1
|
Wang L, Chen SY, Li JL, Dai J, Qin DY, He RQ, Chen G. Anti-inflammatory effects of immunotherapy in clinical treatment and its potential mechanism in alleviating sleeping disorders: A systematic bibliometric study. Hum Vaccin Immunother 2025; 21:2475601. [PMID: 40097368 PMCID: PMC11917172 DOI: 10.1080/21645515.2025.2475601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/15/2025] [Accepted: 03/02/2025] [Indexed: 03/19/2025] Open
Abstract
Sleeping disorders negatively affect cancer patient management, quality of life, and recovery. Immunotherapy, a rising cancer treatment, shows potential to improve sleep quality by reducing inflammation. This study analyzed 255 publications (2000-2024) from the Web of Science Core Collection using bibliometric methods. The US and China dominate research output, with The Mayo Clinic as a key contributor. Core topics are "immunotherapy," "quality of life," and "antibodies." Emerging keywords like "cancer," "encephalitis," and "depression" highlight a shift toward clinical psychology in treating tumors and rare diseases. It is noteworthy that with the rapid expansion of immunotherapy in cancer treatment, clinical trials have shown that it can improve sleep quality in cancer patients by reducing inflammation. As its application in cancer treatment expands, immunotherapy's potential for treating sleep disorders is promising. Future development is expected to improve sleep quality and address clinical issues, offering broad prospects for patient outcomes.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| | - Si-Yan Chen
- Day Chemotherapy Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| | - Jun-Li Li
- Day Chemotherapy Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| | - Jian Dai
- Department of Clinical Psychology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Di-Yuan Qin
- Department of Computer Science and Technology, School of Computer and Electronic Information, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| |
Collapse
|
2
|
Shentu J, Lou H, Duan S. DMB-induced GSDMD-mediated pyroptosis: a novel therapeutic strategy for enhancing anti-tumor immunity. Cell Death Discov 2024; 10:482. [PMID: 39587093 PMCID: PMC11589605 DOI: 10.1038/s41420-024-02248-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024] Open
Affiliation(s)
- Jianqiao Shentu
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Hanqi Lou
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shiwei Duan
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Xu H, Chen C, Chen L, Pan S. Pan-cancer analysis identifies the IRF family as a biomarker for survival prognosis and immunotherapy. J Cell Mol Med 2024; 28:e18084. [PMID: 38130025 PMCID: PMC10844690 DOI: 10.1111/jcmm.18084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 11/08/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
IRF family genes have been shown to be crucial in tumorigenesis and tumour immunity. However, information about the role of IRF in the systematic assessment of pan-cancer and in predicting the efficacy of tumour therapy is still unknown. In this work, we performed a systematic analysis of IRF family genes in 33 tumour samples, including expression profiles, genomics and clinical characteristics. We then applied Single-Sample Gene-Set Enrichment Analysis (ssGSEA) to calculate IRF-scores and analysed the impact of IRF-scores on tumour progression, immune infiltration and treatment efficacy. Our results showed that genomic alterations, including SNPs, CNVs and DNA methylation, can lead to dysregulation of IRFs expression in tumours and participate in regulating multiple tumorigenesis. IRF-score expression differed significantly between 12 normal and tumour samples and the impact on tumour prognosis and immune infiltration depended on tumour type. IRF expression was correlated to drug sensitivity and to the expression of immune checkpoints and immune cell infiltration, suggesting that dysregulation of IRF family expression may be a critical factor affecting tumour drug response. Our study comprehensively characterizes the genomic and clinical profile of IRFs in pan-cancer and highlights their reliability and potential value as predictive markers of oncology drug efficacy. This may provide new ideas for future personalized oncology treatment.
Collapse
Affiliation(s)
- Hua‐Guo Xu
- Department of Laboratory MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Can Chen
- Department of Laboratory MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Lin‐Yuan Chen
- Department of Laboratory MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| | - Shiyang Pan
- Department of Laboratory MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Branch of National Clinical Research Center for Laboratory MedicineNanjingChina
| |
Collapse
|
4
|
Wang Q, Jiang H, Zhang H, Lu W, Wang X, Xu W, Li J, Lv Y, Li G, Cai C, Yu G. β-Glucan-conjugated anti-PD-L1 antibody enhances antitumor efficacy in preclinical mouse models. Carbohydr Polym 2024; 324:121564. [PMID: 37985066 DOI: 10.1016/j.carbpol.2023.121564] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/10/2023] [Accepted: 11/03/2023] [Indexed: 11/22/2023]
Abstract
The use of immune checkpoint blockade (ICB) is a promising approach for clinical cancer treatment. However, most of cancer patients do not respond to anti-PD-1/PD-L1 antibody. In this study, we proposed a novel strategy of antibody-β-glucan conjugates (AGC) to enhance the antitumor immune response to ICB therapy. The AGC were constructed by conjugating an anti-PD-L1 antibody with a β-glucan via click chemistry. This design facilitates the delivery of β-glucan into the tumor microenvironment (TME). Furthermore, the bridging effect mediated by AGC can promote the interaction between tumor cells and dendritic cells (DCs), thereby enhancing immunotherapeutic benefits. In the MC38 tumor-bearing mouse model, AGC demonstrated powerful tumor suppression, achieving a tumor suppression rate of 86.7 %. Immunophenotyping, cytokine analysis, RNA sequencing, and FTY720-treated models were combined to elucidate the mechanism underlying AGC function. Compared with anti-PD-L1 antibody, AGC induced an earlier immune response, infiltration of DCs, and activation of preexisting T cells in the TME, with T cells predominantly proliferating locally rather than migrating from other organs. In conclusion, these data suggest that AGC could serve as a promising strategy to improve ICB therapy with prospects for clinical utilization.
Collapse
Affiliation(s)
- Qian Wang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Hao Jiang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao 266237, China.
| | - Hongli Zhang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Weiqiao Lu
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Xiao Wang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Wenfeng Xu
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jia Li
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Youjing Lv
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Guoyun Li
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao 266237, China
| | - Chao Cai
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao 266237, China
| | - Guangli Yu
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao 266237, China.
| |
Collapse
|
5
|
Perdyan A, Sobocki BK, Balihodzic A, Dąbrowska A, Kacperczyk J, Rutkowski J. The Effectiveness of Cancer Immune Checkpoint Inhibitor Retreatment and Rechallenge-A Systematic Review. Cancers (Basel) 2023; 15:3490. [PMID: 37444600 DOI: 10.3390/cancers15133490] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/24/2023] [Accepted: 07/02/2023] [Indexed: 07/15/2023] Open
Abstract
Despite a great success of immunotherapy in cancer treatment, a great number of patients will become resistant. This review summarizes recent reports on immune checkpoint inhibitor retreatment or rechallenge in order to overcome primary resistance. The systematic review was performed according to PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. The search was performed using PubMed, Web of Science and Scopus. In total, 31 articles were included with a total of 812 patients. There were 16 retreatment studies and 13 rechallenge studies. We identified 15 studies in which at least one parameter (overall response rate or disease control rate) improved or was stable at secondary treatment. Interval treatment, primary response to and the cause of cessation for the first immune checkpoint inhibitors seem to be promising predictors of secondary response. However, high heterogeneity of investigated cohorts and lack of reporting guidelines are limiting factors for current in-depth analysis.
Collapse
Affiliation(s)
- Adrian Perdyan
- 3P-Medicine Laboratory, Medical University of Gdansk, 80-210 Gdansk, Poland
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Bartosz Kamil Sobocki
- Student Scientific Circle of Oncology and Radiotherapy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Amar Balihodzic
- Division of Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Anna Dąbrowska
- Student Scientific Circle of Oncology and Radiotherapy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Justyna Kacperczyk
- The University Clinical Centre in Gdansk, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Jacek Rutkowski
- Department of Oncology and Radiotherapy, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
6
|
Jassas RS, Naeem N, Sadiq A, Mehmood R, Alenazi NA, Al-Rooqi MM, Mughal EU, Alsantali RI, Ahmed SA. Current status of N-, O-, S-heterocycles as potential alkaline phosphatase inhibitors: a medicinal chemistry overview. RSC Adv 2023; 13:16413-16452. [PMID: 37274413 PMCID: PMC10233329 DOI: 10.1039/d3ra01888a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/24/2023] [Indexed: 06/06/2023] Open
Abstract
Heterocycles are a class of compounds that have been found to be potent inhibitors of alkaline phosphatase (AP), an enzyme that plays a critical role in various physiological processes such as bone metabolism, cell growth and differentiation, and has been linked to several diseases such as cancer and osteoporosis. AP is a widely distributed enzyme, and its inhibition has been considered as a therapeutic strategy for the treatment of these diseases. Heterocyclic compounds have been found to inhibit AP by binding to the active site of the enzyme, thereby inhibiting its activity. Heterocyclic compounds such as imidazoles, pyrazoles, and pyridines have been found to be potent AP inhibitors and have been studied as potential therapeutics for the treatment of cancer, osteoporosis, and other diseases. However, the development of more potent and selective inhibitors that can be used as therapeutics for the treatment of various diseases is an ongoing area of research. Additionally, the study of the mechanism of action of heterocyclic AP inhibitors is an ongoing area of research, which could lead to the identification of new targets and new therapeutic strategies. The enzyme known as AP has various physiological functions and is present in multiple tissues and organs throughout the body. This article presents an overview of the different types of AP isoforms, their distribution, and physiological roles. It also discusses the structure and mechanism of AP, including the hydrolysis of phosphate groups. Furthermore, the importance of AP as a clinical marker for liver disease, bone disorders, and cancer is emphasized, as well as its use in the diagnosis of rare inherited disorders such as hypophosphatasia. The potential therapeutic applications of AP inhibitors for different diseases are also explored. The objective of this literature review is to examine the function of alkaline phosphatase in various physiological conditions and diseases, as well as analyze the structure-activity relationships of recently reported inhibitors. The present review summarizes the structure-activity relationship (SAR) of various heterocyclic compounds as AP inhibitors. The SAR studies of these compounds have revealed that the presence of a heterocyclic ring, particularly a pyridine, pyrimidine, or pyrazole ring, in the molecule is essential for inhibitory activity. Additionally, the substitution pattern and stereochemistry of the heterocyclic ring also play a crucial role in determining the potency of the inhibitor.
Collapse
Affiliation(s)
- Rabab S Jassas
- Department of Chemistry, Jamoum University College, Umm Al-Qura University Makkah 21955 Saudi Arabia
| | - Nafeesa Naeem
- Department of Chemistry, University of Gujrat Gujrat 50700 Pakistan
| | - Amina Sadiq
- Department of Chemistry, Govt. College Women University Sialkot 51300 Pakistan
| | - Rabia Mehmood
- Department of Chemistry, Govt. College Women University Sialkot 51300 Pakistan
| | - Noof A Alenazi
- Department of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University Al-kharj 11942 Saudi Arabia
| | - Munirah M Al-Rooqi
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University 21955 Makkah Saudi Arabia
| | | | - Reem I Alsantali
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University P.O. Box 11099 Taif 21944 Saudi Arabia
| | - Saleh A Ahmed
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University 21955 Makkah Saudi Arabia
| |
Collapse
|
7
|
Xiao M, Tang Q, Zeng S, Yang Q, Yang X, Tong X, Zhu G, Lei L, Li S. Emerging biomaterials for tumor immunotherapy. Biomater Res 2023; 27:47. [PMID: 37194085 DOI: 10.1186/s40824-023-00369-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/23/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND The immune system interacts with cancer cells in various intricate ways that can protect the individual from overproliferation of cancer cells; however, these interactions can also lead to malignancy. There has been a dramatic increase in the application of cancer immunotherapy in the last decade. However, low immunogenicity, poor specificity, weak presentation efficiency, and off-target side effects still limit its widespread application. Fortunately, advanced biomaterials effectively contribute immunotherapy and play an important role in cancer treatment, making it a research hotspot in the biomedical field. MAIN BODY This review discusses immunotherapies and the development of related biomaterials for application in the field. The review first summarizes the various types of tumor immunotherapy applicable in clinical practice as well as their underlying mechanisms. Further, it focuses on the types of biomaterials applied in immunotherapy and related research on metal nanomaterials, silicon nanoparticles, carbon nanotubes, polymer nanoparticles, and cell membrane nanocarriers. Moreover, we introduce the preparation and processing technologies of these biomaterials (liposomes, microspheres, microneedles, and hydrogels) and summarize their mechanisms when applied to tumor immunotherapy. Finally, we discuss future advancements and shortcomings related to the application of biomaterials in tumor immunotherapy. CONCLUSION Research on biomaterial-based tumor immunotherapy is booming; however, several challenges remain to be overcome to transition from experimental research to clinical application. Biomaterials have been optimized continuously and nanotechnology has achieved continuous progression, ensuring the development of more efficient biomaterials, thereby providing a platform and opportunity for breakthroughs in tumor immunotherapy.
