1
|
Kwon N, Weng H, Rajora MA, Zheng G. Activatable Photosensitizers: From Fundamental Principles to Advanced Designs. Angew Chem Int Ed Engl 2025; 64:e202423348. [PMID: 39899458 PMCID: PMC11976215 DOI: 10.1002/anie.202423348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
Photodynamic therapy (PDT) is a promising treatment that uses light to excite photosensitizers in target tissue, producing reactive oxygen species and localized cell death. It is recognized as a minimally invasive, clinically approved cancer therapy with additional preclinical applications in arthritis, atherosclerosis, and infection control. A hallmark of ideal PDT is delivering disease-specific cytotoxicity while sparing healthy tissue. However, conventional photosensitizers often suffer from non-specific photoactivation, causing off-target toxicity. Activatable photosensitizers (aPS) have emerged as more precise alternatives, offering controlled activation. Unlike traditional photosensitizers, they remain inert and photoinactive during circulation and off-target accumulation, minimizing collateral damage. These photosensitizers are designed to "turn on" in response to disease-specific biostimuli, enhancing therapeutic selectivity and reducing off-target effects. This review explores the principles of aPS, including quenching mechanisms stemming from activatable fluorescent probes and applied to activatable photosensitizers (RET, PeT, ICT, ACQ, AIE), as well as pathological biostimuli (pH, enzymes, redox conditions, cellular internalization), and bioresponsive constructs enabling quenching and activation. We also provide a critical assessment of unresolved challenges in aPS development, including limitations in targeting precision, selectivity under real-world conditions, and potential solutions to persistent issues (dual-lock, targeting moieties, biorthogonal chemistry and artificial receptors). Additionally, it provides an in-depth discussion of essential research design considerations needed to develop translationally relevant aPS with improved therapeutic outcomes and specificity.
Collapse
Affiliation(s)
- Nahyun Kwon
- Princess Margaret Cancer CentreUniversity Health Network101 College Street, PMCRT 5–354Toronto, ONM5G1L7Canada
| | - Hanyi Weng
- Princess Margaret Cancer CentreUniversity Health Network101 College Street, PMCRT 5–354Toronto, ONM5G1L7Canada
- Department of Medical BiophysicsUniversity of TorontoToronto, ONCanada
| | - Maneesha A. Rajora
- Princess Margaret Cancer CentreUniversity Health Network101 College Street, PMCRT 5–354Toronto, ONM5G1L7Canada
- Department of MedicineUniversity of TorontoToronto, ONCanada
| | - Gang Zheng
- Princess Margaret Cancer CentreUniversity Health Network101 College Street, PMCRT 5–354Toronto, ONM5G1L7Canada
- Department of Medical BiophysicsUniversity of TorontoToronto, ONCanada
| |
Collapse
|
2
|
Jose S, Sharma H, Insan J, Sharma K, Arora V, Puranapanda S, Dhamija S, Eid N, Menon MB. Kinase Inhibitor-Induced Cell-Type Specific Vacuole Formation in the Absence of Canonical ATG5-Dependent Autophagy Initiation Pathway. Mol Cell Biol 2025; 45:99-115. [PMID: 39895059 DOI: 10.1080/10985549.2025.2454421] [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: 02/29/2024] [Revised: 11/20/2024] [Accepted: 01/10/2025] [Indexed: 02/04/2025] Open
Abstract
Pyridinyl-imidazole class p38 MAPKα/β (MAPK14/MAPK11) inhibitors including SB202190 have been shown to induce cell-type specific defective autophagy resulting in micron-scale vacuole formation, cell death, and tumor suppression. We had earlier shown that this is an off-target effect of SB202190. Here we provide evidence that this vacuole formation is independent of ATG5-mediated canonical autophagosome initiation. While SB202190 interferes with autophagic flux in many cell lines parallel to vacuolation, autophagy-deficient DU-145 cells and CRISPR/Cas9 gene-edited ATG5-knockout A549 cells also undergo vacuolation upon SB202190 treatment. Late-endosomal GTPase RAB7 colocalizes with these compartments and RAB7 GTP-binding is essential for SB202190-induced vacuolation. A screen for modulators of SB202190-induced vacuolation revealed molecules including multi-kinase inhibitor sorafenib as inhibitors of vacuolation and sorafenib co-treatment enhanced cytotoxicity of SB202190. Moreover, VE-821, an ATR inhibitor was found to phenocopy the cell-type specific vacuolation response of SB202190. To identify the factors determining the cell-type specificity of vacuolation induced by SB-compounds and VE-821, we compared the transcriptomics data from vacuole-forming and non-vacuole-forming cancer cell lines and identified a gene expression signature that may define sensitivity of cells to these small-molecules. Further analyses using small molecule tools and the gene signature discovered here, could reveal novel mechanisms regulating this interesting anti-cancer phenotype.
Collapse
Affiliation(s)
- Susan Jose
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Himanshi Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Janki Insan
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Khushboo Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Varun Arora
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | | | - Sonam Dhamija
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Manoj B Menon
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
3
|
Shi Y, Zheng H, Wang T, Zhou S, Zhao S, Li M, Cao B. Targeting KRAS: from metabolic regulation to cancer treatment. Mol Cancer 2025; 24:9. [PMID: 39799325 PMCID: PMC11724471 DOI: 10.1186/s12943-024-02216-3] [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: 09/06/2024] [Accepted: 12/25/2024] [Indexed: 01/15/2025] Open
Abstract
The Kirsten rat sarcoma viral oncogene homolog (KRAS) protein plays a key pathogenic role in oncogenesis, cancer progression, and metastasis. Numerous studies have explored the role of metabolic alterations in KRAS-driven cancers, providing a scientific rationale for targeting metabolism in cancer treatment. The development of KRAS-specific inhibitors has also garnered considerable attention, partly due to the challenge of acquired treatment resistance. Here, we review the metabolic reprogramming of glucose, glutamine, and lipids regulated by oncogenic KRAS, with an emphasis on recent insights into the relationship between changes in metabolic mechanisms driven by KRAS mutant and related advances in targeted therapy. We also focus on advances in KRAS inhibitor discovery and related treatment strategies in colorectal, pancreatic, and non-small cell lung cancer, including current clinical trials. Therefore, this review provides an overview of the current understanding of metabolic mechanisms associated with KRAS mutation and related therapeutic strategies, aiming to facilitate the understanding of current challenges in KRAS-driven cancer and to support the investigation of therapeutic strategies.
Collapse
Affiliation(s)
- Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, China
| | - Huiling Zheng
- Department of Gastroenterology, Peking University Third Hospital, Beijing, 100191, China
| | - Tianzhen Wang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction (Peking University), Peking University Third Hospital, Ministry of Education, Beijing, 100191, China
| | - Shengpu Zhou
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, China
| | - Shiqing Zhao
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, China
| | - Mo Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction (Peking University), Peking University Third Hospital, Ministry of Education, Beijing, 100191, China.
| | - Baoshan Cao
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
4
|
Li Y, Feng Y, Chen D. Interfering Nuclear Protein Laminb1 Induces DNA Damage and Reduces Vemurafenib Resistance in Melanoma Cells In Vitro. Cancers (Basel) 2024; 16:4060. [PMID: 39682248 DOI: 10.3390/cancers16234060] [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: 10/31/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Drug resistance poses a substantial clinical challenge in melanoma treatment, yet the underlying mechanism remains elusive. Here, we report the novel role of laminB1, a nuclear structure protein, in regulating the response of BRAF-mutated melanoma cells to vemurafenib. RESULTS Our analysis of clinical samples and existing databases highlights the tight correlation between the laminB1 expression level and melanoma progression and prognosis. Notably, we observe that laminB1 expression is upregulated when BRAF-mutated melanoma cells develop resistance to vemurafenib. The knockdown of laminB1 substantially increases the sensitivity of melanoma cells to vemurafenib. Furthermore, we found laminB1 suppression increases cell apoptosis via the escalation of DNA damage in a vemurafenib-dose-dependent manner. Conversely, protective cell autophagy is negatively regulated by laminB1 suppression. Interestingly, this distinct regulation pattern of apoptosis and autophagy by laminB1 cooperatively promotes the response of BRAF-mutated melanoma cells to vemurafenib. CONCLUSIONS Our findings unveil the potential of laminB1 as both a diagnosis marker and a therapeutic target of melanoma.
Collapse
Affiliation(s)
- Yuan Li
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Yuqing Feng
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Dan Chen
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
5
|
Rahdan F, Abedi F, Dianat-Moghadam H, Sani MZ, Taghizadeh M, Alizadeh E. Autophagy-based therapy for hepatocellular carcinoma: from standard treatments to combination therapy, oncolytic virotherapy, and targeted nanomedicines. Clin Exp Med 2024; 25:13. [PMID: 39621122 PMCID: PMC11611955 DOI: 10.1007/s10238-024-01527-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024]
Abstract
Human hepatocellular carcinoma (HCC) has been identified as a significant cause of mortality worldwide. In recent years, extensive research has been conducted to understand the underlying mechanisms of autophagy in the pathogenesis of the disease, with the aim of developing novel therapeutic agents. Targeting autophagy with conventional therapies in invasive HCC has opened up new opportunities for treatment. However, the emergence of resistance and the immunosuppressive tumor environment highlight the need for combination therapy or specific targeting, as well as an efficient drug delivery system to ensure targeted tumor areas receive sufficient doses without affecting normal cells or tissues. In this review, we discuss the findings of several studies that have explored autophagy as a potential therapeutic approach in HCC. We also outline the potential and limitations of standard therapies for autophagy modulation in HCC treatment. Additionally, we discuss how different combination therapies, nano-targeted strategies, and oncolytic virotherapy could enhance autophagy-based HCC treatment in future research.
Collapse
Affiliation(s)
- Fereshteh Rahdan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Abedi
- Clinical Research Development, Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran.
- Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran.
| | - Maryam Zamani Sani
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Taghizadeh
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Dąbrowska A, Mastalerz J, Wilczyński B, Osiecka B, Choromańska A. Determinants of Photodynamic Therapy Resistance in Cancer Cells. Int J Mol Sci 2024; 25:12069. [PMID: 39596137 PMCID: PMC11594179 DOI: 10.3390/ijms252212069] [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: 10/10/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Photodynamic therapy (PDT) has emerged as a promising therapeutic approach owing to its non-invasive nature and minimal toxicity. PDT involves the administration of a photosensitizing agent (PS), which, upon light activation, induces a photodynamic reaction (PDR), leading to targeted cell destruction. However, developing resistance to PDT poses a significant challenge to its effectiveness. Various factors, including properties and administration of PSs, mediate this resistance. Despite the widespread use of substances like 5-aminolevulinic acid (5-ALA) and protoporphyrin, their efficacy is limited due to restricted tumor penetration and a lack of tumor targeting. To address these limitations, nano-delivery techniques and newer PSs like Aza-BODIPY and its derivatives, which offer enhanced tissue penetration, are being explored. In this paper, we provide an overview of resistance mechanisms in PDT and discuss novel methods, substances, and technologies to overcome resistance to improve clinical outcomes in tumor treatment.
