1
|
Villa J, Cury J, Kessler L, Tan X, Richter CP. Enhancing biocompatibility of the brain-machine interface: A review. Bioact Mater 2024; 42:531-549. [PMID: 39308547 PMCID: PMC11416625 DOI: 10.1016/j.bioactmat.2024.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/05/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
In vivo implantation of microelectrodes opens the door to studying neural circuits and restoring damaged neural pathways through direct electrical stimulation and recording. Although some neuroprostheses have achieved clinical success, electrode material properties, inflammatory response, and glial scar formation at the electrode-tissue interfaces affect performance and sustainability. Those challenges can be addressed by improving some of the materials' mechanical, physical, chemical, and electrical properties. This paper reviews materials and designs of current microelectrodes and discusses perspectives to advance neuroprosthetics performance.
Collapse
Affiliation(s)
- Jordan Villa
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Joaquin Cury
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Lexie Kessler
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Xiaodong Tan
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
- The Hugh Knowles Center, Department of Communication Sciences and Disorders, Northwestern University, USA
| | - Claus-Peter Richter
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
- The Hugh Knowles Center, Department of Communication Sciences and Disorders, Northwestern University, USA
- Department of Communication Sciences and Disorders, Northwestern University, USA
- Department of Biomedical Engineering, Northwestern University, USA
| |
Collapse
|
2
|
Begum S, Irvin SD, Cox CK, Huang Z, Wilson JJ, Monroe JD, Gibert Y. Anti-ovarian cancer migration and toxicity characteristics of a platinum(IV) pro-drug with axial HDAC inhibitor ligands in zebrafish models. Invest New Drugs 2024:10.1007/s10637-024-01479-3. [PMID: 39433643 DOI: 10.1007/s10637-024-01479-3] [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: 06/24/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024]
Abstract
Ovarian cancer is the fifth leading cause of cancer related death in the United States. Cisplatin is a platinum-based anti-cancer drug used against ovarian cancer that enters malignant cells and then damages DNA causing cell death. Typically, ovarian cancer cells become resistant to cisplatin making it necessary to increase subsequent dosage, which usually leads to side-effects including irreversible damage to kidney and auditory system tissue. Ovarian cancer resistance is often associated with upregulation of histone deacetylase (HDAC) enzymes that cause DNA to adopt a closed configuration which reduces the ability of cisplatin to target and damage DNA. Compound B, a platinum(IV) complex with two axial phenylbutyrate (PBA) HDAC inhibitor ligands attached to a cisplatin core, can simultaneously inhibit HDAC activity and damage DNA causing decreased cancer cell viability in cisplatin-sensitive (A2780) and -resistant (A2780cis) ovarian cancer cell lines. However, compound B was not previously evaluated in vivo. As simultaneously inhibiting HDAC-mediated resistance with cisplatin treatment could potentiate the platinum drug's effect, we first confirmed the anti-cancer effect of compound B in the A2780 and A2780cis cell lines using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide spectrophotometric assay. Then, we used zebrafish embryo and transgenic animal models to comparatively analyze the effect of cisplatin, compound B, and controls on general organismal, auditory, and renal system toxicity, and cancer metastasis. We found that lower dosages of compound B (0.3 or 0.6 µM) than of cisplatin (2.0 µM) could cause similar or decreased levels of general, auditory, and renal tissue toxicity, and at 0.6 µM, compound B reduces cancer metastasis more than 2.0 µM cisplatin.
Collapse
Affiliation(s)
- Salma Begum
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Scheldon D Irvin
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Carol K Cox
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Zhouyang Huang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853- 1301, USA
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853- 1301, USA
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA, 93106-9510, USA
| | - Jerry D Monroe
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA
| | - Yann Gibert
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
3
|
Zhang M, Zhang J, Ma Y, Jin Y, Li Y, Wu X. Nephropathy induced by cisplatin results from mitochondrial disruption, impaired energy metabolism, altered expression of renal transporters, and accumulation of urinary toxins. J Trace Elem Med Biol 2024; 86:127553. [PMID: 39427559 DOI: 10.1016/j.jtemb.2024.127553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/09/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND The administration of platinum-based drugs such as cisplatin and its derivatives, which are frequently used during clinical chemotherapy, is highly restricted due to the incidence of nephrotoxicity. The present study focused on investigating cisplatin-induced nephrotoxicity from the perspective of energy metabolism, renal transporter expression and urinary toxin accumulation. METHODS This study investigated cisplatin's toxic effects, including nephrotoxicity, cardiotoxicity, hepatotoxicity, pulmonary toxicity, and splenotoxicity. We used transmission electron microscopy (TEM) and scanning electron microscopy (SEM) to characterize the accumulation of cisplatin in the kidney and the structure of renal mitochondria. The production of reactive oxygen species (ROS) induced by cisplatin in renal tubular epithelial cells was evaluated by in vitro experiments, and apoptosis of renal tubular epithelial cells and alterations to the renal microvasculature were assessed. Metabolites associated with the glycolytic and tricarboxylic acid pathways were measured, and renal transporters expression, autophagy, and urinary toxins (UTs) accumulation were also assessed. RESULTS Our results reveal that cisplatin-induced varying degrees of damage to the heart, liver, spleen, lungs, and kidneys, including inflammatory and fibrotic damage. Accumulation of cisplatin in renal mitochondria disrupted mitochondrial structure and mitochondrial function, as evidenced by decreased levels of glucose 6-phosphate and ribose 5-phosphate and elevated levels of isocitric acid. Cisplatin-induced accumulation of ROS in renal tubular epithelial cells led to apoptosis and, ultimately, constriction or loss of renal microvasculature. Furthermore, dysregulation of renal transporter expression, activation of autophagy and increased accumulation of UTs was observed. CONCLUSION Accumulation of cisplatin in the kidney led to damage to mitochondrial structure and function, apoptosis of renal tubular epithelial cells, constriction or loss of renal microvasculature, dysfunction of renal transporters, activation of autophagy, and accumulation of UTs.
Collapse
Affiliation(s)
- Mingkang Zhang
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Jianping Zhang
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Yanrong Ma
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Yongwen Jin
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Yile Li
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Xin'an Wu
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China.
| |
Collapse
|
4
|
Cederroth CR, Dyhrfjeld-Johnsen J, Canlon B. Pharmacological Approaches to Hearing Loss. Pharmacol Rev 2024; 76:1063-1088. [PMID: 39164117 PMCID: PMC11549935 DOI: 10.1124/pharmrev.124.001195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024] Open
Abstract
Hearing disorders pose significant challenges to individuals experiencing them and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Current treatment options often focus on amplification devices, cochlear implants, or other rehabilitative therapies, leaving a substantial gap regarding effective pharmacological interventions. Advancements in our understanding of the molecular and cellular mechanisms involved in hearing disorders induced by noise, aging, and ototoxicity have opened new avenues for drug development, some of which have led to numerous clinical trials, with promising results. The development of optimal drug delivery solutions in animals and humans can also enhance the targeted delivery of medications to the ear. Moreover, large genome studies contributing to a genetic understanding of hearing loss in humans combined with advanced molecular technologies in animal studies have shown a great potential to increase our understanding of the etiologies of hearing loss. The auditory system exhibits circadian rhythms and temporal variations in its physiology, its vulnerability to auditory insults, and its responsiveness to drug treatments. The cochlear clock rhythms are under the control of the glucocorticoid system, and preclinical evidence suggests that the risk/benefit profile of hearing disorder treatments using chronopharmacological approaches would be beneficial. If translatable to the bedside, such approaches may improve the outcome of clinical trials. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug formulation and delivery as well as optimized timing of drug administration, holds great promise of more effective treatments. SIGNIFICANCE STATEMENT: Hearing disorders pose significant challenges to individuals and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug delivery procedures and optimized timing of drug administration, holds the promise of more effective treatments.
Collapse
Affiliation(s)
- Christopher R Cederroth
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Jonas Dyhrfjeld-Johnsen
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Barbara Canlon
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| |
Collapse
|
5
|
Fawzy MH, Moustafa YM, Khodeer DM, Saeed NM, El-Sayed NM. Doxepin as OCT2 inhibitor ameliorates inflammatory response and modulates PI3K/Akt signaling associated with cisplatin-induced nephrotoxicity in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03473-1. [PMID: 39400714 DOI: 10.1007/s00210-024-03473-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/18/2024] [Indexed: 10/15/2024]
Abstract
Organic cationic transporter 2 (OCT2) was identified as the main transporter involved in the accumulation of cisplatin (CP) in the proximal tubular renal cells, resulting in nephrotoxicity. Doxepin (DOX) is a tricyclic agent with an inhibitory effect on OCT2. This study aimed to explore the possible mechanisms of the renoprotective role of DOX toward CP-induced nephrotoxicity. Rats were randomly divided into six groups: group 1, control; group 2, CP; groups 3, 4, and 5 were treated with graded doses of DOX (5, 10, and 20 mg/kg, respectively) intraperitoneally (ip) once daily for 10 consecutive days and group 6 was treated only with DOX (20 mg/kg). On the seventh day, a single injected dose of CP (10 mg/kg, ip) was given to the rats in groups 2-5. Seventy-two hours after CP injection, rats were sacrificed, and the kidneys were removed for histological and biochemical measurements. DOX ameliorated the CP-induced histopathological alterations. DOX significantly reduced the expression of OCT2, lipid peroxidation marker (MDA), and inflammatory cytokines, including TNF-α, IL-6, IL-1, IL-2, and IL-1β, and increased the activity of antioxidant enzymes. In addition, pre- and co-treatment with DOX significantly reduced the CP-mediated apoptotic effect by reducing the renal tissue expression of BAX and caspase-3 levels, upregulating the expression of Bcl-2, and modulating the phosphorylation of PI3K/Akt signaling cascade. DOX exerts a nephroprotective impact against CP-mediated nephrotoxicity via the inhibition of OCT2, suppression of inflammation, oxidative stress, and apoptotic markers, and modulation of PI3K/Akt signaling cascade.
Collapse
Affiliation(s)
- Mariam H Fawzy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Yasser M Moustafa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University in Cairo, Badr City, Egypt
| | - Dina M Khodeer
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Noha M Saeed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Norhan M El-Sayed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
6
|
Miao DNR, Wilke MAP, Pham J, Ladha F, Singh M, Arsenio J, Luca E, Dabdoub A, Yang W, Yang JJ, Drögemöller BI. Leveraging large-scale datasets and single cell omics data to develop a polygenic score for cisplatin-induced ototoxicity. Hum Genomics 2024; 18:112. [PMID: 39380081 PMCID: PMC11463131 DOI: 10.1186/s40246-024-00679-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Cisplatin-induced ototoxicity (CIO), characterized by irreversible and progressive bilateral hearing loss, is a prevalent adverse effect of cisplatin chemotherapy. Alongside clinical risk factors, genetic variants contribute to CIO and genome-wide association studies (GWAS) have highlighted the polygenicity of this adverse drug reaction. Polygenic scores (PGS), which integrate information from multiple genetic variants across the genome, offer a promising tool for the identification of individuals who are at higher risk for CIO. Integrating large-scale hearing loss GWAS data with single cell omics data holds potential to overcome limitations related to small sample sizes associated with CIO studies, enabling the creation of PGSs to predict CIO risk. RESULTS We utilized a large-scale hearing loss GWAS and murine inner ear single nuclei RNA-sequencing (snRNA-seq) data to develop two polygenic scores: a hearing loss PGS (PGSHL) and a biologically informed PGS for CIO (PGSCIO). The PGSCIO included only variants which mapped to genes that were differentially expressed within cochlear cells that showed differential abundance in the murine snRNA-seq data post-cisplatin treatment. Evaluation of the association of these PGSs with CIO in our target CIO cohort revealed that PGSCIO demonstrated superior performance (P = 5.54 × 10- 5) relative to PGSHL (P = 2.93 × 10- 3). PGSCIO was also associated with CIO in our test cohort (P = 0.04), while the PGSHL did not show a significant association with CIO (P = 0.52). CONCLUSION This study developed the first PGS for CIO using a large-scale hearing loss dataset and a biologically informed filter generated from cisplatin-treated murine inner ear snRNA-seq data. This innovative approach offers new avenues for developing PGSs for pharmacogenomic traits, which could contribute to the implementation of tailored therapeutic interventions. Further, our approach facilitated the identification of specific cochlear cells that may play critical roles in CIO. These novel insights will guide future research aimed at developing targeted therapeutic strategies to prevent CIO.
Collapse
Affiliation(s)
- Deanne Nixie R Miao
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - MacKenzie A P Wilke
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - John Pham
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Feryal Ladha
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Mansumeet Singh
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Janilyn Arsenio
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Emilia Luca
- Sunnybrook Research Institute, Toronto, ON, Canada
| | | | - Wejian Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jun J Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Britt I Drögemöller
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
- CancerCare Manitoba Research Institute, Winnipeg, MB, Canada.
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
- Centre of Aging, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
7
|
Choi SJ, Lee SJ, Lee D, Im GJ, Jung HH, Lee SU, Park E. Protective Effect of Memantine on Cisplatin-Induced Ototoxicity: An In Vitro Study. Otol Neurotol 2024; 45:998-1005. [PMID: 39186064 DOI: 10.1097/mao.0000000000004317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
HYPOTHESIS Memantine, an N -methyl- d -aspartate receptor antagonist, is widely used to treat Alzheimer's disease and has been found to have potential neuroprotective effects. In this study, we evaluated the protective effects of memantine against cisplatin-induced ototoxicity. BACKGROUND Cisplatin is a widely used anticancer drug for various cancers; however, its use is limited by its side effects, including ototoxicity. Several drugs have been developed to reduce cisplatin toxicity. In this study, we treated cisplatin-damaged cochlear hair cells with memantine and evaluated its protective effects. METHOD House Ear Institute Organ of Corti 1 (HEI-OC1) cells and cochlear explants were treated with cisplatin or memantine. Cell viability, apoptotic patterns, reactive oxygen species (ROS) production, Bcl-2/caspase-3 activity, and cell numbers were measured to evaluate the anti-apoptotic and antioxidative effects of memantine. RESULT Memantine treatment significantly improved cell viability and reduced cisplatin-induced apoptosis in auditory cells. Bcl-2/caspase-3 activity was also significantly increased, suggesting anti-apoptotic effects against cisplatin-induced ototoxicity. CONCLUSION Our results suggest that memantine protects against cisplatin-induced ototoxicity in vitro, providing a potential new strategy for preventing hearing loss in patients undergoing cisplatin chemotherapy.
