1
|
Singh P, Khatib MN, R R, Kaur M, Srivastava M, Barwal A, Rajput GVS, Rajput P, Syed R, Sharma G, Kumar S, Shabil M, Pandey S, Brar M, Bushi G, Mehta R, Sah S, Goh KW, Satapathy P, Gaidhane AM, Samal SK. Advancements and challenges in personalized neoantigen-based cancer vaccines. Oncol Rev 2025; 19:1541326. [PMID: 40160263 PMCID: PMC11949952 DOI: 10.3389/or.2025.1541326] [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: 12/07/2024] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
Advancements in personalized neoantigen-based cancer vaccines are ushering in a new era in oncology, targeting unique genetic alterations within tumors to enhance treatment precision and efficacy. Neoantigens, specific to cancer cells and absent in normal tissues, are at the heart of these vaccines, promising to direct the immune system specifically against the tumor, thereby maximizing therapeutic efficacy while minimizing side effects. The identification of neoantigens through genomic and proteomic technologies is central to developing these vaccines, allowing for the precise mapping of a tumor's mutational landscape. Despite advancements, accurately predicting which neoantigens will elicit strong immune responses remains challenging due to tumor variability and the complexity of immune system interactions. This necessitates further refinement of bioinformatics tools and predictive models. Moreover, the efficacy of these vaccines heavily depends on innovative delivery methods that enhance neoantigen presentation to the immune system. Techniques like encapsulating neoantigens in lipid nanoparticles and using viral vectors are critical for improving vaccine stability and delivery. Additionally, these vaccines contribute towards achieving Sustainable Development Goal 3.8, promoting universal health coverage by advancing access to safe and effective cancer treatments. This review delves into the potential of neoantigen-based vaccines to transform cancer treatment, examining both revolutionary advancements and the ongoing challenges they face.
Collapse
Affiliation(s)
- Parminder Singh
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia
| | - Mahalaqua Nazli Khatib
- Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education, Wardha, India
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mandeep Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | | | - Amit Barwal
- Chandigarh Pharmacy College, Chandigarh Group of College, Mohali, Punjab, India
| | - G. V. Siva Rajput
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Pranchal Rajput
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Rukshar Syed
- IES Institute of Pharmacy, IES University, Bhopal, Madhya Pradesh, India
| | - Gajendra Sharma
- New Delhi Institute of Management, Tughlakabad Institutional Area, New Delhi, India
| | - Sunil Kumar
- Department of Microbiology, Graphic Era (Deemed to be University), Dehradun, India
| | - Muhammed Shabil
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Sakshi Pandey
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
| | - Manvinder Brar
- Chitkara Centre for Research and Development, Chitkara University, Solan, Himachal Pradesh, India
| | - Ganesh Bushi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Rachana Mehta
- Clinical Microbiology, RDC, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, India
| | - Sanjit Sah
- Department of Paediatrics, Dr. D. Y. Patil Medical College Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed-to-be-University), Pimpri, Pune, Maharashtra, India
- Department of Public Health Dentistry, Dr. D. Y. Patil Medical College Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed-to-be-University), Pimpri, Pune, Maharashtra, India
- Department of Medicine, Korea Universtiy, Seoul, Republic of Korea
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia
- Faculty of Mathematics and Natural Sciences, Universitas Negeri Padang, Padang, Indonesia
| | - Prakasini Satapathy
- University Center for Research and Development, Chandigarh University, Mohali, Punjab, India
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Abhay M. Gaidhane
- Jawaharlal Nehru Medical College, and Global Health Academy, School of Epidemiology and Public Health, Datta Meghe Institute of Higher Education, Wardha, India
| | - Shailesh Kumar Samal
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
Kirane A, Lee D, Ariyan C. Surgical Considerations in Tumor-Infiltrating Lymphocyte Therapy: Challenges and Opportunities. Transplant Cell Ther 2025; 31:S591-S598. [PMID: 40089328 DOI: 10.1016/j.jtct.2024.11.015] [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: 07/23/2024] [Accepted: 11/24/2024] [Indexed: 03/17/2025]
Abstract
Adoptive T cell therapy (ACT) using tumor-infiltrating lymphocytes (TILs) is a promising personalized immunotherapy approach, spearheaded by Dr. Steven Rosenberg, targeting various cancer types. Despite initial challenges in TIL production, recent advancements have showcased its superiority to immune checkpoint blockade in metastatic melanoma, even after anti-PD-1 therapy failure. The expedited manufacturing process, now around 3 weeks, coupled with the US Food and Drug Administration approval of lifileucel in 2024, is poised to propel TIL therapy into mainstream oncology. This commentary delves into the critical surgical aspects of TIL harvesting, emphasizing the integral role of surgeons in ensuring optimal TIL quality, safety, and therapeutic effectiveness. By shedding light on these considerations, this article aims to guide and enhance collaborative efforts in advancing TIL therapy for patients facing limited treatment options.