Collapse
Affiliation(s)
- Alicja Dąbrowska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (A.D.); (J.M.); (B.W.)
| | - Jakub Mastalerz
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (A.D.); (J.M.); (B.W.)
| | - Bartosz Wilczyński
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (A.D.); (J.M.); (B.W.)
| | - Beata Osiecka
- Department of Clinical and Experimental Pathology, Wroclaw Medical University, T. Marcinkowskiego 1, 50-368 Wroclaw, Poland;
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
7
|
Merlin JPJ, Abrahamse H. Optimizing CRISPR/Cas9 precision: Mitigating off-target effects for safe integration with photodynamic and stem cell therapies in cancer treatment. Biomed Pharmacother 2024; 180:117516. [PMID: 39332185 DOI: 10.1016/j.biopha.2024.117516] [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: 07/12/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 09/29/2024] Open
Abstract
CRISPR/Cas9 precision genome editing has revolutionized cancer treatment by introducing specific alterations to the cancer genome. But the therapeutic potential of CRISPR/Cas9 is limited by off-target effects, which can cause undesired changes to genomic regions and create major safety concerns. The primary emphasis lies in their implications within the realm of cancer photodynamic therapy (PDT), where precision is paramount. PDT is a promising cancer treatment method; nevertheless, its effectiveness is severely limited and readily leads to recurrence due to the therapeutic resistance of cancer stem cells (CSCs). With a focus on targeted genome editing into cancer cells during PDT and stem cell treatment (SCT), the review aims to further the ongoing search for safer and more accurate CRISPR/Cas9-mediated methods. At the core of this exploration are recent advancements and novel techniques that offer promise in mitigating the risks associated with off-target effects. With a focus on cancer PDT and SCT, this review critically assesses the landscape of off-target effects in CRISPR/Cas9 applications, offering a comprehensive knowledge of their nature and prevalence. A key component of the review is the assessment of cutting-edge delivery methods, such as technologies based on nanoparticles (NPs), to optimize the distribution of CRISPR components. Additionally, the study delves into the intricacies of guide RNA design, focusing on advancements that bolster specificity and minimize off-target effects, crucial elements in ensuring the precision required for effective cancer PDT and SCT. By synthesizing insights from various methodologies, including the exploration of innovative genome editing tools and leveraging robust validation methods and bioinformatics tools, the review aspires to chart a course towards more reliable and precise CRISPR-Cas9 applications in cancer PDT and SCT. For safe PDT and SCT integration in cancer therapy, CRISPR/Cas9 precision optimization is essential. Utilizing sophisticated molecular and computational techniques to address off-target effects is crucial to realizing the therapeutic promise of these technologies, which will ultimately lead to the development of individualized and successful cancer treatment strategies. Our long-term goals are to improve precision genome editing for more potent cancer therapy approaches by refining the way CRISPR/Cas9 is integrated with photodynamic and stem cell therapies.
Collapse
Affiliation(s)
- J P Jose Merlin
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, South Africa.
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, South Africa
| |
Collapse
|
8
|
Hlapisi N, Songca SP, Ajibade PA. Capped Plasmonic Gold and Silver Nanoparticles with Porphyrins for Potential Use as Anticancer Agents-A Review. Pharmaceutics 2024; 16:1268. [PMID: 39458600 PMCID: PMC11510308 DOI: 10.3390/pharmaceutics16101268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Photothermal therapy (PTT) and photodynamic therapy (PDT) are potential cancer treatment methods that are minimally invasive with high specificity for malignant cells. Emerging research has concentrated on the application of metal nanoparticles encapsulated in porphyrin and their derivatives to improve the efficacy of these treatments. Gold and silver nanoparticles have distinct optical properties and biocompatibility, which makes them efficient materials for PDT and PTT. Conjugation of these nanoparticles with porphyrin derivatives increases their light absorption and singlet oxygen generation that create a synergistic effect that increases phototoxicity against cancer cells. Porphyrin encapsulation with gold or silver nanoparticles improves their solubility, stability, and targeted tumor delivery. This paper provides comprehensive review on the design, functionalization, and uses of plasmonic silver and gold nanoparticles in biomedicine and how they can be conjugated with porphyrins for synergistic therapeutic effects. Furthermore, it investigates this dual-modal therapy's potential advantages and disadvantages and offers perspectives for future prospects. The possibility of developing gold, silver, and porphyrin nanotechnology-enabled biomedicine for combination therapy is also examined.
Collapse
Affiliation(s)
| | | | - Peter A. Ajibade
- School of Chemistry and Physics, University of KwaZulu-Natal, Private Bag X01, Scottsville, Pietermaritzburg 3209, South Africa; (N.H.); (S.P.S.)
| |
Collapse
|
9
|
Ebrahimi S, Khaleghi Ghadiri M, Stummer W, Gorji A. Enhancing 5-ALA-PDT efficacy against resistant tumor cells: Strategies and advances. Life Sci 2024; 351:122808. [PMID: 38852796 DOI: 10.1016/j.lfs.2024.122808] [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: 04/04/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
As a precursor of protoporphyrin IX (PpIX), an endogenous pro-apoptotic and fluorescent molecule, 5-Aminolevulinic acid (5-ALA) has gained substantial attention for its potential in fluorescence-guided surgery as well as photodynamic therapy (PDT). Moreover, 5-ALA-PDT has been suggested as a promising chemo-radio sensitization therapy for various cancers. However, insufficient 5-ALA-induced PpIX fluorescence and the induction of multiple resistance mechanisms may hinder the 5-ALA-PDT clinical outcome. Reduced efficacy and resistance to 5-ALA-PDT can result from genomic alterations, tumor heterogeneity, hypoxia, activation of pathways related to cell surveillance, production of nitric oxide, and most importantly, deregulated 5-ALA transporter proteins and heme biosynthesis enzymes. Understanding the resistance regulatory mechanisms of 5-ALA-PDT may allow the development of effective personalized cancer therapy. Here, we described the mechanisms underlying resistance to 5-ALA-PTD across various tumor types and explored potential strategies to overcome this resistance. Furthermore, we discussed future approaches that may enhance the efficacy of treatments using 5-ALA-PDT.
Collapse
Affiliation(s)
- Safieh Ebrahimi
- Epilepsy Research Center, Münster University, 48149 Münster, Germany; Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran
| | | | - Walter Stummer
- Department of Neurosurgery, Münster University, 48149 Münster, Germany
| | - Ali Gorji
- Epilepsy Research Center, Münster University, 48149 Münster, Germany; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran; Neuroscience Research Center, Mashhad University of Medical Sciences, 9177948564 Mashhad, Iran.
| |
Collapse
|
10
|
Wahyudianingsih R, Sanjaya A, Jonathan T, Pranggono EH, Achmad D, Hernowo BS. Chemotherapy's effects on autophagy in the treatment of Hodgkin's lymphoma: a scoping review. Discov Oncol 2024; 15:269. [PMID: 38976168 PMCID: PMC11231119 DOI: 10.1007/s12672-024-01142-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Classical Hodgkin Lymphomas (HL) are a unique malignant growth with an excellent initial prognosis. However, 10-30% of patients will still relapse after remission. One primary cellular function that has been the focus of tumor progression is autophagy. This process can preserve cellular homeostasis under stressful conditions. Several studies have shown that autophagy may play a role in developing HL. Therefore, this review aimed to explore chemotherapy's effect on autophagy in HL, and the effects of autophagy on HL. METHODS A scoping review in line with the published PRISMA extension for scoping reviews (PRISMA-ScR) was conducted. A literature search was conducted on the MEDLINE database and the Cochrane Central Register of Controlled Trials (CENTRAL). All results were retrieved and screened, and the resulting articles were synthesized narratively. RESULTS The results showed that some cancer chemotherapy also induces autophagic flux. Although the data on HL is limited, since the mechanisms of action of these drugs are similar, we can infer a similar relationship. However, this increased autophagy activity may reflect a mechanism for increasing tumor growth or a cellular compensation to inhibit its growth. Although evidence supports both views, we argued that autophagy allowed cancer cells to resist cell death, mainly due to DNA damage caused by cytotoxic drugs. CONCLUSION Autophagy reflects the cell's adaptation to survive and explains why chemotherapy generally induces autophagy functions. However, further research on autophagy inhibition is needed as it presents a viable treatment strategy, especially against drug-resistant populations that may arise from HL chemotherapy regimens.
Collapse
Affiliation(s)
- Roro Wahyudianingsih
- Postgraduate Program of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, Indonesia
- Department of Anatomical Pathology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Ardo Sanjaya
- Department of Anatomy, Faculty of Medicine, Maranatha Christian University, Bandung, Indonesia.
| | - Timothy Jonathan
- Undergraduate Program in Medicine, Faculty of Medicine, Maranatha Christian University, Bandung, Indonesia
| | - Emmy Hermiyanti Pranggono
- Department of Internal Medicine, Faculty of Medicine, Universitas Padjadjaran/Rumah Sakit Hasan Sadikin, Bandung, West Java, Indonesia
| | - Dimyati Achmad
- Department of Oncological Surgery, Faculty of Medicine, Universitas Padjadjaran/Rumah Sakit Hasan Sadikin, Bandung, West Java, Indonesia
| | - Bethy Suryawathy Hernowo
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Padjadjaran/Rumah Sakit Hasan Sadikin, Bandung, West Java, Indonesia
| |
Collapse
|
11
|
Kurganovs NJ, Engedal N. To eat or not to eat: a critical review on the role of autophagy in prostate carcinogenesis and prostate cancer therapeutics. Front Pharmacol 2024; 15:1419806. [PMID: 38910881 PMCID: PMC11190189 DOI: 10.3389/fphar.2024.1419806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Around 1 in 7 men will be diagnosed with prostate cancer during their lifetime. Many strides have been made in the understanding and treatment of this malignancy over the years, however, despite this; treatment resistance and disease progression remain major clinical concerns. Recent evidence indicate that autophagy can affect cancer formation, progression, and therapeutic resistance. Autophagy is an evolutionarily conserved process that can remove unnecessary or dysfunctional components of the cell as a response to metabolic or environmental stress. Due to the emerging importance of autophagy in cancer, targeting autophagy should be considered as a potential option in disease management. In this review, along with exploring the advances made on understanding the role of autophagy in prostate carcinogenesis and therapeutics, we will critically consider the conflicting evidence observed in the literature and suggest how to obtain stronger experimental evidence, as the application of current findings in clinical practice is presently not viable.
