumor eradication via selective Pin1 inhibition in cancer-associated fibroblasts and T lymphocytes engagement. Nat Commun 2022; 13:4308. [PMID: 35879297 PMCID: PMC9314377 DOI: 10.1038/s41467-022-31928-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 07/11/2022] [Indexed: 02/05/2023] Open
Abstract
Cancer associated fibroblasts (CAFs) support tumors via multiple mechanisms, including maintaining the immunosuppressive tumor microenvironment and limiting infiltration of immune cells. The prolyl isomerase Pin1, whose overexpression in CAFs has not been fully profiled yet, plays critical roles in tumor initiation and progression. To decipher effects of selective Pin1 inhibition in CAFs on pancreatic cancer, here we formulate a DNA-barcoded micellular system (DMS) encapsulating the Pin1 inhibitor AG17724. DMS functionalized with CAF-targeting anti-FAP-α antibodies (antiCAFs-DMS) can selectively inhibit Pin1 in CAFs, leading to efficacious but transient tumor growth inhibition. We further integrate DNA aptamers (AptT), which can engage CD8+ T lymphocytes, to obtain a bispecific antiCAFs-DMS-AptT system. AntiCAFs-DMS-AptT inhibits tumor growth in subcutaneous and orthotopic pancreatic cancer models. Pharmacological inhibition of the prolyl isomerase PIN1, highly expressed in cancer cells and cancer associated fibroblasts (CAF), has been proposed for cancer therapy. Here the authors report the design of a DNA-barcoded micellular system functionalized with antibodies targeting CAFs and a T cell recruiting aptamer to deliver the PIN1 inhibitor AG17724, showing antitumor response in preclinical models of pancreatic cancer.
Collapse
Affiliation(s)
- Jiaye Liu
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Thyroid and Parathyroid diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China.,Respiratory Health Institute, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Wang
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Chunyang Mu
- Department of Liver Surgery & Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Li
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Bioland Laboratory, Guangzhou, China
| | - Kewei Li
- Department of Pediatric Department, West China Hospital, Sichuan University, Chengdu, China
| | - Shan Li
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Wenshuang Wu
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Thyroid and Parathyroid diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Lingyao Du
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyun Zhang
- Department of Liver Surgery & Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chuan Li
- Department of Liver Surgery & Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Peng
- Department of Liver Surgery & Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Junyi Shen
- Department of Liver Surgery & Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Thyroid and Parathyroid diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Dujiang Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kaixiang Zhang
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Thyroid and Parathyroid diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Qingyang Ning
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Thyroid and Parathyroid diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoying Fu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Zeng
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Yinyun Ni
- Respiratory Health Institute, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zongguang Zhou
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yiguo Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Xiaofeng Zheng
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China.
| | - Tianfu Wen
- Department of Liver Surgery & Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Zhihui Li
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China. .,Laboratory of Thyroid and Parathyroid diseases, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Yong Liu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
259
|
Cai Q, Duan J, Ding L. Prognostic model of immune-related genes for patients with hepatocellular carcinoma. Front Surg 2022; 9:819491. [PMID: 35937592 PMCID: PMC9349350 DOI: 10.3389/fsurg.2022.819491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
Background Immune-related genes (IRGs) are closely connected to the occurrence and development of tumors. Their influence on the prognosis of patients with HCC, however, remains unclear. Methods From the TCGA database, we integrated 365 liver cancer tissues and 50 normal tissues to identify differential immune genes related to prognosis. Multivariate COX analysis was used to establish a new prognostic index on account of IRGs, whereby risk score = (Expression level of HSPA4*0.022) + (Expression level of PSMD14*0.042) + (Expression level of RBP2*0.019) + (Expression level of MAPT*0.197) + (Expression level of TRAF3*0.146) + (Expression level of NDRG1*(0.006) + (Expression level of NRAS*0.027) + (Expression level of IL17D*0.075). Results The risk score was clearly correlated with an unfavorable survival rate and with clinical characteristics. By integrating the immune-related risk score model with clinical features, a nomogram was constructed to predict the survival rate of HCC patients (1-, 3- and 5-year AUC of 0.721, 0.747 and 0.781, respectively). Conclusion We have established a valuable prognostic risk score for HCC patients that may be a better predictor of survival than the present method. With the risk score's strong predictive value for immune cells and functions, it may provide clinical guidance for the diagnosis and prognosis of different immunophenotypes, and provide multiple therapeutic targets for the treatment of HCC patients based on subtype-specific immune molecules.
Collapse
Affiliation(s)
- Qun Cai
- Department of Infectious Diseases and Liver Diseases, Ningbo Medical Center Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
- Correspondence: Qun Cai
| | - Jinnan Duan
- Department of Infectious Diseases, Shaoxing People's Hospital, Shaoxing, China
| | - Liang Ding
- Department of Infectious Diseases, Shaoxing People's Hospital, Shaoxing, China
| |
Collapse
|
260
|
Sewnath CA, Behrens LM, van Egmond M. Targeting myeloid cells with bispecific antibodies as novel immunotherapies of cancer. Expert Opin Biol Ther 2022; 22:983-995. [PMID: 35854649 DOI: 10.1080/14712598.2022.2098675] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Most bispecific antibody (BsAb) therapies focus on stimulating the adaptive immune system, in particular T cells, to promote tumor cell killing. Another method to promote tumor eradication is through the engagement of myeloid cells, including macrophages and neutrophils, which are abundantly present and possess intrinsic cytotoxic mechanisms for tumor cell killing, making them interesting effector cells to recruit for BsAb therapy. AREAS COVERED In this review, we describe the evolving knowledge of the role of macrophages and neutrophils in cancer in scientific literature. Moreover, we address the BsAbs that have been developed over the years to recruit these cell types as effector cells in immunotherapy of cancer. This includes the discussion of BsAbs that target Fc receptors (i.e. FcγR and FcαRI) to induce antibody-dependent cellular phagocytosis (ADCP) by macrophages or trogoptosis via neutrophils, as well as BsAbs that interfere with checkpoint inhibition, including the SIRPα-CD47 pathway. EXPERT OPINION Elucidating the complexity of macrophage and neutrophil heterogeneity in cancer may help to specifically enlist the cytotoxic ability of these cells through targeting Fc receptors and checkpoint pathways, which may further enhance anti-cancer immunity.
Collapse
Affiliation(s)
- Celine An Sewnath
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands.,Cancer Biology and Immunology Program, Cancer Centre Amsterdam, Amsterdam, The Netherlands.,Cancer Immunology Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Leonie M Behrens
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands.,Cancer Biology and Immunology Program, Cancer Centre Amsterdam, Amsterdam, The Netherlands.,Cancer Immunology Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands.,Cancer Biology and Immunology Program, Cancer Centre Amsterdam, Amsterdam, The Netherlands.,Cancer Immunology Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands.,Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam De Boelelaan, Amsterdam, The Netherlands
| |
Collapse
|
261
|
Nilsson LM, Vilhav C, Karlsson JW, Fagman JB, Giglio D, Engström CE, Naredi P, Nilsson JA. Genetics and Therapeutic Responses to Tumor-Infiltrating Lymphocyte Therapy of Pancreatic Cancer Patient-Derived Xenograft Models. GASTRO HEP ADVANCES 2022; 1:1037-1048. [PMID: 39131259 PMCID: PMC11307969 DOI: 10.1016/j.gastha.2022.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 07/11/2022] [Indexed: 08/13/2024]
Abstract
Background and Aims Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide. Checkpoint immunotherapy has not yet shown encouraging results in pancreatic cancer possibly because of a poor immunogenicity and/or an immune suppressive microenvironment. The aim of this study was to develop patient-derived xenograft (PDX) models, compare their genetics to the original biopsies, and assess if autologous tumor-infiltrating lymphocytes (TILs) would have antitumoral activity in pancreatic cancer. Methods We subcutaneously transplanted tumors from 29 patients into NOG mice to generate PDX models. We established TIL cultures and injected them into PDX mice. We analyzed histology and genetics of biopsies and PDX tumors. Results Tumor growths were confirmed in 11 of 29 transplantations. The PDX tumors histologically resembled their original biopsies, but because stromal cells in the PDX model tumors were from mouse, their gene expression differed from the original biopsies. Immune checkpoint ligands other than programmed death ligand-1 (PD-L1) were expressed in pancreatic cancers, but PD-L1 was rarely expressed. When it was expressed, it correlated with tumor take in PDX models. One of the 3 tumors that expressed PD-L1 was an adenosquamous cancer, and another had a mismatch repair deficiency. TILs were expanded from 6 tumors and were injected into NOG or human interleukin-2 transgenic-NOG mice carrying PDX tumors. Regression of tumors could be verified in human interleukin-2 transgenic-NOG mice in 3 of the 6 PDX models treated with autologous TILs, including the adenosquamous PDX model. Conclusion PDX models of pancreatic cancer can be used to learn more about tumor characteristics and biomarkers and to evaluate responses to adoptive cell therapy and combination therapies. The major benefit of the model is that modifications of T cells can be tested in an autologous humanized mouse model to gain preclinical data to support the initiation of a clinical trial.
