1
|
Cavic G, Fahrer AM. Tumor-seeking bacterial missiles. Immunol Cell Biol 2025. [PMID: 39757005 DOI: 10.1111/imcb.12844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
In this Research Highlight, we discuss an exceptionally elegant and far‐reaching study from the Arpaia lab at Columbia University, who have engineered a probiotic Escherichia coli strain to eliminate cancer.
Collapse
Affiliation(s)
- George Cavic
- Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | - Aude M Fahrer
- Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, ACT, Australia
- Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| |
Collapse
|
2
|
Grant M, Ni Lee L, Chinnakannan S, Tong O, Kwok J, Cianci N, Tillman L, Saha A, Pereira Almeida V, Leung C. Unlocking cancer vaccine potential: What are the key factors? Hum Vaccin Immunother 2024; 20:2331486. [PMID: 38564321 PMCID: PMC11657071 DOI: 10.1080/21645515.2024.2331486] [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: 01/08/2024] [Revised: 03/05/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer is a global health challenge, with changing demographics and lifestyle factors producing an increasing burden worldwide. Screening advancements are enabling earlier diagnoses, but current cancer immunotherapies only induce remission in a small proportion of patients and come at a high cost. Cancer vaccines may offer a solution to these challenges, but they have been mired by poor results in past decades. Greater understanding of tumor biology, coupled with the success of vaccine technologies during the COVID-19 pandemic, has reinvigorated cancer vaccine development. With the first signs of efficacy being reported, cancer vaccines may be beginning to fulfill their potential. Solid tumors, however, present different hurdles than infectious diseases. Combining insights from previous cancer vaccine clinical development and contemporary knowledge of tumor immunology, we ask: who are the 'right' patients, what are the 'right' targets, and which are the 'right' modalities to maximize the chances of cancer vaccine success?
Collapse
|
3
|
Giudice GC, Sonpavde GP. Vaccine approaches to treat urothelial cancer. Hum Vaccin Immunother 2024; 20:2379086. [PMID: 39043175 PMCID: PMC11268260 DOI: 10.1080/21645515.2024.2379086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/08/2024] [Indexed: 07/25/2024] Open
Abstract
Bladder cancer (BC) accounts for about 4% of all malignancies. Non-muscle-invasive BC, 75% of cases, is treated with transurethral resection and adjuvant intravesical instillation, while muscle-invasive BC warrants cisplatin-based perioperative chemotherapy. Although immune-checkpoint inhibitors, antibody drug conjugates and targeted agents have provided dramatic advances, metastatic BC remains a generally incurable disease and clinical trials continue to vigorously evaluate novel molecules. Cancer vaccines aim at activating the patient's immune system against tumor cells. Several means of delivering neoantigens have been developed, including peptides, antigen-presenting cells, virus, or nucleic acids. Various improvements are constantly being explored, such as adjuvants use and combination strategies. Nucleic acids-based vaccines are increasingly gaining attention in recent years, with promising results in other malignancies. However, despite the recent advantages, numerous obstacles persist. This review is aimed at describing the different types of cancer vaccines, their evaluations in UC patients and the more recent innovations in this field.
Collapse
Affiliation(s)
- Giulia Claire Giudice
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Guru P. Sonpavde
- AdventHealth Cancer Institute, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
4
|
Chen S, Li B, Liu B. AIEgen-biomacromolecule conjugates: Visualized delivery and light-controlled theranostic platforms. J Control Release 2024; 378:605-618. [PMID: 39716664 DOI: 10.1016/j.jconrel.2024.12.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Biomacromolecules play a critical role in advancing disease diagnosis and treatment. Traditional carriers often lack real-time tracking capabilities, controlled drug release, and may induce adverse effects for delivering biomacromolecules. Aggregation-induced emission luminogens (AIEgens) provide significant advantages in biomacromolecule delivery, enabling real-time fluorescence imaging and reactive oxygen species generation for photodynamic therapy (PDT). This dual functionality allows for the visualization of the biomacromolecule delivery process, providing valuable insights into biodistribution, cellular uptake, and drug-cell interactions. Additionally, the light-responsive nature of AIEgens enables precise spatial-temporal control over cargo release and imaging-guided PDT with minimal side effects. In this perspective, we summarize recent advancements in the use of AIEgens for visualized delivery and light-controlled theranostic applications of biomacromolecules, highlighting their potential to overcome challenges in targeted imaging and precision treatments. Key topics covered include covalent linkage strategies and the biomedical applications of AIEgen-functionalized biomacromolecules, including nucleic acids, proteins, polysaccharides, and lipids. We further highlight AIEgen-functionalized gene therapy and PDT for cancer, peptide transport for disease diagnosis, as well as polysaccharides and lipids delivery in antimicrobial treatments. This perspective concludes by addressing future challenges and opportunities, emphasizing the potential of AIEgen-functionalized biomacromolecules to advance precision theranostics.
Collapse
Affiliation(s)
- Siqin Chen
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585, Singapore
| | - Bowen Li
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585, Singapore
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585, Singapore.
| |
Collapse
|
5
|
Meng Y, Yao Z, Ke X, Hu M, Ren H, Gao S, Zhang H. Extracellular vesicles-based vaccines: Emerging immunotherapies against cancer. J Control Release 2024; 378:438-459. [PMID: 39667569 DOI: 10.1016/j.jconrel.2024.12.010] [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: 08/01/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Cancer vaccines are promising therapeutic approaches to enhance specific T-cell immunity against most solid tumors. By stimulating anti-tumor immunity, clearing minimal residual disease, and minimizing adverse effects, these vaccines target tumor cells and are effective when combined with immune checkpoint blockade or other immunotherapies. However, the development of tumor cell-based vaccines faces quality issues due to poor immunogenicity, tumor heterogeneity, a suppressive tumor immune microenvironment, and ineffective delivery methods. In contrast, extracellular vesicles (EVs), naturally released by cells, are considered the ideal drug carriers and vaccine platforms. EVs offer highly organ-specific targeting, induce broader and more effective immune responses, and demonstrate superior tissue delivery ability. The development of EV vaccines is crucial for advancing cancer immunotherapy. Compared to cell-based vaccines, EV vaccines produced under Good Manufacturing Practices (GMP) offer advantages such as high safety, ease of preservation and transport, and a wide range of sources. This review summarizes the latest research findings on EV vaccine and potential applications in this field. It also highlights novel neoantigens for the development of EV vaccines against cancer.
Collapse
Affiliation(s)
- Yuhua Meng
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Zhimeng Yao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China; Department of Urology Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiurong Ke
- Department of Surgery, Laboratory for Translational Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mengyuan Hu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Hongzheng Ren
- Gongli Hospital of Shanghai Pudong New Area, Department of Pathology, Shanghai, China
| | - Shegan Gao
- College of Clinical Medicine, The First Affiliated Hospital of Henan University of Science and Technology, Henan Key Laboratory of Cancer Epigenetics, Luoyang, Henan, China.
| | - Hao Zhang
- Gongli Hospital of Shanghai Pudong New Area, Department of Pathology, Shanghai, China; Department of Pathology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, Guangdong, China; Department of Thoracic Surgery and General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
6
|
Tang S, Tang R, Chen G, Zhang D, Lin K, Yang H, Fu J, Guo Y, Lin F, Dong X, Huang T, Kong J, Yin X, Ge A, Lin Q, Wu M, Liu X, Zeng Y, Cai Z. Personalized neoantigen hydrogel vaccine combined with PD-1 and CTLA-4 double blockade elicits antitumor response in liver metastases by activating intratumoral CD8 +CD69 + T cells. J Immunother Cancer 2024; 12:e009543. [PMID: 39694701 DOI: 10.1136/jitc-2024-009543] [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] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Liver metastasis is highly aggressive and immune tolerant, and lacks effective treatment strategies. This study aimed to develop a neoantigen hydrogel vaccine (NPT-gels) with high clinical feasibility and further investigate its efficacy and antitumor molecular mechanisms in combination with immune checkpoint inhibitors (ICIs) for the treatment of liver metastases. METHODS The effects of liver metastasis on survival and intratumor T-cell subpopulation infiltration in patients with advanced tumors were investigated using the Surveillance, Epidemiology, and End Results Program (SEER) database and immunofluorescence staining, respectively. NPT-gels were prepared using hyaluronic acid, screened neoantigen peptides, and dual clinical adjuvants [Poly(I:C) and thymosin α-1]. Then, the efficacy and corresponding antitumor molecular mechanisms of NPT-gels combined with programmed death receptor 1 and cytotoxic T-lymphocyte-associated protein 4 double blockade (PCDB) for the treatment of liver metastases were investigated using various preclinical liver metastasis models. RESULTS Liver metastases are associated with poorer 5-year overall survival, characterized by low infiltration of cytotoxic CD8+ T cells and high infiltration of regulatory T cells (Tregs). NPT-gels overcame the challenges faced by conventional neoantigen peptide vaccines by sustaining a durable, high-intensity immune response with a single injection and significantly improving the infiltration of neoantigen-specific T-cell subpopulations in different mice subcutaneous tumor models. Importantly, NPT-gels further combined with PCDB could enhance neoantigen-specific T-cell infiltration and effectively unlock the immunosuppressive microenvironment of liver metastases, showing superior antitumor efficacy and inducing long-term immune memory in various preclinical liver metastasis models without obvious toxicity. Mechanistically, the combined strategy can inhibit Tregs, induce the production and infiltration of neoantigen-specific CD8+CD69+ T cells to enhance the immune response, and potentially elicit antigen-presenting effects in Naïve B_Ighd+ cells and M1-type macrophages. CONCLUSIONS This study demonstrated that NPT-gels combined with PCDB could exert a durable and powerful antitumor immunity by enhancing the recruitment and activation of CD8+CD69+ T cells, which supports the rationale and clinical translation of this combination strategy and provides important evidence for further improving the immunotherapy efficacy of liver metastases in the future.
Collapse
Affiliation(s)
- Shichuan Tang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The First Clinical Medical College of Fujian Medical University, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Ruijing Tang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Geng Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Kongying Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Huan Yang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Jun Fu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Yutong Guo
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Fangzhou Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Xiuqing Dong
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Tingfeng Huang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The First Clinical Medical College of Fujian Medical University, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Jie Kong
- Department of Hepatobiliary, Heze Municipal Hospital, Heze, Shandong, P. R. China
| | - Xiaowei Yin
- Department of Pathology, Shanxian Central Hospital, Heze, Shandong, P. R. China
| | - Aimin Ge
- Department of Pathology, Heze Municipal Hospital, Heze, Shandong, P. R. China
| | - Qizhu Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The First Clinical Medical College of Fujian Medical University, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, Fujian, P. R. China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, P. R. China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, Fujian, P. R. China
| |
Collapse
|
7
|
Wang X, Jiang L, Zhao J, Wu M, Xiong J, Wu X, Weng X. In silico neoantigen screening and HLA multimer-based validation identify immunogenic neopeptide in multifocal lung adenocarcinoma. Front Immunol 2024; 15:1456209. [PMID: 39720721 PMCID: PMC11666526 DOI: 10.3389/fimmu.2024.1456209] [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: 06/28/2024] [Accepted: 11/04/2024] [Indexed: 12/26/2024] Open
Abstract
Background Mutations commonly occur in cancer cells, arising neoantigen as potential targets for personalized immunotherapy of lung adenocarcinoma (LUAD). However, the substantial heterogeneity observed among individuals and distinct foci within the same patient presents significant challenges in formulating immunotherapy strategies. The aim of the work is to characterize the mutation pattern and identify neopeptides across different patients and diverse foci within the same patients with LUAD. Methods Seven lung adenocarcinoma samples and matched tissues/blood are collected from 4 patients with LUAD for whole exome sequencing, mutation signature analysis, HLA binding prediction and neoantigen screening. Dimeric HLA-A2 molecules were prepared by Bac-to-Bac baculovirus expression system to establish a T cell stimulation system based on HLA-A2-coated artificial antigen-presenting cells for the validation of immunogenic neopeptides. Results Similar mutation pattern with predominant missense mutation and high tumor mutation burden was observed across individuals with lung adenocarcinomas and between non-invasive and invasive foci. We screened and identified 3 consistent mutated genes among 100 top genes with highest mutation scores contributed across 4 patients, and 3 mutated peptides among 30 with highest HLA-A2 binding affinity distributed in at least 2 out of 4 foci in the same patient. Notably, LUAD-7-MT peptide encoded by NANOGNB demonstrated higher immunogenicity in promoting CD8+ T cells proliferation and IFN-γ secretion than the corresponding wildtype peptide. Conclusions This study provides an in-depth analysis of mutation characteristics of LUAD and establishes a neoantigen screening and validation system for identifying immunogenicity neopeptide across individual patients and diverse foci in the same patient with multifocal LUAD.