Collapse
Affiliation(s)
- Natalie Jayne Kurganovs
- Autophagy in Cancer Lab, Institute for Cancer Research, Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Nikolai Engedal
- Autophagy in Cancer Lab, Institute for Cancer Research, Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
12
|
Zarrabi A, Perrin D, Kavoosi M, Sommer M, Sezen S, Mehrbod P, Bhushan B, Machaj F, Rosik J, Kawalec P, Afifi S, Bolandi SM, Koleini P, Taheri M, Madrakian T, Łos MJ, Lindsey B, Cakir N, Zarepour A, Hushmandi K, Fallah A, Koc B, Khosravi A, Ahmadi M, Logue S, Orive G, Pecic S, Gordon JW, Ghavami S. Rhabdomyosarcoma: Current Therapy, Challenges, and Future Approaches to Treatment Strategies. Cancers (Basel) 2023; 15:5269. [PMID: 37958442 PMCID: PMC10650215 DOI: 10.3390/cancers15215269] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Rhabdomyosarcoma is a rare cancer arising in skeletal muscle that typically impacts children and young adults. It is a worldwide challenge in child health as treatment outcomes for metastatic and recurrent disease still pose a major concern for both basic and clinical scientists. The treatment strategies for rhabdomyosarcoma include multi-agent chemotherapies after surgical resection with or without ionization radiotherapy. In this comprehensive review, we first provide a detailed clinical understanding of rhabdomyosarcoma including its classification and subtypes, diagnosis, and treatment strategies. Later, we focus on chemotherapy strategies for this childhood sarcoma and discuss the impact of three mechanisms that are involved in the chemotherapy response including apoptosis, macro-autophagy, and the unfolded protein response. Finally, we discuss in vivo mouse and zebrafish models and in vitro three-dimensional bioengineering models of rhabdomyosarcoma to screen future therapeutic approaches and promote muscle regeneration.
Collapse
Affiliation(s)
- Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Türkiye; (A.Z.); (A.Z.)
| | - David Perrin
- Section of Orthopaedic Surgery, Department of Surgery, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; (D.P.); (M.S.)
| | - Mahboubeh Kavoosi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland;
| | - Micah Sommer
- Section of Orthopaedic Surgery, Department of Surgery, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; (D.P.); (M.S.)
- Section of Physical Medicine and Rehabilitation, Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Serap Sezen
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
| | - Parvaneh Mehrbod
- Department of Influenza and Respiratory Viruses, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Bhavya Bhushan
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Science, McGill University, Montreal, QC H3A 0C7, Canada
| | - Filip Machaj
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Jakub Rosik
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Philip Kawalec
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Section of Neurosurgery, Department of Surgery, University of Manitoba, Health Sciences Centre, Winnipeg, MB R3A 1R9, Canada
| | - Saba Afifi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Seyed Mohammadreza Bolandi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Peiman Koleini
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Mohsen Taheri
- Genetics of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran;
| | - Tayyebeh Madrakian
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (T.M.); (M.A.)
| | - Marek J. Łos
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland;
| | - Benjamin Lindsey
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Nilufer Cakir
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
| | - Atefeh Zarepour
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Türkiye; (A.Z.); (A.Z.)
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran;
| | - Ali Fallah
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Tuzla, Istanbul 34956, Türkiye;
| | - Bahattin Koc
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Tuzla, Istanbul 34956, Türkiye;
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Türkiye
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye;
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (T.M.); (M.A.)
| | - Susan Logue
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), 01007 Vitoria-Gasteiz, Spain;
- University Institute for Regenerative Medicine and Oral Implantology–UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, USA;
| | - Joseph W. Gordon
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- College of Nursing, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555 Katowice, Poland
- Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| |
Collapse
|
13
|
Yang F, Xu M, Chen X, Luo Y. Spotlight on porphyrins: Classifications, mechanisms and medical applications. Biomed Pharmacother 2023; 164:114933. [PMID: 37236030 DOI: 10.1016/j.biopha.2023.114933] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/11/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023] Open
Abstract
Photodynamic therapy (PDT) and sonodynamic therapy (SDT) are non-invasive treatment methods with obvious inhibitory effect on tumors and have few side effects, which have been widely concerned and explored by researchers. Sensitizer is the main factor in determining the therapeutic effect of PDT and SDT. Porphyrins, a group of organic compounds widespread in nature, can be activated by light or ultrasound and produce reactive oxygen species. Therefore, porphyrins as sensitizers in PDT have been widely explored and investigated for many years. Herein, we summarize the classical porphyrin compounds and their applications and mechanisms in PDT and SDT. The application of porphyrin in clinical diagnosis and imaging is also discussed. In conclusion, porphyrins have good application prospects in disease treatment as an important part of PDT or SDT, and in clinical diagnosis and imaging.
Collapse
Affiliation(s)
- Fuyu Yang
- National Health Commission Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin 150001, China
| | - Meiqi Xu
- National Health Commission Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin 150001, China
| | - Xiaoyu Chen
- Department of Neonatal, The Fourth Hospital of Harbin Medical University, Harbin
| | - Yakun Luo
- National Health Commission Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
14
|
Vadarevu H, Sorinolu AJ, Munir M, Vivero-Escoto JL. Autophagy Regulation Using Multimodal Chlorin e6-Loaded Polysilsesquioxane Nanoparticles to Improve Photodynamic Therapy. Pharmaceutics 2023; 15:pharmaceutics15051548. [PMID: 37242794 DOI: 10.3390/pharmaceutics15051548] [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: 04/19/2023] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Photodynamic therapy (PDT) is a promising anticancer noninvasive technique that relies on the generation of reactive oxygen species (ROS). Unfortunately, PDT still has many limitations, including the resistance developed by cancer cells to the cytotoxic effect of ROS. Autophagy, which is a stress response mechanism, has been reported as a cellular pathway that reduces cell death following PDT. Recent studies have demonstrated that PDT in combination with other therapies can eliminate anticancer resistance. However, combination therapy is usually challenged by the differences in the pharmacokinetics of the drugs. Nanomaterials are excellent delivery systems for the efficient codelivery of two or more therapeutic agents. In this work, we report on the use of polysilsesquioxane (PSilQ) nanoparticles for the codelivery of chlorin-e6 (Ce6) and an autophagy inhibitor for early- or late-stage autophagy. Our results, obtained from a reactive oxygen species (ROS) generation assay and apoptosis and autophagy flux analyses, demonstrate that the reduced autophagy flux mediated by the combination approach afforded an increase in the phototherapeutic efficacy of Ce6-PSilQ nanoparticles. We envision that the promising results in the use of multimodal Ce6-PSilQ material as a codelivery system against cancer pave the way for its future application with other clinically relevant combinations.
Collapse
Affiliation(s)
- Hemapriyadarshini Vadarevu
- Department of Chemistry, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Adeola Julian Sorinolu
- Civil and Environmental Engineering Department, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Mariya Munir
- Civil and Environmental Engineering Department, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Juan L Vivero-Escoto
- Department of Chemistry, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| |
Collapse
|
15
|
Sun Q, Yang J, Shen W, Lu H, Hou X, Liu Y, Xu Y, Wu Q, Xuan Z, Yang Y, Yin D. Engineering mitochondrial uncoupler synergistic photodynamic nanoplatform to harness immunostimulatory pro-death autophagy/mitophagy. Biomaterials 2022; 289:121796. [PMID: 36108581 DOI: 10.1016/j.biomaterials.2022.121796] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/05/2022] [Accepted: 09/04/2022] [Indexed: 02/06/2023]
Abstract
Generally, autophagy/mitophagy, as a highly conserved lysosomal-based catabolic pathway, compromises the photodynamic therapy (PDT) efficiency by increasing the adaptation of tumor cells toward reactive oxygen species (ROS)-triggered protein damages and mitochondrial destruction. On the other hand, excessively activated autophagy/mitophagy cascades can provoke autophagic cell death and promote the endogenous antigens release of dying cells, thus playing a vital role in initiating the antitumor immune responses. To harness the exquisite immunomodulating effect of pro-death autophagy/mitophagy, we rationally constructed a MnO2 shell-coated multifunctional porphyrinic metal-organic framework (MOF) to load carbonyl cyanide 3-chlorophenylhydrazone (CCCP). The wrapped MnO2 shell could not only prevent premature release of CCCP during blood circulation but also conquer tumor hypoxia by catalyzing the decomposition of intratumoral H2O2. After entering tumor cells, the MnO2 shell could scavenge over-expressed glutathione (GSH), resulting in burst CCCP release and GSH-depletion/O2-generation enhanced PDT. More importantly, the released CCCP acts as a mitochondrial uncoupler can elicit mitochondrial depolarization and mitophagy, which could significantly boost the autophagy/mitophagy levels generated during PDT and consequently convert the pro-survival autophagy/mitophagy to pro-death, leading tumor cells to autophagic and immunogenic cell death. In vivo results reveal that the CCCP synergistic PDT could induce excessive immunostimulatory autophagy/mitophagy associated with T-cell responses and immunological memory, leading to complete ablation of primary tumors and prevention of tumor recurrence and lung metastasis. The effectiveness of this strategy may highlight the pro-death role and immunomodulating effect of autophagy/mitophagy in cancer therapy, providing a novel yet versatile avenue to enhance the efficacy of cancer treatments.
Collapse
Affiliation(s)
- Quanwei Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Jinming Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China; Anhui Provincial Key Laboratory of Research & Development of Chinese Medicine, Hefei 230021 China
| | - Huiyu Lu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Xiaohui Hou
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Yang Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Yujing Xu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Qinghua Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Zihua Xuan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230031, China; Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, China.
