1
|
Ghorbaninezhad F, Nour MA, Farzam OR, Saeedi H, Vanan AG, Bakhshivand M, Jafarlou M, Hatami-Sadr A, Baradaran B. The tumor microenvironment and dendritic cells: Developers of pioneering strategies in colorectal cancer immunotherapy? Biochim Biophys Acta Rev Cancer 2025; 1880:189281. [PMID: 39929377 DOI: 10.1016/j.bbcan.2025.189281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Colorectal cancer (CRC) is the world's third most frequent cancer, and both its incidence and fatality rates are rising. Despite various therapeutic approaches, neither its mortality rate nor its recurrence frequency has decreased significantly. Additionally, conventional treatment approaches, such as chemotherapy and radiotherapy, have several side effects and risks for patients with CRC. Accordingly, the need for alternative and effective treatments for CRC patients is critical. Immunotherapy that utilizes dendritic cells (DCs) harnesses the patient's immune system to combat cancer cells effectively. DCs are the most potent antigen-presenting cells (APCs), which play a vital role in generating anti-cancer T cell responses. A significant barrier to the immune system's ability to eliminate CRC is the establishment of a potent immunosuppressive tumor milieu by malignant cells. Since DCs are frequently defective in this milieu, the tumor setting significantly reduces the effectiveness of DC-based therapy. Determining central mechanisms contributing to tumor growth by unraveling and comprehending the interaction between CRC tumor milieu and DCs may lead to new therapeutic approaches. This study aims to review DC biology and discuss its role in T-cell-mediated anti-tumor immunity, as well as to highlight the immunosuppressive effects of the CRC tumor milieu on the function of DCs. We will also highlight the tumor microenvironment (TME)-related factors that interfere with DC function as a possible therapeutic target to enhance DC-based cell therapy efficacy.
Collapse
Affiliation(s)
- Farid Ghorbaninezhad
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mina Afrashteh Nour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Ghorbani Vanan
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Bakhshivand
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Jafarlou
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Behzad Baradaran
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Sheykhhasan M, Ahmadieh-Yazdi A, Heidari R, Chamanara M, Akbari M, Poondla N, Yang P, Malih S, Manoochehri H, Tanzadehpanah H, Mahaki H, Fayazi Hosseini N, Dirbaziyan A, Al-Musawi S, Kalhor N. Revolutionizing cancer treatment: The power of dendritic cell-based vaccines in immunotherapy. Biomed Pharmacother 2025; 184:117858. [PMID: 39955851 DOI: 10.1016/j.biopha.2025.117858] [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: 10/01/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 02/18/2025] Open
Abstract
In the modern time, cancer immunotherapies have increasingly become vital treatment options, joining long-established methods like surgery, chemotherapy, and radiotherapy treatment. Central to this emerging approach are dendritic cells (DCs), which boast a remarkable ability for antigen presentation. This ability is being leveraged to modulate T and B cell immunity, offering a groundbreaking strategy for tackling cancer. However, the percentage of patients experiencing meaningful benefits from this treatment remains relatively low, underscoring the ongoing necessity for further research and development in this field. This review offers a comprehensive analysis of the present-day progress in dendritic cell (DC)-based vaccines and recent efforts to enhance their efficacy. We explore the intricacies of DC function, from antigen capture to T cell stimulation, and discuss the outcomes of both preclinical and clinical trials across various cancer types. While the results are promising, the real-world application of DC-based vaccines is still nascent, posing multiple challenges that need to be overcome. These obstacles include optimizing the methods for DC generation and antigen loading, overcoming the immunosuppressive nature of the tumor microenvironment, and enhancing specificities of the immunologic response through personalized vaccines. The review concludes by emphasizing prospective opportunities for future research and emphasizing the critical need for extensive clinical trials. These trials are essential to validate the effectivity of DC-based vaccines and solidify their role in the broader spectrum of cancer immunotherapy options.
Collapse
Affiliation(s)
- Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| | - Amirhossein Ahmadieh-Yazdi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Reza Heidari
- Infectious Diseases Research Center, AJA University of Medical Sciences, Tehran, Iran; Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran; Medical Biotechnology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran; Student research committee, AJA University of Medical Sciences, Tehran, Iran
| | - Mohammad Akbari
- Department of Medical School, Faculty of Medical Sciences, Islamic Azad University, Tonekabon Branch, Mazandaran, Iran
| | - Naresh Poondla
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Global Health Research, Saveetha Medical College & Hospital, Chennai, India
| | - Piao Yang
- Department of Molecular Genetics, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Sara Malih
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, USA; Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamed Manoochehri
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Basic Science Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nashmin Fayazi Hosseini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ashkan Dirbaziyan
- Department of Microbiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | | | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran
| |
Collapse
|
3
|
Zhou Z, Zhang Y, Zhou Y, Gu J, Li J, Shao J, Feng N. Construction and verification of a prognostic model for bladder cancer based on disulfidptosis-related angiogenesis genes. PeerJ 2025; 13:e18911. [PMID: 39995998 PMCID: PMC11849515 DOI: 10.7717/peerj.18911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/06/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Bladder cancer (BLCA) is the most common malignancy of the urinary system and one of the most common cancers worldwide. This study seeks to examine the influence of angiogenesis-related genes (ARGs) linked to disulfidptosis on BLCA patients and to formulate a prognostic model for evaluating their prognosis and response to immunotherapy. METHODS This study used sequencing data of BLCA in the Cancer Genome Atlas (TCGA) database. Unsupervised consensus clustering analysis, cox regression analysis, and least absolute shrinkage and selection operator (LASSO) regression analysis were used to screen hub genes and construct a related prognostic risk model. The receiver operating characteristic (ROC) curve and independent prognostic analysis were then used to verify the predictive performance of the signature genes. Clinical characteristics, immune status, and Tumor Mutation Burden (TMB) of the prognostic risk model were evaluated. The expression levels of model genes within standard bladder epithelial cell lines (SV-HUC-1) and bladder cancer cell lines (T24 and SW1710) were quantified through qRT-PCR. RESULTS The constructed prognostic risk model can be used as an independent risk indicator for BLCA and was validated in an external dataset. Immune cell infiltration analysis showed that CD8+T cells, Tregs and dendritic cells were significantly different between the two groups. A significant increase was observed in the Stromal score, Immune score and ESTIMATE score in the high-risk group compared with the low-risk group. The Immune Exclusion score and Tumor Immune Dysfunction and Exclusion (TIDE) score of the high-risk group were higher than those of the low-risk score group. Compared with the normal bladder epithelial cell line (SV-HUC-1), the expression levels of 2 model genes (COL5A2 and SCG2) in bladder cancer cell lines (T24 and SW1710) were significantly elevated. CONCLUSION This study helps us understand the characteristics of disulfidptosis-related subgroups. The characteristics of disulfidptosis-related ARGs may be used to evaluate the prognosis and immunotherapy response of BLCA patients.
Collapse
Affiliation(s)
- Zhihao Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Yuwei Zhang
- Medical School of Nantong University, Nantong, China
| | - Yuhua Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Jiayu Gu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Jufa Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Jianfeng Shao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Ninghan Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| |
Collapse
|
4
|
Zhu Z, Chu Z, Fei F, Wu C, Fei Z, Sun Y, Chen Y, Lu P. Neo-BCV: A Novel Bacterial Liquid Complex Vaccine for Enhancing Dendritic Cell-Mediated Immune Responses Against Lung Cancer. Vaccines (Basel) 2025; 13:64. [PMID: 39852843 PMCID: PMC11768841 DOI: 10.3390/vaccines13010064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND In the past decade, immunotherapy has become a major choice for the treatment of lung cancer, yet its therapeutic efficacy is still relatively limited due to the various immune escape mechanisms of tumors. Based on this, we introduce Neo-BCV, a novel bacterial composite vaccine designed to enhance immune responses against lung cancer. METHODS We investigated the immune enhancing effect of Neo-BCV through in vivo and in vitro experiments, including flow cytometry, RNA-seq, and Western blot. RESULTS We have demonstrated that Neo-BCV can promote Dendritic cells (DCs) maturation and induce DCs differentiation into pro-inflammatory subgroups, significantly enhancing cytotoxic T lymphocyte (CTL)-mediated anti-tumor responses. Transcriptome sequencing revealed that Neo-BCV exerts its effects by specifically inhibiting the JAK2-STAT3 signaling pathway, a crucial regulator of cancer progression, metabolism, and inflammation. Moreover, Neo-BCV significantly improved the immune microenvironment in both tumor and spleen tissues without inducing notable toxic effects in major organs. CONCLUSIONS These findings highlight Neo-BCV's potential as a safe and effective therapeutic strategy, offering a novel avenue for clinical translation in lung cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yun Chen
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China; (Z.Z.); (Z.C.); (F.F.); (C.W.); (Z.F.); (Y.S.)
| | - Peihua Lu
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China; (Z.Z.); (Z.C.); (F.F.); (C.W.); (Z.F.); (Y.S.)
| |
Collapse
|
5
|
Ng AT, Steve T, Jamouss KT, Arham A, Kawtharani S, Assi HI. The challenges and clinical landscape of glioblastoma immunotherapy. CNS Oncol 2024; 13:2415878. [PMID: 39469854 PMCID: PMC11524205 DOI: 10.1080/20450907.2024.2415878] [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: 02/03/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Glioblastoma is associated with a dismal prognosis with the standard of care involving surgery, radiation therapy and temozolomide chemotherapy. This review investigates the features that make glioblastoma difficult to treat and the results of glioblastoma immunotherapy clinical trials so far. There have been over a hundred clinical trials involving immunotherapy in glioblastoma. We report the survival-related outcomes of every Phase III glioblastoma immunotherapy trial with online published results we could find at the time of writing. To date, the DCVax-L vaccine is the only immunotherapy shown to have statistically significant increased median survival compared with standard-of-care in a Phase III trial: 19.3 months versus 16.5 months. However, this trial used an external control group to compare with the intervention which limits its quality of evidence. In conclusion, glioblastoma immunotherapy requires further investigation to determine its significance in improving disease survival.
Collapse
Affiliation(s)
- Andrew Timothy Ng
- Department of Medicine, University of Massachusetts Chan Medical School – Baystate Campus, Springfield, MA01199, USA
| | - Tyler Steve
- Department of Medicine, University of Massachusetts Chan Medical School – Baystate Campus, Springfield, MA01199, USA
| | - Kevin T Jamouss
- Department of Medicine, University of Massachusetts Chan Medical School – Baystate Campus, Springfield, MA01199, USA
| | - Abdul Arham
- Department of Medicine, University of Massachusetts Chan Medical School – Baystate Campus, Springfield, MA01199, USA
| | - Sarah Kawtharani
- Department of Neurosurgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Hazem I Assi
- Department of Hematology and Oncology, American University of Beirut Medical Center, Beirut, 1107 2020, Lebanon
| |
Collapse
|
6
|
Kaviyarasan V, Das A, Deka D, Saha B, Banerjee A, Sharma NR, Duttaroy AK, Pathak S. Advancements in immunotherapy for colorectal cancer treatment: a comprehensive review of strategies, challenges, and future prospective. Int J Colorectal Dis 2024; 40:1. [PMID: 39731596 DOI: 10.1007/s00384-024-04790-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/15/2024] [Indexed: 12/30/2024]
Abstract
PURPOSE Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Metastatic colorectal cancer (mCRC) continues to present significant challenges, particularly in patients with proficient mismatch repair/microsatellite stable (pMMR/MSS) tumors. This narrative review aims to provide recent developments in immunotherapy for CRC treatment, focusing on its efficacy and challenges. METHODS This review discussed the various immunotherapeutic strategies for CRC treatment, including immune checkpoint inhibitors (ICIs) targeting PD-1 and PD-L1, combination therapies involving ICIs with other modalities, chimeric antigen receptor T-cell (CAR-T) cell therapy, and cancer vaccines. The role of the tumor microenvironment and immune evasion mechanisms was also explored to understand their impact on the effectiveness of these therapies. RESULTS This review provides a comprehensive update of recent advancements in immunotherapy for CRC, highlighting the potential of various immunotherapeutic approaches, including immune checkpoint inhibitors, combination therapies, CAR-T therapy, and vaccination strategies. The results of checkpoint inhibitors, particularly in patients with MSI-H/dMMR tumors, which have significant improvements in survival rates have been observed. Furthermore, this review also addresses the challenges faced in treating pMMR/MSS CRC, which remains resistant to immunotherapy. CONCLUSION Immunotherapy plays a significant role in the treatment of CRC, particularly in patients with MSI-H/dMMR tumors. However, many challenges remain, especially in treating pMMR/MSS CRC. This review discussed the need for further research into combination therapies, biomarker development, CAR-T cell therapy, and a deeper understanding of immune evasion mechanisms for CRC treatment.