Collapse
Affiliation(s)
- Xin Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lang Jiang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Zhao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mi Wu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Xiong
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiongwen Wu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiufang Weng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Shi G, Xu Y, Qiu H, Cao F, Xiao ZX, Zhang C, Zha GF. Personalized membrane protein vaccine based on a lipid nanoparticle delivery system prevents postoperative recurrence in colorectal cancer models. Acta Biomater 2024:S1742-7061(24)00716-5. [PMID: 39637957 DOI: 10.1016/j.actbio.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
While accessing tumor neoantigens and developing effective delivery systems have posed significant challenges in therapeutic oncology vaccines, this study introduces a cost- and time-efficient personalized tumor vaccine demonstrating potent anti-tumor effects in a mouse xenograft model. This vaccine utilizes a lipid nanoparticle (C5 LNP) system loaded with membrane protein antigens (mAg) derived from surgically excised tumor tissue. Its safety and efficacy were validated in a B16-OVA murine model. C5/OVA exhibited significant uptake by dendritic cells (DCs), leading to cross-presentation, maturation, and subsequent initiation of robust cell-mediated and humoral immune responses. This resulted in clear tumor growth suppression and extended survival in the B16-OVA model. Furthermore, the personalized C5/mAg vaccine effectively inhibited tumor growth in a colorectal carcinoma model. When combined with anti-PD-1 therapy, it notably increased complete remission (CR) rates in the CT26 xenograft model. Vaccinated mice demonstrated 100 % resistance to tumor rechallenge, underscoring the vaccine's ability to induce long-term immune memory. This study presents a promising personalized cancer vaccine delivery system with potential for both treatment and prevention of carcinoma in clinical applications. STATEMENT OF SIGNIFICANCE: In this paper, we present a scheme for manufacturing personalized tumor vaccines. The vaccine component consists of lipid nanoparticles (LNPs) designated C5 and membrane protein antigens (mAg) derived from autologous tumor tissue surgically resected from the patient. Our study demonstrates that the personalized mAg-C5 vaccine for colorectal carcinoma significantly inhibits tumor growth. Furthermore, conjugation with anti-PD-1 therapy demonstrably increased the complete remission (CR) rate in the murine CT26 xenograft tumor model. Additionally, mice treated with the C5/mAg vaccine exhibited 100 % resistance to tumor growth in a colon carcinoma rechallenge model, indicating the induction of immune memory by the vaccine. Our research results suggest that the mAg-LNP vaccine is a simple, cost-effective treatment that clinicians can easily and quickly integrate into routine practice.
Collapse
Affiliation(s)
- Guangzhao Shi
- Department: Digestive Medicine Centre, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China
| | - Yuandong Xu
- Department: Scientific Research Center, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China
| | - Haowei Qiu
- Department: Scientific Research Center, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China
| | - Fei Cao
- Department: Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China
| | - Ze-Xiu Xiao
- Department: Research and development center, Institution: Shenzhen MagicRNA Biotech, No.459, Qiaokai Road, Guangming District, Shenzhen, Guangdong CN518107, PR China
| | - Changhua Zhang
- Department: Digestive Medicine Centre, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China.
| | - Gao-Feng Zha
- Department: Digestive Medicine Centre, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China; Department: Scientific Research Center, Institution: The Seventh Affiliated Hospital Sun Yat-sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, Guangdong CN518107, PR China.
| |
Collapse
|
9
|
Chen S, Cheng S, Cai J, Liu Z, Li H, Wang P, Li Y, Yang F, Chen K, Qiu M. The current therapeutic cancer vaccines landscape in non-small cell lung cancer. Int J Cancer 2024; 155:1909-1927. [PMID: 39109825 DOI: 10.1002/ijc.35088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/12/2024] [Accepted: 05/29/2024] [Indexed: 10/04/2024]
Abstract
Currently, conventional immunotherapies for the treatment of non-small cell lung cancer (NSCLC) have low response rates and benefit only a minority of patients, particularly those with advanced disease, so novel therapeutic strategies are urgent deeded. Therapeutic cancer vaccines, a form of active immunotherapy, harness potential to activate the adaptive immune system against tumor cells via antigen cross-presentation. Cancer vaccines can establish enduring immune memory and guard against recurrences. Vaccine-induced tumor cell death prompts antigen epitope spreading, activating functional T cells and thereby sustaining a cancer-immunity cycle. The success of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rendered cancer vaccines a promising avenue, especially when combined with immunotherapy or chemoradiotherapy for NSCLC. This review delves into the intricate antitumor immune mechanisms underlying therapeutic cancer vaccines, enumerates the tumor antigen spectrum of NSCLC, discusses different cancer vaccines progress and summarizes relevant clinical trials. Additionally, we analyze the combination strategies, current limitations, and future prospects of cancer vaccines in NSCLC treatment, aiming to offer fresh insights for their clinical application in managing NSCLC. Overall, cancer vaccines offer promising potential for NSCLC treatment, particularly combining with chemoradiotherapy or immunotherapy could further improve survival in advanced patients. Exploring inhaled vaccines or prophylactic vaccines represents a crucial research avenue.
Collapse
Affiliation(s)
- Shaoyi Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Sida Cheng
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Jingsheng Cai
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Zheng Liu
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Haoran Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Peiyu Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yun Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Fan Yang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Kezhong Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Mantang Qiu
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
10
|
Hu W, Bian Y, Ji H. TIL Therapy in Lung Cancer: Current Progress and Perspectives. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2409356. [PMID: 39422665 PMCID: PMC11633538 DOI: 10.1002/advs.202409356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/03/2024] [Indexed: 10/19/2024]
Abstract
Lung cancer remains the most prevalent malignant tumor worldwide and is the leading cause of cancer-related mortality. Although immune checkpoint blockade has revolutionized the treatment of advanced lung cancer, many patients still do not respond well, often due to the lack of functional T cell infiltration. Adoptive cell therapy (ACT) using expanded immune cells has emerged as an important therapeutic modality. Tumor-infiltrating lymphocytes (TIL) therapy is one form of ACT involving the administration of expanded and activated autologous T cells derived from surgically resected cancer tissues and reinfusion into patients and holds great therapeutic potential for lung cancer. In this review, TIL therapy is introduced and its suitability for lung cancer is discussed. Then its historical and clinical developments are summarized, and the methods developed up-to-date to identify tumor-recognizing TILs and optimize TIL composition. Some perspectives toward future TIL therapy for lung cancer are also provided.
Collapse
Affiliation(s)
- Weilei Hu
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
- Key Laboratory of Multi‐Cell SystemsShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yifei Bian
- Key Laboratory of Multi‐Cell SystemsShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hongbin Ji
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
- Key Laboratory of Multi‐Cell SystemsShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
- School of Life Science and TechnologyShanghai Tech UniversityShanghai200120China
| |
Collapse
|
11
|
Ren D, Xiong S, Ren Y, Yang X, Zhao X, Jin J, Xu M, Liang T, Guo L, Weng L. Advances in therapeutic cancer vaccines: Harnessing immune adjuvants for enhanced efficacy and future perspectives. Comput Struct Biotechnol J 2024; 23:1833-1843. [PMID: 38707540 PMCID: PMC11066472 DOI: 10.1016/j.csbj.2024.04.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/07/2024] Open
Abstract
Preventive cancer vaccines are highly effective in preventing viral infection-induced cancer, but advances in therapeutic cancer vaccines with a focus on eliminating cancer cells through immunotherapy are limited. To develop therapeutic cancer vaccines, the integration of optimal adjuvants is a potential strategy to enhance or complement existing therapeutic approaches. However, conventional adjuvants do not satisfy the criteria of clinical trials for therapeutic cancer vaccines. To improve the effects of adjuvants in therapeutic cancer vaccines, effective vaccination strategies must be formulated and novel adjuvants must be identified. This review offers an overview of the current advancements in therapeutic cancer vaccines and highlights in situ vaccination approaches that can be synergistically combined with other immunotherapies by harnessing the adjuvant effects. Additionally, the refinement of adjuvant systems using cutting-edge technologies and the elucidation of molecular mechanisms underlying immunogenic cell death to facilitate the development of innovative adjuvants have been discussed.
Collapse
Affiliation(s)
- Dekang Ren
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Shizheng Xiong
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Yujie Ren
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Xueni Yang
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Xinmiao Zhao
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Jiaming Jin
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Miaomiao Xu
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Li Guo
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lixing Weng
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| |
Collapse
|
12
|
Goswami S, Pauken KE, Wang L, Sharma P. Next-generation combination approaches for immune checkpoint therapy. Nat Immunol 2024; 25:2186-2199. [PMID: 39587347 DOI: 10.1038/s41590-024-02015-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/09/2024] [Indexed: 11/27/2024]
Abstract
Immune checkpoint therapy has revolutionized cancer treatment, leading to dramatic clinical outcomes for a subset of patients. However, many patients do not experience durable responses following immune checkpoint therapy owing to multiple resistance mechanisms, highlighting the need for effective combination strategies that target these resistance pathways and improve clinical responses. The development of combination strategies based on an understanding of the complex biology that regulates human antitumor immune responses has been a major challenge. In this Review, we describe the current landscape of combination therapies. We also discuss how the development of effective combination strategies will require the integration of small, tissue-rich clinical trials, to determine how therapy-driven perturbation of the human immune system affects downstream biological responses and eventual clinical outcomes, reverse translation of clinical observations to immunocompetent preclinical models, to interrogate specific biological pathways and their impact on antitumor immune responses, and novel computational methods and machine learning, to integrate multiple datasets across clinical and preclinical studies for the identification of the most relevant pathways that need to be targeted for successful combination strategies.
Collapse
Affiliation(s)
- Sangeeta Goswami
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- James P Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristen E Pauken
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- James P Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute for Data Sciences in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- James P Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
13
|
Li Q, Zeng H, Liu T, Wang P, Zhang R, Zhao B, Feng T, Yang Y, Wu J, Zheng Y, Zhou B, Shu Y, Xu H, Yang L, Ding Z. A dendritic cell vaccine for both vaccination and neoantigen-reactive T cell preparation for cancer immunotherapy in mice. Nat Commun 2024; 15:10419. [PMID: 39613742 DOI: 10.1038/s41467-024-54650-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 11/15/2024] [Indexed: 12/01/2024] Open
Abstract
Adoptive cell transfer (ACT) using neoantigen-specific T cells is an effective immunotherapeutic strategy. However, the difficult isolation of neoantigen-specific T cells limits the clinical application of ACT. Here, we propose a method to prepare neoantigen-reactive T cells (NRT) for ACT following immunization with a tumor lysate-loaded dendritic cell (DC) vaccine. We show that the DC vaccine not only induces a neoantigen-reactive immune response in lung cancer-bearing mice in vivo, but also facilitate NRT cell preparation in vitro. Adoptive transfer of the NRTs as combinatorial therapy into DC vaccine-immunized, LL/2 tumor-bearing mice allows infiltration of the infused NRTs, as well as the enrichment of neoantigen reactive, non-ACT/NRT T cells into the tumor microenvironment with the function of these neoantigen-reactive T-cell receptors validated in vitro. In summary, we propose a method for preparing NRTs that increases ACT efficacy and paves the way to the design of personalized immunotherapies.
Collapse
Affiliation(s)
- Qing Li
- Department of Biotherapy, Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Peipei Wang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Binyan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tang Feng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuling Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiumei Wu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Zheng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Bailing Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Shu
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Heng Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Zhenyu Ding
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
14
|
Chang Y, Wu L. CapHLA: a comprehensive tool to predict peptide presentation and binding to HLA class I and class II. Brief Bioinform 2024; 26:bbae595. [PMID: 39688477 DOI: 10.1093/bib/bbae595] [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: 06/29/2024] [Revised: 09/13/2024] [Accepted: 12/14/2024] [Indexed: 12/18/2024] Open
Abstract
Human leukocyte antigen class I (HLA-I) and class II (HLA-II) proteins play an essential role in epitope binding and presentation to initiate an immune response. Accurate prediction of peptide-HLA (pHLA) binding and presentation is critical for developing effective immunotherapies. However, current tools can predict antigens exclusively for pHLA-I or pHLA-II, but not both; have constraints on peptide length; and commonly show unsatisfactory predictive accuracy. Here, we developed a convolution and attention-based model, CapHLA, trained with eluted ligand and binding affinity mass spectrometry data, to predict peptide presentation probability (PB) and binding affinities (BA) for HLA-I and HLA-II. In comparison with 11 other methods, CapHLA consistently showed improved performance in predicting pHLA BA and PB, particularly in HLA-II and non-classical peptide length datasets. Using CapHLA PB and BA predictions in combination with antigen expression level (EP) from transcriptomic data, we developed a neoantigen quality model for predicting immunotherapy response. In analyses of clinical response among 276 cancer patients given immunotherapy and overall survival in 7228 cancer patients, our neoantigen quality model outperformed other genetics-based models in predicting response to checkpoint inhibitors and patient prognosis. This study provides a versatile neoantigen screening tool, illustrating the prognostic value of neoantigen quality.