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China; Anhui Provincial Key Laboratory of Research & Development of Chinese Medicine, Hefei 230021 China; Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, China.
| |
Collapse
|
16
|
Ma Z, Lin K, Tang M, Ramachandran M, Qiu R, Li J, Solano LN, Huang Y, De Souza C, Abou-Adas S, Xiang B, Zhang L, Li M, Li Y. A pH-Driven Small-Molecule Nanotransformer Hijacks Lysosomes and Overcomes Autophagy-Induced Resistance in Cancer. Angew Chem Int Ed Engl 2022; 61:e202204567. [PMID: 35791769 PMCID: PMC9995155 DOI: 10.1002/anie.202204567] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Indexed: 11/11/2022]
Abstract
Smart conversion of supramolecular structures in vivo is an attractive strategy in cancer nanomedicine, which is usually achieved via specific peptide sequences. Here we developed a lysosomal targeting small-molecule conjugate, PBC, which self-assembles into nanoparticles at physiological pH and smartly converts to nanofibrils in lysosomes of tumor cells. Such a transformation mechanically leads to lysosomal dysfunction, autophagy inhibition, and unusual cytoplasmic vacuolation, thus granting PBC a unique anticancer activity as a monotherapy. Importantly, the photo-activated PBC elicits significant phototoxicity to lysosomes and shows enormous advantages in overcoming autophagy-caused treatment resistance frequently occurring in conventional phototherapy. This improved phototherapy achieves a complete cure of oral cancer xenografts upon limited administration. Our work provides a new paradigm for the construction of nonpeptide nanotransformers with biomedical activities.
Collapse
Affiliation(s)
- Zhao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine Shandong University, Jinan, Shandong, 250012, China
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Kai Lin
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, 266003, China
| | - Menghuan Tang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Mythili Ramachandran
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Reng Qiu
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Jin Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Lucas N Solano
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Cristabelle De Souza
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Sara Abou-Adas
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Bai Xiang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, 266003, China
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine Shandong University, Jinan, Shandong, 250012, China
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
17
|
Ma Z, Lin K, Tang M, Ramachandran M, Qiu R, Li J, Solano LN, Huang Y, De Souza C, Abou-Adas S, Xiang B, Zhang L, Li M, Li Y. A pH‐Driven Small‐Molecule Nanotransformer Hijacks Lysosomes and Overcomes Autophagy‐Induced Resistance in Cancer. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202204567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Zhao Ma
- Shandong University Cheeloo College of Medicine Medicinal Chemistry CHINA
| | - Kai Lin
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Menghuan Tang
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Mythili Ramachandran
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Reng Qiu
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Jin Li
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Lucas N. Solano
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Yanyu Huang
- University of California Davis Biochemistry and Molecular Medicine UNITED STATES
| | - Cristabelle De Souza
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Sara Abou-Adas
- University of California Davis Biochemistry and Molecular Medicine UNITED STATES
| | - Bai Xiang
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Lanwei Zhang
- Ocean University of China College of Food Science and Engineering CHINA
| | - Minyong Li
- Shandong University Cheeloo College of Medicine Medicinal Chemistry CHINA
| | - Yuanpei Li
- University of California Davis School of Medicine Biochemistry and Molecular Medicine 2700 Stockton Blvd, Suite 2405 95817 Sacramento UNITED STATES
| |
Collapse
|
18
|
Poghosyan S, Frenkel N, Lentzas A, Laoukili J, Rinkes IB, Kranenburg O, Hagendoorn J. Loss of Neuropilin-2 in Murine Mesenchymal-like Colon Cancer Organoids Causes Mesenchymal-to-Epithelial Transition and an Acquired Dependency on Insulin-Receptor Signaling and Autophagy. Cancers (Basel) 2022; 14:cancers14030671. [PMID: 35158941 PMCID: PMC8833430 DOI: 10.3390/cancers14030671] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Many cancer types are reported to have high lymphangiogenic receptor Neuropilin-2 (Nrp2) expression, including colorectal cancer (CRC). Nrp2 is shown to be associated with tumor progression in vivo and poor prognosis in CRC patients. Although the role of Nrp2 is well established in lymphangiogenesis, the tumor cell-intrinsic role of Nrp2 remains elusive. Here, we employed murine CRC tumor-derived mesenchymal-like organoids to induce Nrp2 depletion. We demonstrate that Nrp2 deletion in CRC organoids results in a drastically altered phenotype that is characterized by mesenchymal-to-epithelial transition (MET), and an acquired dependency on IR signaling and autophagy. This phenotype is preserved in subcutaneous tumors generated by CRC organoids. We conclude that there is a complex interaction between Nrp2 and alternative pro-survival mechanisms in aggressive CRC, which could be therapeutically exploited. Abstract Neuropilin-2 (Nrp2), an important regulator of lymphangiogenesis and lymphatic metastasis, has been associated with progression in colorectal cancer (CRC). However, the tumor cell-intrinsic role of Nrp2 in cancer progression is incompletely understood. To address this question, we employed CRISPR-Cas9 technology to generate Nrp2-knockout organoids derived from murine CRC tumors with a mesenchymal phenotype. Transcriptome profiling and tumor tissue analysis showed that Nrp2 loss resulted in mesenchymal-to-epithelial transition (MET), which was accompanied with restored polarity and tight junction stabilization. Signaling pathway analysis revealed that Nrp2-knockout organoids acquire de novo dependency on insulin receptor (IR) signaling and autophagy as alternative survival mechanisms. Combined inhibition of IR signaling and autophagy prevented the stabilization of cell-cell junctions, reduced metabolic activity, and caused profound cell death in Nrp2-knockout organoids. Collectively, the data demonstrate a key role for Nrp2 in maintaining the aggressive phenotype and survival of tumor-derived CRC organoids. The identified connection between Nrp2, insulin receptor signaling and autophagy may guide the development of novel combination-treatment strategies for aggressive CRC.
Collapse
|
19
|
Aniogo EC, George BP, Abrahamse H. Molecular Effectors of Photodynamic Therapy-Mediated Resistance to Cancer Cells. Int J Mol Sci 2021; 22:ijms222413182. [PMID: 34947979 PMCID: PMC8704319 DOI: 10.3390/ijms222413182] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 12/27/2022] Open
Abstract
Photodynamic therapy (PDT) is currently enjoying considerable attention as the subject of experimental research to treat resistant cancers. The preferential accumulation of a non-toxic photosensitizer (PS) in different cellular organelles that causes oxidative damage by combining light and molecular oxygen leads to selective cell killing. However, one major setback, common among other treatment approaches, is tumor relapse and the development of resistance causing treatment failure. PDT-mediated resistance could result from increased drug efflux and decreased localization of PS, reduced light exposure, increased DNA damage repair, and altered expression of survival genes. This review highlights the essential insights of PDT reports in which PDT resistance was observed and which identified some of the molecular effectors that facilitate the development of PDT resistance. We also discuss different perceptions of PDT and how its current limitations can be overturned to design improved cancer resistant treatments.
Collapse
|
20
|
Li L, Li G, Chen M, Cai R. Astragaloside IV enhances the sensibility of lung adenocarcinoma cells to bevacizumab by inhibiting autophagy. Drug Dev Res 2021; 83:461-469. [PMID: 34499759 DOI: 10.1002/ddr.21878] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/24/2022]
Abstract
Bevacizumab (BV) has an inhibitory effect on tumor growth including lung adenocarcinoma. However, its efficacy is greatly affected by drug resistance. Astragaloside IV (AST-IV) is effective in combination with other drugs is effective to treat cancer. This study aimed to investigate the effect of AST-IV on enhancing the sensibility of lung adenocarcinoma cells to BV. A549 cells were treated by different concentrations of BV and AST-IV. Cell viability, cell cycle, and apoptosis were detected by thiazolyl blue tetrazolium bromide (MTT) and flow cytometry, respectively. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and western blotting were performed to detect the expression levels of autophagy- and apoptosis-related proteins, protein kinase B (AKT), and mammalian target of rapamycin (mTOR). The results showed that BV or AST-IV could inhibit the viability and promote the apoptosis of A549 cells in a concentration-dependent manner. Moreover, BV or AST-IV inhibited Bcl-2 expression and increased the expressions of Bax and Cleaved caspase-3, and promoted apoptosis. BV and AST-IV in combination acted synergistically on viability and apoptosis of A549 cells. However, BV alone down-regulated P62 expression, LC3I/LC3II level, the number of cells arrested at S phase and the phosphorylation levels of AKT and mTOR, but upregulated the number of cells arrested at G0/G1 phase and Beclin1 expression, whereas AST-IV alone could reverse the effect of BV on autophagy-related proteins, the phosphorylation levels of AKT and mTOR. This paper demonstrates that AST-IV enhances the effect of BV on inhibiting proliferation and promoting apoptosis of lung adenocarcinoma cells through inhibiting autophagy pathway.
Collapse
Affiliation(s)
- Liang Li
- Department of Thoracic Surgery, Hainan General Hospital, Haikou, China
| | - Gao Li
- Department of Thoracic Surgery, Hainan General Hospital, Haikou, China
| | - Minbiao Chen
- Department of Thoracic Surgery, Hainan General Hospital, Haikou, China
| | - Renzhong Cai
- Department of Thoracic Surgery, Hainan General Hospital, Haikou, China
| |
Collapse
|
21
|
Gamelas SRD, Moura NMM, Habraken Y, Piette J, Neves MGPMS, Faustino MAF. Tetracationic porphyrin derivatives against human breast cancer. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 222:112258. [PMID: 34399205 DOI: 10.1016/j.jphotobiol.2021.112258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/10/2021] [Accepted: 07/09/2021] [Indexed: 01/06/2023]
Abstract
Photodynamic therapy (PDT) is an approved therapeutic approach and an alternative to conventional chemotherapy for the treatment of several types of cancer with the advantages of reducing the side effects and developing resistance mechanisms. Here, was evaluated the photosensitization capabilities of 5,10,15,20-tetrakis[4-(pyridinium-1-yl-methyl)phenyl]porphyrin (3), its N-confused isomer (4) and of the neutral precursors (1) and (2) and the results were compared with the ones obtained with the cationic 5,10,15,20-tetrakis(1-methylpyridinium-4-yl)porphyrin (TMPyP). Both regular porphyrin derivatives 1 and 3 showed higher efficiency to generate singlet oxygen than TMPyP. The PDT assays towards MCF-7 cells under red light irradiation (λ > 640 nm, 23.7 mW cm-2) demonstrated that the cationic porphyrin 3 is an efficient photosensitizer to kill MCF-7 breast cancer cells. The study of the cell death mechanisms induced by the photodynamic process showed that the studied porphyrin 3 and TMPyP caused cell death by autophagic flux and necrosis.