Collapse
Affiliation(s)
- Amanda Kirane
- Stanford University, Department of Surgery, 1201 Welch Road, Stanford, CA 94304.
| | - David Lee
- Stanford University, Department of Surgery, 1201 Welch Road, Stanford, CA 94304
| | - Charlotte Ariyan
- Memorial Sloan Kettering Cancer Center, Department of Surgery, 1275 York Ave, New York, NY 10065
| |
Collapse
|
3
|
Zhang Y, Fu H, Zhao Q. Current status and perspectives of clinical trials for tumor-infiltrating lymphocyte therapy. Clin Transl Oncol 2025; 27:466-472. [PMID: 39078471 DOI: 10.1007/s12094-024-03608-z] [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: 04/03/2024] [Accepted: 07/09/2024] [Indexed: 07/31/2024]
Abstract
Immunotherapies, mainly immune checkpoint inhibitors (ICIs), have revolutionized cancer treatment strategies over the past decade, but their limitations have limited clinical applications. Tumor-infiltrating lymphocyte (TIL) therapy is a type of adoptive cell therapy (ACT), which collects infiltrating lymphocytes at the tumor site and expands them in vitro to obtain TIL final products cloned by various T-cell receptors, subsequently reinfused TIL into the patient, which is effective for the treatment of solid tumors. The approval of Lifileucel for commercialization marks the success of TIL therapy. This review summarizes the current status of clinical trials of TIL treatment. In addition, it is suggested that the current research trend of TIL should focus on improving the survival time of TIL in vivo, reducing drug toxicity, and searching for prognostic markers. Finally, it is expected that TIL therapy can be applied to a more wide range of clinical treatments.
Collapse
Affiliation(s)
- Yunting Zhang
- Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Hongye Fu
- Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Qiong Zhao
- Department of Thoracic Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310022, People's Republic of China.
| |
Collapse
|
4
|
Tang Y, Yang C, Zhao J, Heng H, Peng M, Sun L, Dai L, Chan EWC, Chen S. LTX-315 is a novel broad-spectrum antimicrobial peptide against clinical multidrug-resistant bacteria. J Adv Res 2025:S2090-1232(24)00621-0. [PMID: 39793961 DOI: 10.1016/j.jare.2024.12.044] [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: 10/17/2024] [Revised: 12/17/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025] Open
Abstract
INTRODUCTION Infections stemming from multidrug-resistant bacteria present a substantial threat to public health today. Discovering or synthesizing novel compounds is crucial to alleviate this pressing situation. OBJECTIVE The main purpose of this study is to verify the antibacterial activity of LTX-315 and explore its primary action mode. METHODS Through antibacterial phenotype assay screening, we obtained a potent compound named LTX-315 from diverse drug libraries, 10,926 compounds in total. Then, the bactericidal effect and its action mode were explored through biochemical and chemistry methods such as atime-killing curve, scanning electronic microscopy, isothermal titration calorimetry analysis, and nuclear magnetic resonance. Finally, the efficacy in vivo of LTX-315 against drug-resistant bacteria was proved through amice infection model. RESULTS In this study, LTX-315, an oncolytic peptide, was discovered to effectively eliminate gram-positive and gram-negative pathogens, even for those multidrug-resistant strains. Through strong electrostatic interactions, LTX-315 can bind to the membrane component phosphatidylglycerol (PG) with extremely high affinity (nanomolar level). Strikingly, in contrast to the typical electrostatic interactions of antibacterial peptides, the indole group of LTX-315, situated near the alkyl chain, exhibits significantly enhanced recognition and interaction with PG due to the hydrophobic effect of the alkyl chain. Furthermore, it exerts various impacts on cell membranes, including damaging integrity, increasing permeability, and decreasing membrane fluidity. Additionally, microscopy revealed significant cell disintegration. The influence, in turn, disrupts several physiological activities inside cells, such as increasing the reactive oxygen species level, ultimately leading to cell death. Finally, the efficacy of LTX-315 in vivo against multidrug-resistant and hypervirulent Klebsiella pneumoniae was demonstrated. CONCLUSION The unique mechanism of LTX-315 involves high-affinity binding to PG and subsequent membrane disruption, providing a novel approach against multidrug-resistant bacteria compared to conventional antibiotics. As a potential candidate, it shows promise in effectively treating bacterial infections, particularly those caused by drug-resistant bacteria, thereby addressing the escalating challenge of antibiotic resistance worldwide.
