1
|
Degavre C, Lepez A, Ibanez S, François C, Głowacka K, Guilbaud C, Laloux-Morris F, Esfahani H, Brusa D, Bouzin C, Feron O. In situ endoscopic photodynamic therapy combined with immature DC vaccination induces a robust T cell response against peritoneal carcinomatosis. J Immunother Cancer 2024; 12:e009752. [PMID: 39500528 PMCID: PMC11552574 DOI: 10.1136/jitc-2024-009752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2024] [Indexed: 11/13/2024] Open
Abstract
BACKGROUND Immunogenic cell death (ICD) and ferroptosis have recently emerged as key factors in the anticancer immune response. Among the treatments able to induce ICD and the associated release of danger signals is photodynamic therapy (PDT). Ferroptosis for its part results from lipid peroxidation and is induced by CD8+ T cells to kill nearby cancer cells on IFN-γ production. We aimed to combine the two concepts, that is, to evaluate whether the strong pro-oxidant effects of PDT may promote ferroptosis and antigen release and to develop a procedure for in situ PDT to prepare the soil for highly endocytotic immature dendritic cell (iDC) adoptive transfer. This approach was implemented for managing peritoneal carcinomatosis, a lesion often associated with poor outcomes. METHODS We used three-dimensional (3D) heterotypic spheroids made of cancer cells, exposed them to a white light-activated OR141 photosensitizer (PS), and subsequently complexified them by adding iDC and naive lymphocytes. We next used a model of mouse peritoneal carcinomatosis and administered PDT using laparoscopy to locally induce photoactivation using the endoscope light. The immune response following adoptive transfer of iDC was tracked both in vivo and ex vivo using isolated immune cells from in situ vaccinated mice. RESULTS Cancer cells undergoing PDT-induced cell death significantly increased ICD markers and the infiltration of iDCs in spheroids, relying on ferroptosis. These actions induced the sequential activation of CD8+ and CD4+ T cells as revealed by a significant spheroid 3D structure deterioration and, remarkably, were not recapitulated by conventional ferroptosis inducer RSL3. Using LED light from an endoscope for in situ photoactivation of PS enabled us to apply the vaccination modality in mice with peritoneal tumors. Consecutive intraperitoneal injection of iDCs resulted in delayed tumor growth, increased survival rates, and prevented tumor relapse on rechallenge. CD8+ T cell response was supported by depletion experiments, nodal detection of early activated T cells, and ex vivo T cell-induced cytotoxicity toward spheroids. CONCLUSIONS The combination of in situ PDT locally delivered by an endoscope light and iDC administration induces a durable memory immune response against peritoneal carcinomatosis thereby opening new perspectives for the treatment of a life-threatening condition.
Collapse
Affiliation(s)
- Charline Degavre
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Anouk Lepez
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Sebastien Ibanez
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Clémence François
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Katarzyna Głowacka
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Céline Guilbaud
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Florine Laloux-Morris
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Hrag Esfahani
- IPHY Platform, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Davide Brusa
- CytoFlux-Flow Cytometry and Cell Sorting Platform, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Caroline Bouzin
- Imaging Platform 2IP, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Olivier Feron
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WEL Research Institute, Wavre, Belgium
| |
Collapse
|
2
|
Canti G, Rapozzi V. PDT and antitumor immunity: the beginnings of the story. Photochem Photobiol Sci 2024:10.1007/s43630-024-00627-1. [PMID: 39235681 DOI: 10.1007/s43630-024-00627-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/23/2024] [Indexed: 09/06/2024]
Abstract
This mini-review reports a brief description of the first experiments conducted by Canti's group on the role of photodynamic therapy in generating immunity against cancer. It highlights for the first time the effective role of PDT in the induction of anti-tumor T lymphocytes and shows that this effect is tumor-specific. It has also been reported how this adoptive immunity can improve the efficacy of chemotherapy. These studies have helped to open an important new field of scientific research on the role of PDT-generated immunity and to stimulate today's important new pre-clinical approaches.
Collapse
Affiliation(s)
- Gianfranco Canti
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20129, Milan, Italy.
| | | |
Collapse
|
3
|
Body N, Lefebvre C, Eeckhout S, Léonard AS, Troian-Gautier L, Hermans S, Riant O. Structure-Activity Relationship of Benzophenazine Derivatives for Homogeneous and Heterogenized Photooxygenation Catalysis. Chemistry 2024; 30:e202400242. [PMID: 38805006 DOI: 10.1002/chem.202400242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 05/29/2024]
Abstract
Singlet oxygen is a powerful oxidant used in various applications, such as organic synthesis, medicine, and environmental remediation. Organic and inorganic photosensitizers are commonly used to generate this reactive species through energy transfer with the triplet ground state of oxygen. We describe here a series of novel benzophenazine derivatives as a promising class of photosensitizers for singlet oxygen photosensitization. In this study, we investigated the structure-activity relationship of these benzophenazine derivatives. Akin to a molecular compass, the southern fragment was first functionalized with either aromatic tertiary amines, alkyl tertiary amines, aromatic sulfur groups, alkyl sulfur groups, or cyclic ethers. Enhanced photophysical properties (in terms of triplet excited-state lifetime, absorption wavelength, triplet state energy, and O2 quenching capabilities) were obtained with cyclic ether and sulfur groups. Conversely, the presence of an amine moiety was detrimental to the photocatalysts. The western and northern fragments were also investigated and slightly undesirable to negligible changes in photophysical properties were observed. The most promising candidate was then immobilized on silica nanoparticles and its photoactivity was evaluated in the citronellol photooxidation reaction. A high NMR yield of 97 % in desired product was obtained, with only a slight decrease over several recycling runs (85 % in the fourth run). These results provide insights into the design of efficient photosensitizers for singlet oxygen generation and the development of heterogeneous systems.