Collapse
Affiliation(s)
- Vaishak Kaviyarasan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Alakesh Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Dikshita Deka
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Biki Saha
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India.
| | - Neeta Raj Sharma
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India.
| |
Collapse
|
7
|
Kim EH, Teerdhala SV, Padilla MS, Joseph RA, Li JJ, Haley RM, Mitchell MJ. Lipid nanoparticle-mediated RNA delivery for immune cell modulation. Eur J Immunol 2024; 54:e2451008. [PMID: 39279550 PMCID: PMC11628889 DOI: 10.1002/eji.202451008] [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: 01/12/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/18/2024]
Abstract
Lipid nanoparticles (LNPs) have emerged as the preeminent nonviral drug delivery vehicles for nucleic acid therapeutics, as exemplified by their usage in the mRNA COVID-19 vaccines. As a safe and highly modular delivery platform, LNPs are attractive for a wide range of applications. In addition to vaccines, LNPs are being utilized as platforms for other immunoengineering efforts, especially as cancer immunotherapies by modulating immune cells and their functionality via nucleic acid delivery. In this review, we focus on the methods and applications of LNP-based immunotherapy in five cell types: T cells, NK cells, macrophages, stem cells, and dendritic cells. Each of these cell types has wide-reaching applications in immunotherapy but comes with unique challenges and delivery barriers. By combining knowledge of immunology and nanotechnology, LNPs can be developed for improved immune cell targeting and transfection, ultimately working toward novel clinical therapeutics.
Collapse
Affiliation(s)
- Emily H. Kim
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sridatta V. Teerdhala
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Marshall S. Padilla
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ryann A. Joseph
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jacqueline J. Li
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Rebecca M. Haley
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Michael J. Mitchell
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Abramson Cancer CenterPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Cellular ImmunotherapiesPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Institute for RNA InnovationPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for ImmunologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Cardiovascular InstitutePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Regenerative MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
8
|
Su L, Wang Z, Cai M, Wang Q, Wang M, Yang W, Gong Y, Fang F, Xu L. Single-cell analysis of matrisome-related genes in breast invasive carcinoma: new avenues for molecular subtyping and risk estimation. Front Immunol 2024; 15:1466762. [PMID: 39493752 PMCID: PMC11530991 DOI: 10.3389/fimmu.2024.1466762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Background The incidence of breast cancer remains high and severely affects human health. However, given the heterogeneity of tumor cells, identifying additional characteristics of breast cancer cells is essential for accurate treatment. Purpose This study aimed to analyze the relevant characteristics of matrix genes in breast cancer through the multigroup data of a breast cancer multi-database. Methods The related characteristics of matrix genes in breast cancer were analyzed using multigroup data from the breast cancer multi database in the Cancer Genome Atlas, and the differential genes of breast cancer matrix genes were identified using the elastic net penalty logic regression method. The risk characteristics of matrix genes in breast cancer were determined, and matrix gene expression in different breast cancer cells was evaluated using real-time fluorescent quantitative polymerase chain reaction (PCR). A consensus clustering algorithm was used to identify the biological characteristics of the population based on the matrix molecular subtypes in breast cancer, followed by gene mutation, immune correlation, pathway, and ligand-receptor analyses. Results This study reveals the genetic characteristics of cell matrix related to breast cancer. It is found that 18.1% of stromal genes are related to the prognosis of breast cancer, and these genes are mostly concentrated in the biological processes related to metabolism and cytokines in protein. Five different matrix-related molecular subtypes were identified by using the algorithm, and it was found that the five molecular subtypes were obviously different in prognosis, immune infiltration, gene mutation and drug-making gene analysis. Conclusions This study involved analyzing the characteristics of cell-matrix genes in breast cancer, guiding the precise prevention and treatment of the disease.
Collapse
Affiliation(s)
- Lingzi Su
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhe Wang
- The First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Mengcheng Cai
- The First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Qin Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Man Wang
- The First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Wenxiao Yang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yabin Gong
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fanfu Fang
- The First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Ling Xu
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
9
|
Kyr M, Mudry P, Polaskova K, Dubska LZ, Demlova R, Kubatova J, Hlavackova E, Pilatova KC, Mazanek P, Vejmelkova K, Dusek V, Tinka P, Balaz M, Merta T, Kuttnerova Z, Turekova T, Pavelka Z, Pokorna P, Palova H, Mlnarikova M, Jezova M, Kellnerova R, Kozakova S, Slaby O, Valik D, Sterba J. Personalized dendritic cell vaccine in multimodal individualized combination therapy improves survival in high-risk pediatric cancer patients. Int J Cancer 2024; 155:1443-1454. [PMID: 38958237 DOI: 10.1002/ijc.35062] [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: 08/06/2023] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 07/04/2024]
Abstract
A lot of hope for high-risk cancers is being pinned on immunotherapy but the evidence in children is lacking due to the rarity and limited efficacy of single-agent approaches. Here, we aim to assess the effectiveness of multimodal therapy comprising a personalized dendritic cell (DC) vaccine in children with relapsed and/or high-risk solid tumors using the N-of-1 approach in real-world scenario. A total of 160 evaluable events occurred in 48 patients during the 4-year follow-up. Overall survival of the cohort was 7.03 years. Disease control after vaccination was achieved in 53.8% patients. Comparative survival analysis showed the beneficial effect of DC vaccine beyond 2 years from initial diagnosis (HR = 0.53, P = .048) or in patients with disease control (HR = 0.16, P = .00053). A trend for synergistic effect with metronomic cyclophosphamide and/or vinblastine was indicated (HR = 0.60 P = .225). A strong synergistic effect was found for immune check-point inhibitors (ICIs) after priming with the DC vaccine (HR = 0.40, P = .0047). In conclusion, the personalized DC vaccine was an effective component in the multimodal individualized treatment. Personalized DC vaccine was effective in less burdened or more indolent diseases with a favorable safety profile and synergized with metronomic and/or immunomodulating agents.
Collapse
Affiliation(s)
- Michal Kyr
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Centre, St. Anne's University Hospital in Brno, Brno, Czech Republic
| | - Peter Mudry
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Kristyna Polaskova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lenka Zdrazilova Dubska
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Laboratory Medicine, University Hospital Brno, Brno, Czech Republic
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Central European Advanced Therapy and Immunotherapy Centre (CREATIC), Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Regina Demlova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Laboratory Medicine, University Hospital Brno, Brno, Czech Republic
- Central European Advanced Therapy and Immunotherapy Centre (CREATIC), Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jana Kubatova
- Central European Advanced Therapy and Immunotherapy Centre (CREATIC), Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Eva Hlavackova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Central European Advanced Therapy and Immunotherapy Centre (CREATIC), Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Clinical Immunology and Allergology, St. Anne's University Hospital in Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Katerina Cerna Pilatova
- Department of Laboratory Medicine, University Hospital Brno, Brno, Czech Republic
- Central European Advanced Therapy and Immunotherapy Centre (CREATIC), Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Mazanek
- Department of Pediatric Hematology and Biochemistry, Children's University Hospital Brno, Brno, Czech Republic
| | - Klara Vejmelkova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Centre, St. Anne's University Hospital in Brno, Brno, Czech Republic
| | - Vitezslav Dusek
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Tinka
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Centre, St. Anne's University Hospital in Brno, Brno, Czech Republic
| | - Martin Balaz
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tomas Merta
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zuzana Kuttnerova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Terezia Turekova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zdenek Pavelka
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Petra Pokorna
- Department of Biology, Faculty of Medicine and Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Hana Palova
- Department of Biology, Faculty of Medicine and Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Marie Mlnarikova
- Central European Advanced Therapy and Immunotherapy Centre (CREATIC), Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marta Jezova
- Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Renata Kellnerova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sarka Kozakova
- Department of Pharmacy, University Hospital Brno, Brno, Czech Republic
| | - Ondrej Slaby
- Department of Biology, Faculty of Medicine and Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Dalibor Valik
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Laboratory Medicine, University Hospital Brno, Brno, Czech Republic
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Central European Advanced Therapy and Immunotherapy Centre (CREATIC), Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jaroslav Sterba
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
10
|
Colaço M, Cruz MT, de Almeida LP, Borges O. Mannose and Lactobionic Acid in Nasal Vaccination: Enhancing Antigen Delivery via C-Type Lectin Receptors. Pharmaceutics 2024; 16:1308. [PMID: 39458637 PMCID: PMC11510408 DOI: 10.3390/pharmaceutics16101308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/24/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Nasal vaccines are a promising strategy for enhancing mucosal immune responses and preventing diseases at mucosal sites by stimulating the secretion of secretory IgA, which is crucial for early pathogen neutralization. However, designing effective nasal vaccines is challenging due to the complex immunological mechanisms in the nasal mucosa, which must balance protection and tolerance against constant exposure to inhaled pathogens. The nasal route also presents unique formulation and delivery hurdles, such as the mucous layer hindering antigen penetration and immune cell access. METHODS This review focuses on cutting-edge approaches to enhance nasal vaccine delivery, particularly those targeting C-type lectin receptors (CLRs) like the mannose receptor and macrophage galactose-type lectin (MGL) receptor. It elucidates the roles of these receptors in antigen recognition and uptake by antigen-presenting cells (APCs), providing insights into optimizing vaccine delivery. RESULTS While a comprehensive examination of targeted glycoconjugate vaccine development is outside the scope of this study, we provide key examples of glycan-based ligands, such as lactobionic acid and mannose, which can selectively target CLRs in the nasal mucosa. CONCLUSIONS With the rise of new viral infections, this review aims to facilitate the design of innovative vaccines and equip researchers, clinicians, and vaccine developers with the knowledge to enhance immune defenses against respiratory pathogens, ultimately protecting public health.
Collapse
Affiliation(s)
- Mariana Colaço
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria T. Cruz
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Olga Borges
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
11
|
Li Q, Geng S, Luo H, Wang W, Mo YQ, Luo Q, Wang L, Song GB, Sheng JP, Xu B. Signaling pathways involved in colorectal cancer: pathogenesis and targeted therapy. Signal Transduct Target Ther 2024; 9:266. [PMID: 39370455 PMCID: PMC11456611 DOI: 10.1038/s41392-024-01953-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/25/2024] [Accepted: 08/16/2024] [Indexed: 10/08/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Its complexity is influenced by various signal transduction networks that govern cellular proliferation, survival, differentiation, and apoptosis. The pathogenesis of CRC is a testament to the dysregulation of these signaling cascades, which culminates in the malignant transformation of colonic epithelium. This review aims to dissect the foundational signaling mechanisms implicated in CRC, to elucidate the generalized principles underpinning neoplastic evolution and progression. We discuss the molecular hallmarks of CRC, including the genomic, epigenomic and microbial features of CRC to highlight the role of signal transduction in the orchestration of the tumorigenic process. Concurrently, we review the advent of targeted and immune therapies in CRC, assessing their impact on the current clinical landscape. The development of these therapies has been informed by a deepening understanding of oncogenic signaling, leading to the identification of key nodes within these networks that can be exploited pharmacologically. Furthermore, we explore the potential of integrating AI to enhance the precision of therapeutic targeting and patient stratification, emphasizing their role in personalized medicine. In summary, our review captures the dynamic interplay between aberrant signaling in CRC pathogenesis and the concerted efforts to counteract these changes through targeted therapeutic strategies, ultimately aiming to pave the way for improved prognosis and personalized treatment modalities in colorectal cancer.
Collapse
Affiliation(s)
- Qing Li
- The Shapingba Hospital, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Shan Geng
- Central Laboratory, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Wang
- Chongqing Municipal Health and Health Committee, Chongqing, China
| | - Ya-Qi Mo
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Lu Wang
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Guan-Bin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Jian-Peng Sheng
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
12
|
Li X, Li Z, Ma H, Li X, Zhai H, Li X, Cheng X, Zhao X, Zhao Z, Hao Z. Ovarian cancer: Diagnosis and treatment strategies (Review). Oncol Lett 2024; 28:441. [PMID: 39099583 PMCID: PMC11294909 DOI: 10.3892/ol.2024.14574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/25/2024] [Indexed: 08/06/2024] Open
Abstract
Ovarian cancer is a malignant tumor that seriously endangers health. Early ovarian cancer symptoms are frequently challenging to detect, resulting in a large proportion of patients reaching an advanced stage when diagnosed. Conventional diagnosis relies heavily on serum biomarkers and pathological examination, but their sensitivity and specificity require improvement. Targeted therapy inhibits tumor growth by targeting certain characteristics of tumor cells, such as signaling pathways and gene mutations. However, the effectiveness of targeted therapy varies among individuals due to differences in their unique biological characteristics and requires individualized strategies. Immunotherapy is a promising treatment for ovarian cancer due to its long-lasting antitumor effect. Nevertheless, issues such as variable efficacy, immune-associated adverse effects and drug resistance remain to be resolved. The present review discusses the diagnostic strategies, rationale, treatment strategies and prospects of targeted therapy and immunotherapy for ovarian cancer.
