1
|
Xu B, Yang L, Yang L, Al-Maamari A, Zhang J, Song H, Wang M, Su S, Song Z. Role of glutaminyl-peptide cyclotransferase in breast cancer doxorubicin sensitivity. Cancer Biol Ther 2024; 25:2321767. [PMID: 38417050 PMCID: PMC10903679 DOI: 10.1080/15384047.2024.2321767] [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: 04/02/2023] [Accepted: 02/18/2024] [Indexed: 03/01/2024] Open
Abstract
Doxorubicin (DOX) is one of the most effective and widely used chemotherapeutic drugs. However, DOX resistance is a critical risk problem for breast cancer treatment. Previous studies have demonstrated that metadherin (MTDH) involves in DOX resistance in breast cancer, but the exact mechanism remains unclear. In this study, we found that glutaminyl-peptide cyclotransferase (QPCT) was a MTDH DOX resistance-related downstream gene in breast cancer. Elevated expression of QPCT was found in the GEPIA database, breast cancer tissue, and breast cancer cells. Clinical data showed that QPCT expression was positively associated with poor prognosis in DOX-treated patients. Overexpression of QPCT could promote the proliferation, invasion and migration, and reduce DOX sensitivity in MCF-7 and MDA-MB-231 cells. Mechanistically, MTDH positively regulates the expressions of NF-κB (p65) and QPCT, and NF-κB (p65) directly regulates the expression of QPCT. Therefore, MTDH/NF-κB (p65)/QPCT signal axis was proposed. Collectively, our findings delineate the mechanism by which the MTDH/NF-κB (p65) axis regulate QPCT signaling and suggest that this complex may play an essential role in breast cancer progression and affect DOX sensitivity.
Collapse
Affiliation(s)
- Bin Xu
- Department of Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Liu Yang
- Department of Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lixian Yang
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, Hebei, China
| | - Ahmed Al-Maamari
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jingyu Zhang
- Department of Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Heng Song
- Department of Radiotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Meiqi Wang
- Department of Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Suwen Su
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhenchuan Song
- Department of Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
2
|
Tapaswi A, Cemalovic N, Polemi KM, Sexton JZ, Colacino JA. Applying cell painting in non-tumorigenic breast cells to understand impacts of common chemical exposures. Toxicol In Vitro 2024; 101:105935. [PMID: 39243829 DOI: 10.1016/j.tiv.2024.105935] [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: 05/17/2024] [Revised: 08/02/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
The general population is exposed to many chemicals which have putative, but incompletely understood, links to breast cancer. Cell Painting is a high-content imaging-based in vitro assay that allows for unbiased measurements of concentration-dependent effects of chemical exposures on cellular morphology. We used Cell Painting to measure effects of 16 human exposure relevant chemicals, along with 21 small molecules with known mechanisms of action, in non-tumorigenic mammary epithelial cells, the MCF10A cell line. Using CellProfiler image analysis software, we quantified 3042 morphological features across approximately 1.2 million cells. We used benchmark concentration modeling to identify features both conserved and different across chemicals. Benchmark concentrations were compared to exposure biomarker concentration measurements from the National Health and Nutrition Examination Survey to assess which chemicals induce morphological alterations at human-relevant concentrations. We found significant feature overlaps between chemicals, including similarities between the organochlorine pesticide DDT metabolite p,p'-DDE and an activator of Wnt signaling CHIR99201. We validated these findings by assaying the activation of Wnt, as reflected by translocation of ꞵ-catenin, following p'-p' DDE exposure. Consistent with Wnt signaling activation, low concentration p',p'-DDE (25 nM) significantly enhanced the nuclear translocation of ꞵ-catenin. Overall, these findings highlight the ability of Cell Painting to enhance mode-of-action studies for toxicants which are common in our environment but incompletely characterized with respect to breast cancer risk.
Collapse
Affiliation(s)
- Anagha Tapaswi
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas Cemalovic
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Katelyn M Polemi
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Z Sexton
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Medicinal Chemistry, University of Michigan School of Pharmacy, Ann Arbor, MI, USA
| | - Justin A Colacino
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA; Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA; Program in the Environment, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
3
|
Abbas Z, Afzal S, Fujimura NA, Akram M, Tahir S, Malik K, Ahmed N. Recombinant expression, downstream optimization, and therapeutic evaluation of recombinant human interleukin-37 for cancer therapy. Biotechnol Lett 2024; 46:1269-1291. [PMID: 39424749 DOI: 10.1007/s10529-024-03539-3] [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: 06/09/2024] [Revised: 09/23/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
Interleukin-37 is a cytokine with potent immunosuppressive properties that has been shown to have potential to treat autoimmune and chronic inflammatory diseases, as well as certain types of cancer. IL-37 is a 19 kDa protein which interacts with proteins in receptor-dependent and receptor-independent pathways. The expression of the IL-37 protein cloned into the pET-28a vector was optimized in Rosetta 2(DE3) after comparing its expression with Rosetta-gami 2(DE3) and Rosetta 2(DE3) pLysS, which was then used for the large-scale production of IL-37. IMAC purification of IL-37 yielded > 97% pure 0.9 mg/mL protein from auto-induced fermentation. The IC50 value of IL-37 was < 1 µM, which was similar to that of doxorubicin, and proliferation of > 80% of all cancer cells was inhibited by 100 µg/mL of IL-37 protein. IL-37 may be a promising theragnostic target for cancer due to its comparable IC50 value with that of doxorubicin.
Collapse
Affiliation(s)
- Zaheer Abbas
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Samia Afzal
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Nao Akusa Fujimura
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Akram
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Saad Tahir
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Kausar Malik
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Nadeem Ahmed
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan.
| |
Collapse
|
4
|
Pal C. Small Molecules Targeting Mitochondria: A Mechanistic Approach to Combating Doxorubicin-Induced Cardiotoxicity. Cardiovasc Toxicol 2024:10.1007/s12012-024-09941-7. [PMID: 39495464 DOI: 10.1007/s12012-024-09941-7] [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: 09/19/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Doxorubicin (Dox) is a commonly used chemotherapy drug effective against a range of cancers, but its clinical application is greatly limited by dose-dependent and cumulative cardiotoxicity. Mitochondrial dysfunction is recognized as a key factor in Dox-induced cardiotoxicity, leading to oxidative stress, disrupted calcium balance, and activation of apoptotic pathways. Recent research has emphasized the potential of small molecules that specifically target mitochondria to alleviate these harmful effects. This review provides a comprehensive analysis of small molecules that offer cardioprotection by preserving mitochondrial function in the context of doxorubicin-induced cardiotoxicity (DIC). The mechanisms of action include the reduction of reactive oxygen species (ROS) production, stabilization of mitochondrial membrane potential, enhancement of mitochondrial biogenesis, and modulation of key signaling pathways involved in cell survival and apoptosis. By targeting mitochondria, these small molecules present a promising therapeutic strategy to prevent or reduce the cardiotoxic effects associated with Dox treatment. This review not only discusses the mechanistic actions of these agents but also emphasizes their potential in improving cardiovascular outcomes for cancer patients. Gaining insight into these mechanisms can help in creating more effective strategies to safeguard the heart during chemotherapy, allowing for the ongoing use of Dox with a lower risk to the patient's cardiovascular health. This review highlights the critical role of mitochondria-targeted therapies as a promising approach in addressing DIC.
Collapse
Affiliation(s)
- Chinmay Pal
- Department of Chemistry, Gobardanga Hindu College, North 24 Parganas, West Bengal, 743273, India.
| |
Collapse
|
5
|
Cirillo G, Cappello AR, Curcio M, Fiorillo M, Frattaruolo L, Avena P, Scorzafave L, Dolce V, Nicoletta FP, Iemma F. Novel CD44-Targeted Albumin Nanoparticles: An Innovative Approach to Improve Breast Cancer Treatment. Int J Mol Sci 2024; 25:10560. [PMID: 39408889 PMCID: PMC11477043 DOI: 10.3390/ijms251910560] [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: 09/02/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
This study introduces novel CD44-targeted and redox-responsive nanoparticles (FNPs), proposed as doxorubicin (DOX) delivery devices for breast cancer. A cationized and redox-responsive Human Serum Albumin derivative was synthesized by conjugating Human Serum Albumin with cystamine moieties and then ionically complexing it with HA. The suitability of FNPs for cancer therapy was assessed through physicochemical measurements of size distribution (mean diameter of 240 nm), shape, and zeta potential (15.4 mV). Nanoparticles possessed high DOX loading efficiency (90%) and were able to trigger the drug release under redox conditions of the tumor environment (55% release after 2 h incubation). The use of the carrier increased the cytotoxic effect of DOX by targeting the CD44 protein. It was shown that, upon loading, the cytotoxic effect of DOX was enhanced in relation to CD44 protein expression in both 2D and 3D models. DOX@FNPs significantly decrease cellular metabolism by reducing both oxygen consumption and extracellular acidification rates. Moreover, they decrease the expression of proteins involved in the oxidative phosphorylation pathway, consequently reducing cellular viability and motility, as well as breast cancer stem cells and spheroid formation, compared to free DOX. This new formulation could become pioneering in reducing chemoresistance phenomena and increasing the specificity of DOX in breast cancer patients.
Collapse
Affiliation(s)
| | | | - Manuela Curcio
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende (CS), Italy; (G.C.); (A.R.C.); (L.F.); (P.A.); (L.S.); (F.P.N.); (F.I.)
| | - Marco Fiorillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende (CS), Italy; (G.C.); (A.R.C.); (L.F.); (P.A.); (L.S.); (F.P.N.); (F.I.)
| | | | | | | | | | | | | |
Collapse
|
6
|
Chen J, Xiang Y, Bao R, Zheng Y, Fang Y, Feng J, Wu D, Chen X. Combined Photothermal Chemotherapy for Effective Treatment Against Breast Cancer in Mice Model. Int J Nanomedicine 2024; 19:9973-9987. [PMID: 39360036 PMCID: PMC11446203 DOI: 10.2147/ijn.s473052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Breast cancer ranks among the most prevalent cancers in women, characterized by significant morbidity, disability, and mortality. Presently, chemotherapy is the principal clinical approach for treating breast cancer; however, it is constrained by limited targeting capability and an inadequate therapeutic index. Photothermal therapy, as a non-invasive approach, offers the potential to be combined with chemotherapy to improve tumor cellular uptake and tissue penetration. In this research, a mesoporous polydopamine-coated gold nanorod nanoplatform, encapsulating doxorubicin (Au@mPDA@DOX), was developed. Methods This nanoplatform was constructed by surface coating mesoporous polydopamine (mPDA) onto gold nanorods, and doxorubicin (DOX) was encapsulated in Au@mPDA owing to π-π stacking between mPDA and DOX. The dynamic diameter, zeta potential, absorbance, photothermal conversion ability, and drug release behavior were determined. The cellular uptake, cytotoxicity, deep penetration, and anti-tumor effects were subsequently investigated in 4T1 cells. After that, fluorescence imaging, photothermal imaging and pharmacodynamics studies were utilized to evaluate the anti-tumor effects in tumor-bearing mice model. Results This nanoplatform exhibited high drug loading capacity, excellent photothermal conversion and, importantly, pH/photothermal dual-responsive drug release behavior. The in vitro results revealed enhanced photothermal-facilitated cellular uptake, drug release and tumor penetration of Au@mPDA@DOX under near-infrared irradiation. In vivo studies confirmed that, compared with monotherapy with either chemotherapy or photothermal therapy, the anti-tumor effects of Au@mPDA@DOX are synergistically improved. Conclusion Together with good biosafety and biocompatibility, the Au@mPDA@DOX nanoplatform provides an alternative method for safe and synergistic treatment of breast cancer.