Collapse
Affiliation(s)
- Lisa M. Nilsson
- Department of Surgery, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Australia
| | - Caroline Vilhav
- Department of Surgery, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Joakim W. Karlsson
- Department of Surgery, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Australia
| | - Johan Bourghardt Fagman
- Department of Surgery, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Daniel Giglio
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Cecilia E. Engström
- Department of Surgery, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Peter Naredi
- Department of Surgery, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jonas A. Nilsson
- Department of Surgery, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Australia
| |
Collapse
|
262
|
Cui G, Wang H, Long S, Zhang T, Guo X, Chen S, Kakuchi T, Duan Q, Zhao D. Thermo- and Light-Responsive Polymer-Coated Magnetic Nanoparticles as Potential Drug Carriers. Front Bioeng Biotechnol 2022; 10:931830. [PMID: 35903791 PMCID: PMC9315361 DOI: 10.3389/fbioe.2022.931830] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
A series of thermo- and light-responsive copolymers of poly (N-isopropylacrylamide) (PNIPAM) and 6-[4-(4-methoxy phenyl azo)-phenoxyl-hexyl methacrylate) (AzoMA) (PNIPAM-b-PAzoMA) were synthesized via reversible addition–fragmentation chain transfer (RAFT) radical polymerization. The resulting copolymers had a narrow molecular weight distribution range of 1.06–1.24, in which Mn changed regularly with the monomer concentration. Subsequently, the diblock copolymers were successfully modified on the surface of iron oxide nanoparticles through the interaction between the chemical bonds to prepare Fe3O4@(PNIPAM-b-PAzoMA) nanoparticles. The size of fabricated nanoparticles with excellent thermo-sensitivity and photo-sensitivity was controlled at about 40–50 nm. Cell viability assays suggested that the nanoparticles showed no significant cytotoxicity and potential drug delivery in the tumor microenvironment.
Collapse
Affiliation(s)
- Guihua Cui
- Science and Technology Division, Jilin Medical University, Jilin, China
- Department of Materials Science and Engineering, Changchun University of Science and Technology, Jilin, China
| | - Hao Wang
- Science and Technology Division, Jilin Medical University, Jilin, China
| | - Shengsen Long
- Science and Technology Division, Jilin Medical University, Jilin, China
| | - Tianshuo Zhang
- Science and Technology Division, Jilin Medical University, Jilin, China
| | - Xiaoyu Guo
- Jilin Vocational College of Industry and Technology, Jilin, China
| | - Shuiying Chen
- Department of Materials Science and Engineering, Changchun University of Science and Technology, Jilin, China
| | - Toyoji Kakuchi
- Division of Biotechnology and Macromolecular Chemistry, Graduate School of Engineering, Hokkaido University, Sapporo, Japan
| | - Qian Duan
- Department of Materials Science and Engineering, Changchun University of Science and Technology, Jilin, China
- *Correspondence: Qian Duan, ; Donghai Zhao,
| | - Donghai Zhao
- Science and Technology Division, Jilin Medical University, Jilin, China
- *Correspondence: Qian Duan, ; Donghai Zhao,
| |
Collapse
|
263
|
Xiao M, Lai W, Yao X, Pei H, Fan C, Li L. Programming Receptor Clustering with DNA Probabilistic Circuits for Enhanced Natural Killer Cell Recognition. Angew Chem Int Ed Engl 2022; 61:e202203800. [DOI: 10.1002/anie.202203800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Mingshu Xiao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes School of Chemistry and Molecular Engineering East China Normal University 500 Dongchuan Road Shanghai 200241 P. R. China
| | - Wei Lai
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes School of Chemistry and Molecular Engineering East China Normal University 500 Dongchuan Road Shanghai 200241 P. R. China
| | - Xiaowei Yao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes School of Chemistry and Molecular Engineering East China Normal University 500 Dongchuan Road Shanghai 200241 P. R. China
| | - Hao Pei
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes School of Chemistry and Molecular Engineering East China Normal University 500 Dongchuan Road Shanghai 200241 P. R. China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering Institute of Molecular Medicine Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Li Li
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes School of Chemistry and Molecular Engineering East China Normal University 500 Dongchuan Road Shanghai 200241 P. R. China
| |
Collapse
|
264
|
Behrens LM, van den Berg TK, van Egmond M. Targeting the CD47-SIRPα Innate Immune Checkpoint to Potentiate Antibody Therapy in Cancer by Neutrophils. Cancers (Basel) 2022; 14:cancers14143366. [PMID: 35884427 PMCID: PMC9319280 DOI: 10.3390/cancers14143366] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Immunotherapy aims to engage various immune cells in the elimination of cancer cells. Neutrophils are the most abundant leukocytes in the circulation and have unique mechanisms by which they can kill cancer cells opsonized by antibodies. However, neutrophil effector functions are limited by the inhibitory receptor SIRPα, when it interacts with CD47. The CD47 protein is expressed on all cells in the body and acts as a ‘don’t eat me’ signal to prevent tissue damage. Cancer cells can express high levels of CD47 to circumvent tumor elimination. Thus, blocking the interaction between CD47 and SIRPα may enhance anti-tumor effects by neutrophils in the presence of tumor-targeting monoclonal antibodies. In this review, we discuss CD47-SIRPα as an innate immune checkpoint on neutrophils and explore the preliminary results of clinical trials using CD47-SIRPα blocking agents. Abstract In the past 25 years, a considerable number of therapeutic monoclonal antibodies (mAb) against a variety of tumor-associated antigens (TAA) have become available for the targeted treatment of hematologic and solid cancers. Such antibodies opsonize cancer cells and can trigger cytotoxic responses mediated by Fc-receptor expressing immune cells in the tumor microenvironment (TME). Although frequently ignored, neutrophils, which are abundantly present in the circulation and many cancers, have demonstrated to constitute bona fide effector cells for antibody-mediated tumor elimination in vivo. It has now also been established that neutrophils exert a unique mechanism of cytotoxicity towards antibody-opsonized tumor cells, known as trogoptosis, which involves Fc-receptor (FcR)-mediated trogocytosis of cancer cell plasma membrane leading to a lytic/necrotic type of cell death. However, neutrophils prominently express the myeloid inhibitory receptor SIRPα, which upon interaction with the ‘don’t eat me’ signal CD47 on cancer cells, limits cytotoxicity, forming a mechanism of resistance towards anti-cancer antibody therapeutics. In fact, tumor cells often overexpress CD47, thereby even more strongly restricting neutrophil-mediated tumor killing. Blocking the CD47-SIRPα interaction may therefore potentiate neutrophil-mediated antibody-dependent cellular cytotoxicity (ADCC) towards cancer cells, and various inhibitors of the CD47-SIRPα axis are now in clinical studies. Here, we review the role of neutrophils in antibody therapy in cancer and their regulation by the CD47-SIRPα innate immune checkpoint. Moreover, initial results of CD47-SIRPα blockade in clinical trials are discussed.
Collapse
Affiliation(s)
- Leonie M. Behrens
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.K.v.d.B.); (M.v.E.)
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology Program, 1081 HV Amsterdam, The Netherlands
- Correspondence:
| | - Timo K. van den Berg
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.K.v.d.B.); (M.v.E.)
- Byondis B.V., 6545 CM Nijmegen, The Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (T.K.v.d.B.); (M.v.E.)
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology Program, 1081 HV Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
265
|
Guzzeloni V, Veschini L, Pedica F, Ferrero E, Ferrarini M. 3D Models as a Tool to Assess the Anti-Tumor Efficacy of Therapeutic Antibodies: Advantages and Limitations. Antibodies (Basel) 2022; 11:antib11030046. [PMID: 35892706 PMCID: PMC9326665 DOI: 10.3390/antib11030046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 02/07/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) are an emerging and very active frontier in clinical oncology, with hundred molecules currently in use or being tested. These treatments have already revolutionized clinical outcomes in both solid and hematological malignancies. However, identifying patients who are most likely to benefit from mAbs treatment is currently challenging and limiting the impact of such therapies. To overcome this issue, and to fulfill the expectations of mAbs therapies, it is urgently required to develop proper culture models capable of faithfully reproducing the interactions between tumor and its surrounding native microenvironment (TME). Three-dimensional (3D) models which allow the assessment of the impact of drugs on tumors within its TME in a patient-specific context are promising avenues to progressively fill the gap between conventional 2D cultures and animal models, substantially contributing to the achievement of personalized medicine. This review aims to give a brief overview of the currently available 3D models, together with their specific exploitation for therapeutic mAbs testing, underlying advantages and current limitations to a broader use in preclinical oncology.
Collapse
Affiliation(s)
- Virginia Guzzeloni
- B-Cell Neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (V.G.); (E.F.)
| | - Lorenzo Veschini
- Academic Centre of Reconstructive Science, Faculty of Dentistry Oral & Craniofacial Sciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK;
| | - Federica Pedica
- Pathology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
| | - Elisabetta Ferrero
- B-Cell Neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (V.G.); (E.F.)
| | - Marina Ferrarini
- B-Cell Neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (V.G.); (E.F.)