Collapse
Affiliation(s)
- Xuejiao Li
- Department of Basic Medicine Science, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Zhuocheng Li
- Department of Basic Medicine Science, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Huiling Ma
- Department of Basic Medicine Science, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Xinwei Li
- Department of Basic Medicine Science, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Hongxiao Zhai
- Department of Basic Medicine Science, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Xixi Li
- Department of Ultrasound, Zhengzhou First People's Hospital, Zhengzhou, Henan 450004, P.R. China
| | - Xiaofei Cheng
- Department of Basic Medicine Science, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Xiaohui Zhao
- Department of Basic Medicine Science, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Zhilong Zhao
- Department of Basic Medicine Science, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Zhenhua Hao
- Department of Basic Medicine Science, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
13
|
Chen J, Duan Y, Che J, Zhu J. Dysfunction of dendritic cells in tumor microenvironment and immunotherapy. Cancer Commun (Lond) 2024; 44:1047-1070. [PMID: 39051512 PMCID: PMC11492303 DOI: 10.1002/cac2.12596] [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: 01/31/2024] [Revised: 06/10/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024] Open
Abstract
Dendritic cells (DCs) comprise diverse cell populations that play critical roles in antigen presentation and triggering immune responses in the body. However, several factors impair the immune function of DCs and may promote immune evasion in cancer. Understanding the mechanism of DC dysfunction and the diverse functions of heterogeneous DCs in the tumor microenvironment (TME) is critical for designing effective strategies for cancer immunotherapy. Clinical applications targeting DCs summarized in this report aim to improve immune infiltration and enhance the biological function of DCs to modulate the TME to prevent cancer cells from evading the immune system. Herein, factors in the TME that induce DC dysfunction, such as cytokines, hypoxic environment, tumor exosomes and metabolites, and co-inhibitory molecules, have been described. Furthermore, several key signaling pathways involved in DC dysfunction and signal-relevant drugs evaluated in clinical trials were identified. Finally, this review provides an overview of current clinical immunotherapies targeting DCs, especially therapies with proven clinical outcomes, and explores future developments in DC immunotherapies.
Collapse
Affiliation(s)
- Jie Chen
- Jecho Institute Co., LtdShanghaiP. R. China
| | - Yuhang Duan
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of EducationBeijingP. R. China
- Shanghai Jiao Tong University, School of PharmacyShanghaiP. R. China
| | - Junye Che
- Jecho Institute Co., LtdShanghaiP. R. China
| | - Jianwei Zhu
- Jecho Institute Co., LtdShanghaiP. R. China
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of EducationBeijingP. R. China
- Shanghai Jiao Tong University, School of PharmacyShanghaiP. R. China
| |
Collapse
|
14
|
Lin C, Teng W, Tian Y, Li S, Xia N, Huang C. Immune landscape and response to oncolytic virus-based immunotherapy. Front Med 2024; 18:411-429. [PMID: 38453818 DOI: 10.1007/s11684-023-1048-0] [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: 07/19/2023] [Accepted: 11/15/2023] [Indexed: 03/09/2024]
Abstract
Oncolytic virus (OV)-based immunotherapy has emerged as a promising strategy for cancer treatment, offering a unique potential to selectively target malignant cells while sparing normal tissues. However, the immunosuppressive nature of tumor microenvironment (TME) poses a substantial hurdle to the development of OVs as effective immunotherapeutic agents, as it restricts the activation and recruitment of immune cells. This review elucidates the potential of OV-based immunotherapy in modulating the immune landscape within the TME to overcome immune resistance and enhance antitumor immune responses. We examine the role of OVs in targeting specific immune cell populations, including dendritic cells, T cells, natural killer cells, and macrophages, and their ability to alter the TME by inhibiting angiogenesis and reducing tumor fibrosis. Additionally, we explore strategies to optimize OV-based drug delivery and improve the efficiency of OV-mediated immunotherapy. In conclusion, this review offers a concise and comprehensive synopsis of the current status and future prospects of OV-based immunotherapy, underscoring its remarkable potential as an effective immunotherapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Chaolong Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Wenzhong Teng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Yang Tian
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Shaopeng Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China.
| | - Chenghao Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
15
|
Mantooth SM, Abdou Y, Saez-Ibañez AR, Upadhaya S, Zaharoff DA. Intratumoral delivery of immunotherapy to treat breast cancer: current development in clinical and preclinical studies. Front Immunol 2024; 15:1385484. [PMID: 38803496 PMCID: PMC11128577 DOI: 10.3389/fimmu.2024.1385484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Breast cancer poses one of the largest threats to women's health. Treatment continues to improve for all the subtypes of breast cancer, but some subtypes, such as triple negative breast cancer, still present a significant treatment challenge. Additionally, metastasis and local recurrence are two prevalent problems in breast cancer treatment. A newer type of therapy, immunotherapy, may offer alternatives to traditional treatments for difficult-to-treat subtypes. Immunotherapy engages the host's immune system to eradicate disease, with the potential to induce long-lasting, durable responses. However, systemic immunotherapy is only approved in a limited number of indications, and it benefits only a minority of patients. Furthermore, immune related toxicities following systemic administration of potent immunomodulators limit dosing and, consequently, efficacy. To address these safety considerations and improve treatment efficacy, interest in local delivery at the site of the tumor has increased. Numerous intratumorally delivered immunotherapeutics have been and are being explored clinically and preclinically, including monoclonal antibodies, cellular therapies, viruses, nucleic acids, cytokines, innate immune agonists, and bacteria. This review summarizes the current and past intratumoral immunotherapy clinical landscape in breast cancer as well as current progress that has been made in preclinical studies, with a focus on delivery parameters and considerations.
Collapse
Affiliation(s)
- Siena M. Mantooth
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
| | - Yara Abdou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | | | - David A. Zaharoff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
16
|
Heumann P, Albert A, Gülow K, Tümen D, Müller M, Kandulski A. Insights in Molecular Therapies for Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:1831. [PMID: 38791911 PMCID: PMC11120383 DOI: 10.3390/cancers16101831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
We conducted a comprehensive review of the current literature of published data and clinical trials (MEDLINE), as well as published congress contributions and active recruiting clinical trials on targeted therapies in hepatocellular carcinoma. Combinations of different agents and medical therapy along with radiological interventions were analyzed for the setting of advanced HCC. Those settings were also analyzed in combination with adjuvant situations after resection or radiological treatments. We summarized the current knowledge for each therapeutic setting and combination that currently is or has been under clinical evaluation. We further discuss the results in the background of current treatment guidelines. In addition, we review the pathophysiological mechanisms and pathways for each of these investigated targets and drugs to further elucidate the molecular background and underlying mechanisms of action. Established and recommended targeted treatment options that already exist for patients are considered for systemic treatment: atezolizumab/bevacizumab, durvalumab/tremelimumab, sorafenib, lenvatinib, cabozantinib, regorafenib, and ramucirumab. Combination treatment for systemic treatment and local ablative treatment or transarterial chemoembolization and adjuvant and neoadjuvant treatment strategies are under clinical investigation.
Collapse
Affiliation(s)
- Philipp Heumann
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany (K.G.); (D.T.)
| | | | | | | | | | - Arne Kandulski
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany (K.G.); (D.T.)
| |
Collapse
|
17
|
Yang W, Wu Z, Cai S, Li Z, Wang W, Wu J, Luo H, Ye X. Tumor lymphangiogenesis index reveals the immune landscape and immunotherapy response in lung adenocarcinoma. Front Immunol 2024; 15:1354339. [PMID: 38638428 PMCID: PMC11024352 DOI: 10.3389/fimmu.2024.1354339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/25/2024] [Indexed: 04/20/2024] Open
Abstract
Background Lymphangiogenesis (LYM) has an important role in tumor progression and is strongly associated with tumor metastasis. However, the clinical application of LYM has not progressed as expected. The potential value of LYM needs to be further developed in lung adenocarcinoma (LUAD) patients. Methods The Sequencing data and clinical characteristics of LUAD patients were downloaded from The Cancer Genome Atlas and GEO databases. Multiple machine learning algorithms were used to screen feature genes and develop the LYM index. Immune cell infiltration, immune checkpoint expression, Tumor Immune Dysfunction and Exclusion (TIDE) algorithm and drug sensitivity analysis were used to explore the correlation of LYM index with immune profile and anti-tumor therapy. Results We screened four lymphangiogenic feature genes (PECAM1, TIMP1, CXCL5 and PDGFB) to construct LYM index based on multiple machine learning algorithms. We divided LUAD patients into the high LYM index group and the low LYM index group based on the median LYM index. LYM index is a risk factor for the prognosis of LUAD patients. In addition, there was a significant difference in immune profile between high LYM index and low LYM index groups. LUAD patients in the low LYM index group seemed to benefit more from immunotherapy based on the results of TIDE algorithm. Conclusion Overall, we confirmed that the LYM index is a prognostic risk factor and a valuable predictor of immunotherapy response in LUAD patients, which provides new evidence for the potential application of LYM.
Collapse
Affiliation(s)
- Weichang Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, Jiangxi, China
| | - Zhijian Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shanshan Cai
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhouhua Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Wenjun Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Juan Wu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongdan Luo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoqun Ye
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
18
|
Shebbo S, Binothman N, Darwaish M, Niaz HA, Abdulal RH, Borjac J, Hashem AM, Mahmoud AB. Redefining the battle against colorectal cancer: a comprehensive review of emerging immunotherapies and their clinical efficacy. Front Immunol 2024; 15:1350208. [PMID: 38533510 PMCID: PMC10963412 DOI: 10.3389/fimmu.2024.1350208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer globally and presents a significant challenge owing to its high mortality rate and the limitations of traditional treatment options such as surgery, radiotherapy, and chemotherapy. While these treatments are foundational, they are often poorly effective owing to tumor resistance. Immunotherapy is a groundbreaking alternative that has recently emerged and offers new hope for success by exploiting the body's own immune system. This article aims to provide an extensive review of clinical trials evaluating the efficacy of various immunotherapies, including CRC vaccines, chimeric antigen receptor T-cell therapies, and immune checkpoint inhibitors. We also discuss combining CRC vaccines with monoclonal antibodies, delve into preclinical studies of novel cancer vaccines, and assess the impact of these treatment methods on patient outcomes. This review seeks to provide a deeper understanding of the current state of CRC treatment by evaluating innovative treatments and their potential to redefine the prognosis of patients with CRC.
Collapse
Affiliation(s)
- Salima Shebbo
- Strategic Research and Innovation Laboratories, Taibah University, Madinah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biological Sciences, Beirut Arab University, Debbieh, Lebanon
| | - Najat Binothman
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Chemistry, College of Sciences and Arts, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Manar Darwaish
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Research Program, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Hanan A. Niaz
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Rwaa H. Abdulal
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jamilah Borjac
- Department of Biological Sciences, Beirut Arab University, Debbieh, Lebanon
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- Strategic Research and Innovation Laboratories, Taibah University, Madinah, Saudi Arabia
- College of Applied Medical Sciences, Taibah University, Almadinah Almunawarah, Saudi Arabia
| |
Collapse
|
19
|
da Silva SF, Murta EF, Michelin MA. ICAM2 is related to good prognosis in dendritic cell immunotherapy for cancer. Immunotherapy 2024; 16:173-185. [PMID: 38126167 DOI: 10.2217/imt-2021-0097] [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: 04/22/2021] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Objective: To evaluate the behavior of adhesion molecules ICAM-1 and ICAM-2 in dendritic cell (DC) immunotherapy. Materials & methods: 88 female Balb/c mice were divided into experimental groups. Tumors and lymph nodes were evaluated 7 and 14 days after immunotherapy. Results: Higher mean fluorescence intensity of ICAM-1 in the lymph nodes and tumors in the tumor group at 14 days was observed. Higher mean fluorescence intensity of ICAM-2 in the tumor DC vaccine group was observed after 14 days. A positive correlation was observed in the lymph nodes with ICAM-1 against tumoral volume in the tumor group. A negative correlation was found between ICAM-2 and tumoral volume in the lymph nodes of the tumor group. Conclusion: An increase in ICAM-2 in tumor DC vaccine and a decrease in ICAM-1 suggests the DC vaccine positively influences the immune system and that ICAM-2 could be a marker of good prognosis.
