1
|
Chen CH. Membrane-active peptides for anticancer therapies. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 212:67-116. [PMID: 40122653 DOI: 10.1016/bs.pmbts.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Membrane-active peptides are found in many living organisms and play a critical role in their immune systems by combating various infectious diseases. These host defense peptides employ multiple mechanisms against different microorganisms and possess unique functions, such as anti-inflammatory and immunomodulatory effects, often working in synergy with other antimicrobial agents. Despite extensive research over the past few decades and the identification of thousands of sequences, only a few have been successfully applied in clinical settings and received approval from the U.S. Food and Drug Administration. In this chapter, we explore all peptide therapeutics that have reached the market, as well as candidates in preclinical and clinical trials, to understand their success and potential applications in cancer therapy. Our findings indicate that at least four membrane-active peptide drugs have progressed to preclinical or clinical phases, dmonstrating promising results for cancer treatment. We summarize our insights in this chapter, highlighting the potential of membrane-active anticancer peptide therapeutics and their applications as targeting ligands in various biomedical fields.
Collapse
Affiliation(s)
- Charles H Chen
- Synthetic Biology Group, Research Laboratory of Electronics, Massachusetts Institute of Technology (MIT), Cambridge, MA, United States.
| |
Collapse
|
2
|
Liu H, Shen W, Liu W, Yang Z, Yin D, Xiao C. From oncolytic peptides to oncolytic polymers: A new paradigm for oncotherapy. Bioact Mater 2024; 31:206-230. [PMID: 37637082 PMCID: PMC10450358 DOI: 10.1016/j.bioactmat.2023.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/18/2023] [Accepted: 08/08/2023] [Indexed: 08/29/2023] Open
Abstract
Traditional cancer therapy methods, especially those directed against specific intracellular targets or signaling pathways, are not powerful enough to overcome tumor heterogeneity and therapeutic resistance. Oncolytic peptides that can induce membrane lysis-mediated cancer cell death and subsequent anticancer immune responses, has provided a new paradigm for cancer therapy. However, the clinical application of oncolytic peptides is always limited by some factors such as unsatisfactory bio-distribution, poor stability, and off-target toxicity. To overcome these limitations, oncolytic polymers stand out as prospective therapeutic materials owing to their high stability, chemical versatility, and scalable production capacity, which has the potential to drive a revolution in cancer treatment. This review provides an overview of the mechanism and structure-activity relationship of oncolytic peptides. Then the oncolytic peptides-mediated combination therapy and the nano-delivery strategies for oncolytic peptides are summarized. Emphatically, the current research progress of oncolytic polymers has been highlighted. Lastly, the challenges and prospects in the development of oncolytic polymers are discussed.
Collapse
Affiliation(s)
- Hanmeng Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, Anhui, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, Anhui, 230012, China
| | - Wanguo Liu
- Department of Orthopaedic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Zexin Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, Anhui, 230012, China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| |
Collapse
|
3
|
Targeting of the Interleukin-13 Receptor (IL-13R)α2 Expressing Prostate Cancer by a Novel Hybrid Lytic Peptide. Biomolecules 2023; 13:biom13020356. [PMID: 36830725 PMCID: PMC9953383 DOI: 10.3390/biom13020356] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/31/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
The IL-13Rα2 cell surface receptor is highly expressed in tumours such as prostate cancer. In this report, we evaluated the hypothesis that prostate cancer cells with enhanced IL-13Rα2 expression are a suitable target for the hybrid lytic peptide (Pep-1-Phor21) peptide, which is generated by fusing the IL-13Rα2 specific ligand (Pep-1) and a cell membrane disrupting lytic peptide (Phor21). The expression of IL-13Rα2 mRNA and protein in prostate cancer tissues and cell lines was assessed via real-time PCR (RT-PCR) and immunoblotting. The effect of Pep-1-Phor21 on the viability of prostate cancer cells grown in monolayers (2D) and microtissue spheroids (3D) was assessed via CellTox green cytotoxic assay. IL-13Rα2 expression and Pep-1-Phor21-mediated killing were also determined in the cells treated with epigenetic regulators (Trichostatin A (TSA) and 5-aza-2 deoxycytidine (5-Aza-dC)). The hybrid lytic peptide cytotoxic activity correlated with the expression of IL-13Rα2 in prostate cancer cell lines cultured as monolayers (2D) or 3D spheroids. In addition, TSA or 5-Aza-dC treatment of prostate cancer cells, particularly those with low expression of IL-13Rα2, enhanced the cells' sensitivity to the lytic peptide by increasing IL-13Rα2 expression. These results demonstrate that the Pep-1-Phor21 hybrid lytic peptide has potent and selective anticancer properties against IL-13Rα2-expressing prostate cancer cells.