Collapse
Affiliation(s)
- Soo Jeong Choi
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Soo Jin Lee
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Dabin Lee
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Gi Jung Im
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hak Hyun Jung
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | | | | |
Collapse
|
8
|
Lyu AR, Kim SJ, Park MJ, Park YH. CORM‑2 reduces cisplatin accumulation in the mouse inner ear and protects against cisplatin-induced ototoxicity. J Adv Res 2024; 64:183-194. [PMID: 38030129 PMCID: PMC11464639 DOI: 10.1016/j.jare.2023.11.020] [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: 09/13/2023] [Revised: 11/02/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
INTRODUCTION Cisplatin is a life-saving anticancer compound used to treat multiple solid malignant tumors, while it causes permanent hearing loss. There is no known cure, and the FDA has not approved any preventative treatment for cisplatin-based ototoxicity. OBJECTIVES This study investigated whether the carbon monoxide (CO)-releasing tricarbonyldichlororuthenium (II) dimer, CORM-2, reverses cisplatin-induced hearing impairment and reduces cisplatin accumulation in the mouse inner ear. METHODS Male 6-week-old BALB/c mice were randomly assigned to one of the following groups: control (saline-treated, i.p.), CORM-2 only (30 mg/kg, i.p., four doses), cisplatin only (20 mg/kg, i.p., one dose), and CORM-2 + cisplatin, to determine whether cisplatin-based hearing impairment was alleviated by CORM-2 treatment. RESULTS Our results revealed CORM-2 significantly attenuated cisplatin-induced hearing loss in young adult mice. CORM-2 co-treatment significantly decreased platinum accumulation in the inner ear and activated the plasma membrane repair system of the stria vascularis. Moreover, CORM-2 co-treatment significantly decreased cisplatin-induced inflammation, apoptosis, and cochlear necroptosis. Because the stria vascularis is the likely cochlear entry point of cisplatin, we next focused on the microvasculature. Cisplatin induced increased extravasation of a chromatic tracer (fluorescein isothiocyanate [FITC]-dextran, MW 75 kDa) around the cochlear microvessels at 4 days post-treatment; this extravasation was completely inhibited by CORM-2 co-therapy. CORM-2 co-treatment effectively maintained the integrity of stria vascularis components including endothelial cells, pericytes, and perivascular-resident macrophage-type melanocytes. CONCLUSION CORM-2 co-therapy substantially protects against cisplatin-induced ototoxicity by reducing platinum accumulation and toxic cellular stress responses. These data indicate that CORM-2 co-treatment may be translated into clinical strategy to reduce cisplatin-induced hearing loss.
Collapse
Affiliation(s)
- Ah-Ra Lyu
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Soo Jeong Kim
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Min Jung Park
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea.
| | - Yong-Ho Park
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea; Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea.
| |
Collapse
|
9
|
Li P, Li D, Lu Y, Pan S, Cheng F, Li S, Zhang X, Huo J, Liu D, Liu Z. GSTT1/GSTM1 deficiency aggravated cisplatin-induced acute kidney injury via ROS-triggered ferroptosis. Front Immunol 2024; 15:1457230. [PMID: 39386217 PMCID: PMC11461197 DOI: 10.3389/fimmu.2024.1457230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 08/23/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Cisplatin is a widely used chemotherapeutic agent prescribed to treat solid tumors. However, its clinical application is limited because of cisplatin- induced nephrotoxicity. A known complication of cisplatin is acute kidney injury (AKI). Deletion polymorphisms of GSTM1 and GSTT1, members of the glutathione S-transferase family, are common in humans and are presumed to be associated with various kidney diseases. However, the specific roles and mechanisms of GSTM1 and GSTT1 in cisplatin induced AKI remain unclear. Methods To investigate the roles of GSTM1 and GSTT1 in cisplatin-induced AKI, we generated GSTM1 and GSTT1 knockout mice using CRISPR-Cas9 technology and assessed their kidney function under normal physiological conditions and cisplatin treatment. Using ELISA kits, we measured the levels of oxidative DNA and protein damage, along with MDA, SOD, GSH, and the GSH/GSSG ratio in wild-type and GSTM1/GSTT1 knockout mice following cisplatin treatment. Additionally, oxidative stress levels and the expression of ferroptosis-related proteins in kidney tissues were examined through Western blotting, qPCR, immunohistochemistry, and immunofluorescence techniques. Results Here, we found that GSTT1 and GSTM1 were downregulated in the renal tubular cells of AKI patients and cisplatin-treated mice. Compared with WT mice, Gstm1/Gstt1-DKO mice were phenotypically normal but developed more severe kidney dysfunction and exhibited increased ROS levels and severe ferroptosis after injecting cisplatin. Discussion Our study revealed that GSTM1 and GSTT1 can protect renal tubular cells against cisplatin-induced nephrotoxicity and ferroptosis, and genetic screening for GSTM1 and GSTT1 polymorphisms can help determine a standard cisplatin dose for cancer patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Peipei Li
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Duopin Li
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Yanfang Lu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shaokang Pan
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Fei Cheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shen Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaonan Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Jinling Huo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| |
Collapse
|
10
|
Kessler L, Koo C, Richter CP, Tan X. Hearing loss during chemotherapy: prevalence, mechanisms, and protection. Am J Cancer Res 2024; 14:4597-4632. [PMID: 39417180 PMCID: PMC11477841 DOI: 10.62347/okgq4382] [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: 05/15/2024] [Accepted: 08/23/2024] [Indexed: 10/19/2024] Open
Abstract
Ototoxicity is an often-underestimated sequela for cancer patients undergoing chemotherapy, with an incidence rate exceeding 50%, affecting approximately 4 million individuals worldwide each year. Despite the nearly 2,000 publications on chemotherapy-related ototoxicity in the past decade, the understanding of its prevalence, mechanisms, and preventative or therapeutic measures remains ambiguous and subject to debate. To date, only one drug, sodium thiosulfate, has gained FDA approval for treating ototoxicity in chemotherapy. However, its utilization is restricted. This review aims to offer clinicians and researchers a comprehensive perspective by thoroughly and carefully reviewing available data and current evidence. Chemotherapy-induced ototoxicity is characterized by four primary symptoms: hearing loss, tinnitus, vertigo, and dizziness, originating from both auditory and vestibular systems. Hearing loss is the predominant symptom. Amongst over 700 chemotherapeutic agents documented in various databases, only seven are reported to induce hearing loss. While the molecular mechanisms of the hearing loss caused by the two platinum-based drugs are extensively explored, the pathways behind the action of the other five drugs are primarily speculative, rooted in their therapeutic properties and side effects. Cisplatin attracts the majority of attention among these drugs, encompassing around two-thirds of the literature regarding ototoxicity in chemotherapy. Cisplatin ototoxicity chiefly manifests through the loss of outer hair cells, possibly resulting from damages directly by cisplatin uptake or secondary effects on the stria vascularis. Both direct and indirect influences contribute to cisplatin ototoxicity, while it is still debated which path is dominant or where the primary target of cisplatin is located. Candidates for hearing protection against cisplatin ototoxicity are also discussed, with novel strategies and methods showing promise on the horizon.
Collapse
Affiliation(s)
- Lexie Kessler
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
| | - Chail Koo
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
| | - Claus-Peter Richter
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
- Department of Biomedical Engineering, Northwestern UniversityEvanston, Illinois 60208, USA
- Department of Communication Sciences and Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
| | - Xiaodong Tan
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
| |
Collapse
|
11
|
Elwell CE, Stein E, Lewis A, Hamaway S, Alexis KA, Tanski JM, Barnum TJ, Connelly CM, Tyler LA. Synthesis, characterization and comparative biological activity of a novel set of Cu(II) complexes containing azole-based ligand frames. J Inorg Biochem 2024; 262:112736. [PMID: 39332382 DOI: 10.1016/j.jinorgbio.2024.112736] [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: 06/26/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/29/2024]
Abstract
The synthesis and spectroscopic characterization of three complexes containing a substituted 2-(2-pyridyl)benzothiazole (PyBTh) group in the ligand frame are reported along with the comparative biological activity. The ligands have been substituted at the 6-position with either a methoxy (Py(OMe)BTh) or a methyl group (Py(Me)BTh). Reaction of Py(OMe)BTh with either CuCl2 or Cu(NO3)2·2.5 H2O yielded the monomeric [Cu(Py(OMe)BTh))2(NO3)]NO3·1.5 MeOH, (1·1.5 MeOH) complex or the dimeric [Cu(Py(OMe)BTh)Cl2]2 (2), respectively, with the nuclearity of the complex dependent on the starting Cu(II) salt. Reaction between the methyl substituted ligand and Cu(NO3)2·2.5 H2O resulted in the isolation of Cu(Py(Me)BTh)(NO3)2·0.5 THF (3·0.5 THF). Complexes 1-3 were fully characterized. Cyclic voltammetry measurements were performed on all three complexes as well as on [Cu(PyBTh)2(H2O)](BF4)2 (4), a compound previously reported by us which contains the unsubstituted 2-(2-pyridyl)benzothiazole ligand. The biological activity was studied and included concentration dependent DNA binding and cleavage, antibacterial activity, and cancer cell toxicity. All complexes exhibited DNA cleavage activity, however 2 and 4 were found to be the most potent. Mechanistic studies revealed that the nuclease activity is dependent on an oxidative mechanism reliant principally on O2-. Antibacterial studies revealed complex 4 was more potent compared to 1-3. Cancer cell toxicity studies were carried out on HeLa, PC-3, and MCF7 cells with 1-4, Cu(QBTh)(NO3)2(H2O) and Cu(PyBIm)3(BF4)2. The differences in the observed toxicities suggests the importance of the ligand and its substituents in modulating cell death.
Collapse
Affiliation(s)
- Courtney E Elwell
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Emily Stein
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Adam Lewis
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Stefan Hamaway
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Kennedy A Alexis
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Joseph M Tanski
- Department of Chemistry, Vassar College, Poughkeepsie, NY 12604, United States
| | - Timothy J Barnum
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Colleen M Connelly
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States.
| | - Laurie A Tyler
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States.
| |
Collapse
|
12
|
Burger AVM, Duinkerken CW, van Sluis KE, de Boer JP, Navran A, Lanting CP, Jóźwiak K, Dreschler WA, Balm AJM, Zuur CL. Treatment-related hearing loss in weekly versus triweekly cisplatin chemoradiation for head and neck cancer. Eur Arch Otorhinolaryngol 2024:10.1007/s00405-024-08880-x. [PMID: 39242413 DOI: 10.1007/s00405-024-08880-x] [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: 04/10/2024] [Accepted: 07/29/2024] [Indexed: 09/09/2024]
Abstract
PURPOSE Cisplatin-induced hearing loss is a common side effect in patients treated with cisplatin-based chemoradiation (CRT) for head and neck squamous cell carcinoma. The extent of hearing loss after concurrent CRT was compared between triweekly (3 × 100 mg/m2) and weekly (7 × 40 mg/m2) cisplatin CRT. METHOD This retrospective cohort study was conducted in the Antoni van Leeuwenhoek Hospital and included 129 patients with cisplatin-based CRT for head and neck cancer (72 treated in the triweekly and 57 in the weekly regimen). Baseline and follow-up pure tone audiometry was conducted to assess hearing loss. Clinically relevant hearing loss was defined as a decline upon treatment of ≥ 10 decibel at a pure tone average 1-2-4 kHz and/or 8-10-12.5 kHz. RESULTS The incidence of clinically relevant cisplatin CRT induced hearing loss was 42% in the triweekly versus 19% in the weekly group (p < 0.01). The mean threshold shift at a pure tone average (PTA) 1-2-4 kHz was 9.0 decibel in the triweekly compared to 4.3 decibel in the weekly CRT group (p < 0.01). At PTA 8-10-12.5 kHz, the incidence of clinically relevant hearing loss was 75% in the triweekly compared to 74% in the weekly CRT group (p = 0.87). The mean threshold shift at PTA 8-10-12.5 kHz was 20.2 decibel versus 15.6 decibel, respectively (p = 0.07). CONCLUSION Cisplatin-dose reduction to a weekly cisplatin CRT regimen for head and neck cancer may reduce the incidence of clinically relevant hearing loss at frequencies vital for speech perception.
Collapse
Affiliation(s)
- A V M Burger
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
- Department of Otolaryngology and Head and Neck Surgery, Leiden University Medical Centre, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - C W Duinkerken
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
- Department of Otolaryngology and Head and Neck Surgery, Leiden University Medical Centre, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - K E van Sluis
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - J P de Boer
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A Navran
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C P Lanting
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - K Jóźwiak
- Institute of Biostatistics and Registry Research, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - W A Dreschler
- Department of Audiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - A J M Balm
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
- Department of Maxillofacial Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - C L Zuur
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.
- Department of Otolaryngology and Head and Neck Surgery, Leiden University Medical Centre, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands.
| |
Collapse
|
13
|
Moreno-Gordaliza E, González-Nicolás MÁ, Lázaro A, Barbas C, Gómez-Gómez MM, López-Gonzálvez Á. Untargeted metabolomics analysis of serum and urine unveils the protective effect of cilastatin on altered metabolic pathways during cisplatin-induced acute kidney injury. Biochem Pharmacol 2024; 227:116435. [PMID: 39025411 DOI: 10.1016/j.bcp.2024.116435] [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: 03/15/2024] [Revised: 06/26/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Acute kidney injury (AKI) is one of the most serious complications of cisplatin anticancer therapies. Cilastatin is a highly promising nephroprotective agent to eventually enter clinical use, but its biochemical mechanism is still not fully understood. We have employed an untargeted metabolomics approach based on capillary electrophoresis mass spectrometry (CE-MS) analysis of serum and urine from an in vivo rat model, to explore the metabolic pathways involved in cisplatin-induced AKI and cilastatin nephroprotection. A total of 155 and 76 identified metabolites were found to be significantly altered during cisplatin treatment in urine and serum, respectively. Most of these altered metabolites were either partially or totally recovered by cilastatin and cisplatin co-treatment. The main metabolic pathways disturbed by cisplatin during AKI involved diverse amino acids metabolism and biosynthesis, tricarboxylic acids (TCA) cycle, nicotinate and nicotinamide metabolism, among others. Cilastatin was proved to protect diverse cisplatin-altered pathways involving metabolites related to immunomodulation, inflammation, oxidative stress and amino acid metabolism in proximal tubules. However, cisplatin-altered mitochondrial metabolism (especially, the energy-producing TCA cycle) remained largely unprotected by cilastatin, suggesting an unresolved mitochondrial direct damage. Multivariate analysis allowed effective discrimination of cisplatin-induced AKI and cilastatin renoprotection based on metabolic features. A number of potential serum and urine biomarkers could also be foreseen for cisplatin-induced AKI detection and cilastatin nephroprotection.
Collapse
Affiliation(s)
- Estefanía Moreno-Gordaliza
- Department of Analytical Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid (ROR 02p0gd045), Avda. Computense s/n, 28040 Madrid, Spain.
| | - M Ángeles González-Nicolás
- Renal Physiopathology Laboratory, Department of Nephrology, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Alberto Lázaro
- Renal Physiopathology Laboratory, Department of Nephrology, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; Department of Physiology, School of Medicine, Universidad Complutense de Madrid (ROR 02p0gd045), Avda. Complutense s/n, 28040 Madrid, Spain
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Faculty of Pharmacy, Universidad San Pablo-CEU (CEU Universities), Urbanización Montepríncipe, 28660 Boadilla del Monte, Madrid, Spain
| | - M Milagros Gómez-Gómez
- Department of Analytical Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid (ROR 02p0gd045), Avda. Computense s/n, 28040 Madrid, Spain
| | - Ángeles López-Gonzálvez
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Faculty of Pharmacy, Universidad San Pablo-CEU (CEU Universities), Urbanización Montepríncipe, 28660 Boadilla del Monte, Madrid, Spain
| |
Collapse
|
14
|
Honma Y, Monden N, Yamazaki K, Kano S, Satake H, Kadowaki S, Utsumi Y, Nakatogawa T, Takano R, Fujii K, Koroki Y, Aoyama J, Ouchi S, Ogawa T, McCarthy S, Brookman-May SD, Mundle S, Li J, Thaper D, Nagao T, Tada Y. Apalutamide and Goserelin for Androgen Receptor-Positive Salivary Gland Carcinoma: A Phase II Nonrandomized Clinical Trial, YATAGARASU. Clin Cancer Res 2024; 30:3416-3427. [PMID: 38940667 DOI: 10.1158/1078-0432.ccr-24-0455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/09/2024] [Accepted: 06/26/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE To assess the efficacy and safety of apalutamide plus goserelin for androgen receptor (AR)-positive unresectable or recurrent/metastatic salivary gland carcinoma. PATIENTS AND METHODS This trial was an open-label, single-arm, multicenter phase II study. Patients with histologically confirmed unresectable or recurrent/metastatic salivary gland carcinoma with AR expression were included. The primary endpoint was the overall response rate (ORR) according to RECIST v1.1 by an independent central radiology review in the first 24 response-evaluable (RE) patients who had been observed at least 24 weeks from study initiation (primary RE patients). The efficacy was to be declared when at least 8 of the 24 primary RE patients responded. RESULTS A total of 31 patients were enrolled. In the first 24 primary RE patients with a median follow-up of 7.4 months, confirmed ORR by independent central radiology review was 25.0% [6/24 patients; 95% confidence interval, 9.8%-46.7%; P = 0.11 (one-sided)], which did not meet the predefined criteria of efficacy. Clinical benefit rate (ORR + rate of stable disease for at least 24 weeks) and median progression-free survival were 50.0% and 7.4 months, respectively. Both median duration of response and overall survival were not reached. Exploratory analyses showed a better ORR of 54.5% (6/11) in patients with AR positivity ≥70% and no history of prior systemic therapy. Grade 3 or higher treatment-emergent adverse events were reported in 35.5% (11/31), which included skin rash, anemia, leukopenia, and cancer pain. CONCLUSIONS Although this study did not meet the predefined efficacy criteria, apalutamide plus goserelin showed clinically meaningful efficacy in a subset of patients with AR-positive salivary gland carcinoma and safety consistent with prior experience in prostate cancer.