Collapse
Affiliation(s)
- Junzi Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, People's Republic of China
| | - Yumin Xiang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, People's Republic of China
| | - Rong Bao
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, People's Republic of China
| | - Yuyi Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, People's Republic of China
| | - Yingxi Fang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, People's Republic of China
| | - Jiajia Feng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, People's Republic of China
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, People's Republic of China
| | - Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310053, People's Republic of China
| |
Collapse
|
7
|
Hubiernatorova A, Novak J, Vaskovicova M, Sekac D, Kropyvko S, Hodny Z. Tristetraprolin affects invasion-associated genes expression and cell motility in triple-negative breast cancer model. Cytoskeleton (Hoboken) 2024. [PMID: 39319680 DOI: 10.1002/cm.21934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Tristetraprolin (TTP) is an RNA-binding protein that negatively regulates its target mRNAs and has been shown to inhibit tumor progression and invasion. Tumor invasion requires precise regulation of cytoskeletal components, and dysregulation of cytoskeleton-associated genes can significantly alter cell motility and invasive capability. Several genes, including SH3PXD2A, SH3PXD2B, CTTN, WIPF1, and WASL, are crucial components of the cytoskeleton reorganization machinery and are essential for adequate cell motility. These genes are also involved in invasion processes, with SH3PXD2A, SH3PXD2B, WIPF1, and CTTN being key components of invadopodia-specialized structures that facilitate invasion. However, the regulation of these genes is not well understood. This study demonstrates that ectopic expression of TTP in MDA-MB-231 cells leads to decreased mRNA levels of CTTN and SH3PXD2A, as well as defects in cell motility and actin filament organization. Additionally, doxorubicin significantly increases TTP expression and reduces the mRNA levels of cytoskeleton-associated genes, enhancing our understanding of how doxorubicin may affect the transcriptional profile of cells. However, doxorubicin affects target mRNAs differently than TTP ectopic expression, suggesting it may not be the primary mechanism of doxorubicin in breast cancer (BC) treatment. High TTP expression is considered as a positive prognostic marker in multiple cancers, including BC. Given that doxorubicin is a commonly used drug for treating triple-negative BC, using TTP as a prognostic marker in this cohort of patients might be limited since it might be challenging to understand if high TTP expression occurred due to the favorable physiological state of the patient or as a consequence of treatment.
Collapse
Affiliation(s)
- Anastasiia Hubiernatorova
- Department of Functional Genomics, Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
- Laboratory of Cell Regeneration and Plasticity, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - Josef Novak
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michaela Vaskovicova
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
- Laboratory of DNA Integrity, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - David Sekac
- Laboratory of Cell Regeneration and Plasticity, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Serhii Kropyvko
- Department of Functional Genomics, Institute of Molecular Biology and Genetics NAS of Ukraine, Kyiv, Ukraine
| | - Zdenek Hodny
- Laboratory of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
8
|
Bourang S, Noruzpour M, Jahanbakhsh Godekahriz S, Ebrahimi HAC, Amani A, Asghari Zakaria R, Yaghoubi H. Application of nanoparticles in breast cancer treatment: a systematic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6459-6505. [PMID: 38700795 DOI: 10.1007/s00210-024-03082-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/02/2024] [Indexed: 09/25/2024]
Abstract
It is estimated that cancer is the second leading cause of death worldwide. The primary or secondary cause of cancer-related mortality for women is breast cancer. The main treatment method for different types of cancer is chemotherapy with drugs. Because of less water solubility of chemotherapy drugs or their inability to pass through membranes, their body absorbs them inadequately, which lowers the treatment's effectiveness. Drug specificity and pharmacokinetics can be changed by nanotechnology using nanoparticles. Instead, targeted drug delivery allows medications to be delivered to the targeted sites. In this review, we focused on nanoparticles as carriers in targeted drug delivery, their characteristics, structure, and the previous studies related to breast cancer. It was shown that nanoparticles could reduce the negative effects of chemotherapy drugs while increasing their effectiveness. Lipid-based nanocarriers demonstrated notable results in this instance, and some products that are undergoing various stages of clinical trials are among the examples. Nanoparticles based on metal or polymers demonstrated a comparable level of efficacy. With the number of cancer cases rising globally, many researchers are now looking into novel treatment approaches, particularly the use of nanotechnology and nanoparticles in the treatment of cancer. In order to help clinicians, this article aimed to gather more information about various areas of nanoparticle application in breast cancer therapy, such as modifying their synthesis and physicochemical characterization. It also sought to gain a deeper understanding of the mechanisms underlying the interactions between nanoparticles and biologically normal or infected tissues.
Collapse
Affiliation(s)
- Shima Bourang
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mehran Noruzpour
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Sodabeh Jahanbakhsh Godekahriz
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hossein Ali Ca Ebrahimi
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Amin Amani
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Rasool Asghari Zakaria
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hashem Yaghoubi
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| |
Collapse
|
9
|
Prasad A, Bakr MM, ElMeshad AN. Surface-functionalised polymeric nanoparticles for breast cancer treatment: processes and advances. J Drug Target 2024; 32:770-784. [PMID: 38717907 DOI: 10.1080/1061186x.2024.2353359] [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: 02/20/2024] [Revised: 04/09/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024]
Abstract
The World Health Organization (WHO) reported that of all the non-communicable diseases, cancer is considered the second cause of death worldwide. This has driven the big pharma companies to prioritise anticancer products in their pipeline. In addition, research has focused on exploration of new anticancer molecules and design of suitable dosage forms to achieve effective drug delivery to the tumour site. Nanotechnology is a valuable tool to build nano delivery systems with controlled and targeted drug release properties. Nanoparticles can be fabricated by robust, scalable and economic techniques using various polymers. Moreover, specific functional groups can be introduced to the surface of nanoparticles enabling targeting to a specific tissue; besides, they exhibit versatile drug release patterns according to the rate of polymer degradation. This review outlines the processes and advances in surface functionalisation of nanoparticles employed for treatment of breast cancer. The therapeutic molecules, the polymers used to fabricate nanoparticles, the techniques used to prepare the nanoparticles have been reviewed with a focus on the processes employed to functionalise these nanoparticles with suitable ligands to target different types of breast cancer.
Collapse
Affiliation(s)
- Aprameya Prasad
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Mohamed Mofreh Bakr
- Department of Pharmaceutics, Egyptian Drug Authority, Formerly Known as National Organization for Drug Control and Research, Giza, Egypt
| | - Aliaa N ElMeshad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy and Drug Technology, The Egyptian Chinese University, Cairo, Egypt
| |
Collapse
|
10
|
Alioglu MA, Yilmaz YO, Singh YP, Nagamine M, Celik N, Kim MH, Pal V, Gupta D, Ozbolat IT. Nested Biofabrication: Matryoshka-Inspired Intra-Embedded Bioprinting. SMALL METHODS 2024; 8:e2301325. [PMID: 38111377 PMCID: PMC11187694 DOI: 10.1002/smtd.202301325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/04/2023] [Indexed: 12/20/2023]
Abstract
Engineering functional tissues and organs remains a fundamental pursuit in bio-fabrication. However, the accurate constitution of complex shapes and internal anatomical features of specific organs, including their intricate blood vessels and nerves, remains a significant challenge. Inspired by the Matryoshka doll, here a new method called "Intra-Embedded Bioprinting (IEB)" is introduced building upon existing embedded bioprinting methods. a xanthan gum-based material is used which served a dual role as both a bioprintable ink and a support bath, due to its unique shear-thinning and self-healing properties. IEB's capabilities in organ modeling, creating a miniaturized replica of a pancreas using a photocrosslinkable silicone composite is demonstrated. Further, a head phantom and a Matryoshka doll are 3D printed, exemplifying IEB's capability to manufacture intricate, nested structures. Toward the use case of IEB and employing an innovative coupling strategy between extrusion-based and aspiration-assisted bioprinting, a breast tumor model that included a central channel mimicking a blood vessel, with tumor spheroids bioprinted in proximity is developed. Validation using a clinically-available chemotherapeutic drug illustrated its efficacy in reducing the tumor volume via perfusion over time. This method opens a new way of bioprinting enabling the creation of complex-shaped organs with internal anatomical features.
Collapse
Affiliation(s)
- Mecit Altan Alioglu
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
| | - Yasar Ozer Yilmaz
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- Department of Nanoscience and Nanoengineering, Istanbul Technical University, Istanbul, 34469, Turkey
| | - Yogendra Pratap Singh
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
| | - Momoka Nagamine
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Department of Chemistry, Penn State University, University Park, PA, 16802, USA
| | - Nazmiye Celik
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
| | - Myoung Hwan Kim
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, Penn State University, University Park, PA, 16802, USA
| | - Vaibhav Pal
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Department of Chemistry, Penn State University, University Park, PA, 16802, USA
| | - Deepak Gupta
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
| | - Ibrahim T Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- Materials Research Institute, Penn State University, University Park, PA, 16802, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, 17033, USA
- Penn State Cancer Institute, Penn State University, Hershey, PA, 17033, USA
- Department of Medical Oncology, Cukurova University, Adana, 01130, Turkey
- Biotechnology Research and Application Center, Cukurova University, Adana, 01130, Turkey
| |
Collapse
|
11
|
Teng Y, Li Z, Liu J, Teng L, Li H. Synergistic Effect of Doxorubicin and Blue Light Irradiation on the Antitumor Treatment of HepG2 Cells in Liver Cancer. Molecules 2024; 29:3360. [PMID: 39064938 PMCID: PMC11279636 DOI: 10.3390/molecules29143360] [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: 05/23/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Doxorubicin (DOX) has been an effective antitumor agent for human liver cancer cells; however, an overdose might lead to major side effects appearing in clinical applications. In this work, we present a strategy of combining DOX and blue light (BL) irradiation for the antitumor treatment of HepG2 cells (one typical human liver cancer cell line). It is demonstrated that synergetic DOX and BL can significantly reduce cell proliferation and increase the apoptotic rate of HepG2 cells in comparison to individual DOX treatment. The additional BL irradiation is further helpful for enhancing the inhibition of cell migration and invasion. Analyses of reactive oxygen species (ROS) level and Western blotting reveal that the strategy results in more ROS accumulation, mitochondrial damage, and the upregulation of proapoptotic protein (Bcl-2) and downregulation of antiapoptotic protein (Bax). In addition to the improved therapeutic effect, the non-contact BL irradiation is greatly helpful for reducing the dosage of DOX, and subsequently reduces the side effects caused by the DOX drug. These findings offer a novel perspective for the therapeutic approach toward liver cancer with high efficiency and reduced side effects.