Collapse
Affiliation(s)
- Yang Tang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong; State Key Laboratory of Chemical Biology and Drug Discovery and the Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Chen Yang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong; State Key Laboratory of Chemical Biology and Drug Discovery and the Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Jiamin Zhao
- State Key Laboratory of Chemical Biology and Drug Discovery and the Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Heng Heng
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong; State Key Laboratory of Chemical Biology and Drug Discovery and the Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Mingxiu Peng
- Shenzhen Key Laboratory for Food Biological Safety Control, Food Safety and Technology Research Centre, The Hong Kong PolyU Shenzhen Research Institute, Shenzhen, PR China
| | - Liang Sun
- City University of Hong Kong, Shenzhen Research Institute, Nanshan, PR China
| | - Liang Dai
- Department of Physics, City University of Hong Kong, Kowloon, Hong Kong
| | - Edward Wai-Chi Chan
- State Key Laboratory of Chemical Biology and Drug Discovery and the Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Sheng Chen
- State Key Laboratory of Chemical Biology and Drug Discovery and the Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong; Shenzhen Key Laboratory for Food Biological Safety Control, Food Safety and Technology Research Centre, The Hong Kong PolyU Shenzhen Research Institute, Shenzhen, PR China.
| |
Collapse
|
5
|
Cerrato G, Liu P, Zhao L, Petrazzuolo A, Humeau J, Schmid ST, Abdellatif M, Sauvat A, Kroemer G. AI-based classification of anticancer drugs reveals nucleolar condensation as a predictor of immunogenicity. Mol Cancer 2024; 23:275. [PMID: 39702289 DOI: 10.1186/s12943-024-02189-3] [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: 10/04/2024] [Accepted: 11/28/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Immunogenic cell death (ICD) inducers are often identified in phenotypic screening campaigns by the release or surface exposure of various danger-associated molecular patterns (DAMPs) from malignant cells. This study aimed to streamline the identification of ICD inducers by leveraging cellular morphological correlates of ICD, specifically the condensation of nucleoli (CON). METHODS We applied artificial intelligence (AI)-based imaging analyses to Cell Paint-stained cells exposed to drug libraries, identifying CON as a marker for ICD. CON was characterized using SYTO 14 fluorescent staining and holotomographic microscopy, and visualized by AI-deconvoluted transmitted light microscopy. A neural network-based quantitative structure-activity relationship (QSAR) model was trained to link molecular descriptors of compounds to the CON phenotype, and the classifier was validated using an independent dataset from the NCI-curated mechanistic collection of anticancer agents. RESULTS CON strongly correlated with the inhibition of DNA-to-RNA transcription. Cytotoxic drugs that inhibit RNA synthesis without causing DNA damage were as effective as conventional cytotoxicants in inducing ICD, as demonstrated by DAMPs release/exposure and vaccination efficacy in mice. The QSAR classifier successfully predicted drugs with a high likelihood of inducing CON. CONCLUSIONS We developed AI-based algorithms for predicting CON-inducing drugs based on molecular descriptors and their validation using automated micrographs analysis, offering a new approach for screening ICD inducers with minimized adverse effects in cancer therapy.
Collapse
Affiliation(s)
- Giulia Cerrato
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France.