Collapse
Affiliation(s)
- Minna Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shiying Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinying Tong
- Department of Hemodialysis, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
8
|
Huo CW, Gunadasa I, Gkolia P, Shackleton M, Hunn M. Immune-related adverse events are associated with therapeutic efficacy of immunotherapy in patients with melanoma brain metastases. Melanoma Res 2023; 33:58-65. [PMID: 36545922 DOI: 10.1097/cmr.0000000000000864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Immunotherapy with T-cell checkpoint inhibitors have changed the treatment landscape for patients with melanoma brain metastases (MBMs), offering increased survival compared with historical outcomes. We sought to identify clinical features associated with intracranial tumour responses or progression-free survival (PFS) in patients with MBMs treated with immunotherapy. Patients with MBMs treated with immunotherapy from August 2013 to March 2020 were identified through local databases. Melanoma disease burdens and immune-related adverse events (irAEs) were assessed retrospectively by review of patient medical records. Efficacy was evaluated by determining objective response rates (ORRs) in brain metastases using immune-Response Evaluation Criteria in Solid Tumours criteria, MBM-specific survival and overall PFS. Twenty-six patients were identified as eligible for this study. The presence and volume of extracranial metastases (ECM) were associated with a non-significant trend of reduced intracranial ORRs and PFS. Patients with irAEs, on the other hand, had significantly increased intracranial ORRs and PFS compared to those without irAEs. Severe, grade ≥3 irAEs and co-occurrence of ≥2 irAEs were also significantly associated with longer PFS. The presence and volume of ECM correlated inversely with development and severity of irAEs. We report a strong association between the development of irAEs and favourable melanoma-specific outcomes in patients with MBMs receiving immunotherapy. Contrary to previous studies, we found that co-occurrence of ECM in these patients was associated with fewer irAEs and reduced treatment efficacy.
Collapse
|
9
|
Wang Y, Yan K, Guo Y, Lu Y, Su H, Li H. IP-score correlated to endogenous tumour antigen peptide processing: A candidate clinical response score algorithm of immune checkpoint inhibitors therapy in multiple cohorts. Front Immunol 2023; 13:1085491. [PMID: 36700205 PMCID: PMC9868931 DOI: 10.3389/fimmu.2022.1085491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/13/2022] [Indexed: 01/11/2023] Open
Abstract
The processing of endogenous tumour antigen peptides was essential for anti-tumour immunity in the tumour microenvironment. A high degree of Endogenous tumour antigen peptide processing has been demonstrated to improve the prognosis of carcinoma patients. However, there is insufficient evidence to prove its effect on the clinical response to immune checkpoint inhibitor therapy. To undertake a more in-depth analysis of the effects of the aforementioned genes on immunotherapy, we constructed a gene set evaluation score system relevant to tumour endogenous antigen peptide therapy using the GSVA approach. This rating mechanism is known as IP score (IPs). Immediately afterwards, we used the TCGA pan-cancer cohorts to conduct a comprehensive analysis of 6 genes in the IPs, and the analysis results showed that these six genes were related to the proportion of CD8+ T lymphocytes in a variety of solid tumours. As a prognostic protective factor for solid tumours, patients had better prognosis outcomes in the group with high expression levels of the above genes. We analysed the differential expression of six genes between immune checkpoint inhibitor treatment response and disease progression groups using several treatment cohorts. The results revealed that after treatment with PD-1 or CTLA4 inhibitors, the expression levels of the above six genes were comparatively high in the effective group, but the expression of the signature genes was dramatically downregulated in the ICI-insensitive groups. This indicates that the 6 genes are related to the clinical response to ICI treatment. Finally, we used the GSVA method to evaluate the above signatures, and the results showed that PDCD1, CTAL4, CD274 and LAG3 were significantly higher expressed in the IPs high-expression group; therefore, based on the processing of endogenous antigenic peptides in tumours, a predictive score of clinical response to immune checkpoint inhibitor therapy composed of 6 genes(PSMB8/PSMB9/PSMB10/PSME1/PSME2/IRF1) was constructed, and the role of each independent variable in the signature in the solid tumour microenvironment and the impact on ICI treatment were comprehensively analysed. This study provides a candidate evaluation score for predicting clinical response to immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Yutao Wang
- Department of Urology, Chinese Academy of Medical Sciences, Peking Union Medical College, Peking Union Medical College Hospital, Beijing, China
| | - Kexin Yan
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Ye Guo
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yi Lu
- Department of Urology, Chinese Academy of Medical Sciences, Peking Union Medical College, Peking Union Medical College Hospital, Beijing, China
| | - Hao Su
- Department of Urology, Chinese Academy of Medical Sciences, Peking Union Medical College, Peking Union Medical College Hospital, Beijing, China
| | - Hongjun Li
- Department of Urology, Chinese Academy of Medical Sciences, Peking Union Medical College, Peking Union Medical College Hospital, Beijing, China,*Correspondence: Hongjun Li,
| |
Collapse
|
10
|
Roy R, Singh SK, Misra S. Advancements in Cancer Immunotherapies. Vaccines (Basel) 2022; 11:vaccines11010059. [PMID: 36679904 PMCID: PMC9861770 DOI: 10.3390/vaccines11010059] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Recent work has suggested involvement of the immune system in biological therapies specifically targeting tumor microenvironment. Substantial advancement in the treatment of malignant tumors utilizing immune cells, most importantly T cells that play a key role in cell-mediated immunity, have led to success in clinical trials. Therefore, this article focuses on the therapeutic approaches and developmental strategies to treat cancer. This review emphasizes the immunomodulatory response, the involvement of key tumor-infiltrating cells, the mechanistic aspects, and prognostic biomarkers. We also cover recent advancements in therapeutic strategies.
Collapse
Affiliation(s)
- Ruchi Roy
- UICentre for Drug Discovery, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
- Correspondence:
| | - Sunil Kumar Singh
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Sweta Misra
- UICentre for Drug Discovery, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
11
|
Aquino-Acevedo AN, Knochenhauer H, Castillo-Ocampo Y, Ortiz-León M, Rivera-López YA, Morales-López C, Cruz-Robles ME, Hernández-Cordero ER, Russell S, Whitaker R, Bonilla-Claudio M, Chen DT, Dutil J, Gaillard SL, Yi JS, Previs RA, Armaiz-Pena GN. Stress hormones are associated with inflammatory cytokines and attenuation of T-cell function in the ascites from patients with high grade serous ovarian cancer. Brain Behav Immun Health 2022; 26:100558. [PMID: 36439058 PMCID: PMC9694096 DOI: 10.1016/j.bbih.2022.100558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 10/18/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Mounting evidence suggests that chronic stress and subsequent distress can promote ovarian cancer progression. These altered psychological states have been linked to sustained release of stress hormones, activation of the β-adrenergic receptors in ovarian cancer cells, and induction of pro-tumoral signaling pathways. In addition, data suggest that chronic stress promotes an inflammatory landscape highlighted by increased infiltration of tumor-associated macrophages into the ovarian tumor microenvironment (TME). In ovarian cancer, ascites is a unique TME comprised of tumor, and immune cells, which secrete pro-tumoral cytokines and chemokines that modulate tumor-associated immunity. However, our knowledge about how stress hormones impact the ascites TME remains limited. We hypothesized that the ascites harbors measurable levels of stress hormones, and accumulation of these in the ascites generates a pro-tumorigenic, inflammatory, and immunosuppressive TME. We evaluated ascites samples from 49 patients with high grade serous ovarian cancer (HGSOC) and quantified cortisol and stress hormones metabolites, metanephrine (MN), and normetanephrine (NMN) in all samples. We also measured 38 individual cytokines in the ascites, including several pro-inflammatory cytokines, such as IL-6, which were positively correlated to MN or NMN levels of those samples. Conversely, we found cortisol levels were negatively correlated to several pro-inflammatory cytokines. As T-cells are integral to the TME and our analyses identified cytokines in the ascites known to modulate T-cell function, we characterized ascites-derived T-cells and assessed the impact of stress hormones on the T-cell phenotype. Our data show an altered CD4+/CD8+ T-cell ratio and a heterogeneous expression of exhaustion markers in T-cells from the ascites, while ascites-derived CD8+ T-cells exposed to epinephrine had decreased co-expression CD38 and Granzyme B. To extend these findings to animal models, we subjected ovarian cancer-bearing mice to daily restraint stress, which resulted in increased tumor growth in two models. Congruent with our human analyses, we detected corticosterone, MN, and NMN in the ascites from tumor-bearing mice, and these stress hormones correlated with several inflammatory cytokines. Moreover, daily restraint stress leads to increased CD4+PD-1+/CD8+PD-1+ T-cell ratio in the ovarian tumor microenvironment. Overall, these data highlight a role of stress hormones in the ascites TME as a driver of tumor-associated inflammation, T-cell suppression, and disease progression.
Collapse
Affiliation(s)
- Alexandra N. Aquino-Acevedo
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Hope Knochenhauer
- Department of Obstetrics and Gynecology, Division of Gynecology Oncology, School of Medicine, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Yesenia Castillo-Ocampo
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Melanie Ortiz-León
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Yadiel A. Rivera-López
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Camily Morales-López
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Melanie E. Cruz-Robles
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Elvin R. Hernández-Cordero
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
| | - Shonagh Russell
- Department of Obstetrics and Gynecology, Division of Gynecology Oncology, School of Medicine, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Regina Whitaker
- Department of Obstetrics and Gynecology, Division of Gynecology Oncology, School of Medicine, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Margarita Bonilla-Claudio
- School of Dental Medicine, Ponce Health Sciences University, Ponce, PR, USA
- Division of Cancer Biology, Ponce Research Institute, Ponce, PR, USA
| | - Dung-Tsa Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Julie Dutil
- Department of Basic Sciences, Division of Biochemistry, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
- Division of Cancer Biology, Ponce Research Institute, Ponce, PR, USA
- Division of Women's Health, Ponce Research Institute, Ponce, PR, USA
| | - Stephanie L. Gaillard
- Departments of Oncology and Gynecology and Obstetrics, John Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Division of Medical Oncology, School of Medicine, Duke University, Durham, NC, USA
| | - John S. Yi
- Department of Surgery, Division of Surgical Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - Rebecca A. Previs
- Department of Obstetrics and Gynecology, Division of Gynecology Oncology, School of Medicine, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Guillermo N. Armaiz-Pena
- Department of Basic Sciences, Division of Pharmacology, School of Medicine, Ponce Health Sciences University, Ponce, PR, USA
- Division of Cancer Biology, Ponce Research Institute, Ponce, PR, USA
- Division of Women's Health, Ponce Research Institute, Ponce, PR, USA
| |
Collapse
|
12
|
Chen S, Su X, Mo Z. KCNN4 is a Potential Biomarker for Predicting Cancer Prognosis and an Essential Molecule that Remodels Various Components in the Tumor Microenvironment: A Pan-Cancer Study. Front Mol Biosci 2022; 9:812815. [PMID: 35720112 PMCID: PMC9205469 DOI: 10.3389/fmolb.2022.812815] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
Objectives: Potassium Calcium-Activated Channel Subfamily N Member 4 (KCNN4) is a member of the KCNN family. Studies have revealed that KCNN4 is implicated in various physiological processes as well as promotes the malignant phenotypes of cancer cells. However, little is known about its associations with survival outcomes across varying cancer types. Methods: Herein, we systematically explored the prognostic value of KCNN4 in the pan-cancer dataset retrieved from multiple databases. Next, we performed correlation analysis of KCNN4 expression with tumor mutational burden (TMB) and microsatellite instability (MSI), and immune checkpoint genes (ICGs) to assess its potential as a predictor of immunotherapy efficacy. Afterwards, patients were divided into increased-risk group and decreased-risk group based on the contrasting survival outcomes in various cancer types. Furthermore, the underlying mechanisms of the distinctive effects were analyzed using ESTIMATE, CIBERSORT algorithms, and Gene Set Enrichment Analysis (GSEA) analysis. Results: KCNN4 expression levels were aberrant in transcriptomic and proteomic levels between cancer and normal control tissues in pan-cancer datasets, further survival analysis elucidated that KCNN4 expression was correlated to multiple survival data, and clinical annotations. Besides, KCNN4 expression was correlated to TMB and MSI levels in 14 types and 12 types of pan-cancers, respectively. Meanwhile, different types of cancer have specific tumor-infiltrating immune cell (TICs) profiles. Conclusions: Our results revealed that KCNN4 could be an essential biomarker for remodeling components in the tumor microenvironment (TME), and a robust indicator for predicting prognosis as well as immunotherapy response in pan-cancer patients.
