1
|
Adzavon KP, Zhao W, Khattak SN, Sheng W. Cholesterol-modified peptide nanomicelles as a promising platform for cancer therapy: A review. Int J Biol Macromol 2025; 311:143456. [PMID: 40274168 DOI: 10.1016/j.ijbiomac.2025.143456] [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: 02/21/2025] [Revised: 04/01/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025]
Abstract
Drug resistance, systemic toxicity, low solubility, and rapid clearance are common issues with chemotherapy drugs and other molecules used to treat cancer. The development of new therapeutic compounds and nanotherapy offers a solution to these issues. Therapeutic peptides have attracted great interest among these molecules due to their unique advantages, including low immunogenicity, efficient cellular internalization, deep tissue penetration, and low systemic toxicity. They have shown promise in cancer treatment by inducing apoptosis, necrosis, or cell lysis and promoting immunotherapy. In addition, peptides can deliver a range of cargoes, such as drugs, nucleic acids, imaging agents, and nanoparticles, and can specifically target cancer cells. However, problems such as their short half-life and low solubility limit their therapeutic use. Recent developments have addressed these constraints through structural alterations and nanoparticle formulations. In particular, cholesterol modification makes it possible for peptides to self-assemble into nanomicelles, which enhances their stability, half-life, and cell penetration. In this review, therapeutic peptides are presented as a versatile and successful cancer treatment option. The potential of cholesterol-modified peptide micelles as a reliable drug, nucleic acid, and imaging agent delivery system is also examined.
Collapse
Affiliation(s)
- Kodzo Prosper Adzavon
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Weijian Zhao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Sameena Noor Khattak
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Wang Sheng
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China.
| |
Collapse
|
2
|
Mi Y, Jiang P, Luan J, Feng L, Zhang D, Gao X. Peptide‑based therapeutic strategies for glioma: Current state and prospects. Peptides 2025; 185:171354. [PMID: 39922284 DOI: 10.1016/j.peptides.2025.171354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Glioma is a prevalent form of primary malignant central nervous system tumor, characterized by its cellular invasiveness, rapid growth, and the presence of the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB). Current therapeutic approaches, such as chemotherapy and radiotherapy, have shown limited efficacy in achieving significant antitumor effects. Therefore, there is an urgent demand for new treatments. Therapeutic peptides represent an innovative class of pharmaceutical agents with lower immunogenicity and toxicity. They are easily modifiable via chemical means and possess deep tissue penetration capabilities which reduce side effects and drug resistance. These unique pharmacokinetic characteristics make peptides a rapidly growing class of new therapeutics that have demonstrated significant progress in glioma treatment. This review outlines the efforts and accomplishments in peptide-based therapeutic strategies for glioma. These therapeutic peptides can be classified into four types based on their anti-tumor function: tumor-homing peptides, inhibitor/antagonist peptides targeting cell surface receptors, interference peptides, and peptide vaccines. Furthermore, we briefly summarize the results from clinical trials of therapeutic peptides in glioma, which shows that peptide-based therapeutic strategies exhibit great potential as multifunctional players in glioma therapy.
Collapse
Affiliation(s)
- Yajing Mi
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Pengtao Jiang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Jing Luan
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lin Feng
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Dian Zhang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Xingchun Gao
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China.
| |
Collapse
|
3
|
Duan X, Zou H, Yang J, Liu S, Xu T, Ding J. Melittin-incorporated nanomedicines for enhanced cancer immunotherapy. J Control Release 2024; 375:285-299. [PMID: 39216597 DOI: 10.1016/j.jconrel.2024.08.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Immunotherapy is a rapidly developing and effective strategy for cancer therapy. Among various immunotherapy approaches, peptides have garnered significant attention due to their potent immunomodulatory effects. In particular, melittin emerged as a promising candidate to enhance cancer immunotherapy by inducing immunogenic cell death, promoting the maturation of antigen-presenting cells, activating T cells, enhancing the infiltration and cytotoxicity of effector lymphocytes, and modulating macrophage phenotypes for relieving immunosuppression. However, the clinical application of melittin is limited by poor targeting and systemic toxicity. To overcome these challenges, melittin has been incorporated into biomaterials and related nanotechnologies, resulting in extended circulation time in vivo, improved targeting, reduced adverse effects, and enhanced anti-cancer immunological action. This review provides an in-depth analysis of the immunomodulatory effects of melittin-incorporated nanomedicines and examines their development and challenges for clinical cancer immunotherapy.