Collapse
Affiliation(s)
- Nathalie Body
- Université catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Place Louis Pasteur 1, bte L4.01.02, 1348, Louvain-la-Neuve, Belgium
| | - Corentin Lefebvre
- Université catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Place Louis Pasteur 1, bte L4.01.02, 1348, Louvain-la-Neuve, Belgium
| | - Sarah Eeckhout
- Université catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Place Louis Pasteur 1, bte L4.01.02, 1348, Louvain-la-Neuve, Belgium
| | - Anne-Sophie Léonard
- Université catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Place Louis Pasteur 1, bte L4.01.02, 1348, Louvain-la-Neuve, Belgium
| | - Ludovic Troian-Gautier
- Université catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Place Louis Pasteur 1, bte L4.01.02, 1348, Louvain-la-Neuve, Belgium
- Wel Research Institute, Avenue Pasteur 6, 1300, Wavre, Belgium
| | - Sophie Hermans
- Université catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Place Louis Pasteur 1, bte L4.01.02, 1348, Louvain-la-Neuve, Belgium
| | - Olivier Riant
- Université catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Place Louis Pasteur 1, bte L4.01.02, 1348, Louvain-la-Neuve, Belgium
| |
Collapse
|
4
|
Jiang M, Zhang GH, Yu Y, Zhao YH, Liu J, Zeng Q, Feng MY, Ye F, Xiong DS, Wang L, Zhang YN, Yu L, Wei JJ, He LB, Zhi W, Du XR, Li NJ, Han CL, Yan HQ, Zhou ZT, Miao YB, Wang W, Liu WX. De novo design of a nanoregulator for the dynamic restoration of ovarian tissue in cryopreservation and transplantation. J Nanobiotechnology 2024; 22:330. [PMID: 38862987 PMCID: PMC11167790 DOI: 10.1186/s12951-024-02602-5] [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: 03/27/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
The cryopreservation and transplantation of ovarian tissue underscore its paramount importance in safeguarding reproductive capacity and ameliorating reproductive disorders. However, challenges persist in ovarian tissue cryopreservation and transplantation (OTC-T), including the risk of tissue damage and dysfunction. Consequently, there has been a compelling exploration into the realm of nanoregulators to refine and enhance these procedures. This review embarks on a meticulous examination of the intricate anatomical structure of the ovary and its microenvironment, thereby establishing a robust groundwork for the development of nanomodulators. It systematically categorizes nanoregulators and delves deeply into their functions and mechanisms, meticulously tailored for optimizing ovarian tissue cryopreservation and transplantation. Furthermore, the review imparts valuable insights into the practical applications and obstacles encountered in clinical settings associated with OTC-T. Moreover, the review advocates for the utilization of microbially derived nanomodulators as a potent therapeutic intervention in ovarian tissue cryopreservation. The progression of these approaches holds the promise of seamlessly integrating nanoregulators into OTC-T practices, thereby heralding a new era of expansive applications and auspicious prospects in this pivotal domain.
Collapse
Affiliation(s)
- Min Jiang
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Guo-Hui Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Yuan Yu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yu-Hong Zhao
- School of Clinical Laboratory Medicine, Chengdu Medical College, Chengdu, 610083, China
| | - Jun Liu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Qin Zeng
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Meng-Yue Feng
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Fei Ye
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Dong-Sheng Xiong
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Li Wang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Ya-Nan Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Ling Yu
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Jia-Jing Wei
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Li-Bing He
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Weiwei Zhi
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Xin-Rong Du
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ning-Jing Li
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Chang-Li Han
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - He-Qiu Yan
- School of Clinical Laboratory Medicine, Chengdu Medical College, Chengdu, 610083, China
| | - Zhuo-Ting Zhou
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| | - Wen Wang
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| | - Wei-Xin Liu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China.
| |
Collapse
|
5
|
Thiruppathi J, Vijayan V, Park IK, Lee SE, Rhee JH. Enhancing cancer immunotherapy with photodynamic therapy and nanoparticle: making tumor microenvironment hotter to make immunotherapeutic work better. Front Immunol 2024; 15:1375767. [PMID: 38646546 PMCID: PMC11026591 DOI: 10.3389/fimmu.2024.1375767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Cancer immunotherapy has made tremendous advancements in treating various malignancies. The biggest hurdle to successful immunotherapy would be the immunosuppressive tumor microenvironment (TME) and low immunogenicity of cancer cells. To make immunotherapy successful, the 'cold' TME must be converted to 'hot' immunostimulatory status to activate residual host immune responses. To this end, the immunosuppressive equilibrium in TME should be broken, and immunogenic cancer cell death ought to be induced to stimulate tumor-killing immune cells appropriately. Photodynamic therapy (PDT) is an efficient way of inducing immunogenic cell death (ICD) of cancer cells and disrupting immune-restrictive tumor tissues. PDT would trigger a chain reaction that would make the TME 'hot' and have ICD-induced tumor antigens presented to immune cells. In principle, the strategic combination of PDT and immunotherapy would synergize to enhance therapeutic outcomes in many intractable tumors. Novel technologies employing nanocarriers were developed to deliver photosensitizers and immunotherapeutic to TME efficiently. New-generation nanomedicines have been developed for PDT immunotherapy in recent years, which will accelerate clinical applications.
Collapse
Affiliation(s)
- Jayalakshmi Thiruppathi
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Combinatorial Tumor Immunotherapy Medical Research Center (MRC), Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
| | - Veena Vijayan
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - In-Kyu Park
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- Department of Radiology, Biomolecular Theranostics (BiT) Laboratory, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Joon Haeng Rhee
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Combinatorial Tumor Immunotherapy Medical Research Center (MRC), Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea
| |
Collapse
|
6
|
Fan W, Tang J, Tang S, Lin Z, Li M, Zhang Z, Wu D. Bibliometric analysis of photodynamic therapy and immune response from 1989-2023. Front Pharmacol 2024; 15:1299253. [PMID: 38288443 PMCID: PMC10822948 DOI: 10.3389/fphar.2024.1299253] [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: 09/22/2023] [Accepted: 01/03/2024] [Indexed: 01/31/2024] Open
Abstract
Objective: Photodynamic therapy (PDT) is a minimally invasive treatment approach for precancerous and cancerous lesions, known for its ability to activate the host immune response. This study conducted a bibliometric analysis to identify the research trends and hotspots related to the immune response in PDT. Methods: We analyzed articles and reviews published from 1989 to 2023, retrieved from the Web of Science database. Using Citespace and VOSviewer, we visualized the distribution patterns of these studies in time and space. Results: The analysis revealed a substantial increase in the number of publications on PDT-related immune response since 1989. A total of 1,688 articles from 1,701 institutions were included in this analysis. Among thei nstitutions, the Chinese Academy of Sciences demonstrated exceptional productivity and a willingness to collaborate with others. Additionally, 8,567 authors contributed to the field, with Mladen Korbelik, Michael R. Hamblin, and Wei R. Chen being the most prolific contributors. The current research focus revolves around novel strategies to enhance antitumor immunity in PDT, including PDT-based dendritic cell vaccines, combination therapies with immune checkpoint inhibitors (ICIs), and the use of nanoparticles for photosensitizer delivery. Furthermore, genes such as CD8A, TNF, CD4, IFNG, CD274, IL6, IL10, CALR, HMGB1, and CTLA4 have been evaluated in the context of PDT-related immunity. Conclusion: PDT not only achieves tumor ablation but also stimulates the immune response, bolstering antitumor immunity. This study highlights the emerging hotspots in PDT-related immune response research and provides valuable insights for future investigations aimed at further enhancing antitumor immunity.