Collapse
|
4
|
Pei P, Chen L, Fan R, Zhou XR, Feng S, Liu H, Guo Q, Yin H, Zhang Q, Sun F, Peng L, Wei P, He C, Qiao R, Wang Z, Luo SZ. Computer-Aided Design of Lasso-like Self-Assembling Anticancer Peptides with Multiple Functions for Targeted Self-Delivery and Cancer Treatments. ACS NANO 2022; 16:13783-13799. [PMID: 36099446 DOI: 10.1021/acsnano.2c01014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Anticancer peptides are promising drug candidates for cancer treatment, but the short circulation time and low delivery efficiency limit their clinical applications. Herein, we designed several lasso-like self-assembling anticancer peptides (LASAPs) integrated with multiple functions by a computer-aided approach. Among these LASAPs, LASAP1 (CRGDKGPDCGKAFRRFLGALFKALSHLL, 1-9 disulfide bond) was determined to be superior to the others because it can self-assemble into homogeneous nanoparticles and exhibits improved stability in serum. Thus, LASAP1 was chosen for proving the design idea. LASAP1 can self-assemble into nanoparticles displaying iRGD on the surface because of its amphiphilic structure and accumulate to the tumor site after injection because of the EPR effect and iRGD targeting to αVβ3 integrin. The nanoparticles could disassemble in the acidic microenvironment of the solid tumor, and cleaved by the overexpressed hK2, which was secreted by prostate tumor cells, to release the effector peptide PTP-7b (FLGALFKALSHLL), which was further activated by the acidic pH. Therefore, LASAP1 could target the orthotopic prostate tumor in the model mice after intraperitoneal injection and specifically inhibit tumor growth, with low systematic toxicity. Combining the multiple targeting functions, LASAP1 represents a promising design of self-delivery of peptide drugs for targeted cancer treatments.
Collapse
Affiliation(s)
- Pengfei Pei
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Long Chen
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Ruru Fan
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Xi-Rui Zhou
- Division of Metrology in Chemistry, National Institute of Metrology, Beijing 100029, P.R. China
| | - Shan Feng
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Hangrui Liu
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Quanqiang Guo
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Huiwei Yin
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Qiang Zhang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Fude Sun
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Liang Peng
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Peng Wei
- School of Traditional Chinese Medicine, School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Chengzhi He
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Renzhong Qiao
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Shi-Zhong Luo
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| |
Collapse
|
5
|
Pagan L, Yfanti C, Rijneveld R, Todd M, Jongste P, Feijen JJ, Klaassen ES, Bouwes Bavinck JN, Struijk L, de Koning MNC, Prestegarden L, Niemeyer-van der Kolk T, van Poelgeest MIE, Rissmann R. Results of a randomized, placebo-controlled, first-in-human trial of topical CY-002 in patients with cutaneous warts. J Eur Acad Dermatol Venereol 2022; 36:e773-e775. [PMID: 35699238 DOI: 10.1111/jdv.18291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/05/2022] [Indexed: 11/30/2022]
Affiliation(s)
- L Pagan
- Centre for Human Drug Research, Leiden, The Netherlands.,Department of Gynaecology and Obstetrics, Leiden University Medical Centre, Leiden, The Netherlands
| | - C Yfanti
- Centre for Human Drug Research, Leiden, The Netherlands
| | - R Rijneveld
- Centre for Human Drug Research, Leiden, The Netherlands
| | - M Todd
- Centre for Human Drug Research, Leiden, The Netherlands
| | - P Jongste
- Centre for Human Drug Research, Leiden, The Netherlands
| | - J J Feijen
- Centre for Human Drug Research, Leiden, The Netherlands
| | - E S Klaassen
- Centre for Human Drug Research, Leiden, The Netherlands
| | - J N Bouwes Bavinck
- Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - L Struijk
- Viroclinics-DDL, DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | - M N C de Koning
- Viroclinics-DDL, DDL Diagnostic Laboratory, Rijswijk, The Netherlands
| | | | | | - M I E van Poelgeest
- Centre for Human Drug Research, Leiden, The Netherlands.,Department of Gynaecology and Obstetrics, Leiden University Medical Centre, Leiden, The Netherlands
| | - R Rissmann