Collapse
Affiliation(s)
- Shaohua Chen
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Xiaotao Su
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zengnan Mo
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- *Correspondence: Zengnan Mo,
| |
Collapse
|
13
|
Alekseenko IV, Pleshkan VV, Kuzmich AI, Kondratieva SA, Sverdlov ED. Gene-Immune Therapy of Cancer: Approaches and Problems. RUSS J GENET+ 2022; 58:491-506. [DOI: 10.1134/s1022795422040020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/10/2021] [Accepted: 10/14/2021] [Indexed: 01/05/2025]
|
14
|
Zhang X, Li H, Lv X, Hu L, Li W, Zi M, He Y. Impact of Diets on Response to Immune Checkpoint Inhibitors (ICIs) Therapy against Tumors. Life (Basel) 2022; 12:409. [PMID: 35330159 PMCID: PMC8951256 DOI: 10.3390/life12030409] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy has revolutionized the established therapeutics against tumors. As the major immunotherapy approach, immune checkpoint inhibitors (ICIs) achieved remarkable success in the treatment of malignancies. However, the clinical gains are far from universal and durable, because of the primary and secondary resistance of tumors to the therapy, or side effects induced by ICIs. There is an urgent need to find safe combinatorial strategies that enhance the response of ICIs for tumor treatment. Diets have an excellent safety profile and have been shown to play pleiotropic roles in tumor prevention, growth, invasion, and metastasis. Accumulating evidence suggests that dietary regimens bolster not only the tolerability but also the efficacy of tumor immunotherapy. In this review, we discussed the mechanisms by which tumor cells evade immune surveillance, focusing on describing the intrinsic and extrinsic mechanisms of resistance to ICIs. We also summarized the impacts of different diets and/or nutrients on the response to ICIs therapy. Combinatory treatments of ICIs therapy with optimized diet regimens own great potential to enhance the efficacy and durable response of ICIs against tumors, which should be routinely considered in clinical settings.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Clinical Nutrition, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China;
| | - Huiqin Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (H.L.); (L.H.); (M.Z.)
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Xiupeng Lv
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China;
| | - Li Hu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (H.L.); (L.H.); (M.Z.)
- Department of Geriatrics, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Wen Li
- Department of Endocrinology, The Third People’s Hospital of Yunnan Province, Kunming 650011, China;
| | - Meiting Zi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (H.L.); (L.H.); (M.Z.)
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Yonghan He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China; (H.L.); (L.H.); (M.Z.)
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| |
Collapse
|
15
|
Liu M, Gao Y, Yuan Y, Shi S, Wu J, Tian J, Zhang J. An evidence mapping and scientometric analysis of the top-100 most cited clinical trials of anti-PD-1/PD-L1 drugs to treat cancers. Biomed Pharmacother 2021; 143:112238. [PMID: 34649362 DOI: 10.1016/j.biopha.2021.112238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE To gain a deeper understanding of the hot topics and future prospects of programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) inhibitors treatment of cancer through scientometric analysis of the top-100 most cited clinical trials. MATERIALS AND METHODS We searched the Web of Science Core Collection database from 1980 to June 2019. Two reviewers independently screened the top-100 most cited clinical trials that defined by the National Institutes of Health starting from the most cited article. Title, year of publication, citations, type of cancer, and focused aspects of outcomes were extracted from included clinical trials. VOSviewer software (version 1.6.9) and Excel 2016 were used to do statistical analysis. The evidence mapping was used to present the relationship between cancers, drugs, citations, and outcomes, etc. RESULTS: The top-100 most cited clinical trials published from 2010 to 2018 in nine journals with high impact factor (IF) (IF2018:6.68-70.67), and Lancet Oncology (USA) published the most clinical trials (n = 29, IF2018 = 35.3856). The total number of citations of the top-100 most cited clinical trials was from 59 to 5606. 920 authors from 34 countries and 458 organizations participated in publishing the top-100 most cited clinical trials. The USA (n = 95) and Memorial Sloan-Kettering Cancer Center (n = 31) contributed the most publications. Based on the evidence mapping, there are 25 different types of cancers (e.g. lung cancer, melanoma, and renal cell cancer) and five focused aspects of outcomes (e.g. safety and efficacy). CONCLUSION The USA was the dominant country. Anti-PD-1/PD-L1 drugs were widely used to treat lung cancer, melanoma, renal cell cancer, and Hodgkin lymphoma. More exploration should be done to explore the use of anti-PD-1/PD-L1 drugs to treat more type of cancers in future research.
Collapse
Affiliation(s)
- Ming Liu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, China
| | - Ya Gao
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, China
| | - Yuan Yuan
- Gansu Provincial Maternity and Child-care Hospital, Lanzhou 730000, China
| | - Shuzhen Shi
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100000, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, China.
| | - Junhua Zhang
- Evidence-Based Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
16
|
Abdin SM, Paasch D, Morgan M, Lachmann N. CARs and beyond: tailoring macrophage-based cell therapeutics to combat solid malignancies. J Immunother Cancer 2021; 9:jitc-2021-002741. [PMID: 34462325 PMCID: PMC8407221 DOI: 10.1136/jitc-2021-002741] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2021] [Indexed: 12/20/2022] Open
Abstract
Recent understanding of the role and contribution of immune cells in disease onset and progression has pioneered the field of immunotherapies. Use of genetic engineering to deliver, correct or enhance immune cells has been clinically successful, especially in the field of cancer immunotherapy. Indeed, one of the most attractive approaches is the introduction of chimeric antigen receptors (CARs) to immune cells, such as T cells. Recent studies revealed that adapting this platform for use in macrophages may widen the spectrum of CAR applications for better control of solid tumors and, thus, extend this treatment strategy to more patients with cancer. Given the novel insights into tumor-associated macrophages and new targeting strategies to boost anticancer therapy, this review aims to provide an overview of the current status of the role of macrophages in cancer therapy. The various genetic engineering approaches that can be used to optimize macrophages for use in oncology are discussed, with special attention dedicated to the implication of the CAR platform on macrophages for anticancer therapy. The current clinical status, challenges and future perspective of macrophage-based drugs are highlighted.
Collapse
Affiliation(s)
- Shifaa M Abdin
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Daniela Paasch
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Michael Morgan
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Nico Lachmann
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany .,REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany.,RESIST, Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
17
|
Teng CF, Wang T, Shih FY, Shyu WC, Jeng LB. Therapeutic efficacy of dendritic cell vaccine combined with programmed death 1 inhibitor for hepatocellular carcinoma. J Gastroenterol Hepatol 2021; 36:1988-1996. [PMID: 33462840 DOI: 10.1111/jgh.15398] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/15/2020] [Accepted: 01/01/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Hepatocellular carcinoma (HCC) remains a serious cause of cancer-related deaths worldwide. Developing new therapeutic strategies is urgently needed to improve the outcomes of HCC patients. Dendritic cell (DC)-based vaccines and programmed death 1 (PD-1) immune checkpoint inhibitors have been regarded as potential immunotherapeutics for HCC. However, the therapeutic efficacy of combining these two treatments for HCC remains to be evaluated. METHODS In this study, DCs were derived from mouse bone marrow and pulsed with mouse HCC cell lysates to generate a DC vaccine. A monoclonal antibody that blocks the interaction of mouse PD-1 with its ligands was used as a PD-1 inhibitor. An orthotopic HCC mouse model was established to assess the effect of a DC vaccine in combination with a PD-1 inhibitor on overall survival and tumor volume. RESULTS Compared with the untreated control, single treatment with a DC vaccine or PD-1 inhibitor prolonged the overall survival and reduced the tumor volume of HCC mice. Further, compared with the single treatment with the DC vaccine or the PD-1 inhibitor, a combination treatment using both agents elicited a higher cytotoxicity of T cells against HCC cells and resulted in a better overall survival, smaller tumor volume, and greater tumor cell apoptosis in HCC mice. CONCLUSIONS Our results suggest that a combination treatment with DC vaccine and PD-1 inhibitor may be a promising therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Chiao-Fang Teng
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan.,Research Center for Cancer Biology, China Medical University, Taichung, Taiwan
| | - Ting Wang
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| | - Fu-Ying Shih
- Ph.D. Program for Biotech Pharmaceutical Industry, School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Woei-Cherng Shyu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Occupational Therapy, Asia University, Taichung, Taiwan.,Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
18
|
El Meskini R, Atkinson D, Kulaga A, Abdelmaksoud A, Gumprecht M, Pate N, Hayes S, Oberst M, Kaplan IM, Raber P, Van Dyke T, Sharan SK, Hollingsworth R, Day CP, Merlino G, Weaver Ohler Z. Distinct Biomarker Profiles and TCR Sequence Diversity Characterize the Response to PD-L1 Blockade in a Mouse Melanoma Model. Mol Cancer Res 2021; 19:1422-1436. [PMID: 33888600 DOI: 10.1158/1541-7786.mcr-20-0881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/09/2021] [Accepted: 04/19/2021] [Indexed: 11/16/2022]
Abstract
Only a subset of patients responds to immune checkpoint blockade (ICB) in melanoma. A preclinical model recapitulating the clinical activity of ICB would provide a valuable platform for mechanistic studies. We used melanoma tumors arising from an Hgftg;Cdk4R24C/R24C genetically engineered mouse (GEM) model to evaluate the efficacy of an anti-mouse PD-L1 antibody similar to the anti-human PD-L1 antibodies durvalumab and atezolizumab. Consistent with clinical observations for ICB in melanoma, anti-PD-L1 treatment elicited complete and durable response in a subset of melanoma-bearing mice. We also observed tumor growth delay or regression followed by recurrence. For early treatment assessment, we analyzed gene expression profiles, T-cell infiltration, and T-cell receptor (TCR) signatures in regressing tumors compared with tumors exhibiting no response to anti-PD-L1 treatment. We found that CD8+ T-cell tumor infiltration corresponded to response to treatment, and that anti-PD-L1 gene signature response indicated an increase in antigen processing and presentation, cytokine-cytokine receptor interaction, and natural killer cell-mediated cytotoxicity. TCR sequence data suggest that an anti-PD-L1-mediated melanoma regression response requires not only an expansion of the TCR repertoire that is unique to individual mice, but also tumor access to the appropriate TCRs. Thus, this melanoma model recapitulated the variable response to ICB observed in patients and exhibited biomarkers that differentiate between early response and resistance to treatment, providing a valuable platform for prediction of successful immunotherapy. IMPLICATIONS: Our melanoma model recapitulates the variable response to anti-PD-L1 observed in patients and exhibits biomarkers that characterize early antibody response, including expansion of the TCR repertoire.