Collapse
Affiliation(s)
- Xuefeng Duan
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, PR China
| | - Haoyang Zou
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, PR China
| | - Jiazhen Yang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, PR China.
| | - Shixian Liu
- Department of Orthopedics, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, PR China
| | - Tianmin Xu
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, PR China.
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, PR China.
| |
Collapse
|
4
|
Wang L, Zheng J, Tan Z, Zhang Y, Wang H. A novel bispecific peptide targeting PD-1 and PD-L1 with combined antitumor activity of T-cells derived from the patients with TSCC. Int Immunopharmacol 2024; 138:112582. [PMID: 38981226 DOI: 10.1016/j.intimp.2024.112582] [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: 04/18/2024] [Revised: 06/08/2024] [Accepted: 06/25/2024] [Indexed: 07/11/2024]
Abstract
Programmed death-1 (PD-1) and programmed death ligand-1 (PD-L1) are key immune checkpoints (ICs) that critically influence immunotherapy. Tumor resistance to single IC-targeting drugs has increased interest in dual-target drugs, which have shown feasibility for cancer treatment. In this study, we aimed to develop dual-target peptide drugs targeting the PD-1/PD-L1 pathway and to evaluate their efficacy compared to functional antibodies in enhancing the cytotoxicity of human T cells against tongue squamous carcinoma cell lines. Through sequence analysis and peptide truncation, we modified a pre-existing peptide named nABPD-1 targeting PD-1. Subsequently, we obtained two novel peptides named nABPD-2 and nABPD-3, with nABPD-2 showing an enhanced affinity for human PD-1 protein compared to nABPD-1. Importantly, nABPD-2 exhibited dual-targeting capability, possessing a high affinity for both PD-L1 and PD-1. Furthermore, nABPD-2 effectively promoted the cytotoxicity of human T cells against tongue squamous carcinoma cell lines, surpassing the efficacy of anti-PD-1 or anti-PD-L1 functional antibodies alone. Considering that nABPD-2 has lower production costs and dose requirements, it can potentially be used in therapeutic applications.
Collapse
Affiliation(s)
- Lili Wang
- Department of Oral and Maxillofacial Surgery, Clinical Laboratory, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Junheng Zheng
- Zhuhai Taisujian Biotechnology Co., Ltd, Zhuhai, Guangdong, China; Cheerland Taisujian BioPharm. Co., Ltd, Shenzhen, Guangdong, China
| | - Zhihao Tan
- Cheerland Taisujian BioPharm. Co., Ltd, Shenzhen, Guangdong, China
| | - Yan Zhang
- Laboratory of Cancer and Stem Cell Biology, Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou Higher Education Mega Center, Guangzhou 510006, China
| | - Hua Wang
- Department of Oral and Maxillofacial Surgery, Clinical Laboratory, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Department of Oral and Maxillofacial Surgery, Oral Medical Center, Shenzhen Qianhai Taikang Hospital, Shenzhen, China; Zhuhai Taisujian Biotechnology Co., Ltd, Zhuhai, Guangdong, China; Cheerland Taisujian BioPharm. Co., Ltd, Shenzhen, Guangdong, China.