Collapse
Affiliation(s)
- Saulo Fm da Silva
- Oncology Research Institute (IPON), Federal University of Triângulo Mineiro (UFTM), Uberaba, MG, Brazil
| | - Eddie Fc Murta
- Oncology Research Institute (IPON), Federal University of Triângulo Mineiro (UFTM), Uberaba, MG, Brazil
- Department of Gynecology and Obstetrics, Federal University of Triangulo Mineiro (UFTM), Uberaba, MG, 38.025-350, Brazil
| | - Márcia A Michelin
- Oncology Research Institute (IPON), Federal University of Triângulo Mineiro (UFTM), Uberaba, MG, Brazil
- Immunology Discipline, Federal University of Triângulo Mineiro (UFTM), Uberaba, MG, 38.025-500, Brazil
| |
Collapse
|
20
|
Foley CR, Swan SL, Swartz MA. Engineering Challenges and Opportunities in Autologous Cellular Cancer Immunotherapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:188-198. [PMID: 38166251 PMCID: PMC11155266 DOI: 10.4049/jimmunol.2300642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 01/04/2024]
Abstract
The use of a patient's own immune or tumor cells, manipulated ex vivo, enables Ag- or patient-specific immunotherapy. Despite some clinical successes, there remain significant barriers to efficacy, broad patient population applicability, and safety. Immunotherapies that target specific tumor Ags, such as chimeric Ag receptor T cells and some dendritic cell vaccines, can mount robust immune responses against immunodominant Ags, but evolving tumor heterogeneity and antigenic downregulation can drive resistance. In contrast, whole tumor cell vaccines and tumor lysate-loaded dendritic cell vaccines target the patient's unique tumor antigenic repertoire without prior neoantigen selection; however, efficacy can be weak when lower-affinity clones dominate the T cell pool. Chimeric Ag receptor T cell and tumor-infiltrating lymphocyte therapies additionally face challenges related to genetic modification, T cell exhaustion, and immunotoxicity. In this review, we highlight some engineering approaches and opportunities to these challenges among four classes of autologous cell therapies.
Collapse
Affiliation(s)
- Colleen R. Foley
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Sheridan L. Swan
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Melody A. Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
- Committee on Immunology, University of Chicago, Chicago, Illinois
- Ben May Department of Cancer Research, University of Chicago, Chicago, Illinois
| |
Collapse
|
21
|
Ying Li CM, Li R, Drew P, Price T, Smith E, Maddern GJ, Tomita Y, Fenix K. Clinical application of cytokine-induced killer (CIK) cell therapy in colorectal cancer: Current strategies and future challenges. Cancer Treat Rev 2024; 122:102665. [PMID: 38091655 DOI: 10.1016/j.ctrv.2023.102665] [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: 10/27/2023] [Revised: 11/23/2023] [Accepted: 11/26/2023] [Indexed: 01/01/2024]
Abstract
Colorectal cancer (CRC) remains a significant global health burden and is the second leading cause of cancer-related death. Cytokine induced killer (CIK) cell therapy is an immunotherapy which has the potential to meet this need. Clinical trials of CIK cell therapy for the management of CRC have reported improved clinical outcomes. However, production and delivery protocols varied significantly, and many studies were reported only in Chinese language journals. Here we present the most comprehensive review of the clinical CIK cell therapy trials for CRC management to date. We accessed both English and Chinese language clinical studies, and summarise how CIK cell therapy has been implemented, from manufacturing to patient delivery. We discuss current challenges that impede wider adoption of CIK cell therapy in CRC management.
Collapse
Affiliation(s)
- Celine Man Ying Li
- Department of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia
| | - Runhao Li
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia; Medical Oncology, The Queen Elizabeth Hospital and The University of Adelaide, Woodville, SA 5011, Australia
| | - Paul Drew
- Department of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia
| | - Timothy Price
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia; Medical Oncology, The Queen Elizabeth Hospital and The University of Adelaide, Woodville, SA 5011, Australia
| | - Eric Smith
- Department of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia; Medical Oncology, The Queen Elizabeth Hospital and The University of Adelaide, Woodville, SA 5011, Australia
| | - Guy J Maddern
- Department of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia
| | - Yoko Tomita
- Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia; Medical Oncology, The Queen Elizabeth Hospital and The University of Adelaide, Woodville, SA 5011, Australia
| | - Kevin Fenix
- Department of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia; Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Woodville, SA 5011, Australia.
| |
Collapse
|
22
|
Alotaibi N, Aldahlawi A, Zaher K, Basingab F, Alrahimi J. Optimizing the generation of mature bone marrow-derived dendritic cells in vitro: a factorial study design. J Genet Eng Biotechnol 2023; 21:144. [PMID: 38017248 PMCID: PMC10684437 DOI: 10.1186/s43141-023-00597-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/09/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Factorial design is a simple, yet elegant method to investigate the effect of multiple factors and their interaction on a specific response simultaneously. Hence, this type of study design reaches the best optimization conditions of a process. Although the interaction between the variables is widely prevalent in cell culture procedures, factorial design per se is infrequently utilized in improving cell culture output. Therefore, we aim to optimize the experimental conditions for generating mature bone marrow-derived dendritic cells (BMDCs). Two different variables were investigated, including the concentrations of the inducing factors and the starting density of the bone marrow mononuclear cells. In the current study, we utilized the design of experiments (DoE), a statistical approach, to systematically assess the impact of factors with varying levels on cell culture outcomes. Herein, we apply a two-factor, two-level (22) factorial experiment resulting in four conditions that are run in triplicate. The two variables investigated here are cytokines combinations with two levels, granulocyte-macrophage colony-stimulating factor (GM-CSF) alone or with interleukin-4 (IL4). The other parameter is cell density with two different concentrations, 2 × 106 and 4 × 106 cells/mL. Then, we measured cell viability using the trypan blue exclusion method, and a flow cytometer was used to detect the BMDCs expressing the markers FITC-CD80, CD86, CD83, and CD14. BMDC marker expression levels were calculated using arbitrary units (AU) of the mean fluorescence intensity (MFI). RESULTS The current study showed that the highest total viable cells and cells yield obtained were in cell group seeded at 2 × 106 cells/mL and treated with GM-CSF and IL-4. Importantly, the expression of the co-stimulatory molecules CD83 and CD80/CD86 were statistically significant for cell density of 2 × 106 cells/mL (P < 0.01, two-way ANOVA). Bone marrow mononuclear cells seeded at 4 × 106 in the presence of the cytokine mix less efficiently differentiated and matured into BMDCs. Statistical analysis via two-way ANOVA revealed an interaction between cell density and cytokine combinations. CONCLUSION The analysis of this study indicates a substantial interaction between cytokines combinations and cell densities on BMDC maturation. However, higher cell density is not associated with optimizing DC maturation. Notably, applying DoE in bioprocess designs increases experimental efficacy and reliability while minimizing experiments, time, and process costs.
Collapse
Affiliation(s)
- Najla Alotaibi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
- College of Health, Oregon State University, Corvallis, OR, USA.
| | - Alia Aldahlawi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Kawther Zaher
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatemah Basingab
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jehan Alrahimi
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
23
|
Elwakeel A, Bridgewater HE, Bennett J. Unlocking Dendritic Cell-Based Vaccine Efficacy through Genetic Modulation-How Soon Is Now? Genes (Basel) 2023; 14:2118. [PMID: 38136940 PMCID: PMC10743214 DOI: 10.3390/genes14122118] [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: 10/13/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
The dendritic cell (DC) vaccine anti-cancer strategy involves tumour-associated antigen loading and maturation of autologous ex vivo cultured DCs, followed by infusion into the cancer patient. This strategy stemmed from the idea that to induce a robust anti-tumour immune response, it was necessary to bypass the fundamental immunosuppressive mechanisms of the tumour microenvironment that dampen down endogenous innate immune cell activation and enable tumours to evade immune attack. Even though the feasibility and safety of DC vaccines have long been confirmed, clinical response rates remain disappointing. Hence, the full potential of DC vaccines has yet to be reached. Whether this cellular-based vaccination approach will fully realise its position in the immunotherapy arsenal is yet to be determined. Attempts to increase DC vaccine immunogenicity will depend on increasing our understanding of DC biology and the signalling pathways involved in antigen uptake, maturation, migration, and T lymphocyte priming to identify amenable molecular targets to improve DC vaccine performance. This review evaluates various genetic engineering strategies that have been employed to optimise and boost the efficacy of DC vaccines.
Collapse
Affiliation(s)
- Ahmed Elwakeel
- Centre for Health and Life Sciences (CHLS), Coventry University, Coventry CV1 5FB, UK; (A.E.); (H.E.B.)
| | - Hannah E. Bridgewater
- Centre for Health and Life Sciences (CHLS), Coventry University, Coventry CV1 5FB, UK; (A.E.); (H.E.B.)
| | - Jason Bennett
- Department of Biological Sciences, Faculty of Science and Engineering, University of Limerick, V94 T9PX Limerick, Ireland
| |
Collapse
|
24
|
Geng K, Rice-Boucher PJ, Kashentseva EA, Dmitriev IP, Lu ZH, Goedegebuure SP, Gillanders WE, Curiel DT. Engineering a Novel Modular Adenoviral mRNA Delivery Platform Based on Tag/Catcher Bioconjugation. Viruses 2023; 15:2277. [PMID: 38005953 PMCID: PMC10674448 DOI: 10.3390/v15112277] [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: 10/24/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
mRNA vaccines have attracted widespread research attention with clear advantages in terms of molecular flexibility, rapid development, and potential for personalization. However, current mRNA vaccine platforms have not been optimized for induction of CD4/CD8 T cell responses. In addition, the mucosal administration of mRNA based on lipid nanoparticle technology faces challenges in clinical translation. In contrast, adenovirus-based vaccines induce strong T cell responses and have been approved for intranasal delivery. To leverage the inherent strengths of both the mRNA and adenovirus platforms, we developed a novel modular adenoviral mRNA delivery platform based on Tag/Catcher bioconjugation. Specifically, we engineered adenoviral vectors integrating Tag/Catcher proteins at specific locales on the Ad capsid proteins, allowing us to anchor mRNA to the surface of engineered Ad viruses. In proof-of-concept studies, the Ad-mRNA platform successfully mediated mRNA delivery and could be optimized via the highly flexible modular design of both the Ad-mRNA and protein bioconjugation systems.
Collapse
Affiliation(s)
- Kexin Geng
- Department of Radiation Oncology, Biologic Therapeutics Center, Washington University School of Medicine, St. Louis, MO 63110, USA; (K.G.); (P.J.R.-B.); (E.A.K.); (I.P.D.); (Z.H.L.)
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in Saint Louis, St. Louis, MO 63130-4899, USA
| | - Paul J. Rice-Boucher
- Department of Radiation Oncology, Biologic Therapeutics Center, Washington University School of Medicine, St. Louis, MO 63110, USA; (K.G.); (P.J.R.-B.); (E.A.K.); (I.P.D.); (Z.H.L.)
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in Saint Louis, St. Louis, MO 63130-4899, USA
| | - Elena A. Kashentseva
- Department of Radiation Oncology, Biologic Therapeutics Center, Washington University School of Medicine, St. Louis, MO 63110, USA; (K.G.); (P.J.R.-B.); (E.A.K.); (I.P.D.); (Z.H.L.)
| | - Igor P. Dmitriev
- Department of Radiation Oncology, Biologic Therapeutics Center, Washington University School of Medicine, St. Louis, MO 63110, USA; (K.G.); (P.J.R.-B.); (E.A.K.); (I.P.D.); (Z.H.L.)
| | - Zhi Hong Lu
- Department of Radiation Oncology, Biologic Therapeutics Center, Washington University School of Medicine, St. Louis, MO 63110, USA; (K.G.); (P.J.R.-B.); (E.A.K.); (I.P.D.); (Z.H.L.)
| | - S. Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.P.G.); (W.E.G.)
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110, USA
| | - William E. Gillanders
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.P.G.); (W.E.G.)
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David T. Curiel
- Department of Radiation Oncology, Biologic Therapeutics Center, Washington University School of Medicine, St. Louis, MO 63110, USA; (K.G.); (P.J.R.-B.); (E.A.K.); (I.P.D.); (Z.H.L.)