Collapse
Affiliation(s)
- Rajaa El Meskini
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland.
| | - Devon Atkinson
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Alan Kulaga
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Abdalla Abdelmaksoud
- Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research (CCR), National Cancer Institute, Bethesda, Maryland.,Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Michelle Gumprecht
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Nathan Pate
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | | | | | | | - Terry Van Dyke
- Mouse Cancer Genetics Program, CCR, NCI/NIH, Frederick, Maryland
| | - Shyam K Sharan
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland.,Mouse Cancer Genetics Program, CCR, NCI/NIH, Frederick, Maryland
| | | | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, CCR, NCI/NIH, Bethesda, Maryland
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, CCR, NCI/NIH, Bethesda, Maryland
| | - Zoë Weaver Ohler
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland.
| |
Collapse
|
19
|
Shi Y, Li D, He C, Chen X. Design of an Injectable Polypeptide Hydrogel Depot Containing the Immune Checkpoint Blocker Anti-PD-L1 and Doxorubicin to Enhance Antitumor Combination Therapy. Macromol Biosci 2021; 21:e2100049. [PMID: 33871152 DOI: 10.1002/mabi.202100049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/29/2021] [Indexed: 12/11/2022]
Abstract
Combination therapy can be used to enhance the therapeutic response and decrease side effects during cancer treatment. In this study, a system is developed to locally deliver the immune checkpoint blockade antibody targeting programmed death-ligand 1 (anti-PD-L1 or aPD-L1) and doxorubicin (Dox), by an injectable, biocompatible polypeptide hydrogel as a drug depot. The localized and sustained release of Dox after the intratumoral injection of the co-loaded hydrogel induces immunogenic tumor cell death, thus promoting an antitumor immunological response. The tumor inhibitory effect is significantly enhanced by the simultaneous release of aPD-L1 at the tumor site thanks to its action on the inhibition of the PD-1/PD-L1 pathway and restoration of the tumor-killing effect of cytotoxic T cells. Treatment of the B16F10 melanoma model with the aPD-L1 and Dox co-loaded hydrogel leads to a remarkable inhibition of tumor progression and prolongation of animal survival.
Collapse
Affiliation(s)
- Yingge Shi
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China.,University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Dong Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China.,University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Chaoliang He
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China.,University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China.,University of Science and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
20
|
Mondal P, Kaur B, Natesh J, Meeran SM. The emerging role of miRNA in the perturbation of tumor immune microenvironment in chemoresistance: Therapeutic implications. Semin Cell Dev Biol 2021; 124:99-113. [PMID: 33865701 DOI: 10.1016/j.semcdb.2021.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/16/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023]
Abstract
Chemoresistance is a major hindrance in cancer chemotherapies, a leading cause of tumor recurrence and cancer-related deaths. Cancer cells develop numerous strategies to elude immune attacks and are regulated by immunological factors. Cancer cells can alter the expression of several immune modulators to upregulate the activities of immune checkpoint pathways. Targeting the immune checkpoint inhibitors is a part of the cancer immunotherapy altered during carcinogenesis. These immune modulators have the capability to reprogram the tumor microenvironment, thereby change the efficacy of chemotherapeutics. In general, the sensitivity of drugs is reduced in the immunosuppressive tumor microenvironment, resulting in chemoresistance and tumor relapse. The regulation of microRNAs (miRNAs) is well established in cancer initiation, progression, and therapy. Intriguingly, miRNA affects cancer immune surveillance and immune response by targeting immune checkpoint inhibitors in the tumor microenvironment. miRNAs alter the gene expression at the post-transcriptional level, which modulates both innate and adaptive immune systems. Alteration of tumor immune microenvironment influences drug sensitivity towards cancer cells. Besides, the expression profile of immune-modulatory miRNAs can be used as a potential biomarker to predict the response and clinical outcomes in cancer immunotherapy and chemotherapy. Recent evidences have revealed that cancer-derived immune-modulatory miRNAs might be promising targets to counteract cancer immune escape, thereby increasing drug efficacy. In this review, we have compiled the role of miRNAs in overcoming the chemoresistance by modulating tumor microenvironment and discussed their preclinical and clinical implications.
Collapse
Affiliation(s)
- Priya Mondal
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Bhavjot Kaur
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, India
| | - Jagadish Natesh
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Syed Musthapa Meeran
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
21
|
Zhu Q, Sun F, Li T, Zhou M, Ye J, Ji A, Wang H, Ding C, Chen H, Xu Z, Yu H. Engineering Oxaliplatin Prodrug Nanoparticles for Second Near-Infrared Fluorescence Imaging-Guided Immunotherapy of Colorectal Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007882. [PMID: 33690984 DOI: 10.1002/smll.202007882] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/18/2021] [Indexed: 06/12/2023]
Abstract
Colorectal cancer (CRC) ranks as the third common and the fourth lethal cancer type worldwide. Immune checkpoint blockade therapy demonstrates great efficacy in a subset of metastatic CRC patients, but precise activation of the antitumor immune response at the tumor site is still challenging. Here a versatile prodrug nanoparticle for second near-infrared (NIR-II) fluorescence imaging-guided combinatory immunotherapy of CRC is reported. The prodrug nanoparticles are constructed with a polymeric oxaliplatin prodrug (PBOXA) and a donor-spacer-acceptor-spacer-donor type small molecular fluorophore TQTCD. The later displays large Stokes shift (>300 nm), fluorescence emission over 1000 nm, and excellent photothermal conversion performance for NIR-II fluorescence imaging-guided photothermal therapy (PTT). The prodrug nanoparticles show seven times higher intratumoral OXA accumulation than free oxaliplatin. TQTCD-based PTT and PBOXA-induced chemotherapy trigger immunogenic cell death of the tumor cells and elicit antitumor immune response in a spatiotemporally controllable manner. Further combination of the prodrug nanoparticle-based PTT/chemotherapy with programmed death ligand 1 blockade significantly promotes intratumoral infiltration of the cytotoxic T lymphocytes and eradicates the CRC tumors. The NIR-II fluorescence imaging-guided immunotherapy may provide a promising approach for CRC treatment.
Collapse
Affiliation(s)
- Qiurong Zhu
- College of Chemistry and Chemical Engineering of Inner Mongolia University, Inner Mongolia University, Huhhot, 010021, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Fang Sun
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Tianliang Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Mengxue Zhou
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Jiayi Ye
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Aiyan Ji
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Hui Wang
- College of Chemistry and Chemical Engineering of Inner Mongolia University, Inner Mongolia University, Huhhot, 010021, China
| | - Chunyong Ding
- School of Pharmacy, Shanghai Jiaotong University, Shanghai, 200241, China
| | - Hao Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Zhiai Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| |
Collapse
|
22
|
Correlation Between Immune-related Adverse Event (IRAE) Occurrence and Clinical Outcome in Patients With Metastatic Renal Cell Carcinoma (mRCC) Treated With Nivolumab: IRAENE Trial, an Italian Multi-institutional Retrospective Study. Clin Genitourin Cancer 2020; 18:477-488. [DOI: 10.1016/j.clgc.2020.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/06/2020] [Accepted: 05/12/2020] [Indexed: 01/15/2023]
|
23
|
Kuang Z, Pu P, Wu M, Wu Z, Wang L, Li Y, Zhang S, Jing H, Wu W, Chen B, Liu J. A Novel Bispecific Antibody with PD-L1-assisted OX40 Activation for Cancer Treatment. Mol Cancer Ther 2020; 19:2564-2574. [PMID: 32999045 DOI: 10.1158/1535-7163.mct-20-0226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/19/2020] [Accepted: 09/24/2020] [Indexed: 11/16/2022]
Abstract
Immunotherapy using OX40 agonist antibodies shows great preclinical efficacy in mouse tumor models. But in a clinical setting, OX40 agonist antibody alone or in combination with checkpoint blockade exhibits only modest efficacy due to lack of sufficient activation. We hypothesized that the limited antitumor activity in patients may due to insufficient clustering of OX40 antibody in the tumor. To test this hypothesis, we generated a tetravalent programmed death ligand-1 (PD-L1)/OX40 BsAb by fusing two PD-L1 VHH fragments to the C-terminus of a nonblocking agonistic anti-OX40 antibody. The resulting BsAb had intact function of each parental antibody, including efficiently blocking PD1/PD-L1 interaction and inducing OX40 activation. In addition, this BsAb showed significantly enhanced potency in activation of OX40-expressing T cells when PD-L1-expressing tumor cells or dendrite cells were present, through PD-L1-mediated cross-linking of OX40. Moreover, the BsAb exhibited superior antitumor activities over the parental monospecific antibodies alone or in combination in multiple in vivo tumor models. These results demonstrated a great potential for further clinical development of the potent immunostimulatory PD-L1/OX40 bispecific antibody.
Collapse
Affiliation(s)
- Zhihui Kuang
- Drug Discovery, Innovent Biologics Co, Suzhou, Jiangsu, P.R. China
| | - Pu Pu
- Drug Discovery, Innovent Biologics Co, Suzhou, Jiangsu, P.R. China
| | - Min Wu
- Drug Discovery, Innovent Biologics Co, Suzhou, Jiangsu, P.R. China
| | - Zhihai Wu
- Drug Discovery, Innovent Biologics Co, Suzhou, Jiangsu, P.R. China
| | - Li Wang
- Drug Discovery, Innovent Biologics Co, Suzhou, Jiangsu, P.R. China
| | - Yiming Li
- Drug Discovery, Innovent Biologics Co, Suzhou, Jiangsu, P.R. China
| | - Shaofei Zhang
- Drug Discovery, Innovent Biologics Co, Suzhou, Jiangsu, P.R. China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Shaanxi, Xi'an, P.R. China
| | - Hua Jing
- Drug Discovery, Innovent Biologics Co, Suzhou, Jiangsu, P.R. China
| | - Weiwei Wu
- Drug Discovery, Innovent Biologics Co, Suzhou, Jiangsu, P.R. China
| | - Bingliang Chen
- Drug Discovery, Innovent Biologics Co, Suzhou, Jiangsu, P.R. China
| | - Junjian Liu
- Drug Discovery, Innovent Biologics Co, Suzhou, Jiangsu, P.R. China.
| |
Collapse
|
24
|
Raimondi A, Sepe P, Zattarin E, Mennitto A, Stellato M, Claps M, Guadalupi V, Verzoni E, de Braud F, Procopio G. Predictive Biomarkers of Response to Immunotherapy in Metastatic Renal Cell Cancer. Front Oncol 2020; 10:1644. [PMID: 32903369 PMCID: PMC7434861 DOI: 10.3389/fonc.2020.01644] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction In the last decades, the therapeutic decision-making approach to metastatic renal cell cancer (mRCC) has dramatically changed thanks to the introduction in the treatment scenario of, first, anti-angiogenic agents and, afterward, immune-checkpoint inhibitors (ICIs). Immunotherapy is now the standard of care in pretreated mRCC patients and has recently entered even the first line setting. Nevertheless, in mRCC as well as in other tumor settings, a durable and clinically meaningful benefit from treatment with ICIs is not obtained for all patients treated. Therefore, the necessity to identify and validate predictive biomarkers of response to immunotherapy has emerged, in order to design the optimal treatment strategy for mRCC patients. Discussion In this review, we present and discuss the most promising predictive biomarkers of response to ICIs in mRCC with the recent data available. In details, the first marker that was investigated is the immunohistochemical expression of programmed death receptor ligand 1 (PD-L1), showing a negative prognostic role in mRCC, but the debate about its potential predictive value is still open. Additionally, the high heterogeneity in PD-L1 determination methods adds complexity to this issue. Second, the tumor mutational or neoantigen burden is an emerging biomarker of increased response to immunotherapy, hypothesizing that the higher the TMB, the higher is the production of neoantigens, and thus the stimulation of anti-tumor immune response, even though controversial results have been obtained. Third, the tumor microenvironment, namely the different populations of the immune infiltrate, plays a key role in tumor progression and in the response to immunotherapy. Finally, several studies have collected evidence on the potential association of the occurrence of immune-related adverse events (irAEs) with the benefit from ICIs, first in non-small cell lung cancer (NSCLC) and melanoma, and recently even in mRCC. Conclusion Several promising biomarkers of response to immunotherapy with ICIs have been identified, though without conclusive results upon their potential predictive value in mRCC. Therefore, the results of the exploratory analyses of the recently presented first-line trials and hopefully of future prospective, biomarker-driven studies could provide useful tools to be applied in the everyday clinical practice.