Collapse
Affiliation(s)
- Yoshitaka Honma
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Nobuya Monden
- Department of Head and Neck, Surgery, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Keisuke Yamazaki
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medical, and Dental Sciences, Niigata University, Niigata, Japan
| | - Satoshi Kano
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Hironaga Satake
- Cancer Treatment Center, Kansai Medical University Hospital, Osaka & Department of Medical Oncology, Kochi Medical School, Kochi, Japan
| | - Shigenori Kadowaki
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Aichi, Japan
| | - Yoshitaka Utsumi
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | | | - Ryo Takano
- Janssen Pharmaceutical K.K., Tokyo, Japan
| | - Koji Fujii
- Janssen Pharmaceutical K.K., Tokyo, Japan
| | | | | | | | | | - Sharon McCarthy
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Sabine D Brookman-May
- Janssen Research & Development, Spring House, Pennsylvania, USA
- Ludwig-Maximilians University (LMU), Munich, Germany
| | - Suneel Mundle
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Jinhui Li
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Daksh Thaper
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Toshitaka Nagao
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Yuichiro Tada
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare, Mita Hospital, Tokyo, Japan
| |
Collapse
|
15
|
Schaeffers AWMA, Burger AVM, Duinkerken CW, van Sluis KE, de Boer JP, van der Molen L, Hoetink AE, Al-Mamgani A, Jóźwiak K, Devriese LA, de Bree R, Zuur CL. The association between skeletal muscle mass and sensorineural hearing loss upon cisplatin-based chemoradiotherapy in patients with head and neck squamous cell carcinoma. Head Neck 2024. [PMID: 39096016 DOI: 10.1002/hed.27907] [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: 12/14/2023] [Revised: 06/05/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
INTRODUCTION Patients with head and neck squamous cell carcinoma (HNSCC) treated with cisplatin-based chemoradiotherapy (CRT) frequently experience irreversible sensorineural hearing loss (SNHL). Patients with low lumbar skeletal muscle index (LSMI) may experience higher serum peak dosages of cisplatin. This study investigated whether pre-treatment low LSMI is associated with increased SNHL upon cisplatin-based CRT. MATERIALS AND METHODS LSMI was assessed using routine pre-treatment CT scans. Pure tone audiometry was performed at baseline and at follow-up to assess treatment-related SNHL. Linear mixed models were used to reveal a potential association between the continuous variable LSMI and SNHL. RESULTS This retrospective cohort study included 81 patients and found a significant association between low LSMI and increased treatment-related SNHL at pure tone frequencies vital for the perception of speech (averaged of 1, 2, and 4 kHz) (p = 0.048). CONCLUSIONS HNSCC patients with low LSMI suffer increased treatment-related SNHL upon cisplatin-based CRT.
Collapse
Affiliation(s)
- Anouk W M A Schaeffers
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Anouk V M Burger
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Otolaryngology and Head and Neck Surgery, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Charlotte W Duinkerken
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Otolaryngology and Head and Neck Surgery, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Klaske E van Sluis
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jan Paul de Boer
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lisette van der Molen
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Alex E Hoetink
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, UMC Brain Centre, Utrecht, the Netherlands
| | - Abrahim Al-Mamgani
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Katarzyna Jóźwiak
- Institute of Biostatistics and Registry Research, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Lot A Devriese
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Remco de Bree
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Charlotte L Zuur
- Department of Head and Neck Surgery and Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Otolaryngology and Head and Neck Surgery, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| |
Collapse
|
16
|
Magagnoli J, Cummings TH, Hardin JW, Sutton SS, Ambati J. Fluoxetine, fluvoxamine, and hearing loss or tinnitus after cisplatin treatment: A retrospective cohort study. J Investig Med 2024; 72:579-586. [PMID: 38597272 DOI: 10.1177/10815589241247796] [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] [Indexed: 04/11/2024]
Abstract
Cisplatin use is often limited by its ototoxic side effects, which can lead to irreversible hearing loss. Preventing cisplatin-induced ototoxicity is crucial to improve patient outcomes. Fluoxetine and fluvoxamine, both selective serotonin reuptake inhibitors antidepressants, inhibit the NLR pyrin domain-containing protein 3 inflammasome, a potential therapeutic target for preventing ototoxicity. However, human studies have not evaluated if these antidepressants may protect against cisplatin-induced ototoxicity. The object of this study is to assess the association between fluoxetine or fluvoxamine use and incidence of hearing loss or tinnitus in a large cohort of patients receiving cisplatin chemotherapy. We use a retrospective cohort study within the U.S. Department of Veterans Affairs healthcare system. Adult patients with cancer who received cisplatin chemotherapy between 2000 and 2023 are included. Incidence of ototoxicity, defined by international classification of diseases revision 9-CM or international classification of diseases revision 10-CM diagnoses of hearing loss or tinnitus is compared between concurrent use of fluoxetine or fluvoxamine and cisplatin alone. A total of 20,552 patients were included. Of those, 489 received cisplatin and fluoxetine or fluvoxamine. After propensity score adjustment, the hazard of ototoxicity was lower in the group receiving fluoxetine or fluvoxamine compared to the group receiving cisplatin alone (HR = 0.62, 95% CI = (0.41-0.94)). Fluoxetine or fluvoxamine use may be associated with a reduced risk of cisplatin-induced ototoxicity. Randomized clinical trials are needed to confirm these findings and establish the efficacy of the medications in ototoxicity prevention. Further research is also warranted to investigate the potential mechanisms underlying this protective effect.
Collapse
Affiliation(s)
- Joseph Magagnoli
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC, USA
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina College of Pharmacy, Columbia, SC, USA
| | - Tammy H Cummings
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC, USA
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina College of Pharmacy, Columbia, SC, USA
| | - James W Hardin
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC, USA
- Department of Epidemiology and Biostatistics, University of South Carolina, Columbia, SC, USA
| | - S Scott Sutton
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC, USA
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina College of Pharmacy, Columbia, SC, USA
| | - Jayakrishna Ambati
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VI, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VI, USA
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VI, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VI, USA
| |
Collapse
|
17
|
Jiang W, Fan S, Zhu Z, Huang H, Tan Y, Peng Y. Design, synthesis and mechanistic studies of novel arylformylhydrazone butylphenyltin complexes as potential anticancer agents. Bioorg Chem 2024; 149:107502. [PMID: 38805912 DOI: 10.1016/j.bioorg.2024.107502] [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: 03/31/2024] [Revised: 05/17/2024] [Accepted: 05/25/2024] [Indexed: 05/30/2024]
Abstract
Many diorganotin complexes with various alkyl groups exhibit excellent in vitro anticancer activity. However, most diorganotin is the same alkyl group, and the asymmetric alkyl R group has been rarely reported. Hence, in this paper, twenty butylphenyl mixed dialkyltin arylformylhydrazone complexes have been synthesized by microwave "one-pot" reaction with arylformylhydrazine, substituted α-keto acid or its sodium salt and butylphenyltin dichloride. The crystal structures of nine complexes were determined, indicating that the complexes C1, C2, C11, C12, and C16 ∼ C19 possessed a central symmetric structure of a dinuclear Sn2O2 tetrahedral ring; while the complex C9 is a trinuclear tin-oxygen cluster with a 6-membered ring encased in a 12-membered macrocyclic structure. The inhibiting activity of complexes was tested against the human cell lines NCI-H460, MCF-7, HepG2, Huh-7 and HL-7702. Complex C2 demonstrated the optimal inhibitory effect on HepG2 cells, with an IC50 value of 0.82 ± 0.03 μM. Cellular biology experiments revealed that complex C2 could induce apoptosis and G2/M phase cell cycle arrest in HepG2 and Huh-7 cells. The complex also caused the collapse of the mitochondrial membrane potential and increased intracellular reactive oxygen species in HepG2 and Huh-7 cells. Western blot analysis further clarified that complex C2 could induce cell apoptosis through the mitochondrial pathway along with the release of reactive oxygen species.
Collapse
Affiliation(s)
- Wujiu Jiang
- Key Laboratory of Green Chemistry, Jiangxi Province, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China; Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China.
| | - Shanji Fan
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421000, China
| | - Zhihua Zhu
- Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China
| | - Huifen Huang
- Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China
| | - Yuxing Tan
- Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China
| | - Yiyuan Peng
- Key Laboratory of Green Chemistry, Jiangxi Province, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China.
| |
Collapse
|
18
|
Chen LT, Vogel A, Hsu C, Chen MH, Fang W, Pangarsa EA, Sharma A, Ikeda M, Park JO, Tan CK, Regala E, Tai D, Tanasanvimon S, Charoentum C, Chee CE, Lui A, Sow J, Oh DY, Ueno M, Ramaswamy A, Jeo WS, Zhou J, Curigliano G, Yoshino T, Bai LY, Pentheroudakis G, Chiang NJ, Cervantes A, Chen JS, Ducreux M. Pan-Asian adapted ESMO Clinical Practice Guidelines for the diagnosis, treatment and follow-up of patients with biliary tract cancer. ESMO Open 2024; 9:103647. [PMID: 39232586 PMCID: PMC11410730 DOI: 10.1016/j.esmoop.2024.103647] [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: 04/29/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 09/06/2024] Open
Abstract
The European Society for Medical Oncology (ESMO) Clinical Practice Guidelines for the diagnosis, treatment and follow-up of patients with biliary tract cancer (BTC), published in late 2022 were adapted in December 2023, according to established standard methodology, to produce the Pan-Asian adapted (PAGA) ESMO consensus guidelines for the management of Asian patients with BTC. The adapted guidelines presented in this manuscript represent the consensus opinions reached by a panel of Asian experts in the treatment of patients with BTC representing the oncological societies of China (CSCO), Indonesia (ISHMO), India (ISMPO), Japan (JSMO), Korea (KSMO), Malaysia (MOS), the Philippines (PSMO), Singapore (SSO), Taiwan (TOS) and Thailand (TSCO), co-ordinated by ESMO and the Taiwan Oncology Society (TOS). The voting was based on scientific evidence and was independent of the current treatment practices, drug access restrictions and reimbursement decisions in the different regions of Asia. Drug access and reimbursement in the different regions of Asia are discussed separately in the manuscript. The aim is to provide guidance for the optimisation and harmonisation of the management of patients with BTC across the different countries and regions of Asia, drawing on the evidence provided by both Western and Asian trials, whilst respecting the differences in screening practices and molecular profiling, as well as age and stage at presentation. Attention is drawn to the disparity in the drug approvals and reimbursement strategies, between the different countries.
Collapse
Affiliation(s)
- L-T Chen
- Kaohsiung Medical University Hospital, Center for Cancer Research, Kaohsiung Medical University, Kaohsiung; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.
| | - A Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Medical School of Hannover, Hannover, Germany; Division of Gastroenterology and Hepatology, Toronto General Hospital, Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada
| | - C Hsu
- Department of Oncology, National Taiwan University Hospital, Taipei; Department of Medical Oncology, National Taiwan University Cancer Center, Taipei
| | - M-H Chen
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - W Fang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - E A Pangarsa
- Haematology Medical Oncology Division, Department of Oncology, Faculty of Medicine, Diponegoro University/Dr. Kariadi Hospital, Semarang, Indonesia
| | - A Sharma
- Department of Medical Oncology, Max Institute of Cancer Care, Max Super Specialty Hospital, Saket, New Delhi, India
| | - M Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - J O Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - C K Tan
- Department of Oncology and Nuclear Medicine, Thomson Hospital Kota Damansara, Petaling Jaya, Selangor, Malaysia
| | - E Regala
- Clinical Division Building, University of Santo Tomas Hospital, Sampaloc, Manila, Philippines
| | - D Tai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - S Tanasanvimon
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok
| | - C Charoentum
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - C E Chee
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - A Lui
- Department of Internal Medicine, Metro Davao Medical and Research Center, Davao City; Section of Medical Oncology, Department of Internal Medicine, Southern Philippines Medical Center, Davao City, The Philippines
| | - J Sow
- Department of Oncology, Curie Oncology Kuala Lumpur, Kuala Lumpur, Malaysia
| | - D-Y Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - M Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - A Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Parel, Mumbai, India
| | - W S Jeo
- Division of Digestive Surgery, Department of General Surgery, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - J Zhou
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - G Curigliano
- Istituto Europeo di Oncologia, Milano, IRCCS, Milano; Department of Oncology and Haematology, University of Milano, Milano, Italy
| | - T Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - L-Y Bai
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | | | - N-J Chiang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - A Cervantes
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia; CIBERONC. Instituto de Salud Carlos III, Madrid, Spain
| | - J-S Chen
- Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - M Ducreux
- INSERM U1279, Université Paris-Saclay, Villejuif; Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| |
Collapse
|
19
|
Azam F, Alharbi H, Alshangiti A, Zar Gul AR, Bukhari N, Ouda M, Anwar Hussain S, Ibnshamsah F. Eligibility Criteria for Different Platinum-Based Chemotherapy Regimens in Metastatic Urothelial Carcinoma. Cureus 2024; 16:e66520. [PMID: 39246966 PMCID: PMC11380918 DOI: 10.7759/cureus.66520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction Bladder cancer is one of the most prevalent cancers worldwide, with significant morbidity and mortality rates. Treatment options for metastatic urothelial carcinoma (mUC) primarily include platinum-based chemotherapy. Cisplatin-based chemotherapy is conventionally used for treating mUC, but many patients are ineligible due to various factors such as poor performance status, creatinine clearance, neuropathy, and cardiac function. Carboplatin-based therapy is another alternative, which typically yields less favorable outcomes. Some centers use split-dose cisplatin for treating patients with comorbidities and impaired renal function, broadening cisplatin's spectrum. While eligibility criteria for full-dose cisplatin are well-established, those for split-dose cisplatin and carboplatin lack strong evidence. This study aims to assess the recommended criteria for full-dose cisplatin, split-dose cisplatin, and carboplatin regimens in real-world settings, including hematological parameters for patients with mUC. Methods A cross-sectional web-based survey was conducted among 136 oncologists from 21 countries, assessing criteria such as creatinine clearance, Eastern Cooperative Oncology Group (ECOG) performance status (PS), neurotoxicity, hearing loss, heart failure classification, and hematological parameters. Results The survey revealed diverse preferences among 113 oncologists treating mUC, regarding the eligibility criteria for each chemotherapy regimen with 81% prioritizing full-dose cisplatin, 21% split-dose cisplatin, and 14% carboplatin regimens. Criteria for all three regimens included specific thresholds. For full-dose cisplatin, the preferred criteria included creatinine clearance ≥60 mL/min, ECOG PS ≤1, grade 1 neuropathy, grade 1 deafness, New York Heart Association (NYHA) heart failure ≤class II with ≥50% cardiac ejection fraction, and normal blood parameters. Split-dose cisplatin criteria were creatinine clearance ≥40 mL/min, ECOG PS ≤2, grade 1 neuropathy, grade 1 deafness, NYHA heart failure ≤class II with ≥50% cardiac ejection fraction, and normal blood parameters. Carboplatin eligibility criteria were creatinine clearance ≥30, ECOG PS ≤2, grade ≤2 neuropathy, grade ≤2 deafness, NYHA heart failure ≤class II with ≥50% cardiac ejection fraction, and normal blood parameters. Hematological parameters were deemed crucial for all regimens, particularly stringent for carboplatin-based chemotherapy. Conclusion The study underscores the importance of renal function and hematological parameters in determining chemotherapy eligibility for patients with mUC. It highlights the importance of precise treatment criteria in mUC management, with hematological factors playing a significant role. Standardized criteria and further research are warranted to optimize treatment outcomes and minimize adverse events associated with chemotherapy regimens. Understanding the preferences of oncologists globally can facilitate tailored treatment approaches and improve patient care in the management of mUC.