| |
Collapse
|
5
|
Zhong S(J, Liu X, Kaneko T, Feng Y, Hovey O, Yang K, Ezra S, Ha SD, Kim S, McCormick JK, Liu H, Li SSC. Peptide Blockers of PD-1-PD-L1 Interaction Reinvigorate PD-1-Suppressed T Cells and Curb Tumor Growth in Mice. Cells 2024; 13:1193. [PMID: 39056775 PMCID: PMC11274521 DOI: 10.3390/cells13141193] [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: 05/03/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The programmed cell death protein 1 (PD-1) plays a critical role in cancer immune evasion. Blocking the PD-1-PD-L1 interaction by monoclonal antibodies has shown remarkable clinical efficacy in treating certain types of cancer. However, antibodies are costly to produce, and antibody-based therapies can cause immune-related adverse events. To address the limitations associated with current PD-1/PD-L1 blockade immunotherapy, we aimed to develop peptide-based inhibitors of the PD-1/PD-L1 interaction as an alternative means to PD-1/PD-L1 blockade antibodies for anti-cancer immunotherapy. Through the functional screening of peptide arrays encompassing the ectodomains of PD-1 and PD-L1, followed by the optimization of the hit peptides for solubility and stability, we have identified a 16-mer peptide, named mL7N, with a remarkable efficacy in blocking the PD-1/PD-L1 interaction both in vitro and in vivo. The mL7N peptide effectively rejuvenated PD-1-suppressed T cells in multiple cellular systems designed to recapitulate the PD-1/PD-L1 interaction in the context of T-cell receptor signaling. Furthermore, PA-mL7N, a chimera of the mL7N peptide coupled to albumin-binding palmitic acid (PA), significantly promoted breast cancer cell killing by peripheral blood mononuclear cells ex vivo and significantly curbed tumor growth in a syngeneic mouse model of breast cancer. Our work raises the prospect that mL7N may serve as a prototype for the development of a new line of peptide-based immunomodulators targeting the PD-1/PD-L1 immune checkpoint with potential applications in cancer treatment.
Collapse
Affiliation(s)
- Shanshan (Jenny) Zhong
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (S.Z.); (X.L.); (T.K.); (Y.F.); (O.H.); (K.Y.); (S.E.)
| | - Xiaoling Liu
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (S.Z.); (X.L.); (T.K.); (Y.F.); (O.H.); (K.Y.); (S.E.)
| | - Tomonori Kaneko
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (S.Z.); (X.L.); (T.K.); (Y.F.); (O.H.); (K.Y.); (S.E.)
| | - Yan Feng
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (S.Z.); (X.L.); (T.K.); (Y.F.); (O.H.); (K.Y.); (S.E.)
| | - Owen Hovey
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (S.Z.); (X.L.); (T.K.); (Y.F.); (O.H.); (K.Y.); (S.E.)
| | - Kyle Yang
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (S.Z.); (X.L.); (T.K.); (Y.F.); (O.H.); (K.Y.); (S.E.)
| | - Sally Ezra
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (S.Z.); (X.L.); (T.K.); (Y.F.); (O.H.); (K.Y.); (S.E.)
| | - Soon-Duck Ha
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (S.-D.H.); (S.K.); (J.K.M.)
| | - Sung Kim
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (S.-D.H.); (S.K.); (J.K.M.)
| | - John K. McCormick
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (S.-D.H.); (S.K.); (J.K.M.)
| | - Huadong Liu
- School of Health and Life Sciences, Health and Rehabilitation University, 369 Dengyun Rd, Qingdao 266071, China;
| | - Shawn Shun-Cheng Li
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; (S.Z.); (X.L.); (T.K.); (Y.F.); (O.H.); (K.Y.); (S.E.)