- Centre for Human Drug Research, Leiden, The Netherlands.,Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.,Leiden Skin Institute, Leiden, The Netherlands
| |
Collapse
|
6
|
González-Cruz AO, Hernández-Juárez J, Ramírez-Cabrera MA, Balderas-Rentería I, Arredondo-Espinoza E. Peptide-based drug-delivery systems: A new hope for improving cancer therapy. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
7
|
Chen CH, Liu Y, Eskandari A, Ghimire J, Lin LC, Fang Z, Wimley WC, Ulmschneider JP, Suntharalingam K, Hu CJ, Ulmschneider MB. Integrated Design of a Membrane-Lytic Peptide-Based Intravenous Nanotherapeutic Suppresses Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105506. [PMID: 35246961 PMCID: PMC9069370 DOI: 10.1002/advs.202105506] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/12/2022] [Indexed: 05/30/2023]
Abstract
Membrane-lytic peptides offer broad synthetic flexibilities and design potential to the arsenal of anticancer therapeutics, which can be limited by cytotoxicity to noncancerous cells and induction of drug resistance via stress-induced mutagenesis. Despite continued research efforts on membrane-perforating peptides for antimicrobial applications, success in anticancer peptide therapeutics remains elusive given the muted distinction between cancerous and normal cell membranes and the challenge of peptide degradation and neutralization upon intravenous delivery. Using triple-negative breast cancer as a model, the authors report the development of a new class of anticancer peptides. Through function-conserving mutations, the authors achieved cancer cell selective membrane perforation, with leads exhibiting a 200-fold selectivity over non-cancerogenic cells and superior cytotoxicity over doxorubicin against breast cancer tumorspheres. Upon continuous exposure to the anticancer peptides at growth-arresting concentrations, cancer cells do not exhibit resistance phenotype, frequently observed under chemotherapeutic treatment. The authors further demonstrate efficient encapsulation of the anticancer peptides in 20 nm polymeric nanocarriers, which possess high tolerability and lead to effective tumor growth inhibition in a mouse model of MDA-MB-231 triple-negative breast cancer. This work demonstrates a multidisciplinary approach for enabling translationally relevant membrane-lytic peptides in oncology, opening up a vast chemical repertoire to the arms race against cancer.
Collapse
Affiliation(s)
- Charles H. Chen
- Department of ChemistryKing's College LondonLondonSE1 1DBUK
- Synthetic Biology GroupResearch Laboratory of ElectronicsMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Yu‐Han Liu
- Institute of Biomedical SciencesAcademia SinicaTaipei115Taiwan
| | | | - Jenisha Ghimire
- Department of Biochemistry and Molecular BiologyTulane UniversityNew OrleansLA70112USA
| | | | - Zih‐Syun Fang
- Institute of Biomedical SciencesAcademia SinicaTaipei115Taiwan
| | - William C. Wimley
- Department of Biochemistry and Molecular BiologyTulane UniversityNew OrleansLA70112USA
| | - Jakob P. Ulmschneider
- Department of PhysicsInstitute of Natural SciencesShanghai Jiao Tong UniversityShanghai200240China
| | | | | | | |
Collapse
|
8
|
Humeau J, Le Naour J, Galluzzi L, Kroemer G, Pol JG. Trial watch: intratumoral immunotherapy. Oncoimmunology 2021; 10:1984677. [PMID: 34676147 PMCID: PMC8526014 DOI: 10.1080/2162402x.2021.1984677] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023] Open
Abstract
While chemotherapy and radiotherapy remain the first-line approaches for the management of most unresectable tumors, immunotherapy has emerged in the past two decades as a game-changing treatment, notably with the clinical success of immune checkpoint inhibitors. Immunotherapies aim at (re)activating anticancer immune responses which occur in two main steps: (1) the activation and expansion of tumor-specific T cells following cross-presentation of tumor antigens by specialized myeloid cells (priming phase); and (2) the immunological clearance of malignant cells by these antitumor T lymphocytes (effector phase). Therapeutic vaccines, adjuvants, monoclonal antibodies, cytokines, immunogenic cell death-inducing agents including oncolytic viruses, anthracycline-based chemotherapy and radiotherapy, as well as adoptive cell transfer, all act at different levels of this cascade to (re)instate cancer immunosurveillance. Intratumoral delivery of these immunotherapeutics is being tested in clinical trials to promote superior antitumor immune activity in the context of limited systemic toxicity.