Collapse
Affiliation(s)
- Faisal Azam
- Department of Adult Medical Oncology, King Fahad Specialist Hospital, Dammam, SAU
| | - Hulayel Alharbi
- Department of Adult Medical Oncology, King Fahad Specialist Hospital, Dammam, SAU
| | | | - Abdul Rehman Zar Gul
- Department of Adult Medical Oncology, King Fahad Specialist Hospital, Dammam, SAU
- Department of Medical Oncology, National Cancer Center of Care and Research, Doha, QAT
| | - Nedal Bukhari
- Department of Adult Medical Oncology, King Fahad Specialist Hospital, Dammam, SAU
- Department of Medical Oncology, Prince Sultan Military Medical City, Riyadh, SAU
| | - Mohamed Ouda
- Department of Medical Affairs, Merck Limited Saudi Arabia, an Affiliate of Merck KGaA, Riyadh, SAU
| | - Syed Anwar Hussain
- Department of Adult Medical Oncology, King Fahad Specialist Hospital, Dammam, SAU
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, GBR
| | - Fahad Ibnshamsah
- Department of Adult Medical Oncology, King Fahad Specialist Hospital, Dammam, SAU
| |
Collapse
|
20
|
Oliveira CA, Mercês ÉAB, Portela FS, Malheiro LFL, Silva HBL, De Benedictis LM, De Benedictis JM, Silva CCDE, Santos ACL, Rosa DP, Velozo HS, de Jesus Soares T, de Brito Amaral LS. An integrated view of cisplatin-induced nephrotoxicity, hepatotoxicity, and cardiotoxicity: characteristics, common molecular mechanisms, and current clinical management. Clin Exp Nephrol 2024; 28:711-727. [PMID: 38678166 DOI: 10.1007/s10157-024-02490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/18/2024] [Indexed: 04/29/2024]
Abstract
Cisplatin (CP) is a chemotherapy drug widely prescribed to treat various neoplasms. Although fundamental for the therapeutic action of the drug, its cytotoxic mechanisms trigger adverse effects in several tissues, such as the kidney, liver, and heart, which limit its clinical use. In this sense, studies point to an essential role of damage to nuclear and mitochondrial DNA associated with oxidative stress, inflammation, and apoptosis in the pathophysiology of tissue injuries. Due to the limitation of effective preventive and therapeutic measures against CP-induced toxicity, new strategies with potential cytoprotective effects have been studied. Therefore, this article is timely in reviewing the characteristics and main molecular mechanisms common to renal, hepatic, and cardiac toxicity previously described, in addition to addressing the main validated strategies for the current management of these adverse events in clinical practice. We also handle the main promising antioxidant substances recently presented in the literature to encourage the development of new research that consolidates their potential preventive and therapeutic effects against CP-induced cytotoxicity.
Collapse
Affiliation(s)
- Caroline Assunção Oliveira
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Érika Azenathe Barros Mercês
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Fernanda Santos Portela
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Lara Fabiana Luz Malheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | | | | | | | | | | | | | - Helloisa Souza Velozo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Liliany Souza de Brito Amaral
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
| |
Collapse
|
21
|
Zhang B, Hu Y, Du H, Han S, Ren L, Cheng H, Wang Y, Gao X, Zheng S, Cui Q, Tian L, Liu T, Sun J, Chai R. Tissue engineering strategies for spiral ganglion neuron protection and regeneration. J Nanobiotechnology 2024; 22:458. [PMID: 39085923 PMCID: PMC11293049 DOI: 10.1186/s12951-024-02742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
Cochlear implants can directly activate the auditory system's primary sensory neurons, the spiral ganglion neurons (SGNs), via circumvention of defective cochlear hair cells. This bypass restores auditory input to the brainstem. SGN loss etiologies are complex, with limited mammalian regeneration. Protecting and revitalizing SGN is critical. Tissue engineering offers a novel therapeutic strategy, utilizing seed cells, biomolecules, and scaffold materials to create a cellular environment and regulate molecular cues. This review encapsulates the spectrum of both human and animal research, collating the factors contributing to SGN loss, the latest advancements in the utilization of exogenous stem cells for auditory nerve repair and preservation, the taxonomy and mechanism of action of standard biomolecules, and the architectural components of scaffold materials tailored for the inner ear. Furthermore, we delineate the potential and benefits of the biohybrid neural interface, an incipient technology in the realm of implantable devices. Nonetheless, tissue engineering requires refined cell selection and differentiation protocols for consistent SGN quality. In addition, strategies to improve stem cell survival, scaffold biocompatibility, and molecular cue timing are essential for biohybrid neural interface integration.
Collapse
Affiliation(s)
- Bin Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yangnan Hu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Haoliang Du
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
| | - Shanying Han
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lei Ren
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Hong Cheng
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yusong Wang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xin Gao
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Shasha Zheng
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Qingyue Cui
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Lei Tian
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Tingting Liu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Jiaqiang Sun
- Department of Otolaryngology-Head and Neck Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China.
| |
Collapse
|
22
|
Goya L, Mateos R. Antioxidant and Anti-inflammatory Effects of Marine Phlorotannins and Bromophenols Supportive of Their Anticancer Potential. Nutr Rev 2024:nuae066. [PMID: 38894623 DOI: 10.1093/nutrit/nuae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024] Open
Abstract
Following the goal of optimizing nutrition, the food industry has been continuously working on food reformulation, nutritional patterns, functional foods development, and the general promotion of a healthy lifestyle. To this end, the scientific community has been increasingly investigating natural compounds that could prevent or treat chronic diseases. Phlorotannins and bromophenols are phenolic compounds particularly present in marine organisms. There is extensive evidence that shows their potential in the prevention of noncommunicable diseases, including cancer, the second cause of mortality worldwide. Numerous studies have demonstrated the anticarcinogenic activity of polyphenolic algae compounds both in cell culture and experimental animal models. Although recent reviews are also available, the present update focuses on the most recent findings related to the antioxidant/anti-inflammatory effect of seaweed phenolics, as well as their regulatory capacity for new molecular targets. Additionally, the review addresses and discusses the close link between inflammation and oxidative stress, along with their relationship with tumor onset and progression, including the most recent findings supporting this correlation. Although clinical studies are still needed to support this evidence, phlorotannins and bromophenols constitute an emerging bioactive group with high potential as chemopreventive agents and/or potential adjuvants for existing cancer therapies.
Collapse
Affiliation(s)
- Luis Goya
- Department of Metabolism and Nutrition, Institute of Food Science, Technology, and Nutrition (ICTAN-CSIC), Spanish National Research Council (CSIC), 28040 Madrid, Spain
| | - Raquel Mateos
- Department of Metabolism and Nutrition, Institute of Food Science, Technology, and Nutrition (ICTAN-CSIC), Spanish National Research Council (CSIC), 28040 Madrid, Spain
| |
Collapse
|
23
|
Mohtadi S, Salehcheh M, Tabandeh MR, Khorsandi L, Khodayar MJ. Ketotifen counteracts cisplatin-induced acute kidney injury in mice via targeting NF-κB/NLRP3/Caspase-1 and Bax/Bcl2/Caspase-3 signaling pathways. Biomed Pharmacother 2024; 175:116797. [PMID: 38776675 DOI: 10.1016/j.biopha.2024.116797] [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: 02/15/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024] Open
Abstract
Cisplatin (CIS) stands as one of the most effective chemotherapy drugs currently available. Despite its anticancer properties, the clinical application of CIS is restricted due to nephrotoxicity. Our research aimed to specify the impact of ketotifen fumarate (KET) against nephrotoxicity induced by CIS in mice. Male NMRI mice were treated with KET (0.4, 0.8, and 1.6 mg/kg, ip) for seven days. On the fourth day of the study, a single dose of CIS (13 mg/kg, ip) was administered, and the mice were sacrificed on the eighth day. The results indicated that administration of KET attenuated CIS-induced elevation of BUN and Cr in the serum, as well as renal KIM-1 levels. This improvement was accompanied by a significant reduction in kidney tissue damage, which was supported by histopathological examinations. Likewise, the decrease in the ratio of GSH to GSSG and antioxidant enzyme activities (CAT, SOD, and GPx), and the increase in lipid peroxidation marker (TBARS) were reversed in KET-treated mice. The ELISA results revealed that KET-treated mice ameliorated CIS-induced elevation in the renal levels of TNF-α, IL-1β, and IL-18. Western blot analysis exhibited that KET suppressed the activation of the transcription factor NF-κB and the NLRP3 inflammasome in the kidney of CIS-treated mice. Moreover, KET treatment reversed the changes in the protein expression of markers related to apoptosis (Bax, Bcl2, Caspase-3, and p53). Interestingly, KET significantly enhanced the cytotoxicity of CIS in HeLa cells. In conclusion, this study provides valuable insights into the promising effects of KET in mitigating CIS-induced nephrotoxicity.
Collapse
Affiliation(s)
- Shokooh Mohtadi
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Salehcheh
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Reza Tabandeh
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran; Stem Cells and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Layasadat Khorsandi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Khodayar
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
24
|
Maruyama A, Kawashima Y, Fukunaga Y, Makabe A, Nishio A, Tsutsumi T. Susceptibility of mouse cochlear hair cells to cisplatin ototoxicity largely depends on sensory mechanoelectrical transduction channels both Ex Vivo and In Vivo. Hear Res 2024; 447:109013. [PMID: 38718672 DOI: 10.1016/j.heares.2024.109013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/28/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024]
Abstract
Cisplatin, a highly effective chemotherapeutic drug for various human cancers, induces irreversible sensorineural hearing loss as a side effect. Currently there are no highly effective clinical strategies for the prevention of cisplatin-induced ototoxicity. Previous studies have indicated that short-term cisplatin ototoxicity primarily affects the outer hair cells of the cochlea. Therefore, preventing the entry of cisplatin into hair cells may be a promising strategy to prevent cisplatin ototoxicity. This study aimed to investigate the entry route of cisplatin into mouse cochlear hair cells. The competitive inhibitor of organic cation transporter 2 (OCT2), cimetidine, and the sensory mechanoelectrical transduction (MET) channel blocker benzamil, demonstrated a protective effect against cisplatin toxicity in hair cells in cochlear explants. Sensory MET-deficient hair cells explanted from Tmc1Δ;Tmc2Δ mice were resistant to cisplatin toxicity. Cimetidine showed an additive protective effect against cisplatin toxicity in sensory MET-deficient hair cells. However, in the apical turn, cimetidine, benzamil, or genetic ablation of sensory MET channels showed limited protective effects, implying the presence of other entry routes for cisplatin to enter the hair cells in the apical turn. Systemic administration of cimetidine failed to protect cochlear hair cells from ototoxicity caused by systemically administered cisplatin. Notably, outer hair cells in MET-deficient mice exhibited no apparent deterioration after systemic administration of cisplatin, whereas the outer hair cells in wild-type mice showed remarkable deterioration. The susceptibility of mouse cochlear hair cells to cisplatin ototoxicity largely depends on the sensory MET channel both ex vivo and in vivo. This result justifies the development of new pharmaceuticals, such as a specific antagonists for sensory MET channels or custom-designed cisplatin analogs which are impermeable to sensory MET channels.
Collapse
MESH Headings
- Cisplatin/toxicity
- Animals
- Ototoxicity/prevention & control
- Ototoxicity/metabolism
- Ototoxicity/physiopathology
- Mechanotransduction, Cellular/drug effects
- Organic Cation Transporter 2/metabolism
- Organic Cation Transporter 2/genetics
- Organic Cation Transporter 2/antagonists & inhibitors
- Cimetidine/pharmacology
- Antineoplastic Agents/toxicity
- Hair Cells, Auditory/drug effects
- Hair Cells, Auditory/metabolism
- Hair Cells, Auditory/pathology
- Hair Cells, Auditory, Outer/drug effects
- Hair Cells, Auditory, Outer/pathology
- Hair Cells, Auditory, Outer/metabolism
- Mice, Inbred C57BL
- Mice
- Membrane Proteins
Collapse
Affiliation(s)
- Ayako Maruyama
- Department of Otolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Yoshiyuki Kawashima
- Department of Otolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan.
| | - Yoko Fukunaga
- Department of Otolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan; Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54, Kawara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ayane Makabe
- Department of Otolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Ayako Nishio
- Department of Otolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Takeshi Tsutsumi
- Department of Otolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| |
Collapse
|
25
|
Patel S, Sathyanathan V, Salaman SD. Molecular mechanisms underlying cisplatin-induced nephrotoxicity and the potential ameliorative effects of essential oils: A comprehensive review. Tissue Cell 2024; 88:102377. [PMID: 38626527 DOI: 10.1016/j.tice.2024.102377] [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: 11/17/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/18/2024]
Abstract
Since the Middle Ages, essential oils (EO) have been widely used for bactericidal, virucidal, fungicidal, insecticidal, medicinal and cosmetic applications, nowadays in pharmaceutical, agricultural and food industries. Recently, EO have emerged as promising adjuvant therapies to mitigate the toxicities induced by anti - cancerous drugs; among them cisplatin induced renal damage amelioration remain remarkable. Cisplatin (cis-diaminedichloroplatinum II, CDDP) is renowned as one of the most effective anti-neoplastic agents, widely used as a broad-spectrum anti-tumor agent for various solid tumors. However, its clinical use is hampered by several side effects, notably nephrotoxicity and acute kidney injury, which arise from the accumulation of CDDP in the proximal tubular epithelial cells (PTECs). To better understand and analyze the molecular mechanisms of CDDP-induced renal damage, it is crucial to investigate potential interventions to protect against cisplatin-mediated nephrotoxicity. These EO have shown the ability to counteract oxidative stress, reduce inflammation, prevent apoptosis, and exert estrogenic effects, all contributing to renal protection. In this review, we have made an effort to summarize the molecular mechanisms and exploring new interventions by which we can pave the way for safer and more effective cancer management in the future.