| |
Collapse
|
6
|
Wang Q, Huang H, Liang P, Wang L, Zheng J, Zhang Y, Wang H. Development of PD-1 blockade peptide-cell conjugates to enhance cellular therapies for T-cell acute lymphoblastic leukemia. Med Oncol 2023; 41:14. [PMID: 38078948 DOI: 10.1007/s12032-023-02235-y] [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: 07/22/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023]
Abstract
Blockade of the interaction of the immune checkpoint receptor programmed cell death protein (PD)-1 and its ligand PD-L1 has been found to be a promising cancer treatment. Our previous studies identified that nABPD1 competed with PD-L1 to bind PD-1. The aim of this study was to evaluate the efficacy and safety of anti-tumor immunotherapy of ICIK cells conjugated with peptides in vivo and in vitro. Here, we synthesized the nABPD1 derivatives SBP1 and SBP2 and showed that their binding efficiency to PD-1-positive improving cytokine-induced killer (ICIK) cells was 98 and 82%, respectively. The cytotoxicity of ICIK cells to T-cell acute lymphoblastic leukemia (T-ALL) cells was increased by conjugating with SBP1 or SBP2, which was 2 times higher than that of ICIK cells alone. Furthermore, mice experiments showed that the fluorescence intensity of leukemia cells in T-ALL xenograft models was reduced by more than 95%, indicating that the peptides enhanced the therapeutic effect in vivo, while morphological evaluations showed that the peptides had no toxicity to important organs. Therefore, peptide-cell conjugates (PCCs) may be a novel method to improve the efficacy of cancer immunotherapy by blocking PD-1 in T-ALL patients.
Collapse
Affiliation(s)
- Quanxiao Wang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan West Road, Guangzhou, 510055, Guangdong, China
| | - Hongxing Huang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan West Road, Guangzhou, 510055, Guangdong, China
| | - Peisheng Liang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan West Road, Guangzhou, 510055, Guangdong, China
| | - Lili Wang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan West Road, Guangzhou, 510055, Guangdong, China
| | - Junheng Zheng
- Guangzhou Yidai Pharmaceutical Co., Ltd, Guangzhou, Guangdong, China
- Zhuhai Taisujian Biotechnology Co., Ltd, Zhuhai, Guangdong, China
- Cheerland Taisujian BioPharm. Co., Ltd, Shenzhen, Guangdong, China
| | - Yan Zhang
- Laboratory of Cancer and Stem Cell Biology, Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou Higher Education Mega Center, Guangzhou, China
| | - Hua Wang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan West Road, Guangzhou, 510055, Guangdong, China.
- Guangzhou Yidai Pharmaceutical Co., Ltd, Guangzhou, Guangdong, China.
- Zhuhai Taisujian Biotechnology Co., Ltd, Zhuhai, Guangdong, China.
- Cheerland Taisujian BioPharm. Co., Ltd, Shenzhen, Guangdong, China.
| |
Collapse
|
7
|
Cao H, Wu T, Zhou X, Xie S, Sun H, Sun Y, Li Y. Progress of research on PD-1/PD-L1 in leukemia. Front Immunol 2023; 14:1265299. [PMID: 37822924 PMCID: PMC10562551 DOI: 10.3389/fimmu.2023.1265299] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 09/13/2023] [Indexed: 10/13/2023] Open
Abstract
Leukemia cells prevent immune system from clearing tumor cells by inducing the immunosuppression of the bone marrow (BM) microenvironment. In recent years, further understanding of the BM microenvironment and immune landscape of leukemia has resulted in the introduction of several immunotherapies, including checkpoint inhibitors, T-cell engager, antibody drug conjugates, and cellular therapies in clinical trials. Among them, the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis is a significant checkpoint for controlling immune responses, the PD-1 receptor on tumor-infiltrating T cells is bound by PD-L1 on leukemia cells. Consequently, the activation of tumor reactive T cells is inhibited and their apoptosis is promoted, preventing the rejection of the tumor by immune system and thus resulting in the occurrence of immune tolerance. The PD-1/PD-L1 axis serves as a significant mechanism by which tumor cells evade immune surveillance, and PD-1/PD-L1 checkpoint inhibitors have been approved for the treatment of lymphomas and varieties of solid tumors. However, the development of drugs targeting PD-1/PD-L1 in leukemia remains in the clinical-trial stage. In this review, we tally up the basic research and clinical trials on PD-1/PD-L1 inhibitors in leukemia, as well as discuss the relevant toxicity and impacts of PD-1/PD-L1 on other immunotherapies such as hematopoietic stem cell transplantation, bi-specific T-cell engager, chimeric antigen receptor T-cell immunotherapy.