Collapse
Affiliation(s)
- Alessandra Raimondi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Pierangela Sepe
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Emma Zattarin
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessia Mennitto
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco Stellato
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Melanie Claps
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valentina Guadalupi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Verzoni
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Oncology and Hemato-Oncology Department, University of Milan, Milan, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
25
|
Zahavi D, Weiner L. Monoclonal Antibodies in Cancer Therapy. Antibodies (Basel) 2020; 9:E34. [PMID: 32698317 PMCID: PMC7551545 DOI: 10.3390/antib9030034] [Citation(s) in RCA: 368] [Impact Index Per Article: 73.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/11/2020] [Accepted: 07/04/2020] [Indexed: 12/19/2022] Open
Abstract
Monoclonal antibody-based immunotherapy is now considered to be a main component of cancer therapy, alongside surgery, radiation, and chemotherapy. Monoclonal antibodies possess a diverse set of clinically relevant mechanisms of action. In addition, antibodies can directly target tumor cells while simultaneously promoting the induction of long-lasting anti-tumor immune responses. The multifaceted properties of antibodies as a therapeutic platform have led to the development of new cancer treatment strategies that will have major impacts on cancer care. This review focuses on the known mechanisms of action, current clinical applications for the treatment of cancer, and mechanisms of resistance of monoclonal antibody therapy. We further discuss how monoclonal antibody-based strategies have moved towards enhancing anti-tumor immune responses by targeting immune cells instead of tumor antigens as well as some of the current combination therapies.
Collapse
Affiliation(s)
- David Zahavi
- Tumor Biology Training Program, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, 3800 Reservoir Rd NW, Washington, DC 20007, USA;
| | - Louis Weiner
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, 3800 Reservoir Rd NW, Washington, DC 20007, USA
| |
Collapse
|
26
|
Jiang J, Chen ZP, Zhu HP, Zhang YQ, Qian XL, Zhang M, Ni C, Zuo Y. Responses of metastatic primary fallopian tube carcinoma to pembrolizumab and nab-paclitaxel: A case report. Medicine (Baltimore) 2020; 99:e21203. [PMID: 32664168 PMCID: PMC7360280 DOI: 10.1097/md.0000000000021203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
RATIONALE Primary fallopian tube carcinoma (PFTC) is an extremely rare but invasive malignancy with a dismal prognosis. Very few data exist on the salvage treatment for patients with PFTC. Here we report a case showing an impressive response to immunotherapy combined with chemotherapy, which have never been reported before on patients with metastatic PFTC. PATIENT CONCERNS A 42-year-old woman, who was diagnosed with PFTC in 2010, had been failed of multiple systemic therapies and antiangiogenic therapy because of the disease recurrence and progression. DIAGNOSIS Metastatic primary fallopian tube carcinoma. INTERVENTIONS The patient underwent surgery in May 2010 and had multi-line chemotherapies plus an anti-vascular endothelial growth factor (anti-VEGF) monoclonal antibody for about 9 years. Due to treatment failure the patient accepted the immunotherapy with the checkpoint inhibitor, pembrolizumab, combined with nab-paclitaxel from December 2018 to April 2019. OUTCOMES The patient showed a complete response after 6 cycles treatment. Thus far, the patient is taking pembrolizumab as maintenance and remains in good health. LESSONS Pembrolizumab combined with chemotherapy for treatment of PFTC may provide a positive antitumor effect in multiple metastatic lesions, but more clinical evidence is needed to confirm the efficacy and safety.
Collapse
|
27
|
Hazama D, Yin Y, Murata Y, Matsuda M, Okamoto T, Tanaka D, Terasaka N, Zhao J, Sakamoto M, Kakuchi Y, Saito Y, Kotani T, Nishimura Y, Nakagawa A, Suga H, Matozaki T. Macrocyclic Peptide-Mediated Blockade of the CD47-SIRPα Interaction as a Potential Cancer Immunotherapy. Cell Chem Biol 2020; 27:1181-1191.e7. [PMID: 32640189 DOI: 10.1016/j.chembiol.2020.06.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/30/2020] [Accepted: 06/12/2020] [Indexed: 12/12/2022]
Abstract
Medium-sized macrocyclic peptides are an alternative to small compounds and large biomolecules as a class of pharmaceutics. The CD47-SIRPα signaling axis functions as an innate immune checkpoint that inhibits phagocytosis in phagocytes and has been implicated as a promising target for cancer immunotherapy. The potential of macrocyclic peptides that target this signaling axis as immunotherapeutic agents has remained unknown, however. Here we have developed a macrocyclic peptide consisting of 15 amino acids that binds to the ectodomain of mouse SIRPα and efficiently blocks its interaction with CD47 in an allosteric manner. The peptide markedly promoted the phagocytosis of antibody-opsonized tumor cells by macrophages in vitro as well as enhanced the inhibitory effect of anti-CD20 or anti-gp75 antibodies on tumor formation or metastasis in vivo. Our results suggest that allosteric inhibition of the CD47-SIRPα interaction by macrocyclic peptides is a potential approach to cancer immunotherapy.
Collapse
Affiliation(s)
- Daisuke Hazama
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yizhen Yin
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yoji Murata
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Makoto Matsuda
- Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Takeshi Okamoto
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Daisuke Tanaka
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Naohiro Terasaka
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Jinxuan Zhao
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Mariko Sakamoto
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yuka Kakuchi
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yasuyuki Saito
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takenori Kotani
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Atsushi Nakagawa
- Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Takashi Matozaki
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| |
Collapse
|
28
|
Teng CF, Wang T, Wu TH, Lin JH, Shih FY, Shyu WC, Jeng LB. Combination therapy with dendritic cell vaccine and programmed death ligand 1 immune checkpoint inhibitor for hepatocellular carcinoma in an orthotopic mouse model. Ther Adv Med Oncol 2020; 12:1758835920922034. [PMID: 32565925 PMCID: PMC7288802 DOI: 10.1177/1758835920922034] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/03/2020] [Indexed: 02/06/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is among the most common and lethal human cancers worldwide. Despite remarkable advances in treatment, high mortality in HCC patients remains a big challenge. To develop novel therapeutic strategies for HCC is thus urgently needed to improve patient survival. Dendritic cells (DC)-based vaccines can induce tumor-specific immunity and have emerged as a promising approach for treating HCC patients; however, its effectiveness needs to be improved. Recently, blockade of programmed death ligand 1 (PD-L1) immune checkpoint pathway has been shown to enhance anti-tumor immune responses and exhibited great potential in HCC therapy. Methods In this study, we generated DC vaccine by pulsing the C57BL/6J mouse bone marrow-derived DC with mouse hepatoma Hep-55.1C cell lysate. We developed a therapeutic strategy combining DC vaccine and PD-L1 inhibitor for HCC and evaluated its efficacy in an orthotopic HCC mouse model in which Hep-55.1C cells were directly injected into left liver lobe of C57BL/6J mouse. Results Compared with a control group of mice, groups of mice treated with DC vaccine or PD-L1 inhibitor had significantly improved overall survival, reduced tumor volume, and increased tumor cell apoptosis. Remarkably, combination treatment with DC vaccine and PD-L1 inhibitor led to considerably longer overall survival, smaller tumor volume, and higher tumor cell apoptosis of mice than either treatment alone in a dose-dependent manner through inducing a stronger anti-tumor cytotoxic T cell response. Conclusion Our data suggested that combination therapy with DC vaccine and PD-L1 inhibitor might have great promise as a novel treatment strategy for HCC.
Collapse
Affiliation(s)
- Chiao-Fang Teng
- Graduate Institute of Biomedical Sciences, China Medical University, No.91, Hsueh-Shih Rd., Northern Dist., Taichung City 404Organ Transplantation Center, China Medical University Hospital, Taichung Research Center for Cancer Biology, China Medical University, Taichung
| | - Ting Wang
- Organ Transplantation Center, China Medical University Hospital, Taichung
| | - Tzu-Hua Wu
- Organ Transplantation Center, China Medical University Hospital, Taichung
| | - Jia-Hui Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung
| | - Fu-Ying Shih
- Program for Biotech Pharmaceutical Industry, School of Pharmacy, China Medical University, Taichung
| | - Woei-Cherng Shyu
- Graduate Institute of Biomedical Sciences, China Medical University, No.91, Hsueh-Shih Rd., Northern Dist., Taichung City 404 Department of Occupational Therapy, Asia University, Taichung Department of Neurology, China Medical University Hospital, Taichung Translational Medicine Research Center, China Medical University Hospital, Taichung
| | - Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, No.2, Yude Rd., Northern Dist., Taichung City 404
| |
Collapse
|
29
|
da Costa AC, Sodergren M, Jayant K, Santa Cruz F, Spalding D, Pai M, Habib N. Radiofrequency combined with immunomodulation for hepatocellular carcinoma: State of the art and innovations. World J Gastroenterol 2020; 26:2040-2048. [PMID: 32536773 PMCID: PMC7267689 DOI: 10.3748/wjg.v26.i17.2040] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/25/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver tumor and has been considered a very immunogenic tumor. The treatment with radiofrequency ablation (RFA) has been established as the standard ablative therapy for early HCC, and is currently recognized as the main ablative tool for HCC tumors < 5 cm in size; however, progression and local recurrence remain the main disadvantages of this approach. To solve this clinical problem, recent efforts were concentrated on multimodal treatment, combining different strategies, including the combination of RFA and immunotherapy. This article reviewed the combination treatment of RFA with immunotherapy and found that this treatment strategy leads to an increased response of anti-tumor T cells, significantly reduces the risk of recurrence and improves survival rates compared to RFA alone. This review highlighted scientific evidence that supports the current recommendations for pre-clinical studies, and discuss the need for further research on this topic.