Collapse
Affiliation(s)
- Yun Teng
- State Key Laboratory of Superhard Materials, College of Physics, Jilin University, Changchun 130012, China;
| | - Zhige Li
- School of Life Sciences, Jilin University, Changchun 130012, China;
| | - Junsong Liu
- State Key Laboratory of Superhard Materials, College of Physics, Jilin University, Changchun 130012, China;
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun 130012, China;
| | - Hongdong Li
- State Key Laboratory of Superhard Materials, College of Physics, Jilin University, Changchun 130012, China;
| |
Collapse
|
12
|
Alshehade SA, Almoustafa HA, Alshawsh MA, Chik Z. Flow cytometry-based quantitative analysis of cellular protein expression in apoptosis subpopulations: A protocol. Heliyon 2024; 10:e33665. [PMID: 39040270 PMCID: PMC11260931 DOI: 10.1016/j.heliyon.2024.e33665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Flow cytometry techniques utilizing dual staining with annexin V and propidium iodide (PI) provide a robust method for quantitatively analyzing apoptosis induction. Annexin V binds phosphatidylserine exposed on the outer leaflet of the plasma membrane during early apoptosis, while PI permeates late apoptotic/necrotic cells. Simultaneous staining allows differentiation of viable, early apoptotic, and late apoptotic/necrotic populations. This approach can be enhanced by using fluorochrome-conjugated antibodies to stain specific proteins, enabling the simultaneous tracking of protein expression changes in defined cell subpopulations during apoptosis. This multiparametric approach provides key insights into signaling regulation and the mechanisms underlying the apoptotic response to cytotoxic treatments. Here we present a protocol that combines annexin V-FITC/PI staining with APC-conjugated antibody labeling in MDA-MB-231 breast cancer cells treated with doxorubicin. This protocol enables both the quantitative assessment of apoptosis induction and the tracking of decreased CD44 expression from viable to apoptotic cells. This protocol also provides guidelines for appropriate filter selection, compensation controls, gating strategies, and troubleshooting. This robust protocol holds significant potential for elucidating signaling networks involved in apoptosis and therapeutic resistance across various cellular models.
Collapse
Affiliation(s)
- Salah Abdalrazak Alshehade
- Department of Pharmacology, Faculty of Pharmacy & Bio Medical Sciences, MAHSA University, 42610, Selangor, Malaysia
| | - Hassan A. Almoustafa
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
- Universiti Malaya Bioequivalence and Testing Centre (UBAT), Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Mohammed Abdullah Alshawsh
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
- School of Clinical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
| | - Zamri Chik
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
- Universiti Malaya Bioequivalence and Testing Centre (UBAT), Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Martins SA, Costa RR, Brito A, Reis RL, Alves NM, Pashkuleva I, Soares da Costa D. Multifunctional calcium-based nanocarriers for synergistic treatment of triple-negative breast cancer. J Colloid Interface Sci 2024; 674:500-512. [PMID: 38943911 DOI: 10.1016/j.jcis.2024.06.159] [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: 04/08/2024] [Revised: 06/14/2024] [Accepted: 06/22/2024] [Indexed: 07/01/2024]
Abstract
Targeted breast cancer therapies hold the potential to improve the efficiency of drug delivery to the pathology site without impacting the viability and function of healthy cells. Herein, we developed multifunctional nanocarriers that target simultaneously several downstream signaling processes in triple negative breast cancer cells. The system comprises pH sensitive CaCO3 nanoparticles (NPs) as carriers of the anticancer drug doxorubicin (DOX). The NPs were coated in a layer-by-layer (LbL) fashion using poly-l-lysine and hyaluronic acid to target receptors overexpressed in breast cancer (e.g. CD44, RHAMM). Spheroids of the triple-negative Hs578T cell line were used as a 3D model to assess the therapeutic potential of this system. Our results showed that the NPs act via a synergistic mechanism that combines Ca2+ overload causing cell calcification and DNA damage by DOX. The LbL coating was crucial for the protection of the healthy cells, i.e. it provides NPs with targeting capacity. The overall data suggests that the LbL-coated NPs loaded with DOX hold great potential for the treatment of breast cancer.
Collapse
Affiliation(s)
- Sara A Martins
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Rui R Costa
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Alexandra Brito
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Natália M Alves
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Iva Pashkuleva
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Diana Soares da Costa
- 3B's Research Group, I3Bs - Research Institute On Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, 4805-017, Barco Guimarães, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
14
|
Bisht A, Avinash D, Sahu KK, Patel P, Das Gupta G, Kurmi BD. A comprehensive review on doxorubicin: mechanisms, toxicity, clinical trials, combination therapies and nanoformulations in breast cancer. Drug Deliv Transl Res 2024:10.1007/s13346-024-01648-0. [PMID: 38884850 DOI: 10.1007/s13346-024-01648-0] [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] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
Doxorubicin is a key treatment for breast cancer, but its effectiveness often comes with significant side effects. Its actions include DNA intercalation, topoisomerase II inhibition, and reactive oxygen species generation, leading to DNA damage and cell death. However, it can also cause heart problems and low blood cell counts. Current trials aim to improve doxorubicin therapy by adjusting doses, using different administration methods, and combining it with targeted treatments or immunotherapy. Nanoformulations show promise in enhancing doxorubicin's effectiveness by improving drug delivery, reducing side effects, and overcoming drug resistance. This review summarizes recent progress and difficulties in using doxorubicin for breast cancer, highlighting its mechanisms, side effects, ongoing trials, and the potential impact of nanoformulations. Understanding these different aspects is crucial in optimizing doxorubicin's use and improving outcomes for breast cancer patients. This review examines the toxicity of doxorubicin, a drug used in breast cancer treatment, and discusses strategies to mitigate adverse effects, such as cardioprotective agents and liposomal formulations. It also discusses clinical trials evaluating doxorubicin-based regimens, the evolving landscape of combination therapies, and the potential of nanoformulations to optimize delivery and reduce systemic toxicity. The review also discusses the potential of liposomes, nanoparticles, and polymeric micelles to enhance drug accumulation within tumor tissues while sparing healthy organs.
Collapse
Affiliation(s)
- Anjali Bisht
- Department of Pharmaceutical Quality Assurance, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Dubey Avinash
- Department of Pharmaceutical Quality Assurance, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, 17 km Stone, NH-2, Chaumuhan, Mathura, 281406, UP, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
15
|
Xu Z, Li J, Yan N, Liu X, Deng Y, Song Y. Phosphatidylserine and/or Sialic Acid Modified Liposomes Increase Uptake by Tumor-associated Macrophages and Enhance the Anti-tumor Effect. AAPS PharmSciTech 2024; 25:125. [PMID: 38834759 DOI: 10.1208/s12249-024-02837-3] [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: 01/07/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
DOX liposomes have better therapeutic effects and lower toxic side effects. The targeting ability of liposomes is one of the key factors affecting the therapeutic effect of DOX liposomes. This study developed two types of targeted liposomes. Sialic acid (SA)-modified liposomes were designed to target the highly expressed Siglec-1 receptor on tumor-associated macrophages surface. Phosphatidylserine (PS)-modified liposomes were designed to promote phagocytosis by monocyte-derived macrophages through PS apoptotic signaling. In order to assess and compare the therapeutic potential of different targeted pathways in the context of anti-tumor treatment, we compared four phosphatidylserine membrane materials (DOPS, DSPS, DPPS and DMPS) and found that liposomes prepared using DOPS as material could significantly improve the uptake ability of RAW264.7 cells for DOX liposomes. On this basis, normal DOX liposomes (CL-DOX) and SA-modified DOX liposomes (SAL-DOX), PS-modified DOX liposomes (PS-CL-DOX), SA and PS co-modified DOX liposomes (PS-SAL-DOX) were prepared. The anti-tumor cells function of each liposome on S180 and RAW264.7 in vitro was investigated, and it was found that SA on the surface of liposomes can increase the inhibitory effect. In vivo efficacy results exhibited that SAL-DOX and PS-CL-DOX were superior to other groups in terms of ability to inhibit tumor growth and tumor inhibition index, among which SAL-DOX had the best anti-tumor effect. Moreover, SAL-DOX group mice had high expression of IFN-γ as well as IL-12 factors, which could significantly inhibit mice tumor growth, improve the immune microenvironment of the tumor site, and have excellent targeted delivery potential.
Collapse
Affiliation(s)
- Zihan Xu
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Jie Li
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Na Yan
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China.
| |
Collapse
|
16
|
Jørgensen AR, Bue M, Hanberg P, Petersen EK, Harlev C, Hansen J, Baad-Hansen T, Safwat A, Stilling M. Doxorubicin concentrations in bone tumour-relevant tissues after bolus and continuous infusion: a randomized porcine microdialysis study. Cancer Chemother Pharmacol 2024; 93:555-564. [PMID: 38332155 PMCID: PMC11130026 DOI: 10.1007/s00280-023-04637-1] [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: 09/23/2023] [Accepted: 12/24/2023] [Indexed: 02/10/2024]
Abstract
PURPOSE Doxorubicin is a widely used chemotherapeutic drug that can be administered intravenously as both a bolus infusion and a continuous infusion. The latter is believed to lower the risk of cardiotoxicity, which is a critical long-term complication of doxorubicin treatment. The local tissue concentrations of doxorubicin will be reflected in both treatment efficacy and toxicity, but very limited information is available. The aim of this study was to measure the concentration of doxorubicin after continuous and bolus infusion in tissue compartments around a typical location of a bone tumour. METHODS Sixteen pigs (female, Danish Landrace, mean weight 77 kg) were randomized into two groups of eight. Both groups received an intravenous infusion of 150 mg doxorubicin; Group 1 received a bolus infusion (10-15 min) and Group 2 received a continuous infusion (6 h). Before infusion, microdialysis catheters were placed intravenously and in four bone tumour-relevant tissue compartments (cancellous bone, subcutaneous tissue, synovial fluid of the knee joint and muscle tissue). Sampling was done (n = 15) over 24 h, and venous blood samples were collected as a reference. RESULTS Area under the concentration-time curve (AUC0-24 h) for plasma (total concentration) was significantly different between the two groups, while peak drug concentration (Cmax) was significantly higher in two compartments (plasma and synovial fluid of the knee joint) in Group 1 compared to Group 2. Overall, the unbound tissue concentrations were extremely low with values below 0.20 µg/mL. CONCLUSION The pharmacokinetic profile for doxorubicin in the investigated tissues is very similar when comparing bolus and 6 h continuous infusion.