Collapse
Affiliation(s)
- Yunjian Chang
- Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Ligang Wu
- Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| |
Collapse
|
15
|
Rosenberg-Mogilevsky A, Siegfried Z, Karni R. Generation of tumor neoantigens by RNA splicing perturbation. Trends Cancer 2024:S2405-8033(24)00227-9. [PMID: 39578174 DOI: 10.1016/j.trecan.2024.10.008] [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: 07/19/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
Immunotherapy has revolutionized cancer treatment, but the limited availability of tumor-specific neoantigens still remains a challenge. The potential of alternative mRNA splicing-derived neoantigens as a source of new immunotherapy targets has gained significant attention. Tumors exhibit unique splicing changes and splicing factor mutations which are prevalent in various cancers and play a crucial role in neoantigen production. We present advances in splicing modulation approaches, including small-molecule drugs, decoy and splice-switching antisense oligonucleotides (SSOs), CRISPR, small interfering RNAs (siRNAs), and nonsense-mediated RNA decay (NMD) inhibition, that can be adapted to enhance antitumor immune responses. Finally, we explore the clinical implications of these approaches, highlighting their potential to transform cancer immunotherapy and broaden its efficacy.
Collapse
Affiliation(s)
- Adi Rosenberg-Mogilevsky
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University and Hadassah Medical School, Jerusalem, Israel
| | - Zahava Siegfried
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University and Hadassah Medical School, Jerusalem, Israel
| | - Rotem Karni
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University and Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
16
|
Zhang G, Huang X, Liang T. mRNA vaccines for gastrointestinal malignancies: cutting-edge advances and future perspectives. Sci Bull (Beijing) 2024:S2095-9273(24)00825-9. [PMID: 39581833 DOI: 10.1016/j.scib.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Affiliation(s)
- Gang Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou 310003, China
| | - Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou 310003, China.
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou 310003, China.
| |
Collapse
|
17
|
Yi X, Zhao H, Hu S, Dong L, Dou Y, Li J, Gao Q, Zhang B. Tumor-associated antigen prediction using a single-sample gene expression state inference algorithm. CELL REPORTS METHODS 2024; 4:100906. [PMID: 39561714 DOI: 10.1016/j.crmeth.2024.100906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/13/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024]
Abstract
We developed a Bayesian-based algorithm to infer gene expression states in individual samples and incorporated it into a workflow to identify tumor-associated antigens (TAAs) across 33 cancer types using RNA sequencing (RNA-seq) data from the Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA). Our analysis identified 212 candidate TAAs, with 78 validated in independent RNA-seq datasets spanning seven cancer types. Eighteen of these TAAs were further corroborated by proteomics data, including 10 linked to liver cancer. We predicted that 38 peptides derived from these 10 TAAs would bind strongly to HLA-A02, the most common HLA allele. Experimental validation confirmed significant binding affinity and immunogenicity for 21 of these peptides. Notably, approximately 64% of liver tumors expressed one or more TAAs associated with these 21 peptides, positioning them as promising candidates for liver cancer therapies, such as peptide vaccines or T cell receptor (TCR)-T cell treatments. This study highlights the power of integrating computational and experimental approaches to discover TAAs for immunotherapy.
Collapse
Affiliation(s)
- Xinpei Yi
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hongwei Zhao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of China, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Shunjie Hu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of China, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Liangqing Dong
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of China, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yongchao Dou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jing Li
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of China, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
18
|
Yang Y, Zheng P, Duan B, Yang Y, Zheng X, Li W, Liu Q, Hu Y, Ma Y. A personalized vaccine combining immunogenic cell death-induced cells and nanosized antigens for enhanced antitumor immunity. J Control Release 2024; 376:1271-1287. [PMID: 39515613 DOI: 10.1016/j.jconrel.2024.10.060] [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: 07/04/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
The tumor vaccine aims to activate the immune system, promote antitumor cellular responses, and restore immune recognition and clearance of tumor cells. However, the low immunogenicity and heterogeneity of tumor antigens, along with immunosuppressive mechanisms, severely hinder tumor vaccines from achieving an efficient and sustained antitumor effect. Herein, we developed a combined vaccine strategy that utilizes immunogenic cell death (ICD) to elicit a broad spectrum of antigen-specific responses in a whole-cell-based manner. Additionally, we introduced nanosized antigens to intensify immune responses targeting a key tumor antigen. The combination of mitoxantrone (MTX) and curcumin (Cur) optimized ICD properties in TC-1 tumor cells, as evidenced by increased release of "find me" signals, such as HMGB1 and ATP, and enhanced exposure of the "eat me" signal, CALR, compared to either MTX or Cur alone. Correspondingly, the ICD cells induced by the combination produced more significant antitumor effects in vivo. Furthermore, the ICD cells in combination with E7-HBcAg VLPs or E7-Q11 nanofibers induced more intense effector cell responses to the antigen included in the nanovaccines, as well as a broad spectrum of antigens provided by tumor cells, and significantly suppressed the growth of established tumors compared with either ICD cells, VLPs, or nanofibers alone. In conclusion, the combination of ICD cells and nanosized antigens produced synergistic antitumor effects and elicited robust and comprehensive antitumor immunity, presenting an attractive strategy for developing personalized tumor vaccines.
Collapse
Affiliation(s)
- Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China; Cell Biology & Molecular Biology Laboratory of Experimental Teaching Center, Faculty of Basic Medical Science, Kunming Medical University, Chunrong West Road, Kunming 650500, China
| | - Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China; Kunming Medical University Graduate School, Chunrong West Road, Kunming 650500, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China
| | - Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China; Kunming Medical University Graduate School, Chunrong West Road, Kunming 650500, China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China; School of Life Sciences, Yunnan University, No.2 Cuihu North Road, Kunming 650091, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, No.935 Jiaoling Road, Kunming 650118, China; State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
19
|
Zhao L, Liu Y, Jin F, Hu K, Lv M, Zhou Y, Zhao W, Hu Y, Wu J, Yang Y, Wang W. Multifunctional nanoparticles potentiate in-situ tumor vaccines via reversing insufficient Photothermal therapy by disrupting tumor vasculature. J Control Release 2024; 376:842-860. [PMID: 39401677 DOI: 10.1016/j.jconrel.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/25/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024]
Abstract
Photothermal therapy can trigger immunogenic cell death and release personalized in-situ tumor vaccine, activating immune responses to eliminate systemic tumors beyond the irradiated zone. However, the immune response of the in-situ tumor vaccines is often undermined by the residual tumor cells and their induced immunosuppressive tumor microenvironment (TME), which is attributed to insufficient photothermal effects stemming from the limited accumulation of photosensitizers. To overcome these limitations, we developed multi-functional nanoparticles (VI@Gd-NPs) that integrate a tumor vasculature-specific disrupting agent (Vadimezan, Phase III clinical drug), a photosensitizer (Indocyanine Green, ICG), and a magnetic resonance imaging contrast agent (Gadolinium, Gd) through chemical self-assembly. By selectively disrupting the tumor vasculature, these nanoparticles enhance the intratumoral delivery of photosensitizers (ICG and blood cells), and Gd. With the guidance of Gd-enhanced MRI, the improved delivery facilitates comprehensive photothermal ablation and regulates the TME, further initiating the in-situ tumor vaccine. Notably, this approach significantly enhances anti-tumor immune responses, improves survival rates, and reduces tumor recurrence and metastasis in various animal models. Moreover, depleting CD8+ T cells reverses these therapeutic benefits, highlighting the critical role of adaptive T cell immunity. Therefore, the VI@Gd-NPs treatment holds great potential for reigniting the in-situ tumor vaccine of photothermal therapy.
Collapse
Affiliation(s)
- Lili Zhao
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yiran Liu
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Fangfei Jin
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Kaiyuan Hu
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Miao Lv
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yuehua Zhou
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Weijun Zhao
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Yong Yang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Wenguang Wang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
20
|
Jonker AH, Tataru EA, Graessner H, Dimmock D, Jaffe A, Baynam G, Davies J, Mitkus S, Iliach O, Horgan R, Augustine EF, Bateman-House A, Pasmooij AMG, Yu T, Synofzik M, Douville J, Lapteva L, Brooks PJ, O'Connor D, Aartsma-Rus A. The state-of-the-art of N-of-1 therapies and the IRDiRC N-of-1 development roadmap. Nat Rev Drug Discov 2024:10.1038/s41573-024-01059-3. [PMID: 39496921 DOI: 10.1038/s41573-024-01059-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 11/06/2024]
Abstract
In recent years, a small number of people with rare diseases caused by unique genetic variants have been treated with therapies developed specifically for them. This pioneering field of genetic N-of-1 therapies is evolving rapidly, giving hope for the individualized treatment of people living with very rare diseases. In this Review, we outline the concept of N-of-1 individualized therapies, focusing on genetic therapies, and illustrate advances and challenges in the field using cases for which therapies have been successfully developed. We discuss why the traditional drug development and reimbursement pathway is not fit for purpose in this field, and outline the pragmatic, regulatory and ethical challenges this poses for future access to N-of-1 therapies. Finally, we provide a roadmap for N-of-1 individualized therapy development.
Collapse
Affiliation(s)
| | - Elena-Alexandra Tataru
- French National Institute of Health and Medical Research (INSERM), Paris, France
- International Rare Diseases Research Consortium (IRDiRC), Paris, France
| | - Holm Graessner
- Centre for Rare Diseases, University Hospital Tübingen, Tübingen, Germany
| | | | - Adam Jaffe
- School of Clinical Medicine, University of New South Wales, Faculty of Medicine, Sydney, New South Wales, Australia
| | - Gareth Baynam
- Rare Care Centre, Perth Children's Hospital, Perth, Western Australia, Australia
- University of Western Australia, Faculty of Health and Medical Sciences, Division of Paediatrics and Telethon Kids Institute, Perth, Western Australia, Australia
- University of Notre Dame, Medical Faculty, Sydney, New South Wales, Australia
| | - James Davies
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- NIHR Blood and Transplant Research Unit in Precision Cellular Therapeutics, University of Oxford, Oxford, UK
| | - Shruti Mitkus
- Patient Services, Global Genes, Aliso Viejo, CA, USA
| | - Oxana Iliach
- Regulatory Strategy and Policy, Certara, Toronto, Ontario, Canada
- Canadian Organization for Rare Disorders (CORD), Toronto, Ontario, Canada
| | | | | | - Alison Bateman-House
- Division of Medical Ethics, Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Anna Maria Gerdina Pasmooij
- Science Department, Dutch Medicines Evaluation Board, Utrecht, the Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Tim Yu
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - Matthis Synofzik
- Research Division Translational Genomics of Neurodegenerative Diseases, Hertie-Institute of Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Division of Translational Genomics of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Julie Douville
- ASO Discovery and Development, n-Lorem Foundation, Carlsbad, CA, USA
| | - Larissa Lapteva
- Division of Clinical Evaluation Pharmacology and Toxicology, Food and Drug Administration, Silver Spring, MD, USA
| | - Philip John Brooks
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Daniel O'Connor
- Regulatory Policy & Early Access, The Association of the British Pharmaceutical Industry (ABPI), London, UK
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
21
|
Liu Q, Wu P, Lei J, Bai P, Zhong P, Yang M, Wei P. Old concepts, new tricks: How peptide vaccines are reshaping cancer immunotherapy? Int J Biol Macromol 2024; 279:135541. [PMID: 39270889 DOI: 10.1016/j.ijbiomac.2024.135541] [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: 07/01/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
Over the past few decades, research on cancer immunotherapy has firmly established immune cells as key players in effective cancer treatment. Peptide vaccines directly targeting immune cells have demonstrated immense potential due to their specificity and applicability. However, developing peptide vaccines to generate tumor-reactive T cells remains challenging, primarily due to suboptimal immunogenicity and overcoming the immunosuppressive tumor microenvironment (TME). In this review, we discuss various elements of effective peptide vaccines, including antigen selection, peptide epitope optimization, vaccine adjuvants, and the combination of multiple immunotherapies, in addition to recent advances in tumor neoantigens as well as epitopes bound by non-classical human leukocyte antigen (HLA) molecules, to increase the understanding of cancer peptide vaccines and provide multiple references for the design of subsequent T cell-based peptide vaccines.