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in Saint Louis, St. Louis, MO 63130-4899, USA
| |
Collapse
|
25
|
Esperante D, Gutiérrez MIM, Issa ME, Schcolnik-Cabrera A, Mendlovic F. Similarities and divergences in the metabolism of immune cells in cancer and helminthic infections. Front Oncol 2023; 13:1251355. [PMID: 38044996 PMCID: PMC10690632 DOI: 10.3389/fonc.2023.1251355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/16/2023] [Indexed: 12/05/2023] Open
Abstract
Energetic and nutritional requirements play a crucial role in shaping the immune cells that infiltrate tumor and parasite infection sites. The dynamic interaction between immune cells and the microenvironment, whether in the context of tumor or helminth infection, is essential for understanding the mechanisms of immunological polarization and developing strategies to manipulate them in order to promote a functional and efficient immune response that could aid in the treatment of these conditions. In this review, we present an overview of the immune response triggered during tumorigenesis and establishment of helminth infections, highlighting the transition to chronicity in both cases. We discuss the energetic demands of immune cells under normal conditions and in the presence of tumors and helminths. Additionally, we compare the metabolic changes that occur in the tumor microenvironment and the infection site, emphasizing the alterations that are induced to redirect the immune response, thereby promoting the survival of cancer cells or helminths. This emerging discipline provides valuable insights into disease pathogenesis. We also provide examples of novel strategies to enhance immune activity by targeting metabolic pathways that shape immune phenotypes, with the aim of achieving positive outcomes in cancer and helminth infections.
Collapse
Affiliation(s)
- Diego Esperante
- Plan de Estudios Combinados en Medicina (PECEM), Facultad de Medicina, Universidad Nacional Autonóma de México (UNAM), Mexico City, Mexico
| | - Mónica Itzel Martínez Gutiérrez
- Plan de Estudios Combinados en Medicina (PECEM), Facultad de Medicina, Universidad Nacional Autonóma de México (UNAM), Mexico City, Mexico
| | - Mark E. Issa
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Alejandro Schcolnik-Cabrera
- Département de Biochimie et Médicine Moléculaire, Université de Montréal, Succursale Centre-Ville, Montréal, QC, Canada
- Department of Immunology-Oncology, Maisonneuve-Rosemont Hospital Research Centre, Montréal, QC, Canada
| | - Fela Mendlovic
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Facultad de Ciencias de la Salud, Universidad Anáhuac México Norte, Huixquilucan, Mexico
| |
Collapse
|
26
|
Subtirelu RC, Teichner EM, Ashok A, Parikh C, Talasila S, Matache IM, Alnemri AG, Anderson V, Shahid O, Mannam S, Lee A, Werner T, Revheim ME, Alavi A. Advancements in dendritic cell vaccination: enhancing efficacy and optimizing combinatorial strategies for the treatment of glioblastoma. Front Neurol 2023; 14:1271822. [PMID: 38020665 PMCID: PMC10644823 DOI: 10.3389/fneur.2023.1271822] [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: 08/02/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Glioblastomas (GBM) are highly invasive, malignant primary brain tumors. The overall prognosis is poor, and management of GBMs remains a formidable challenge, necessitating novel therapeutic strategies such as dendritic cell vaccinations (DCVs). While many early clinical trials demonstrate an induction of an antitumoral immune response, outcomes are mixed and dependent on numerous factors that vary between trials. Optimization of DCVs is essential; the selection of GBM-specific antigens and the utilization of 18F-fludeoxyglucose Positron Emission Tomography (FDG-PET) may add significant value and ultimately improve outcomes for patients undergoing treatment for glioblastoma. This review provides an overview of the mechanism of DCV, assesses previous clinical trials, and discusses future strategies for the integration of DCV into glioblastoma treatment protocols. To conclude, the review discusses challenges associated with the use of DCVs and highlights the potential of integrating DCV with standard therapies.
Collapse
Affiliation(s)
- Robert C. Subtirelu
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Eric M. Teichner
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Arjun Ashok
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Chitra Parikh
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sahithi Talasila
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Irina-Mihaela Matache
- Department of Physiology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ahab G. Alnemri
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Victoria Anderson
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Osmaan Shahid
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Sricharvi Mannam
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Andrew Lee
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Thomas Werner
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Mona-Elisabeth Revheim
- Division of Technology and Innovation, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
27
|
Rumler S. Non-cellular immunotherapies in pediatric central nervous system tumors. Front Immunol 2023; 14:1242911. [PMID: 37885882 PMCID: PMC10598668 DOI: 10.3389/fimmu.2023.1242911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Central nervous system (CNS) tumors are the second most common type of cancer and the most common cause of cancer death in pediatric patients. New therapies are desperately needed for some of the most malignant of all cancers. Immunotherapy has emerged in the past two decades as an additional avenue to augment/replace traditional therapies (such as chemotherapy, surgery, and radiation therapy). This article first discusses the unique nature of the pediatric CNS immune system and how it interacts with the systemic immune system. It then goes on to review three important and widely studied types of immune therapies: checkpoint inhibitors, vaccines, and radiation therapy, and touches on early studies of antibody-mediated immunogenic therapies, Finally, the article discusses the importance of combination immunotherapy for pediatric CNS tumors, and addresses the neurologic toxicities associated with immunotherapies.
Collapse
Affiliation(s)
- Sarah Rumler
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
28
|
Mohamed AA, Caussat T, Kelly S, Johansen PM, Lucke-Wold B. Choroid plexus tumors: A spectrum from benign to malignant. TUMOR DISCOVERY 2023; 2:1057. [PMID: 37799733 PMCID: PMC10552314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Choroid plexus tumors (CPT) are believed to originate from outgrowths of the choroid plexus. Despite their broad spectrum of symptoms, invasive nature, and prognosis, most CPTs typically exhibit similar presentations due to their relationship with the cerebral ventricles, as well as the mechanical obstruction and mass effect associated with their growth. In addition, these tumors mainly affect the pediatric population, further complicating the differentiation between benign and malignant subtypes. The World Health Organization classifies CPTs into three grades, namely, grades I, II, or III, based on their mitotic activity, which determine the benign or malignant nature of the tumors. CPTs classified by the World Health Organization (WHO) include choroid plexus papillomas (CPP), atypical CPPs (aCPP), and malignant choroid plexus carcinomas (CPC). Choroid plexus adenomas represent an additional category of benign CPTs not officially classified by the WHO. Despite the variations in histology, immunohistochemistry, imaging, treatment, and prognosis, CPTs cannot be reliably distinguished based solely on clinical presentation. Therefore, in this review, we aim to provide a comprehensive overview of each tumor subtype, along with the current management approach and emerging treatments.
Collapse
Affiliation(s)
- Ali A. Mohamed
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Thomas Caussat
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Sophie Kelly
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Phillip M. Johansen
- Department of Neurosurgery, University of South Florida, Orlando, Florida, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
29
|
Shirman Y, Lubovsky S, Shai A. HER2-Low Breast Cancer: Current Landscape and Future Prospects. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:605-616. [PMID: 37600670 PMCID: PMC10439285 DOI: 10.2147/bctt.s366122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 08/09/2023] [Indexed: 08/22/2023]
Abstract
More than 50% of breast cancers are currently defined as "Human epidermal growth factor receptor 2 (HER2) low breast cancer (BC)", with HER2 immunohistochemistry (IHC) scores of +1 or +2 with a negative fluorescence in situ hybridization (FISH) test. In most studies that compared the clinical and biological characteristics of HER2-low BC with HER2-negative BC, HER2-low was not associated with unique clinical and molecular characteristics, and it seems that the importance of HER2 in these tumors is being a docking site for the antibody portion of antibody drug conjugates (ADCs). Current pathological methods may underestimate the proportion of BCs that express low levels of HER2 due to analytical limitations and tumor heterogeneity. In this review we summarize and contextualize the most recent literature on HER2-low breast cancers, including clinical and translational studies We also review the challenges of assessing low HER2 expression in BC and discuss the current and future therapeutic landscape for these tumors.
Collapse
Affiliation(s)
- Yelena Shirman
- Division of Oncology, Rambam Health Care Campus, Haifa, Israel
| | | | - Ayelet Shai
- Division of Oncology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
30
|
Noon A, Galban S. Therapeutic avenues for targeting treatment challenges of diffuse midline gliomas. Neoplasia 2023; 40:100899. [PMID: 37030112 PMCID: PMC10119952 DOI: 10.1016/j.neo.2023.100899] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Diffuse midline glioma (DMG) is the leading cause of brain tumor-related deaths in children. DMG typically presents with variable neurologic symptoms between ages 3 and 10. Currently, radiation remains the standard therapy for DMG to halt progression and reduce tumor bulk to minimize symptoms. However, tumors recur in almost 100% of patients and thus, DMG is still considered an incurable cancer with a median survival of 9-12 months. Surgery is generally contraindicated due to the delicate organization of the brainstem, where DMG is located. Despite extensive research efforts, no chemotherapeutic agents, immune therapies, or molecularly targeted therapies have been approved to provide survival benefit. Furthermore, the efficacy of therapies is limited by poor blood-brain barrier penetration and inherent resistance mechanisms of the tumor. However, novel drug delivery approaches, along with recent advances in molecularly targeted therapies and immunotherapies, have advanced to clinical trials and may provide viable future treatment options for DMG patients. This review seeks to evaluate current therapeutics at the preclinical stage and those that have advanced to clinical trials and to discuss the challenges of drug delivery and inherent resistance to these therapies.
Collapse
Affiliation(s)
- Aleeha Noon
- College of Medicine, California Northstate University, 9700 W Taron Drive, Elk Grove, CA 95757, USA
| | - Stefanie Galban
- Center for Molecular Imaging, The University of Michigan Medical School, BSRB A502, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Department of Radiology, The University of Michigan Medical School, BSRB A502, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Rogel Cancer Center, The University of Michigan Medical School, 1500 E Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
31
|
Fang X, Lan H, Jin K, Qian J. Pancreatic cancer and exosomes: role in progression, diagnosis, monitoring, and treatment. Front Oncol 2023; 13:1149551. [PMID: 37287924 PMCID: PMC10242099 DOI: 10.3389/fonc.2023.1149551] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 05/05/2023] [Indexed: 06/09/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most dangerous diseases that threaten human life, and investigating the details affecting its progression or regression is particularly important. Exosomes are one of the derivatives produced from different cells, including tumor cells and other cells such as Tregs, M2 macrophages, and MDSCs, and can help tumor growth. These exosomes perform their actions by affecting the cells in the tumor microenvironment, such as pancreatic stellate cells (PSCs) that produce extracellular matrix (ECM) components and immune cells that are responsible for killing tumor cells. It has also been shown that pancreatic cancer cell (PCC)-derived exosomes at different stages carry molecules. Checking the presence of these molecules in the blood and other body fluids can help us in the early stage diagnosis and monitoring of PC. However, immune system cell-derived exosomes (IEXs) and mesenchymal stem cell (MSC)-derived exosomes can contribute to PC treatment. Immune cells produce exosomes as part of the mechanisms involved in the immune surveillance and tumor cell-killing phenomenon. Exosomes can be modified in such a way that their antitumor properties are enhanced. One of these methods is drug loading in exosomes, which can significantly increase the effectiveness of chemotherapy drugs. In general, exosomes form a complex intercellular communication network that plays a role in developing, progressing, diagnosing, monitoring, and treating pancreatic cancer.
Collapse
Affiliation(s)
- Xingliang Fang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People’s Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang, Zhejiang, China
| |
Collapse
|
32
|
Watowich MB, Gilbert MR, Larion M. T cell exhaustion in malignant gliomas. Trends Cancer 2023; 9:270-292. [PMID: 36681605 PMCID: PMC10038906 DOI: 10.1016/j.trecan.2022.12.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/21/2023]
Abstract
Despite advances in understanding tumor biology, malignant gliomas remain incurable. While immunotherapy has improved outcomes in other cancer types, comparable efficacy has not yet been demonstrated for primary cancers of the central nervous system (CNS). T cell exhaustion, defined as a progressive decrease in effector function, sustained expression of inhibitory receptors, metabolic dysfunction, and distinct epigenetic and transcriptional alterations, contributes to the failure of immunotherapy in the CNS. Herein, we describe recent advances in understanding the drivers of T cell exhaustion in the glioma microenvironment. We discuss the extrinsic and intrinsic factors that contribute to exhaustion and highlight potential avenues for reversing this phenotype. Our ability to directly target specific immunosuppressive drivers in brain cancers would be a major advance in immunotherapy.