- Onco-Pheno-Screen Platform, Centre de Recherche des Cordeliers, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
| | - Peng Liu
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France
- Onco-Pheno-Screen Platform, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Liwei Zhao
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France
- Onco-Pheno-Screen Platform, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Adriana Petrazzuolo
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- International Centre for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Juliette Humeau
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche en Cancérologie de Lyon (CRCL), Equipe Oncopharmacologie, Faculté Rockfeller, Lyon, France
| | - Sophie Theresa Schmid
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Mahmoud Abdellatif
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Allan Sauvat
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France.
- Onco-Pheno-Screen Platform, Centre de Recherche des Cordeliers, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France.
- Onco-Pheno-Screen Platform, Centre de Recherche des Cordeliers, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
- Centre de Recherche des Cordeliers, 15 Rue de l'École de Médecine, Paris, 75006, France.
| |
Collapse
|
6
|
D'Avanzo C, Blaeschke F, Lysandrou M, Ingelfinger F, Zeiser R. Advances in cell therapy: progress and challenges in hematological and solid tumors. Trends Pharmacol Sci 2024; 45:1119-1134. [PMID: 39603960 DOI: 10.1016/j.tips.2024.10.016] [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: 08/12/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 11/29/2024]
Abstract
Cell-based therapies harness the endogenous ability of the immune system to fight cancer and have shown promising results in the treatment of hematological malignancies. However, their clinical application beyond B cell malignancies is hampered by numerous hurdles, ranging from relapsed disease to a hostile tumor microenvironment (TME). Recent advances in cell engineering and TME modulation may expand the applicability of these therapies to a wider range of cancers, creating new treatment possibilities. Breakthroughs in advanced gene editing and sophisticated cell engineering, have also provided promising solutions to longstanding challenges. In this review, we examine the challenges and future directions of the most prominent cell-based therapies, including chimeric antigen receptor (CAR)-T cells, tumor-infiltrating lymphocytes (TILs), and natural killer (NK) cells, and emerging modalities. We provide a comprehensive analysis of emerging cell types and combination strategies translated into clinical trials, offering insights into the next generation of cell-based cancer treatments.
Collapse
Affiliation(s)
- Claudia D'Avanzo
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Franziska Blaeschke
- German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; Heidelberg University Hospital, Heidelberg, Germany
| | - Memnon Lysandrou
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florian Ingelfinger
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
7
|
Zare-Zardini H, Saberian E, Jenča A, Ghanipour-Meybodi R, Jenča A, Petrášová A, Jenčová J. From defense to offense: antimicrobial peptides as promising therapeutics for cancer. Front Oncol 2024; 14:1463088. [PMID: 39445062 PMCID: PMC11496142 DOI: 10.3389/fonc.2024.1463088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
Antimicrobial peptides (AMPs), naturally occurring components of innate immunity, are emerging as a promising new class of anticancer agents. This review explores the potential of AMPs as a novel class of anticancer agents. AMPs, naturally occurring peptides with broad-spectrum antimicrobial activity, exhibit several characteristics that make them attractive candidates for cancer therapy, including selectivity for cancer cells, broad-spectrum activity, and immunomodulatory effects. Analysis of a dataset of AMPs with anticancer activity reveals that their effectiveness is influenced by various structural properties, including net charge, length, Boman index, and hydrophobicity. These properties contribute to their ability to target and disrupt cancer cell membranes, interfere with intracellular processes, and modulate the immune response. The review highlights the promising potential of AMPs as a new frontier in cancer treatment, offering hope for more effective and less toxic therapies. AMPs demonstrate promising potential in cancer therapy through multiple mechanisms, including direct cytotoxicity, immune response modulation, and targeting of the tumor microenvironment, as evidenced by extensive preclinical studies in animal models showing tumor regression, metastasis inhibition, and improved survival rates. AMPs show significant potential as cancer therapeutics through their direct cytotoxicity, immune response modulation, and tumor microenvironment targeting, with promising results from preclinical studies and early-phase clinical trials. Future research should focus on optimizing AMP properties, developing novel delivery strategies, and exploring synergistic combination therapies to fully realize their potential as effective cancer treatments, while addressing challenges related to stability, delivery, and potential toxicity.