Collapse
Affiliation(s)
- Juliette Humeau
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC H3C 3J7, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Julie Le Naour
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Institut Universitaire de France, Paris, France
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Jonathan G. Pol
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
| |
Collapse
|
9
|
Abdel-Salam MAL, Pinto B, Cassali G, Bueno L, Pêgas G, Oliveira F, Silva I, Klein A, de Souza-Fagundes EM, de Lima ME, Carvalho-Tavares J. LyeTx I-b Peptide Attenuates Tumor Burden and Metastasis in a Mouse 4T1 Breast Cancer Model. Antibiotics (Basel) 2021; 10:1136. [PMID: 34572719 PMCID: PMC8466574 DOI: 10.3390/antibiotics10091136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 12/20/2022] Open
Abstract
Cationic anticancer peptides have exhibited potent anti-proliferative and anti-inflammatory effects in neoplastic illness conditions. LyeTx I-b is a synthetic peptide derived from Lycosa erythrognatha spider venom that previously showed antibiotic activity in vitro and in vivo. This study focused on the effects of LyeTxI-b on a 4T1 mouse mammary carcinoma model. Mice with a palpable tumor in the left flank were subcutaneously or intratumorally injected with LyeTx I-b (5 mg/kg), which significantly decreased the tumor volume and metastatic nodules. Histological analyses showed a large necrotic area in treated primary tumors compared to the control. LyeTxI-b reduced tumor growth and lung metastasis in the 4T1 mouse mammary carcinoma model with no signs of toxicity in healthy or cancerous mice. The mechanism of action of LyeTx I-b on the 4T1 mouse mammary carcinoma model was evaluated in vitro and is associated with induction of apoptosis and cell proliferation inhibition. Furthermore, LyeTx I-b seems to be an efficient regulator of the 4T1 tumor microenvironment by modulating several cytokines, such as TGF-β, TNF-α, IL-1β, IL-6, and IL-10, in primary tumor and lung, spleen, and brain. LyeTx I-b also plays a role in leukocytes rolling and adhesion into spinal cord microcirculation and in the number of circulating leukocytes. These data suggest a potent antineoplastic efficacy ofLyeTx I-b.
Collapse
Affiliation(s)
- Mostafa A. L. Abdel-Salam
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (M.A.L.A.-S.); (B.P.); (E.M.d.S.-F.)
| | - Bárbara Pinto
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (M.A.L.A.-S.); (B.P.); (E.M.d.S.-F.)
| | - Geovanni Cassali
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (G.C.); (G.P.); (F.O.)
| | - Lilian Bueno
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Gabriela Pêgas
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (G.C.); (G.P.); (F.O.)
| | - Fabrício Oliveira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (G.C.); (G.P.); (F.O.)
| | - Irismara Silva
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (I.S.); (A.K.)
| | - André Klein
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (I.S.); (A.K.)
| | - Elaine Maria de Souza-Fagundes
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (M.A.L.A.-S.); (B.P.); (E.M.d.S.-F.)
| | - Maria Elena de Lima
- Programa de Pós-Graduação em Medicina-Biomedicina, Faculdade Santa Casa de Belo Horizonte, Belo Horizonte 30110-005, Brazil
| | - Juliana Carvalho-Tavares
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (M.A.L.A.-S.); (B.P.); (E.M.d.S.-F.)
| |
Collapse
|
10
|
Vitale I, Yamazaki T, Wennerberg E, Sveinbjørnsson B, Rekdal Ø, Demaria S, Galluzzi L. Targeting Cancer Heterogeneity with Immune Responses Driven by Oncolytic Peptides. Trends Cancer 2021; 7:557-572. [PMID: 33446447 DOI: 10.1016/j.trecan.2020.12.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023]
Abstract
Accumulating preclinical and clinical evidence indicates that high degrees of heterogeneity among malignant cells constitute a considerable obstacle to the success of cancer therapy. This calls for the development of approaches that operate - or enable established treatments to operate - despite such intratumoral heterogeneity (ITH). In this context, oncolytic peptides stand out as promising therapeutic tools based on their ability to drive immunogenic cell death associated with robust anticancer immune responses independently of ITH. We review the main molecular and immunological pathways engaged by oncolytic peptides, and discuss potential approaches to combine these agents with modern immunotherapeutics in support of superior tumor-targeting immunity and efficacy in patients with cancer.