- Correspondence:
| |
Collapse
|
266
|
Wang F, Yang L, Xiao M, Zhang Z, Shen J, Anuchapreeda S, Tima S, Chiampanichayakul S, Xiao Z. PD-L1 regulates cell proliferation and apoptosis in acute myeloid leukemia by activating PI3K-AKT signaling pathway. Sci Rep 2022; 12:11444. [PMID: 35794161 PMCID: PMC9259561 DOI: 10.1038/s41598-022-15020-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/16/2022] [Indexed: 01/02/2023] Open
Abstract
As immune checkpoint inhibitors (ICIs) continue to advance, more evidence has emerged that anti-PD-1/PD-L1 immunotherapy is an effective treatment against cancers. Known as the programmed death ligand-1 (PD-L1), this co-inhibitory ligand contributes to T cell exhaustion by interacting with programmed death-1 (PD-1) receptor. However, cancer-intrinsic signaling pathways of the PD-L1 molecule are not well elucidated. Therefore, the present study aimed to evaluate the regulatory network of PD-L1 and lay the basis of successful use of anti-PD-L1 immunotherapy in acute myeloid leukemia (AML). Data for AML patients were extracted from TCGA and GTEx databases. The downstream signaling pathways of PD-L1 were identified via Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. The key PD-L1 related genes were selected by weighted gene co-expression network analysis (WGCNA), MCC algorithm and Molecular Complex Detection (MCODE). The CCK-8 assay was used to assess cell proliferation. Flow cytometry was used to determine cell apoptosis and cell cycle. Western blotting was used to identify the expression of the PI3K-AKT signaling pathway. PD-L1 was shown to be elevated in AML patients when compared with the control group, and high PD-L1 expression was associated with poor overall survival rate. The ECM-receptor interaction, as well as the PI3K-AKT signaling pathway, were important PD-L1 downstream pathways. All three analyses found eight genes (ITGA2B, ITGB3, COL6A5, COL6A6, PF4, NMU, AGTR1, F2RL3) to be significantly associated with PD-L1. Knockdown of PD-L1 inhibited AML cell proliferation, induced cell apoptosis and G2/M cell cycle arrest. Importantly, PD-L1 knockdown reduced the expression of PI3K and p-AKT, but PD-L1 overexpression increased their expression. The current study elucidates the main regulatory network and downstream targets of PD-L1 in AML, assisting in the understanding of the underlying mechanism of anti-PD-1/PD-L1 immunotherapy and paving the way for clinical application of ICIs in AML.
Collapse
Affiliation(s)
- Fang Wang
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Liqiong Yang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Mintao Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Zhuo Zhang
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Research Center of Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai, Thailand
| | - Singkome Tima
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Research Center of Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai, Thailand
| | - Sawitree Chiampanichayakul
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand. .,Research Center of Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai, Thailand.
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China. .,South Sichuan Institute of Translational Medicine, Luzhou, China.
| |
Collapse
|
267
|
Chen C, Wang R, Chen X, Hou Y, Jiang J. Targeting CD47 as a Novel Immunotherapy for Breast Cancer. Front Oncol 2022; 12:924740. [PMID: 35860564 PMCID: PMC9289165 DOI: 10.3389/fonc.2022.924740] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/07/2022] [Indexed: 11/23/2022] Open
Abstract
Nowadays, breast cancer has become the most common cancer worldwide with a high mortality rate. Immune checkpoint blockade holds great promise in tumor‐targeted therapy, and CD47 blockade as one immune therapy is undergoing various preclinical studies and clinical trials to demonstrate its safety and efficacy in breast cancer. In this review, we summarized different therapeutic mechanisms targeting CD47 and its prognostic role and therapeutic value in breast cancer.
Collapse
Affiliation(s)
- Can Chen
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Runlu Wang
- Respiratory Division, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xi Chen
- Department of Dermatology, First People’s Hospital, Huzhou, China
| | - Yulong Hou
- Department of Surgery, Huzhou Central Hospital, Huzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- *Correspondence: Jingting Jiang,
| |
Collapse
|
268
|
A Novel Prognostic Signature Revealed the Interaction of Immune Cells in Tumor Microenvironment Based on Single-Cell RNA Sequencing for Lung Adenocarcinoma. J Immunol Res 2022; 2022:6555810. [PMID: 35812244 PMCID: PMC9270162 DOI: 10.1155/2022/6555810] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/12/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022] Open
Abstract
Background The tumor immune microenvironment (TIME) played an important role in immunotherapy prognosis and treatment response. Immune cells constitute a large part of the tumor microenvironment and regulate tumor progression. Our research is dedicated to studying the infiltrating immune cell in lung adenocarcinoma (LUAD) and seeking potential targets. Methods The scRNA-seq data were collected from our FDZSH and two public datasets. The code for cell-type mapping algorithms was downloaded from the CIBERSORTx portal. The bioinformatics data of LUAD patients could be approached from The Cancer Genome Atlas (TCGA) portal. Weighted gene coexpression network analysis (WGCNA) and least absolute shrinkage and selection operator (LASSO) analyses were performed to construct a risk model. TIMER2 and TIDE helped with the immune infiltration estimation, while PROGENy helped the cancer-related pathways' enrichment analysis. GSE31210 dataset and IMVigor ICB therapy cohort validated our findings as the external validation datasets. Results We clustered the scRNA-seq dataset (integrating our FDZSH datasets and other public datasets) into 23 subpopulations. After curated cell annotation, we implemented Cibersort and WGCNA analysis to anchor the brown module and natural killer cell cluster1 due to the most relationship with tumor trait. The overlap of the brown module gene, natural killer cell signature, and DEGs of tumor and adjacent normal samples was screened by LASSO Cox regression. The obtained 5-gene risk model showed an excellent prognostic performance in the validation dataset. Furthermore, there was a correlation between risk score and tumor-infiltrating immune cells and tumor genomics abnormity. Patients with higher risk scores had a significantly lower immunotherapy response rate. Conclusion Our observations implied that immune cells played a pivotal role in TIME and established a 5-gene signature (including IDH2, ADRB2, SFTPC, CCDC69, and CCND2) on the basement of nature killer markers targeted by WGCNA analysis. The significance of clinical outcome and immunotherapy response prediction was validated robustly.
Collapse
|
269
|
Kashiwagi S, Asano Y, Takada K, Goto W, Kouhashi R, Yabumoto A, Tauchi Y, Morisaki T, Ogisawa K, Shibutani M, Tanaka H, Ohira M. Validation of the Optimum Timing of Assessment of Tumor Infiltrating Lymphocytes During Preoperative Chemotherapy for Breast Cancer. CANCER DIAGNOSIS & PROGNOSIS 2022; 2:443-451. [PMID: 35813011 PMCID: PMC9254105 DOI: 10.21873/cdp.10127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND/AIM Tumor microenvironment (TME) assessment is considered to play an important role in the prediction of prognosis and therapeutic response following breast cancer treatment. No consensus has been reached regarding evaluation methods despite reports on the utilization of tumor-infiltrating lymphocytes (TILs) for immune TME (iTME) monitoring. Optimum timing of iTME assessment has not yet been established. PATIENTS AND METHODS Two hundred thirty-nine patients were treated with neoadjuvant chemotherapy (NAC). During the period from diagnostic needle biopsy to NAC initiation for breast cancer, the optimal evaluation timing was examined using a receiver operating characteristic (ROC) curve analysis. RESULTS A significant correlation between TILs and pathological complete response (pCR) was only observed in the short-term group (≤35 days) (p=0.033). Prognostic analysis revealed that in the short-term group, patients with high TIL levels had a significantly better survival prognosis relative to those with low TIL levels (>35 days) [disease-free survival (DFS): p=0.001, overall survival (OS): p=0.021]. TILs were identified as an independent factor affecting DFS in a multivariate analysis (p=0.008, hazard ratio=0.130). CONCLUSION TIL assessment during NAC for breast cancer is a prognostic predictor only when performed at ≤35 days before NAC initiation.
Collapse
Affiliation(s)
- Shinichiro Kashiwagi
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yuka Asano
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Koji Takada
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Wataru Goto
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Rika Kouhashi
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Akimichi Yabumoto
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yukie Tauchi
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tamami Morisaki
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kana Ogisawa
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masatsune Shibutani
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Tanaka
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaichi Ohira
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
270
|
Sui Y, Li J, Qu J, Fang T, Zhang H, Zhang J, Wang Z, Xia M, Dai Y, Wang D. Dual-Responsive Nanovaccine for Cytosolic Delivery of Antigens to Boost Cellular Immune Responses and Cancer Immunotherapy. Asian J Pharm Sci 2022; 17:583-595. [PMID: 36101894 PMCID: PMC9459061 DOI: 10.1016/j.ajps.2022.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Yang Sui
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ji Li
- Department of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiqiang Qu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ting Fang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Hongyan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jian Zhang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zheran Wang
- Department of Mathematics and Statistics, Auburn University, Auburn, AL 36849, USA
- Corresponding authors.
| | - Mingyu Xia
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding authors.
| | - Yinghui Dai
- Department of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding authors.
| | - Dongkai Wang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding authors.
| |
Collapse
|
271
|
Ting NLN, Lau HCH, Yu J. Cancer pharmacomicrobiomics: targeting microbiota to optimise cancer therapy outcomes. Gut 2022; 71:1412-1425. [PMID: 35277453 PMCID: PMC9185832 DOI: 10.1136/gutjnl-2021-326264] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 02/25/2022] [Indexed: 02/06/2023]
Abstract
Despite the promising advances in novel cancer therapy such as immune checkpoint inhibitors (ICIs), limitations including therapeutic resistance and toxicity remain. In recent years, the relationship between gut microbiota and cancer has been extensively studied. Accumulating evidence reveals the role of microbiota in defining cancer therapeutic efficacy and toxicity. Unlike host genetics, microbiota can be easily modified via multiple strategies, including faecal microbiota transplantation (FMT), probiotics and antibiotics. Preclinical studies have identified the mechanisms on how microbes influence cancer treatment outcomes. Clinical trials have also demonstrated the potential of microbiota modulation in cancer treatments. Herein, we review the mechanistic insights of gut microbial interactions with chemotherapy and ICIs, particularly focusing on the interplay between gut bacteria and the pharmacokinetics (eg, metabolism, enzymatic degradation) or pharmacodynamics (eg, immunomodulation) of cancer treatment. The translational potential of basic findings in clinical settings is then explored, including using microbes as predictive biomarkers and microbial modulation by antibiotics, probiotics, prebiotics, dietary modulations and FMT. We further discuss the current limitations of gut microbiota modulation in patients with cancer and suggest essential directions for future study. In the era of personalised medicine, it is crucial to understand the microbiota and its interactions with cancer. Manipulating the gut microbiota to augment cancer therapeutic responses can provide new insights into cancer treatment.