Collapse
MESH Headings
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Combined Modality Therapy/methods
- Disease Progression
- Disease-Free Survival
- Humans
- Immunotherapy/methods
- Liver/immunology
- Liver/pathology
- Liver/surgery
- Liver Neoplasms/immunology
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/prevention & control
- Radiofrequency Ablation/instrumentation
- Radiofrequency Ablation/methods
- Randomized Controlled Trials as Topic
- Review Literature as Topic
- Survival Rate
Collapse
Affiliation(s)
- Adriano Carneiro da Costa
- Department of Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom
| | - Mikael Sodergren
- Department of Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom
| | - Kumar Jayant
- Department of Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom
| | - Fernando Santa Cruz
- Department of Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom
| | - Duncan Spalding
- Department of Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom
| | - Madhava Pai
- Department of Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom
| | - Nagy Habib
- Department of Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom
| |
Collapse
|
30
|
Zaher DM, El‐Gamal MI, Omar HA, Aljareh SN, Al‐Shamma SA, Ali AJ, Zaib S, Iqbal J. Recent advances with alkaline phosphatase isoenzymes and their inhibitors. Arch Pharm (Weinheim) 2020; 353:e2000011. [DOI: 10.1002/ardp.202000011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/09/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Dana M. Zaher
- Sharjah Institute for Medical ResearchSharjah United Arab Emirates
| | - Mohammed I. El‐Gamal
- Sharjah Institute for Medical ResearchSharjah United Arab Emirates
- College of PharmacySharjah United Arab Emirates
- Department of Medicinal ChemistryFaculty of PharmacyMansoura Egypt
| | - Hany A. Omar
- Sharjah Institute for Medical ResearchSharjah United Arab Emirates
- College of PharmacySharjah United Arab Emirates
- Department of PharmacologyFaculty of PharmacyBeni‐Suef Egypt
| | | | | | - Aya J. Ali
- College of PharmacySharjah United Arab Emirates
| | - Sumera Zaib
- Centre for Advanced Drug ResearchCOMSATS University Islamabad Abbottabad Campus Abbottabad Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug ResearchCOMSATS University Islamabad Abbottabad Campus Abbottabad Pakistan
| |
Collapse
|
31
|
Lanitis T, Ambavane A, Zheng Y, Schlichting M, Phatak H. Value assessment of immuno-oncology in the treatment of rare tumors in the era of accelerated conditional approvals. Future Oncol 2019; 15:4057-4067. [PMID: 31687852 DOI: 10.2217/fon-2019-0456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: To conduct a value assessment of an immuno-oncology (IO) therapy for a rare cancer and evaluate whether existing frameworks consider challenges associated with valuing IOs for rare cancers. Materials & methods: Value frameworks developed by American Society of Clinical Oncologists, Memorial Sloan Kettering Cancer Center and National Comprehensive Cancer Network were used to estimate the value of an IO therapy in a rare cancer based on single-arm trial data and retrospective studies. Results: Paucity of direct evidence comparing rare cancer treatments and lack of acceptance of indirect comparisons hinder appropriate value assessment. Measurement of value based on short-term outcomes may not capture the value of IOs, where survival is often characterized by a plateau. Conclusion: Further work is required to factor in nuances associated with rare cancers and guide end users of the frameworks. To capture true value, multiple or more holistic value assessments are required.
Collapse
Affiliation(s)
- Tereza Lanitis
- Evidera, The Ark, 201 Talgarth Road, Hammersmith, London W6 8BJ, United Kingdom
| | - Apoorva Ambavane
- Evidera, The Ark, 201 Talgarth Road, Hammersmith, London W6 8BJ, United Kingdom
| | - Ying Zheng
- EMD Serono Inc, One Technology Place, Rockland, MA 02370, USA
| | | | - Hemant Phatak
- EMD Serono Inc, One Technology Place, Rockland, MA 02370, USA
| |
Collapse
|
32
|
Omar HA, El‐Serafi AT, Hersi F, Arafa EA, Zaher DM, Madkour M, Arab HH, Tolba MF. Immunomodulatory MicroRNAs in cancer: targeting immune checkpoints and the tumor microenvironment. FEBS J 2019; 286:3540-3557. [DOI: 10.1111/febs.15000] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 05/29/2019] [Accepted: 07/12/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Hany A. Omar
- Sharjah Institute for Medical Research University of Sharjah UAE
- Department of Pharmacology, Faculty of Pharmacy Beni‐Suef University Egypt
| | - Ahmed T. El‐Serafi
- Sharjah Institute for Medical Research University of Sharjah UAE
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine Suez Canal University Ismailia Egypt
| | - Fatema Hersi
- Sharjah Institute for Medical Research University of Sharjah UAE
| | - El‐Shaimaa A. Arafa
- Department of Clinical Sciences, College of Pharmacy and Health Sciences Ajman University UAE
| | - Dana M. Zaher
- Sharjah Institute for Medical Research University of Sharjah UAE
| | - Mohamed Madkour
- Sharjah Institute for Medical Research University of Sharjah UAE
| | - Hany H. Arab
- Department of Biochemistry, Faculty of Pharmacy Cairo University Egypt
- Biochemistry Division and GTMR Unit, Department of Pharmacology and Toxicology, Faculty of Pharmacy Taif University Saudi Arabia
| | - Mai F. Tolba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy Ain Shams University Cairo Egypt
- Biology Department, School of Sciences and Engineering The American University in Cairo New Cairo Egypt
| |
Collapse
|
33
|
Geng A, Cui H, Zhang L, Chen X, Li H, Lu T, Zhu Y. Discovery of novel phenoxybenzamide analogues as Raf/HDAC dual inhibitors. Bioorg Med Chem Lett 2019; 29:1605-1608. [DOI: 10.1016/j.bmcl.2019.04.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/22/2019] [Accepted: 04/27/2019] [Indexed: 12/13/2022]
|
34
|
Ugurel S, Spassova I, Wohlfarth J, Drusio C, Cherouny A, Melior A, Sucker A, Zimmer L, Ritter C, Schadendorf D, Becker JC. MHC class-I downregulation in PD-1/PD-L1 inhibitor refractory Merkel cell carcinoma and its potential reversal by histone deacetylase inhibition: a case series. Cancer Immunol Immunother 2019; 68:983-990. [PMID: 30993371 PMCID: PMC11028125 DOI: 10.1007/s00262-019-02341-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 04/11/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Merkel cell carcinoma (MCC) is an aggressive skin cancer in which PD-1/PD-L1 blockade has shown remarkable response rates. However, a significant proportion of patients shows primary or secondary resistance against PD-1/PD-L1 inhibition, with HLA class-I downregulation and insufficient influx of CD8+ T cells into the tumor as possible immune escape mechanisms. Histone deacetylase inhibitors (HDACi) have been demonstrated to reverse low HLA class-I expression caused by epigenetic downregulation of the antigen machinery (APM) in vitro and in pre-clinical models in vivo. CASE PRESENTATIONS We report four cases of patients with metastatic MCC who did not respond to immunotherapy by PD-1/PD-L1 blockade. Two of the patients received, subsequently, the HDACi panobinostat in combination with PD-1/PD-L1 blockade. Tumor biopsies of the patients were analyzed for cellular and molecular markers of antigen processing and presentation as well as the degree of T-cell infiltration. RESULTS AND CONCLUSION Low expression of APM-related genes associated with low HLA class-I surface expression was observed in all MCC patients, progressing on PD-1/PD-L1 blockade. In one evaluable patient, of the two treated with the combination therapy of the HDACi, panobinostat and PD-1/PD-L1 blockade, reintroduction of HLA class-I-related genes, enhanced HLA class-I surface expression, and elevated CD8+ T-cell infiltration into the MCC tumor tissue were observed; however, these changes did not translate into a clinical benefit. Our findings suggest that HDACi may be useful to overcome HLA class-I downregulation as a resistance mechanism against anti-PD-1/PD-L1 antibodies in MCC patients. Prospective clinical trials are needed to evaluate this notion.
Collapse
Affiliation(s)
- Selma Ugurel
- Department of Dermatology, University Clinic Essen, Essen, Germany
| | - Ivelina Spassova
- Translational Skin Cancer Research (TSCR), University Clinic Essen, Universitätsstrasse 1, S05 T05 B, 45141, Essen, Germany
| | | | - Christina Drusio
- Department of Dermatology, University Clinic Essen, Essen, Germany
| | - Angela Cherouny
- Translational Skin Cancer Research (TSCR), University Clinic Essen, Universitätsstrasse 1, S05 T05 B, 45141, Essen, Germany
| | - Anita Melior
- Translational Skin Cancer Research (TSCR), University Clinic Essen, Universitätsstrasse 1, S05 T05 B, 45141, Essen, Germany
| | - Antje Sucker
- Department of Dermatology, University Clinic Essen, Essen, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Clinic Essen, Essen, Germany
| | - Cathrin Ritter
- Translational Skin Cancer Research (TSCR), University Clinic Essen, Universitätsstrasse 1, S05 T05 B, 45141, Essen, Germany
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung, DKTK) Partner Site Essen, Deutsches Krebsforschungsinstitut (DKFZ), Heidelberg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Clinic Essen, Essen, Germany
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung, DKTK) Partner Site Essen, Deutsches Krebsforschungsinstitut (DKFZ), Heidelberg, Germany
| | - Jürgen C Becker
- Department of Dermatology, University Clinic Essen, Essen, Germany.
- Translational Skin Cancer Research (TSCR), University Clinic Essen, Universitätsstrasse 1, S05 T05 B, 45141, Essen, Germany.
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung, DKTK) Partner Site Essen, Deutsches Krebsforschungsinstitut (DKFZ), Heidelberg, Germany.
| |
Collapse
|
35
|
Verzoni E, Cartenì G, Cortesi E, Giannarelli D, De Giglio A, Sabbatini R, Buti S, Rossetti S, Cognetti F, Rastelli F, Sobrero A, Turci D, Sternberg CN, Porta C, Cappuzzo F, Tortora G, Tassinari D, Panni S, Pazzola A, Surico G, Raimondi A, De Giorgi U, Procopio G. Real-world efficacy and safety of nivolumab in previously-treated metastatic renal cell carcinoma, and association between immune-related adverse events and survival: the Italian expanded access program. J Immunother Cancer 2019; 7:99. [PMID: 30944023 PMCID: PMC6448290 DOI: 10.1186/s40425-019-0579-z] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 03/26/2019] [Indexed: 12/17/2022] Open
Abstract
Background The Italian Renal Cell Cancer Early Access Program was an expanded access program that allowed access to nivolumab, for patients (pts) with metastatic renal cell carcinoma (mRCC) prior to regulatory approval. Methods Pts with previously treated advanced or mRCC were eligible to receive nivolumab 3 mg/kg every 2 weeks. Pts included in the analysis had received ≥1 dose of nivolumab and were monitored for drug-related adverse events (drAEs) using CTCAE v.4.0. Immune-related (ir) AEs were defined as AEs displaying a certain, likely or possible correlation with immunotherapy (cutaneous, endocrine, hepatic, gastro-intestinal and pulmonary). The association between overall survival (OS) and irAEs was assessed, and associations between variables were evaluated with a logistic regression model. Results A total of 389 pts were enrolled between July 2015 and April 2016. Overall, the objective response rate was 23.1%. At a median follow-up of 12 months, the median progression-free survival was 4.5 months (95% CI 3.7–6.2) and the 12-month overall survival rate was 63%. Any grade and grade 3–4 drAEs were reported in 124 (32%) and 27 (7%) of pts, respectively, and there were no treatment-related deaths. Any grade irAEs occurred in 76 (20%) of patients, 8% cutaneous, 4% endocrine, 2% hepatic, 5% gastro-intestinal and 1% pulmonary. Of the 22 drAEs inducing treatment discontinuation, 10 (45%) were irAEs. Pts with drAEs had a significantly longer survival than those without drAEs (median OS 22.5 versus 16.4 months, p = 0.01). Pts with irAEs versus without irAEs had a more significant survival benefit (median OS not reached versus 16.8 months, p = 0.002), confirmed at the landmark analysis at 6 weeks. The occurrence of irAEs displayed a strong association with OS in univariable (HR 0.48, p = 0.003) and multivariable (HR 0.57, p = 0.02) analysis. Conclusions The appearance of irAEs strongly correlates with survival benefit in a real-life population of mRCC pts treated with nivolumab. Electronic supplementary material The online version of this article (10.1186/s40425-019-0579-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elena Verzoni
- Medical Oncology-Genitourinary Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Venezian 1, 20133, Milan, Italy.