Collapse
Affiliation(s)
- Huizhen Cao
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Tianyu Wu
- Department of Gastrointestinal Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Xue Zhou
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Shuyang Xie
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Hongfang Sun
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| | - Yunxiao Sun
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, China
| |
Collapse
|
8
|
Vadevoo SMP, Gurung S, Lee HS, Gunassekaran GR, Lee SM, Yoon JW, Lee YK, Lee B. Peptides as multifunctional players in cancer therapy. Exp Mol Med 2023; 55:1099-1109. [PMID: 37258584 PMCID: PMC10318096 DOI: 10.1038/s12276-023-01016-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 06/02/2023] Open
Abstract
Peptides exhibit lower affinity and a shorter half-life in the body than antibodies. Conversely, peptides demonstrate higher efficiency in tissue penetration and cell internalization than antibodies. Regardless of the pros and cons of peptides, they have been used as tumor-homing ligands for delivering carriers (such as nanoparticles, extracellular vesicles, and cells) and cargoes (such as cytotoxic peptides and radioisotopes) to tumors. Additionally, tumor-homing peptides have been conjugated with cargoes such as small-molecule or chemotherapeutic drugs via linkers to synthesize peptide-drug conjugates. In addition, peptides selectively bind to cell surface receptors and proteins, such as immune checkpoints, receptor kinases, and hormone receptors, subsequently blocking their biological activity or serving as hormone analogs. Furthermore, peptides internalized into cells bind to intracellular proteins and interfere with protein-protein interactions. Thus, peptides demonstrate great application potential as multifunctional players in cancer therapy.
Collapse
Affiliation(s)
- Sri Murugan Poongkavithai Vadevoo
- Department of Biochemistry and Cell Biology, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Department of Biomedical Science, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Cell & Matrix Research Institute, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Smriti Gurung
- Department of Biochemistry and Cell Biology, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Department of Biomedical Science, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Cell & Matrix Research Institute, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Hyun-Su Lee
- Department of Physiology, Daegu Catholic University School of Medicine, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu, 42472, Republic of Korea
| | - Gowri Rangaswamy Gunassekaran
- Department of Biochemistry and Cell Biology, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Department of Biomedical Science, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Cell & Matrix Research Institute, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Seok-Min Lee
- Department of Biochemistry and Cell Biology, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Department of Biomedical Science, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Cell & Matrix Research Institute, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Jae-Won Yoon
- Department of Biochemistry and Cell Biology, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Department of Biomedical Science, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Cell & Matrix Research Institute, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Yun-Ki Lee
- Department of Biochemistry and Cell Biology, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Department of Biomedical Science, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Cell & Matrix Research Institute, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea.
- Department of Biomedical Science, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea.
- Cell & Matrix Research Institute, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea.
| |
Collapse
|
9
|
Awad RM, Breckpot K. Novel technologies for applying immune checkpoint blockers. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 382:1-101. [PMID: 38225100 DOI: 10.1016/bs.ircmb.2023.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Cancer cells develop several ways to subdue the immune system among others via upregulation of inhibitory immune checkpoint (ICP) proteins. These ICPs paralyze immune effector cells and thereby enable unfettered tumor growth. Monoclonal antibodies (mAbs) that block ICPs can prevent immune exhaustion. Due to their outstanding effects, mAbs revolutionized the field of cancer immunotherapy. However, current ICP therapy regimens suffer from issues related to systemic administration of mAbs, including the onset of immune related adverse events, poor pharmacokinetics, limited tumor accessibility and immunogenicity. These drawbacks and new insights on spatiality prompted the exploration of novel administration routes for mAbs for instance peritumoral delivery. Moreover, novel ICP drug classes that are adept to novel delivery technologies were developed to circumvent the drawbacks of mAbs. We therefore review the state-of-the-art and novel delivery strategies of ICP drugs.