Collapse
Affiliation(s)
- Ilio Vitale
- Italian Institute for Genomic Medicine (IIGM), Istituto Di Ricovero e Cura a Carattere Scientifico (IRCSS) Candiolo, Torino, Italy; Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia (FPO)-IRCCS, Candiolo, Italy
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Erik Wennerberg
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Baldur Sveinbjørnsson
- Lytix Biopharma, Oslo, Norway; Department of Medical Biology, University of Tromsø, Tromsø, Norway; Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Øystein Rekdal
- Lytix Biopharma, Oslo, Norway; Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, USA; Université de Paris, Paris, France.
| |
Collapse
|
11
|
Aasen SN, Espedal H, Holte CF, Keunen O, Karlsen TV, Tenstad O, Maherally Z, Miletic H, Hoang T, Eikeland AV, Baghirov H, Olberg DE, Pilkington GJ, Sarkar G, Jenkins RB, Sundstrøm T, Bjerkvig R, Thorsen F. Improved Drug Delivery to Brain Metastases by Peptide-Mediated Permeabilization of the Blood-Brain Barrier. Mol Cancer Ther 2019; 18:2171-2181. [PMID: 31467182 DOI: 10.1158/1535-7163.mct-19-0160] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/06/2019] [Accepted: 08/19/2019] [Indexed: 11/16/2022]
Abstract
Patients with melanoma have a high risk of developing brain metastasis, which is associated with a dismal prognosis. During early stages of metastasis development, the blood-brain barrier (BBB) is likely intact, which inhibits sufficient drug delivery into the metastatic lesions. We investigated the ability of the peptide, K16ApoE, to permeabilize the BBB for improved treatment with targeted therapies preclinically. Dynamic contrast enhanced MRI (DCE-MRI) was carried out on NOD/SCID mice to study the therapeutic window of peptide-mediated BBB permeabilization. Further, both in vivo and in vitro assays were used to determine K16ApoE toxicity and to obtain mechanistic insight into its action on the BBB. The therapeutic impact of K16ApoE on metastases was evaluated combined with the mitogen-activated protein kinase pathway inhibitor dabrafenib, targeting BRAF mutated melanoma cells, which is otherwise known not to cross the intact BBB. Our results from the DCE-MRI experiments showed effective K16ApoE-mediated BBB permeabilization lasting for up to 1 hour. Mechanistic studies showed a dose-dependent effect of K16ApoE caused by induction of endocytosis. At concentrations above IC50, the peptide additionally showed nonspecific disturbances on plasma membranes. Combined treatment with K16ApoE and dabrafenib reduced the brain metastatic burden in mice and increased animal survival, and PET/CT showed that the peptide also facilitated the delivery of compounds with molecular weights as large as 150 kDa into the brain. To conclude, we demonstrate a transient permeabilization of the BBB, caused by K16ApoE, that facilitates enhanced drug delivery into the brain. This improves the efficacy of drugs that otherwise do not cross the intact BBB.