Collapse
Affiliation(s)
- Matthew B Watowich
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mioara Larion
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
33
|
Minaei N, Ramezankhani R, Tamimi A, Piryaei A, Zarrabi A, Aref AR, Mostafavi E, Vosough M. Immunotherapeutic approaches in Hepatocellular carcinoma: Building blocks of hope in near future. Eur J Cell Biol 2023; 102:151284. [PMID: 36584598 DOI: 10.1016/j.ejcb.2022.151284] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/30/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary hepatic cancer and is among the major causes of mortality due to cancer. Due to the lack of efficient conventional therapeutic options for this cancer, particularly in advanced cases, novel treatments including immunotherapy have been considered. However, despite the encouraging clinical outcomes after implementing these innovative approaches, such as oncolytic viruses (OVs), adoptive cell therapies (ACT), immune checkpoint blockades (ICBs), and cancer vaccines, several factors have restricted their therapeutic effect. The main concern is the existence of an immunosuppressive tumor microenvironment (TME). Combination of different ICBs or ICBs plus tyrosine kinase inhibitors have shown promising results in overcoming these limiting factors to some extent. Combination of programmed cell death ligand-1 (PD-L1) antibody Atezolizumab and vascular endothelial growth factor (VEGF) antibody Bevacizumab has become the standard of care in the first-line therapy for untestable HCC, approved by regulatory agencies. This paper highlighted a wide overview of the direct and indirect immunotherapeutic strategies proposed for the treatment of HCC patients and the common challenges that have hindered their further clinical applications.
Collapse
Affiliation(s)
- Neda Minaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Roya Ramezankhani
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran; Department of Development and Regeneration, KU Leuven Stem Cell Institute, Leuven, Belgium
| | - Atena Tamimi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital-Huddinge, Sweden.
| |
Collapse
|
34
|
Lee SW, Lee H, Lee KW, Kim MJ, Kang SW, Lee YJ, Kim H, Kim YM. CD8α+ dendritic cells potentiate antitumor and immune activities against murine ovarian cancers. Sci Rep 2023; 13:98. [PMID: 36596856 PMCID: PMC9810613 DOI: 10.1038/s41598-022-27303-7] [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: 07/15/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Dendritic cell (DC)-based immunotherapies have been shown to be a potential treatment option for various cancers; however, the exact strategies in ovarian cancer remain unknown. Here, we report the effectiveness of mouse CD8α+ DCs derived from bone marrow hematopoietic stem cells (BM-HSCs), equivalent to human CD141+ DCs, which have proven to be a highly superior subset. Mono-DCs from monocytes and stem-DCs from HSCs were characterized by CD11c+ CD80+ CD86+ and CD8α+ Clec9a+ expression, respectively. Despite a lower dose compared with Mono-DCs, mice treated with pulsed Stem-DCs showed a reduced amount of ascitic fluid and lower body weights compared with those of vehicle-treated mice. These mice treated with pulsed stem-DCs appeared to have fewer tumor implants, which were usually confined in the epithelium of tumor-invaded organs. All mice treated with DCs showed longer survival than the vehicle group, especially in the medium/high dose pulsed Stem-DC treatment groups. Moreover, the stem-DC-treated group demonstrated a low proportion of myeloid-derived suppressor cells and regulatory T cells, high interleukin-12 and interferon-γ levels, and accumulation of several tumor-infiltrating lymphocytes. Together, these results indicate that mouse CD8α+ DCs derived from BM-HSCs decrease tumor progression and enhance antitumor immune responses against murine ovarian cancer, suggesting that better DC vaccines can be used as an effective immunotherapy in EOC treatment. Further studies are necessary to develop potent DC vaccines using human CD141+ DCs.
Collapse
Affiliation(s)
- Shin-Wha Lee
- grid.267370.70000 0004 0533 4667Department of Obstetrics and Gynecology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| | - Hyunah Lee
- grid.497660.aPharmicell Co., Seoul, Republic of Korea
| | - Kyung-Won Lee
- grid.413967.e0000 0001 0842 2126Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Min-Je Kim
- grid.413967.e0000 0001 0842 2126Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Sung Wan Kang
- grid.413967.e0000 0001 0842 2126Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Young-Jae Lee
- grid.267370.70000 0004 0533 4667Department of Obstetrics and Gynecology, GangNeung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Republic of Korea
| | - HyunSoo Kim
- grid.497660.aPharmicell Co., Seoul, Republic of Korea
| | - Yong-Man Kim
- grid.267370.70000 0004 0533 4667Department of Obstetrics and Gynecology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| |
Collapse
|
35
|
Elahi Z, Angel PW, Butcher SK, Rajab N, Choi J, Deng Y, Mintern JD, Radford K, Wells CA. The Human Dendritic Cell Atlas: An Integrated Transcriptional Tool to Study Human Dendritic Cell Biology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2352–2361. [PMID: 36427009 PMCID: PMC9719841 DOI: 10.4049/jimmunol.2200366] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/09/2022] [Indexed: 12/24/2022]
Abstract
Dendritic cells (DCs) are functionally diverse and are present in most adult tissues, but deep understanding of human DC biology is hampered by relatively small numbers of these in circulation and their short lifespan in human tissues. We built a transcriptional atlas of human DCs by combining samples from 14 expression profiling studies derived from 10 laboratories. We identified significant gene expression variation of DC subset-defining markers across tissue type and upon viral or bacterial stimulation. We further highlight critical gaps between in vitro-derived DC subsets and their in vivo counterparts and provide evidence that monocytes or cord blood progenitor in vitro-differentiated DCs fail to capture the repertoire of primary DC subsets or behaviors. In constructing a reference DC atlas, we provide an important resource for the community wishing to identify and annotate tissue-specific DC subsets from single-cell datasets, or benchmark new in vitro models of DC biology.
Collapse
Affiliation(s)
- Zahra Elahi
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul W. Angel
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Suzanne K. Butcher
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Nadia Rajab
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Jarny Choi
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Yidi Deng
- Melbourne Integrative Genomics, School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria, Australia
| | - Justine D. Mintern
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia; and
| | - Kristen Radford
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Christine A. Wells
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
36
|
V Deligiorgi M, T Trafalis D. Refining personalized diagnosis, treatment and exploitation of hypothyroidism related to solid nonthyroid cancer. Per Med 2022; 20:87-105. [DOI: 10.2217/pme-2022-0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Hypothyroidism in the setting of cancer is a puzzling entity due to the dual role of the thyroid hormones (TH) in cancer – promoting versus inhibitory – and the complexity of the hypothyroidism itself. The present review provides a comprehensive overview of the personalized approach to hypothyroidism in patients with solid nonthyroid cancer, focusing on current challenges, unmet needs and future perspectives. Major electronic databases were searched from January 2011 until March 2022. The milestones of the refinement of such a personalized approach are prompt diagnosis, proper TH replacement and development of interventions and/or pharmaceutical agents to exploit hypothyroidism or, on the contrary, TH replacement as an anticancer strategy. Further elucidation of the dual role of TH in cancer – especially of the interference of TH signaling with the hallmarks of cancer – is anticipated to inform decision-making and optimize patient selection.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Department of Pharmacology – Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Building 16, 1st Floor, 75 Mikras Asias, Goudi, Athens, 11527, Greece
| | - Dimitrios T Trafalis
- Department of Pharmacology – Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Building 16, 1st Floor, 75 Mikras Asias, Goudi, Athens, 11527, Greece
| |
Collapse
|
37
|
Parsonidis P, Beis G, Iliopoulos AC, Papasotiriou I. Adoptive transfer of activated immune cells against solid tumors: A preliminary study. Cell Immunol 2022; 382:104616. [PMID: 36219944 DOI: 10.1016/j.cellimm.2022.104616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND This study presents preliminary results concerning the effectiveness of a novel immunotherapy in cancer. The proposed adoptive cellular therapy product contains a mixture of effector immune cells, specifically macrophages, NK cells, dendritic cells, cytotoxic T lymphocytes and monoclonal antibody producing plasma cells. METHODS The results were based on both descriptive and inferential statistical analysis of data concerning 17 cancer patients. Particularly, performance scales such as clinical condition, Karnofsky-Index, ECOG index and symptom's scale were evaluated post therapy administration (4 months). Furthermore, circulating tumor cells (CTCs) and a specific tumor marker (EpCAM) were measured pre- and post-cellular therapy. RESULTS The results revealed a positive evaluation for clinical condition (70.59 %), Karnofsky-Index (88.23 %), ECOG index (94.12 %), and symptoms' scale (64.70 %). In addition, statistically significant reductions were found for both CTCs (p = 0.0016) and EpCAM positive cells (p = 0.0005), post-therapy, which were related to large size effects, namely 0.77 and 0.85, respectively. No cytokine storm, anaphylaxis or severe adverse events were observed with 4 months follow up evaluation. CONCLUSIONS These preliminary results indicate that the proposed cellular therapy can be considered for further studies in clinical trials.
Collapse
Affiliation(s)
| | - Georgios Beis
- Research Genetic Cancer Centre S.A., Florina, Greece
| | | | | |
Collapse
|
38
|
Saha C, Bojdo J, Dunne NJ, Duary RK, Buckley N, McCarthy HO. Nucleic acid vaccination strategies for ovarian cancer. Front Bioeng Biotechnol 2022; 10:953887. [PMID: 36420446 PMCID: PMC9677957 DOI: 10.3389/fbioe.2022.953887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 10/26/2022] [Indexed: 09/19/2023] Open
Abstract
High grade serous carcinoma (HGSC) is one of the most lethal ovarian cancers that is characterised by asymptomatic tumour growth, insufficient knowledge of malignant cell origin and sub-optimal detection. HGSC has been recently shown to originate in the fallopian tube and not in the ovaries. Conventional treatments such as chemotherapy and surgery depend upon the stage of the disease and have resulted in higher rates of relapse. Hence, there is a need for alternative treatments. Differential antigen expression levels have been utilised for early detection of the cancer and could be employed in vaccination strategies using nucleic acids. In this review the different vaccination strategies in Ovarian cancer are discussed and reviewed. Nucleic acid vaccination strategies have been proven to produce a higher CD8+ CTL response alongside CD4+ T-cell response when compared to other vaccination strategies and thus provide a good arena for antitumour immune therapy. DNA and mRNA need to be delivered into the intracellular matrix. To overcome ineffective naked delivery of the nucleic acid cargo, a suitable delivery system is required. This review also considers the suitability of cell penetrating peptides as a tool for nucleic acid vaccine delivery in ovarian cancer.
Collapse
Affiliation(s)
- Chayanika Saha
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
| | - James Bojdo
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
| | - Nicholas J. Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, Ireland
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- Advanced Processing Technology Research Centre, Dublin City University, Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Raj Kumar Duary
- Department of Food Engineering and Technology, Tezpur University, Tezpur, India
| | - Niamh Buckley
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
| | - Helen O. McCarthy
- School of Pharmacy, Queen’s University of Belfast, Belfast, United Kingdom
- School of Chemical Sciences, Dublin City University, Dublin, Ireland
| |
Collapse
|
39
|
Fu C, Ma T, Zhou L, Mi QS, Jiang A. Dendritic Cell-Based Vaccines Against Cancer: Challenges, Advances and Future Opportunities. Immunol Invest 2022; 51:2133-2158. [PMID: 35946383 DOI: 10.1080/08820139.2022.2109486] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
As the most potent professional antigen presenting cells, dendritic cells (DCs) have the ability to activate both naive CD4 and CD8 T cells. Recognized for their exceptional ability to cross-present exogenous antigens to prime naive antigen-specific CD8 T cells, DCs play a critical role in generating CD8 T cell immunity, as well as mediating CD8 T cell tolerance to tumor antigens. Despite the ability to potentiate host CD8 T cell-mediated anti-tumor immunity, current DC-based cancer vaccines have not yet achieved the promised success clinically with the exception of FDA-approved Provenge. Interestingly, recent studies have shown that type 1 conventional DCs (cDC1s) play a critical role in cross-priming tumor-specific CD8 T cells and determining the anti-tumor efficacy of cancer immunotherapies including immune checkpoint blockade (ICB). Together with promising clinical results in neoantigen-based cancer vaccines, there is a great need for DC-based vaccines to be further developed and refined either as monotherapies or in combination with other immunotherapies. In this review, we will present a brief review of DC development and function, discuss recent progress, and provide a perspective on future directions to realize the promising potential of DC-based cancer vaccines.