Collapse
Affiliation(s)
- Hadi Zare-Zardini
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| | - Elham Saberian
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | | | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | - Adriána Petrášová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | - Janka Jenčová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| |
Collapse
|
8
|
Rencinai A, Tollapi E, Marianantoni G, Brunetti J, Henriquez T, Pini A, Bracci L, Falciani C. Branched oncolytic peptides target HSPGs, inhibit metastasis, and trigger the release of molecular determinants of immunogenic cell death in pancreatic cancer. Front Mol Biosci 2024; 11:1429163. [PMID: 39417004 PMCID: PMC11479992 DOI: 10.3389/fmolb.2024.1429163] [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: 05/07/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Immunogenic cell death (ICD) can be exploited to treat non-immunoreactive tumors that do not respond to current standard and innovative therapies. Not all chemotherapeutics trigger ICD, among those that do exert this effect, there are anthracyclines, irinotecan, some platinum derivatives and oncolytic peptides. We studied two new branched oncolytic peptides, BOP7 and BOP9 that proved to elicit the release of damage-associated molecular patterns DAMPS, mediators of ICD, in pancreatic cancer cells. The two BOPs selectively bound and killed tumor cells, particularly PANC-1 and Mia PaCa-2, but not cells of non-tumor origin such as RAW 264.7, CHO-K1 and pgsA-745. The cancer selectivity of the two BOPs may be attributed to their repeated cationic sequences, which enable multivalent binding to heparan sulfate glycosaminoglycans (HSPGs), bearing multiple anionic sulfation patterns on cancer cells. This interaction of BOPs with HSPGs not only fosters an anti-metastatic effect in vitro, as demonstrated by reduced adhesion and migration of PANC-1 cancer cells, but also shows promising tumor-specific cytotoxicity and low hemolytic activity. Remarkably, the cytotoxicity induced by BOPs triggers the release of DAMPs, particularly HMGB1, IFN-β and ATP, by dying cells, persisting longer than the cytotoxicity of conventional chemotherapeutic agents such as irinotecan and daunorubicin. An in vivo assay in nude mice showed an encouraging 20% inhibition of tumor grafting and growth in a pancreatic cancer model by BOP9.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chiara Falciani
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| |
Collapse
|
9
|
Li XQ, Yamazaki T, He T, Alam MM, Liu J, Trivett AL, Sveinbjørnsson B, Rekdal Ø, Galluzzi L, Oppenheim JJ, Yang D. LTX-315 triggers anticancer immunity by inducing MyD88-dependent maturation of dendritic cells. Front Immunol 2024; 15:1332922. [PMID: 38545099 PMCID: PMC10967226 DOI: 10.3389/fimmu.2024.1332922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/22/2024] [Indexed: 04/10/2024] Open
Abstract
LTX-315 is a synthetic cationic oncolytic peptide with potent anticancer activity but limited toxicity for non-malignant cells. LTX-315 induces both immunogenic tumor cell death and generation of tumor-specific immune responses in multiple experimental tumor models. Given the central role of dendritic cell (DC) maturation in the induction of antigen-specific immunity, we investigated the effect of LTX-315 treatment on the maturation of tumor-infiltrating DCs (TiDCs) and the generation of anti-melanoma immunity. We found that LTX-315 treatment induces the maturation of DCs, both indirectly through the release of cancer cell-derived damage-associated molecular patterns (DAMPs)/alarmins and nucleic acids (DNA and RNA) capable of triggering distinct Toll-like receptor (TLR) signaling, and, directly by activating TLR7. The latter results in the ignition of multiple intracellular signaling pathways that promotes DC maturation, including NF-κB, mitogen activated protein kinases (MAPKs), and inflammasome signaling, as well as increased type 1 interferon production. Critically, the effects of LTX-315 on DCs the consequent promotion of anti-melanoma immunity depend on the cytosolic signal transducer myeloid differentiation response gene 88 (MyD88). These results cast light on the mechanisms by which LTX-315 induces DC maturation and hence elicits anticancer immunity, with important implications for the use of LTX-315 as an anticancer immunotherapeutic.
Collapse
Affiliation(s)
- Xiao-Qing Li
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin Medical University, Tianjin, China
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Tianzhen He
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Md Masud Alam
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Jia Liu
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Anna L. Trivett
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | | | | | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
- Sandra and Edward Meyer Cancer Center, New York, NY, United States
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States
| | - Joost J. Oppenheim
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - De Yang
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| |
Collapse
|