Collapse
Affiliation(s)
- Synnøve Nymark Aasen
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway.,Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Centre, University of Bergen, Bergen, Norway
| | - Heidi Espedal
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Centre, University of Bergen, Bergen, Norway.,Molecular Imaging Center, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Christopher Florian Holte
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Centre, University of Bergen, Bergen, Norway
| | - Olivier Keunen
- Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | | | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Zaynah Maherally
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Hrvoje Miletic
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Centre, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Tuyen Hoang
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Centre, University of Bergen, Bergen, Norway
| | | | - Habib Baghirov
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Dag Erlend Olberg
- Department of Pharmaceutical Chemistry, University of Oslo, Oslo, Norway.,Norwegian Cyclotron Center, Oslo University Hospital, Oslo, Norway
| | - Geoffrey John Pilkington
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Gobinda Sarkar
- Division of Experimental Pathology, Mayo Clinic, Rochester, Minnesota
| | - Robert B Jenkins
- Division of Experimental Pathology, Mayo Clinic, Rochester, Minnesota
| | - Terje Sundstrøm
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Centre, University of Bergen, Bergen, Norway.,Department of Neurosurgery, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Rolf Bjerkvig
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Centre, University of Bergen, Bergen, Norway.,Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Frits Thorsen
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Centre, University of Bergen, Bergen, Norway. .,Molecular Imaging Center, Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
12
|
Neundorf I. Antimicrobial and Cell-Penetrating Peptides: How to Understand Two Distinct Functions Despite Similar Physicochemical Properties. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1117:93-109. [PMID: 30980355 DOI: 10.1007/978-981-13-3588-4_7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antimicrobial and cell-penetrating peptides are both classes of membrane-active peptides sharing similar physicochemical properties. Both kinds of peptides have attracted much attention owing to their specific features. AMPs disrupt cell membranes of bacteria and display urgently needed antibiotic substances with alternative modes of action. Since the multidrug resistance of bacterial pathogens is a more and more raising concern, AMPs have gained much interest during the past years. On the other side, CPPs enter eukaryotic cells without substantially affecting the plasma membrane. They can be used as drug delivery platforms and have proven their usefulness in various applications. However, although both groups of peptides are quite similar, their intrinsic activity is often different, and responsible factors are still in discussion. The aim of this chapter is to summarize and shed light on recent findings and concepts dealing with differences and similarities of AMPs and CPPs and to understand these different functions.
Collapse
Affiliation(s)
- Ines Neundorf
- Department of Chemistry, Institute for Biochemistry, University of Cologne, Cologne, Germany.
| |
Collapse
|
13
|
Yang G, Huang T, Wang Y, Wang H, Li Y, Yu K, Dong L. Sustained Release of Antimicrobial Peptide from Self-Assembling Hydrogel Enhanced Osteogenesis. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2018; 29:1812-1824. [PMID: 30035666 DOI: 10.1080/09205063.2018.1504191] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Biomaterials have been widely used in bone infection and osteomyelitis resulting from their versatile functionalities. As far as we know, the appearance of osteomyelitis was mainly caused by bacteria. Therefore, a biomaterial that can cure bone infection and promote osteogenesis may become an ideal candidate for the treatment of osteomyelitis. Cationic antimicrobial peptides (AMPs) have been proved to have an excellent ability to kill bacteria, fungi, viruses, and parasites. However, the application of AMPs in bone infection and osteomyelitis is quite limited. Here, we designed a new hydrogel that has an inhibitory effect on the proliferation of S. aureus and enhances osteogenesis. RADA16 self-assembling peptide has been applied for AMPs delivery. In this study, we demonstrated that RADA16 could form a stable structure and afford the sustained release of AMPs. The interwoven nanofiber morphology was detected by field emission scanning electron microscopy. The sustained release study revealed that the release of AMPs could be obtained until 28 days. In vitro research showed this new self-assembling hydrogel could promote the proliferation of bone mesenchymal stem cells (BMSCs) and inhibited the growth of S. aureus. More importantly, the results in vivo also proved that RADA16-AMP self-assembling peptide had an excellent effect on bone formation. Our findings implied that we successfully combined RADA16 and AMPs together and laid the foundation for the application of this new hydrogel and open new avenues for biomaterials.