Collapse
Affiliation(s)
- Qingyang Liu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Peihua Wu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Jun Lei
- Hubei Key Laboratory of Cell Homeostasis, State Key Laboratory of Virology, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Laboratory Medicine, Xixi Hospital of Hangzhou, Hangzhou, China
| | - Peng Bai
- In Vivo Pharmacology Unit, WuXi AppTec, Nantong, Jiangsu, China
| | - Peiluan Zhong
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Min Yang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China.
| | - Pengcheng Wei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China.
| |
Collapse
|
22
|
Oli AN, Adejumo SA, Rowaiye AB, Ogidigo JO, Hampton-Marcell J, Ibeanu GC. Tumour Immunotherapy and Applications of Immunological Products: A Review of Literature. J Immunol Res 2024; 2024:8481761. [PMID: 39483536 PMCID: PMC11527548 DOI: 10.1155/2024/8481761] [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: 03/20/2024] [Revised: 09/23/2024] [Accepted: 09/28/2024] [Indexed: 11/03/2024] Open
Abstract
Malignant tumors, characterized by uncontrolled cell proliferation, are a leading global health challenge, responsible for over 9.7 million deaths in 2022, with new cases expected to rise to 35 million annually by 2050. Immunotherapy is preferred to other cancer therapies, offering precise targeting of malignant cells while simultaneously strengthening the immune system's complex responses. Advances in this novel field of science have been closely linked to a deeper knowledge of tumor biology, particularly the intricate interplay between tumor cells, the immune system, and the tumor microenvironment (TME), which are central to cancer progression and immune evasion. This review offers a comprehensive analysis of the molecular mechanisms that govern these interactions, emphasizing their critical role in the development of effective immunotherapeutic products. We critically evaluate the current immunotherapy approaches, including cancer vaccines, adoptive T cell therapies, and cytokine-based treatments, highlighting their efficacy and safety. We also explore the latest advancements in combination therapies, which synergistically integrate multiple immunotherapeutic strategies to overcome resistance and enhance therapeutic outcomes. This review offers key insights into the future of cancer immunotherapy with a focus on advancing more effective and personalized treatment strategies.
Collapse
Affiliation(s)
- Angus Nnamdi Oli
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka 420211, Nigeria
| | - Samson Adedeji Adejumo
- Department of Biological Sciences, University of Illinois, Chicago, 845 West Taylor, Chicago 60607, Illinois, USA
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Federal University Oye Ekiti, Oye, Ekiti State, Nigeria
| | - Adekunle Babajide Rowaiye
- National Biotechnology Development Agency, Abuja 900211, Nigeria
- Department of Pharmaceutical Science, North Carolina Central University, Durham 27707, North Carolina, USA
| | | | - Jarrad Hampton-Marcell
- Department of Biological Sciences, University of Illinois, Chicago, 845 West Taylor, Chicago 60607, Illinois, USA
| | - Gordon C. Ibeanu
- Department of Pharmaceutical Science, North Carolina Central University, Durham 27707, North Carolina, USA
| |
Collapse
|
23
|
Baharom F, Hermans D, Delamarre L, Seder RA. Vax-Innate: improving therapeutic cancer vaccines by modulating T cells and the tumour microenvironment. Nat Rev Immunol 2024:10.1038/s41577-024-01091-9. [PMID: 39433884 DOI: 10.1038/s41577-024-01091-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/23/2024]
Abstract
T cells have a critical role in mediating antitumour immunity. The success of immune checkpoint inhibitors (ICIs) for cancer treatment highlights how enhancing endogenous T cell responses can mediate tumour regression. However, mortality remains high for many cancers, especially in the metastatic setting. Based on advances in the genetic characterization of tumours and identification of tumour-specific antigens, individualized therapeutic cancer vaccines targeting mutated tumour antigens (neoantigens) are being developed to generate tumour-specific T cells for improved therapeutic responses. Early clinical trials using individualized neoantigen vaccines for patients with advanced disease had limited clinical efficacy despite demonstrated induction of T cell responses. Therefore, enhancing T cell activity by improving the magnitude, quality and breadth of T cell responses following vaccination is one current goal for improving outcome against metastatic tumours. Another major consideration is how T cells can be further optimized to function within the tumour microenvironment (TME). In this Perspective, we focus on neoantigen vaccines and propose a new approach, termed Vax-Innate, in which vaccination through intravenous delivery or in combination with tumour-targeting immune modulators may improve antitumour efficacy by simultaneously increasing the magnitude, quality and breadth of T cells while transforming the TME into a largely immunostimulatory environment for T cells.
Collapse
Affiliation(s)
| | - Dalton Hermans
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
| | | | - Robert A Seder
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
24
|
Xu H, Hu R, Dong X, Kuang L, Zhang W, Tu C, Li Z, Zhao Z. ImmuneApp for HLA-I epitope prediction and immunopeptidome analysis. Nat Commun 2024; 15:8926. [PMID: 39414796 PMCID: PMC11484853 DOI: 10.1038/s41467-024-53296-0] [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: 06/27/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
Advances in mass spectrometry accelerates the characterization of HLA ligandome, necessitating the development of efficient methods for immunopeptidomics analysis and (neo)antigen prediction. We develop ImmuneApp, an interpretable deep learning framework trained on extensive HLA ligand datasets, which improves the prediction of HLA-I epitopes, prioritizes neoepitopes, and enhances immunopeptidomics deconvolution. ImmuneApp extracts informative embeddings and identifies key residues for pHLA binding. We also present a more accurate model-based deconvolution approach and systematically analyzed 216 multi-allelic immunopeptidomics samples, identifying 835,551 ligands restricted to over 100 HLA-I alleles. Our investigation reveals the effectiveness of the composite model, denoted as ImmuneApp-MA, which integrates mono- and multi-allelic data to enhance predictive performance. Leveraging ImmuneApp-MA as a pre-trained model, we built ImmuneApp-Neo, an immunogenicity predictor that outperforms existing methods for prioritizing immunogenic neoepitope. ImmuneApp demonstrates its utility across various immunopeptidomics datasets, which will promote the discovery of novel neoantigens and the development of new immunotherapies.
Collapse
Affiliation(s)
- Haodong Xu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Ruifeng Hu
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Center for Advanced Parkinson Research, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Genomics and Bioinformatics Hub, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xianjun Dong
- Center for Advanced Parkinson Research, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Genomics and Bioinformatics Hub, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Lan Kuang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Wenchao Zhang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
25
|
Fan T, Xu C, Wu J, Cai Y, Cao W, Shen H, Zhang M, Zhu H, Yang J, Zhu Z, Ma X, Ren J, Huang L, Li Q, Tang Y, Yu B, Chen C, Xu M, Wang Q, Xu Z, Chen F, Liang S, Zhong Z, Jamroze A, Tang DG, Li H, Dong C. Lipopolyplex-formulated mRNA cancer vaccine elicits strong neoantigen-specific T cell responses and antitumor activity. SCIENCE ADVANCES 2024; 10:eadn9961. [PMID: 39392882 PMCID: PMC11468906 DOI: 10.1126/sciadv.adn9961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 09/06/2024] [Indexed: 10/13/2024]
Abstract
mRNA neoantigen cancer vaccine inducing neoantigen-specific T cell responses holds great promise for cancer immunotherapy; however, its clinical translation remains challenging because of suboptimal neoantigen prediction accuracy and low delivery efficiency, which compromise the in vivo therapeutic efficacy. We present a lipopolyplex (LPP)-formulated mRNA cancer vaccine encoding tandem neoantigens as a cancer therapeutic regimen. The LPP-formulated mRNA vaccines elicited robust neoantigen-specific CD8+ T cell responses in three syngeneic murine tumor models (CT26, MC38, and B16F10) to suppress tumor growth. Prophylactic cancer vaccine treatment completely prevented tumor development, and long-lasting memory T cells protected mice from tumor cell rechallenge. Combining the vaccine with immune checkpoint inhibitor further boosted the antitumor activity. Of note, LPP-based personalized cancer vaccine was administered in two cancer patients and induced meaningful neoantigen-specific T cell and clinical responses. In conclusion, we demonstrated that the LPP-based mRNA vaccine can elicit strong antitumor immune responses, and the results support further clinical evaluation of the therapeutic mRNA cancer vaccine.
Collapse
Affiliation(s)
- Ting Fan
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Congcong Xu
- StemiRNA Therapeutics Inc., Shanghai, China
- Biomedical Polymers Laboratory, College of Chemistry Chemical Engineering and Materials Science and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
- International College of Pharmaceutical Innovation, Soochow University, Suzhou 215123, China
| | - Jichuan Wu
- StemiRNA Therapeutics Inc., Shanghai, China
- Biomedical Polymers Laboratory, College of Chemistry Chemical Engineering and Materials Science and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yihua Cai
- StemiRNA Therapeutics Inc., Shanghai, China
| | - Wanlu Cao
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Haifa Shen
- StemiRNA Therapeutics Inc., Shanghai, China
| | - Mingna Zhang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Hanfei Zhu
- StemiRNA Therapeutics Inc., Shanghai, China
| | - Jingxian Yang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Zhounan Zhu
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xiaopin Ma
- StemiRNA Therapeutics Inc., Shanghai, China
| | - Jiale Ren
- StemiRNA Therapeutics Inc., Shanghai, China
| | - Lei Huang
- StemiRNA Therapeutics Inc., Shanghai, China
| | - Qianyun Li
- StemiRNA Therapeutics Inc., Shanghai, China
| | | | - Bo Yu
- StemiRNA Therapeutics Inc., Shanghai, China
| | | | | | - Qiuhe Wang
- StemiRNA Therapeutics Inc., Shanghai, China
| | - Zhuya Xu
- StemiRNA Therapeutics Inc., Shanghai, China
| | | | - Shujing Liang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Zhixian Zhong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Anmbreen Jamroze
- Department of Pharmacology & Therapeutics and Experimental Therapeutics (ET) Graduate Program, University at Buffalo and Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Dean G. Tang
- Department of Pharmacology & Therapeutics and Experimental Therapeutics (ET) Graduate Program, University at Buffalo and Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hangwen Li
- StemiRNA Therapeutics Inc., Shanghai, China
| | - Chunyan Dong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
26
|
Yang R, Cui J. Advances and applications of RNA vaccines in tumor treatment. Mol Cancer 2024; 23:226. [PMID: 39385255 PMCID: PMC11463124 DOI: 10.1186/s12943-024-02141-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
Compared to other types of tumor vaccines, RNA vaccines have emerged as promising alternatives to conventional vaccine therapy due to their high efficiency, rapid development capability, and potential for low-cost manufacturing and safe drug delivery. RNA vaccines mainly include mRNA, circular RNA (circRNA), and Self-amplifying mRNA(SAM). Different RNA vaccine platforms for different tumors have shown encouraging results in animal and human models. This review comprehensively describes the advances and applications of RNA vaccines in antitumor therapy. Future directions for extending this promising vaccine platform to a wide range of therapeutic uses are also discussed.
Collapse
Affiliation(s)
- Ruohan Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
27
|
Chi WY, Hu Y, Huang HC, Kuo HH, Lin SH, Kuo CTJ, Tao J, Fan D, Huang YM, Wu AA, Hung CF, Wu TC. Molecular targets and strategies in the development of nucleic acid cancer vaccines: from shared to personalized antigens. J Biomed Sci 2024; 31:94. [PMID: 39379923 PMCID: PMC11463125 DOI: 10.1186/s12929-024-01082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/01/2024] [Indexed: 10/10/2024] Open
Abstract
Recent breakthroughs in cancer immunotherapies have emphasized the importance of harnessing the immune system for treating cancer. Vaccines, which have traditionally been used to promote protective immunity against pathogens, are now being explored as a method to target cancer neoantigens. Over the past few years, extensive preclinical research and more than a hundred clinical trials have been dedicated to investigating various approaches to neoantigen discovery and vaccine formulations, encouraging development of personalized medicine. Nucleic acids (DNA and mRNA) have become particularly promising platform for the development of these cancer immunotherapies. This shift towards nucleic acid-based personalized vaccines has been facilitated by advancements in molecular techniques for identifying neoantigens, antigen prediction methodologies, and the development of new vaccine platforms. Generating these personalized vaccines involves a comprehensive pipeline that includes sequencing of patient tumor samples, data analysis for antigen prediction, and tailored vaccine manufacturing. In this review, we will discuss the various shared and personalized antigens used for cancer vaccine development and introduce strategies for identifying neoantigens through the characterization of gene mutation, transcription, translation and post translational modifications associated with oncogenesis. In addition, we will focus on the most up-to-date nucleic acid vaccine platforms, discuss the limitations of cancer vaccines as well as provide potential solutions, and raise key clinical and technical considerations in vaccine development.