Collapse
Affiliation(s)
- Wanting Fan
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Jianming Tang
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Su Tang
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Zhengshen Lin
- Department of Stomatology, The People’s Hospital of Baoan Shenzhen, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Mohan Li
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Zheng Zhang
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| | - Donglei Wu
- Department of Stomatology, Shenzhen People’s Hospital, Shenzhen, China
| |
Collapse
|
7
|
Calvillo-Rodríguez KM, Lorenzo-Anota HY, Rodríguez-Padilla C, Martínez-Torres AC, Scott-Algara D. Immunotherapies inducing immunogenic cell death in cancer: insight of the innate immune system. Front Immunol 2023; 14:1294434. [PMID: 38077402 PMCID: PMC10701401 DOI: 10.3389/fimmu.2023.1294434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/01/2023] [Indexed: 12/18/2023] Open
Abstract
Cancer immunotherapies include monoclonal antibodies, cytokines, oncolytic viruses, cellular therapies, and other biological and synthetic immunomodulators. These are traditionally studied for their effect on the immune system's role in eliminating cancer cells. However, some of these therapies have the unique ability to directly induce cytotoxicity in cancer cells by inducing immunogenic cell death (ICD). Unlike general immune stimulation, ICD triggers specific therapy-induced cell death pathways, based on the release of damage-associated molecular patterns (DAMPs) from dying tumour cells. These activate innate pattern recognition receptors (PRRs) and subsequent adaptive immune responses, offering the promise of sustained anticancer drug efficacy and durable antitumour immune memory. Exploring how onco-immunotherapies can trigger ICD, enhances our understanding of their mechanisms and potential for combination strategies. This review explores the complexities of these immunotherapeutic approaches that induce ICD, highlighting their implications for the innate immune system, addressing challenges in cancer treatment, and emphasising the pivotal role of ICD in contemporary cancer research.
Collapse
Affiliation(s)
- Kenny Misael Calvillo-Rodríguez
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Helen Yarimet Lorenzo-Anota
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
- The Institute for Obesity Research, Tecnológico de Monterrey, Monterrey, NL, Mexico
| | - Cristina Rodríguez-Padilla
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Ana Carolina Martínez-Torres
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Daniel Scott-Algara
- Département d'Immunologie, Unité de Biologie Cellulaire des Lymphocytes, Pasteur Institute, Paris, France
| |
Collapse
|
8
|
Ibarra AMC, Aguiar EMG, Ferreira CBR, Siqueira JM, Corrêa L, Nunes FD, Franco ALDS, Cecatto RB, Hamblin MR, Rodrigues MFSD. Photodynamic therapy in cancer stem cells - state of the art. Lasers Med Sci 2023; 38:251. [PMID: 37919479 DOI: 10.1007/s10103-023-03911-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/14/2023] [Indexed: 11/04/2023]
Abstract
Despite significant efforts to control cancer progression and to improve oncology treatment outcomes, recurrence and tumor resistance are frequently observed in cancer patients. These problems are partly related to the presence of cancer stem cells (CSCs). Photodynamic therapy (PDT) has been developed as a therapeutic approach for solid tumors; however, it remains unclear how this therapy can affect CSCs. In this review, we focus on the effects of PDT on CSCs and the possible changes in the CSC population after PDT exposure. Tumor response to PDT varies according to the photosensitizer and light parameters employed, but most studies have reported the successful elimination of CSCs after PDT. However, some studies have reported that CSCs were more resistant to PDT than non-CSCs due to the increased efflux of photosensitizer molecules and the action of autophagy. Additionally, using different PDT approaches to target the CSCs resulted in increased sensitivity, reduction of sphere formation, invasiveness, stem cell phenotype, and improved response to chemotherapy. Lastly, although mainly limited to in vitro studies, PDT, combined with targeted therapies and/or chemotherapy, could successfully target CSCs in different solid tumors and promote the reduction of stemness, suggesting a promising therapeutic approach requiring evaluation in robust pre-clinical studies.
Collapse
Affiliation(s)
- Ana Melissa C Ibarra
- Postgraduate Program in Biophotonics Applied to Health Sciences, Nove de Julho University - UNINOVE, São Paulo, Brazil
| | | | - Cássia B R Ferreira
- Postgraduate Program in Biophotonics Applied to Health Sciences, Nove de Julho University - UNINOVE, São Paulo, Brazil
| | | | - Luciana Corrêa
- School of Dentistry, University of São Paulo - FOUSP, São Paulo, Brazil
| | - Fabio D Nunes
- School of Dentistry, University of São Paulo - FOUSP, São Paulo, Brazil
| | | | - Rebeca B Cecatto
- Postgraduate Program in Biophotonics Applied to Health Sciences, Nove de Julho University - UNINOVE, São Paulo, Brazil
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Johannesburg, South Africa
| | - Maria Fernanda S D Rodrigues
- Postgraduate Program in Biophotonics Applied to Health Sciences, Nove de Julho University - UNINOVE, São Paulo, Brazil.
| |
Collapse
|
9
|
Jia J, Wu X, Long G, Yu J, He W, Zhang H, Wang D, Ye Z, Tian J. Revolutionizing cancer treatment: nanotechnology-enabled photodynamic therapy and immunotherapy with advanced photosensitizers. Front Immunol 2023; 14:1219785. [PMID: 37860012 PMCID: PMC10582717 DOI: 10.3389/fimmu.2023.1219785] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/20/2023] [Indexed: 10/21/2023] Open
Abstract
Nanotechnology-enhanced photodynamic therapy (PDT) and immunotherapy are emerging as exciting cancer therapeutic methods with significant potential for improving patient outcomes. By combining these approaches, synergistic effects have been observed in preclinical studies, resulting in enhanced immune responses to cancer and the capacity to conquer the immunosuppressive tumor microenvironment (TME). Despite challenges such as addressing treatment limitations and developing personalized cancer treatment strategies, the integration of nanotechnology-enabled PDT and immunotherapy, along with advanced photosensitizers (PSs), represents an exciting new avenue in cancer treatment. Continued research, development, and collaboration among researchers, clinicians, and regulatory agencies are crucial for further advancements and the successful implementation of these promising therapies, ultimately benefiting cancer patients worldwide.