Collapse
Affiliation(s)
- Andrea René Jørgensen
- Aarhus Microdialysis Research Group, Orthopaedic Research Unit, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, J112, 8200, Aarhus N, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark.
| | - Mats Bue
- Aarhus Microdialysis Research Group, Orthopaedic Research Unit, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, J112, 8200, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
- Department of Orthopaedic Surgery, Aarhus University Hospital, Aarhus N, Denmark
| | - Pelle Hanberg
- Aarhus Microdialysis Research Group, Orthopaedic Research Unit, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, J112, 8200, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Elisabeth Krogsgaard Petersen
- Aarhus Microdialysis Research Group, Orthopaedic Research Unit, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, J112, 8200, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Christina Harlev
- Aarhus Microdialysis Research Group, Orthopaedic Research Unit, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, J112, 8200, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Jakob Hansen
- Department of Forensic Medicine, Aarhus University, Aarhus N, Denmark
| | - Thomas Baad-Hansen
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
- Department of Orthopaedic Surgery, Aarhus University Hospital, Aarhus N, Denmark
| | - Akmal Safwat
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus N, Denmark
| | - Maiken Stilling
- Aarhus Microdialysis Research Group, Orthopaedic Research Unit, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, J112, 8200, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
- Department of Orthopaedic Surgery, Aarhus University Hospital, Aarhus N, Denmark
| |
Collapse
|
17
|
Mirzayans R. Changing the Landscape of Solid Tumor Therapy from Apoptosis-Promoting to Apoptosis-Inhibiting Strategies. Curr Issues Mol Biol 2024; 46:5379-5396. [PMID: 38920994 PMCID: PMC11202608 DOI: 10.3390/cimb46060322] [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: 04/17/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
The many limitations of implementing anticancer strategies under the term "precision oncology" have been extensively discussed. While some authors propose promising future directions, others are less optimistic and use phrases such as illusion, hype, and false hypotheses. The reality is revealed by practicing clinicians and cancer patients in various online publications, one of which has stated that "in the quest for the next cancer cure, few researchers bother to look back at the graveyard of failed medicines to figure out what went wrong". The message is clear: Novel therapeutic strategies with catchy names (e.g., synthetic "lethality") have not fulfilled their promises despite decades of extensive research and clinical trials. The main purpose of this review is to discuss key challenges in solid tumor therapy that surprisingly continue to be overlooked by the Nomenclature Committee on Cell Death (NCCD) and numerous other authors. These challenges include: The impact of chemotherapy-induced genome chaos (e.g., multinucleation) on resistance and relapse, oncogenic function of caspase 3, cancer cell anastasis (recovery from late stages of apoptosis), and pitfalls of ubiquitously used preclinical chemosensitivity assays (e.g., cell "viability" and tumor growth delay studies in live animals) that score such pro-survival responses as "lethal" events. The studies outlined herein underscore the need for new directions in the management of solid tumors.
Collapse
Affiliation(s)
- Razmik Mirzayans
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
18
|
M G A, K S A, B S U, P L R, H P S, J S, Joseph MM, T T S. HER2 siRNA Facilitated Gene Silencing Coupled with Doxorubicin Delivery: A Dual Responsive Nanoplatform Abrogates Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2024; 16:25710-25726. [PMID: 38739808 DOI: 10.1021/acsami.4c02532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The present study investigated the concurrent delivery of antineoplastic drug, doxorubicin, and HER2 siRNA through a targeted theranostic metallic gold nanoparticle designed using polysaccharide, PSP001. The as-synthesized HsiRNA@PGD NPs were characterized in terms of structural, functional, physicochemical, and biological properties. HsiRNA@PGD NPs exposed adequate hydrodynamic size, considerable ζ potential, and excellent drug/siRNA loading and encapsulation efficiency. Meticulous exploration of the biocompatible dual-targeted nanoconjugate exhibited an appealing biocompatibility and pH-sensitive cargo release kinetics, indicating its safety for use in clinics. HsiRNA@PGD NPs deciphered competent cancer cell internalization, enhanced cytotoxicity mediated via the induction of apoptosis, and excellent downregulation of the overexpressing target HER2 gene. Further in vivo explorations in the SKBR3 xenograft breast tumor model revealed the appealing tumor reduction properties, selective accumulation in the tumor site followed by significant suppression of the HER2 gene which contributed to the exclusive abrogation of breast tumor mass by the HsiRNA@PGD NPs. Compared to free drugs or the monotherapy constructs, the dual delivery approach produced a synergistic suppression of breast tumors both in vitro and in vivo. Hence the drawings from these findings implicate that the as-synthesized HsiRNA@PGD NPs could offer a promising platform for chemo-RNAi combinational breast cancer therapy.
Collapse
Affiliation(s)
- Archana M G
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre (RCC), Thiruvananthapuram 695011, Kerala, India
| | - Anusree K S
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre (RCC), Thiruvananthapuram 695011, Kerala, India
| | - Unnikrishnan B S
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre (RCC), Thiruvananthapuram 695011, Kerala, India
- Centre for Nanotechnology, Indian Institute of Technology (IIT), Roorkee 247667, Uttarakhand, India
| | - Reshma P L
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre (RCC), Thiruvananthapuram 695011, Kerala, India
| | - Syama H P
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre (RCC), Thiruvananthapuram 695011, Kerala, India
| | - Sreekutty J
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre (RCC), Thiruvananthapuram 695011, Kerala, India
| | - Manu M Joseph
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
- Department of Life Sciences, CHRIST University, Banglore 560029, India
| | - Sreelekha T T
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research, Regional Cancer Centre (RCC), Thiruvananthapuram 695011, Kerala, India
| |
Collapse
|
19
|
Pan R, Lin C, Yang X, Xie Y, Gao L, Yu L. The influence of spheroid maturity on fusion dynamics and micro-tissue assembly in 3D tumor models. Biofabrication 2024; 16:035016. [PMID: 38663395 DOI: 10.1088/1758-5090/ad4392] [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: 11/23/2023] [Accepted: 04/25/2024] [Indexed: 07/02/2024]
Abstract
Three-dimensional (3D) cell culture has been used in many fields of biology because of its unique advantages. As a representative of the 3D systems, 3D spheroids are used as building blocks for tissue construction. Larger tumor aggregates can be assembled by manipulating or stacking the tumor spheroids. The motivation of this study is to investigate the behavior of the cells distributed at different locations of the spheroids in the fusion process and the mechanism behind it. To this aim, spheroids with varying grades of maturity or age were generated for fusion to assemble micro-tumor tissues. The dynamics of the fusion process, the motility of the cells distributed in different heterogeneous architecture sites, and their reactive oxygen species profiles were studied. We found that the larger the spheroid necrotic core, the slower the fusion rate of the spheroid. The cells that move were mainly distributed on the spheroid's surface during fusion. In addition to dense microfilament distribution and low microtubule content, the reactive oxygen content was high in the fusion site, while the non-fusion site was the opposite. Last, multi-spheroids with different maturities were fused to complex micro-tissues to mimic solid tumors and evaluate Doxorubicin's anti-tumor efficacy.
Collapse
Affiliation(s)
- Rong Pan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing 400715, People's Republic of China
| | - Chenyu Lin
- Institute for Developmental and Biology and Regenerative Medicine, Southwest University, Chongqing 400715, People's Republic of China
| | - Xiaoyan Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing 400715, People's Republic of China
| | - Yuanyuan Xie
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing 400715, People's Republic of China
| | - Lixia Gao
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, College of Pharmacy & International Academy of Targeted Therapeutics and Innovation, Chongqing University of Arts and Sciences, Chongqing 402160, People's Republic of China
| | - Ling Yu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute for Clean Energy and Advanced Materials, School of Materials and Energy, Southwest University, Chongqing 400715, People's Republic of China
| |
Collapse
|
20
|
Tapaswi A, Cemalovic N, Polemi KM, Sexton JZ, Colacino JA. Applying Cell Painting in Non-Tumorigenic Breast Cells to Understand Impacts of Common Chemical Exposures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591893. [PMID: 38746407 PMCID: PMC11092634 DOI: 10.1101/2024.04.30.591893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
There are a substantial number of chemicals to which individuals in the general population are exposed which have putative, but still poorly understood, links to breast cancer. Cell Painting is a high-content imaging-based in vitro assay that allows for rapid and unbiased measurements of the concentration-dependent effects of chemical exposures on cellular morphology. We optimized the Cell Painting assay and measured the effect of exposure to 16 human exposure relevant chemicals, along with 21 small molecules with known mechanisms of action, for 48 hours in non-tumorigenic mammary epithelial cells, the MCF10A cell line. Through unbiased imaging analyses using CellProfiler, we quantified 3042 morphological features across approximately 1.2 million cells. We used benchmark concentration modeling to quantify significance and dose-dependent directionality to identify morphological features conserved across chemicals and find features that differentiate the effects of toxicants from one another. Benchmark concentrations were compared to chemical exposure biomarker concentration measurements from the National Health and Nutrition Examination Survey to assess which chemicals induce morphological alterations at human-relevant concentrations. Morphometric fingerprint analysis revealed similar phenotypes between small molecules and prioritized NHANES-toxicants guiding further investigation. A comparison of feature fingerprints via hypergeometric analysis revealed significant feature overlaps between chemicals when stratified by compartment and stain. One such example was the similarities between a metabolite of the organochlorine pesticide DDT (p,p'-DDE) and an activator of canonical Wnt signaling CHIR99201. As CHIR99201 is a known Wnt pathway activator and its role in β-catenin translocation is well studied, we studied the translocation of β-catenin following p'-p' DDE exposure in an orthogonal high-content imaging assay. Consistent with activation of Wnt signaling, low dose p',p'-DDE (25nM) significantly enhances the nuclear translocation of β-catenin. Overall, these findings highlight the ability of Cell Painting to enhance mode-of-action studies for toxicants which are common exposures in our environment but have previously been incompletely characterized with respect to breast cancer risk.
Collapse
Affiliation(s)
- Anagha Tapaswi
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas Cemalovic
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Katelyn M Polemi
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Z Sexton
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Medicinal Chemistry, University of Michigan School of Pharmacy, Ann Arbor, MI, USA
| | - Justin A Colacino
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA
- Program in the Environment, University of Michigan, Ann Arbor MI, USA
| |
Collapse
|
21
|
Bilgin S. Apoptotic effect of 5-fluorouracil-doxorubicin combination on colorectal cancer cell monolayers and spheroids. Mol Biol Rep 2024; 51:603. [PMID: 38698270 DOI: 10.1007/s11033-024-09562-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: 01/25/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Drug combination studies help to improve new treatment approaches for colon cancer. Tumor spheroids (3D) are better models than traditional 2-dimensional cultures (2D) to evaluate cellular responses to chemotherapy drugs. The cultivation of cancer cells in 2D and 3D cultures affects the apoptotic process, which is a major factor influencing the response of cancer cells to chemotherapeutic drugs. In this study, the antiproliferative effects of 5-fluorouracil (5-FU) and doxorubicin (DOX) were investigated separately and in combination using 2D and 3D cell culture models on two different colon cancer cell lines, HT-29 (apoptosis-resistant cells) and Caco-2 2 (apoptosis-susceptible cells). METHODS The effect of the drugs on the proliferation of both colon cancer cells was determined by performing an MTT assay in 2D culture. The apoptotic effect of 5-FU and DOX, both as single agents and in combination, was assessed in 2D and 3D cultures through quantitative real-time polymerase chain reaction analysis. The expression of apoptotic genes, such as caspases, p53, Bax, and Bcl-2, was quantified. RESULTS It was found that the mRNA expression of proapoptotic genes was significantly upregulated, whereas the mRNA expression of the antiapoptotic Bcl-2 gene was significantly downregulated in both colon cancer models treated with 5-FU, DOX, and 5-FU + DOX. CONCLUSION The results indicated that the 5-FU + DOX combination therapy induces apoptosis and renders 5-FU and DOX more effective at lower concentrations compared to their alone use. This study reveals promising results in reducing the potential side effects of treatment by enabling the use of lower drug doses.