Collapse
Affiliation(s)
- Wei-Yu Chi
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Yingying Hu
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hsin-Che Huang
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hui-Hsuan Kuo
- Pharmacology PhD Program, Weill Cornell Medicine, New York, NY, USA
| | - Shu-Hong Lin
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas Graduate School of Biomedical Sciences at Houston and MD Anderson Cancer Center, Houston, TX, USA
| | - Chun-Tien Jimmy Kuo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Julia Tao
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Darrell Fan
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Yi-Min Huang
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Annie A Wu
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - T-C Wu
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Obstetrics and Gynecology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
28
|
Qu L, Cui G, Sun Y, Ye R, Sun Y, Meng F, Wang S, Zhong Z. A Biomimetic Autophagosomes-Based Nanovaccine Boosts Anticancer Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409590. [PMID: 39194369 DOI: 10.1002/adma.202409590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/01/2024] [Indexed: 08/29/2024]
Abstract
Personalized cancer vaccines based on tumor cell lysates offer promise for cancer immunotherapy yet fail to elicit a robust therapeutic effect due to the weak immunogenicity of tumor antigens. Autophagosomes, obtained from pleural effusions and ascites of cancer patients, have been identified as abundant reservoirs of tumor neoantigens that exhibit heightened immunogenicity. However, their potential as personalized cancer vaccines have been constrained by suboptimal lymphatic-targeting performances and challenges in antigen-presenting cell endocytosis. Here,a reinforced biomimetic autophagosome-based (BAPs) nanovaccine generated by precisely amalgamating autophagosome-derived neoantigens and two types of adjuvants capable of targeting lymph nodes is developed to potently elicit antitumor immunity. The redox-responsive BAPs facilitate cytosolic vaccine opening within antigen-presenting cells, thereby exposing adjuvants and antigens to stimulate a strong immune response. BAPs evoke broad-spectrum T-cell responses, culminating in the effective eradication of 71.4% of established tumors. Notably, BAPs vaccination triggers enduring T-cell responses that confer robust protection, with 100% of mice shielded against tumor rechallenge and a significant reduction in tumor incidence by 87.5%. Furthermore, BAPs synergize with checkpoint blockade therapy to inhibit tumor growth in the poorly immunogenic breast cancer model. The biomimetic approach presents a powerful nanovaccine formula with high versatility for personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Liping Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Guanhong Cui
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yinping Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Ruonan Ye
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yu Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Shenqiang Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
29
|
Cui C, Ott PA, Wu CJ. Advances in Vaccines for Melanoma. Hematol Oncol Clin North Am 2024; 38:1045-1060. [PMID: 39079791 PMCID: PMC11524149 DOI: 10.1016/j.hoc.2024.05.009] [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] [Indexed: 09/03/2024]
Abstract
Personalized neoantigen vaccines have achieved major advancements in recent years, with studies in melanoma leading progress in the field. Early clinical trials have demonstrated their feasibility, safety, immunogenicity, and potential efficacy. Advances in sequencing technologies and neoantigen prediction algorithms have substantively improved the identification and prioritization of neoantigens. Innovative delivery platforms now support the rapid and flexible production of vaccines. Several ongoing efforts in the field are aimed at improving the integration of large datasets, refining the training of prediction models, and ensuring the functional validation of vaccine immunogenicity.
Collapse
Affiliation(s)
- Can Cui
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Patrick A Ott
- Harvard Medical School, Boston, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine J Wu
- Harvard Medical School, Boston, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
30
|
Gonzalez-Melero L, Santos-Vizcaino E, Varela-Calvino R, Gomez-Tourino I, Asumendi A, Boyano MD, Igartua M, Hernandez RM. PLGA-PEI nanoparticle covered with poly(I:C) for personalised cancer immunotherapy. Drug Deliv Transl Res 2024; 14:2788-2803. [PMID: 38427275 PMCID: PMC11525302 DOI: 10.1007/s13346-024-01557-2] [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] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
Melanoma is the main cause of death among skin cancers and its incidence worldwide has been experiencing an appalling increase. However, traditional treatments lack effectiveness in advanced or metastatic patients. Immunotherapy, meanwhile, has been shown to be an effective treatment option, but the rate of cancers responding remains far from ideal. Here we have developed a personalized neoantigen peptide-based cancer vaccine by encapsulating patient derived melanoma neoantigens in polyethylenimine (PEI)-functionalised poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) and coating them with polyinosinic:polycytidylic acid (poly(I:C)). We found that PLGA NPs can be effectively modified to be coated with the immunoadjuvant poly(I:C), as well as to encapsulate neoantigens. In addition, we found that both dendritic cells (DCs) and lymphocytes were effectively stimulated. Moreover, the developed NP was found to have a better immune activation profile than NP without poly(I:C) or without antigen. Our results demonstrate that the developed vaccine has a high capacity to activate the immune system, efficiently maturing DCs to present the antigen of choice and promoting the activity of lymphocytes to exert their cytotoxic function. Therefore, the immune response generated is optimal and specific for the elimination of melanoma tumour cells.
Collapse
Affiliation(s)
- Lorena Gonzalez-Melero
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain
| | - Ruben Varela-Calvino
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Santiago de Compostela, Santiago, Spain
| | - Iria Gomez-Tourino
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Santiago, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
| | - Aintzane Asumendi
- Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Maria Dolores Boyano
- Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
31
|
Zhu S, Liu C, Jin Y, Zhang H, Zhou M, Xu C, Shao J, Liu Q, Wei J, Shen J, Liu B. An Advanced Intrahepatic Cholangiocarcinoma Patient Benefits from Personalized Immunotherapy. Inflammation 2024; 47:1699-1705. [PMID: 38492185 DOI: 10.1007/s10753-024-02003-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/15/2024] [Accepted: 03/04/2024] [Indexed: 03/18/2024]
Abstract
Advanced intrahepatic cholangiocarcinoma (ICC) is a highly aggressive malignancy characterized by limited response to standard therapeutic modalities, such as radiotherapy, chemotherapy, and targeted therapy. The prognosis for patients with advanced ICC is exceedingly bleak, with an overall survival of less than 1 year. In recent years, personalized neoantigen vaccines have emerged as a promising approach to augment the immune response against tumors. Clinical investigations are currently underway to evaluate the efficacy of neoantigen-based peptide, DNA, and dendritic cell vaccines. Herein, we present a noteworthy case of advanced ICC patients who experienced disease progression following relapse and subsequently received immunotherapy with a personalized neoantigen nanovaccine. This innovative treatment strategy involved the administration of a custom-designed neoantigen-based peptide nanovaccine tailored to the patient's specific gene mutation profile subsequent to failure of first-line therapy. The clinical efficacy and anti-tumor immune responses were evaluated using various methods, including imaging, interferon-γ ELISPOT assay, and intracellular cytokine staining. Notably, the neoantigen nanovaccine elicited a robust and specific tumor-killing effect mediated by T cells, resulting in a durable response lasting up to 25 months. These findings highlight the potential of neoantigen-based immunotherapy as a novel therapeutic avenue for the management of advanced ICC.
Collapse
Affiliation(s)
- Sihui Zhu
- Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
- Comprehensive Cancer Centre of Nanjing International Hospital, Medical School of Nanjing University, Nanjing, China
- Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
| | - Chenxi Liu
- Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
- Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
| | - Yunchen Jin
- Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
- Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
| | - Hailong Zhang
- Department of Radiology of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Mingzhen Zhou
- Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
- Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
| | - Chen Xu
- Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
- Comprehensive Cancer Centre of Nanjing International Hospital, Medical School of Nanjing University, Nanjing, China
- Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
| | - Jie Shao
- Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
- Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
| | - Qin Liu
- Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
- Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
| | - Jia Wei
- Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
- Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China
| | - Jie Shen
- Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China.
- Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China.
| | - Baorui Liu
- Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China.
- Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Jiangsu Province, 210008, Nanjing, China.
| |
Collapse
|
32
|
Tapescu I, Madsen PJ, Lowenstein PR, Castro MG, Bagley SJ, Fan Y, Brem S. The transformative potential of mRNA vaccines for glioblastoma and human cancer: technological advances and translation to clinical trials. Front Oncol 2024; 14:1454370. [PMID: 39399167 PMCID: PMC11466887 DOI: 10.3389/fonc.2024.1454370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Originally devised for cancer control, mRNA vaccines have risen to the forefront of medicine as effective instruments for control of infectious disease, notably their pivotal role in combating the COVID-19 pandemic. This review focuses on fundamental aspects of the development of mRNA vaccines, e.g., tumor antigens, vector design, and precise delivery methodologies, - highlighting key technological advances. The recent, promising success of personalized mRNA vaccines against pancreatic cancer and melanoma illustrates the potential value for other intractable, immunologically resistant, solid tumors, such as glioblastoma, as well as the potential for synergies with a combinatorial, immunotherapeutic approach. The impact and progress in human cancer, including pancreatic cancer, head and neck cancer, bladder cancer are reviewed, as are lessons learned from first-in-human CAR-T cell, DNA and dendritic cell vaccines targeting glioblastoma. Going forward, a roadmap is provided for the transformative potential of mRNA vaccines to advance cancer immunotherapy, with a particular focus on the opportunities and challenges of glioblastoma. The current landscape of glioblastoma immunotherapy and gene therapy is reviewed with an eye to combinatorial approaches harnessing RNA science. Preliminary preclinical and clinical data supports the concept that mRNA vaccines could be a viable, novel approach to prolong survival in patients with glioblastoma.
Collapse
Affiliation(s)
- Iulia Tapescu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Peter J. Madsen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Pedro R. Lowenstein
- Department of Neurosurgery, The University of Michigan, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, The University of Michigan, Ann Arbor, MI, United States
| | - Maria G. Castro
- Department of Neurosurgery, The University of Michigan, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, United States
| | - Stephen J. Bagley
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Yi Fan
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Steven Brem
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, United States
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
33
|
Wu DW, Jia SP, Xing SJ, Ma HL, Wang X, Tang QY, Li ZW, Wu Q, Bai M, Zhang XY, Fu XF, Jia MM, Tang Y, Chen L, Li N. Personalized neoantigen cancer vaccines: current progression, challenges and a bright future. Clin Exp Med 2024; 24:229. [PMID: 39325256 PMCID: PMC11427492 DOI: 10.1007/s10238-024-01436-7] [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: 05/08/2024] [Accepted: 07/12/2024] [Indexed: 09/27/2024]
Abstract
Tumor neoantigens possess specific immunogenicity and personalized therapeutic vaccines based on neoantigens which have shown promising results in some clinical trials, with broad application prospects. However, the field is developing rapidly and there are currently few relevant review articles. Summarizing and analyzing the status of global personalized neoantigen vaccine clinical trials will provide important data for all stakeholders in drug development. Based on the Trialtrove database, a retrospective analysis was conducted using trial quantity as a key indicator for neo-adjuvant and adjuvant therapy anti-PD-1/PD-L1 clinical trials initiated before the end of 2022. The time trend of newly initiated trials was investigated. The sponsor type, host country, treatment mode, combination strategy, tested drugs, and targeted cancer types of these trials were summarized. As of December 2022, a total of 199 trials were included in the analysis. Among these studies, Phase I studies were the most numerous (119, 59.8%), and Phase I studies have been the predominant study type since 2015. Peptide vaccines were the largest neoantigen vaccines type, accounting for 64.8% of all clinical trials. Based on peptide delivery platforms, the proportion of trials was highest for the DC system (32, 16.1%), followed by LNP (11, 5.5%), LPX (11, 5.5%), and viruses (7, 3.5%). Most vaccines were applied in trials as a monotherapy (133/199, 66.8%), meanwhile combining immunotherapeutic drugs was the most common form for combination therapy. In terms of indications, the largest number of trials involved three or more unspecified solid tumors (50/199, 25.1%), followed by non-small cell lung cancer (24/199, 12.1%) and pancreatic cancer (15/199, 7.5%). The clinical development of personalized neoantigen cancer vaccines is still in the early stage. A clear shift in delivery systems from peptides to DC and liposomal platforms, with the largest number of studies in Asia, collectively marks a new era in the field. The adjuvant or maintenance therapy, and the combination treatment with ICIs are becoming the important clinical development orientation. As research on tumor-immune interactions intensifies, the design, development, and application of neoantigen vaccines are bound to develop rapidly, which will bring a new revolution in the future cancer treatment.