Collapse
Affiliation(s)
- Jiedong Jia
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Xue Wu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Gongwei Long
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jie Yu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Wei He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiping Zhang
- Institute of Reproduction Health Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongwen Wang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Tian
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- Department of Urology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical, Beijing, China
| |
Collapse
|
10
|
Yu S, Xiao H, Ma L, Zhang J, Zhang J. Reinforcing the immunogenic cell death to enhance cancer immunotherapy efficacy. Biochim Biophys Acta Rev Cancer 2023; 1878:188946. [PMID: 37385565 DOI: 10.1016/j.bbcan.2023.188946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Immunogenic cell death (ICD) has been a revolutionary modality in cancer treatment since it kills primary tumors and prevents recurrent malignancy simultaneously. ICD represents a particular form of cancer cell death accompanied by production of damage-associated molecular patterns (DAMPs) that can be recognized by pattern recognition receptors (PRRs), which enhances infiltration of effector T cells and potentiates antitumor immune responses. Various treatment methods can elicit ICD involving chemo- and radio-therapy, phototherapy and nanotechnology to efficiently convert dead cancer cells into vaccines and trigger the antigen-specific immune responses. Nevertheless, the efficacy of ICD-induced therapies is restrained due to low accumulation in the tumor sites and damage of normal tissues. Thus, researchers have been devoted to overcoming these problems with novel materials and strategies. In this review, current knowledge on different ICD modalities, various ICD inducers, development and application of novel ICD-inducing strategies are summarized. Moreover, the prospects and challenges are briefly outlined to provide reference for future design of novel immunotherapy based on ICD effect.
Collapse
Affiliation(s)
- Sihui Yu
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Hongyang Xiao
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Li Ma
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Jiarong Zhang
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
11
|
Huis in ‘t Veld RV, Heuts J, Ma S, Cruz LJ, Ossendorp FA, Jager MJ. Current Challenges and Opportunities of Photodynamic Therapy against Cancer. Pharmaceutics 2023; 15:pharmaceutics15020330. [PMID: 36839652 PMCID: PMC9965442 DOI: 10.3390/pharmaceutics15020330] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Photodynamic therapy (PDT) is an established, minimally invasive treatment for specific types of cancer. During PDT, reactive oxygen species (ROS) are generated that ultimately induce cell death and disruption of the tumor area. Moreover, PDT can result in damage to the tumor vasculature and induce the release and/or exposure of damage-associated molecular patterns (DAMPs) that may initiate an antitumor immune response. However, there are currently several challenges of PDT that limit its widespread application for certain indications in the clinic. METHODS A literature study was conducted to comprehensively discuss these challenges and to identify opportunities for improvement. RESULTS The most notable challenges of PDT and opportunities to improve them have been identified and discussed. CONCLUSIONS The recent efforts to improve the current challenges of PDT are promising, most notably those that focus on enhancing immune responses initiated by the treatment. The application of these improvements has the potential to enhance the antitumor efficacy of PDT, thereby broadening its potential application in the clinic.
Collapse
Affiliation(s)
- Ruben V. Huis in ‘t Veld
- Department of Ophthalmology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
- Department of Radiology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
- Correspondence:
| | - Jeroen Heuts
- Department of Immunology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Sen Ma
- Department of Ophthalmology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Luis J. Cruz
- Department of Radiology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Ferry A. Ossendorp
- Department of Immunology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| |
Collapse
|
12
|
Sasaki M, Tanaka M, Kojima Y, Nishie H, Shimura T, Kubota E, Kataoka H. Anti-tumor immunity enhancement by photodynamic therapy with talaporfin sodium and anti-programmed death 1 antibody. Mol Ther Oncolytics 2023; 28:118-131. [PMID: 36726602 PMCID: PMC9867957 DOI: 10.1016/j.omto.2022.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 12/31/2022] [Indexed: 01/04/2023] Open
Abstract
Photodynamic therapy (PDT) is a relatively non-invasive anti-cancer therapy that employs a photosensitizer with a specific wavelength of light irradiation. PDT induces direct cell killing and enhancement effects on tumor immunity, but its underlying mechanism remains unknown. Here, we perform a basic analysis of the anti-tumor effect of talaporfin sodium (TS)-PDT as well as its synergism with the immune checkpoint inhibitor anti-programmed death 1 (anti-PD-1) antibody. We estimate the cell death mechanism induced by TS-PDT and the induction of damage-associated molecular patterns (DAMPs) by TS-PDT in vitro. We establish a syngeneic mouse model of bilateral flank tumors and verify the enhancement of the abscopal effect on the non-irradiated side. TS-PDT induced apoptosis, necrosis, and autophagy-associated cell death in vitro. TS-PDT induced the release and/or expression of DAMPs in vitro. Tumor growth was inhibited in the TS-PDT and anti-PD-1 antibody combination group compared with other single-treatment or non-treatment groups in vivo. In summary, TS-PDT induces the release and/or expression of DAMPs, indicating that it activates innate immunity. PD-1 blockage enhances the anti-tumor immunity induced by TS-PDT. Thus, our results demonstrate that the combination of TS-PDT and anti-PD-1 antibody can potentially be used for anti-tumor therapy.
Collapse
Affiliation(s)
- Makiko Sasaki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Science, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Mamoru Tanaka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Science, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan,Corresponding author: Mamoru Tanaka, MD, PhD, Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| | - Yuki Kojima
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Science, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Hirotada Nishie
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Science, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Science, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Eiji Kubota
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Science, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Science, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
13
|
Moghassemi S, Dadashzadeh A, Camboni A, Feron O, Azevedo RB, Amorim CA. Ex vivo purging of cancer cells from ovarian tissue using photodynamic therapy: a novel strategy to restore fertility in leukemia patients. Hum Reprod Open 2023; 2023:hoad005. [PMID: 36895885 PMCID: PMC9991580 DOI: 10.1093/hropen/hoad005] [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: 10/14/2022] [Revised: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