Collapse
Affiliation(s)
- Sema Bilgin
- Department of Medical Laboratory Techniques, Tokat Vocational School of Health Services, Tokat Gaziosmanpasa University, 60000, Tokat, Turkey.
| |
Collapse
|
22
|
Dutta RS, Elhassan GO, Devi TB, Bhattacharjee B, Singh M, Jana BK, Sahu S, Mazumder B, Sahu RK, Khan J. Enhanced efficacy of β-carotene loaded solid lipid nanoparticles optimized and developed via central composite design on breast cancer cell lines. Heliyon 2024; 10:e28457. [PMID: 38586388 PMCID: PMC10998123 DOI: 10.1016/j.heliyon.2024.e28457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/16/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024] Open
Abstract
β-carotene is obtained from both plants and animals and has been the subject of intense research because of its provitamin-A, antioxidant, and anticancer effects. Its limited absorption and oxidative degradation significantly reduce its antitumor efficacy when taken orally. In our study, we utilize a central composite design to develop "bio-safe and highly bio-compatible" solid lipid nanoparticles (SLNs) by using only the combination of palmitic acid and poloxamer-407, a block co-polymer as a surfactant. The current research aim to develop and characterize SLNs loaded with β-carotene to improve their bioavailability and therapeutic efficacy. In addition, the improved cytotoxicity of solid lipid nanoparticles loaded with β-carotene was screened in-vitro in human breast cancer cell lines (MCF-7). The nanoparticles exhibits good stability, as indicated by their mean zeta potential of -26.3 ± 1.3 mV. The particles demonstrated high drug loading and entrapment capabilities. The fabricated nanoparticle's prolonged release potential was shown by the in-vitro release kinetics, which showed a first-order release pattern that adhered to the Higuchi model and showed a slow, linear, and steady release over 48 h. Moreover, a diffusion-type release mechanism was used to liberate β-carotene from the nanoparticles. For six months, the nanoparticles also showed a notable degree of physical stability. Lastly, using the MTT assay, the anti-cancer properties of β-carotene-loaded solid lipid nanoparticles were compared with intact β-carotene on MCF-7 cell lines. The cytotoxicity tests have shown that the encapsulation of β-carotene in the lipid bilayers of the optimized formulation does not interfere with the anti-cancer activity of the drug. When compared to standard β-carotene, β-carotene loaded SLNs showed enhanced anticancer efficacy and it is a plausible therapeutic candidate for enhancing the solubility of water-insoluble and degradation-sensitive biotherapeutics like β-carotene.
Collapse
Affiliation(s)
- Rajat Subhra Dutta
- School of Pharmaceutical Sciences, Girijananda Chowdhury University-Tezpur Campus, 784501, Assam, India
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Gamal Osman Elhassan
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraidah, 52571, Saudi Arabia
| | | | - Bedanta Bhattacharjee
- School of Pharmaceutical Sciences, Girijananda Chowdhury University-Tezpur Campus, 784501, Assam, India
| | - Mohini Singh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Bani Kumar Jana
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Supriya Sahu
- School of Pharmaceutical Sciences, Girijananda Chowdhury University-Tezpur Campus, 784501, Assam, India
| | - Bhaskar Mazumder
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Ram Kumar Sahu
- Department of Pharmaceutical Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Chauras Campus, Tehri Garhwal, Uttarakhand, India
| | - Jiyauddin Khan
- School of Pharmacy, Management and Science University, 40100, Shah Alam, Selangor, Malaysia
| |
Collapse
|
23
|
Wang T, Xing G, Fu T, Ma Y, Wang Q, Zhang S, Chang X, Tong Y. Role of mitochondria in doxorubicin-mediated cardiotoxicity: From molecular mechanisms to therapeutic strategies. Cell Stress Chaperones 2024; 29:349-357. [PMID: 38485043 PMCID: PMC10999808 DOI: 10.1016/j.cstres.2024.03.003] [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: 12/22/2023] [Accepted: 03/09/2024] [Indexed: 04/05/2024] Open
Abstract
This comprehensive review delves into the pivotal role of mitochondria in doxorubicin-induced cardiotoxicity, a significant complication limiting the clinical use of this potent anthracycline chemotherapeutic agent. Doxorubicin, while effective against various malignancies, is associated with dose-dependent cardiotoxicity, potentially leading to irreversible cardiac damage. The review meticulously dissects the molecular mechanisms underpinning this cardiotoxicity, particularly focusing on mitochondrial dysfunction, a central player in this adverse effect. Central to the discussion is the concept of mitochondrial quality control, including mitochondrial dynamics (fusion/fission balance) and mitophagy. The review presents evidence linking aberrations in these processes to cardiotoxicity in doxorubicin-treated patients. It elucidates how doxorubicin disrupts mitochondrial dynamics, leading to an imbalance between mitochondrial fission and fusion, and impairs mitophagy, culminating in the accumulation of dysfunctional mitochondria and subsequent cardiac cell damage. Furthermore, the review explores emerging therapeutic strategies targeting mitochondrial dysfunction. It highlights the potential of modulating mitochondrial dynamics and enhancing mitophagy to mitigate doxorubicin-induced cardiac damage. These strategies include pharmacological interventions with mitochondrial fission inhibitors, fusion promoters, and agents that modulate mitophagy. The review underscores the promising results from preclinical studies while advocating for more extensive clinical trials to validate these approaches in human patients. In conclusion, this review offers valuable insights into the intricate relationship between mitochondrial dysfunction and doxorubicin-mediated cardiotoxicity. It underscores the need for continued research into targeted mitochondrial therapies as a means to improve the cardiac safety profile of doxorubicin, thereby enhancing the overall treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Tianen Wang
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Guoli Xing
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tong Fu
- Brandeis University, Waltham, MA, USA
| | - Yanchun Ma
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qi Wang
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shuxiang Zhang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xing Chang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ying Tong
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
24
|
Jørgensen AR, Hansen J, Bue M, Hanberg P, Stilling M. Microdialysis as a sampling tool for the chemotherapeutic agent Doxorubicin. J Pharm Biomed Anal 2024; 239:115872. [PMID: 38039870 DOI: 10.1016/j.jpba.2023.115872] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/13/2023] [Accepted: 11/21/2023] [Indexed: 12/03/2023]
Abstract
Doxorubicin is a chemotherapeutic agent used for more than fifty years to treat a great variety of cancers in both children and adults. Despite hereof, pharmacokinetic knowledge is almost solely based on systemic plasma concentrations. Microdialysis is a catheter-based pharmacokinetic sampling tool enabling simultaneous target site sampling of unbound molecules of interest. The aim of this study was to thoroughly evaluate the feasibility of applying microdialysis for sampling of Doxorubicin in both in vitro experiments and an in vivo setting. Doxorubicin relative recovery by gain and by loss was tested for different catheter types, perfusion fluids, concentrations and collection vials. Adsorption tests revealed polystyrene/santoprene vials to be the biggest contributor of unwanted adsorption between Doxorubicin and the microdialysis equipment, and confirmed LoBind Eppendorf tubes to be a suitable alternative. The methodological combination of polyamide membranes, saline as perfusion fluid and LoBind Eppendorf sampling tubes demonstrated no statistically significant differences for relative recovery by gain and by loss, and the relative recovery was also found to be concentration independent. We conclude, that a proper microdialysis set-up can be used to collect samples containing concentrations of the chemotherapeutic drug Doxorubicin in vitro and in vivo, which encourage future pharmacokinetic studies to evaluate current treatment regimens to find the most effective and least toxic anti-neoplastic treatment for the patients.
Collapse
Affiliation(s)
- Andrea René Jørgensen
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Unit, Aarhus University Hospital, Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark.
| | - Jakob Hansen
- Department of Forensic Medicine, Aarhus University, Aarhus N, Denmark
| | - Mats Bue
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Unit, Aarhus University Hospital, Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark; Department of Orthopaedic Surgery, Aarhus University Hospital, Aarhus N, Denmark
| | - Pelle Hanberg
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Unit, Aarhus University Hospital, Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Maiken Stilling
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Unit, Aarhus University Hospital, Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark; Department of Orthopaedic Surgery, Aarhus University Hospital, Aarhus N, Denmark
| |
Collapse
|
25
|
Dougherty BV, Moore CJ, Rawls KD, Jenior ML, Chun B, Nagdas S, Saucerman JJ, Kolling GL, Wallqvist A, Papin JA. Identifying metabolic adaptations characteristic of cardiotoxicity using paired transcriptomics and metabolomics data integrated with a computational model of heart metabolism. PLoS Comput Biol 2024; 20:e1011919. [PMID: 38422168 PMCID: PMC10931521 DOI: 10.1371/journal.pcbi.1011919] [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: 12/13/2022] [Revised: 03/12/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Improvements in the diagnosis and treatment of cancer have revealed long-term side effects of chemotherapeutics, particularly cardiotoxicity. Here, we present paired transcriptomics and metabolomics data characterizing in vitro cardiotoxicity to three compounds: 5-fluorouracil, acetaminophen, and doxorubicin. Standard gene enrichment and metabolomics approaches identify some commonly affected pathways and metabolites but are not able to readily identify metabolic adaptations in response to cardiotoxicity. The paired data was integrated with a genome-scale metabolic network reconstruction of the heart to identify shifted metabolic functions, unique metabolic reactions, and changes in flux in metabolic reactions in response to these compounds. Using this approach, we confirm previously seen changes in the p53 pathway by doxorubicin and RNA synthesis by 5-fluorouracil, we find evidence for an increase in phospholipid metabolism in response to acetaminophen, and we see a shift in central carbon metabolism suggesting an increase in metabolic demand after treatment with doxorubicin and 5-fluorouracil.
Collapse
Affiliation(s)
- Bonnie V. Dougherty
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Connor J. Moore
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Kristopher D. Rawls
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Matthew L. Jenior
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Bryan Chun
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sarbajeet Nagdas
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Glynis L. Kolling
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland, United States of America
| | - Jason A. Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
26
|
Mirzayans R, Andrais B, Murray D. Single-Cell MTT: A Simple and Sensitive Assay for Determining the Viability and Metabolic Activity of Polyploid Giant Cancer Cells (PGCCs). Methods Mol Biol 2024; 2825:293-308. [PMID: 38913317 DOI: 10.1007/978-1-0716-3946-7_17] [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: 06/25/2024]
Abstract
Solid tumors and tumor-derived cell lines commonly contain highly enlarged (giant) cancer cells that enter a state of transient dormancy (active sleep) after they are formed, but retain viability, secrete growth promoting factors, and exhibit the ability to generate rapidly proliferating progeny with stem cell-like properties. Giant cells with a highly enlarged nucleus or multiple nuclei are often called polyploid giant cancer cells (PGCCs). Although PGCCs constitute only a subset of cells within a solid tumor/tumor-derived cell line, their frequency can increase markedly following exposure to ionizing radiation or chemotherapeutic drugs. In this chapter we outline a simple and yet highly sensitive cell-based assay, called single-cell MTT, that we have optimized for determining the viability and metabolic activity of PGCCs before and after exposure to anticancer agents. The assay measures the ability of individual PGCCs to convert the MTT tetrazolium salt to its water insoluble formazan metabolite. In addition to evaluating PGCCs, this assay is also a powerful tool for determining the viability and metabolic activity of cancer cells undergoing premature senescence following treatment with anticancer agents, as well as for distinguishing dead cancer cells and dying cells (e.g., exhibiting features of apoptosis, ferroptosis, etc.) that have the potential to resume proliferation through a process called anastasis.