Collapse
Affiliation(s)
- Sara R D Gamelas
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Nuno M M Moura
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Yvette Habraken
- Laboratory of Gene Expression and Cancer, GIGA-Molecular Biology of Diseases, B34, University of Liège, Avenue de l'Hôpital 11, 4000 Liège, Belgium.
| | - Jacques Piette
- Laboratory of Virology and Immunology, GIGA-Molecular Biology of Diseases, GIGA B34, University of Liège, Avenue de l'Hôpital 11, 4000 Liège, Belgium
| | - Maria G P M S Neves
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Maria A F Faustino
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
22
|
Broadwater D, Medeiros HCD, Lunt RR, Lunt SY. Current Advances in Photoactive Agents for Cancer Imaging and Therapy. Annu Rev Biomed Eng 2021; 23:29-60. [PMID: 34255992 DOI: 10.1146/annurev-bioeng-122019-115833] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Photoactive agents are promising complements for both early diagnosis and targeted treatment of cancer. The dual combination of diagnostics and therapeutics is known as theranostics. Photoactive theranostic agents are activated by a specific wavelength of light and emit another wavelength, which can be detected for imaging tumors, used to generate reactive oxygen species for ablating tumors, or both. Photodynamic therapy (PDT) combines photosensitizer (PS) accumulation and site-directed light irradiation for simultaneous imaging diagnostics and spatially targeted therapy. Although utilized since the early 1900s, advances in the fields of cancer biology, materials science, and nanomedicine have expanded photoactive agents to modern medical treatments. In this review we summarize the origins of PDT and the subsequent generations of PSs and analyze seminal research contributions that have provided insight into rational PS design, such as photophysics, modes of cell death, tumor-targeting mechanisms, and light dosing regimens. We highlight optimizable parameters that, with further exploration, can expand clinical applications of photoactive agents to revolutionize cancer diagnostics and treatment.
Collapse
Affiliation(s)
- Deanna Broadwater
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Hyllana C D Medeiros
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Richard R Lunt
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan 48824, USA; , .,Department of Physics and Astronomy, Michigan State University, East Lansing, Michigan 48824, USA
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA.,Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan 48824, USA; ,
| |
Collapse
|
23
|
Xie J, Wang Y, Choi W, Jangili P, Ge Y, Xu Y, Kang J, Liu L, Zhang B, Xie Z, He J, Xie N, Nie G, Zhang H, Kim JS. Overcoming barriers in photodynamic therapy harnessing nano-formulation strategies. Chem Soc Rev 2021; 50:9152-9201. [PMID: 34223847 DOI: 10.1039/d0cs01370f] [Citation(s) in RCA: 265] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Photodynamic therapy (PDT) has been extensively investigated for decades for tumor treatment because of its non-invasiveness, spatiotemporal selectivity, lower side-effects, and immune activation ability. It can be a promising treatment modality in several medical fields, including oncology, immunology, urology, dermatology, ophthalmology, cardiology, pneumology, and dentistry. Nevertheless, the clinical application of PDT is largely restricted by the drawbacks of traditional photosensitizers, limited tissue penetrability of light, inefficient induction of tumor cell death, tumor resistance to the therapy, and the severe pain induced by the therapy. Recently, various photosensitizer formulations and therapy strategies have been developed to overcome these barriers. Significantly, the introduction of nanomaterials in PDT, as carriers or photosensitizers, may overcome the drawbacks of traditional photosensitizers. Based on this, nanocomposites excited by various light sources are applied in the PDT of deep-seated tumors. Modulation of cell death pathways with co-delivered reagents promotes PDT induced tumor cell death. Relief of tumor resistance to PDT with combined therapy strategies further promotes tumor inhibition. Also, the optimization of photosensitizer formulations and therapy procedures reduces pain in PDT. Here, a systematic summary of recent advances in the fabrication of photosensitizers and the design of therapy strategies to overcome barriers in PDT is presented. Several aspects important for the clinical application of PDT in cancer treatment are also discussed.
Collapse
Affiliation(s)
- Jianlei Xie
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Institute of Microscale Optoelectronics, and Otolaryngology Department and Biobank of the First Affiliated Hospital, Shenzhen Second People's Hospital, Health Science Center, Shenzhen University, Shenzhen 518060, P. R. China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
El-Hussein A, Manoto SL, Ombinda-Lemboumba S, Alrowaili ZA, Mthunzi-Kufa P. A Review of Chemotherapy and Photodynamic Therapy for Lung Cancer Treatment. Anticancer Agents Med Chem 2021; 21:149-161. [PMID: 32242788 DOI: 10.2174/1871520620666200403144945] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/15/2020] [Accepted: 02/20/2020] [Indexed: 11/22/2022]
Abstract
Cancer is among the leading causes of mortality and morbidity worldwide. Among the different types of cancers, lung cancer is considered to be the leading cause of death related to cancer and the most commonly diagnosed form of such disease. Chemotherapy remains a dominant treatment modality for many types of cancers at different stages. However, in many cases, cancer cells develop drug resistance and become nonresponsive to chemotherapy, thus, necessitating the exploration of alternative and /or complementary treatment modalities. Photodynamic Therapy (PDT) has emerged as an effective treatment modality for various malignant neoplasia and tumors. In PDT, the photochemical interaction of light, Photosensitizer (PS) and molecular oxygen produces Reactive Oxygen Species (ROS), which induces cell death. Combination therapy, by using PDT and chemotherapy, can promote synergistic effect against this fatal disease with the elimination of drug resistance, and enhancement of the efficacy of cancer eradication. In this review, we give an overview of chemotherapeutic modalities, PDT, and the different types of drugs associated with each therapy. Furthermore, we also explored the combined use of chemotherapy and PDT in the course of lung cancer treatment and how this approach could be the last resort for thousands of patients that have been diagnosed by this fatal disease.
Collapse
Affiliation(s)
- Ahmed El-Hussein
- National Institute of Laser Enhanced Science, Cairo University, Giza, Egypt
| | - Sello L Manoto
- Council for Scientific and Industrial Research (CSIR), National Laser Centre, Pretoria, South Africa
| | | | - Ziya A Alrowaili
- Physics Department, College of Science, Jouf University, Jouf, Saudi Arabia
| | - Patience Mthunzi-Kufa
- Council for Scientific and Industrial Research (CSIR), National Laser Centre, Pretoria, South Africa
| |
Collapse
|
25
|
Lim SM, Mohamad Hanif EA, Chin SF. Is targeting autophagy mechanism in cancer a good approach? The possible double-edge sword effect. Cell Biosci 2021; 11:56. [PMID: 33743781 PMCID: PMC7981910 DOI: 10.1186/s13578-021-00570-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a conserved cellular process required to maintain homeostasis. The hallmark of autophagy is the formation of a phagophore that engulfs cytosolic materials for degradation and recycling to synthesize essential components. Basal autophagy is constitutively active under normal conditions and it could be further induced by physiological stimuli such as hypoxia, nutrient starvation, endoplasmic reticulum stress,energy depletion, hormonal stimulation and pharmacological treatment. In cancer, autophagy is highly context-specific depending on the cell type, tumour microenvironment, disease stage and external stimuli. Recently, the emerging role of autophagy as a double-edged sword in cancer has gained much attention. On one hand, autophagy suppresses malignant transformation by limiting the production of reactive oxygen species and DNA damage during tumour development. Subsequently, autophagy evolved to support the survival of cancer cells and promotes the tumourigenicity of cancer stem cells at established sites. Hence, autophagy is an attractive target for cancer therapeutics and researchers have been exploiting the use of autophagy modulators as adjuvant therapy. In this review, we present a summary of autophagy mechanism and controlling pathways, with emphasis on the dual-role of autophagy (double-edged sword) in cancer. This is followed by an overview of the autophagy modulation for cancer treatment and is concluded by a discussion on the current perspectives and future outlook of autophagy exploitation for precision medicine.
Collapse
Affiliation(s)
- Su Min Lim
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, W. Persekutuan, 56000, Kuala Lumpur, Malaysia
| | - Ezanee Azlina Mohamad Hanif
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, W. Persekutuan, 56000, Kuala Lumpur, Malaysia
| | - Siok-Fong Chin
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, W. Persekutuan, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
26
|
Zhang L, Ouyang S, Zhang H, Qiu M, Dai Y, Wang S, Wang Y, Ou J. Graphene oxide induces dose-dependent lung injury in rats by regulating autophagy. Exp Ther Med 2021; 21:462. [PMID: 33747194 DOI: 10.3892/etm.2021.9893] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Graphene is a two-dimensional structured material with a hexagonal honeycomb lattice composed of carbon atoms. The biological effects of graphene oxide (GO) have been extensively investigated, as it has been widely used in biological research due to its increased hydrophilicity/biocompatibility. However, the exact mechanisms underlying GO-associated lung toxicity have not yet been fully elucidated. The aim of the present study was to determine the role of GO in lung injury induction, as well as its involvement in oxidative stress, inflammation and autophagy. The results revealed that lower concentrations of GO (5 and 10 mg/kg) did not cause significant lung injury, but the administration of GO at higher concentrations (50 and 100 mg/kg) induced lung edema, and increased lung permeability and histopathological lung changes. High GO concentrations also induced oxidative injury and inflammatory reactions in the lung, demonstrated by increased levels of oxidative products [malondialdehyde(MDA) and 8-hydroxydeoxyguanosine (8-OHdG)] and inflammatory factors (TNF-α, IL-6, IL-1β and IL-8). The autophagy inhibitors 3-methyladenine (3-MA) and chloroquine (CLQ) inhibited autophagy in the lung and attenuated GO-induced lung injury, as demonstrated by a reduced lung wet-to-dry weight ratio, lower levels of protein in the bronchoalveolar lavage fluid, and a reduced lung injury score. Furthermore, 3-MA and CLQ significantly reduced the levels of MDA, 8-OHdG and inflammatory factors in lung tissue, suggesting that autophagy also mediates the development of oxidative injury and inflammation in the lung. Finally, autophagy was directly inhibited in BEAS-2B cells by short hairpin RNA-mediated autophagy protein 5 (ATG5) knockdown, which were then treated with GO. Cell viability, as well as the extent of injury (indicated by lactate dehydrogenase level) and oxidative stress were determined. The results revealed that ATG5 knockdown-induced autophagic inhibition significantly decreased cellular injury and oxidative stress, suggesting that autophagy induction is a key event that leads to lung injury during exposure to GO. In conclusion, the findings of the present study indicated that GO causes lung injury in a dose-dependent manner by inducing autophagy.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Geotechnical Engineering, College of Civil Engineering, Tongji University, Shanghai 200092, P.R. China
| | - Shuge Ouyang
- Cambridge International Exam Centre in Shanghai Experimental School, Shanghai 200092, P.R. China
| | - Hongbo Zhang
- Chongming Branch of Xinhua Hospital Affiliated to The Medical College of Shanghai Jiaotong University, Shanghai 202150, P.R. China
| | - Mingke Qiu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Yuxin Dai
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Shuqing Wang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Yang Wang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| | - Jingmin Ou
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
27
|
Li Z, Teng M, Wang Y, Wang Q, Feng Y, Xiao Z, Li C, Zeng K. The mechanism of 5-aminolevulinic acid photodynamic therapy in promoting endoplasmic reticulum stress in the treatment of HR-HPV-infected HeLa cells. PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2021; 37:348-359. [PMID: 33513285 DOI: 10.1111/phpp.12663] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/04/2021] [Accepted: 01/24/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND 5-aminoketovaleric acid, as a precursor of the strong photosensitizer protoporphyrin IX (PpIX), mainly enters the mitochondria after entering the cell, and the formed PpIX is also mainly localized in the mitochondria. So at present the research on the mechanism of 5-aminoketovalerate photodynamic therapy (ALA-PDT) mainly focuses on its impact on mitochondria. There are few reports on whether ALA-PAT can affect the endoplasmic reticulum and trigger endoplasmic reticulum stress (ERS). AIMS/OBJECTIVES Here we investigated the effects of ALA-PDT on endoplasmic reticulum and its underlying mechanisms in high-risk human papillomavirus (HR-HPV) infection. MATERIALS AND METHODS The human cervical cancer cell line HeLa (containing whole genome of HR-HPV18) was treated with ALAPDT, and cell viability, ROS production, the level of Ca2+ in the cytoplasm and apoptosis were evaluated by CCK8, immunofluorescence and flow cytometry, respectively. The protein expression of the markers of ERS and autophagy and CamKKβ-AMPK pathway was examined by western blot. RESULTS The results showed that ALA-PDT inhibited cell viability of HeLa cells in vitro; ALA-PDT induced autophagy in HeLa cells ; ALA-PDT induced autophagy via the Ca2+-CamKKβ-AMPK pathway, which could be suppressed by the inhibition of ERS;ALA-PDT induced ERS-specific apoptosis via the activation of caspase 12. CONCLUSIONS Our study demonstrated that ALA-PDT could exert a killing effect by inducing HeLa cell apoptosis, including endoplasmic reticulum-specific apoptosis. Meanwhile, ERS via the Ca2+ -CamKKβ-AMPK pathway promoted the occurrence of autophagy in HeLa cells. Inhibition of autophagy could increase the apoptosis rate of HeLa cells after ALA-PDT, suggesting that autophagy may be one of the mechanisms of PDT resistance; The Ca2+-CamKKβ-AMPK pathway and autophagy may be targets to improve the killing effect of ALA-PDT in treating HR-HPV infection.