Collapse
Affiliation(s)
- Nick Lung-Ngai Ting
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
272
|
Catanzaro E, Feron O, Skirtach AG, Krysko DV. Immunogenic Cell Death and Role of Nanomaterials Serving as Therapeutic Vaccine for Personalized Cancer Immunotherapy. Front Immunol 2022; 13:925290. [PMID: 35844506 PMCID: PMC9280641 DOI: 10.3389/fimmu.2022.925290] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/02/2022] [Indexed: 07/20/2023] Open
Abstract
Immunogenic cell death (ICD) is a rapidly growing research area representing one of the emerging therapeutic strategies of cancer immunotherapy. ICD is an umbrella term covering several cell death modalities including apoptosis, necroptosis, ferroptosis and pyroptosis, and is the product of a balanced combination of adjuvanticity (damage-associated molecular patterns and chemokines/cytokines) and antigenicity (tumor associated antigens). Only a limited number of anti-cancer therapies are available to induce ICD in experimental cancer therapies and even much less is available for clinical use. To overcome this limitation, nanomaterials can be used to increase the immunogenicity of cancer cells killed by anti-cancer therapy, which in themselves are not necessarily immunogenic. In this review, we outline the current state of knowledge of ICD modalities and discuss achievements in using nanomaterials to increase the immunogenicity of dying cancer cells. The emerging trends in modulating the immunogenicity of dying cancer cells in experimental and translational cancer therapies and the challenges facing them are described. In conclusion, nanomaterials are expected to drive further progress in their use to increase efficacy of anti-cancer therapy based on ICD induction and in the future, it is necessary to validate these strategies in clinical settings, which will be a challenging research area.
Collapse
Affiliation(s)
- Elena Catanzaro
- Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Olivier Feron
- Cancer Translational Research Laboratory, Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - André G. Skirtach
- Cancer Research Institute Ghent, Ghent, Belgium
- Nano-BioTechnology Laboratory, Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Dmitri V. Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
273
|
Zhang Y, Zhang J, Wang W, Guo X, Hou L, Zhang T, Wang B, Kou F, Huan M, He W, Zhou S, Zhang B. Eliciting an Immunostimulatory Tumor Microenvironment to Enhance the Anti‐tumor Efficacy by Targeted Cancer Immunotherapy. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Ya‐Xuan Zhang
- Department of Pharmaceutics School of Pharmacy Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine Xi'an 710032 China
- Xi'an Maternity and Child Healthcare Hospital Xi'an 710002 China
| | - Jun‐Jie Zhang
- Department of Pharmaceutics School of Pharmacy Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine Xi'an 710032 China
| | - Wei Wang
- Department of Pharmaceutics School of Pharmacy Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine Xi'an 710032 China
| | - Xiao‐Ying Guo
- College of Chemistry &Environment Engineering Baise University Baise 533000 China
| | - Li‐Shuang Hou
- Department of Pharmaceutics School of Pharmacy Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine Xi'an 710032 China
| | - Tang‐Rui Zhang
- Department of Pharmaceutics School of Pharmacy Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine Xi'an 710032 China
| | - Bao‐Long Wang
- Department of Pharmaceutics School of Pharmacy Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine Xi'an 710032 China
| | - Fang Kou
- Department of Pharmaceutics School of Pharmacy Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine Xi'an 710032 China
| | - Meng‐Lei Huan
- Department of Pharmaceutics School of Pharmacy Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine Xi'an 710032 China
| | - Wei He
- Department of Chemistry School of Pharmacy Fourth Military Medical University Xi'an 710032 China
| | - Si‐Yuan Zhou
- Department of Pharmaceutics School of Pharmacy Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine Xi'an 710032 China
| | - Bang‐Le Zhang
- Department of Pharmaceutics School of Pharmacy Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine Xi'an 710032 China
| |
Collapse
|
274
|
Sun D, Zhang A, Gao B, Zou L, Huang H, Zhao X, Xu D. Identification of Alternative Splicing-Related Genes CYB561 and FOLH1 in the Tumor-Immune Microenvironment for Endometrial Cancer Based on TCGA Data Analysis. Front Genet 2022; 13:770569. [PMID: 35836577 PMCID: PMC9274141 DOI: 10.3389/fgene.2022.770569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 06/13/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Advanced and recurrent endometrial cancer EC remains controversial. Immunotherapy will play a landmark role in cancer treatment, and alternative splicing (AS) of messenger RNA (mRNA) may offer the potential of a broadened target space.Methods: We downloaded the clinical information and mRNA expression profiles from The Cancer Genome Atlas (TCGA) database. Hub genes were extracted from 11 AS-related genes to analyze the correlation between clinical parameters and the tumor-immune microenvironment. We also analyzed the correlations between the copy numbers, gene expressions of hub genes, and immune cells. The correlation between the risk score and the six most important checkpoint genes was also investigated. The ESTIMATE algorithm was finally performed on each EC sample based on the high- and low-risk groups.Results: The risk score was a reliable and stable independent risk predictor in the Uterine Corpus Endometrial Carcinoma (UCEC) cohort. CYB561|42921|AP and FOLH1|15817|ES were extracted. The expression of CYB561 and FOLH1 decreased gradually with the increased grade and International Federation of Gynecology and Obstetrics (FIGO) stage (p < 0.05). Gene copy number changes in CYB561 and FOLH1 led to the deletion number of myeloid DC cells and T cell CD8+. Low expression of both CYB561 and FOLH1 was associated with poor prognosis (p < 0.001). The checkpoint genes, CTLA-4 and PDCD1, exhibited a negative correlation with the risk score of AS in UCEC.Conclusion: AS-related gene signatures were related to the immune-tumor microenvironment and prognosis. These outcomes were significant for studying EC’s immune-related mechanisms and exploring novel prognostic predictors and precise therapy methods.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dabao Xu
- *Correspondence: Xingping Zhao, ; Dabao Xu,
| |
Collapse
|
275
|
Xu Y, Jiang Z, Kuang X, Chen X, Liu H. Research Trends in Immune Checkpoint Blockade for Melanoma: Visualization and Bibliometric Analysis. J Med Internet Res 2022; 24:e32728. [PMID: 35759331 PMCID: PMC9274394 DOI: 10.2196/32728] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/13/2021] [Accepted: 03/26/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Melanoma is one of the most life-threatening skin cancers; immune checkpoint blockade is widely used in the treatment of melanoma because of its remarkable efficacy. OBJECTIVE This study aimed to conduct a comprehensive bibliometric analysis of research conducted in recent decades on immune checkpoint blockade for melanoma, while exploring research trends and public interest in this topic. METHODS We summarized the articles in the Web of Science Core Collection on immune checkpoint blockade for melanoma in each year from 1999 to 2020. The R package bibliometrix was used for data extraction and visualization of the distribution of publication year and the top 10 core authors. Keyword citation burst analysis and cocitation networks were calculated with CiteSpace. A Gunn online world map was used to evaluate distribution by country and region. Ranking was performed using the Standard Competition Ranking method. Coauthorship analysis and co-occurrence were analyzed and visualized with VOSviewer. RESULTS After removing duplicates, a total of 9169 publications were included. The distribution of publications by year showed that the number of publications rose sharply from 2015 onwards and either reached a peak in 2020 or has yet to reach a peak. The geographical distribution indicated that there was a large gap between the number of publications in the United States and other countries. The coauthorship analysis showed that the 149 top institutions were grouped into 8 clusters, each covering approximately a single country, suggesting that international cooperation among institutions should be strengthened. The core author extraction revealed changes in the most prolific authors. The keyword analysis revealed clustering and top citation bursts. The cocitation analysis of references from 2010 to 2020 revealed the number of citations and the centrality of the top articles. CONCLUSIONS This study revealed trends in research and public interest in immune checkpoint blockade for melanoma. Our findings suggest that the field is growing rapidly, has several core authors, and that the United States is taking the lead position. Moreover, cooperation between countries should be strengthened, and future research hot spots might focus on deeper exploration of drug mechanisms, prediction of treatment efficacy, prediction of adverse events, and new modes of administration, such as combination therapy, which may pave the way for further research.