Collapse
Affiliation(s)
- Razmik Mirzayans
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB, Canada.
| | - Bonnie Andrais
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB, Canada
| | - David Murray
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB, Canada
| |
Collapse
|
27
|
Zhang S, Chen M, Geng Z, Liu T, Li S, Yu Q, Cao L, Liu D. Potential Application of Self-Assembled Peptides and Proteins in Breast Cancer and Cervical Cancer. Int J Mol Sci 2023; 24:17056. [PMID: 38069380 PMCID: PMC10706889 DOI: 10.3390/ijms242317056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Ongoing research is gradually broadening the idea of cancer treatment, with attention being focused on nanoparticles to improve the stability, therapeutic efficacy, targeting, and other important metrics of conventional drugs and traditional drug delivery methods. Studies have demonstrated that drug delivery carriers based on biomaterials (e.g., protein nanoparticles and lipids) and inorganic materials (e.g., metal nanoparticles) have potential anticancer effects. Among these carriers, self-assembled proteins and peptides, which are highly biocompatible and easy to standardize and produce, are strong candidates for the preparation of anticancer drugs. Breast cancer (BC) and cervical cancer (CC) are two of the most common and deadly cancers in women. These cancers not only threaten lives globally but also put a heavy burden on the healthcare system. Despite advances in medical care, the incidence of these two cancers, particularly CC, which is almost entirely preventable, continues to rise, and the mortality rate remains steady. Therefore, there is still a need for in-depth research on these two cancers to develop more targeted, efficacious, and safe therapies. This paper reviews the types of self-assembling proteins and peptides (e.g., ferritin, albumin, and virus-like particles) and natural products (e.g., soy and paclitaxel) commonly used in the treatment of BC and CC and describes the types of drugs that can be delivered using self-assembling proteins and peptides as carriers (e.g., siRNAs, DNA, plasmids, and mRNAs). The mechanisms (including self-assembly) by which the natural products act on CC and BC are discussed. The mechanism of action of natural products on CC and BC and the mechanism of action of self-assembled proteins and peptides have many similarities (e.g., NF-KB and Wnt). Thus, natural products using self-assembled proteins and peptides as carriers show potential for the treatment of BC and CC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lingling Cao
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.Z.); (M.C.); (Z.G.); (T.L.); (S.L.); (Q.Y.)
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.Z.); (M.C.); (Z.G.); (T.L.); (S.L.); (Q.Y.)
| |
Collapse
|
28
|
Abdelmaksoud NM, Abulsoud AI, Doghish AS, Abdelghany TM. From resistance to resilience: Uncovering chemotherapeutic resistance mechanisms; insights from established models. Biochim Biophys Acta Rev Cancer 2023; 1878:188993. [PMID: 37813202 DOI: 10.1016/j.bbcan.2023.188993] [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: 07/24/2023] [Revised: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023]
Abstract
Despite the tremendous advances in cancer treatment, resistance to chemotherapeutic agents impedes higher success rates and accounts for major relapses in cancer therapy. Moreover, the resistance of cancer cells to chemotherapy is linked to low efficacy and high recurrence of cancer. To stand up against chemotherapy resistance, different models of chemotherapy resistance have been established to study various molecular mechanisms of chemotherapy resistance. Consequently, this review is going to discuss different models of induction of chemotherapy resistance, highlighting the most common mechanisms of cancer resistance against different chemotherapeutic agents, including overexpression of efflux pumps, drug inactivation, epigenetic modulation, and epithelial-mesenchymal transition. This review aims to open a new avenue for researchers to lower the resistance to the existing chemotherapeutic agents, develop new therapeutic agents with low resistance potential, and establish possible prognostic markers for chemotherapy resistance.
Collapse
Affiliation(s)
- Nourhan M Abdelmaksoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, 3 Cairo-Belbeis Desert Road, P.O. Box 3020 El Salam, 11785 Cairo, Egypt.
| | - Ahmed I Abulsoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, 3 Cairo-Belbeis Desert Road, P.O. Box 3020 El Salam, 11785 Cairo, Egypt; Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11823, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11823, Egypt
| | - Tamer M Abdelghany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11884, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, 3 Cairo-Belbeis Desert Road, P.O. Box 3020 El Salam, 11785 Cairo, Egypt.
| |
Collapse
|
29
|
Alioglu MA, Yilmaz YO, Singh YP, Nagamine M, Celik N, Kim MH, Pal V, Gupta D, Ozbolat IT. Nested biofabrication: Matryoshka-inspired Intra-embedded Bioprinting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.560028. [PMID: 37808743 PMCID: PMC10557751 DOI: 10.1101/2023.09.28.560028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Engineering functional tissues and organs remains a fundamental pursuit in biofabrication. However, the accurate constitution of complex shapes and internal anatomical features of specific organs, including their intricate blood vessels and nerves, remains a significant challenge. Inspired by the Matryoshka doll, we here introduce a new method called 'Intra-Embedded Bioprinting (IEB),' building upon existing embedded bioprinting methods. We used a xanthan gum-based material, which served a dual role as both a bioprintable ink and a support bath, due to its unique shear-thinning and self-healing properties. We demonstrated IEB's capabilities in organ modelling, creating a miniaturized replica of a pancreas using a photocrosslinkable silicone composite. Further, a head phantom and a Matryoshka doll were 3D printed, exemplifying IEB's capability to manufacture intricate, nested structures. Towards the use case of IEB and employing innovative coupling strategy between extrusion-based and aspiration-assisted bioprinting, we developed a breast tumor model that included a central channel mimicking a blood vessel, with tumor spheroids bioprinted in proximity. Validation using a clinically-available chemotherapeutic drug illustrated its efficacy in reducing the tumor volume via perfusion over time. This method opens a new way of bioprinting enabling the creation of complex-shaped organs with internal anatomical features.
Collapse
|
30
|
Mattioli R, Ilari A, Colotti B, Mosca L, Fazi F, Colotti G. Doxorubicin and other anthracyclines in cancers: Activity, chemoresistance and its overcoming. Mol Aspects Med 2023; 93:101205. [PMID: 37515939 DOI: 10.1016/j.mam.2023.101205] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/31/2023]
Abstract
Anthracyclines have been important and effective treatments against a number of cancers since their discovery. However, their use in therapy has been complicated by severe side effects and toxicity that occur during or after treatment, including cardiotoxicity. The mode of action of anthracyclines is complex, with several mechanisms proposed. It is possible that their high toxicity is due to the large set of processes involved in anthracycline action. The development of resistance is a major barrier to successful treatment when using anthracyclines. This resistance is based on a series of mechanisms that have been studied and addressed in recent years. This work provides an overview of the anthracyclines used in cancer therapy. It discusses their mechanisms of activity, toxicity, and chemoresistance, as well as the approaches used to improve their activity, decrease their toxicity, and overcome resistance.
Collapse
Affiliation(s)
- Roberto Mattioli
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy
| | - Beatrice Colotti
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Luciana Mosca
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy.
| |
Collapse
|
31
|
Rossi V, Govoni M, Di Stefano G. Lactate Can Modulate the Antineoplastic Effects of Doxorubicin and Relieve the Drug's Oxidative Damage on Cardiomyocytes. Cancers (Basel) 2023; 15:3728. [PMID: 37509389 PMCID: PMC10378253 DOI: 10.3390/cancers15143728] [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: 05/24/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Doxorubicin (DOXO) is currently administered as the first-choice therapy for a variety of malignancies. Cancer cells exhibit enhanced glycolysis and lactate production. This metabolite affects gene expression and can play a role in chemoresistance. AIM OF THIS STUDY We investigated whether the enhanced lactate levels that characterize neoplastic tissues can modify the response of cancer cells to DOXO. METHODS After exposing cancer cells to increased lactate levels, we examined whether this metabolite could interfere with the principal mechanisms responsible for the DOXO antineoplastic effect. RESULTS Increased lactate levels did not affect DOXO-induced topoisomerase poisoning but offered protection against the oxidative damage caused by the drug. This protection was related to changes in gene expression caused by the combined action of DOXO and lactate. Oxidative damage significantly contributed to the heavy cardiotoxicity following DOXO treatment. In cultured cardiomyocytes, we confirmed that DOXO-induced DNA damage and oxidative stress can be significantly mitigated by exposing the cells to increased lactate levels. CONCLUSIONS In addition to contributing to elucidating the effects of the combined action of DOXO and lactate, our results suggest a possible method to reduce the heavy drug cardiotoxicity, a major side effect leading to therapy discontinuation.
Collapse
Affiliation(s)
- Valentina Rossi
- Department of Medical and Surgical Sciences (DIMEC), Section of General Pathology, University of Bologna, 40126 Bologna, Italy
| | - Marzia Govoni
- Department of Medical and Surgical Sciences (DIMEC), Section of General Pathology, University of Bologna, 40126 Bologna, Italy
| | - Giuseppina Di Stefano
- Department of Medical and Surgical Sciences (DIMEC), Section of General Pathology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
32
|
Rushing BR, Molina S, Sumner S. Metabolomics Analysis Reveals Altered Metabolic Pathways and Response to Doxorubicin in Drug-Resistant Triple-Negative Breast Cancer Cells. Metabolites 2023; 13:865. [PMID: 37512572 PMCID: PMC10383792 DOI: 10.3390/metabo13070865] [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: 06/14/2023] [Revised: 07/07/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
This study aimed to investigate metabolic changes following the acquisition of resistance to doxorubicin in the triple-negative breast cancer (TNBC) cell line MDA-MB-231. Two drug-resistant cell lines, DOX-RES-50 and DOX-RES-100, were generated by treating MDA-MB-231 cells with doxorubicin for 24 h and allowing them to recover for six weeks. Both drug-resistant cell lines demonstrated an increase in doxorubicin IC50 values, indicating acquired drug resistance. Metabolomics analysis showed clear separation between the parental MDA-MB-231 cell line and the drug-resistant cell lines. Pathway analysis revealed that arginine and proline metabolism, glutathione metabolism, and beta-alanine metabolism were significantly perturbed in the drug-resistant cell lines compared to the parental cell line. After matching signals to an in-house library of reference standards, significant decreases in short- and medium-chain acylcarnitines and significant increases in long-chain acylcarnitines, 5-oxoproline, and 7-ketodeoxycholic acid were observed in the resistant cell lines as compared to the parental MDA-MB-231 cell line. In addition to baseline metabolic differences, we also investigated differences in metabolic responses in resistant cell lines upon a second exposure at multiple concentrations. Results indicate that whereas the parental MDA-MB-231 cell line had many metabolites that responded to doxorubicin in a dose-dependent manner, the two resistant cell lines lost a dose-dependent response for the majority of these metabolites. The study's findings provide insight into how metabolism is altered during the acquisition of resistance in TNBC cells and how the metabolic response to doxorubicin changes upon repeated treatment. This information can potentially identify novel targets to prevent or reverse multi-drug resistance in TNBC, and also demonstrate the usefulness of metabolomics technology in identifying new mechanisms of drug resistance in cancer and potential drug targets.