STUDY QUESTION Is it possible to purge leukemia cells from ovarian tissue (OT) fragments before transplantation? SUMMARY ANSWER Our photodynamic therapy (PDT) approach has been shown to efficiently destroy leukemia cells from tumor-infiltration mimicking models (TIMs), indicating the feasibility of this technique to purge OT samples. WHAT IS KNOWN ALREADY Autotransplantation of cryopreserved OT is the most suitable option to preserve fertility for prepubertal girls and women who require immediate cancer treatment. Up until now, more than 200 live births have already been reported after OT cryopreservation and transplantation. Leukemia is the 12th most common cancer in Europe among prepubertal girls and women of reproductive age and in 2020, the estimated number of new leukemia cases was higher than 33 000 in girls between 0 and 19 years old. Unfortunately, once their health has been restored, autotransplantation of cryopreserved OT for leukemia patients is not advised due to the high risk of transferring malignant cells back to the patient leading to leukemia recurrence. STUDY DESIGN SIZE DURATION To safely transplant the OT from leukemia patients and restore their fertility, our goal was to develop a PDT strategy to eliminate leukemia ex vivo. To this end, we designed OR141-loaded niosomes (ORN) to create the most effective formulation for ex vivo purging of acute myelogenous leukemia cells from OT fragments (n = 4). Moreover, to ensure that such treatments are not harmful to follicle survival and development so they can be deemed a potential fertility restoration alternative, the effect of the ORN-based PDT purging procedure on follicles was assessed after xenografting the photodynamic-treated OT in SCID mice (n = 5). The work was carried out between September 2020 and April 2022 at the Catholic University of Louvain. PARTICIPANTS/MATERIALS SETTING METHODS After establishing the best ORN formulation, our PDT approach was used to eradicate HL60 cells from ex vivo TIMs prepared by microinjection of a cancer cell suspension into OT fragments. The purging efficiency was analyzed by droplet digital polymerase chain reaction and immunohistochemical analyses. Additionally, we evaluated the effect of ORN-based PDT on follicle density, survival and development, and tissue quality in terms of fibrotic areas and vascularization after 7-day xenotransplantation to immunodeficient mice. MAIN RESULTS AND THE ROLE OF CHANCE The ex vivo purging of TIMs demonstrated that our PDT strategy could selectively eradicate the malignant cells from tissue fragments without affecting OT normal cells, as evidenced by PCR and immunohistochemical analysis. Regarding the effect of our PDT approach on follicle population and OT quality, our results after xenotransplantation revealed no significant difference between the follicle density of control (non-treated, grafted OT) and PDT-treated groups (2.38 ± 0.63 and 3.21 ± 1.94 morphologically normal follicles/mm2, respectively). In addition, our findings showed that the control and PDT-treated OT could be equally vascularized (7.65 ± 1.45% and 9.89 ± 2.21%, respectively). Similarly, the proportions of fibrotic area did not differ between the control (15.96 ± 5.94%) and PDT-treated groups (13.32 ± 3.05%). LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION This study did not use OT fragments from leukemia patients, but TIMs created after injection of HL60 cells into OT from healthy patients. Therefore, while the results are promising, whether our PDT approach will be equally successful in eliminating malignant cells from leukemia patients remains to be assessed. WIDER IMPLICATIONS OF THE FINDINGS Our results showed that the purging procedure causes no significant impairment effect on follicle development and tissue quality, suggesting that our novel PDT procedure could be a promising strategy to destroy leukemia cells in fragments of OT, allowing safe transplantation in cancer survivors. STUDY FUNDING/COMPETING INTERESTS This study was supported by grants from the Fonds National de la Recherche Scientifique de Belgique (FNRS-PDR Convention grant number T.0004.20 awarded to C.A.A.); Fondation Louvain (awarded to C.A.A.; a Ph.D. scholarship awarded to S.M., as part of a legacy from Mr Frans Heyes, and a Ph.D. scholarship awarded to A.D. as part of a legacy from Mrs. Ilse Schirmer); and Foundation Against Cancer (grant number 2018-042 awarded to A.C.). The authors declare no competing interests.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Alessandra Camboni
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.,Service d'Anatomie Pathologique, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Olivier Feron
- Pôle de Pharmacologie et Thérapeutique, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Ricardo Bentes Azevedo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília DF, Brazil
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
14
|
Kong C, Chen X. Combined Photodynamic and Photothermal Therapy and Immunotherapy for Cancer Treatment: A Review. Int J Nanomedicine 2022; 17:6427-6446. [PMID: 36540374 PMCID: PMC9760263 DOI: 10.2147/ijn.s388996] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/03/2022] [Indexed: 12/23/2022] Open
Abstract
Photoactivation therapy based on photodynamic therapy (PDT) and photothermal therapy (PTT) has been identified as a tumour ablation modality for numerous cancer indications, with photosensitisers and photothermal conversion agents playing important roles in the phototherapy process, especially in recent decades. In addition, the iteration of nanotechnology has strongly promoted the development of phototherapy in tumour treatment. PDT can increase the sensitivity of tumour cells to PTT by interfering with the tumour microenvironment, whereas the heat generated by PTT can increase blood flow, improve oxygen supply and enhance the PDT therapeutic effect. In addition, tumour cell debris generated by phototherapy can serve as tumour-associated antigens, evoking antitumor immune responses. In this review, the research progress of phototherapy, and its research effects in combination with immunotherapy on the treatment of tumours are mainly outlined, and issues that may need continued attention in the future are raised.
Collapse
Affiliation(s)
- Cunqing Kong
- Department of medical imaging center, central hospital affiliated to Shandong first medical university, Jinan, People’s Republic of China
| | - Xingcai Chen
- Department of Human Anatomy and Center for Genomics and Personalized Medicine, Nanning, People’s Republic of China,Correspondence: Xingcai Chen, Email
| |
Collapse
|
15
|
Moghassemi S, Dadashzadeh A, Camboni A, Feron O, Azevedo RB, Amorim CA. Photodynamic therapy using OR141-loaded nanovesicles for eradication of leukemic cells from ovarian tissue. Photodiagnosis Photodyn Ther 2022; 40:103139. [PMID: 36198387 DOI: 10.1016/j.pdpdt.2022.103139] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 12/14/2022]
Abstract
In 2020, the estimated number of new leukemia cases was higher than 30,000 in girls between 0 and 19 years old. Due to cancer treatment, some of these patients may lose both endocrine and reproductive functions. Transplantation of cryopreserved ovarian tissue is not advised after cancer remission because it has a high risk of reintroducing malignant cells in the patient, potentially leading to leukemia recurrence. To safely transplant the ovarian tissue from these patients and restore their fertility, our goal was to develop a photodynamic therapy (PDT) strategy to eliminate leukemia ex vivo. To this end, we designed, optimized, and characterized OR141-loaded niosomes (ORN) to develop the most effective formulation for ex vivo purging ovarian fragments from chronic myelogenous leukemia cells. After establishing the best ORN formulation, the PDT efficiency of optimized ORN was determined for human ovarian stromal cells and acute myeloid leukemia cell line (HL60). Blank niosomes treatment on ovarian stromal cells causes no significant toxicity, showing that the composition of the nanoparticle is not toxic. On the other hand, the in vitro studies showed that while ovarian stromal cells were still viable (82.04 ± 2.79%) after the treatment by 0.5 µM ORN, the same treatment yielded 95.43 ± 3.89% toxicity and cell death in the cancer cells. In conclusion, our results showed that our novel PDT procedure could be a promising strategy to destroy leukemia cells in ovarian tissue fragments allowing safe transplantation in cancer survivors.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Alessandra Camboni
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Service d'Anatomie Pathologique, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Olivier Feron
- Pôle de Pharmacologie et Thérapeutique, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Ricardo Bentes Azevedo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
16
|
Moghassemi S, Dadashzadeh A, de Azevedo RB, Amorim CA. Secure transplantation by tissue purging using photodynamic therapy to eradicate malignant cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2022; 234:112546. [PMID: 36029759 DOI: 10.1016/j.jphotobiol.2022.112546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/07/2022] [Accepted: 08/16/2022] [Indexed: 12/17/2022]
Abstract