Collapse
Affiliation(s)
- Chunmei Fu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| | - Tianle Ma
- Department of Computer Science and Engineering, School of Engineering and Computer Science, Oakland University, Rochester, Michigan, USA
| | - Li Zhou
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| | - Aimin Jiang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, Michigan, USA
| |
Collapse
|
40
|
Brückner M, Fichter M, da Costa Marques R, Landfester K, Mailänder V. PEG Spacer Length Substantially Affects Antibody-Based Nanocarrier Targeting of Dendritic Cell Subsets. Pharmaceutics 2022; 14:pharmaceutics14081614. [PMID: 36015239 PMCID: PMC9414227 DOI: 10.3390/pharmaceutics14081614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/27/2022] [Accepted: 07/31/2022] [Indexed: 02/01/2023] Open
Abstract
Successful cell targeting depends on the controlled positioning of cell-type-specific antibodies on the nanocarrier’s (NC) surface. Uncontrolled antibody immobilization results in unintended cell uptake due to Fc-mediated cell interaction. Consequently, precise immobilization of the Fc region towards the nanocarrier surface is needed with the Fab regions staying freely accessible for antigen binding. Moreover, the antibody needs to be a certain distance from the nanocarrier surface, influencing the targeting performance after formation of the biomolecular corona. This can be achieved by using PEG linker molecules. Here we demonstrate cell type-specific targeting for dendritic cells (DC) as cellular key regulators of immune responses. However, to date, dendritic cell targeting experiments using different linker lengths still need to be conducted. Consequently, we focused on the surface modification of nanocarriers with different molecular weight PEG linkers (0.65, 2, and 5 kDa), and their ability to reduce undesired cell uptake, while achieving efficient DC targeting via covalently immobilized antibodies (stealth targeting). Our findings demonstrate that the PEG linker length significantly affects active dendritic cell targeting from cell lines (DC2.4) to primary cells (BMDCs, splenocytic conventional DCs type 1 (cDC1)). While antibody-functionalized nanocarriers with a shorter PEG length (0.65 kDa) showed the best targeting in DC2.4, a longer PEG length (5 kDa) was required to specifically accumulate in BMDCs and splenocytic cDC1. Our study highlights that these crucial aspects must be considered when targeting dendritic cell subsets, which are of great importance in the fields of cancer immunotherapy and vaccine development.
Collapse
Affiliation(s)
- Maximilian Brückner
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.B.); (M.F.); (R.d.C.M.)
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany;
| | - Michael Fichter
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.B.); (M.F.); (R.d.C.M.)
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany;
| | - Richard da Costa Marques
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.B.); (M.F.); (R.d.C.M.)
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany;
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany;
| | - Volker Mailänder
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.B.); (M.F.); (R.d.C.M.)
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany;
- Correspondence:
| |
Collapse
|
41
|
Biscari L, Kaufman CD, Farré C, Huhn V, Pacini MF, Balbi CB, Gómez KA, Pérez AR, Alloatti A. Immunization With Lipopolysaccharide-Activated Dendritic Cells Generates a Specific CD8+ T Cell Response That Confers Partial Protection Against Infection With Trypanosoma cruzi. Front Cell Infect Microbiol 2022; 12:897133. [PMID: 35903201 PMCID: PMC9318436 DOI: 10.3389/fcimb.2022.897133] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Lipopolysaccharide (LPS) induces the activation of dendritic cells (DCs) throughout the engagement of toll-like receptor 4. LPS-activated DCs show increased capacity to process and present pathogen-derived antigens to activate naïve T cells. DCs-based vaccines have been successfully used to treat some cancer types, and lately transferred to the field of infectious diseases, in particular against HIV. However, there is no vaccine or DC therapy for any parasitic disease that is currently available. The immune response against Trypanosoma cruzi substantially relies on T cells, and both CD4+ and CD8+ T lymphocytes are required to control parasite growth. Here, we develop a vaccination strategy based on DCs derived from bone marrow, activated with LPS and loaded with TsKb20, an immunodominant epitope of the trans-sialidase family of proteins. We extensively characterized the CD8+ T cell response generated after immunization and compared three different readouts: a tetramer staining, ELISpot and Activation-Induced Marker (AIM) assays. To our knowledge, this work shows for the first time a proper set of T cell markers to evaluate specific CD8+ T cell responses in mice. We also show that our immunization scheme confers protection against T. cruzi, augmenting survival and reducing parasite burden in female but not male mice. We conclude that the immunization with LPS-activated DCs has the potential to prime significant CD8+ T cell responses in C57BL/6 mice independently of the sex, but this response will only be effective in female, possibly due to mice sexual dimorphisms in the response generated against T. cruzi.
Collapse
Affiliation(s)
- Lucía Biscari
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Cintia Daniela Kaufman
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Cecilia Farré
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Victoria Huhn
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - María Florencia Pacini
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Camila Bulfoni Balbi
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Karina Andrea Gómez
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular Dr. Héctor N. Torres (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ana Rosa Pérez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Andrés Alloatti
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
- *Correspondence: Andrés Alloatti,
| |
Collapse
|
42
|
Deligiorgi MV, Trafalis DT. The Clinical Relevance of Hypothyroidism in Patients with Solid Non-Thyroid Cancer: A Tantalizing Conundrum. J Clin Med 2022; 11:3417. [PMID: 35743483 PMCID: PMC9224934 DOI: 10.3390/jcm11123417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023] Open
Abstract
Hypothyroidism in patients with solid non-thyroid cancer is a tantalizing entity, integrating an intriguing thyroid hormones (THs)-cancer association with the complexity of hypothyroidism itself. The present narrative review provides a comprehensive overview of the clinical relevance of hypothyroidism in solid non-thyroid cancer. Hypothyroidism in patients with solid non-thyroid cancer is reminiscent of hypothyroidism in the general population, yet also poses distinct challenges due to the dual role of THs in cancer: promoting versus inhibitory. Close collaboration between oncologists and endocrinologists will enable the prompt and personalized diagnosis and treatment of hypothyroidism in patients with solid non-thyroid cancer. Clinical data indicate that hypothyroidism is a predictor of a decreased or increased risk of solid non-thyroid cancer and is a prognostic factor of favorable or unfavorable prognosis in solid non-thyroid cancer. However, the impact of hypothyroidism with respect to the risk and/or prognosis of solid non-thyroid cancer is not a consistent finding. To harness hypothyroidism, or THs replacement, as a personalized anticancer strategy for solid non-thyroid cancer, four prerequisites need to be fulfilled, namely: (i) deciphering the dual THs actions in cancer; (ii) identifying interventions in THs status and developing agents that block tumor-promoting THs actions and/or mimic anticancer THs actions; (iii) appropriate patient selection; and (iv) counteracting current methodological limitations.
Collapse
Affiliation(s)
- Maria V. Deligiorgi
- Department of Pharmacology—Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Str., Goudi, 11527 Athens, Greece;
| | | |
Collapse
|
43
|
Parsonidis P, Papasotiriou I. Adoptive Cellular Transfer Immunotherapies for Cancer. Cancer Treat Res Commun 2022; 32:100575. [PMID: 35679756 DOI: 10.1016/j.ctarc.2022.100575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 06/15/2023]
Abstract
Patients with cancer require efficient treatment approaches as the mortality rate due to their disease is high. Conventional therapies, like chemotherapy and radiation, have severe side effects. Drug discovery is focusing on the development of alternative strategies that could have beneficial effects to the patients. Cellular therapies are potential therapeutics, and the generation of new products is growing fast. The concept involves the isolation of immune cells, ex vivo activation and reinfusion into the patient. The goal is to boost the immune cells to fight cancer cells. Different immune cells can be used, including dendritic cells, T cells, NK cells, macrophages and B lymphocytes. Some products have already gained FDA approval, while many more are currently in clinical trials. Research is focusing on the improvement of the function of the cells that may require genetic modification or combination with other therapies. Finally, it is crucial to develop novel technologies that could be used in monitoring of the immune profile of patients that have received a cellular therapy to assess the efficacy of the treatment.
Collapse
|
44
|
Modak M, Mattes AK, Reiss D, Skronska-Wasek W, Langlois R, Sabarth N, Konopitzky R, Ramírez F, Lehr K, Mayr T, Kind D, Viollet C, Swee LK, Petschenka J, El Kasmi KC, Noessner E, Kitt K, Pflanz S. CD206+ tumor-associated macrophages cross-present tumor antigen and drive anti-tumor immunity. JCI Insight 2022; 7:155022. [PMID: 35503656 PMCID: PMC9220841 DOI: 10.1172/jci.insight.155022] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 04/22/2022] [Indexed: 11/29/2022] Open
Abstract
In many solid cancers, tumor-associated macrophages (TAM) represent the predominant myeloid cell population. Antigen (Ag) cross-presentation leading to tumor Ag–directed cytotoxic CD8+ T cell responses is crucial for antitumor immunity. However, the role of recruited monocyte-derived macrophages, including TAM, as potential cross-presenting cells is not well understood. Here, we show that primary human as well as mouse CD206+ macrophages are effective in functional cross-presentation of soluble self-Ag and non–self-Ag, including tumor-associated Ag (TAA), as well as viral Ag. To confirm the presence of cross-presenting TAM in vivo, we performed phenotypic and functional analysis of TAM from B16-F10 and CT26 syngeneic tumor models and have identified CD11b+F4/80hiCD206+ TAM to effectively cross-present TAA. We show that CD11b+CD206+ TAM represent the dominant tumor-infiltrating myeloid cell population, expressing a unique cell surface repertoire, promoting Ag cross-presentation and Ag-specific CD8+ T cell activation comparable with cross-presenting CLEC9A+ DCs (cDC1). The presence of cross-presenting CD206+ TAM is associated with reduced tumor burden in mouse syngeneic tumor models and with improved overall survival in cutaneous melanoma patients. Therefore, the demonstration of effective Ag cross-presentation capabilities of CD206+ TAM, including their clinical relevance, expands our understanding of TAM phenotypic diversity and functional versatility.
Collapse
Affiliation(s)
- Madhura Modak
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Ann-Kathrin Mattes
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Daniela Reiss
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Wioletta Skronska-Wasek
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Rebecca Langlois
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Nicolas Sabarth
- Department of Biotherapeutics Discovery, Boehringer Ingelheim RCV GmbH & Co KG., Vienna, Austria
| | - Renate Konopitzky
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmbH & Co. KG, Vienna, Austria
| | - Fidel Ramírez
- Department of Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Katharina Lehr
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Tobias Mayr
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - David Kind
- Department of Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Coralie Viollet
- Department of Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Lee Kim Swee
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Jutta Petschenka
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Karim Christian El Kasmi
- Department of Immunology and Respiratory, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Elfriede Noessner
- Immunoanalytics- Research Group Tissue Control of Immunocytes, Deutsches Forschungszentrum für Gesundheit und Umwelt, Helmholtz Zentrum, Munich, Germany
| | - Kerstin Kitt
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Stefan Pflanz
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
45
|
Kalus P, De Munck J, Vanbellingen S, Carreer L, Laeremans T, Broos K, Dufait I, Schwarze JK, Van Riet I, Neyns B, Breckpot K, Aerts JL. Oncolytic Herpes Simplex Virus Type 1 Induces Immunogenic Cell Death Resulting in Maturation of BDCA-1 + Myeloid Dendritic Cells. Int J Mol Sci 2022; 23:ijms23094865. [PMID: 35563257 PMCID: PMC9103433 DOI: 10.3390/ijms23094865] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023] Open
Abstract
Recently, a paradigm shift has been established for oncolytic viruses (OVs) as it was shown that the immune system plays an important role in the specific killing of tumor cells by OVs. OVs have the intrinsic capacity to provide the right signals to trigger anti-tumor immune responses, on the one hand by delivering virus-derived innate signals and on the other hand by inducing immunogenic cell death (ICD), which is accompanied by the release of various damage-associated molecules from infected tumor cells. Here, we determined the ICD-inducing capacity of Talimogene laherparepvec (T-VEC), a herpes simplex virus type 1 based OV, and benchmarked this to other previously described ICD (e.g., doxorubicin) and non-ICD inducing agents (cisplatin). Furthermore, we studied the capability of T-VEC to induce the maturation of human BDCA-1+ myeloid dendritic cells (myDCs). We found that T-VEC treatment exerts direct and indirect anti-tumor effects as it induces tumor cell death that coincides with the release of hallmark mediators of ICD, while simultaneously contributing to the maturation of BDCA-1+ myDCs. These results unequivocally cement OVs in the category of cancer immunotherapy.