Collapse
Affiliation(s)
- Saraswati Patel
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, India.
| | - V Sathyanathan
- Department of Pharmacognosy, Apollo College of Pharmacy, Kanchipuram, Tamil Nadu, India
| | - Samsi D Salaman
- Department of Pharmacognosy, Apollo College of Pharmacy, Kanchipuram, Tamil Nadu, India
| |
Collapse
|
26
|
Yao Z, Xiao Y, Li W, Kong S, Tu H, Guo S, Liu Z, Ma L, Qiao R, Wang S, Chang M, Zhao X, Zhang Y, Xu L, Sun D, Fu X. FDA-Approved Tedizolid Phosphate Prevents Cisplatin-Induced Hearing Loss Without Decreasing Its Anti-tumor Effect. J Assoc Res Otolaryngol 2024; 25:259-275. [PMID: 38622383 DOI: 10.1007/s10162-024-00945-2] [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: 08/02/2023] [Accepted: 03/04/2024] [Indexed: 04/17/2024] Open
Abstract
PURPOSE Cisplatin is a low-cost clinical anti-tumor drug widely used to treat solid tumors. However, its use could damage cochlear hair cells, leading to irreversible hearing loss. Currently, there appears one drug approved in clinic only used for reducing ototoxicity associated with cisplatin in pediatric patients, which needs to further explore other candidate drugs. METHODS Here, by screening 1967 FDA-approved drugs to protect cochlear hair cell line (HEI-OC1) from cisplatin damage, we found that Tedizolid Phosphate (Ted), a drug indicated for the treatment of acute infections, had the best protective effect. Further, we evaluated the protective effect of Ted against ototoxicity in mouse cochlear explants, zebrafish, and adult mice. The mechanism of action of Ted was further explored using RNA sequencing analysis and verified. Meanwhile, we also observed the effect of Ted on the anti-tumor effect of cisplatin. RESULTS Ted had a strong protective effect on hair cell (HC) loss induced by cisplatin in zebrafish and mouse cochlear explants. In addition, when administered systemically, it protected mice from cisplatin-induced hearing loss. Moreover, antitumor studies showed that Ted had no effect on the antitumor activity of cisplatin both in vitro and in vivo. RNA sequencing analysis showed that the otoprotective effect of Ted was mainly achieved by inhibiting phosphorylation of ERK. Consistently, ERK activator aggravated the damage of cisplatin to HCs. CONCLUSION Collectively, these results showed that FDA-approved Ted protected HCs from cisplatin-induced HC loss by inhibiting ERK phosphorylation, indicating its potential as a candidate for preventing cisplatin ototoxicity in clinical settings.
Collapse
Affiliation(s)
- Zhiwei Yao
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Yu Xiao
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
- School of Life Science, Shandong University, Qingdao, 266237, China
| | - Wen Li
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China.
| | - Shuhui Kong
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250000, China
| | - Hailong Tu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Siwei Guo
- School of Life Science, Shandong University, Qingdao, 266237, China
| | - Ziyi Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Lushun Ma
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Ruifeng Qiao
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250000, China
| | - Song Wang
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Miao Chang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Xiaoxu Zhao
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yuan Zhang
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Lei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250000, China.
| | - Daqing Sun
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Xiaolong Fu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China.
| |
Collapse
|
27
|
Kedeshian K, Hong M, Hoffman L, Kita A. N-acetylcysteine microparticles reduce cisplatin-induced RSC96 Schwann cell toxicity. Laryngoscope Investig Otolaryngol 2024; 9:e1256. [PMID: 38765675 PMCID: PMC11099882 DOI: 10.1002/lio2.1256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/03/2023] [Accepted: 04/16/2024] [Indexed: 05/22/2024] Open
Abstract
Objectives Cisplatin is known to cause inner ear dysfunction. There is growing evidence that cisplatin-induced demyelination of spiral or Scarpa's ganglion neurons may play an additional role in drug-induced ototoxicity alongside afferent neuron injury. As Schwann cells produce myelin, there may be an opportunity to reduce ototoxic inner ear damage by promoting Schwann cell viability. This work describes a cellular model of cisplatin-induced Schwann cell injury and investigates the ability of the antioxidant N-acetylcysteine to promote Schwann cell viability. A local delivery system of drug-eluting microparticles was then fabricated, characterized, and investigated for bioactivity. Methods RSC96 rat Schwann cells were dosed with varying concentrations of cisplatin to obtain a dose curve and identify the lethal concentration of 50% of the cells (LC50). In subsequent experiments, RSC96 cells were co-treated with cisplatin and both resuspended or eluted N-acetylcysteine. Cell viability was assessed with the CCK8 assay. Results The LC50 dose of cisplatin was determined to be 3.76 μM (p = 2.2 x 10-16). When co-dosed with cisplatin and a therapeutic concentration of resuspended or eluted N-acetylcysteine, Schwann cells had an increased viability compared to cells dosed with cisplatin alone. Conclusion RSC96 Schwann cell injury following cisplatin insult is characterized in this in vitro model. Cisplatin caused injury at physiologic concentrations and N-acetylcysteine improved cell viability and mitigated this injury. N-acetylcysteine was packaged into microparticles and eluted N-acetylcysteine retained its ability to increase cell viability, thus demonstrating promise as a therapeutic to offset cisplatin-induced ototoxicity. Level of Evidence N/A Laryngoscope, 2023.
Collapse
Affiliation(s)
- Katherine Kedeshian
- Department of Head and Neck SurgeryDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCaliforniaUSA
| | - Michelle Hong
- Department of Head and Neck SurgeryDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCaliforniaUSA
| | - Larry Hoffman
- Department of Head and Neck SurgeryDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCaliforniaUSA
- Vestibular Neuroscience Laboratory, Brain Research InstituteDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCaliforniaUSA
| | - Ashley Kita
- Department of Head and Neck SurgeryDavid Geffen School of Medicine at University of California Los AngelesLos AngelesCaliforniaUSA
| |
Collapse
|
28
|
Saeed Y, Zhong R, Sun Z. Advances in traditional herbal formulation based nano-vaccine for cancer immunotherapy: Unraveling the enigma of complex tumor environment and multidrug resistance. Int Immunopharmacol 2024; 132:111948. [PMID: 38554445 DOI: 10.1016/j.intimp.2024.111948] [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: 01/19/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/01/2024]
Abstract
Cancer is attributed to uncontrolled cell growth and is among the leading causes of death with no known effective treatment while complex tumor microenvironment (TME) and multidrug resistance (MDR) are major challenges for developing an effective therapeutic strategy. Advancement in cancer immunotherapy has been limited by the over-activation of the host immune response that ultimately affects healthy tissues or organs and leads to a feeble response of the patient's immune system against tumor cells. Besides, traditional herbal medicines (THM) have been well-known for their essential role in the treatment of cancer and are considered relatively safe due to their compatibility with the human body. Yet, poor solubility, low bio-availability, and lack of understanding about their pathophysiological mechanism halt their clinical application. Moreover, considering the complex TME and drug resistance, the most precarious and least discussed concerns for developing THM-based nano-vaccination, are identification of specific biomarkers for drug inhibitory protein and targeted delivery of bioactive ingredients of THM on the specific sites in tumor cells. The concept of THM-based nano-vaccination indicates immunomodulation of TME by THM-based bioactive adjuvants, exerting immunomodulatory effects, via targeted inhibition of key proteins involved in the metastasis of cancer. However, this concept is at its nascent stage and very few preclinical studies provided the evidence to support clinical translation. Therefore, we attempted to capsulize previously reported studies highlighting the role of THM-based nano-medicine in reducing the risk of MDR and combating complex tumor environments to provide a reference for future study design by discussing the challenges and opportunities for developing an effective and safe therapeutic strategy against cancer.
Collapse
Affiliation(s)
- Yasmeen Saeed
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan 512005, China.
| | - Ruimin Zhong
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan 512005, China
| | - Zhanghua Sun
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan 512005, China
| |
Collapse
|
29
|
Pasdelou MP, Byelyayeva L, Malmström S, Pucheu S, Peytavy M, Laullier H, Hodges DB, Tzafriri AR, Naert G. Ototoxicity: a high risk to auditory function that needs to be monitored in drug development. Front Mol Neurosci 2024; 17:1379743. [PMID: 38756707 PMCID: PMC11096496 DOI: 10.3389/fnmol.2024.1379743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Hearing loss constitutes a major global health concern impacting approximately 1.5 billion people worldwide. Its incidence is undergoing a substantial surge with some projecting that by 2050, a quarter of the global population will experience varying degrees of hearing deficiency. Environmental factors such as aging, exposure to loud noise, and the intake of ototoxic medications are implicated in the onset of acquired hearing loss. Ototoxicity resulting in inner ear damage is a leading cause of acquired hearing loss worldwide. This could be minimized or avoided by early testing of hearing functions in the preclinical phase of drug development. While the assessment of ototoxicity is well defined for drug candidates in the hearing field - required for drugs that are administered by the otic route and expected to reach the middle or inner ear during clinical use - ototoxicity testing is not required for all other therapeutic areas. Unfortunately, this has resulted in more than 200 ototoxic marketed medications. The aim of this publication is to raise awareness of drug-induced ototoxicity and to formulate some recommendations based on available guidelines and own experience. Ototoxicity testing programs should be adapted to the type of therapy, its indication (targeting the ear or part of other medications classes being potentially ototoxic), and the number of assets to test. For multiple molecules and/or multiple doses, screening options are available: in vitro (otic cell assays), ex vivo (cochlear explant), and in vivo (in zebrafish). In assessing the ototoxicity of a candidate drug, it is good practice to compare its ototoxicity to that of a well-known control drug of a similar class. Screening assays provide a streamlined and rapid method to know whether a drug is generally safe for inner ear structures. Mammalian animal models provide a more detailed characterization of drug ototoxicity, with a possibility to localize and quantify the damage using functional, behavioral, and morphological read-outs. Complementary histological measures are routinely conducted notably to quantify hair cells loss with cochleogram. Ototoxicity studies can be performed in rodents (mice, rats), guinea pigs and large species. However, in undertaking, or at the very least attempting, all preclinical investigations within the same species, is crucial. This encompasses starting with pharmacokinetics and pharmacology efficacy studies and extending through to toxicity studies. In life read-outs include Auditory Brainstem Response (ABR) and Distortion Product OtoAcoustic Emissions (DPOAE) measurements that assess the activity and integrity of sensory cells and the auditory nerve, reflecting sensorineural hearing loss. Accurate, reproducible, and high throughput ABR measures are fundamental to the quality and success of these preclinical trials. As in humans, in vivo otoscopic evaluations are routinely carried out to observe the tympanic membrane and auditory canal. This is often done to detect signs of inflammation. The cochlea is a tonotopic structure. Hair cell responsiveness is position and frequency dependent, with hair cells located close to the cochlea apex transducing low frequencies and those at the base transducing high frequencies. The cochleogram aims to quantify hair cells all along the cochlea and consequently determine hair cell loss related to specific frequencies. This measure is then correlated with the ABR & DPOAE results. Ototoxicity assessments evaluate the impact of drug candidates on the auditory and vestibular systems, de-risk hearing loss and balance disorders, define a safe dose, and optimize therapeutic benefits. These types of studies can be initiated during early development of a therapeutic solution, with ABR and otoscopic evaluations. Depending on the mechanism of action of the compound, studies can include DPOAE and cochleogram. Later in the development, a GLP (Good Laboratory Practice) ototoxicity study may be required based on otic related route of administration, target, or known potential otic toxicity.
Collapse
|
30
|
Meijer AJM, Diepstraten FA, van den Heuvel-Eibrink MM, Bleyer A. Prevention of cisplatin-induced hearing loss in children: achievements and challenges for evidence-based implementation of sodium Thiosulfate. Front Oncol 2024; 14:1336714. [PMID: 38562178 PMCID: PMC10982326 DOI: 10.3389/fonc.2024.1336714] [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: 11/11/2023] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Ototoxicity is a devastating direct, irreversible side effect of platinum use in children with cancer, with its consequent effect on speech, language and social development, quality of life and adult productivity. Cisplatin, an essential chemotherapeutic agent for the treatment of solid tumors in children, is a DNA cross-linking agent. Which causes hearing loss in 50-70% of cisplatin treated children. Fortunately, to prevent hearing loss, sodium thiosulfate (STS), which binds to cisplatin, and reduces the superoxides in both tumor and outer hair cells of the cochlea has now been discovered to be an effective and safe otoprotectant if administered correctly. The aim of this perspective paper is to explore the key safety issues and challenges important for pediatric oncologists and pharmacists when considering the clinical use of STS as an otoprotectant for children and adolescents receiving cisplatin. These include: the choice of the formulation; the timing, both that of the STS in relation to cisplatin as well as the timing of the cisplatin infusion itself; the dosing; the challenge left by the definition of localized versus disseminated disease and the difference in indication for STS, between cisplatin treated patients and those receiving another platinum chemotherapeutic agent, carboplatin.
Collapse
Affiliation(s)
| | | | - Marry M. van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Wilhelmina Childrens Hospital, University of Utrecht, Utrecht, Netherlands
| | - Archie Bleyer
- Radiation Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
31
|
Dutta A, Thakur S, Dey DK, Kumar A. Cisplatin and Starvation Differently Sensitize Autophagy in Renal Carcinoma: A Potential Therapeutic Pathway to Target Variegated Drugs Resistant Cancerous Cells. Cells 2024; 13:471. [PMID: 38534315 DOI: 10.3390/cells13060471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/26/2024] [Accepted: 03/03/2024] [Indexed: 03/28/2024] Open
Abstract
Cisplatin, a powerful chemotherapy medication, has long been a cornerstone in the fight against cancer due to chemotherapeutic failure. The mechanism of cisplatin resistance/failure is a multifaceted and complex issue that consists mainly of apoptosis inhibition through autophagy sensitization. Currently, researchers are exploring ways to regulate autophagy in order to tip the balance in favor of effective chemotherapy. Based on this notion, the current study primarily identifies the differentially expressed genes (DEGs) in cisplatin-treated autophagic ACHN cells through the Illumina Hi-seq platform. A protein-protein interaction network was constructed using the STRING database and KEGG. GO classifiers were implicated to identify genes and their participating biological pathways. ClueGO, David, and MCODE detected ontological enrichment and sub-networking. The network topology was further examined using 12 different algorithms to identify top-ranked hub genes through the Cytoscape plugin Cytohubba to identify potential targets, which established profound drug efficacy under an autophagic environment. Considerable upregulation of genes related to autophagy and apoptosis suggests that autophagy boosts cisplatin efficacy in malignant ACHN cells with minimal harm to normal HEK-293 growth. Furthermore, the determination of cellular viability and apoptosis by AnnexinV/FITC-PI assay corroborates with in silico data, indicating the reliability of the bioinformatics method followed by qRT-PCR. Altogether, our data provide a clear molecular insight into drug efficacy under starved conditions to improve chemotherapy and will likely prompt more clinical trials on this aspect.