Collapse
Affiliation(s)
- Blake R Rushing
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| | - Sabrina Molina
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| | - Susan Sumner
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| |
Collapse
|
33
|
Jordan HA, Thomas SN. Novel proteomic technologies to address gaps in pre-clinical ovarian cancer biomarker discovery efforts. Expert Rev Proteomics 2023; 20:439-450. [PMID: 38116719 DOI: 10.1080/14789450.2023.2295861] [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: 08/05/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
INTRODUCTION An estimated 20,000 women in the United States will receive a diagnosis of ovarian cancer in 2023. Late-stage diagnosis is associated with poor prognosis. There is a need for novel diagnostic biomarkers for ovarian cancer to improve early-stage detection and novel prognostic biomarkers to improve patient treatment. AREAS COVERED This review provides an overview of the clinicopathological features of ovarian cancer and the currently available biomarkers and treatment options. Two affinity-based platforms using proximity extension assays (Olink) and DNA aptamers (SomaLogic) are described in the context of highly reproducible and sensitive multiplexed assays for biomarker discovery. Recent developments in ion mobility spectrometry are presented as novel techniques to apply to the biomarker discovery pipeline. Examples are provided of how these aforementioned methods are being applied to biomarker discovery efforts in various diseases, including ovarian cancer. EXPERT OPINION Translating novel ovarian cancer biomarkers from candidates in the discovery phase to bona fide biomarkers with regulatory approval will have significant benefits for patients. Multiplexed affinity-based assay platforms and novel mass spectrometry methods are capable of quantifying low abundance proteins to aid biomarker discovery efforts by enabling the robust analytical interrogation of the ovarian cancer proteome.
Collapse
Affiliation(s)
- Helen A Jordan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Stefani N Thomas
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
34
|
Li F, Cao Y, Kan X, Li D, Li Y, Huang C, Liu P. AS1411-conjugated doxorubicin-loaded silver nanotriangles for targeted chemo-photothermal therapy of breast cancer. Nanomedicine (Lond) 2023; 18:1077-1094. [PMID: 37650546 DOI: 10.2217/nnm-2023-0158] [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: 09/01/2023] Open
Abstract
Background: Combination therapy has attracted tremendous interest for its great potential in treating cancers. Materials & methods: Based on chitosan-coated silver nanotriangles, polyethylene glycol, AS1411 aptamer and doxorubicin, a multifunctional nanocomposite (AS1411-DOX-AgNTs) was constructed and characterized. Then the photothermal properties, ability to target breast cancer cells and anti-breast cancer effect of AS1411-DOX-AgNTs were evaluated. Results: AS1411-DOX-AgNTs were successfully fabricated and showed excellent photothermal conversion efficiency, breast cancer cell and tumor targeting ability. Compared with single treatments, the combination of AS1411-DOX-AgNTs with near-infrared irradiation possessed the strongest anti-breast cancer effect in vitro and in vivo. Conclusion: AS1411-DOX-AgNTs hold great potential in targeted DOX delivery and combined chemo-photothermal therapy for breast cancer.
Collapse
Affiliation(s)
- Fan Li
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yuyu Cao
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Xuechun Kan
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Dongdong Li
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yan Li
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Cheng Huang
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Peidang Liu
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
- Jiangsu Key Laboratory for Biomaterials & Devices, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| |
Collapse
|
35
|
Demina PA, Khaydukov KV, Babayeva G, Varaksa PO, Atanova AV, Stepanov ME, Nikolaeva ME, Krylov IV, Evstratova II, Pokrovsky VS, Zhigarkov VS, Akasov RA, Egorova TV, Khaydukov EV, Generalova AN. Upconversion Nanoparticles Intercalated in Large Polymer Micelles for Tumor Imaging and Chemo/Photothermal Therapy. Int J Mol Sci 2023; 24:10574. [PMID: 37445751 DOI: 10.3390/ijms241310574] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Frontiers in theranostics are driving the demand for multifunctional nanoagents. Upconversion nanoparticle (UCNP)-based systems activated by near-infrared (NIR) light deeply penetrating biotissue are a powerful tool for the simultaneous diagnosis and therapy of cancer. The intercalation into large polymer micelles of poly(maleic anhydride-alt-1-octadecene) provided the creation of biocompatible UCNPs. The intrinsic properties of UCNPs (core@shell structure NaYF4:Yb3+/Tm3+@NaYF4) embedded in micelles delivered NIR-to-NIR visualization, photothermal therapy, and high drug capacity. Further surface modification of micelles with a thermosensitive polymer (poly-N-vinylcaprolactam) exhibiting a conformation transition provided gradual drug (doxorubicin) release. In addition, the decoration of UCNP micelles with Ag nanoparticles (Ag NPs) synthesized in situ by silver ion reduction enhanced the cytotoxicity of micelles at cell growth temperature. Cell viability assessment on Sk-Br-3, MDA-MB-231, and WI-26 cell lines confirmed this effect. The efficiency of the prepared UCNP complex was evaluated in vivo by Sk-Br-3 xenograft regression in mice for 25 days after peritumoral injection and photoactivation of the lesions with NIR light. The designed polymer micelles hold promise as a photoactivated theranostic agent with quattro-functionalities (NIR absorption, photothermal effect, Ag NP cytotoxicity, and Dox loading) that provides imaging along with chemo- and photothermal therapy enhanced with Ag NPs.
Collapse
Affiliation(s)
- Polina A Demina
- Federal Scientific Research Center «Crystallography and Photonics» of the Russian Academy of Sciences, 119333 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
- Institute of Physics, Technology, and Informational Systems, Moscow State Pedagogical University, 119435 Moscow, Russia
| | - Kirill V Khaydukov
- Federal Scientific Research Center «Crystallography and Photonics» of the Russian Academy of Sciences, 119333 Moscow, Russia
- Institute of Physics, Technology, and Informational Systems, Moscow State Pedagogical University, 119435 Moscow, Russia
| | - Gulalek Babayeva
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 115478 Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Pavel O Varaksa
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 115478 Moscow, Russia
| | - Alexandra V Atanova
- Federal Scientific Research Center «Crystallography and Photonics» of the Russian Academy of Sciences, 119333 Moscow, Russia
| | - Maxim E Stepanov
- Institute of Physics, Technology, and Informational Systems, Moscow State Pedagogical University, 119435 Moscow, Russia
| | - Maria E Nikolaeva
- Institute of Physics, Technology, and Informational Systems, Moscow State Pedagogical University, 119435 Moscow, Russia
| | - Ivan V Krylov
- Federal Scientific Research Center «Crystallography and Photonics» of the Russian Academy of Sciences, 119333 Moscow, Russia
| | - Irina I Evstratova
- Institute of Physics, Technology, and Informational Systems, Moscow State Pedagogical University, 119435 Moscow, Russia
| | - Vadim S Pokrovsky
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, 115478 Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Vyacheslav S Zhigarkov
- Federal Scientific Research Center «Crystallography and Photonics» of the Russian Academy of Sciences, 119333 Moscow, Russia
| | - Roman A Akasov
- Federal Scientific Research Center «Crystallography and Photonics» of the Russian Academy of Sciences, 119333 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
- Institute of Physics, Technology, and Informational Systems, Moscow State Pedagogical University, 119435 Moscow, Russia
- Institute of Molecular Theranostics, Sechenov University, 119991 Moscow, Russia
| | - Tatiana V Egorova
- Institute of Physics, Technology, and Informational Systems, Moscow State Pedagogical University, 119435 Moscow, Russia
| | - Evgeny V Khaydukov
- Federal Scientific Research Center «Crystallography and Photonics» of the Russian Academy of Sciences, 119333 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
- Institute of Physics, Technology, and Informational Systems, Moscow State Pedagogical University, 119435 Moscow, Russia
- Institute of Molecular Theranostics, Sechenov University, 119991 Moscow, Russia
| | - Alla N Generalova
- Federal Scientific Research Center «Crystallography and Photonics» of the Russian Academy of Sciences, 119333 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
| |
Collapse
|
36
|
Babalola AA, Adelowo AR, Da-Silva OF, Ikeji CN, Owoeye O, Rocha JBT, Adedara IA, Farombi EO. Attenuation of doxorubicin-induced hypothalamic-pituitary-testicular axis dysfunction by diphenyl diselenide involves suppression of hormonal deficits, oxido-inflammatory stress and caspase 3 activity in rats. J Trace Elem Med Biol 2023; 79:127254. [PMID: 37379681 DOI: 10.1016/j.jtemb.2023.127254] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 05/27/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Doxorubicin (DOX) is one of the popular anti-cancer drugs in the world and several literatures have implicated it in various toxicities especially cardiotoxicity and reproductive toxicity. Diphenyl diselenide (DPDS) is well acknowledged for its compelling pharmacological effects in numerous disease models and chemically-mediated toxicity. This study was carried out to investigate the effect of DPDS on DOX-induced changes in the reproductive indices of male Wistar rats. METHODS Rats were intraperitoneally injected with 7.5 mg/kg body weight of DOX alone once followed by treatment with DPDS at 5 and 10 mg/kg for seven successive days. Excised hypothalamus, testes and epididymis were processed for biochemical and histological analyses. RESULTS DPDS treatment significantly (p < 0.05) abated DOX-induced oxidative damage by decreasing the levels of oxidative stress indices such as hydrogen peroxide, reactive oxygen and nitrogen species, and lipid peroxidation with a respective improvement in the level of glutathione in the hypothalamic, testicular and epididymal tissues of DOX-treated rats. The activities of antioxidant enzymes such as catalase, superoxide dismutase, glutathione S-transferase and glutathione peroxidase were upregulated in the DPDS co-treated group. DPDS co-treatment alleviates the burden of DOX-induced inflammation by significant reductions in myeloperoxidase activity, levels of nitric oxide and tumor necrosis factor alpha with concomitant decline in the activity of caspase-3, an apoptotic biomarker. Consequently, significant improvement in the spermiogram, levels of reproductive hormones (follicle stimulating hormone, luteinizing hormone, prolactin, serum testosterone and intra-testicular testosterone) levels in the DPDS co-treatment group in comparison to DOX alone-treated group were observed. Histology results of the testes and epididymis showed that DPDS significantly alleviated pathological lesions induced by DOX in the animals. CONCLUSION DPDS may modulate reproductive toxicity associated with DOX therapy in male cancer patients.