| | | | - Enrico Cortesi
- Radiology, Oncology and Pathology, Policlinico Umberto I, Rome, Italy
| | - Diana Giannarelli
- Biostatistical Unit, Regina Elena National Cancer Institute - IRCCS, Rome, Italy
| | | | - Roberto Sabbatini
- Oncology and Hematology Department, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Sebastiano Buti
- Medical Oncology, Azienda Ospedaliera di Parma, Parma, Italy
| | - Sabrina Rossetti
- Urology and Gynecology, Istituto Nazionale Tumori - IRCCS - Fondazione Pascale, Naples, Italy
| | - Francesco Cognetti
- Medical Oncology, Regina Elena National Cancer Institute - IRCCS, Rome, Italy
| | | | | | - Daniele Turci
- Medical Oncology, Ospedale Santa Maria delle Croci, Ravenna, Italy
| | - Cora N Sternberg
- Medical Oncology, Azienda Ospedaliera S. Camillo Forlanini, Rome, Italy
| | - Camillo Porta
- Department of Internal Medicine, University of Pavia and Division of Translational Oncology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | | | | | | | - Stefano Panni
- Medical Oncology, Istituti Ospitalieri di Cremona, Cremona, Italy
| | - Antonio Pazzola
- Medical Oncology, Ospedale Civile SS Annunziata, Sassari, Italy
| | | | - Alessandra Raimondi
- Medical Oncology-Genitourinary Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Venezian 1, 20133, Milan, Italy
| | - Ugo De Giorgi
- Urologic-Gynecologic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.), Meldola, Italy
| | - Giuseppe Procopio
- Medical Oncology-Genitourinary Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Venezian 1, 20133, Milan, Italy
| | | |
Collapse
|
36
|
Porcu M, De Silva P, Solinas C, Battaglia A, Schena M, Scartozzi M, Bron D, Suri JS, Willard-Gallo K, Sangiolo D, Saba L. Immunotherapy Associated Pulmonary Toxicity: Biology Behind Clinical and Radiological Features. Cancers (Basel) 2019; 11:cancers11030305. [PMID: 30841554 PMCID: PMC6468855 DOI: 10.3390/cancers11030305] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/17/2019] [Accepted: 02/26/2019] [Indexed: 12/22/2022] Open
Abstract
The broader use of immune checkpoint blockade in clinical routine challenges clinicians in the diagnosis and management of side effects which are caused by inflammation generated by the activation of the immune response. Nearly all organs can be affected by immune-related toxicities. However, the most frequently reported are: fatigue, rash, pruritus, diarrhea, nausea/vomiting, arthralgia, decreased appetite and abdominal pain. Although these adverse events are usually mild, reversible and not frequent, an early diagnosis is crucial. Immune-related pulmonary toxicity was most frequently observed in trials of lung cancer and of melanoma patients treated with the combination of the anti-cytotoxic T lymphocyte antigen (CTLA)-4 and the anti-programmed cell death-1 (PD-1) antibodies. The most frequent immune-related adverse event in the lung is represented by pneumonitis due to the development of infiltrates in the interstitium and in the alveoli. Clinical symptoms and radiological patterns are the key elements to be considered for an early diagnosis, rendering the differential diagnosis crucial. Diagnosis of immune-related pneumonitis may imply the temporary or definitive suspension of immunotherapy, along with the start of immuno-suppressive treatments. The aim of this work is to summarize the biological bases, clinical and radiological findings of lung toxicity under immune checkpoint blockade, underlining the importance of multidisciplinary teams for an optimal early diagnosis of this side effect, with the aim to reach an improved patient care.
Collapse
Affiliation(s)
- Michele Porcu
- Department of Radiology, University Hospital of Cagliari, 09042 Monserrato (Cagliari), Italy.
| | - Pushpamali De Silva
- Molecular Immunology Unit, Institut Jules Bordet, Universitè Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
- Clinical and Experimental Hematology, Institute Jules Bordet, Universitè Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
| | - Cinzia Solinas
- Molecular Immunology Unit, Institut Jules Bordet, Universitè Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
- Department of Medical Oncology and Hematology, Regional Hospital of Aosta, 11100 Aosta, Italy.
| | - Angelo Battaglia
- Department of Medical Oncology and Hematology, Regional Hospital of Aosta, 11100 Aosta, Italy.
| | - Marina Schena
- Department of Medical Oncology and Hematology, Regional Hospital of Aosta, 11100 Aosta, Italy.
| | - Mario Scartozzi
- Department of Medical Oncology, University Hospital of Cagliari, 09042 Monserrato (Cagliari), Italy.
| | - Dominique Bron
- Clinical and Experimental Hematology, Institute Jules Bordet, Universitè Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
| | - Jasjit S Suri
- Lung Diagnostic Division, Global Biomedical Technologies, Inc., Roseville, CA 95661, USA.
- AtheroPoint™ LLC, Roseville, CA 95661, USA.
| | - Karen Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet, Universitè Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
| | - Dario Sangiolo
- Department of Oncology, University of Torino, 10043 Orbassano (Torino), Italy.
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo (Torino), Italy.
| | - Luca Saba
- Department of Radiology, University Hospital of Cagliari, 09042 Monserrato (Cagliari), Italy.
| |
Collapse
|
37
|
The Risks and Benefits of Immune Checkpoint Blockade in Anti-AChR Antibody-Seropositive Non-Small Cell Lung Cancer Patients. Cancers (Basel) 2019; 11:cancers11020140. [PMID: 30682845 PMCID: PMC6407108 DOI: 10.3390/cancers11020140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Anti-programmed cell death 1 (PD-1) monoclonal antibodies (Abs) unleash an immune response to cancer. However, a disruption of the immune checkpoint function by blocking PD-1/PD-ligand 1(PD-L1) signaling may trigger myasthenia gravis (MG) as a life-threatening immune-related adverse event. MG is a neuromuscular disease and is closely associated with being positive for anti-acetylcholine receptor (anti-AChR) Abs, which are high specific and diagnostic Abs for MG. Methods: A 72-year-old man was diagnosed with chemotherapy-refractory lung squamous cell carcinoma and nivolumab was selected as the third-line regimen. We describe the first report of an anti-AChR Ab-seropositive lung cancer patient achieving a durable complete response (CR) to an anti-PD-1 antibody therapy. To further explore this case, we performed multiplex immunofluorescence analysis on a pretreatment tumor. Results: The patient achieved a durable CR without developing MG. However, the levels of anti-AChR Abs were elevated during two years of anti-PD-1 antibody therapy. The tumor of the subclinical MG patient had high PD-L1 expression and an infiltrated–inflamed tumor immune microenvironment. Conclusions: This study suggests that immune checkpoint inhibitors can be safely used and provide the benefits for advanced cancer patients with immunologically ‘hot’ tumor even if anti-AChR Abs are positive. Although careful monitoring clinical manifestation in consultation with neurologist is needed, immune checkpoint inhibitors should be considered as a treatment option for asymptomatic anti-AChR Ab-seropositive cancer patients.
Collapse
|
38
|
Omar HA, Tolba MF. Tackling molecular targets beyond PD-1/PD-L1: Novel approaches to boost patients' response to cancer immunotherapy. Crit Rev Oncol Hematol 2019; 135:21-29. [PMID: 30819443 DOI: 10.1016/j.critrevonc.2019.01.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/14/2019] [Accepted: 01/18/2019] [Indexed: 12/31/2022] Open
Abstract
In the new era of immunotherapy, which has changed the clinical oncology practice guidelines, there is a pressing need for finding novel approaches to tune up the clinical outcomes of immunotherapy and extend its benefits to a wider cohort of cancer patients. Several non-classical molecular immune targets beyond PD-1/PD-L1 signaling were shown to be engaged as feedback resistance circuits to shut down the antitumor immune response mediated by the classical immune checkpoint inhibitors. Those include T-cell inducible co-stimulator (ICOS), CD40, CD47, V-domain Ig suppressor of T-cell activation (VISTA), cyclin-dependent kinase (CDK)12, enhancer of Zeste homolog 2 (EZH2), toll-like receptors (TLRs) and OX-40 (CD134). Herein we critically discussed the latest studies concerned with understanding the mechanisms involved in the negative clinical response to classical immunotherapies and strategies to optimize the efficacy of cancer immunotherapy through novel combinatorial approaches.
Collapse
Affiliation(s)
- Hany A Omar
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Pharmacology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Mai F Tolba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt; Biology Department, School of Sciences and Engineering, The American University in Cairo, New Cairo, Egypt.
| |
Collapse
|
39
|
Targeting Multiple Receptors to Increase Checkpoint Blockade Efficacy. Int J Mol Sci 2019; 20:ijms20010158. [PMID: 30621125 PMCID: PMC6337574 DOI: 10.3390/ijms20010158] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/20/2018] [Accepted: 12/28/2018] [Indexed: 12/30/2022] Open
Abstract
Immune checkpoint blockade therapy is a powerful treatment strategy for many cancer types. Many patients will have limited responses to monotherapy targeted to a single immune checkpoint. Both inhibitory and stimulatory immune checkpoints continue to be discovered. Additionally, many receptors previously identified to play a role in tumor formation and progression are being found to have immunomodulatory components. The success of immunotherapy depends on maximizing pro-anti-tumor immunity while minimizing immunosuppressive signaling. Combining immune checkpoint targeted approaches with each other or with other receptor targets is a promising schema for future therapeutic regimen designs.
Collapse
|
40
|
Onesti CE, Frères P, Jerusalem G. Atypical patterns of response to immune checkpoint inhibitors: interpreting pseudoprogression and hyperprogression in decision making for patients' treatment. J Thorac Dis 2019; 11:35-38. [PMID: 30863564 DOI: 10.21037/jtd.2018.12.47] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Concetta Elisa Onesti
- Laboratory of Human Genetics, GIGA Research Institute, University of Liège, Liège, Belgium.,Department of Medical Oncology, University Hospital (CHU Liège), Liège, Belgium.,Laboratory of Medical Oncology, GIGA Research Institute, University of Liège, Liège, Belgium
| | - Pierre Frères
- Department of Medical Oncology, University Hospital (CHU Liège), Liège, Belgium
| | - Guy Jerusalem
- Department of Medical Oncology, University Hospital (CHU Liège), Liège, Belgium.,Laboratory of Medical Oncology, GIGA Research Institute, University of Liège, Liège, Belgium
| |
Collapse
|
41
|
Alekseenko IV, Monastyrskaya GS, Sverdlov ED. Emerging Potential of Cancer Therapy—Binary Direct Interactions of Cancer and Stromal Cells. RUSS J GENET+ 2018. [DOI: 10.1134/s1022795418120025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
Kumar P, Bhattacharya P, Prabhakar BS. A comprehensive review on the role of co-signaling receptors and Treg homeostasis in autoimmunity and tumor immunity. J Autoimmun 2018; 95:77-99. [PMID: 30174217 PMCID: PMC6289740 DOI: 10.1016/j.jaut.2018.08.007] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/22/2018] [Accepted: 08/26/2018] [Indexed: 12/22/2022]
Abstract
The immune system ensures optimum T-effector (Teff) immune responses against invading microbes and tumor antigens while preventing inappropriate autoimmune responses against self-antigens with the help of T-regulatory (Treg) cells. Thus, Treg and Teff cells help maintain immune homeostasis through mutual regulation. While Tregs can contribute to tumor immune evasion by suppressing anti-tumor Teff response, loss of Treg function can result in Teff responses against self-antigens leading to autoimmune disease. Thus, loss of homeostatic balance between Teff/Treg cells is often associated with both cancer and autoimmunity. Co-stimulatory and co-inhibitory receptors, collectively known as co-signaling receptors, play an indispensable role in the regulation of Teff and Treg cell expansion and function and thus play critical roles in modulating autoimmune and anti-tumor immune responses. Over the past three decades, considerable efforts have been made to understand the biology of co-signaling receptors and their role in immune homeostasis. Mutations in co-inhibitory receptors such as CTLA4 and PD1 are associated with Treg dysfunction, and autoimmune diseases in mice and humans. On the other hand, growing tumors evade immune surveillance by exploiting co-inhibitory signaling through expression of CTLA4, PD1 and PDL-1. Immune checkpoint blockade (ICB) using anti-CTLA4 and anti-PD1 has drawn considerable attention towards co-signaling receptors in tumor immunology and created renewed interest in studying other co-signaling receptors, which until recently have not been as well studied. In addition to co-inhibitory receptors, co-stimulatory receptors like OX40, GITR and 4-1BB have also been widely implicated in immune homeostasis and T-cell stimulation, and use of agonistic antibodies against OX40, GITR and 4-1BB has been effective in causing tumor regression. Although ICB has seen unprecedented success in cancer treatment, autoimmune adverse events arising from ICB due to loss of Treg homeostasis poses a major obstacle. Herein, we comprehensively review the role of various co-stimulatory and co-inhibitory receptors in Treg biology and immune homeostasis, autoimmunity, and anti-tumor immunity. Furthermore, we discuss the autoimmune adverse events arising upon targeting these co-signaling receptors to augment anti-tumor immune responses.