Collapse
Affiliation(s)
- Yantao Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zixi Jiang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xinwei Kuang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
276
|
Nie W, Chen J, Wang B, Gao X. Nonviral vector system for cancer immunogene therapy. MEDCOMM – BIOMATERIALS AND APPLICATIONS 2022. [DOI: 10.1002/mba2.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Wen Nie
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu PR China
| | - Jing Chen
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu PR China
| | - Bilan Wang
- Department of Pharmacy West China Second University Hospital of Sichuan University Chengdu PR China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu PR China
| |
Collapse
|
277
|
Priesner C, Hildebrandt M. Advanced Therapy Medicinal Products and the Changing Role of Academia. Transfus Med Hemother 2022; 49:158-162. [PMID: 35813600 PMCID: PMC9209977 DOI: 10.1159/000524392] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/21/2022] [Indexed: 09/22/2023] Open
Abstract
Academic institutions coin the ATMP landscape but do not possess an industry-like capacity to vigorously pursue the full developmental pathway to marketing authorization. At the same time, industry has fostered clinical trials with ATMPs, brought the first products to marketing authorization, and defined novel modes of interaction with academia. A regulatory niche for local manufacturing of ATMPs within an academic institution had been foreseen in Regulation (EU) 1394/2007 under the term "Hospital Exemption" but remained ill-defined. Manufacture in close proximity to the patient is difficult to accomplish, as "point of care" systems for the manufacture of ATMPs have encountered regulatory challenges hovering between process and product. The efforts and costs for the development of ATMPs continue to be dramatically underestimated, and few academic centers were persistent enough to invest in the GMP infrastructure needed and to recruit personnel trained in ATMP development. As a consequence, the contribution by hospitals to ATMP development has shifted from the finished ATMP toward the procurement of starting materials, selected manufacturing steps, storage of the product, clinical application, and participation in clinical trials. As the development and use of cell-based therapies and ATMPs continue to attract and challenge clinicians and scientists, this review aims to discuss logistical, financial, and regulatory issues that might contribute to the changing role of Academia in ATMP development, with an outlook into possible developments in the future and proposals for ways to reshape the academic environment under the auspices of what might truly have been meant by the hospital exemption clause.
Collapse
Affiliation(s)
- Christoph Priesner
- TUMCells Interdisciplinary Center for Cellular Therapies, Technical University of Munich, München, Germany
| | - Martin Hildebrandt
- Department of Internal Medicine III, Hematology and Oncology, Technical University of Munich Medical School, München, Germany
| |
Collapse
|
278
|
Sanmamed MF, Berraondo P, Rodriguez-Ruiz ME, Melero I. Charting roadmaps towards novel and safe synergistic immunotherapy combinations. NATURE CANCER 2022; 3:665-680. [PMID: 35764745 DOI: 10.1038/s43018-022-00401-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
Checkpoint inhibitor-based cancer immunotherapy is often combined in the clinic with other immunotherapy strategies, targeted therapies, chemotherapy or standard-of-care treatments to achieve superior therapeutic efficacy. The large number of immunotherapy combinations that are currently undergoing clinical testing necessitate the establishment of faithful criteria to prioritize optimal combinations with evidence of synergy, to determine their safety and optimal sequence of administration and to identify biomarkers of therapy resistance and response. In this review, we focus on recent developments in immunotherapy combinations and reflect on how combinations should be optimized to maximize the impact of immunotherapy in clinical oncology.
Collapse
Affiliation(s)
- Miguel F Sanmamed
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Maria E Rodriguez-Ruiz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain.
| |
Collapse
|
279
|
Shang Q, Dong Y, Su Y, Leslie F, Sun M, Wang F. Local scaffold-assisted delivery of immunotherapeutic agents for improved cancer immunotherapy. Adv Drug Deliv Rev 2022; 185:114308. [PMID: 35472398 DOI: 10.1016/j.addr.2022.114308] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 12/18/2022]
Abstract
Cancer immunotherapy, which reprograms a patient's own immune system to eradicate cancer cells, has been demonstrated as a promising therapeutic strategy clinically. Immune checkpoint blockade (ICB) therapies, cytokine therapies, cancer vaccines, and chimeric antigen receptor (CAR) T cell therapies utilize immunotherapy techniques to relieve tumor immune suppression and/or activate cellular immune responses to suppress tumor growth, metastasis and recurrence. However, systemic administration is often hampered by limited drug efficacy and adverse side effects due to nonspecific tissue distribution of immunotherapeutic agents. Advancements in local scaffold-based delivery systems facilitate a controlled release of therapeutic agents into specific tissue sites through creating a local drug reservoir, providing a potent strategy to overcome previous immunotherapy limitations by improving site-specific efficacy and minimizing systemic toxicity. In this review, we summarized recent advances in local scaffold-assisted delivery of immunotherapeutic agents to reeducate the immune system, aiming to amplify anticancer efficacy and minimize immune-related adverse events. Additionally, the challenges and future perspectives of local scaffold-assisted cancer immunotherapy for clinical translation and applications are discussed.
Collapse
Affiliation(s)
- Qi Shang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Yabing Dong
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Yun Su
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21231, United States
| | - Faith Leslie
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD 21218, United States; Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, United States
| | - Mingjiao Sun
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD 21218, United States; Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, United States; Department of Ophthalmology, School of Medicine, The Johns Hopkins University, Baltimore, MD 21231, United States
| | - Feihu Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| |
Collapse
|
280
|
Cao Y, Wei D, Yang L, Luo Z, Yu P, Li H, Zhang C, Liu X, Wu F, Wu M, Zeng Y. Nanoplatform Self-Assembly from Small Molecules of Porphyrin Derivatives for NIR-II Fluorescence Imaging Guided Photothermal-Immunotherapy. Adv Healthc Mater 2022; 11:e2102526. [PMID: 35134273 DOI: 10.1002/adhm.202102526] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/21/2022] [Indexed: 01/12/2023]
Abstract
Combinatorial photothermal and immunotherapy have demonstrated great potential to remove primary tumors, suppress metastases, and prevent tumor recurrence. However, this strategy still confronts patients with many limitations, such as complex components, sophisticated construction, and inadequate therapeutic efficacy. In this work, small molecules of porphyrin derivatives (PPor) which can self-assemble into monodispersed nanoparticles without supplement of any other ingredients or surfactants are developed. The formed PPor nanoparticles (PPor NPs) exhibit highly photothermal conversion efficiency of 70% and NIR-II luminous abilities originate from the strong intramolecular charge transfer (ICT) effect of D-A structure under 808 nm laser irradiation, thus achieving NIR-II fluorescence imaging guided photothermal therapy (PTT) against primary tumors with a high cure rate. More importantly, tumor-associated antigens (TAAs), together with damage-associated molecular patterns (DAMPs) released from PTT-treated cancer cells, are proved to elicit immune responses to some degree. After combination with programmed cell death-1 (PD-1) antibodies, a robust systematic antitumor immunity is generated to restrain both primary and abscopal tumors growth, prolong survival, and prevent pulmonary metastasis on an aggressive 4T1 murine breast tumor model. Thus, this study provides a promising therapeutic paradigm with porphyrin derivatives nano-assembly as phototheranostic agents for the treatment of aggressive tumors with high efficiency.
Collapse
Affiliation(s)
- Yanbing Cao
- College of Chemical Engineering, Fuzhou University, Fuzhou, 350108, P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - De Wei
- Department of Neurosurgery, Fujian Provincial Hospital South Branch, Fuzhou, 350001, P. R. China
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, P. R. China
| | - Lixia Yang
- Key Laboratory for Green Chemical Process of the Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, P. R. China
| | - Zijin Luo
- Key Laboratory for Green Chemical Process of the Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, P. R. China
| | - Peiwen Yu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Hao Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Cao Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Xiaolong Liu
- College of Chemical Engineering, Fuzhou University, Fuzhou, 350108, P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Fengshou Wu
- Key Laboratory for Green Chemical Process of the Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, P. R. China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, P. R. China
| |
Collapse
|
281
|
Xu ZH, Wang WQ, Lou WH, Liu L. Insight of pancreatic cancer: recommendations for improving its therapeutic efficacy in the next decade. JOURNAL OF PANCREATOLOGY 2022; 5:58-68. [DOI: 10.1097/jp9.0000000000000093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Pancreatic cancer is one of the most malignant digestive system tumors. The effectiveness of pancreatic cancer treatment is still dismal, and the 5-year survival rate is only about 10%. Further improving the diagnosis and treatment of pancreatic cancer is the top priority of oncology research and clinical practice. Based on the existing clinical and scientific research experience, the review provides insight into the hotspots and future directions for pancreatic cancer, which focuses on early detection, early diagnosis, molecular typing and precise treatment, new drug development and regimen combination, immunotherapy, database development, model establishment, surgical technology and strategy change, as well as innovation of traditional Chinese medicine and breakthrough of treatment concept.