Collapse
Affiliation(s)
- Guoli Yang
- a Department of Implantology , Stomatology Hospital, School of Medicine, Zhejiang University , Hangzhou , P. R. China
| | - Tingben Huang
- a Department of Implantology , Stomatology Hospital, School of Medicine, Zhejiang University , Hangzhou , P. R. China
| | - Ying Wang
- c Department of Oral Medicine , Stomatology Hospital, School of Medicine, Zhejiang University , Hangzhou , P. R. China
| | - Huiming Wang
- b Department of Oral and Maxillofacial Surgery , Stomatology Hospital, School of Medicine, Zhejiang University , Hangzhou , P. R. China
| | - Yongzheng Li
- a Department of Implantology , Stomatology Hospital, School of Medicine, Zhejiang University , Hangzhou , P. R. China
| | - Ke Yu
- a Department of Implantology , Stomatology Hospital, School of Medicine, Zhejiang University , Hangzhou , P. R. China
| | - Lingling Dong
- c Department of Oral Medicine , Stomatology Hospital, School of Medicine, Zhejiang University , Hangzhou , P. R. China
| |
Collapse
|
14
|
Piotrowska U, Sobczak M, Oledzka E. Current state of a dual behaviour of antimicrobial peptides-Therapeutic agents and promising delivery vectors. Chem Biol Drug Des 2017; 90:1079-1093. [DOI: 10.1111/cbdd.13031] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/14/2017] [Accepted: 05/18/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Urszula Piotrowska
- Chair of Inorganic and Analytical Chemistry; Department of Biomaterials Chemistry; Faculty of Pharmacy with the Laboratory Medicine Division; Medical University of Warsaw; Warsaw Poland
| | - Marcin Sobczak
- Chair of Inorganic and Analytical Chemistry; Department of Biomaterials Chemistry; Faculty of Pharmacy with the Laboratory Medicine Division; Medical University of Warsaw; Warsaw Poland
| | - Ewa Oledzka
- Chair of Inorganic and Analytical Chemistry; Department of Biomaterials Chemistry; Faculty of Pharmacy with the Laboratory Medicine Division; Medical University of Warsaw; Warsaw Poland
| |
Collapse
|
15
|
Gronewold A, Horn M, Ranđelović I, Tóvári J, Muñoz Vázquez S, Schomäcker K, Neundorf I. Characterization of a Cell-Penetrating Peptide with Potential Anticancer Activity. ChemMedChem 2017; 12:42-49. [PMID: 27860402 PMCID: PMC5516705 DOI: 10.1002/cmdc.201600498] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/03/2016] [Indexed: 12/25/2022]
Abstract
Cell-penetrating peptides (CPPs) are still an interesting and viable alternative for drug delivery applications. CPPs contain considerably high amounts of positively charged amino acids, imparting them with cationic character. Tumor cells are characterized by an enhanced anionic nature of their membrane surface, a property that could be used by CPPs to target these cells. We recently identified a branched CPP that displays a high internalization capacity while exhibiting selectivity for certain tumor cell types. In this study we elucidated this observation in greater detail by investigating the underlying mechanism behind the cellular uptake of this peptide. An additional cytotoxicity screen against several cancer cell lines indeed demonstrates high cytotoxic activity against cancer cells over normal fibroblasts. Furthermore, we show that this feature can be used for delivering the anticancer drug actinomycin D with high efficiency in the MCF-7 cancer cell line.
Collapse
Affiliation(s)
- Anja Gronewold
- University of CologneDepartment of ChemistryInstitute of BiochemistryZuelpicher Str. 4750674CologneGermany
| | - Mareike Horn
- University of CologneDepartment of ChemistryInstitute of BiochemistryZuelpicher Str. 4750674CologneGermany
| | - Ivan Ranđelović
- National Institute of OncologyDepartment of Experimental PharmacologyRáth Gy. u. 7–9112BudapestHungary
| | - József Tóvári
- National Institute of OncologyDepartment of Experimental PharmacologyRáth Gy. u. 7–9112BudapestHungary
| | - Sergio Muñoz Vázquez
- University Hospital of CologneDepartment of Nuclear MedicineKerpener Str. 6250937CologneGermany
| | - Klaus Schomäcker
- University Hospital of CologneDepartment of Nuclear MedicineKerpener Str. 6250937CologneGermany
| | - Ines Neundorf
- University of CologneDepartment of ChemistryInstitute of BiochemistryZuelpicher Str. 4750674CologneGermany
| |
Collapse
|
16
|
Reinhardt A, Neundorf I. Design and Application of Antimicrobial Peptide Conjugates. Int J Mol Sci 2016; 17:E701. [PMID: 27187357 PMCID: PMC4881524 DOI: 10.3390/ijms17050701] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/25/2016] [Accepted: 05/04/2016] [Indexed: 12/17/2022] Open
Abstract
Antimicrobial peptides (AMPs) are an interesting class of antibiotics characterized by their unique antibiotic activity and lower propensity for developing resistance compared to common antibiotics. They belong to the class of membrane-active peptides and usually act selectively against bacteria, fungi and protozoans. AMPs, but also peptide conjugates containing AMPs, have come more and more into the focus of research during the last few years. Within this article, recent work on AMP conjugates is reviewed. Different aspects will be highlighted as a combination of AMPs with antibiotics or organometallic compounds aiming to increase antibacterial activity or target selectivity, conjugation with photosensitizers for improving photodynamic therapy (PDT) or the attachment to particles, to name only a few. Owing to the enormous resonance of antimicrobial conjugates in the literature so far, this research topic seems to be very attractive to different scientific fields, like medicine, biology, biochemistry or chemistry.