Collapse
Affiliation(s)
- Da-Wei Wu
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuo-Peng Jia
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shu-Jun Xing
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hai-Lan Ma
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xin Wang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Clinical Trials Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Qi-Yu Tang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zi-Wei Li
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Qing Wu
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Min Bai
- Department of Hematology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Xin-Yong Zhang
- Department of Medical Oncology, Beijing Chest Hospital of Capital Medical University, Beijing, 100021, China
| | - Xiao-Feng Fu
- Beijing Likang Life Science, Beijing, 100000, China
| | | | - Yu Tang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Li Chen
- Beijing Likang Life Science, Beijing, 100000, China.
| | - Ning Li
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
34
|
Gupta DS, Gupta DS, Abjani NK, Dave Y, Apte K, Kaur G, Kaur D, Saini AK, Sharma U, Haque S, Tuli HS. Vaccine-based therapeutic interventions in lung cancer management: A recent perspective. Med Oncol 2024; 41:249. [PMID: 39316239 DOI: 10.1007/s12032-024-02489-0] [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: 07/05/2024] [Accepted: 08/24/2024] [Indexed: 09/25/2024]
Abstract
The incidence of lung cancer continues to grow globally, contributing to an ever-increasing load on healthcare systems. Emerging evidence has indicated lowered efficacy of conventional treatment strategies, such as chemotherapy, surgical interventions and radiotherapy, prompting the need for exploring alternative interventions. A growing focus on immunotherapy and the development of personalized medicine has paved the way for vaccine-based delivery in lung cancer. With various prominent targets such as CD8+T cells and PD-L1, immune-targeted, anti-cancer vaccines have been evaluated in both, pre-clinical and clinical settings, to improve therapeutic outcomes. However, there are a number of challenges that must be addressed, including the scalability of such delivery systems, heterogeneity of lung cancers, and long-term safety as well as efficacy. In addition to this, natural compounds, in combination with immunotherapy, have gained considerable research interest in recent times. This makes it necessary to explore their role in synergism with immune-targeted agents. The authors of this review aim to offer an overview of recent advances in our understanding of lung cancer pathogenesis, detection and management strategies, and the emergence of immunotherapy with a special focus on vaccine delivery. This finding is supported with evidence from testing in non-human and human models, showcasing promising results. Prospects for phytotherapy have also been discussed, in order to combat some pitfalls and limitations. Finally, the future perspectives of vaccine usage in lung cancer management have also been discussed, to offer a holistic perspective to readers, and to prompt further research in the domain.
Collapse
Affiliation(s)
- Dhruv Sanjay Gupta
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 56, India
| | - Daksh Sanjay Gupta
- Vivekanand Education Society's College of Pharmacy, Chembur, Mumbai, Maharashtra, 400074, India
| | - Nosheen Kamruddin Abjani
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 56, India
| | - Yash Dave
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 56, India
| | - Ketaki Apte
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 56, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 56, India.
| | - Damandeep Kaur
- University Center for Research & Development (UCRD), Chandigarh University, Gharuan, Mohali, Punjab, 140413, India
| | - Adesh Kumar Saini
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India
| | - Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda, 151001, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India.
| |
Collapse
|
35
|
Ji M, Yu Q, Yang XZ, Yu X, Wang J, Xiao C, An NA, Han C, Li CY, Ding W. Long-range alternative splicing contributes to neoantigen specificity in glioblastoma. Brief Bioinform 2024; 25:bbae503. [PMID: 39401143 PMCID: PMC11472750 DOI: 10.1093/bib/bbae503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/14/2024] [Indexed: 10/17/2024] Open
Abstract
Recent advances in neoantigen research have accelerated the development of immunotherapies for cancers, such as glioblastoma (GBM). Neoantigens resulting from genomic mutations and dysregulated alternative splicing have been studied in GBM. However, these studies have primarily focused on annotated alternatively-spliced transcripts, leaving non-annotated transcripts largely unexplored. Circular ribonucleic acids (circRNAs), abnormally regulated in tumors, are correlated with the presence of non-annotated linear transcripts with exon skipping events. But the extent to which these linear transcripts truly exist and their functions in cancer immunotherapies remain unknown. Here, we found the ubiquitous co-occurrence of circRNA biogenesis and alternative splicing across various tumor types, resulting in large amounts of long-range alternatively-spliced transcripts (LRs). By comparing tumor and healthy tissues, we identified tumor-specific LRs more abundant in GBM than in normal tissues and other tumor types. This may be attributable to the upregulation of the protein quaking in GBM, which is reported to promote circRNA biogenesis. In total, we identified 1057 specific and recurrent LRs in GBM. Through in silico translation prediction and MS-based immunopeptidome analysis, 16 major histocompatibility complex class I-associated peptides were identified as potential immunotherapy targets in GBM. This study revealed long-range alternatively-spliced transcripts specifically upregulated in GBM may serve as recurrent, immunogenic tumor-specific antigens.
Collapse
Affiliation(s)
- Mingjun Ji
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing 100871, China
| | - Qing Yu
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing 100871, China
| | - Xin-Zhuang Yang
- Center for Bioinformatics, National Infrastructures for Translational Medicine, Institute of Clinical Medicine and Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Dongcheng District, Beijing 100730, China
| | - Xianhong Yu
- Academic Department, Shanghai MobiDrop Co., Ltd., Room 308, Building 1, No. 351 Guoshoujing Road, Shanghai Free Trade Pilot Zone, Shanghai 200000, China
| | - Jiaxin Wang
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing 100871, China
| | - Chunfu Xiao
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing 100871, China
| | - Ni A An
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Chuanhui Han
- School of Basic Medical Sciences, Peking University, No. 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Chuan-Yun Li
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing 100871, China
- Chinese Institute for Brain Research, No. 26 Science Park Road, Changping District, Beijing 102206, China
- Southwest United Graduate School, 121 Dajie, Wuhua District, Kunming 650092, China
| | - Wanqiu Ding
- State Key Laboratory of Protein and Plant Gene Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing 100871, China
- Bioinformatics Core Facility, Institute of Molecular Medicine, College of Future Technology, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing 100871, China
| |
Collapse
|
36
|
Hou W, Shen L, Zhu Y, Wang X, Du T, Yang F, Zhu Y. Fullerene Derivatives for Tumor Treatment: Mechanisms and Application. Int J Nanomedicine 2024; 19:9771-9797. [PMID: 39345909 PMCID: PMC11430870 DOI: 10.2147/ijn.s476601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/28/2024] [Indexed: 10/01/2024] Open
Abstract
Fullerenes hold tremendous potential as alternatives to conventional chemotherapy or radiotherapy for tumor treatment due to their abilities to photodynamically kill tumor cells, destroy the tumor vasculature, inhibit tumor metastasis and activate anti-tumor immune responses, while protecting normal tissue through antioxidative effects. The symmetrical hollow molecular structures of fullerenes with abundant C=C bonds allow versatile chemical modification with diverse functional groups, metal clusters and biomacromolecules to synthesize a wide range of fullerene derivatives with increased water solubility, improved biocompatibility, enhanced photodynamic properties and stronger targeting abilities. This review introduces the anti-tumor mechanisms of fullerenes and summarizes the most recent works on the functionalization of fullerenes and the application of fullerene derivatives in tumor treatment. This review aims to serve as a valuable reference for further development and clinical application of anti-tumor fullerene derivatives.
Collapse
Affiliation(s)
- Wenjia Hou
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, People's Republic of China
| | - Lan Shen
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Yimin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Xuanjia Wang
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Tianyu Du
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Fang Yang
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, People's Republic of China
| |
Collapse
|
37
|
Wu Y, Liang X, Sun Y, Ning J, Dai Y, Jin S, Xu Y, Chen S, Pan L. A general pHLA-CD80 scaffold fusion protein to promote efficient antigen-specific T cell-based immunotherapy. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200827. [PMID: 39027379 PMCID: PMC11255371 DOI: 10.1016/j.omton.2024.200827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/23/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024]
Abstract
Inadequate antigen-specific T cells activation hampers immunotherapy due to complex antigen presentation. In addition, therapeutic in vivo T cell expansion is constrained by slow expansion rates and limited functionality. Herein, we introduce a model fusion protein termed antigen-presenting cell-mimic fusion protein (APC-mimic), designed to greatly mimicking the natural antigen presentation pattern of antigen-presenting cells and directly expand T cells both in vitro and in vivo. The APC-mimic comprises the cognate peptide-human leukocyte antigen (pHLA) complex and the co-stimulatory marker CD80, which are natural ligands on APCs. Following a single stimulation, APC-mimic leads to an approximately 400-fold increase in the polyclonal expansion of antigen-specific T cells compared with the untreated group in vitro without the requirement for specialized antigen-presenting cells. Through the combination of single-cell TCR sequencing (scTCR-seq) and single-cell RNA sequencing (scRNA-seq), we identify an approximately 600-fold monoclonal expansion clonotype among these polyclonal clonotypes. It also exhibits suitability for in vivo applications confirmed in the OT-1 mouse model. Furthermore, T cells expanded by APC-mimic effectively inhibits tumor growth in adoptive cell transfer (ACT) murine models. These findings pave the way for the versatile APC-mimic platform for personalized therapeutics, enabling direct expansion of polyfunctional antigen-specific T cell subsets in vitro and in vivo.
Collapse
Affiliation(s)
- Yue Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiao Liang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yanping Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiangtao Ning
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yukun Dai
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shijie Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yingchun Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shuqing Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Precision Medicine on Tumor Therapeutics, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311200, China
| | - Liqiang Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
38
|
Zheng W, Li S, Shi Z, Su K, Ding Y, Zhang L, Tang Q, Han J, Zhao H, Wang F, Zhang H, Hong Z. Recombinant ferritin-based nanoparticles as neoantigen carriers significantly inhibit tumor growth and metastasis. J Nanobiotechnology 2024; 22:562. [PMID: 39272180 PMCID: PMC11401311 DOI: 10.1186/s12951-024-02837-2] [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: 06/28/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Tumor neoantigen peptide-based vaccines, systemic immunotherapies that enhance antitumor immunity by activating and expanding antigen-specific T cells, have achieved remarkable results in the treatment of a variety of solid tumors. However, how to effectively deliver neoantigens to induce robust antitumor immune responses remains a major obstacle. RESULTS Here, we developed a safe and effective neoantigen peptide delivery system (neoantigen-ferritin nanoparticles, neoantigen-FNs) that successfully achieved effective lymph node targeting and induced robust antitumor immune responses. The genetically engineered self-assembled particles neoantigen-FNs with a size of 12 nm were obtained by fusing a neoantigen with optimized ferritin, which rapidly drainage to and continuously accumulate in lymph nodes. The neoantigen-FNs vaccine induced a greater quantity and quality of antigen-specific CD8+ T cells and resulted in significant growth control of multiple tumors, dramatic inhibition of melanoma metastasis and regression of established tumors. In addition, no obvious toxic side effects were detected in the various models, indicating the high safety of optimized ferritin as a vaccine carrier. CONCLUSIONS Homogeneous and safe neoantigen-FNs could be a very promising system for neoantigen peptide delivery because of their ability to efficiently drainage to lymph nodes and induce efficient antitumor immune responses.
Collapse
Affiliation(s)
- Wei Zheng
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Shixiong Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Zhongliang Shi
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Kailing Su
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Yu Ding
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Luyue Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Qian Tang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Jiani Han
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Han Zhao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Fengwei Wang
- School of Medicine, Nankai University, Tianjin, 300071, PR China
- People's Hospital of Tianjin, Tianjin, 300180, PR China
| | - Hongru Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
- Nankai International Advanced Research Institute (SHENZHEN FUTIAN), Shenzhen, 518045, PR China.
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
- Nankai International Advanced Research Institute (SHENZHEN FUTIAN), Shenzhen, 518045, PR China.
| |
Collapse
|
39
|
Mei Y, Liu Y, Liu W, Chen M, Liu X, Wang S, Mou J, Xing H, Tang K, Tian Z, Rao Q, Wang M, Gu R, Qiu S, Wang J. Identifying ADGRG1 as a specific marker for tumor-reactive T cells in acute myeloid leukemia. Exp Hematol Oncol 2024; 13:92. [PMID: 39243082 PMCID: PMC11380426 DOI: 10.1186/s40164-024-00560-0] [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: 06/26/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024] Open
Abstract
Besides chemotherapy and hematopoietic stem cell transplantation (HSCT), autologous T cells can also serve as a new treatment approach for AML patients. However, the features of tumor-reactive T cells and their distinctive markers still lack full description. To evaluate the characteristics of tumor-reactive T cells, we collected bone marrow (BM) T cells from newly diagnosed AML patients with RUNX1::RUNX1T1 as examples for paired single-cell RNA sequencing and single-cell V(D)J sequencing. Based on the STARTRAC-like algorithm, we defined bystander T cells and tumor-reactive T cells. Compared with bystander T cells, tumor-reactive T cells presented as senescent-like cytotoxic terminally differentiated T cells (Temra) with upregulated NK-related markers. Additionally, we found ADGRG1 could serve as the specific marker of CD8+ T tumor-reactive T cell and validated it through the Runx1Runx1t1/+; Mx1-Cre mouse model. In chimeric antigen receptor (CAR)-T and target cell system, ADGRG1 was selectively upregulated upon antigen-TCR encounter. Moreover, ADGRG1+CD8+ T cells released a higher level of IFN-γ and showed higher cell-killing ability when exposed to matched AML blasts. Together, our findings depict the single-cell profile of tumor-reactive T cells in AML BM and propose that ADGRG1 can act as an indicator of T cell tumor reactivity in AML, which may be further harnessed for adoptive cell therapy and tumor-reactive TCR enrichment.