Collapse
Affiliation(s)
- Zhi-Hang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Quan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Hui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
282
|
Guo B, Zang Y. A Bayesian phase I/II biomarker-based design for identifying subgroup-specific optimal dose for immunotherapy. Stat Methods Med Res 2022; 31:1104-1119. [PMID: 35191780 PMCID: PMC9305985 DOI: 10.1177/09622802221080753] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immunotherapy is an innovative treatment that enlists the patient's immune system to battle tumors. The optimal dose for treating patients with an immunotherapeutic agent may differ according to their biomarker status. In this article, we propose a biomarker-based phase I/II dose-finding design for identifying subgroup-specific optimal dose for immunotherapy (BSOI) that jointly models the immune response, toxicity, and efficacy outcomes. We propose parsimonious yet flexible models to borrow information across different types of outcomes and subgroups. We quantify the desirability of the dose using a utility function and adopt a two-stage dose-finding algorithm to find the optimal dose for each subgroup. Simulation studies show that the BSOI design has desirable operating characteristics in selecting the subgroup-specific optimal doses and allocating patients to those optimal doses, and outperforms conventional designs.
Collapse
Affiliation(s)
- Beibei Guo
- Department of Experimental Statistics, 5779Louisiana State University, Baton Rouge, USA
| | - Yong Zang
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University, Indianapolis, USA
| |
Collapse
|
283
|
Nong C, Guan P, Li L, Zhang H, Hu H. Tumor immunotherapy: Mechanisms and clinical applications. MEDCOMM – ONCOLOGY 2022. [DOI: 10.1002/mog2.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Cheng Nong
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Pengbo Guan
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Li Li
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Huiyuan Zhang
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Hongbo Hu
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
- Chongqing International Institution for Immunology Chongqing China
| |
Collapse
|
284
|
Tomita Y, Goto Y, Sakata S, Imamura K, Minemura A, Oka K, Hayashi A, Jodai T, Akaike K, Anai M, Hamada S, Iyama S, Saruwatari K, Saeki S, Takahashi M, Ikeda T, Sakagami T. Clostridium butyricum therapy restores the decreased efficacy of immune checkpoint blockade in lung cancer patients receiving proton pump inhibitors. Oncoimmunology 2022; 11:2081010. [PMID: 35655708 PMCID: PMC9154751 DOI: 10.1080/2162402x.2022.2081010] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yusuke Tomita
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiko Goto
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shinya Sakata
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kosuke Imamura
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Ayaka Minemura
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Kentaro Oka
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Atsushi Hayashi
- R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Takayuki Jodai
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kimitaka Akaike
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Moriyasu Anai
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shohei Hamada
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shinji Iyama
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichi Saruwatari
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Sho Saeki
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Tokunori Ikeda
- Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Takuro Sakagami
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
285
|
Çuburu N, Finn OJ, Van Der Burg SH. Editorial: Cancer Prevention: Targeting Premalignant Epithelial Neoplasms in the Era of Cancer Immunotherapy and Vaccines. Front Immunol 2022; 13:924099. [PMID: 35686125 PMCID: PMC9171359 DOI: 10.3389/fimmu.2022.924099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/11/2022] [Indexed: 11/21/2022] Open
Affiliation(s)
- Nicolas Çuburu
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute (NIH), Bethesda, MA, United States
| | - Olivera J. Finn
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | | |
Collapse
|
286
|
Abdel Sater AH, Bouferraa Y, Amhaz G, Haibe Y, Lakkiss AE, Shamseddine A. From Tumor Cells to Endothelium and Gut Microbiome: A Complex Interaction Favoring the Metastasis Cascade. Front Oncol 2022; 12:804983. [PMID: 35600385 PMCID: PMC9117727 DOI: 10.3389/fonc.2022.804983] [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: 10/29/2021] [Accepted: 04/12/2022] [Indexed: 11/30/2022] Open
Abstract
Metastasis is a complicated process through which tumor cells disseminate to distant organs and adapt to novel tumor microenvironments. This multi-step cascade relies on the accumulation of genetic and epigenetic alterations within the tumor cells as well as the surrounding non-tumor stromal cells. Endothelial cells constitute a major player in promoting metastasis formation either by inducing the growth of tumor cells or by directing them towards dissemination in the blood or lymph. In fact, the direct and indirect interactions between tumor and endothelial cells were shown to activate several mechanisms allowing cancer cells’ invasion and extravasation. On the other side, gastrointestinal cancer development was shown to be associated with the disruption of the gut microbiome. While several proposed mechanisms have been investigated in this regard, gut and tumor-associated microbiota were shown to impact the gut endothelial barrier, increasing the dissemination of bacteria through the systemic circulation. This bacterial dislocation allows the formation of an inflammatory premetastatic niche in the distant organs promoting the metastatic cascade of primary tumors. In this review, we discuss the role of the endothelial cells in the metastatic cascade of tumors. We will focus on the role of the gut vascular barrier in the regulation metastasis. We will also discuss the interaction between this vascular barrier and the gut microbiota enhancing the process of metastasis. In addition, we will try to elucidate the different mechanisms through which this bacterial dislocation prepares the favorable metastatic niche at distant organs allowing the dissemination and successful deposition of tumor cells in the new microenvironments. Finally, and given the promising results of the studies combining immune checkpoint inhibitors with either microbiota alterations or anti-angiogenic therapy in many types of cancer, we will elaborate in this review the complex interaction between these 3 factors and their possible therapeutic combination to optimize response to treatment.
Collapse
Affiliation(s)
- Ali H Abdel Sater
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Youssef Bouferraa
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ghid Amhaz
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yolla Haibe
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ahmed El Lakkiss
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Shamseddine
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
287
|
González-Navarro EA, Español M, Egri N, Castellà M, Calderón H, España C, Guijarro C, Heredia L, Pascal M, Juan Otero M. What will (and should) be improved in CAR immunotherapy? INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 370:149-161. [PMID: 35798504 DOI: 10.1016/bs.ircmb.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Chimeric antigen receptor (CAR) is probably one of the most successful proposals for cancer treatment, especially hematological diseases for which several Advanced Therapies Medicinal Products (ATMP) have been approved worldwide by drug agencies. But, despite this unprecedented success in the oncology and cell/gene therapy fields, there are a lot of aspects that could (and should) be improved in the multiple aspects that involve this complex therapy: from the design of the chimeric molecule to the clinical protocols of use of the engineered T-cells, including even the regulatory rules that they are currently restricting the development of these hopeful therapies. In this chapter, we will try to summarize the main aspects that can (and probably should) be improved for the expansion of immunotherapy with CAR proposals beyond onco-hematology.
Collapse
Affiliation(s)
- Europa Azucena González-Navarro
- Servei d'Immunologia. Hospital Clínic de Barcelona. IDIBAPS. Universitat de Barcelona. Plataforma d'Immunoteràpia Hospital Clínic-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Marta Español
- Servei d'Immunologia. Hospital Clínic de Barcelona. IDIBAPS. Universitat de Barcelona. Plataforma d'Immunoteràpia Hospital Clínic-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Natalia Egri
- Servei d'Immunologia. Hospital Clínic de Barcelona. IDIBAPS. Universitat de Barcelona. Plataforma d'Immunoteràpia Hospital Clínic-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Maria Castellà
- Servei d'Immunologia. Hospital Clínic de Barcelona. IDIBAPS. Universitat de Barcelona. Plataforma d'Immunoteràpia Hospital Clínic-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Hugo Calderón
- Servei d'Immunologia. Hospital Clínic de Barcelona. IDIBAPS. Universitat de Barcelona. Plataforma d'Immunoteràpia Hospital Clínic-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Carolina España
- Servei d'Immunologia. Hospital Clínic de Barcelona. IDIBAPS. Universitat de Barcelona. Plataforma d'Immunoteràpia Hospital Clínic-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Carla Guijarro
- Servei d'Immunologia. Hospital Clínic de Barcelona. IDIBAPS. Universitat de Barcelona. Plataforma d'Immunoteràpia Hospital Clínic-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Libertad Heredia
- Servei d'Immunologia. Hospital Clínic de Barcelona. IDIBAPS. Universitat de Barcelona. Plataforma d'Immunoteràpia Hospital Clínic-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Mariona Pascal
- Servei d'Immunologia. Hospital Clínic de Barcelona. IDIBAPS. Universitat de Barcelona. Plataforma d'Immunoteràpia Hospital Clínic-Hospital Sant Joan de Déu, Barcelona, Spain
| | - Manel Juan Otero
- Servei d'Immunologia. Hospital Clínic de Barcelona. IDIBAPS. Universitat de Barcelona. Plataforma d'Immunoteràpia Hospital Clínic-Hospital Sant Joan de Déu, Barcelona, Spain.