The field of photodynamic therapy (PDT) for treating various malignant neoplasms has been given researchers' attention due to its ability to be a selective and minimally invasive cancer therapy strategy. The possibility of tumor cell infection and hence high recurrence rates in cancer patients tends to restrict autologous transplantation. So, the photodynamic tissue purging process, which consists of selective photoinactivation of the malignant cells in the graft, is defined as a compromising strategy to purify contaminated tissues before transplantation. In this strategy, the direct malignant cells' death results from the reactive oxygen species (ROS) generation through the activation of a photosensitizer (PS) by light exposure in the presence of oxygen. Since new PS generations can effectively penetrate the tissue, PDT could be an ideal ex vivo tissue purging protocol that eradicates cancer cells derived from various malignancies. The challenge is that the applied pharmacologic ex vivo tissue purging should efficiently induce tumor cells with minor influence on normal tissue cells. This review aims to provide an overview of the current status of the most effective PDT strategies and PS development concerning their potential application in ex vivo purging before hematopoietic stem cell or ovarian tissue transplantation.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Ricardo Bentes de Azevedo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília DF, Brazil
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
17
|
Chen C, Wu C, Yu J, Zhu X, Wu Y, Liu J, Zhang Y. Photodynamic-based combinatorial cancer therapy strategies: Tuning the properties of nanoplatform according to oncotherapy needs. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214495] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
18
|
Combinatorial Therapeutic Approaches with Nanomaterial-Based Photodynamic Cancer Therapy. Pharmaceutics 2022; 14:pharmaceutics14010120. [PMID: 35057015 PMCID: PMC8780767 DOI: 10.3390/pharmaceutics14010120] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/11/2021] [Accepted: 12/28/2021] [Indexed: 12/27/2022] Open
Abstract
Photodynamic therapy (PDT), in which a light source is used in combination with a photosensitizer to induce local cell death, has shown great promise in therapeutically targeting primary tumors with negligible toxicity and minimal invasiveness. However, numerous studies have shown that noninvasive PDT alone is not sufficient to completely ablate tumors in deep tissues, due to its inherent shortcomings. Therefore, depending on the characteristics and type of tumor, PDT can be combined with surgery, radiotherapy, immunomodulators, chemotherapy, and/or targeted therapy, preferably in a patient-tailored manner. Nanoparticles are attractive delivery vehicles that can overcome the shortcomings of traditional photosensitizers, as well as enable the codelivery of multiple therapeutic drugs in a spatiotemporally controlled manner. Nanotechnology-based combination strategies have provided inspiration to improve the anticancer effects of PDT. Here, we briefly introduce the mechanism of PDT and summarize the photosensitizers that have been tested preclinically for various cancer types and clinically approved for cancer treatment. Moreover, we discuss the current challenges facing the combination of PDT and multiple cancer treatment options, and we highlight the opportunities of nanoparticle-based PDT in cancer therapies.
Collapse
|
19
|
Understanding and improving cellular immunotherapies against cancer: From cell-manufacturing to tumor-immune models. Adv Drug Deliv Rev 2021; 179:114003. [PMID: 34653533 DOI: 10.1016/j.addr.2021.114003] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment (TME) is shaped by dynamic metabolic and immune interactions between precancerous and cancerous tumor cells and stromal cells like epithelial cells, fibroblasts, endothelial cells, and hematopoietically-derived immune cells. The metabolic states of the TME, including the hypoxic and acidic niches, influence the immunosuppressive phenotypes of the stromal and immune cells, which confers resistance to both host-mediated tumor killing and therapeutics. Numerous in vitro TME platforms for studying immunotherapies, including cell therapies, are being developed. However, we do not yet understand which immune and stromal components are most critical and how much model complexity is needed to answer specific questions. In addition, scalable sourcing and quality-control of appropriate TME cells for reproducibly manufacturing these platforms remain challenging. In this regard, lessons from the manufacturing of immunomodulatory cell therapies could provide helpful guidance. Although immune cell therapies have shown unprecedented results in hematological cancers and hold promise in solid tumors, their manufacture poses significant scale, cost, and quality control challenges. This review first provides an overview of the in vivo TME, discussing the most influential cell populations in the tumor-immune landscape. Next, we summarize current approaches for cell therapies against cancers and the relevant manufacturing platforms. We then evaluate current immune-tumor models of the TME and immunotherapies, highlighting the complexity, architecture, function, and cell sources. Finally, we present the technical and fundamental knowledge gaps in both cell manufacturing systems and immune-TME models that must be addressed to elucidate the interactions between endogenous tumor immunity and exogenous engineered immunity.
Collapse
|
20
|
Nkune NW, Simelane NWN, Montaseri H, Abrahamse H. Photodynamic Therapy-Mediated Immune Responses in Three-Dimensional Tumor Models. Int J Mol Sci 2021; 22:12618. [PMID: 34884424 PMCID: PMC8657498 DOI: 10.3390/ijms222312618] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising non-invasive phototherapeutic approach for cancer therapy that can eliminate local tumor cells and produce systemic antitumor immune responses. In recent years, significant efforts have been made in developing strategies to further investigate the immune mechanisms triggered by PDT. The majority of in vitro experimental models still rely on the two-dimensional (2D) cell cultures that do not mimic a three-dimensional (3D) cellular environment in the human body, such as cellular heterogeneity, nutrient gradient, growth mechanisms, and the interaction between cells as well as the extracellular matrix (ECM) and therapeutic resistance to anticancer treatments. In addition, in vivo animal studies are highly expensive and time consuming, which may also show physiological discrepancies between animals and humans. In this sense, there is growing interest in the utilization of 3D tumor models, since they precisely mimic different features of solid tumors. This review summarizes the characteristics and techniques for 3D tumor model generation. Furthermore, we provide an overview of innate and adaptive immune responses induced by PDT in several in vitro and in vivo tumor models. Future perspectives are highlighted for further enhancing PDT immune responses as well as ideal experimental models for antitumor immune response studies.
Collapse
Affiliation(s)
| | | | | | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Johannesburg 2028, South Africa; (N.W.N.); (N.W.N.S.); (H.M.)
| |
Collapse
|
21
|
Sun Z, Deng G, Peng X, Xu X, Liu L, Peng J, Ma Y, Zhang P, Wen A, Wang Y, Yang Z, Gong P, Jiang W, Cai L. Intelligent photothermal dendritic cells restart the cancer immunity cycle through enhanced immunogenic cell death. Biomaterials 2021; 279:121228. [PMID: 34717198 DOI: 10.1016/j.biomaterials.2021.121228] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/17/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DCs) play a pivotal role in initiating antigen-specific tumor immunity. However, the abnormal function of DCs owing to the immunosuppressive tumor microenvironment (TME) and the insufficient number of tumor infiltrating DCs could promote immune tolerance and tumor immune escape. Thus, there is great potential to employ DCs to induce efficient antitumor immunity. In this paper, we developed intelligent DCs (iDCs), which consist of nanoparticles loaded with photothermal agents (IR-797) and coated with a mature DC membrane. The DC cell membrane on the surface of iDCs preserves the ability to present antigens and prime T cells. The iDCs can also enter the lymph node and stimulate T cells. The activated T cells reduced the expression of heat shock proteins (HSPs) in tumor cells, rendering them more sensitive to heat stress. Subsequently, we used mild photothermal therapy (42-45 °C) to induce immunogenic cell death and contribute to a synergistic antitumor effect. iDCs as a refined and precise system in combination with DC-based immunotherapy and thermal therapy can be stored long-term and on a large scale, so they can be applied in many patients.