Collapse
Affiliation(s)
- Yihan Mei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yu Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Wenbing Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Manling Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xiaoyu Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Shangshang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Junli Mou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Kejing Tang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Zheng Tian
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Qing Rao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Runxia Gu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Shaowei Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC), Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
40
|
Pavelescu LA, Enache RM, Roşu OA, Profir M, Creţoiu SM, Gaspar BS. Predictive Biomarkers and Resistance Mechanisms of Checkpoint Inhibitors in Malignant Solid Tumors. Int J Mol Sci 2024; 25:9659. [PMID: 39273605 PMCID: PMC11395316 DOI: 10.3390/ijms25179659] [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: 08/22/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Predictive biomarkers for immune checkpoint inhibitors (ICIs) in solid tumors such as melanoma, hepatocellular carcinoma (HCC), colorectal cancer (CRC), non-small cell lung cancer (NSCLC), endometrial carcinoma, renal cell carcinoma (RCC), or urothelial carcinoma (UC) include programmed cell death ligand 1 (PD-L1) expression, tumor mutational burden (TMB), defective deoxyribonucleic acid (DNA) mismatch repair (dMMR), microsatellite instability (MSI), and the tumor microenvironment (TME). Over the past decade, several types of ICIs, including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors, anti-programmed cell death 1 (PD-1) antibodies, anti-programmed cell death ligand 1 (PD-L1) antibodies, and anti-lymphocyte activation gene-3 (LAG-3) antibodies have been studied and approved by the Food and Drug Administration (FDA), with ongoing research on others. Recent studies highlight the critical role of the gut microbiome in influencing a positive therapeutic response to ICIs, emphasizing the importance of modeling factors that can maintain a healthy microbiome. However, resistance mechanisms can emerge, such as increased expression of alternative immune checkpoints, T-cell immunoglobulin (Ig), mucin domain-containing protein 3 (TIM-3), LAG-3, impaired antigen presentation, and alterations in the TME. This review aims to synthesize the data regarding the interactions between microbiota and immunotherapy (IT). Understanding these mechanisms is essential for optimizing ICI therapy and developing effective combination strategies.
Collapse
Affiliation(s)
- Luciana Alexandra Pavelescu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Robert Mihai Enache
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Oana Alexandra Roşu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Monica Profir
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Bogdan Severus Gaspar
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
41
|
Wu T, Hu Y, Tang LV. Gene therapy for polygenic or complex diseases. Biomark Res 2024; 12:99. [PMID: 39232780 PMCID: PMC11375922 DOI: 10.1186/s40364-024-00618-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
Gene therapy utilizes nucleic acid drugs to treat diseases, encompassing gene supplementation, gene replacement, gene silencing, and gene editing. It represents a distinct therapeutic approach from traditional medications and introduces novel strategies for genetic disorders. Over the past two decades, significant advancements have been made in the field of gene therapy, leading to the approval of various gene therapy drugs. Gene therapy was initially employed for treating genetic diseases and cancers, particularly monogenic conditions classified as orphan diseases due to their low prevalence rates; however, polygenic or complex diseases exhibit higher incidence rates within populations. Extensive research on the etiology of polygenic diseases has unveiled new therapeutic targets that offer fresh opportunities for their treatment. Building upon the progress achieved in gene therapy for monogenic diseases and cancers, extending its application to polygenic or complex diseases would enable targeting a broader range of patient populations. This review aims to discuss the strategies of gene therapy, methods of gene editing (mainly CRISPR-CAS9), and carriers utilized in gene therapy, and highlight the applications of gene therapy in polygenic or complex diseases focused on applications that have either entered clinical stages or are currently undergoing clinical trials.
Collapse
Affiliation(s)
- Tingting Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| | - Liang V Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| |
Collapse
|
42
|
Jiang D, Xi B, Tan W, Chen Z, Wei J, Hu M, Lu X, Chen D, Cai H, Du H. NeoaPred: a deep-learning framework for predicting immunogenic neoantigen based on surface and structural features of peptide-human leukocyte antigen complexes. BIOINFORMATICS (OXFORD, ENGLAND) 2024; 40:btae547. [PMID: 39276157 PMCID: PMC11419954 DOI: 10.1093/bioinformatics/btae547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/13/2024] [Accepted: 09/12/2024] [Indexed: 09/16/2024]
Abstract
MOTIVATION Neoantigens, derived from somatic mutations in cancer cells, can elicit anti-tumor immune responses when presented to autologous T cells by human leukocyte antigen. Identifying immunogenic neoantigens is crucial for cancer immunotherapy development. However, the accuracy of current bioinformatic methods remains unsatisfactory. Surface and structural features of peptide-HLA class I (pHLA-I) complexes offer valuable insight into the immunogenicity of neoantigens. RESULTS We present NeoaPred, a deep-learning framework for neoantigen prediction. NeoaPred accurately constructs pHLA-I complex structures, with 82.37% of the predicted structures showing an RMSD of < 1 Å. Using these structures, NeoaPred integrates differences in surface, structural, and atom group features between the mutant peptide and its wild-type counterpart to predict a foreignness score. This foreignness score is an effective factor for neoantigen prediction, achieving an AUROC (Area Under the Receiver Operating Characteristic Curve) of 0.81 and an AUPRC (Area Under the Precision-Recall Curve) of 0.54 in the test set, outperforming existing methods. AVAILABILITY AND IMPLEMENTATION The source code is released under an Apache v2.0 license and is available at the GitHub repository (https://github.com/Dulab2020/NeoaPred).
Collapse
Affiliation(s)
- Dawei Jiang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Binbin Xi
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Wenchong Tan
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Zixi Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jinfen Wei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Meiling Hu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Dong Chen
- Fangrui Institute of Innovative Drugs, South China University of Technology, Guangzhou 510006, China
| | - Hongmin Cai
- School of Computer Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
43
|
Clemen R, Miebach L, Singer D, Freund E, von Woedtke T, Weltmann K, Bekeschus S. Oxidized Melanoma Antigens Promote Activation and Proliferation of Cytotoxic T-Cell Subpopulations. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404131. [PMID: 38958560 PMCID: PMC11434111 DOI: 10.1002/advs.202404131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/30/2024] [Indexed: 07/04/2024]
Abstract
Increasing evidence suggests the role of reactive oxygen and nitrogen species (RONS) in regulating antitumor immune effects and immunosuppression. RONS modify biomolecules and induce oxidative post-translational modifications (oxPTM) on proteins that can alarm phagocytes. However, it is unclear if and how protein oxidation by technical means could be a strategy to foster antitumor immunity and therapy. To this end, cold gas plasma technology producing various RONS simultaneously to oxidize the two melanoma-associated antigens MART and PMEL is utilized. Cold plasma-oxidized MART (oxMART) and PMEL (oxPMEL) are heavily decorated with oxPTMs as determined by mass spectrometry. Immunization with oxidized MART or PMEL vaccines prior to challenge with viable melanoma cells correlated with significant changes in cytokine secretion and altered T-cell differentiation of tumor-infiltrated leukocytes (TILs). oxMART promoted the activity of cytotoxic central memory T-cells, while oxPMEL led to increased proliferation of cytotoxic effector T-cells. Similar T-cell results are observed after incubating splenocytes of tumor-bearing mice with B16F10 melanoma cells. This study, for the first time, provides evidence of the importance of oxidative modifications of two melanoma-associated antigens in eliciting anticancer immunity.
Collapse
Affiliation(s)
- Ramona Clemen
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 217489GreifswaldGermany
| | - Lea Miebach
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 217489GreifswaldGermany
| | - Debora Singer
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 217489GreifswaldGermany
- Department of Dermatology and VenerologyRostock University Medical CenterStrempelstr. 1318057RostockGermany
| | - Eric Freund
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 217489GreifswaldGermany
- Department of NeurosurgeryWien University Medical CenterVienna1090Austria
| | - Thomas von Woedtke
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 217489GreifswaldGermany
- Institute for Hygiene and Environmental MedicineGreifswald University Medical CenterFerdinand‐Sauerbruch‐Str.17475GreifswaldGermany
| | - Klaus‐Dieter Weltmann
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 217489GreifswaldGermany
| | - Sander Bekeschus
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 217489GreifswaldGermany
- Department of Dermatology and VenerologyRostock University Medical CenterStrempelstr. 1318057RostockGermany
| |
Collapse
|
44
|
Salek M, Förster JD, Becker JP, Meyer M, Charoentong P, Lyu Y, Lindner K, Lotsch C, Volkmar M, Momburg F, Poschke I, Fröhling S, Schmitz M, Offringa R, Platten M, Jäger D, Zörnig I, Riemer AB. optiPRM: A Targeted Immunopeptidomics LC-MS Workflow With Ultra-High Sensitivity for the Detection of Mutation-Derived Tumor Neoepitopes From Limited Input Material. Mol Cell Proteomics 2024; 23:100825. [PMID: 39111711 PMCID: PMC11405902 DOI: 10.1016/j.mcpro.2024.100825] [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: 04/26/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 09/08/2024] Open
Abstract
Personalized cancer immunotherapies such as therapeutic vaccines and adoptive transfer of T cell receptor-transgenic T cells rely on the presentation of tumor-specific peptides by human leukocyte antigen class I molecules to cytotoxic T cells. Such neoepitopes can for example arise from somatic mutations and their identification is crucial for the rational design of new therapeutic interventions. Liquid chromatography mass spectrometry (LC-MS)-based immunopeptidomics is the only method to directly prove actual peptide presentation and we have developed a parameter optimization workflow to tune targeted assays for maximum detection sensitivity on a per peptide basis, termed optiPRM. Optimization of collision energy using optiPRM allows for the improved detection of low abundant peptides that are very hard to detect using standard parameters. Applying this to immunopeptidomics, we detected a neoepitope in a patient-derived xenograft from as little as 2.5 × 106 cells input. Application of the workflow on small patient tumor samples allowed for the detection of five mutation-derived neoepitopes in three patients. One neoepitope was confirmed to be recognized by patient T cells. In conclusion, optiPRM, a targeted MS workflow reaching ultra-high sensitivity by per peptide parameter optimization, makes the identification of actionable neoepitopes possible from sample sizes usually available in the clinic.
Collapse
Affiliation(s)
- Mogjiborahman Salek
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Jonas D Förster
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Jonas P Becker
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Marten Meyer
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Department of Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Pornpimol Charoentong
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Heidelberg, Germany
| | - Yanhong Lyu
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Katharina Lindner
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), NCT Heidelberg, A Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Catharina Lotsch
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Michael Volkmar
- T Cell Discovery Platform, Helmholtz Institute for Translational Oncology (HI-TRON) Mainz - A Helmholtz Institute of the DKFZ, Mainz, Germany
| | - Frank Momburg
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Isabel Poschke
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), NCT Heidelberg, A Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Stefan Fröhling
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Division of Translational Medical Oncology, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, A Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany; National Center for Tumor Diseases (NCT), NCT Dresden, A PARTNership between DKFZ, University Hospital Carl Gustav Carus, Faculty of Medicine Carl Gustav Carus of TU Dresden and Helmholtz Center Dresden-Rossendorf, Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, A Partnership Between DKFZ, University Hospital Carl Gustav Carus, Faculty of Medicine Carl Gustav Carus of TU Dresden, Helmholtz Center Dresden-Rossendorf and Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Dresden, Germany
| | - Rienk Offringa
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Platten
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), NCT Heidelberg, A Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience (MCTN), Heidelberg University, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany; Helmholtz Institute for Translational Oncology, Mainz (HI-TRON Mainz) - A Helmholtz Institute of the DKFZ, Mainz, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Angelika B Riemer
- Division of Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany; Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany.