Collapse
Affiliation(s)
- Ankita Dutta
- Advanced Nanoscale Molecular Oncology Laboratory (ANMOL), Department of Biotechnology, University of North Bengal, Siliguri 734013, West Bengal, India
| | - Subarna Thakur
- Department of Bioinformatics, University of North Bengal, Siliguri 734013, West Bengal, India
| | - Debasish Kumar Dey
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Anoop Kumar
- Advanced Nanoscale Molecular Oncology Laboratory (ANMOL), Department of Biotechnology, University of North Bengal, Siliguri 734013, West Bengal, India
| |
Collapse
|
32
|
Luo Y, Zhang J, Jiao Y, Huang H, Ming L, Song Y, Niu Y, Tang X, Liu L, Li Y, Jiang Y. Dihydroartemisinin abolishes cisplatin-induced nephrotoxicity in vivo. J Nat Med 2024; 78:439-454. [PMID: 38351420 DOI: 10.1007/s11418-024-01783-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/15/2024] [Indexed: 02/29/2024]
Abstract
Dihydroartemisinin (DHA), a derivative of artemisinin which is primarily used to treat malaria in clinic, also confers protective effect on lipopolysaccharide-induced nephrotoxicity. While, the activities of DHA in cisplatin (CDDP)-caused nephrotoxicity are elusive. To investigate the role and underlying mechanism of DHA in CDDP-induced nephrotoxicity. Mice were randomly separated into four groups: normal, CDDP, and DHA (25 and 50 mg/kg were orally injected 1 h before CDDP for consecutive 10 days). All mice except the normal were single injected intraperitoneally with CDDP (22 mg/kg) for once on the 7th day. Combined with quantitative proteomics and bioinformatics analysis, the impact of DHA on renal cell apoptosis, oxidative stress, biochemical indexes, and inflammation in mice were investigated. Moreover, a human hepatocellular carcinoma cells xenograft model was established to elucidate the impact of DHA on tumor-related effects of CDDP. DHA reduced the levels of creatinine (CREA) (p < 0.01) and blood urea nitrogen (BUN) (p < 0.01), reversed CDDP-induced oxidative, inflammatory, and apoptosis indexes (p < 0.01). Mechanistically, DHA attenuated CDDP-induced inflammation by inhibiting nuclear factor κB p65 (NFκB p65) expression, and suppressed CDDP-induced renal cell apoptosis by inhibiting p63-mediated endogenous and exogenous apoptosis pathways. Additionally, DHA alone significantly decreased the tumor weight and did not destroy the antitumor effect of CDDP, and did not impact AST and ALT. In conclusion, DHA prevents CDDP-triggered nephrotoxicity via reducing inflammation, oxidative stress, and apoptosis. The mechanisms refer to inhibiting NFκB p65-regulated inflammation and alleviating p63-mediated mitochondrial endogenous and Fas death receptor exogenous apoptosis pathway.
Collapse
Affiliation(s)
- Yan Luo
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, School of Pharmacy, Scientific Research Center, Gannan Medical University, Ganzhou, 341000, China
| | - Jiaxing Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yue Jiao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research On Prevention and Treatment of Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Huang
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, Gannan Medical University, Jiangxi, China
| | - Liangshan Ming
- Institute for Advanced Study, Jiangxi University of Chinese Medicine, Jiangxi, China
| | - Yunlei Song
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, School of Pharmacy, Scientific Research Center, Gannan Medical University, Ganzhou, 341000, China
| | - Yanlong Niu
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, Gannan Medical University, Jiangxi, China
| | - Xiaolu Tang
- Department of Human Anatomy, School of Basic Medical Science, Gannan Medical University, Jiangxi, China
| | - Liwei Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Li
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| | - Yumao Jiang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, School of Pharmacy, Scientific Research Center, Gannan Medical University, Ganzhou, 341000, China.
- Artemisinin Research Center, and Institute of Chinese Materia Medical, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
33
|
Padovani BN, Morales Fénero C, Paredes LC, do Amaral MA, Domínguez-Amorocho O, Cipelli M, Gomes JMM, da Silva EM, Silva LM, Vieira RDS, Pereira MT, Cruz MC, Câmara NOS. Cisplatin Toxicity Causes Neutrophil-Mediated Inflammation in Zebrafish Larvae. Int J Mol Sci 2024; 25:2363. [PMID: 38397041 PMCID: PMC10889180 DOI: 10.3390/ijms25042363] [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: 12/22/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Cisplatin is an antineoplastic agent used to treat various tumors. In mammals, it can cause nephrotoxicity, tissue damage, and inflammation. The release of inflammatory mediators leads to the recruitment and infiltration of immune cells, particularly neutrophils, at the site of inflammation. Cisplatin is often used as an inducer of acute kidney injury (AKI) in experimental models, including zebrafish (Danio rerio), due to its accumulation in kidney cells. Current protocols in larval zebrafish focus on studying its effect as an AKI inducer but ignore other systematic outcomes. In this study, cisplatin was added directly to the embryonic medium to assess its toxicity and impact on systemic inflammation using locomotor activity analysis, qPCR, microscopy, and flow cytometry. Our data showed that larvae exposed to cisplatin at 7 days post-fertilization (dpf) displayed dose-dependent mortality and morphological changes, leading to a decrease in locomotion speed at 9 dpf. The expression of pro-inflammatory cytokines such as interleukin (il)-12, il6, and il8 increased after 48 h of cisplatin exposure. Furthermore, while a decrease in the number of neutrophils was observed in the glomerular region of the pronephros, there was an increase in neutrophils throughout the entire animal after 48 h of cisplatin exposure. We demonstrate that cisplatin can have systemic effects in zebrafish larvae, including morphological and locomotory defects, increased inflammatory cytokines, and migration of neutrophils from the hematopoietic niche to other parts of the body. Therefore, this protocol can be used to induce systemic inflammation in zebrafish larvae for studying new therapies or mechanisms of action involving neutrophils.
Collapse
Affiliation(s)
- Barbara Nunes Padovani
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil; (B.N.P.); (L.C.P.); (O.D.-A.); (M.C.); (L.M.S.); (R.d.S.V.); (M.T.P.); (M.C.C.)
| | - Camila Morales Fénero
- Department of Microbiology and Environmental Toxicology, Biomedical Sciences, University of California Santa, Santa Cruz, CA 95064, USA
| | - Lais Cavalieri Paredes
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil; (B.N.P.); (L.C.P.); (O.D.-A.); (M.C.); (L.M.S.); (R.d.S.V.); (M.T.P.); (M.C.C.)
| | - Mariana Abrantes do Amaral
- Nephrology Division, Department of Medicine, Federal University of Sao Paulo, São Paulo 04023062, Brazil; (M.A.d.A.); (E.M.d.S.)
| | - Omar Domínguez-Amorocho
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil; (B.N.P.); (L.C.P.); (O.D.-A.); (M.C.); (L.M.S.); (R.d.S.V.); (M.T.P.); (M.C.C.)
| | - Marcella Cipelli
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil; (B.N.P.); (L.C.P.); (O.D.-A.); (M.C.); (L.M.S.); (R.d.S.V.); (M.T.P.); (M.C.C.)
| | | | - Eloisa Martins da Silva
- Nephrology Division, Department of Medicine, Federal University of Sao Paulo, São Paulo 04023062, Brazil; (M.A.d.A.); (E.M.d.S.)
| | - Luísa Menezes Silva
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil; (B.N.P.); (L.C.P.); (O.D.-A.); (M.C.); (L.M.S.); (R.d.S.V.); (M.T.P.); (M.C.C.)
| | - Raquel de Souza Vieira
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil; (B.N.P.); (L.C.P.); (O.D.-A.); (M.C.); (L.M.S.); (R.d.S.V.); (M.T.P.); (M.C.C.)
| | - Mariana Tominaga Pereira
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil; (B.N.P.); (L.C.P.); (O.D.-A.); (M.C.); (L.M.S.); (R.d.S.V.); (M.T.P.); (M.C.C.)
| | - Mario Costa Cruz
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil; (B.N.P.); (L.C.P.); (O.D.-A.); (M.C.); (L.M.S.); (R.d.S.V.); (M.T.P.); (M.C.C.)
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508000, Brazil; (B.N.P.); (L.C.P.); (O.D.-A.); (M.C.); (L.M.S.); (R.d.S.V.); (M.T.P.); (M.C.C.)
| |
Collapse
|
34
|
Wang SX, Streit A. Shared features in ear and kidney development - implications for oto-renal syndromes. Dis Model Mech 2024; 17:dmm050447. [PMID: 38353121 PMCID: PMC10886756 DOI: 10.1242/dmm.050447] [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] [Indexed: 02/16/2024] Open
Abstract
The association between ear and kidney anomalies has long been recognized. However, little is known about the underlying mechanisms. In the last two decades, embryonic development of the inner ear and kidney has been studied extensively. Here, we describe the developmental pathways shared between both organs with particular emphasis on the genes that regulate signalling cross talk and the specification of progenitor cells and specialised cell types. We relate this to the clinical features of oto-renal syndromes and explore links to developmental mechanisms.
Collapse
Affiliation(s)
- Scarlet Xiaoyan Wang
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Andrea Streit
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| |
Collapse
|
35
|
Alqahtani NF, Alfaifi MY, Shati AA, Elbehairi SEI, Elshaarawy RFM, Serag WM, Hassan YA, El-Sayed WN. Exploring the chondroitin sulfate nanogel's potential in combating nephrotoxicity induced by cisplatin and doxorubicin-An in-vivo study on rats. Int J Biol Macromol 2024; 258:128839. [PMID: 38134998 DOI: 10.1016/j.ijbiomac.2023.128839] [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: 07/21/2023] [Revised: 12/02/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
In this study, we aim to unveil the potential of itaconyl chondroitin sulfate nanogel (ICSNG) in tackling chronic kidney diseases triggered by the administration of CDDP and doxorubicin (Adriamycin, ADR). To that end, the new drug delivery system (ICSNG) was initially prepared, characterized, and loaded with the target drugs. Thereafter, the in-vivo studies were performed using five equally divided groups of 100 male Sprague-Dawley (SD) rats. Biochemical evaluation and immunohistochemistry studies have revealed the renal toxicity and the ameliorative effects of ICSNG on renal function. When ICSNG-based treatments were contrasted with the CDDP and ADR infected groups, they significantly increased paraoxonase-1 (PON-1), superoxide dismutase (SOD), catalase (CAT) and albumin activity and significantly decreased nitric oxide (NO), tumor necrosis factor alpha (TNF-α), creatinine, urea, and cyclooxygenase-2 (COX-2) activity (p < 0.001). The findings of the current study imply that ICSNG may be able to lessen renal inflammation and damage in chronic kidney disorders brought on by the administration of CDDP and ADR. Interestingly, according to the estimated selectivity indices, the ICSNG-encapsulated drugs have demonstrated superior selectivity for cancer MCF-7 cells, over healthy HSF cells, in comparison to the bare drugs.
Collapse
Affiliation(s)
- Norah F Alqahtani
- Department of Chemistry, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - Ali A Shati
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | | | - Reda F M Elshaarawy
- Department of Chemistry, Faculty of Science, Suez University, 43533 Suez, Egypt; Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany.
| | - Waleed M Serag
- Department of Chemistry, Faculty of Science, Suez University, 43533 Suez, Egypt
| | - Yasser A Hassan
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Al-Kitab University, Kirkuk, Iraq; Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Al-Qalam University College, Kirkuk, Iraq; Department of pharmaceutics and Pharmaceutical Technology, Faculty of pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - W N El-Sayed
- Department of Chemistry, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| |
Collapse
|
36
|
Džidić-Krivić A, Sher EK, Kusturica J, Farhat EK, Nawaz A, Sher F. Unveiling drug induced nephrotoxicity using novel biomarkers and cutting-edge preventive strategies. Chem Biol Interact 2024; 388:110838. [PMID: 38104745 DOI: 10.1016/j.cbi.2023.110838] [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: 09/18/2023] [Revised: 12/03/2023] [Accepted: 12/15/2023] [Indexed: 12/19/2023]
Abstract
Drug-induced nephrotoxicity is still a significant obstacle in pharmacotherapy of various diseases and it accounts for around 25 % of serious side-effects reported after drug administration. Furthermore, some groups of drugs such as nonsteroidal anti-inflammatory drugs, antibiotics, antiviral drugs, antifungal drugs, immunosuppressants, and chemotherapeutic drugs have the "preference" for damaging the kidney and are often referred to as the kidney's "silent killer". Clinically, the onset of acute kidney injury associated with drug administration is registered in approximately 20 % of patients and many of them develop chronic kidney disease vulnerability. However, current knowledge about the mechanisms underlying this dangerous phenomenon is still insufficient with many unknowns. Hence, the valuable use of these drugs in clinical practice is significantly limited. The main aim of this study is to draw attention to commonly prescribed nephrotoxic drugs by clinicians or drugs bought over the counter. In addition, the complex relationship between immunological, vascular and inflammatory events that promote kidney damage is discussed. The practical use of this knowledge could be implemented in the engineering of novel biomarkers for early detection of drug-associated kidney damage such as Kidney Injury Molecule (KIM-1), lipocalin associated with neutrophil gelatinase (NGAL) and various microRNAs. In addition, the utilization of artificial intelligence (AI) for the development of computer algorithms that could detect kidney damage at an early stage should be further explored. Therefore, this comprehensive review provides a new outlook on drug nephrotoxicity that opens the door for further clinical research of novel potential drugs or natural products for the prevention of drug-induced nephrotoxicity and accessible education.
Collapse
Affiliation(s)
- Amina Džidić-Krivić
- Department of Neurology, Cantonal Hospital Zenica, Zenica, 72000, Bosnia and Herzegovina; International Society of Engineering Science and Technology, Nottingham, United Kingdom
| | - Emina K Sher
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, United Kingdom.
| | - Jasna Kusturica
- Faculty of Medicine,Univerisity of Sarajevo, Sarajevo, 71000, Bosnia and Herzegovina
| | - Esma K Farhat
- International Society of Engineering Science and Technology, Nottingham, United Kingdom; Department of Food and Nutrition Research, Faculty of Food Technology, Juraj Strossmayer University of Osijek, Osijek, 31000, Croatia
| | - Asma Nawaz
- International Society of Engineering Science and Technology, Nottingham, United Kingdom; Department of Biochemistry, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Farooq Sher
- Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, United Kingdom.
| |
Collapse
|
37
|
Hayashi D, Shirai T, Terauchi R, Tsuchida S, Mizoshiri N, Mori Y, Shimomura S, Mazda O, Takahashi K. A Natural Organic Compound "Decursin" Has Both Antitumor and Renal Protective Effects: Treatment for Osteosarcoma. JOURNAL OF ONCOLOGY 2023; 2023:5445802. [PMID: 38130464 PMCID: PMC10735716 DOI: 10.1155/2023/5445802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/24/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023]
Abstract
Osteosarcoma is a rare malignant tumor that commonly occurs in children. Anticancer drugs, for example, cisplatin, aid in postsurgery recovery but induce side effects such as renal damage, affecting the life prognosis of patients. Decursin which is one of the bioactive components has been reported for its anti-inflammatory, antioxidant, and antitumor effects, but the effect on osteosarcoma is unexplained. In this study, the research theme was to examine the sensitizing effect of decursin and its influence on cisplatin-induced nephrotoxicity. The cell viability and half maximal inhibitory concentration (IC50), apoptosis induction, and effect on cell cycle and Akt pathways were examined. In vivo, we examine the effects of decursin on tumors and mice bodies. Additionally, the effects of the cisplatin-decursin combination were evaluated in vitro and in vivo. Decursin suppressed cell viability and induced apoptosis via the cell cycle. Decursin also inhibited the Akt pathway by suppressing the phosphorylation of Akt. It enhanced apoptosis induction and lowered cell viability in combination with cisplatin. The increasing tumor volume was suppressed in the decursin-administrated group with further suppression in combination with cisplatin compared to sole cisplatin administration. The decrease in renal function and renal epithelial cell damage caused by cisplatin was improved by the combinatorial treatment with decursin. Therefore, decursin demonstrated an antitumor effect on the osteosarcoma cells and a renal protective effect in combination with cisplatin. Therefore, decursin is a prospective therapeutic agent against osteosarcoma.