Collapse
Affiliation(s)
- Zhijia Li
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Muzhou Teng
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yajie Wang
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qi Wang
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yingjun Feng
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zixuan Xiao
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Changxing Li
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kang Zeng
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Martins WK, Belotto R, Silva MN, Grasso D, Suriani MD, Lavor TS, Itri R, Baptista MS, Tsubone TM. Autophagy Regulation and Photodynamic Therapy: Insights to Improve Outcomes of Cancer Treatment. Front Oncol 2021; 10:610472. [PMID: 33552982 PMCID: PMC7855851 DOI: 10.3389/fonc.2020.610472] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/03/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is considered an age-related disease that, over the next 10 years, will become the most prevalent health problem worldwide. Although cancer therapy has remarkably improved in the last few decades, novel treatment concepts are needed to defeat this disease. Photodynamic Therapy (PDT) signalize a pathway to treat and manage several types of cancer. Over the past three decades, new light sources and photosensitizers (PS) have been developed to be applied in PDT. Nevertheless, there is a lack of knowledge to explain the main biochemical routes needed to trigger regulated cell death mechanisms, affecting, considerably, the scope of the PDT. Although autophagy modulation is being raised as an interesting strategy to be used in cancer therapy, the main aspects referring to the autophagy role over cell succumbing PDT-photoinduced damage remain elusive. Several reports emphasize cytoprotective autophagy, as an ultimate attempt of cells to cope with the photo-induced stress and to survive. Moreover, other underlying molecular mechanisms that evoke PDT-resistance of tumor cells were considered. We reviewed the paradigm about the PDT-regulated cell death mechanisms that involve autophagic impairment or boosted activation. To comprise the autophagy-targeted PDT-protocols to treat cancer, it was underlined those that alleviate or intensify PDT-resistance of tumor cells. Thereby, this review provides insights into the mechanisms by which PDT can be used to modulate autophagy and emphasizes how this field represents a promising therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Waleska K Martins
- Laboratory of Cell and Membrane, Anhanguera University of São Paulo, São Paulo, Brazil
| | - Renata Belotto
- Perola Byington Hospital Gynecology - Lasertherapy Clinical Research Department, São Paulo, Brazil
| | - Maryana N Silva
- Laboratory of Cell and Membrane, Anhanguera University of São Paulo, São Paulo, Brazil
| | - Daniel Grasso
- CONICET, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maynne D Suriani
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia, Brazil
| | - Tayná S Lavor
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia, Brazil
| | - Rosangela Itri
- Institute of Physics, University of São Paulo, São Paulo, Brazil
| | | | - Tayana M Tsubone
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
29
|
Role of Bcl-2 Family Proteins in Photodynamic Therapy Mediated Cell Survival and Regulation. Molecules 2020; 25:molecules25225308. [PMID: 33203053 PMCID: PMC7696921 DOI: 10.3390/molecules25225308] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022] Open
Abstract
Photodynamic therapy (PDT) is a treatment modality that involves three components: combination of a photosensitizer, light and molecular oxygen that leads to localized formation of reactive oxygen species (ROS). The ROS generated from this promising therapeutic modality can be lethal to the cell and leads to consequential destruction of tumor cells. However, sometimes the ROS trigger a stress response survival mechanism that helps the cells to cope with PDT-induced damage, resulting in resistance to the treatment. One preferred mechanism of cell death induced by PDT is apoptosis, and B-cell lymphoma 2 (Bcl-2) family proteins have been described as a major determinant of life or death decision of the death pathways. Apoptosis is a cellular self-destruction mechanism to remove old cells through the biological event of tissue homeostasis. The Bcl-2 family proteins act as a critical mediator of a life–death decision of cells in maintaining tissue homeostasis. There are several reports that show cancer cells developing resistance due to the increased interaction of the pro-survival Bcl-2 family proteins. However, the key mechanisms leading to apoptosis evasion and drug resistance have not been adequately understood. Therefore, it is critical to understand the mechanisms of PDT resistance, as well as the Bcl-2 family proteins, to give more insight into the treatment outcomes. In this review, we describe the role of Bcl-2 gene family proteins’ interaction in response to disease progression and PDT-induced resistance mechanisms.
Collapse
|
30
|
Tsubone TM, Martins WK, Franco MSF, Silva MN, Itri R, Baptista MS. Cellular compartments challenged by membrane photo-oxidation. Arch Biochem Biophys 2020; 697:108665. [PMID: 33159891 DOI: 10.1016/j.abb.2020.108665] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/20/2020] [Accepted: 10/31/2020] [Indexed: 12/18/2022]
Abstract
The lipid composition impacts directly on the structure and function of the cytoplasmic as well as organelle membranes. Depending on the type of membrane, specific lipids are required to accommodate, intercalate, or pack membrane proteins to the proper functioning of the cells/organelles. Rather than being only a physical barrier that separates the inner from the outer spaces, membranes are responsible for many biochemical events such as cell-to-cell communication, protein-lipid interaction, intracellular signaling, and energy storage. Photochemical reactions occur naturally in many biological membranes and are responsible for diverse processes such as photosynthesis and vision/phototaxis. However, excessive exposure to light in the presence of absorbing molecules produces excited states and other oxidant species that may cause cell aging/death, mutations and innumerable diseases including cancer. At the same time, targeting key compartments of diseased cells with light can be a promising strategy to treat many diseases in a clinical procedure called Photodynamic Therapy. Here we analyze the relationships between membrane alterations induced by photo-oxidation and the biochemical responses in mammalian cells. We specifically address the impact of photosensitization reactions in membranes of different organelles such as mitochondria, lysosome, endoplasmic reticulum, and plasma membrane, and the subsequent responses of eukaryotic cells.
Collapse
Affiliation(s)
| | | | - Marcia S F Franco
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, SP, Brazil
| | | | - Rosangela Itri
- Department of Applied Physics, Institute of Physics, University of São Paulo, SP, Brazil
| | - Mauricio S Baptista
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, SP, Brazil.
| |
Collapse
|
31
|
Song C, Xu W, Wu H, Wang X, Gong Q, Liu C, Liu J, Zhou L. Photodynamic therapy induces autophagy-mediated cell death in human colorectal cancer cells via activation of the ROS/JNK signaling pathway. Cell Death Dis 2020; 11:938. [PMID: 33130826 PMCID: PMC7603522 DOI: 10.1038/s41419-020-03136-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Evidence has shown that m-THPC and verteporfin (VP) are promising sensitizers in photodynamic therapy (PDT). In addition, autophagy can act as a tumor suppressor or a tumor promoter depending on the photosensitizer (PS) and the cancer cell type. However, the role of autophagy in m-THPC- and VP-mediated PDT in in vitro and in vivo models of human colorectal cancer (CRC) has not been reported. In this study, m-THPC-PDT or VP-PDT exhibited significant phototoxicity, inhibited proliferation, and induced the generation of large amounts of reactive oxygen species (ROS) in CRC cells. From immunoblotting, fluorescence image analysis, and transmission electron microscopy, we found extensive autophagic activation induced by ROS in cells. In addition, m-THPC-PDT or VP-PDT treatment significantly induced apoptosis in CRC cells. Interestingly, the inhibition of m-THPC-PDT-induced autophagy by knockdown of ATG5 or ATG7 substantially inhibited the apoptosis of CRC cells. Moreover, m-THPC-PDT treatment inhibited tumorigenesis of subcutaneous HCT116 xenografts. Meanwhile, antioxidant treatment markedly inhibited autophagy and apoptosis induced by PDT in CRC cells by inactivating JNK signaling. In conclusion, inhibition of autophagy can remarkably alleviate PDT-mediated anticancer efficiency in CRC cells via inactivation of the ROS/JNK signaling pathway. Our study provides evidence for the therapeutic application of m-THPC and VP in CRC.