Collapse
Affiliation(s)
- Andre Reinhardt
- Department of Chemistry, Institute of Biochemistry, University of Cologne, Zuelpicher Str. 47, D-50674 Cologne, Germany.
| | - Ines Neundorf
- Department of Chemistry, Institute of Biochemistry, University of Cologne, Zuelpicher Str. 47, D-50674 Cologne, Germany.
| |
Collapse
|
17
|
Development of a lytic peptide derived from BH3-only proteins. Cell Death Discov 2016; 2:16008. [PMID: 27551502 PMCID: PMC4979451 DOI: 10.1038/cddiscovery.2016.8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/08/2016] [Accepted: 01/12/2016] [Indexed: 12/28/2022] Open
Abstract
Despite great advances in cancer therapy, drug resistance is a difficult hurdle to overcome that requires development of anticancer agents with novel and effective modes of action. In a number of studies, lytic peptides have shown remarkable ability to eliminate cancer cells through a different way from traditional treatments. Lytic peptides are positively charged, amphiphilic, and are efficient at binding and disrupting the negatively charged cell membrane of cancer cells. In this study, we described the anticancer properties of a lytic peptide that was developed on the basis of the alignment of amphiphilic BH3 peptides. Our results demonstrated that the positive charge and conformation constraint were favourable for efficient cancer cell elimination. Artificial BCL-2 homology 3 peptides (ABH3) exhibited effective anticancer effects against a series of cancer cell lines in vitro and in HeLa human cervical tumour xenografts in vivo. ABH3 induced cell death in an apoptosis-independent manner through the lytic properties of the peptide that caused disruption of cell membrane. Our results showed that charge tuning and conformation constraining in a lytic peptide could be applied to optimise the anticancer activity of lytic peptides. These results also suggest that ABH3 may be a promising beginning for the development of additional lytic peptides as anticancer reagents.
Collapse
|
18
|
The Cytolytic Amphipathic β(2,2)-Amino Acid LTX-401 Induces DAMP Release in Melanoma Cells and Causes Complete Regression of B16 Melanoma. PLoS One 2016; 11:e0148980. [PMID: 26881822 PMCID: PMC4755540 DOI: 10.1371/journal.pone.0148980] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/26/2016] [Indexed: 01/23/2023] Open
Abstract
In the present study we examined the ability of the amino acid derivative LTX-401 to induce cell death in cancer cell lines, as well as the capacity to induce regression in a murine melanoma model. Mode of action studies in vitro revealed lytic cell death and release of danger-associated molecular pattern molecules, preceded by massive cytoplasmic vacuolization and compromised lysosomes in treated cells. The use of a murine melanoma model demonstrated that the majority of animals treated with intratumoural injections of LTX-401 showed complete and long-lasting remission. Taken together, these results demonstrate the potential of LTX-401 as an immunotherapeutic agent for the treatment of solid tumors.
Collapse
|
19
|
The antimicrobial peptide pardaxin exerts potent anti-tumor activity against canine perianal gland adenoma. Oncotarget 2016; 6:2290-301. [PMID: 25544775 PMCID: PMC4385852 DOI: 10.18632/oncotarget.2959] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 12/09/2014] [Indexed: 12/26/2022] Open
Abstract
Pardaxin is an antimicrobial peptide of 33 amino acids, originally isolated from marine fish. We previously demonstrated that pardaxin has anti-tumor activity against murine fibrosarcoma, both in vitro and in vivo. In this study, we examined the anti-tumor activity, toxicity profile, and maximally-tolerated dose of pardaxin treatment in dogs with different types of refractory tumor. Local injection of pardaxin resulted in a significant reduction of perianal gland adenoma growth between 28 and 38 days post-treatment. Surgical resection of canine histiocytomas revealed large areas of ulceration, suggesting that pardaxin acts like a lytic peptide. Pardaxin treatment was not associated with significant variations in blood biochemical parameters or secretion of immune-related proteins. Our findings indicate that pardaxin has strong therapeutic potential for treating perianal gland adenomas in dogs. These data justify the veterinary application of pardaxin, and also provide invaluable information for veterinary medicine and future human clinical trials.
Collapse
|