Collapse
Affiliation(s)
- Zhihong Sun
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Qindao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, 264000, PR China
| | - Guanjun Deng
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xinghua Peng
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiuli Xu
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jiaofeng Peng
- Instrumental Analysis Center of Shenzhen University, Shenzhen University, Shenzhen, 518055, China
| | - Yifan Ma
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; HRYZ Biotech Co., Shenzhen, 518057, PR China
| | - Pengfei Zhang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Austin Wen
- Pomona College, 333 N College Way, Claremont, CA, 91711, USA
| | - Yifan Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhaogang Yang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ping Gong
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
22
|
Aru B, Günay A, Demirel GY, Gürek AG, Atilla D. Evaluation of histone deacetylase inhibitor substituted zinc and indium phthalocyanines for chemo- and photodynamic therapy. RSC Adv 2021; 11:34963-34978. [PMID: 35494743 PMCID: PMC9042886 DOI: 10.1039/d1ra05404j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/14/2021] [Indexed: 01/03/2023] Open
Abstract
In this study, we synthesized and characterized 3-hydroxypyridin-2-thione (3-HPT) bearing zinc (ZnPc-1 and ZnPc-2) and indium (InPc-1 and InPc-2) phthalocyanine (Pc) derivatives, either non-peripherally or peripherally substituted as photosensitizer (PS) agents and evaluated their anti-cancer efficacy on two breast cancer cell lines, MDA-MB-231 and MCF-7 as well as a human endothelial cell line, HUVEC. Our results indicated different localization patterns between ZnPcs and InPcs in addition to enhanced effects on the mitochondrial network for InPcs. Moreover, peripheral or non-peripheral substitution of HDACi moieties altered cellular localization between ZnPc-1 and ZnPc-2, leading to increased IC50 values along with decreased anti-cancer activity for non-peripheral substitution. When considering the compounds' differential effects in vitro, our data indicates that further research is required to determine the ideal Pcs for anti-cancer PDT treatments since the core metals of the compounds have affected the cellular localization, and positioning of the chemotherapeutic residues may inhibit cellular penetrance.
Collapse
Affiliation(s)
- Başak Aru
- Faculty of Medicine, Immunology Department, Yeditepe University 34755 Ataşehir İstanbul Turkey
| | - Aysel Günay
- Department of Chemistry, Gebze Technical University 41400 Gebze Kocaeli Turkey
| | | | - Ayşe Gül Gürek
- Department of Chemistry, Gebze Technical University 41400 Gebze Kocaeli Turkey
| | - Devrim Atilla
- Department of Chemistry, Gebze Technical University 41400 Gebze Kocaeli Turkey
| |
Collapse
|
23
|
Gunaydin G, Gedik ME, Ayan S. Photodynamic Therapy for the Treatment and Diagnosis of Cancer-A Review of the Current Clinical Status. Front Chem 2021; 9:686303. [PMID: 34409014 PMCID: PMC8365093 DOI: 10.3389/fchem.2021.686303] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/19/2021] [Indexed: 12/24/2022] Open
Abstract
Photodynamic therapy (PDT) has been used as an anti-tumor treatment method for a long time and photosensitizers (PS) can be used in various types of tumors. Originally, light is an effective tool that has been used in the treatment of diseases for ages. The effects of combination of specific dyes with light illumination was demonstrated at the beginning of 20th century and novel PDT approaches have been developed ever since. Main strategies of current studies are to reduce off-target effects and improve pharmacokinetic properties. Given the high interest and vast literature about the topic, approval of PDT as the first drug/device combination by the FDA should come as no surprise. PDT consists of two stages of treatment, combining light energy with a PS in order to destruct tumor cells after activation by light. In general, PDT has fewer side effects and toxicity than chemotherapy and/or radiotherapy. In addition to the purpose of treatment, several types of PSs can be used for diagnostic purposes for tumors. Such approaches are called photodynamic diagnosis (PDD). In this Review, we provide a general overview of the clinical applications of PDT in cancer, including the diagnostic and therapeutic approaches. Assessment of PDT therapeutic efficacy in the clinic will be discussed, since identifying predictors to determine the response to treatment is crucial. In addition, examples of PDT in various types of tumors will be discussed. Furthermore, combination of PDT with other therapy modalities such as chemotherapy, radiotherapy, surgery and immunotherapy will be emphasized, since such approaches seem to be promising in terms of enhancing effectiveness against tumor. The combination of PDT with other treatments may yield better results than by single treatments. Moreover, the utilization of lower doses in a combination therapy setting may cause less side effects and better results than single therapy. A better understanding of the effectiveness of PDT in a combination setting in the clinic as well as the optimization of such complex multimodal treatments may expand the clinical applications of PDT.
Collapse
Affiliation(s)
- Gurcan Gunaydin
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - M. Emre Gedik
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Seylan Ayan
- Department of Chemistry, Bilkent University, Ankara, Turkey
| |
Collapse
|
24
|
Jin F, Liu D, Xu X, Ji J, Du Y. Nanomaterials-Based Photodynamic Therapy with Combined Treatment Improves Antitumor Efficacy Through Boosting Immunogenic Cell Death. Int J Nanomedicine 2021; 16:4693-4712. [PMID: 34267518 PMCID: PMC8275223 DOI: 10.2147/ijn.s314506] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Benefiting from the rapid development of nanotechnology, photodynamic therapy (PDT) is arising as a novel non-invasive clinical treatment for specific cancers, which exerts direct efficacy in destroying primary tumors by generating excessive cytotoxic reactive oxygen species (ROS). Notably, PDT-induced cell death is related to T cell-mediated antitumor immune responses through induction of immunogenic cell death (ICD). However, ICD elicited via PDT is not strong enough and is limited by immunosuppressive tumor microenvironment (ITM). Therefore, it is necessary to improve PDT efficacy through enhancing ICD with the combination of synergistic tumor therapies. Herein, the recent progress of nanomaterials-based PDT combined with chemotherapy, photothermal therapy, radiotherapy, and immunotherapy, employing ICD-boosted treatments is reviewed. An outlook about the future application in clinics of nanomaterials-based PDT strategies is also mentioned.