Collapse
Affiliation(s)
- Changfeng Song
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Wen Xu
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Hongkun Wu
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, 200003, P.R. China
| | - Xiaotong Wang
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Qianyi Gong
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P.R. China
| | - Chang Liu
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, 200003, P.R. China
| | - Jianwen Liu
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, P.R. China.
| | - Lin Zhou
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, 200003, P.R. China.
| |
Collapse
|
32
|
He C, Xia J, Gao Y, Chen Z, Wan X. Chlorin A-mediated photodynamic therapy induced apoptosis in human cholangiocarcinoma cells via impaired autophagy flux. Am J Transl Res 2020; 12:5080-5094. [PMID: 33042407 PMCID: PMC7540121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 02/02/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Photodynamic therapy (PDT) is a promising strategy for multiple cancers. Chlorin e6 and its derivative 131-[2'-(2-pyridyl)ethylamine] Chlorin e6 (Chlorin A) are effective photosensitizers, although their cytotoxic mechanisms have not yet been fully characterized. METHODS Cell viability and apoptosis were evaluated by CCK8 assay, TUNEL assay, and Annexin V/PI staining. The expression levels of different proteins were analyzed by Western blot analysis and immunofluorescence. The crosstalk between autophagy, endoplasmic reticulum stress (ERS), and mitochondrial dysfunction was investigated using reactive oxygen species (ROS) scavenger N-acetyl cysteine (NAC), PERK inhibitor GSK2606414, autophagy inhibitor 3-MA, and mitochondrial stabilizer elamipretide. Furthermore, the extent of ROS production, lysosomal damage, autophagy flux, and mitochondrial membrane potential (MMP) were tracked using established probes. An in vivo xenograft model of cholangiocarcinoma (CCA) was established in BALB/c-nude mice by inoculation with EGI-1 cells, and Chlorin A was administered topically or intravenously, followed by light irradiation. RESULTS Chlorin A-PDT decreased the viability of CCA cells and induced apoptosis. Intriguingly, Chlorin A-PDT promoted autophagy via activation of ROS-induced ERS-related PERK/p-eif2α/CHOP axis, and blocked the ensuing autophagy flux by lysosomal damage. The PERK inhibitor GSK2606414 and NAC alleviated apoptosis and autophagy induced by Chlorin A-PDT. Furthermore, mitochondrial dysfunction aggravated ERS, and stabilizing the mitochondria reduced both apoptosis and autophagy. Finally, Chlorin A-PDT significantly reduced tumor growth in vivo. CONCLUSIONS Chlorin A-PDT induced apoptosis in CCA cells by initiating autophagy and impaired the autophagy flux via ROS-mediated ERS and lysosomal damage.
Collapse
Affiliation(s)
- Chongxin He
- Shanghai Key Laboratory of Pancreatic Diseases and Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Jie Xia
- Department of Gastroenterology, The Second Hospital of Changzhou Affiliated to Nanjing Medical UniversityChangzhou, China
| | - Yinghua Gao
- Department of Pharmaceutical Science and Technology, College of Chemistry and Biology, Donghua UniversityShanghai, China
| | - Zhilong Chen
- Department of Pharmaceutical Science and Technology, College of Chemistry and Biology, Donghua UniversityShanghai, China
| | - Xinjian Wan
- Shanghai Key Laboratory of Pancreatic Diseases and Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| |
Collapse
|
33
|
Chemo-photodynamic therapy by pulmonary delivery of gefitinib nanoparticles and 5-aminolevulinic acid for treatment of primary lung cancer of rats. Photodiagnosis Photodyn Ther 2020; 31:101807. [PMID: 32404298 DOI: 10.1016/j.pdpdt.2020.101807] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/16/2020] [Accepted: 04/29/2020] [Indexed: 12/16/2022]
Abstract
Lung cancer is a severe disease with high mortality. Chemotherapy is one major treatment for lung cancer. However, systemic chemotherapeutics usually distribute throughout the body without specific lung distribution so that serious side effects are unavoidable. Photodynamic therapy (PDT) is occasionally used for lung cancer treatment but photosensitizers are also systemically administered and the bronchoscopic intervention under anesthesia may hurt lung tissues. Here, we combined inhaled chemotherapeutics and photosensitizers for chemo-photodynamic therapy (CPDT) of primary lung cancer of rats with external laser light irradiation. Gefitinib PLGA nanoparticles (GNPs) were prepared. The anti-cancer effects of GNPs and/or a common photosensitizer 5-aminolevulinic acid (5-ALA) were explored on A549 cells (adenocarcinomic human alveolar basal epithelial cells) and primary lung cancer rats after intratracheal administration. External light irradiation was applied due to its higher safety compared to internal light irradiation that may result in injuries after a laser optic fiber was intubated into the lung. The remarkable synergistic effect of CPDT was confirmed although the single therapies were also effective, where the high anti-lung cancer effects were shown and some typical lung cancer markers, including CD31, VEGF, NF-κB p65 and Bcl-2, significantly decreased. Moreover, the treatments attenuated inflammation with the downregulation of TNF-α. The combination of pulmonary drug delivery and chemo-photodynamic therapy is a promising strategy for treatment of lung cancer.
Collapse
|
34
|
Zou H, Wang F, Zhou JJ, Liu X, He Q, Wang C, Zheng YW, Wen Y, Xiong L. Application of photodynamic therapy for liver malignancies. J Gastrointest Oncol 2020; 11:431-442. [PMID: 32399283 PMCID: PMC7212095 DOI: 10.21037/jgo.2020.02.10] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 02/14/2020] [Indexed: 12/11/2022] Open
Abstract
Liver malignancies include primary and metastatic tumors. Limited progress has been achieved in improving the survival rate of patients with advanced stage liver cancer and who are unsuitable for surgery. Apart from surgery, chemoradiotherapy, trans-arterial chemoembolization and radiofrequency ablation, a novel therapeutic modality is needed for the clinical treatment of liver cancer. Photodynamic therapy (PDT) is a novel strategy for treating patients with advanced cancers; it uses a light-triggered cytotoxic photosensitizer and a laser light. PDT provides patients with a potential treatment approach with minimal invasion and low toxicity, that is, the whole course of treatment is painless, harmless, and repeatable. Therefore, PDT has been considered an effective palliative treatment for advanced liver cancers. To date, PDT has been used to treat hepatocellular carcinoma, cholangiocarcinoma, hepatoblastoma and liver metastases. Clinical outcomes reveal that PDT can be considered a promising treatment modality for all liver cancers to improve the quality and quantity of life of patients. Despite the advances achieved with this approach, several challenges still impede the application of PDT to liver malignancies. In this review, we focus on the recent advancements and discuss the future prospects of PDT in treating liver malignancies.
Collapse
Affiliation(s)
- Heng Zou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Fusheng Wang
- Department of General Surgery, Fuyang People’s Hospital, Fuyang 236000, China
| | - Jiang-Jiao Zhou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xi Liu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qing He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Cong Wang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yan-Wen Zheng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yu Wen
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Li Xiong
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
35
|
The Switch between Protective and Nonprotective Autophagy; Implications for Autophagy Inhibition as a Therapeutic Strategy in Cancer. BIOLOGY 2020; 9:biology9010012. [PMID: 31947952 PMCID: PMC7168293 DOI: 10.3390/biology9010012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 12/30/2019] [Accepted: 01/02/2020] [Indexed: 01/18/2023]
Abstract
Autophagy, a process of cellular self-degradation and cell survival whereby the cell generates energy and metabolic intermediates under conditions of stress (i.e., nutrient deprivation), is also commonly induced in tumor cells in response to chemotherapy and radiation. While chemotherapy-induced autophagy and radiation-induced autophagy are generally considered to have cytoprotective functions, thereby reducing tumor cell sensitivity (and potentially conferring resistance) to various treatment modalities, autophagy can also be nonprotective; furthermore, the nature of the autophagy can be altered via the “autophagic switch” depending on such factors as the p53 status of the tumor cells. Defective or compromised autophagy has also been associated with neurodegenerative diseases, raising concerns as to the impact of autophagy inhibition on normal tissue function. Furthermore, the impact of autophagy inhibition on the immune system response to therapy as well as the influence of autophagy inhibition in combination with chemotherapy or radiation on critical tissue sites such as the bone marrow remain uncertain. These are factors requiring serious consideration within the context of current clinical efforts to exploit autophagy inhibition as a therapeutic strategy in cancer.
Collapse
|
36
|
Rodrigues SD, Santos SS, Meireles T, Romero N, Glorieux G, Pecoits-Filho R, Zhang DD, Nakao LS. Uremic toxins promote accumulation of oxidized protein and increased sensitivity to hydrogen peroxide in endothelial cells by impairing the autophagic flux. Biochem Biophys Res Commun 2019; 523:123-129. [PMID: 31837804 DOI: 10.1016/j.bbrc.2019.12.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 12/05/2019] [Indexed: 01/12/2023]
Abstract
Chronic kidney disease (CKD) is associated with high mortality rates, mainly due to cardiovascular diseases (CVD). Uremia has been considered a relevant risk factor for CVD in CKD patients, since uremic toxins (UTs) promote systemic and vascular inflammation, oxidative stress and senescence. Here, we demonstrate that uremic toxins indoxyl sulfate (IxS), p-cresyl sulfate (pCS) and indole acetic acid (IAA) are incorporated by human endothelial cells and inhibit the autophagic flux, demonstrated by cellular p62 accumulation. Moreover, isolated and mixed UTs impair the lysosomal stage of autophagy, as determined by cell imaging of the mRFP-GFP-LC3 protein. Endothelial cells exposed to UTs display accumulation of carbonylated proteins and increased sensitivity to hydrogen peroxide. Rapamycin, an autophagy activator which induces both autophagosome formation and clearance, prevented these effects. Collectively, our findings demonstrate that accumulation of oxidized proteins and enhanced cell sensitivity to hydrogen peroxide are consequences of impaired autophagic flux. These data provide evidence that UTs-induced impaired autophagy may be a novel contributor to endothelial dysfunction.