Collapse
Affiliation(s)
- Daichi Hayashi
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshiharu Shirai
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Ryu Terauchi
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Shinji Tsuchida
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Naoki Mizoshiri
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yuki Mori
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Seiji Shimomura
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Osam Mazda
- Department of Immunology, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kenji Takahashi
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
38
|
Guidotti L, Tomassi E, Marracci S, Lai M, Lapi D, Pesi R, Pucci L, Novellino E, Albi E, Garcia-Gil M. Effects of Nutraceuticals on Cisplatin-Induced Cytotoxicity in HEI-OC1 Cells. Int J Mol Sci 2023; 24:17416. [PMID: 38139245 PMCID: PMC10743635 DOI: 10.3390/ijms242417416] [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: 11/09/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Cisplatin is a chemotherapeutic drug for the treatment of several solid tumors, whose use is limited by its nephrotoxicity, neurotoxicity, ototoxicity, and development of resistance. The toxicity is caused by DNA cross-linking, increase in reactive oxygen species and/or depletion of cell antioxidant defenses. The aim of the work was to study the effect of antioxidant compounds (Lisosan G, Taurisolo®) or hydrogen sulfide (H2S)-releasing compounds (erucin) in the auditory HEI-OC1 cell line treated with cisplatin. Cell viability was determined using the MTT assay. Caspase and sphingomyelinase activities were measured by fluorometric and colorimetric methods, respectively. Expression of transcription factors, apoptosis hallmarks and genes codifying for antioxidant response proteins were measured by Western blot and/or RT-qPCR. Lisosan G, Taurisolo® and erucin did not show protective effects. Sodium hydrosulfide (NaHS), a donor of H2S, increased the viability of cisplatin-treated cells and the transcription of heme oxygenase 1, superoxide dismutase 2, NAD(P)H quinone dehydrogenase type 1 and the catalytic subunit of glutamate-cysteine ligase and decreased reactive oxygen species (ROS), the Bax/Bcl2 ratio, caspase-3, caspase-8 and acid sphingomyelinase activity. Therefore, NaHS might counteract the cytotoxic effect of cisplatin by increasing the antioxidant response and by reducing ROS levels and caspase and acid sphingomyelinase activity.
Collapse
Affiliation(s)
- Lorenzo Guidotti
- General Physiology Unit, Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy; (L.G.); (S.M.); (D.L.)
| | - Elena Tomassi
- Institute of Agricultural Biology and Biotechnology, Italian National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (E.T.); (L.P.)
| | - Silvia Marracci
- General Physiology Unit, Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy; (L.G.); (S.M.); (D.L.)
| | - Michele Lai
- Retrovirus Centre, Department of Translational Medicine and New Technologies in Medicine and Surgery, University of Pisa, Strada Statale del Brennero 2, 56127 Pisa, Italy;
| | - Dominga Lapi
- General Physiology Unit, Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy; (L.G.); (S.M.); (D.L.)
| | - Rossana Pesi
- Biochemistry Unit, Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy;
| | - Laura Pucci
- Institute of Agricultural Biology and Biotechnology, Italian National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (E.T.); (L.P.)
| | - Ettore Novellino
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Elisabetta Albi
- Department of Pharmaceutical Sciences, Interno Orto Botanico, University of Perugia, Via Romana, 06126 Perugia, Italy;
| | - Mercedes Garcia-Gil
- General Physiology Unit, Department of Biology, University of Pisa, Via San Zeno 31, 56127 Pisa, Italy; (L.G.); (S.M.); (D.L.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56124 Pisa, Italy
| |
Collapse
|
39
|
Kushwaha R, Singh V, Peters S, Yadav AK, Dolui D, Saha S, Sarkar S, Dutta A, Koch B, Sadhukhan T, Banerjee S. Density Functional Theory-Guided Photo-Triggered Anticancer Activity of Curcumin-Based Zinc(II) Complexes. J Phys Chem B 2023; 127:10266-10278. [PMID: 37988143 DOI: 10.1021/acs.jpcb.3c02382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Photodynamic therapy (PDT) has evolved as a new therapeutic modality for cancer treatment with fewer side effects and drug resistance. Curcumin exhibits PDT activity, but its low bioavailability restricts its clinical application. Here, the bioavailability of curcumin was increased by its complex formation with the Zn(II) center. For a structure-activity relationship study, Zn(II)-based complexes (1-3) comprising N^N-based ligands (2,2'-bipyridine in 1 and 2 or 1,10-phenanthroline in 3) and O^O-based ligands (acetylacetone in 1, monoanionic curcumin in 2 and 3) were synthesized and thoroughly characterized. The X-ray structure of the control complex, 1, indicated a square pyramidal shape of the molecules. Photophysical and TD-DFT studies indicated the potential of 2 and 3 as good visible light type-II photosensitizers for PDT. Guided by the TD-DFT studies, the low-energy visible light-triggered singlet oxygen (1O2) generation efficacy of 2 and 3 was explored in solution and in cancer cells. As predicted by the TD-DFT calculations, these complexes produced 1O2 efficiently in the cytosol of MCF-7 cancer cells and ultimately displayed excellent apoptotic anticancer activity in the presence of light. Moreover, the molecular docking investigation showed that complexes 2 and 3 have very good binding affinities with caspase-9 and p-53 proteins and could activate them for cellular apoptosis. Further molecular dynamics simulations confirmed the stability of 3 in the caspase-9 protein binding site.
Collapse
Affiliation(s)
- Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Virendra Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Silda Peters
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Ashish K Yadav
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Dependu Dolui
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| | - Sukanta Saha
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| | - Sujit Sarkar
- Prescience Insilico Pvt. Ltd., Bengaluru, Karnataka 560066, India
| | - Arnab Dutta
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| | - Biplob Koch
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Tumpa Sadhukhan
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
40
|
Han JM, Song HY, Kim KI, Byun EB. Protective Effects of Bombyx batryticatus Protein-Rich Extract Against Cisplatin-Induced Nephrotoxicity in HEK293 Cells and a Mouse Model. J Med Food 2023; 26:927-938. [PMID: 38064431 DOI: 10.1089/jmf.2023.k.0182] [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] [Indexed: 12/18/2023] Open
Abstract
Cisplatin, a potent and prominent chemotherapeutic drug, has considerable side effects, including nephrotoxicity, which limits its therapeutic application and efficacy. Therefore, the development of agents that protect normal cells while preserving cisplatin's chemotherapeutic properties is of utmost importance. This study aimed to explore the protective effects of Bombyx batryticatus protein-rich extract (BBPE) against cisplatin-induced nephrotoxicity in a cisplatin-treated mouse model and human embryonic kidney (HEK293) cells. Apoptosis was assessed in HEK293 cells to determine the cytoprotective effects of BBPE and its effects on the generation of cisplatin-induced reactive oxygen species (ROS) and mitochondrial transmembrane potential (MTP) collapse. Although cisplatin induced nephrotoxicity in HEK293 cells, pretreatment with BBPE showed significant protective effects against cisplatin-induced nephrotoxicity by regulating the expression levels of pro- and antiapoptotic proteins. The cytoprotective effects of BBPE were mediated by decreased ROS production and MTP loss in cisplatin-treated HEK293 cells. The in vitro results were confirmed in the cisplatin-treated mouse model. Pretreatment with BBPE protected against cisplatin-induced nephrotoxicity by restoring malondialdehyde, superoxide dismutase, and catalase levels in kidney tissue and blood urea nitrogen and creatinine serum levels. Furthermore, histopathological assessment and terminal dUTP nick end-labeling staining showed that BBPE mitigated cisplatin-induced nephrotoxicity in kidney tissues. Overall, BBPE may act as a potent agent for alleviating cisplatin-induced nephrotoxicity, thereby increasing the safety of cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Jeong Moo Han
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, Korea
| | - Ha-Yeon Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Korea
| | - Kwang-Il Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Korea
| | - Eui-Baek Byun
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Korea
| |
Collapse
|
41
|
Liu X, Gao L, Huang X, Deng R, Wu S, Peng Y, Lu J. Huangqi-Danshen decoction protects against cisplatin-induced acute kidney injury in mice. Front Pharmacol 2023; 14:1236820. [PMID: 38034992 PMCID: PMC10687478 DOI: 10.3389/fphar.2023.1236820] [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: 06/08/2023] [Accepted: 10/24/2023] [Indexed: 12/02/2023] Open
Abstract
Background: Acute kidney injury (AKI) induced by cisplatin remains a major impediment to the clinical application of cisplatin, necessitating urgent exploration for promising solutions. Huangqi-Danshen decoction (HDD), a Chinese herbal preparation, has been shown by our group to have a reno-protective effect in adenine-induced chronic kidney disease mice and diabetic db/db mice. However, the effect of HDD on cisplatin-induced AKI and its underlying mechanisms are unknown. Methods: The AKI model was established by intraperitoneal injection of cisplatin (20 mg/kg) in C57BL/6 mice. The mice in the treatment group were administrated with HDD (6.8 g/kg/d) for 5 consecutive days before cisplatin challenge. After 72 h cisplatin injection, blood and kidney tissue were subsequently collected for biochemical detection, histopathological evaluation, Western blot analysis, immunohistochemical staining, and terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling assay. Ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry was used to detect changes in renal metabolites. Results: The results showed that HDD significantly reduced serum creatinine and blood urea nitrogen levels and alleviated renal histopathological injury in cisplatin-induced AKI mice. And HDD treatment demonstrated a significant inhibition in apoptosis, inflammation, and oxidative stress in AKI mice. Moreover, non-target metabolomics revealed that HDD significantly restored 165 altered metabolites in AKI mice. Subsequent enrichment analysis and pathway analysis of these metabolites indicated that nicotinate and nicotinamide metabolism was the primary pathway affected by HDD intervention. Further investigation showed that HDD could upregulate nicotinamide adenine dinucleotide (NAD+) biosynthesis-related enzymes quinolinate phosphoribosyltransferase, nicotinamide mononucleotide adenylyltransferase 1, and nicotinamide phosphoribosyltransferase to replenish NAD+ content in the kidney of AKI mice. Conclusion: In summary, HDD exerted a protective effect against cisplatin-induced AKI and suppressed apoptosis, inflammation, and oxidative stress in the kidney of AKI mice, which may be attributed to the modulation of NAD+ biosynthesis.
Collapse
Affiliation(s)
- Xinhui Liu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Liwen Gao
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xi Huang
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Ruyu Deng
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Shanshan Wu
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yu Peng
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jiandong Lu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
42
|
Kedeshian K, Hong M, Hoffman L, Kita A. N-acetylcysteine Microparticles Reduce Cisplatin-induced RSC96 Schwann Cell Toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.31.564430. [PMID: 37961184 PMCID: PMC10635004 DOI: 10.1101/2023.10.31.564430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Objectives Cisplatin is known to cause inner ear dysfunction. There is growing evidence that cisplatin-induced demyelination of spiral or Scarpa's ganglion neurons may play an additional role in drug-induced ototoxicity alongside afferent neuron injury. As Schwann cells produce myelin, there may be an opportunity to reduce ototoxic inner ear damage by promoting Schwann cell viability. This work describes a cellular model of cisplatin-induced Schwann cell injury and investigates the ability of the antioxidant N-acetylcysteine to promote Schwann cell viability. A local delivery system of drug-eluting microparticles was then fabricated, characterized, and investigated for bioactivity. Methods RSC96 rat Schwann cells were dosed with varying concentrations of cisplatin to obtain a dose curve and identify the lethal concentration of 50% of the cells (LC 50 ). In subsequent experiments, RSC96 cells were co-treated with cisplatin and both resuspended or eluted N-acetylcysteine. Cell viability was assessed with the CCK8 assay. Results The LC 50 dose of cisplatin was determined to be 3.76 μM (p=2.2 × 10 -16 ). When co-dosed with cisplatin and therapeutic concentration of resuspended or eluted N-acetylcysteine, Schwann cells had an increased viability compared to cells dosed with cisplatin alone. Conclusion RSC96 Schwann cell injury following cisplatin insult is characterized in this in vitro model. Cisplatin caused injury at physiologic concentrations and N-acetylcysteine improved cell viability and mitigated this injury. N-acetylcysteine was packaged into microparticles and eluted N-acetylcysteine retained its ability to increase cell viability, thus demonstrating promise as a therapeutic to offset cisplatin-induced ototoxicity. Lay Summary Cisplatin is a chemotherapeutic agent known to cause balance and hearing problems through damage to the inner ear. This project explored cisplatin injury in a Schwann cell culture model and packaged an antioxidant into microparticles suitable for future drug delivery applications.
Collapse
|
43
|
Janky K, Steyger PS. Mechanisms and Impact of Aminoglycoside-Induced Vestibular Deficits. Am J Audiol 2023; 32:746-760. [PMID: 37319406 PMCID: PMC10721243 DOI: 10.1044/2023_aja-22-00199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/26/2023] [Accepted: 03/06/2023] [Indexed: 06/17/2023] Open
Abstract
PURPOSE Acquired vestibulotoxicity from hospital-prescribed medications such as aminoglycoside antibiotics affects as many as 40,000 people each year in North America. However, there are no current federally approved drugs to prevent or treat the debilitating and permanent loss of vestibular function caused by bactericidal aminoglycoside antibiotics. This review will cover our current understanding of the impact of, and mechanisms underlying, aminoglycoside-induced vestibulotoxicity and highlight the gaps in our knowledge that remain. CONCLUSIONS Aminoglycoside-induced vestibular deficits have long-term impacts on patients across the lifespan. Additionally, the prevalence of aminoglycoside-induced vestibulotoxicity appears to be greater than cochleotoxicity. Thus, monitoring for vestibulotoxicity should be independent of auditory monitoring and encompass patients of all ages from young children to older adults before, during, and after aminoglycoside therapy.
Collapse
Affiliation(s)
- Kristen Janky
- Department of Audiology, Boys Town National Research Hospital, Omaha, NE
| | - Peter S. Steyger
- Bellucci Translational Hearing Center, Creighton University, Omaha, NE
| |
Collapse
|
44
|
Fan YW, Lu IC, Hsu MY, Kuo WT, Wu SY, Lan SH, Wang PY, Chen CY, Liu HS, Su CL. Synthetic lethality in human bladder cancer cells by curcumin via concurrent Aurora A inhibition and autophagy induction. J Nutr Biochem 2023; 121:109438. [PMID: 37666476 DOI: 10.1016/j.jnutbio.2023.109438] [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: 06/20/2022] [Revised: 01/24/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
Combination therapies to induce mixed-type cell death and synthetic lethality have the potential to overcome drug resistance in cancer. In this study, we demonstrated that the curcumin-enhanced cytotoxicity of cisplatin/carboplatin in combination with gemcitabine was associated with Aurora A suppression-mediated G2/M arrest, and thus apoptosis, as well as MEK/ERK-mediated autophagy in human bladder cancer cells. Animal study data confirmed that curcumin combined with cisplatin/gemcitabine reduced tumorigenesis of xenograft in mice and this phenomenon was associated with elevated expressions of p-ERK and reduced p-Aurora A in tumors. Gene analyses using data repositories further revealed that reduced Aurora A expression alone did not significantly elevate the sensitivity of human bladder carcinoma cells to these anticancer drugs. Unlike other major cancer types, human bladder urothelial carcinoma tissue coexpressed higher AURKA and lower MAP1LC3B than normal tissue, and reduced Aurora A and induction of autophagy have been clinically associated with a better prognosis in patients with early but not advanced stage bladder cancer. Therefore, our results suggest that treatment strategies can utilize the synthetic lethal pair to concurrently suppress oncogenic Aurora A and induce autophagy by coadministrating curcumin with anticancer drugs for early-stage bladder cancer with high expression of Aurora A.