Collapse
Affiliation(s)
- Feiyang Jin
- Institute of Pharmaceutics, College of Pharmaceutics Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Di Liu
- Institute of Pharmaceutics, College of Pharmaceutics Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Xiaoling Xu
- Institute of Pharmaceutics, College of Pharmaceutics Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Jiansong Ji
- Department of Radiology, Lishui Hospital of Zhejiang University, Lishui, 323000, People's Republic of China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutics Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| |
Collapse
|
25
|
Current Prospects for Treatment of Solid Tumors via Photodynamic, Photothermal, or Ionizing Radiation Therapies Combined with Immune Checkpoint Inhibition (A Review). Pharmaceuticals (Basel) 2021; 14:ph14050447. [PMID: 34068491 PMCID: PMC8151935 DOI: 10.3390/ph14050447] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/21/2022] Open
Abstract
Photodynamic therapy (PDT) causes selective damage to tumor cells and vasculature and also triggers an anti-tumor immune response. The latter fact has prompted the exploration of PDT as an immune-stimulatory adjuvant. PDT is not the only cancer treatment that relies on electromagnetic energy to destroy cancer tissue. Ionizing radiation therapy (RT) and photothermal therapy (PTT) are two other treatment modalities that employ photons (with wavelengths either shorter or longer than PDT, respectively) and also cause tissue damage and immunomodulation. Research on the three modalities has occurred in different “silos”, with minimal interaction between the three topics. This is happening at a time when immune checkpoint inhibition (ICI), another focus of intense research and clinical development, has opened exciting possibilities for combining PDT, PTT, or RT with ICI to achieve improved therapeutic benefits. In this review, we surveyed the literature for studies that describe changes in anti-tumor immunity following the administration of PDT, PTT, and RT, including efforts to combine each modality with ICI. This information, collected all in one place, may make it easier to recognize similarities and differences and help to identify new mechanistic hypotheses toward the goal of achieving optimized combinations and tumor cures.
Collapse
|
26
|
Alzeibak R, Mishchenko TA, Shilyagina NY, Balalaeva IV, Vedunova MV, Krysko DV. Targeting immunogenic cancer cell death by photodynamic therapy: past, present and future. J Immunother Cancer 2021; 9:e001926. [PMID: 33431631 PMCID: PMC7802670 DOI: 10.1136/jitc-2020-001926] [Citation(s) in RCA: 290] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 12/18/2022] Open
Abstract
The past decade has witnessed major breakthroughs in cancer immunotherapy. This development has been largely motivated by cancer cell evasion of immunological control and consequent tumor resistance to conventional therapies. Immunogenic cell death (ICD) is considered one of the most promising ways to achieve total tumor cell elimination. It activates the T-cell adaptive immune response and results in the formation of long-term immunological memory. ICD can be triggered by many anticancer treatment modalities, including photodynamic therapy (PDT). In this review, we first discuss the role of PDT based on several classes of photosensitizers, including porphyrins and non-porphyrins, and critically evaluate their potential role in ICD induction. We emphasize the emerging trend of ICD induction by PDT in combination with nanotechnology, which represents third-generation photosensitizers and involves targeted induction of ICD by PDT. However, PDT also has some limitations, including the reduced efficiency of ICD induction in the hypoxic tumor microenvironment. Therefore, we critically evaluate strategies for overcoming this limitation, which is essential for increasing PDT efficiency. In the final part, we suggest several areas for future research for personalized cancer immunotherapy, including strategies based on oxygen-boosted PDT and nanoparticles. In conclusion, the insights from the last several years increasingly support the idea that PDT is a powerful strategy for inducing ICD in experimental cancer therapy. However, most studies have focused on mouse models, but it is necessary to validate this strategy in clinical settings, which will be a challenging research area in the future.
Collapse
Affiliation(s)
- Razan Alzeibak
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Tatiana A Mishchenko
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Natalia Y Shilyagina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Irina V Balalaeva
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Maria V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| | - Dmitri V Krysko
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
- Cell Death Investigation and Therapy Laboratory (CDIT), Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
27
|
Trempolec N, Degavre C, Doix B, Brusa D, Corbet C, Feron O. Acidosis-Induced TGF-β2 Production Promotes Lipid Droplet Formation in Dendritic Cells and Alters Their Potential to Support Anti-Mesothelioma T Cell Response. Cancers (Basel) 2020; 12:cancers12051284. [PMID: 32438640 PMCID: PMC7281762 DOI: 10.3390/cancers12051284] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 11/23/2022] Open
Abstract
For poorly immunogenic tumors such as mesothelioma there is an imperious need to understand why antigen-presenting cells such as dendritic cells (DCs) are not prone to supporting the anticancer T cell response. The tumor microenvironment (TME) is thought to be a major contributor to this DC dysfunction. We have reported that the acidic TME component promotes lipid droplet (LD) formation together with epithelial-to-mesenchymal transition in cancer cells through autocrine transforming growth factor-β2 (TGF-β2) signaling. Since TGF-β is also a master regulator of immune tolerance, we have here examined whether acidosis can impede immunostimulatory DC activity. We have found that exposure of mesothelioma cells to acidosis promotes TGF-β2 secretion, which in turn leads to LD accumulation and profound metabolic rewiring in DCs. We have further documented how DCs exposed to the mesothelioma acidic milieu make the anticancer vaccine less efficient in vivo, with a reduced extent of both DC migratory potential and T cell activation. Interestingly, inhibition of TGF-β2 signaling and diacylglycerol O-acyltransferase (DGAT), the last enzyme involved in triglyceride synthesis, led to a significant restoration of DC activity and anticancer immune response. In conclusion, our study has identified that acidic mesothelioma milieu drives DC dysfunction and altered T cell response through pharmacologically reversible TGF-β2-dependent mechanisms.
Collapse
Affiliation(s)
- Natalia Trempolec
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200 Brussels, Belgium; (N.T.); (C.D.); (B.D.); (C.C.)
| | - Charline Degavre
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200 Brussels, Belgium; (N.T.); (C.D.); (B.D.); (C.C.)
| | - Bastien Doix
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200 Brussels, Belgium; (N.T.); (C.D.); (B.D.); (C.C.)
| | - Davide Brusa
- Institut de Recherche Expérimentale et Clinique (IREC) Flow Cytometry Platform, UCLouvain, B-1200 Brussels, Belgium;
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200 Brussels, Belgium; (N.T.); (C.D.); (B.D.); (C.C.)
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, B-1200 Brussels, Belgium; (N.T.); (C.D.); (B.D.); (C.C.)
- Correspondence: ; Tel.: +32-2-7645264; Fax: +32-2-7645269
| |
Collapse
|