Collapse
Affiliation(s)
- Silvia D Rodrigues
- Department of Basic Pathology, Universidade Federal do Paraná, 81531-980, Curitiba, Brazil
| | - Sabrina S Santos
- Department of Basic Pathology, Universidade Federal do Paraná, 81531-980, Curitiba, Brazil
| | - Tassiana Meireles
- Department of Basic Pathology, Universidade Federal do Paraná, 81531-980, Curitiba, Brazil
| | - Natalia Romero
- Cell Analysis Division, Agilent Technologies, 02421, Lexington, MA, USA
| | - Griet Glorieux
- Department of Nephrology, Ghent University Hospital, 9000, Ghent, Belgium
| | - Roberto Pecoits-Filho
- Center for Health and Biological Sciences, Pontific Catholic University of Paraná, 80215-901, Curitiba, Brazil
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, University of Arizona, 85721, Tucson, AZ, USA
| | - Lia S Nakao
- Department of Basic Pathology, Universidade Federal do Paraná, 81531-980, Curitiba, Brazil.
| |
Collapse
|
37
|
Xie J, Wang S, Li Z, Ao C, Wang J, Wang L, Peng X, Zeng K. 5-aminolevulinic acid photodynamic therapy reduces HPV viral load via autophagy and apoptosis by modulating Ras/Raf/MEK/ERK and PI3K/AKT pathways in HeLa cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2019; 194:46-55. [PMID: 30925276 DOI: 10.1016/j.jphotobiol.2019.03.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/11/2019] [Accepted: 03/20/2019] [Indexed: 01/09/2023]
Abstract
Human papillomavirus (HPV) infection is linked to several diseases, the most prominent of which are cervical cancer and genital condyloma acuminatum. Previous studies have suggested an effective role for 5-aminolevulinic acid photodynamic therapy (ALA-PDT) against various cancers by the induction of autophagy and apoptosis. However, few reports have focused on the effectiveness of ALA-PDT on HPV related disorders. To identify the role of ALA-PDT in the context of HPV infection, we initially investigated 111 patients suffering from genital condyloma acuminatum. HPV viral load detected before and after ALA-PDT treatment was compared during this procedure; a significant difference was noted. HeLa (HPV18) cells were exposed to ALA-PDT in vitro to further explore the underlying mechanisms. Western blot analysis showed that ALA-PDT induces LC3II and p62 expression, along with the up regulation of caspase-3 and cleaved caspase-3. Our study also demonstrated that ALA-PDT treatment inhibits the proliferation of HeLa cells in a dose dependent manner and effectively reduces HPV viral load via autophagy and apoptosis by regulating the Ras/Raf/MEK/ERK and PI3K/AKT/mTOR pathways. Hydroxychloroquine (HCQ), although it inhibited autophagy degradation, functioned to activate reactive oxygen species (ROS) levels of ALA-PDT to enhance the observed effect. These findings suggest strategies for the improvement of PDT efficacy in patients.
Collapse
Affiliation(s)
- Jiajia Xie
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Sijia Wang
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhijia Li
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chunping Ao
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jingying Wang
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Li Wang
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoming Peng
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kang Zeng
- Department of Dermatology and Venereology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
38
|
EtNBSe-PDT inhibited proliferation and induced autophagy of HNE-1 cells via downregulating the Wnt/β-catenin signaling pathway. Photodiagnosis Photodyn Ther 2019; 26:65-72. [PMID: 30831261 DOI: 10.1016/j.pdpdt.2019.02.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/19/2019] [Accepted: 02/25/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Increasing evidence has suggested that autophagy may play a resistant role during photodynamic therapy (PDT). The Wnt/β-catenin pathway is tightly involved in cell proliferation and autophagy. In this study, we aimed to determine the influence of 5-Ethylamino-9-diethylaminobenzo[a]phenoselenazinium (EtNBSe) mediated PDT (EtNBSe-PDT) on autophagy, proliferation and Wnt/β-catenin pathway in human NPC cell line (HNE-1 cells), and further explore the underlying crosstalk between them. METHODS Cell viability and proliferation was evaluated by MTT assay. Autophagy and Wnt/β-catenin signaling pathway was analyzed by western blotting and immunofluorescence. RESULTS It was revealed that EtNBSe-PDT significantly impeded the viability and proliferation of HNE-1 cells. Meanwhile EtNBSe-PDT could notably induce autophagy in HNE-1 cells accompanied with the inhibition of Wnt/β-catenin pathway. The Wnt/β-catenin pathway activator Wnt agonist was found to partially counteract the inhibitory proliferation of HNE-1 cells and suppress the autophagy induced by EtNBSe-PDT. In addition, pretreatment with the autophagy inhibitor 3-methyladenine (3-MA) or Wnt agonist showed the potential in enhancing the cytotoxic effect of EtNBSe-PDT (cell survival from 50.71 ± 4.16% to 24.53 ± 4.27% and from 52.64 ± 3.54% to 35.74 ± 4.27% respectively). CONCLUSION Taken together, this study demonstrated that EtNBSe-PDT suppressed viability and proliferation, and induced autophagy of HNE-1 cells via downregulating the Wnt/β-catenin pathway. The autophagy further constituted the cytoprotective mechanisms involved in HNE-1 cells, which suggested that the combination of EtNBSe-PDT and autophagy inhibitors may be a promising strategy for the treatment of human NPC.
Collapse
|
39
|
Wan SS, Zhang L, Zhang XZ. An ATP-Regulated Ion Transport Nanosystem for Homeostatic Perturbation Therapy and Sensitizing Photodynamic Therapy by Autophagy Inhibition of Tumors. ACS CENTRAL SCIENCE 2019; 5:327-340. [PMID: 30834321 PMCID: PMC6396388 DOI: 10.1021/acscentsci.8b00822] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Indexed: 05/08/2023]
Abstract
In this article, an adenosine-triphosphate-regulated (ATP-regulated) ion transport nanosystem [SQU@PCN, porphyrinic porous coordination network (PCN) incorporated with squaramide (SQU)] was designed and synthesized for homeostatic perturbation therapy (HPT) and sensitizing photodynamic therapy (PDT) of tumors. It was found that this nanotransporter SQU@PCN easily accumulated in tumor sites while avoiding metabolic clearance and side effects. In response to a high expression of ATP in the tumor, SQU@PCN was decomposed because of the strong coordination of ATP with metal ligand of PCN. Subsequently, incorporated SQU was released and then simultaneously transported chloride ions across membrane of the cell and lysosome along with the chloride ion concentration gradient. On one hand, influx of chloride ions by SQU increased intracellular ion concentration, which disrupted ion homeostasis and further induced tumor cell apoptosis. On the other hand, SQU-medicated coupling transport of H+/Cl- across the lysosomal membrane alkalized the lysosome, resulting in inhibition of autophagy. This SQU-mediated autophagy inhibition would sensitize PCN-based PDT since activated autophagy by traditional PDT would resist and weaken the therapeutic efficacy. In vivo animal test results revealed that combined HPT and sensitized PDT could realize tumor eradication while blocking metastasis, which provided a paradigm for complementary multimodal tumor treatment.
Collapse
Affiliation(s)
| | | | - Xian-Zheng Zhang
- Key Laboratory of Biomedical
Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
40
|
Duan X, Chen B, Cui Y, Zhou L, Wu C, Yang Z, Wen Y, Miao X, Li Q, Xiong L, He J. Ready player one? Autophagy shapes resistance to photodynamic therapy in cancers. Apoptosis 2018; 23:587-606. [PMID: 30288638 DOI: 10.1007/s10495-018-1489-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Photodynamic therapy (PDT) is a procedure used in cancer therapy that has been shown to be useful for certain indications. Considerable evidence suggests that PDT might be superior to conventional modalities for some indications. In this report, we examine the relationship between PDT responsiveness and autophagy, which can exert a cytoprotective effect. Autophagy is an essential physiological process that maintains cellular homeostasis by degrading dysfunctional or impaired cellular components and organelles via a lysosome-based pathway. Autophagy, which includes macroautophagy and microautophagy, can be a factor that decreases or abolishes responses to various therapeutic protocols. We systematically discuss the mechanisms underlying cell-fate decisions elicited by PDT; analyse the principles of PDT-induced autophagy, macroautophagy and microautophagy; and present evidence to support the notion that autophagy is a critical mechanism in resistance to PDT. A combined strategy involving autophagy inhibitors may be able to further enhance PDT efficacy. Finally, we provide suggestions for future studies, note where our understanding of the relevant molecular regulators is deficient, and discuss the correlations among PDT-induced resistance and autophagy, especially microautophagy.
Collapse
Affiliation(s)
- Xian Duan
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Bo Chen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yanan Cui
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Zhou
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Chenkai Wu
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhulin Yang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiongying Miao
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Qinglong Li
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Xiong
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China.
| | - Jun He
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
41
|
Autophagy and its potent modulators from phytochemicals in cancer treatment. Cancer Chemother Pharmacol 2018; 83:17-26. [PMID: 30353226 DOI: 10.1007/s00280-018-3707-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022]
Abstract
Autophagy is a ubiquitous catabolic process by which damaged or harmful intracellular components are delivered to the lysosomes for self-digestion and recycling. It is critical in cancer treatment. Therapy-induced autophagy predominantly acts as a pro-survival mechanism, but progressive autophagy can lead to non-apoptotic cell death, also known as autophagic cell death. Plants or herbs contain various natural compounds that are widely used in the treatment of many types of malignancies. Emerging evidence indicates that phytochemicals targeting the autophagic pathway are promising agents for cancer treatment. However, these compounds play different roles in autophagy. In this review, we discussed the role of autophagy in cancer development and therapy, and focussed on elucidating the anti-cancer activities of autophagic modulators, especially phytochemicals. Notably, we described a novel premise that the dynamic role of phytochemicals should be evaluated in regulation of autophagy in cancer.
Collapse
|
42
|
Typical and Atypical Inducers of Lysosomal Cell Death: A Promising Anticancer Strategy. Int J Mol Sci 2018; 19:ijms19082256. [PMID: 30071644 PMCID: PMC6121368 DOI: 10.3390/ijms19082256] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 07/30/2018] [Accepted: 07/30/2018] [Indexed: 12/22/2022] Open
Abstract
Lysosomes are conservative organelles with an indispensable role in cellular degradation and the recycling of macromolecules. However, in light of recent findings, it has emerged that the role of lysosomes in cancer cells extends far beyond cellular catabolism and includes a variety of cellular pathways, such as proliferation, metastatic potential, and drug resistance. It has been well described that malignant transformation leads to alterations in lysosomal structure and function, which, paradoxically, renders cancer cells more sensitive to lysosomal destabilization. Furthermore, lysosomes are implicated in the regulation and execution of cell death in response to diverse stimuli and it has been shown that lysosome-dependent cell death can be utilized to overcome apoptosis and drug resistance. Thus, the purpose of this review is to characterize the role of lysosome in cancer therapy and to describe how these organelles impact treatment resistance. We summarized the characteristics of typical inducers of lysosomal cell death, which exert its function primarily via alterations in the lysosomal compartment. The review also presents other anticancer agents with the predominant mechanism of action different from lysosomal destabilization, the activity of which is influenced by lysosomal signaling, including classical chemotherapeutics, kinase inhibitors, monoclonal antibodies, as well as photodynamic therapy.
Collapse
|