| |
Collapse
|
288
|
Zhou Z, Pang Y, Sun W. 淋巴结-肿瘤双靶向型外泌体: 双效协同肿瘤免疫治疗. CHINESE SCIENCE BULLETIN-CHINESE 2022. [DOI: 10.1360/tb-2022-0385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
289
|
Xiao M, Lai W, Yao X, Pei H, Fan C, Li L. Programming Receptor Clustering with DNA Probabilistic Circuits for Enhanced Natural Killer Cell Recognition. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202203800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Mingshu Xiao
- East China Normal University School of Chemistry and Molecular Engineering 500 Dongchuan Road 200241 Shanghai CHINA
| | - Wei Lai
- East China Normal University School of Chemistry and Molecular Engineering 500 Dongchuan Road 200241 Shanghai CHINA
| | - Xiaowei Yao
- East China Normal University School of Chemistry and Molecular Engineering 500 Dongchuan Road 200241 Shanghai CHINA
| | - Hao Pei
- East China Normal University School of Chemistry and Molecular Engineering 500 Dongchuan Road 200241 Shanghai CHINA
| | - Chunhai Fan
- Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital School of Chemistry and Chemical Engineering, Institute of Molecular Medicine 800 Dongchuan Road 200240 Shanghai CHINA
| | - Li Li
- East China Normal University School of Chemistry and Molecular Engineering No. 500 Dongchuan Road 200241 Shanghai CHINA
| |
Collapse
|
290
|
Zhao Y, Xu H, Zhang M, Li L. Single-Cell RNA-Seq and Bulk RNA-Seq Reveal Intratumoral Heterogeneity and Tumor Microenvironment Characteristics in Diffuse Large B-Cell Lymphoma. Front Genet 2022; 13:881345. [PMID: 35601491 PMCID: PMC9116505 DOI: 10.3389/fgene.2022.881345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/22/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Diffuse large B-cell lymphoma (DLBCL) is the most common histologic subtype of non-Hodgkin’s lymphoma (NHL) with highly heterogeneous genetic and phenotypic features. Therefore, a comprehensive understanding of cellular diversity and intratumoral heterogeneity is essential to elucidate the mechanisms driving DLBCL progression and to develop new therapeutic approaches. Methods: We analyzed single-cell transcriptomic data from 2 reactive lymph node tissue samples and 2 DLBCL lymph node biopsy tissue samples to explore the transcriptomic landscape of DLBCL. In addition, we constructed a prognostic model based on the genes obtained from differential analysis. Results: Based on gene expression profiles at the single cell level, we identified and characterized different subpopulations of malignant and immune cells. Malignant cells exhibited a high degree of inter-tumor heterogeneity. Tumor-infiltrating regulatory CD4+ T cells showed highly immunosuppressive properties and exhausted cytotoxic CD8+ T cells were highly expressed with markers of exhaustion. Cell communication analysis identified complex interactions between malignant cells and other cell subpopulations. In addition, the prognostic model we constructed allows for monitoring the prognosis of DLBCL patients. Conclusion: This study provides an in-depth dissection of the transcriptional features of malignant B cells and tumor microenvironment (TME) in DLBCL and provides new insights into the tumor heterogeneity of DLBCL.
Collapse
|
291
|
Bôle-Richard E, Pemmaraju N, Caël B, Daguindau E, Lane AA. CD123 and More: How to Target the Cell Surface of Blastic Plasmacytoid Dendritic Cell Neoplasm. Cancers (Basel) 2022; 14:2287. [PMID: 35565416 PMCID: PMC9099711 DOI: 10.3390/cancers14092287] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and aggressive leukemia derived from plasmacytoid dendritic cells (pDCs). It is associated with a remarkably poor prognosis and unmet need for better therapies. Recently, the first-in-class CD123-targeting therapy, tagraxofusp, was approved for treatment of BPDCN. Other CD123-targeting strategies are in development, including bispecific antibodies and combination approaches with tagraxofusp and other novel agents. In other blood cancers, adoptive T-cell therapy using chimeric antigen receptor (CAR)-modified T cells represents a promising new avenue in immunotherapy, showing durable remissions in some relapsed hematologic malignancies. Here, we report on novel and innovative therapies in development to target surface molecules in BPDCN currently in clinical trials or in preclinical stages. We also discuss new cell surface targets that may have implications for future BPDCN treatment.
Collapse
Affiliation(s)
- Elodie Bôle-Richard
- INSERM, EFS BFC, UMR1098, RIGHT, University of Bourgogne Franche-Comté, Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besancon, France; (B.C.); (E.D.)
| | - Naveen Pemmaraju
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Blandine Caël
- INSERM, EFS BFC, UMR1098, RIGHT, University of Bourgogne Franche-Comté, Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besancon, France; (B.C.); (E.D.)
| | - Etienne Daguindau
- INSERM, EFS BFC, UMR1098, RIGHT, University of Bourgogne Franche-Comté, Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besancon, France; (B.C.); (E.D.)
- Service Hématologie, CHU Besançon, F-25000 Besancon, France
| | - Andrew A. Lane
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
292
|
Lewin TD, Avignon B, Tovaglieri A, Cabon L, Gjorevski N, Hutchinson LG. An in silico Model of T Cell Infiltration Dynamics Based on an Advanced in vitro System to Enhance Preclinical Decision Making in Cancer Immunotherapy. Front Pharmacol 2022; 13:837261. [PMID: 35586042 PMCID: PMC9108393 DOI: 10.3389/fphar.2022.837261] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/11/2022] [Indexed: 01/04/2023] Open
Abstract
Cancer immunotherapy often involves the use of engineered molecules to selectively bind and activate T cells located within tumour tissue. Fundamental to the success of such treatments is the presence or recruitment of T cells localised within the tumour microenvironment. Advanced organ-on-a-chip systems provide an in vitro setting in which to investigate how novel molecules influence the spatiotemporal dynamics of T cell infiltration into tissue, both in the context of anti-tumour efficacy and off-tumour toxicity. While highly promising, the complexity of these systems is such that mathematical modelling plays a crucial role in the quantitative evaluation of experimental results and maximising the mechanistic insight derived. We develop a mechanistic, mathematical model of a novel microphysiological in vitro platform that recapitulates T cell infiltration into epithelial tissue, which may be normal or transformed. The mathematical model describes the spatiotemporal dynamics of infiltrating T cells in response to chemotactic cytokine signalling. We integrate the model with dynamic imaging data to optimise key model parameters. The mathematical model demonstrates a good fit to the observed experimental data and accurately describes the distribution of infiltrating T cells. This model is designed to complement the in vitro system; with the potential to elucidate complex biological mechanisms, including the mode of action of novel therapies and the drivers of safety events, and, ultimately, improve the efficacy-safety profile of T cell-targeted cancer immunotherapies.
Collapse
|
293
|
The interplay between anticancer challenges and the microbial communities from the gut. Eur J Clin Microbiol Infect Dis 2022; 41:691-711. [PMID: 35353280 DOI: 10.1007/s10096-022-04435-2] [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: 12/22/2021] [Accepted: 03/15/2022] [Indexed: 11/03/2022]
Abstract
Cancer being an increasing burden on human health, the use of anticancer drugs has risen over the last decades. The physiological effects of these drugs are not only perceived by the host's cells but also by the microbial cells it harbors as commensals, notably the gut microbiota. Since the early '50 s, the cytotoxicity of anticancer chemotherapy was evaluated on bacteria revealing some antimicrobial activities that result in an established perturbation of the gut microbiota. This perturbation can affect the host's health through dysbiosis, which can lead to multiple complications, but has also been shown to have a direct effect on the treatment efficiency.We, therefore, conducted a review of literature focusing on this triangular relationship involving the microbial communities from the gut, the host's disease, and the anticancer treatment. We focused specifically on the antimicrobial effects of anticancer chemotherapy, their impact on mutagenesis in bacteria, and the perspectives of using bacteria-based tools to help in the diagnostic and treatment of cancer.
Collapse
|
294
|
Romero PJ, Gulley JL, Hwu P, Dean M, Million-Weaver S. Celebrating a decade of the Journal for ImmunoTherapy of Cancer. J Immunother Cancer 2022; 10:jitc-2022-005207. [PMID: 35618287 PMCID: PMC9125750 DOI: 10.1136/jitc-2022-005207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- Pedro J Romero
- Ludwig Institute for Cancer Research, Epalinges, Switzerland
| | - James L Gulley
- NCI, National Institutes of Health, Bethesda, Maryland, USA
| | - Patrick Hwu
- Administration, Moffitt Cancer Center, Tampa, Florida, USA
| | - Mary Dean
- Society for Immunotherapy of Cancer, Milwaukee, Wisconsin, USA
| | | |
Collapse
|
295
|
Liu C, Liu X, Xiang X, Pang X, Chen S, Zhang Y, Ren E, Zhang L, Liu X, Lv P, Wang X, Luo W, Xia N, Chen X, Liu G. A nanovaccine for antigen self-presentation and immunosuppression reversal as a personalized cancer immunotherapy strategy. NATURE NANOTECHNOLOGY 2022; 17:531-540. [PMID: 35410368 DOI: 10.1038/s41565-022-01098-0] [Citation(s) in RCA: 192] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/10/2022] [Indexed: 05/23/2023]
Abstract
The strategy of combining a vaccine with immune checkpoint inhibitors has been widely investigated in cancer management, but the complete response rate for this strategy is still unresolved. We describe a genetically engineered cell membrane nanovesicle that integrates antigen self-presentation and immunosuppression reversal (ASPIRE) for cancer immunotherapy. The ASPIRE nanovaccine is derived from recombinant adenovirus-infected dendritic cells in which specific peptide-major histocompatibility complex class I (pMHC-I), anti-PD1 antibody and B7 co-stimulatory molecules are simultaneously anchored by a programmed process. ASPIRE can markedly improve antigen delivery to lymphoid organs and generate broad-spectrum T-cell responses that eliminate established tumours. This work presents a powerful vaccine formula that can directly activate both native T cells and exhausted T cells, and suggests a general strategy for personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Chao Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Xue Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Xinchu Xiang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Xin Pang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Siyuan Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Yunming Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - En Ren
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Lili Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Xuan Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Peng Lv
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Xiaoyong Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Wenxin Luo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Clinical Imaging Research Centre, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, Singapore.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China.