Collapse
Affiliation(s)
- Philipp Kalus
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Vrije Universiteit Brussel (VUB), 1000 Brussels, Belgium; (P.K.); (J.D.M.); (S.V.); (L.C.); (T.L.)
| | - Jolien De Munck
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Vrije Universiteit Brussel (VUB), 1000 Brussels, Belgium; (P.K.); (J.D.M.); (S.V.); (L.C.); (T.L.)
| | - Sarah Vanbellingen
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Vrije Universiteit Brussel (VUB), 1000 Brussels, Belgium; (P.K.); (J.D.M.); (S.V.); (L.C.); (T.L.)
| | - Laura Carreer
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Vrije Universiteit Brussel (VUB), 1000 Brussels, Belgium; (P.K.); (J.D.M.); (S.V.); (L.C.); (T.L.)
| | - Thessa Laeremans
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Vrije Universiteit Brussel (VUB), 1000 Brussels, Belgium; (P.K.); (J.D.M.); (S.V.); (L.C.); (T.L.)
| | - Katrijn Broos
- Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel (VUB), 1000 Brussels, Belgium; (K.B.); (K.B.)
| | - Inès Dufait
- Department of Radiotherapy, Laboratory of Translational Radiation Oncology, Supportive Care and Physics, Vrije Universiteit Brussel (VUB), 1000 Brussels, Belgium;
| | - Julia K. Schwarze
- Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), 1000 Brussels, Belgium; (J.K.S.); (B.N.)
| | - Ivan Van Riet
- Stem Cell Laboratory, Departement of Hematology, Universitair Ziekenhuis Brussel (UZ Brussel), 1000 Brussels, Belgium;
| | - Bart Neyns
- Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), 1000 Brussels, Belgium; (J.K.S.); (B.N.)
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel (VUB), 1000 Brussels, Belgium; (K.B.); (K.B.)
| | - Joeri L. Aerts
- Laboratory for Neuro-Aging and Viro-Immunotherapy (NAVI), Vrije Universiteit Brussel (VUB), 1000 Brussels, Belgium; (P.K.); (J.D.M.); (S.V.); (L.C.); (T.L.)
- Correspondence:
| |
Collapse
|
46
|
Johnson AO, Fowler SB, Webster CI, Brown AJ, James DC. Bioinformatic Design of Dendritic Cell-Specific Synthetic Promoters. ACS Synth Biol 2022; 11:1613-1626. [PMID: 35389220 PMCID: PMC9016764 DOI: 10.1021/acssynbio.2c00027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
Next-generation DNA vectors for cancer
immunotherapies and vaccine
development require promoters eliciting predefined transcriptional
activities specific to target cell types, such as dendritic cells
(DCs), which underpin immune response. In this study, we describe
the de novo design of DC-specific synthetic promoters via in silico assembly of cis-transcription
factor response elements (TFREs) that harness the DC transcriptional
landscape. Using computational genome mining approaches, candidate
TFREs were identified within promoter sequences of highly expressed
DC-specific genes or those exhibiting an upregulated expression during
DC maturation. Individual TFREs were then screened in vitro in a target DC line and off-target cell lines derived from skeletal
muscle, fibroblast, epithelial, and endothelial cells using homotypic
(TFRE repeats in series) reporter constructs. Based on these data,
a library of heterotypic promoter assemblies varying in the TFRE composition,
copy number, and sequential arrangement was constructed and tested in vitro to identify DC-specific promoters. Analysis of
the transcriptional activity and specificity of these promoters unraveled
underlying design rules, primarily TFRE composition, which govern
the DC-specific synthetic promoter activity. Using these design rules,
a second library of exclusively DC-specific promoters exhibiting varied
transcriptional activities was generated. All DC-specific synthetic
promoter assemblies exhibited >5-fold activity in the target DC
line
relative to off-target cell lines, with transcriptional activities
ranging from 8 to 67% of the nonspecific human cytomegalovirus (hCMV-IE1)
promoter. We show that bioinformatic analysis of a mammalian cell
transcriptional landscape is an effective strategy for de
novo design of cell-type-specific synthetic promoters with
precisely controllable transcriptional activities.
Collapse
Affiliation(s)
- Abayomi O. Johnson
- Department of Chemical and Biological Engineering, University of Sheffield, Mappin Street, Sheffield S1 3JD, U.K
- SynGenSys Limited, Freeths LLP, Norfolk Street, Sheffield S1 2JE, U.K
| | - Susan B. Fowler
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge CB21 6GH, U.K
| | - Carl I. Webster
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB21 6GH, U.K
| | - Adam J. Brown
- Department of Chemical and Biological Engineering, University of Sheffield, Mappin Street, Sheffield S1 3JD, U.K
- SynGenSys Limited, Freeths LLP, Norfolk Street, Sheffield S1 2JE, U.K
| | - David C. James
- Department of Chemical and Biological Engineering, University of Sheffield, Mappin Street, Sheffield S1 3JD, U.K
- SynGenSys Limited, Freeths LLP, Norfolk Street, Sheffield S1 2JE, U.K
| |
Collapse
|
47
|
Žilionytė K, Bagdzevičiūtė U, Mlynska A, Urbštaitė E, Paberalė E, Dobrovolskienė N, Krasko JA, Pašukonienė V. Functional antigen processing and presentation mechanism as a prerequisite factor of response to treatment with dendritic cell vaccines and anti-PD-1 in preclinical murine LLC1 and GL261 tumor models. Cancer Immunol Immunother 2022; 71:2691-2700. [PMID: 35364740 DOI: 10.1007/s00262-022-03190-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/14/2022] [Indexed: 12/15/2022]
Abstract
Low efficacy of cancer immunotherapy encourages the search for possible resistance mechanisms and biomarkers that would predict the outcome of immunotherapy in oncology patients. Most cancer immunotherapies act on T lymphocytes, which can specifically recognize and kill tumor cells. However, for immunotherapy-activated T lymphocytes to be able to perform these functions, proper tumor Ag processing and surface presentation by MHC-I molecule is important. Knowing the significance of Ag processing and presentation mechanism (APM) in anti-tumor immune response, we sought to evaluate how the functionality of APM affects tumor immune microenvironment and response to dendritic cell vaccines (DCV) and anti-PD-1. By comparing murine Lewis lung carcinoma LLC1 and glioma GL261 models a decreased expression of APM-related genes, such as Psmb8, Psmb9, Psmb10, Tap1, Tap2, Erap1, B2m, and low expression of surface MHC-I molecule were found in LLC1 cells. Changes in APM-related gene expression affected the ability of T lymphocytes to recognize and kill LLC1 cells, resulting in the absence of cytotoxic immune response and resistance to DCV and anti-PD-1. An emerging cytotoxic immune reaction and sensitivity to DCV and anti-PD-1 were observed in GL261 tumors where APM remained functional. This study demonstrates that one of the possible mechanisms of tumor resistance to immunotherapy is a dysfunctional APM and reveals a predictive potential of APM-related gene set expression for the personalization of dendritic cell vaccine and anti-PD-1 therapies in murine pre-treated tumors.
Collapse
Affiliation(s)
- Karolina Žilionytė
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania. .,Life Sciences Center, Vilnius University, Vilnius, Lithuania.
| | - Ugnė Bagdzevičiūtė
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Agata Mlynska
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | | | - Emilija Paberalė
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | | | - Jan Aleksander Krasko
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Vita Pašukonienė
- Laboratory of Immunology, National Cancer Institute, Vilnius, Lithuania.,Department of Chemistry and Bioengineering, Vilnius Gediminas Technical University, Vilnius, Lithuania
| |
Collapse
|
48
|
Agostinetto E, Losurdo A, Nader-Marta G, Santoro A, Punie K, Barroso R, Popovic L, Solinas C, Kok M, de Azambuja V, Lambertini M. Progress and pitfalls in the use of immunotherapy for patients with triple negative breast cancer. Expert Opin Investig Drugs 2022; 31:567-591. [PMID: 35240902 DOI: 10.1080/13543784.2022.2049232] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Triple negative breast cancer (TNBC) is an area of high unmet medical need in terms of new effective treatment strategies. Although breast cancer is traditionally considered a 'cold' tumor type, TNBC is the most appropriate subtype for immunotherapeutic strategies; this is due to the high level of tumor infiltrating lymphocytes, PD-L1 expression and tumor mutational burden relative to other breast cancer subtypes. AREAS COVERED This review examines the use of immunotherapeutic strategies in early and advanced TNBC. The paper summarizes data on novel promising immunomodulatory approaches that have been explored in early phase trials and discusses the pitfalls and limitations often encountered in clinical research. EXPERT OPINION PD-1-blockade is approved for stage II/III TNBC and for first-line treatment of PD-L1-positive TNBC patients with metastatic disease and should be considered standard of care. However, question marks and difficulties remain; these include the identification of predictive biomarkers to select patients who benefit from the addition of PD1-blockade and the balance between efficacy and long-term toxicity for an individual patient. Numerous treatment combinations and new immunotherapeutic strategies beyond PD1 blockade are being evaluated, thus reflecting a promising evolution of a more personalized approach, and extended clinical benefit in TNBC.
Collapse
Affiliation(s)
- Elisa Agostinetto
- Academic Trials Promoting Team, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium.,Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| | - Agnese Losurdo
- Humanitas Research Hospital - IRCCS, Humanitas Cancer Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Guilherme Nader-Marta
- Academic Trials Promoting Team, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy.,Humanitas Research Hospital - IRCCS, Humanitas Cancer Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | | | - Lazar Popovic
- Oncology Institute of Vojvodina, Faculty of Medicine, University Novi Sad, Novi Sad, Serbia
| | - Cinzia Solinas
- Medical Oncology, ATS Sardegna, Ospedale San Francesco, Nuoro, Italy
| | - Marleen Kok
- Departments of Medical Oncology, Tumor Biology & Immunology. Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Vandro de Azambuja
- Academic Trials Promoting Team, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy.,Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
49
|
Espinar-Buitrago M, Muñoz-Fernández MA. New Approaches to Dendritic Cell-Based Therapeutic Vaccines Against HIV-1 Infection. Front Immunol 2022; 12:719664. [PMID: 35058917 PMCID: PMC8763680 DOI: 10.3389/fimmu.2021.719664] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
Due to the success of combined antiretroviral therapy (cART) in recent years, the pathological outcome of Human Immunodeficiency Virus type 1 (HIV-1) infection has improved substantially, achieving undetectable viral loads in most cases. Nevertheless, the presence of a viral reservoir formed by latently infected cells results in patients having to maintain treatment for life. In the absence of effective eradication strategies against HIV-1, research efforts are focused on obtaining a cure. One of these approaches is the creation of therapeutic vaccines. In this sense, the most promising one up to now is based on the establishing of the immunological synapse between dendritic cells (DCs) and T lymphocytes (TL). DCs are one of the first cells of the immune system to encounter HIV-1 by acting as antigen presenting cells, bringing about the interaction between innate and adaptive immune responses mediated by TL. Furthermore, TL are the end effector, and their response capacity is essential in the adaptive elimination of cells infected by pathogens. In this review, we summarize the knowledge of the interaction between DCs with TL, as well as the characterization of the specific T-cell response against HIV-1 infection. The use of nanotechnology in the design and improvement of vaccines based on DCs has been researched and presented here with a special emphasis.
Collapse
Affiliation(s)
- Marisierra Espinar-Buitrago
- Section Head Immunology, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Ma Angeles Muñoz-Fernández
- Section Head Immunology, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Spanish Human Immunodeficiency Virus- Hospital Gregorio Marañón (HIV-HGM) BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
50
|
Plesca I, Müller L, Böttcher JP, Medyouf H, Wehner R, Schmitz M. Tumor-associated human dendritic cell subsets: phenotype, functional orientation, and clinical relevance. Eur J Immunol 2022; 52:1750-1758. [PMID: 35106759 DOI: 10.1002/eji.202149487] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DCs) play a pivotal role in orchestrating innate and adaptive antitumor immunity. Activated DCs can produce large amounts of various proinflammatory cytokines, initiate T cell responses, and exhibit direct cytotoxicity against tumor cells. They also efficiently enhance the antitumoral properties of natural killer cells and T lymphocytes. Based on these capabilities, immunogenic DCs promote tumor elimination and are associated with improved survival of patients. Furthermore, they can essentially contribute to the clinical efficacy of immunotherapeutic strategies for cancer patients. However, depending on their intrinsic properties and the tumor microenvironment, DCs can be rendered dysfunctional and mediate tolerance by producing immunosuppressive cytokines and activating regulatory T cells. Such tolerogenic DCs can foster tumor progression and are linked to poor prognosis of patients. Here, we focus on recent studies exploring the phenotype, functional orientation, and clinical relevance of tumor-infiltrating conventional DC1, conventional DC2, plasmacytoid DCs, and monocyte-derived DCs in translational and clinical settings. In addition, recent findings demonstrating the influence of DCs on the efficacy of immunotherapeutic strategies are summarized. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ioana Plesca
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Luise Müller
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Jan P Böttcher
- Institute of Molecular Immunology and Experimental Oncology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Hind Medyouf
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt/Main, Germany.,Frankfurt Cancer Institute, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rebekka Wehner
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|