Collapse
Affiliation(s)
- Prabhakaran Kumar
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL, USA
| | - Palash Bhattacharya
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL, USA
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois-College of Medicine, Chicago, IL, USA; Department of Ophthalmology, Associate Dean for Technological Innovation and Training, University of Illinois College of Medicine, Room E-705, (M/C 790), 835 S. Wolcott Ave, Chicago, IL, 60612, USA.
| |
Collapse
|
43
|
Wang J, Jebbawi F, Bellanger AP, Beldi G, Millon L, Gottstein B. Immunotherapy of alveolar echinococcosis via PD-1/PD-L1 immune checkpoint blockade in mice. Parasite Immunol 2018; 40:e12596. [PMID: 30315719 PMCID: PMC6587932 DOI: 10.1111/pim.12596] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/04/2018] [Accepted: 10/08/2018] [Indexed: 01/01/2023]
Abstract
The growth potential of the tumour‐like Echinococcus multilocularis metacestode (causing alveolar echinococcosis, AE) is directly dependent upon the nature/function of the periparasitic adaptive host immune‐mediated processes. PD‐1/PD‐L1 pathway (programmed cell death 1), which inhibits lymphocytic proliferation in tumour development, is over‐expressed at the chronic stage of AE. We tested the impact of a PD‐1/PD‐L1 pathway blockade on the outcome of both chronic AE (intraperitoneal metacestode inoculation, secondary AE and SAE) and acute AE (peroral egg infection, primary AE and PAE). To assess the parasite proliferation potential, we measured parasite mass weight for SAE and liver lesion number for PAE. In both models, the parasite load was significantly decreased in response to anti‐PD‐L1 antibody treatment. In SAE, anti‐PDL1 administration was associated with increased Th1 response parameters and decreased Treg responses, while in PAE anti‐PDL1 administration was associated with fewer lesions in the liver and decreased Treg/Th2 responses. Our findings highly suggested that a PD‐1/PD‐L1 pathway blockade triggered the host immune responses in favour of an immune‐mediated control of E. multilocularis proliferation. Based on this, future studies that combine PD‐1/PD‐L1 blockade with a parasitostatic albendazole medication may yield in a putatively curative therapeutic approach to control alveolar echinococcosis.
Collapse
Affiliation(s)
- Junhua Wang
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Parasitology, University of Bern, Bern, Switzerland
| | - Fadi Jebbawi
- Department of Visceral Surgery and Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Anne-Pauline Bellanger
- Chrono-Environment UMR/CNRS 6249, University of Bourgogne Franche-Comté, Besançon, France.,Parasitology Mycology Department, University Hospital Jean Minjoz, Besancon, France
| | - Guido Beldi
- Department of Visceral Surgery and Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Laurence Millon
- Chrono-Environment UMR/CNRS 6249, University of Bourgogne Franche-Comté, Besançon, France.,Parasitology Mycology Department, University Hospital Jean Minjoz, Besancon, France
| | - Bruno Gottstein
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Parasitology, University of Bern, Bern, Switzerland
| |
Collapse
|
44
|
Johnson MD. PD-L1 expression in meningiomas. J Clin Neurosci 2018; 57:149-151. [DOI: 10.1016/j.jocn.2018.08.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 08/13/2018] [Indexed: 01/12/2023]
|
45
|
Cui C, Yu B, Jiang Q, Li X, Shi K, Yang Z. The roles of PD-1/PD-L1 and its signalling pathway in gastrointestinal tract cancers. Clin Exp Pharmacol Physiol 2018; 46:3-10. [PMID: 30161295 DOI: 10.1111/1440-1681.13028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/24/2018] [Accepted: 08/26/2018] [Indexed: 12/31/2022]
Abstract
Cancer immunotherapy has been increasingly applied in the treatment of advanced malignancies. Consequently, immune checkpoints have become a major concern. As PD-1 is an important immunomodulatory protein, the blockade of PD-1 and its ligand PD-L1 is a promising tumour immunotherapy for human carcinoma. In this review, we first discuss the role of the PD-1/PD-L1 interaction in gastrointestinal tract cancers. Targeting PD-1 and PD-L1 in immune cells and tumour cells may show remarkable efficiency in gastrointestinal tract cancers. Second, the PD-1/PD-L1-associated signalling pathway involved in cancer immunotherapy in gastrointestinal tract cancers is discussed. Most importantly, this review summarizes the PD-1/PD-L1-targeted immunotherapy combinations with relevant signalling pathways, which may result in a breakthrough for the treatment of gastrointestinal tract cancers, such as gastric cancer, colorectal cancer and liver cancer. Meanwhile, the review provides a deeper insight into the mechanism of checkpoint blockade immunotherapies.
Collapse
Affiliation(s)
- Chunguo Cui
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bo Yu
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qi Jiang
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xingfang Li
- 2nd Hospital of Jilin University, Changchun City, China
| | - Kaiyao Shi
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zecheng Yang
- 2nd Hospital of Jilin University, Changchun City, China
| |
Collapse
|
46
|
Sverdlov E. Missed Druggable Cancer Hallmark: Cancer-Stroma Symbiotic Crosstalk as Paradigm and Hypothesis for Cancer Therapy. Bioessays 2018; 40:e1800079. [DOI: 10.1002/bies.201800079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/15/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Eugene Sverdlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences; Ulitsa Miklukho-Maklaya, 16/10 117997 Moscow Russia
| |
Collapse
|
47
|
Rataj F, Kraus FBT, Chaloupka M, Grassmann S, Heise C, Cadilha BL, Duewell P, Endres S, Kobold S. PD1-CD28 Fusion Protein Enables CD4+ T Cell Help for Adoptive T Cell Therapy in Models of Pancreatic Cancer and Non-hodgkin Lymphoma. Front Immunol 2018; 9:1955. [PMID: 30214445 PMCID: PMC6125378 DOI: 10.3389/fimmu.2018.01955] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/08/2018] [Indexed: 01/22/2023] Open
Abstract
Background: Interaction of the programmed death receptor 1 (PD-1) and its ligand, PD-L1, suppresses T cell activity and permits tumors to evade T cell-mediated immune surveillance. We have recently demonstrated that antigen-specific CD8+ T cells transduced with a PD1-CD28 fusion protein are protected from PD-1-mediated inhibition. We have now investigated the potential of PD1-CD28 fusion protein-transduced CD4+ T cells alone or in combination with CD8+ T cells for immunotherapy of pancreatic cancer and non-Hodgkin lymphoma. Methods: OVA-specific CD4+ and CD8+ were retrovirally transduced with the PD1-CD28 fusion protein. Cytokine release, proliferation, cytotoxic activity, and phenotype of transduced T cells were assessed in the context of Panc02-OVA (murine pancreatic cancer model) and E.G7-PD-L1 (murine T cell lymphoma model) cells. Results: Stimulation of PD1-CD28 fusion protein-transduced CD4+ T cells with anti-CD3 and recombinant PD-L1 induced specific T cell activation, as measured by IFN-y release and T cell proliferation. Coculture with Panc02-OVA or E.G7-PD-L1 tumor cells also led to specific activation of CD4+ T cells. Cytokine release and T cell proliferation was most effective when tumor cells simultaneously encountered genetically engineered CD4+ and CD8+ T cells. Synergy between both cell populations was also observed for specific tumor cell lysis. T cell cytotoxicity was mediated via granzyme B release and mediated enhanced tumor control in vivo. Transduced CD4+ and CD8+ T cells in co-culture with tumor cells developed a predominant central memory phenotype over time. Different ratios of CD4+ and CD8+ transduced T cells led to a significant increase of IFN-y and IL-2 secretion positively correlating with CD4+ T cell numbers used. Mechanistically, IL-2 and MHC-I were central to the synergistic activity of CD4+ and CD8+ T cells, since neutralization of IL-2 prevented the crosstalk between these cell populations. Conclusion: PD1-CD28 fusion protein-transduced CD4+ T cells significantly improved anti-tumoral effect of fusion protein-transduced CD8+ T cells. Thus, our results indicate that PD1-CD28 fusion protein-transduced CD4+ T cells have the potential to overcome the PD-1-PD-L1 immunosuppressive axis in pancreatic cancer and non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Felicitas Rataj
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Fabian B T Kraus
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Michael Chaloupka
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Simon Grassmann
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Constanze Heise
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Bruno L Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Peter Duewell
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany.,German Cancer Research Center (DKTK), Partner Site Munich, Heidelberg, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Member of the German Center for Lung Research (DZL), Munich, Germany.,German Cancer Research Center (DKTK), Partner Site Munich, Heidelberg, Germany
| |
Collapse
|
48
|
Ito M, Oshima Y, Yajima S, Suzuki T, Nanami T, Shiratori F, Funahashi K, Nemoto T, Shimada H. Is high serum programmed death ligand 1 level a risk factor for poor survival in patients with gastric cancer? Ann Gastroenterol Surg 2018; 2:313-318. [PMID: 30003194 PMCID: PMC6036390 DOI: 10.1002/ags3.12175] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 05/04/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND AIM Although the clinicopathological significance of the expression of programmed death ligand 1(PD-L1) in various cancer tissues has been reported, serum PD-L1 level has not been evaluated in patients with surgically treated gastric cancer. Therefore, we evaluated the clinicopathological characteristics and prognostic significance of preoperative serum PD-L1 levels in patients with gastric cancer. PATIENTS AND METHODS Serum samples were obtained before surgery from 152 patients with gastric cancer, including 75 patients with stage I, 31 with stage II, 23 with stage III, and 23 with stage IV gastric cancer. The samples were analyzed using enzyme-linked immunosorbent assay to detect soluble PD-L1. Using the median serum PD-L1 level of 50 pg/mL, patients were divided into two groups, namely high serum and low serum PD-L1 level groups. Clinicopathological characteristics and prognosis were compared between these two groups using univariate and multivariate analysis. RESULTS Serum PD-L1 level was significantly associated with older age, positive cancer antigen 19-9 (CA19-9), C-reactive protein levels, and albumin levels but not with tumor stage. Patients in the high serum PD-L1 group showed significantly worse overall survival and recurrence-free survival than those in the low serum PD-L1 group (P < .05). Multivariate analysis showed that high serum PD-L1 level was an independent risk factor for poor overall survival (P = .02). CONCLUSION High serum PD-L1 level was a prognostic factor for reduced overall survival in patients with surgically treated gastric cancer.
Collapse
Affiliation(s)
- Masaaki Ito
- Department of SurgeryToho University School of MedicineTokyoJapan
- Department of Clinical OncologyToho University School of MedicineTokyoJapan
| | - Yoko Oshima
- Department of SurgeryToho University School of MedicineTokyoJapan
| | - Satoshi Yajima
- Department of SurgeryToho University School of MedicineTokyoJapan
| | - Takashi Suzuki
- Department of SurgeryToho University School of MedicineTokyoJapan
| | - Tatsuki Nanami
- Department of SurgeryToho University School of MedicineTokyoJapan
| | | | | | - Tetsuo Nemoto
- Department of Surgical PathologyToho University School of MedicineTokyoJapan
| | - Hideaki Shimada
- Department of SurgeryToho University School of MedicineTokyoJapan
- Department of Clinical OncologyToho University School of MedicineTokyoJapan
| |
Collapse
|
49
|
Design, synthesis and anticancer evaluation of novel spirobenzo[h]chromene and spirochromane derivatives with dual EGFR and B-RAF inhibitory activities. Eur J Med Chem 2018; 150:567-578. [DOI: 10.1016/j.ejmech.2018.03.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 01/16/2023]
|