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China.
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China.
| |
Collapse
|
296
|
Feng Z, He X, Zhang X, Wu Y, Xing B, Knowles A, Shan Q, Miller S, Hojnacki T, Ma J, Katona BW, Gade TPF, Siegel DL, Schrader J, Metz DC, June CH, Hua X. Potent suppression of neuroendocrine tumors and gastrointestinal cancers by CDH17CAR T cells without toxicity to normal tissues. NATURE CANCER 2022; 3:581-594. [PMID: 35314826 DOI: 10.1038/s43018-022-00344-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 02/09/2022] [Indexed: 12/15/2022]
Abstract
Gastrointestinal cancers (GICs) and neuroendocrine tumors (NETs) are often refractory to therapy after metastasis. Adoptive cell therapy using chimeric antigen receptor (CAR) T cells, though remarkably efficacious for treating leukemia, is yet to be developed for solid tumors such as GICs and NETs. Here we isolated a llama-derived nanobody, VHH1, and found that it bound cell surface adhesion protein CDH17 upregulated in GICs and NETs. VHH1-CAR T cells (CDH17CARTs) killed both human and mouse tumor cells in a CDH17-dependent manner. CDH17CARTs eradicated CDH17-expressing NETs and gastric, pancreatic and colorectal cancers in either tumor xenograft or autochthonous mouse models. Notably, CDH17CARTs do not attack normal intestinal epithelial cells, which also express CDH17, to cause toxicity, likely because CDH17 is localized only at the tight junction between normal intestinal epithelial cells. Thus, CDH17 represents a class of previously unappreciated tumor-associated antigens that is 'masked' in healthy tissues from attack by CAR T cells for developing safer cancer immunotherapy.
Collapse
Affiliation(s)
- Zijie Feng
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Xin He
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Xuyao Zhang
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yuan Wu
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Bowen Xing
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Alison Knowles
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Qiaonan Shan
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel Miller
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Taylor Hojnacki
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jian Ma
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Bryson W Katona
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Division of Gastroenterology and Hepatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Terence P F Gade
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Don L Siegel
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jörg Schrader
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David C Metz
- Division of Gastroenterology and Hepatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Xianxin Hua
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
297
|
Chen W, Huang Y, Pan W, Xu M, Chen L. Strategies for developing PD-1 inhibitors and future directions. Biochem Pharmacol 2022; 202:115113. [DOI: 10.1016/j.bcp.2022.115113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023]
|
298
|
Soldierer M, Bister A, Haist C, Thivakaran A, Cengiz SC, Sendker S, Bartels N, Thomitzek A, Smorra D, Hejazi M, Uhrberg M, Scheckenbach K, Monzel C, Wiek C, Reinhardt D, Niktoreh N, Hanenberg H. Genetic Engineering and Enrichment of Human NK Cells for CAR-Enhanced Immunotherapy of Hematological Malignancies. Front Immunol 2022; 13:847008. [PMID: 35464442 PMCID: PMC9022481 DOI: 10.3389/fimmu.2022.847008] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 01/11/2023] Open
Abstract
The great clinical success of chimeric antigen receptor (CAR) T cells has unlocked new levels of immunotherapy for hematological malignancies. Genetically modifying natural killer (NK) cells as alternative CAR immune effector cells is also highly promising, as NK cells can be transplanted across HLA barriers without causing graft-versus-host disease. Therefore, off-the-shelf usage of CAR NK cell products might allow to widely expand the clinical indications and to limit the costs of treatment per patient. However, in contrast to T cells, manufacturing suitable CAR NK cell products is challenging, as standard techniques for genetically engineering NK cells are still being defined. In this study, we have established optimal lentiviral transduction of primary human NK cells by systematically testing different internal promoters for lentiviral CAR vectors and comparing lentiviral pseudotypes and viral entry enhancers. We have additionally modified CAR constructs recognizing standard target antigens for acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) therapy—CD19, CD33, and CD123—to harbor a CD34-derived hinge region that allows efficient detection of transduced NK cells in vitro and in vivo and also facilitates CD34 microbead-assisted selection of CAR NK cell products to >95% purity for potential clinical usage. Importantly, as most leukemic blasts are a priori immunogenic for activated primary human NK cells, we developed an in vitro system that blocks the activating receptors NKG2D, DNAM-1, NKp30, NKp44, NKp46, and NKp80 on these cells and therefore allows systematic testing of the specific killing of CAR NK cells against ALL and AML cell lines and primary AML blasts. Finally, we evaluated in an ALL xenotransplantation model in NOD/SCID-gamma (NSG) mice whether human CD19 CAR NK cells directed against the CD19+ blasts are relying on soluble or membrane-bound IL15 production for NK cell persistence and also in vivo leukemia control. Hence, our study provides important insights into the generation of pure and highly active allogeneic CAR NK cells, thereby advancing adoptive cellular immunotherapy with CAR NK cells for human malignancies further.
Collapse
Affiliation(s)
- Maren Soldierer
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Arthur Bister
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Corinna Haist
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Aniththa Thivakaran
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sevgi Can Cengiz
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stephanie Sendker
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nina Bartels
- Department of Experimental Medical Physics, Heinrich Heine University, Düsseldorf, Germany
| | - Antonia Thomitzek
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Denise Smorra
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Maryam Hejazi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Kathrin Scheckenbach
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Cornelia Monzel
- Department of Experimental Medical Physics, Heinrich Heine University, Düsseldorf, Germany
| | - Constanze Wiek
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Dirk Reinhardt
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Naghmeh Niktoreh
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Helmut Hanenberg
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
299
|
Brennan L, Brouwer-Visser J, Nüesch E, Karpova M, Heller A, Gaire F, Schneider M, Gomes B, Korski K. T-Cell Heterogeneity in Baseline Tumor Samples: Implications for Early Clinical Trial Design and Analysis. Front Immunol 2022; 13:760763. [PMID: 35558070 PMCID: PMC9086966 DOI: 10.3389/fimmu.2022.760763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background In early stage clinical trials, changes to levels of tumor infiltrating lymphocytes (TILs) in the tumor microenvironment (TME) are critical biomarkers of the mechanism of action of novel immunotherapies. However, baseline heterogeneity of tumor samples, both between and within patients, and the resultant impact on the validity of clinical trial data is not well defined. Here we identify and quantify the impact of baseline variables on the heterogeneity of FoxP3+ and proliferating CD8+ T-cells levels (MKi67+CD8A+) in the TME both between and within patients for the purpose of informing clinical trial design and analysis. Methods We compared levels of FoxP3+ and MKi67+CD8+ cell densities (counts/mm2) from >1000 baseline tumor samples from clinical trials and commercially available sources. Using multivariate hierarchical regression techniques, we investigated whether inter-person heterogeneity of activated or regulatory T-cells could be attributed to baseline characteristics including demographics, indication, lesion type, tissue of excision, biopsy method, prior cancer treatment, and tissue type i.e., "fresh" or "archival" status. We also sought to characterize within-patient heterogeneity by lesion type and tissue type. Results Prior cancer treatment with hormone therapy or chemotherapy that induces immunogenic cell death may alter the TME. Archival tissue is an unreliable substitute for fresh tissue for determining baseline TIL levels. Baseline and on treatment biopsies should be matched by lesion type to avoid bias.
Collapse
Affiliation(s)
- Laura Brennan
- Roche Pharma Research and Early Development, Early Biomarker Development Oncology, Roche Innovation Center New York, Little Falls, NJ, United States
| | - Jurriaan Brouwer-Visser
- Roche Pharma Research and Early Development, Early Biomarker Development Oncology, Roche Innovation Center New York, Little Falls, NJ, United States
| | - Eveline Nüesch
- Roche Innovation Center Basel, Roche Pharma and Early Development, Basel, Switzerland
| | - Maria Karpova
- Roche Innovation Center Munich, Roche Pharma and Early Development, Penzberg, Germany
| | - Astrid Heller
- Roche Innovation Center Munich, Roche Pharma and Early Development, Penzberg, Germany
| | - Fabien Gaire
- Roche Innovation Center Munich, Roche Pharma and Early Development, Penzberg, Germany
| | - Meike Schneider
- Roche Innovation Center Basel, Roche Pharma and Early Development, Basel, Switzerland
| | - Bruno Gomes
- Roche Innovation Center Basel, Roche Pharma and Early Development, Basel, Switzerland
| | - Konstanty Korski
- Roche Innovation Center Munich, Roche Pharma and Early Development, Penzberg, Germany
| |
Collapse
|
300
|
Xiao J, Lu Y, Lu D, Chen W, Hu W, Zhao Y, Chen S. Co‐delivery of paclitaxel and
CXCL1 shRNA
via cationic polymeric micelles for synergistic therapy against ovarian cancer. POLYM INT 2022. [DOI: 10.1002/pi.6406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Jingjing Xiao
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Yingying Lu
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Deng Lu
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Wulian Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science Fudan University Shanghai 200433 PR China
| | - Weiguo Hu
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Yuqing Zhao
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Shouzhen Chen
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| |
Collapse
|