| |
Collapse
|
45
|
Phan T, Fan D, Melstrom LG. Developing Vaccines in Pancreatic Adenocarcinoma: Trials and Tribulations. Curr Oncol 2024; 31:4855-4884. [PMID: 39329989 PMCID: PMC11430674 DOI: 10.3390/curroncol31090361] [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: 06/19/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
Pancreatic adenocarcinoma represents one of the most challenging malignancies to treat, with dismal survival rates despite advances in therapeutic modalities. Immunotherapy, particularly vaccines, has emerged as a promising strategy to harness the body's immune system in combating this aggressive cancer. This abstract reviews the trials and tribulations encountered in the development of vaccines targeting pancreatic adenocarcinoma. Key challenges include the immunosuppressive tumor microenvironment, the heterogeneity of tumor antigens, and a limited understanding of immune evasion mechanisms employed by pancreatic cancer cells. Various vaccine platforms, including peptide-based, dendritic cell-based, and viral vector-based vaccines, have been explored in preclinical and clinical settings. However, translating promising results from preclinical models to clinical efficacy has proven elusive. In recent years, mRNA vaccines have emerged as a promising immunotherapeutic strategy in the fight against various cancers, including pancreatic adenocarcinoma. We will discuss the potential applications, opportunities, and challenges associated with mRNA vaccines in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Thuy Phan
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Darrell Fan
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Laleh G. Melstrom
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA;
| |
Collapse
|
46
|
Cieri N, Hookeri N, Stromhaug K, Li L, Keating J, Díaz-Fernández P, Gómez-García de Soria V, Stevens J, Kfuri-Rubens R, Shao Y, Kooshesh KA, Powell K, Ji H, Hernandez GM, Abelin J, Klaeger S, Forman C, Clauser KR, Sarkizova S, Braun DA, Penter L, Kim HT, Lane WJ, Oliveira G, Kean LS, Li S, Livak KJ, Carr SA, Keskin DB, Muñoz-Calleja C, Ho VT, Ritz J, Soiffer RJ, Neuberg D, Stewart C, Getz G, Wu CJ. Systematic identification of minor histocompatibility antigens predicts outcomes of allogeneic hematopoietic cell transplantation. Nat Biotechnol 2024:10.1038/s41587-024-02348-3. [PMID: 39169264 DOI: 10.1038/s41587-024-02348-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/02/2024] [Indexed: 08/23/2024]
Abstract
T cell alloreactivity against minor histocompatibility antigens (mHAgs)-polymorphic peptides resulting from donor-recipient (D-R) disparity at sites of genetic polymorphisms-is at the core of the therapeutic effect of allogeneic hematopoietic cell transplantation (allo-HCT). Despite the crucial role of mHAgs in graft-versus-leukemia (GvL) and graft-versus-host disease (GvHD) reactions, it remains challenging to consistently link patient-specific mHAg repertoires to clinical outcomes. Here we devise an analytic framework to systematically identify mHAgs, including their detection on HLA class I ligandomes and functional verification of their immunogenicity. The method relies on the integration of polymorphism detection by whole-exome sequencing of germline DNA from D-R pairs with organ-specific transcriptional- and proteome-level expression. Application of this pipeline to 220 HLA-matched allo-HCT D-R pairs demonstrated that total and organ-specific mHAg load could independently predict the occurrence of acute GvHD and chronic pulmonary GvHD, respectively, and defined promising GvL targets, confirmed in a validation cohort of 58 D-R pairs, for the prevention or treatment of post-transplant disease recurrence.
Collapse
Affiliation(s)
- Nicoletta Cieri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nidhi Hookeri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kari Stromhaug
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Liang Li
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Julia Keating
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paula Díaz-Fernández
- Department of Immunology, Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Valle Gómez-García de Soria
- Department of Hematology, Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Jonathan Stevens
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Raphael Kfuri-Rubens
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Yiren Shao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Kaila Powell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Helen Ji
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gabrielle M Hernandez
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Jennifer Abelin
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Susan Klaeger
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Proteomic and Genomic Technologies, Genentech Inc, South San Francisco, CA, USA
| | - Cleo Forman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Karl R Clauser
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Siranush Sarkizova
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - David A Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Haesook T Kim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - William J Lane
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Leslie S Kean
- Harvard Medical School, Boston, MA, USA
- Division Hematology/Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Shuqiang Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth J Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Steven A Carr
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Computer Science, Metropolitan College, Boston University, Boston, MA, USA
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Cecilia Muñoz-Calleja
- Department of Immunology, Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Vincent T Ho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Robert J Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Donna Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Chip Stewart
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Gad Getz
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
47
|
Meng W, Takeuchi Y, Ward JP, Sultan H, Arthur CD, Mardis ER, Artyomov MN, Lichti CF, Schreiber RD. Improvement of Tumor Neoantigen Detection by High-Field Asymmetric Waveform Ion Mobility Mass Spectrometry. Cancer Immunol Res 2024; 12:988-1006. [PMID: 38768391 PMCID: PMC11456315 DOI: 10.1158/2326-6066.cir-23-0900] [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: 10/26/2023] [Revised: 03/05/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
Cancer neoantigens have been shown to elicit cancer-specific T-cell responses and have garnered much attention for their roles in both spontaneous and therapeutically induced antitumor responses. Mass spectrometry (MS) profiling of tumor immunopeptidomes has been used, in part, to identify MHC-bound mutant neoantigen ligands. However, under standard conditions, MS-based detection of such rare but clinically relevant neoantigens is relatively insensitive, requiring 300 million cells or more. Here, to quantitatively define the minimum detectable amounts of therapeutically relevant MHC-I and MHC-II neoantigen peptides, we analyzed different dilutions of immunopeptidomes isolated from the well-characterized T3 mouse methylcholanthrene (MCA)-induced cell line by MS. Using either data-dependent acquisition or parallel reaction monitoring (PRM), we established the minimum amount of material required to detect the major T3 neoantigens in the presence or absence of high field asymmetric waveform ion mobility spectrometry (FAIMS). This analysis yielded a 14-fold enhancement of sensitivity in detecting the major T3 MHC-I neoantigen (mLama4) with FAIMS-PRM compared with PRM without FAIMS, allowing ex vivo detection of this neoantigen from an individual 100 mg T3 tumor. These findings were then extended to two other independent MCA-sarcoma lines (1956 and F244). This study demonstrates that FAIMS substantially increases the sensitivity of MS-based characterization of validated neoantigens from tumors.
Collapse
Affiliation(s)
- Wei Meng
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Yoshiko Takeuchi
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Jeffrey P. Ward
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Hussein Sultan
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Cora D. Arthur
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Elaine R. Mardis
- The Steve and Cindy Rasmussen Institute for Genomic Medicine at Nationwide Children’s Hospital, Columbus, OH 43215, U.S.A
| | - Maxim N. Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Cheryl F. Lichti
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| | - Robert D. Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, U.S.A
| |
Collapse
|
48
|
Gong N, Alameh MG, El-Mayta R, Xue L, Weissman D, Mitchell MJ. Enhancing in situ cancer vaccines using delivery technologies. Nat Rev Drug Discov 2024; 23:607-625. [PMID: 38951662 DOI: 10.1038/s41573-024-00974-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/03/2024]
Abstract
In situ cancer vaccination refers to any approach that exploits tumour antigens available at a tumour site to induce tumour-specific adaptive immune responses. These approaches hold great promise for the treatment of many solid tumours, with numerous candidate drugs under preclinical or clinical evaluation and several products already approved. However, there are challenges in the development of effective in situ cancer vaccines. For example, inadequate release of tumour antigens from tumour cells limits antigen uptake by immune cells; insufficient antigen processing by antigen-presenting cells restricts the generation of antigen-specific T cell responses; and the suppressive immune microenvironment of the tumour leads to exhaustion and death of effector cells. Rationally designed delivery technologies such as lipid nanoparticles, hydrogels, scaffolds and polymeric nanoparticles are uniquely suited to overcome these challenges through the targeted delivery of therapeutics to tumour cells, immune cells or the extracellular matrix. Here, we discuss delivery technologies that have the potential to reduce various clinical barriers for in situ cancer vaccines. We also provide our perspective on this emerging field that lies at the interface of cancer vaccine biology and delivery technologies.
Collapse
Affiliation(s)
- Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, Center for BioAnalytical Chemistry, Hefei National Research Center for Physical Science at the Microscale, University of Science and Technology of China, Hefei, China
| | - Mohamad-Gabriel Alameh
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, George Mason University, Fairfax, VA, USA
| | - Rakan El-Mayta
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
49
|
Desai N, Chavda V, Singh TRR, Thorat ND, Vora LK. Cancer Nanovaccines: Nanomaterials and Clinical Perspectives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401631. [PMID: 38693099 DOI: 10.1002/smll.202401631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/30/2024] [Indexed: 05/03/2024]
Abstract
Cancer nanovaccines represent a promising frontier in cancer immunotherapy, utilizing nanotechnology to augment traditional vaccine efficacy. This review comprehensively examines the current state-of-the-art in cancer nanovaccine development, elucidating innovative strategies and technologies employed in their design. It explores both preclinical and clinical advancements, emphasizing key studies demonstrating their potential to elicit robust anti-tumor immune responses. The study encompasses various facets, including integrating biomaterial-based nanocarriers for antigen delivery, adjuvant selection, and the impact of nanoscale properties on vaccine performance. Detailed insights into the complex interplay between the tumor microenvironment and nanovaccine responses are provided, highlighting challenges and opportunities in optimizing therapeutic outcomes. Additionally, the study presents a thorough analysis of ongoing clinical trials, presenting a snapshot of the current clinical landscape. By curating the latest scientific findings and clinical developments, this study aims to serve as a comprehensive resource for researchers and clinicians engaged in advancing cancer immunotherapy. Integrating nanotechnology into vaccine design holds immense promise for revolutionizing cancer treatment paradigms, and this review provides a timely update on the evolving landscape of cancer nanovaccines.
Collapse
Affiliation(s)
- Nimeet Desai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, 502285, India
| | - Vivek Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, 380009, India
| | | | - Nanasaheb D Thorat
- Limerick Digital Cancer Research Centre (LDCRC), University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
- Department of Physics, Bernal Institute, Castletroy, Limerick, V94T9PX, Ireland
- Nuffield Department of Women's & Reproductive Health, Medical Science Division, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| |
Collapse
|
50
|
Taylor JL, Kokolus KM, Basse PH, Filderman JN, Cosgrove CE, Watkins SC, Gambotto A, Lowe DB, Edwards RP, Kalinski P, Storkus WJ. Therapeutic Anti-Tumor Efficacy of DC-Based Vaccines Targeting TME-Associated Antigens Is Improved When Combined with a Chemokine-Modulating Regimen and/or Anti-PD-L1. Vaccines (Basel) 2024; 12:777. [PMID: 39066414 PMCID: PMC11281486 DOI: 10.3390/vaccines12070777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
We previously reported that dendritic cell (DC)-based vaccines targeting antigens expressed by tumor-associated vascular endothelial cells (VECs) and pericytes effectively control tumor growth in translational mouse tumor models. In the current report, we examined whether the therapeutic benefits of such tumor blood vessel antigen (TBVA)-targeted vaccines could be improved by the cotargeting of tumor antigens in the s.c. B16 melanoma model. We also evaluated whether combination vaccines incorporating anti-PD-L1 checkpoint blockade and/or a chemokine-modulating (CKM; IFNα + TLR3-L [rintatolimod] + Celecoxib) regimen would improve T cell infiltration/functionality in tumors yielding enhanced treatment benefits. We report that DC-peptide or DC-tumor lysate vaccines coordinately targeting melanoma antigens and TBVAs were effective in slowing B16 growth in vivo and extending survival, with superior outcomes observed for DC-peptide-based vaccines. Peptide-based vaccines that selectively target either melanoma antigens or TBVAs elicited a CD8+ T cell repertoire recognizing both tumor cells and tumor-associated VECs and pericytes in vitro, consistent with a treatment-induced epitope spreading mechanism. Notably, combination vaccines including anti-PD-L1 + CKM yielded superior therapeutic effects on tumor growth and animal survival, in association with the potentiation of polyfunctional CD8+ T cell reactivity against both tumor cells and tumor-associated vascular cells and a pro-inflammatory TME.
Collapse
Affiliation(s)
- Jennifer L. Taylor
- Departments of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (J.L.T.); (C.E.C.)
| | - Kathleen M. Kokolus
- Departments of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (K.M.K.); (P.H.B.); (P.K.)
| | - Per H. Basse
- Departments of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (K.M.K.); (P.H.B.); (P.K.)
| | - Jessica N. Filderman
- Departments of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Chloe E. Cosgrove
- Departments of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (J.L.T.); (C.E.C.)
| | - Simon C. Watkins
- Departments of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Andrea Gambotto
- Departments of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Devin B. Lowe
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA;
| | - Robert P. Edwards
- Departments of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Pawel Kalinski
- Departments of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (K.M.K.); (P.H.B.); (P.K.)
- Departments of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
- Departments of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Walter J. Storkus
- Departments of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (J.L.T.); (C.E.C.)
- Departments of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA; (K.M.K.); (P.H.B.); (P.K.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Departments of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Departments of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- W1151 Thomas E. Starzl Biomedical Sciences Tower, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| |
Collapse
|