Collapse
Affiliation(s)
- Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
10
|
Lee WS, Kim DS, Kim JH, Heo Y, Yang H, Go EJ, Kim JH, Lee SJ, Ahn BC, Yum JS, Chon HJ, Kim C. Intratumoral immunotherapy using a TLR2/3 agonist, L-pampo, induces robust antitumor immune responses and enhances immune checkpoint blockade. J Immunother Cancer 2022; 10:jitc-2022-004799. [PMID: 35764365 PMCID: PMC9240943 DOI: 10.1136/jitc-2022-004799] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Toll-like receptors (TLRs) are critical innate immune sensors that elicit antitumor immune responses in cancer immunotherapy. Although a few TLR agonists have been approved for the treatment of patients with early-stage superficial cancers, their therapeutic efficacy is limited in patient with advanced invasive cancers. Here, we identified the therapeutic role of a TLR2/3 agonist, L-pampo (LP), which promotes antitumor immunity and enhances the immune checkpoint blockade. METHODS We generated LP by combining a TLR2 agonist, Pam3CSK4, with a TLR3 agonist, Poly (I:C). Immune responses to stimulation with various TLR agonists were compared. Tumor-bearing mice were intratumorally treated with LP, and their tumor sizes were measured. The antitumor effects of LP treatment were determined using flow cytometry, multiplexed imaging, and NanoString nCounter immune profiling. The immunotherapeutic potential of LP in combination with α-programmed cell death protein-1 (PD-1) or α-cytotoxic T-lymphocytes-associated protein 4 (CTLA-4) was evaluated in syngeneic MC38 colon cancer and B16F10 melanoma. RESULTS The LP treatment induced a potent activation of T helper 1 (Th1) and 2 (Th2)-mediated immunity, tumor cell apoptosis, and immunogenic tumor cell death. Intratumoral LP treatment effectively inhibited tumor progression by activating tumor-specific T cell immunity. LP-induced immune responses were mediated by CD8+ T cells and interferon-γ, but not by CD4+ T cells and CD25+ T cells. LP simultaneously activated TLR2 and TLR3 signaling, thereby extensively changing the immune-related gene signatures within the tumor microenvironment (TME). Moreover, intratumoral LP treatment led to systemic abscopal antitumor effects in non-injected distant tumors. Notably, LP treatment combined with ɑPD-1 and ɑCTLA-4 further enhanced the efficacy of monotherapy, resulting in complete tumor regression and prolonged overall survival. Furthermore, LP-based combination immunotherapy elicited durable antitumor immunity with tumor-specific immune memory in colon cancer and melanoma. CONCLUSIONS Our study demonstrated that intratumoral LP treatment improves the innate and adaptive antitumor immunity within the TME and enhances the efficacy of αPD-1 and αCTLA-4 immune checkpoint blockade.
Collapse
Affiliation(s)
- Won Suk Lee
- Laboratory of Translational Immuno-Oncology, Seongnam, Gyeonggi-do, Korea (the Republic of).,Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Dong Sung Kim
- Laboratory of Translational Immuno-Oncology, Seongnam, Gyeonggi-do, Korea (the Republic of).,Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Jeong Hun Kim
- Laboratory of Translational Immuno-Oncology, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Yoonki Heo
- CHA Vaccine Institute, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Hannah Yang
- Laboratory of Translational Immuno-Oncology, Seongnam, Gyeonggi-do, Korea (the Republic of).,Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Eun-Jin Go
- Laboratory of Translational Immuno-Oncology, Seongnam, Gyeonggi-do, Korea (the Republic of).,Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Jin Hyoung Kim
- Laboratory of Translational Immuno-Oncology, Seongnam, Gyeonggi-do, Korea (the Republic of).,Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Seung Joon Lee
- Laboratory of Translational Immuno-Oncology, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Byung Cheol Ahn
- CHA Vaccine Institute, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Jung Sun Yum
- CHA Vaccine Institute, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Hong Jae Chon
- Laboratory of Translational Immuno-Oncology, Seongnam, Gyeonggi-do, Korea (the Republic of) .,Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of).,Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-do, Korea (the Republic of)
| | - Chan Kim
- Laboratory of Translational Immuno-Oncology, Seongnam, Gyeonggi-do, Korea (the Republic of) .,Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi-do, Korea (the Republic of).,Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-do, Korea (the Republic of)
| |
Collapse
|