Collapse
Affiliation(s)
- Ya-Wen Fan
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei, Taiwan
| | - I-Ching Lu
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Man-Yuan Hsu
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Wan-Ting Kuo
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shan-Ying Wu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Hui Lan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pao-Yuan Wang
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ching-Ying Chen
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hsiao-Sheng Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center of Infectious Disease and Signaling Research Center, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Master of Science Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Li Su
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei, Taiwan; Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei, Taiwan.
| |
Collapse
|
45
|
Yoshioka T, Goda M, Kanda M, Itobayashi S, Sugimoto Y, Izawa‐Ishizawa Y, Yagi K, Aizawa F, Miyata K, Niimura T, Hamano H, Sakurada T, Zamami Y, Ishizawa K. Valproic acid treatment attenuates cisplatin-induced kidney injury by suppressing proximal tubular cell damage. Clin Transl Sci 2023; 16:2369-2381. [PMID: 37700528 PMCID: PMC10651653 DOI: 10.1111/cts.13638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023] Open
Abstract
Cisplatin treatment is effective against several types of carcinomas. However, it frequently leads to kidney injury, which warrants effective prevention methods. Sodium valproic acid is a prophylactic drug candidate with a high potential for clinical application against cisplatin-induced kidney injury. Therefore, in this study, we aimed to elucidate the mechanism underlying the prophylactic effect of valproic acid on cisplatin-induced kidney injury in a mouse model and HK2 and PODO cells with cisplatin-induced toxicity. In the mouse model of cisplatin-induced kidney injury, various renal function parameters and tubular damage scores were worsened by cisplatin, but they were significantly improved upon combination with valproic acid. No difference was observed in cisplatin accumulation between the cisplatin-treated and valproic acid-treated groups in whole blood and the kidneys. The mRNA expression levels of proximal tubular damage markers, apoptosis markers, and inflammatory cytokines significantly increased in the cisplatin group 72 h after cisplatin administration but significantly decreased upon combination with valproic acid. In HK2 cells, a human proximal tubular cell line, the cisplatin-induced decrease in cell viability was significantly suppressed by co-treatment with valproic acid. Valproic acid may inhibit cisplatin-induced kidney injury by suppressing apoptosis, inflammatory responses, and glomerular damage throughout the kidneys by suppressing proximal tubular cell damage. However, prospective controlled trials need to evaluate these findings before their practical application.
Collapse
Affiliation(s)
- Toshihiko Yoshioka
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Mitsuhiro Goda
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Masaya Kanda
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Sayuri Itobayashi
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
| | - Yugo Sugimoto
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
| | - Yuki Izawa‐Ishizawa
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of General MedicineTaoka HospitalTokushimaJapan
| | - Kenta Yagi
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Clinical Research Center for Developmental TherapeuticsTokushima University HospitalTokushimaJapan
| | - Fuka Aizawa
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Koji Miyata
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
| | - Takahiro Niimura
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Clinical Research Center for Developmental TherapeuticsTokushima University HospitalTokushimaJapan
| | - Hirofumi Hamano
- Department of PharmacyOkayama University HospitalOkayamaJapan
| | - Takumi Sakurada
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Yoshito Zamami
- Department of PharmacyOkayama University HospitalOkayamaJapan
| | - Keisuke Ishizawa
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
- Clinical Research Center for Developmental TherapeuticsTokushima University HospitalTokushimaJapan
| |
Collapse
|
46
|
Golmohammadi M, Elmaghraby DA, Ramírez-Coronel AA, Rakhimov N, Mohammed SS, Romero-Parra RM, Jawad MA, Zamanian MY, Soltani A, Taheri N, Kianifar F, Vousooghi N. A comprehensive view on the quercetin impact on bladder cancer: Focusing on oxidative stress, cellular, and molecular mechanisms. Fundam Clin Pharmacol 2023; 37:900-909. [PMID: 36960597 DOI: 10.1111/fcp.12896] [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: 12/12/2022] [Revised: 02/27/2023] [Accepted: 03/22/2023] [Indexed: 03/25/2023]
Abstract
Bladder cancer (BC) is known as a prevalent genitourinary malignancy and has a significant mortality rate worldwide. Despite recent therapeutic approaches, the recurrence rate is high, highlighting the need for a new strategy to reduce the BC cell progression. Quercetin, a flavonoid compound, demonstrated promising anticancer properties and could be used in the management of various malignancies such as BC. This comprehensive review summarized quercetin's cellular and molecular mechanisms underlying anticancer activities. The study's findings indicated that quercetin prevents the proliferation of the human BC cell line, promotes apoptosis of BIU-87 cells, reduces the expression of p-P70S6K, and induces apoptosis by p-AMPK. Moreover, quercetin restricts tumor growth through the AMPK/mTOR cascade and prevents colony formation of human BC cells by triggering DNA damage. Studying this review article will help researchers better understand quercetin's functional role in the prevention and treatment of BC.
Collapse
Affiliation(s)
- Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1988873554, Iran
| | - Dalia Ahmed Elmaghraby
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, 31982, Saudi Arabia
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Cuenca, Ecuador
- University of Palermo, Buenos Aires, Argentina
- Research group in educational statistics, National University of Education, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, CES University, Medellín, Colombia
| | - Nodir Rakhimov
- Department of Oncology, Samarkand State Medical University, Amir Temur Street 18, Samarkand, Uzbekistan
| | | | | | | | - Mohammad Yasin Zamanian
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran
| | - Afsaneh Soltani
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Taheri
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Farzaneh Kianifar
- School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Nasim Vousooghi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| |
Collapse
|
47
|
Katanić Stanković JS, Selaković D, Rosić G. Oxidative Damage as a Fundament of Systemic Toxicities Induced by Cisplatin-The Crucial Limitation or Potential Therapeutic Target? Int J Mol Sci 2023; 24:14574. [PMID: 37834021 PMCID: PMC10572959 DOI: 10.3390/ijms241914574] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/11/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023] Open
Abstract
Cisplatin, an inorganic complex of platinum, is a chemotherapeutic drug that has been used for 45 years. Despite the progress of pharmaceutical sciences and medicine and the successful application of other platinum complexes for the same purpose, cisplatin is still the therapy of choice in many cancers. Treatment for testicular, ovarian, head and neck, urothelial, cervical, esophageal, breast, and pulmonary malignancies is still unthinkable without the use of this drug. However, cisplatin is also known for many side effects, of which the most pronounced are nephrotoxicity leading to acute renal failure, neurotoxicity, and ototoxicity. Mechanistic studies have proven that one of the conditions that plays a major role in the development of cisplatin-induced toxicities is oxidative stress. Knowing the fact that numerous antioxidants can be used to reduce oxidative stress, thereby reducing tissue lesions, organ failure, and apoptosis at the cellular level, many studies have defined antioxidants as a priority for investigation as a cotreatment. To investigate the mechanism of antioxidant action in vivo, many animal models have been employed. In the last few years, studies have mostly used rodents and zebrafish models. In this article, some of the most recent investigations that used animal models are listed, and the advantages and disadvantages of such experimental studies are pointed out.
Collapse
Affiliation(s)
- Jelena S. Katanić Stanković
- Department of Science, Institute for Information Technologies Kragujevac, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | - Dragica Selaković
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia;
| | - Gvozden Rosić
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia;
| |
Collapse
|
48
|
Morelli C, Formica V, Bossi P, Rofei M, Guerriero S, Riondino S, Argirò R, Pucci N, Cenci T, Savino L, Rinaldi CG, Garaci F, Orlandi A, D’Angelillo RM, Arkenau HT, Roselli M. Untailored vs. Gender- and Body-Mass-Index-Tailored Skeletal Muscle Mass Index (SMI) to Assess Sarcopenia in Advanced Head and Neck Squamous Cell Carcinoma (HNSCC). Cancers (Basel) 2023; 15:4716. [PMID: 37835410 PMCID: PMC10571960 DOI: 10.3390/cancers15194716] [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: 08/01/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
(1) Background: Sarcopenia lasting >1 year might be considered a chronic condition in many HNSCC patients. CT-scan-derived Skeletal Muscle Mass Index (SMI) is an established surrogate of sarcopenia; yet, the cut-off reported in the literature (literature-based, lb-SMI < 43.2) is mainly based on the risk of chemoradiotherapy-induced toxicity, and the optimal value to discriminate OS is under-investigated. (2) Methods: The effect on OS of the lb-SMI cutoff was compared with an untailored OS-oriented SMI cutoff obtained in a cohort of consecutive advanced HNSCC patients treated with primary chemoradiotherapy, bio-chemotherapy or chemo-immunotherapy (cohort-specific, cs-SMI cutoff). Gender- and BMI-tailored (gt-SMI and bt-SMI) cut-offs were also evaluated. Cutoff values were identified by using the maximally selected rank statistics for OS. (3) Results: In 115 HNSCC patients, the cs-SMI cutoff was 31.50, which was lower compared to the lb-SMI reported cut-off. The optimal cut-off separately determined in females, males, overweight and non-overweight patients were 46.02, 34.37, 27.32 and 34.73, respectively. gt-SMI categorization had the highest effect on survival (p < 0.0001); its prognostic value was independent of the treatment setting or the primary location and was retained in a multivariate cox-regression analysis for OS including other HNSCC-specific prognostic factors (p = 0.0004). (4) Conclusions: A tailored SMI assessment would improve clinical management of sarcopenia in chemoradiotherapy-, bio-chemotherapy- or chemo-immunotherapy-treated HNSCC patients. Gender-based SMI could be used for prognostication in HNSCC patients.
Collapse
Affiliation(s)
- Cristina Morelli
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.M.); (M.R.); (S.G.); (S.R.); (M.R.)
| | - Vincenzo Formica
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.M.); (M.R.); (S.G.); (S.R.); (M.R.)
| | - Paolo Bossi
- Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, “ASST Spedali Civili di Brescia”, University of Brescia, 25123 Brescia, Italy;
| | - Michela Rofei
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.M.); (M.R.); (S.G.); (S.R.); (M.R.)
| | - Simona Guerriero
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.M.); (M.R.); (S.G.); (S.R.); (M.R.)
| | - Silvia Riondino
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.M.); (M.R.); (S.G.); (S.R.); (M.R.)
| | - Renato Argirò
- Interventional Radiology Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Noemi Pucci
- Neuroradiology Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (N.P.); (F.G.)
| | - Tonia Cenci
- Anatomic Pathology, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.C.); (L.S.); (A.O.)
| | - Luca Savino
- Anatomic Pathology, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.C.); (L.S.); (A.O.)
| | - Carla G. Rinaldi
- Radiation Oncology, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00152 Rome, Italy; (C.G.R.); (R.M.D.)
| | - Francesco Garaci
- Neuroradiology Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (N.P.); (F.G.)
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.C.); (L.S.); (A.O.)
| | - Rolando M. D’Angelillo
- Radiation Oncology, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00152 Rome, Italy; (C.G.R.); (R.M.D.)
| | | | - Mario Roselli
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.M.); (M.R.); (S.G.); (S.R.); (M.R.)
| |
Collapse
|
49
|
Gao X, Yin Y, Liu S, Dong K, Wang J, Guo C. Fucoidan-proanthocyanidins nanoparticles protect against cisplatin-induced acute kidney injury by activating mitophagy and inhibiting mtDNA-cGAS/STING signaling pathway. Int J Biol Macromol 2023:125541. [PMID: 37355076 DOI: 10.1016/j.ijbiomac.2023.125541] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023]
Abstract
Fucoidan (FU) is a natural polymer from marine organisms, which has been widely studied and applied in drug delivery. In this study, FU nanoparticles loaded with proanthocyanidins (PCs) (FU/PCs NPs) were prepared and their effect and mechanism in protecting cisplatin-induced acute kidney injury (AKI) were studied. The in vitro studies confirmed that FU/PCs NPs increased the antioxidant activity of free PCs and protected the death of human kidney proximal tubule (HK-2) cells induced by cisplatin. Further mechanism studies showed that FU/PCs NPs protected the mitochondrial damage induced by cisplatin, activated mitophagy, inhibited the release of mitochondrial DNA (mtDNA), and inhibited the cGAS/STING signal pathway. The in vivo results also indicated that FU/PCs NPs protected cisplatin-induced AKI, including inhibiting the increase of blood urea nitrogen (BUN) and serum creatinine (SCr) levels induced by cisplatin. The mechanism studies confirmed that cisplatin induced an increase in the expression of mitophagy-related protein Pink/Pakrin, mitochondrial mtDNA release and cGAS/STING expression in mice kidney tissues. Pre-administration of FU/PCs NPs further activated mitophagy, as well as inhibiting mtDNA release and cGAS/STING expression. In conclusion, our research proved the role of mitophagy-mtDNA-cGAS/STING signal was involved in cisplatin-induced AKI.
Collapse
Affiliation(s)
- Xintao Gao
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yulan Yin
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Shuai Liu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Kehong Dong
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jing Wang
- Department of Biology Science and Technology, Baotou Teacher's College, Baotou 014030, China
| | - Chuanlong Guo
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266273, Shandong, China.
| |
Collapse
|
50
|
Li Y, Zhang T, Song Q, Gao D, Li Y, Jie H, Huang P, Zheng G, Yang J, He J. Cisplatin ototoxicity mechanism and antagonistic intervention strategy: a scope review. Front Cell Neurosci 2023; 17:1197051. [PMID: 37323582 PMCID: PMC10267334 DOI: 10.3389/fncel.2023.1197051] [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: 03/30/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Cisplatin is a first-line chemotherapeutic agent in the treatment of malignant tumors with remarkable clinical effects and low cost. However, the ototoxicity and neurotoxicity of cisplatin greatly limit its clinical application. This article reviews the possible pathways and molecular mechanisms of cisplatin trafficking from peripheral blood into the inner ear, the toxic response of cisplatin to inner ear cells, as well as the cascade reactions leading to cell death. Moreover, this article highlights the latest research progress in cisplatin resistance mechanism and cisplatin ototoxicity. Two effective protective mechanisms, anti-apoptosis and mitophagy activation, and their interaction in the inner ear are discussed. Additionally, the current clinical preventive measures and novel therapeutic agents for cisplatin ototoxicity are described. Finally, this article also forecasts the prospect of possible drug targets for mitigating cisplatin-induced ototoxicity. These include the use of antioxidants, inhibitors of transporter proteins, inhibitors of cellular pathways, combination drug delivery methods, and other mechanisms that have shown promise in preclinical studies. Further research is needed to evaluate the efficacy and safety of these approaches.
Collapse
Affiliation(s)
- Yingru Li
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Tianyang Zhang
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Qiang Song
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Dekun Gao
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yue Li
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Huiqun Jie
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Ping Huang
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Guiliang Zheng
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jun Yang
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jingchun He
- Department of Otorhinolaryngology–Head and Neck Surgery, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Ear Institute, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| |
Collapse
|