Collapse
Affiliation(s)
- Adesina A Babalola
- Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adedoyin R Adelowo
- Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oluwatobiloba F Da-Silva
- Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Cynthia N Ikeji
- Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olatunde Owoeye
- Department of Anatomy, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Joao B T Rocha
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Isaac A Adedara
- Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Ebenezer O Farombi
- Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
37
|
Mekhileri NV, Major G, Lim K, Mutreja I, Chitcholtan K, Phillips E, Hooper G, Woodfield T. Biofabrication of Modular Spheroids as Tumor-Scale Microenvironments for Drug Screening. Adv Healthc Mater 2023; 12:e2201581. [PMID: 36495232 PMCID: PMC11468982 DOI: 10.1002/adhm.202201581] [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: 06/30/2022] [Revised: 11/13/2022] [Indexed: 12/14/2022]
Abstract
To streamline the drug discovery pipeline, there is a pressing need for preclinical models which replicate the complexity and scale of native tumors. While there have been advancements in the formation of microscale tumor units, these models are cell-line dependent, time-consuming and have not improved clinical trial success rates. In this study, two methods for generating 3D tumor microenvironments are compared, rapidly fabricated hydrogel microspheres and traditional cell-dense spheroids. These modules are then bioassembled into 3D printed thermoplastic scaffolds, using an automated biofabrication process, to form tumor-scale models. Modules are formed with SKOV3 and HFF cells as monocultures and cocultures, and the fabrication efficiency, cell architecture, and drug response profiles are characterized, both as single modules and as multimodular constructs. Cell-encapsulated Gel-MA microspheres are fabricated with high-reproducibility and dimensions necessary for automated tumor-scale bioassembly regardless of cell type, however, only cocultured spheroids form compact modules suitable for bioassembly. Chemosensitivity assays demonstrate the reduced potency of doxorubicin in coculture bioassembled constructs and a ≈five-fold increase in drug resistance of cocultured cells in 3D modules compared with 2D monolayers. This bioassembly system is efficient and tailorable so that a variety of relevant-sized tumor constructs could be developed to study tumorigenesis and modernize drug discovery.
Collapse
Affiliation(s)
- Naveen Vijayan Mekhileri
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Gretel Major
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Khoon Lim
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Isha Mutreja
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and GynaecologyGynaecological Cancer Research GroupUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Elisabeth Phillips
- Mackenzie Cancer Research GroupDepartment of Pathology and Biomedical ScienceUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Gary Hooper
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Tim Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| |
Collapse
|
38
|
Liu X, Chen J, Chen W, Xu Y, Shen Y, Xu X. Targeting IGF2BP3 in Cancer. Int J Mol Sci 2023; 24:ijms24119423. [PMID: 37298373 DOI: 10.3390/ijms24119423] [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: 03/10/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
RNA-binding proteins (RBPs) can regulate multiple pathways by binding to RNAs, playing a variety of functions, such as localization, stability, and immunity. In recent years, with the development of technology, researchers have discovered that RBPs play a key role in the N6-methyladenosine (m6A) modification process. M6A methylation is the most abundant form of RNA modification in eukaryotes, which is defined as methylation on the sixth N atom of adenine in RNA. Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) is one of the components of m6A binding proteins, which plays an important role in decoding m6A marks and performing various biological functions. IGF2BP3 is abnormally expressed in many human cancers, often associated with poor prognosis. Here, we summarize the physiological role of IGF2BP3 in organisms and describe its role and mechanism in tumors. These data suggest that IGF2BP3 may be a valuable therapeutic target and prognostic marker in the future.
Collapse
Affiliation(s)
- Xin Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiayu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wenliang Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yangtao Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yang Shen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ximing Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
39
|
Liu J, Zhang Q, Wang C, Yang J, Yang S, Wang T, Wang B. Knockdown of BAP31 Overcomes Hepatocellular Carcinoma Doxorubicin Resistance through Downregulation of Survivin. Int J Mol Sci 2023; 24:ijms24087622. [PMID: 37108785 PMCID: PMC10142662 DOI: 10.3390/ijms24087622] [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: 03/16/2023] [Revised: 04/14/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
The expression of B-cell receptor associated protein 31 (BAP31) is increased in many tumor types, and it is reported to participate in proliferation, migration, and apoptosis. However, the relationship between BAP31 and chemoresistance is uncertain. This study investigated the role of BAP31 in regulating the doxorubicin (Dox) resistance of hepatocellular carcinoma (HCC). The expression of proteins was assessed by Western blotting. The correlation between BAP31 expression and Dox resistance was examined by MTT and colony formation assays. Apoptosis was analyzed by flow cytometry and TdT-mediated dUTP nick end labeling assays. Western blot and immunofluorescence analyses were performed in the knockdown cell lines to explore the possible mechanisms. In this study, BAP31 was strongly expressed, and knockdown of BAP31 increased Dox chemosensitivity in cancer cells. Furthermore, the expression of BAP31 was higher in the Dox-resistant HCC cells than that in their parental cells; knockdown of BAP31 reduced the half maximal inhibitory concentration value and overcame Dox resistance in Dox-resistant HCC cells. In HCC cells, knockdown of BAP31 increased Dox-induced apoptosis and enhanced Dox chemosensitivity in vitro and in vivo. The potential mechanism by which BAP31 increased Dox-induced apoptosis is that BAP31 inhibited survivin expression by promoting FoxO1 nucleus-cytoplasm translocation. Knockdown of BAP31 and survivin had a synergistic effect on Dox chemosensitivity by enhancing the apoptosis of HCC cells. These findings reveal that BAP31 knockdown enhances Dox chemosensitivity through the downregulation of survivin, suggesting that BAP31 is a potential therapeutic target for improving the treatment response of HCC with resistance to Dox.
Collapse
Affiliation(s)
- Jingjing Liu
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Qi Zhang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Changli Wang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Jiaying Yang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Sheng Yang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Tianyi Wang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Bing Wang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| |
Collapse
|
40
|
Antar SA, Abd-Elsalam M, Abdo W, Abdeen A, Abdo M, Fericean L, Raslan NA, Ibrahim SF, Sharif AF, Elalfy A, Nasr HE, Zaid AB, Atia R, Atwa AM, Gebba MA, Alzokaky AA. Modulatory Role of Autophagy in Metformin Therapeutic Activity toward Doxorubicin-Induced Nephrotoxicity. TOXICS 2023; 11:273. [PMID: 36977038 PMCID: PMC10052439 DOI: 10.3390/toxics11030273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 06/18/2023]
Abstract
Doxorubicin (DOX) is a frequent chemotherapeutic drug used to treat various malignant tumors. One of the key factors that diminish its therapeutic importance is DOX-induced nephrotoxicity. The first-line oral antidiabetic drug is metformin (Met), which also has antioxidant properties. The purpose of our study was to investigate the underlying molecular mechanisms for the potential protective effects of Met on DOX-triggered nephrotoxicity. Four animal groups were assigned as follows; animals received vehicle (control group), 200 mg/kg Met (Met group), DOX 15 mg/kg DOX (DOX group), and a combination of DOX and Met (DOX/Met group). Our results demonstrated that DOX administration caused marked histological alterations of widespread inflammation and tubular degeneration. Notably, the DOX-induced dramatic up-regulation of the nuclear factor-kappa B/P65 (NF-κB/P65), microtubule-associated protein light chain 3B (LC3B), neutrophil gelatinase-associated lipocalin (NGAL), interleukin-1beta (IL-1β), 8-hydroxy-2' -deoxyguanosine (8-OHdG), and Beclin-1 in renal tissue. A marked increase in the malondialdehyde (MDA) tissue level and a decrease in the total antioxidant capacity (TAC) were also recorded in DOX-exposed animals. Interestingly, Met could minimize all histopathological changes as well as the disruptions caused by DOX in the aforementioned measures. Thus, Met provided a workable method for suppressing the nephrotoxicity that occurred during the DOX regimen via the deactivation of the Beclin-1/LC3B pathway.
Collapse
Affiliation(s)
- Samar A. Antar
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA
| | - Marwa Abd-Elsalam
- Department of Histology, Faculty of Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Walied Abdo
- Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt
| | - Mohamed Abdo
- Department of Animal Histology and Anatomy, School of Veterinary Medicine, Badr University in Cairo (BUC), Badr City 32897, Egypt
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32897, Egypt
| | - Liana Fericean
- Department of Biology and Plant Protection, Faculty of Agriculture, University of Life Sciences “King Michael I” from Timișoara, Calea Aradului 119, CUI 3487181, 300645 Timisoara, Romania
| | - Nahed A. Raslan
- Clinical Pharmacy Program, College of Health Sciences and Nursing, Al-Rayan Colleges, Medina 42541, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11651, Egypt
| | - Samah F. Ibrahim
- Department of Clinical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Asmaa F. Sharif
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Tanta University, Tanta 31111, Egypt
| | - Amira Elalfy
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Hend E. Nasr
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Ahmed B. Zaid
- Department of Clinical Pathology, National Liver Institute, Menoufia University, Shibin Elkom 32511, Egypt
| | - Rania Atia
- Department of Physiology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
- Department of Basic Medical Science, Faculty of Applied Medical Science, Al-Baha University, Al-Baha 65779, Saudi Arabia
| | - Ahmed M. Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo 11829, Egypt
| | - Mohammed A. Gebba
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Amany A. Alzokaky
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11651, Egypt
| |
Collapse
|
41
|
Pavlik T, Gudkova V, Razvolyaeva D, Pavlova M, Kostukova N, Miloykovich L, Kolik L, Konchekov E, Shimanovskii N. The Role of Autophagy and Apoptosis in the Combined Action of Plasma-Treated Saline, Doxorubicin, and Medroxyprogesterone Acetate on K562 Myeloid Leukaemia Cells. Int J Mol Sci 2023; 24:ijms24065100. [PMID: 36982174 PMCID: PMC10049101 DOI: 10.3390/ijms24065100] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
The anti-cancer properties of plasma-treated solutions (PTS) and their interaction with drugs are one of the most popular topics in modern plasma medicine. Our research involved comparing the effects of four physiological saline solutions (0.9% NaCl, Ringer’s solution, Hank’s Balanced Salt Solution, Hank’s Balanced Salt Solution with amino acids added in concentrations observed in the human blood) treated with cold atmospheric plasma and studying the combined cytotoxic effect of PTS with doxorubicin and medroxyprogesterone acetate (MPA). Analysis of the effect of the studied agents on the formation of radicals in the incubation medium, the vitality of K562 myeloid leukaemia cells, and the processes of autophagy and apoptosis in them revealed two key findings. The first is that when using PTS and doxorubicin-containing PTS, autophagy is the predominant process in cancer cells. The second is that combining PTS with MPA enhances apoptotic processes. It was hypothesised that while autophagy is stimulated by the accumulation of reactive oxygen species in the cell, apoptosis is stimulated through specific cell progesterone receptors.
Collapse
Affiliation(s)
- Tatyana Pavlik
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991 Moscow, Russia
- Faculty of Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Correspondence:
| | - Victoria Gudkova
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991 Moscow, Russia
- Faculty of Science, Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Darya Razvolyaeva
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991 Moscow, Russia
- Faculty of Science, Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Marina Pavlova
- Faculty of Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Nadejda Kostukova
- Faculty of Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Lilia Miloykovich
- Faculty of Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Leonid Kolik
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Evgeny Konchekov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991 Moscow, Russia
| | - Nikolay Shimanovskii
- Faculty of Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
42
|
Kumar U, Castellanos-Uribe M, May ST, Yagüe E. Adaptive resistance is not responsible for long-term drug resistance in a cellular model of triple negative breast cancer. Gene 2023; 850:146930. [PMID: 36195266 DOI: 10.1016/j.gene.2022.146930] [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: 07/01/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 06/16/2023]
Abstract
Resistance to cancer therapeutics represents a leading cause of mortality and is particularly important in cancers, such as triple negative breast cancer, for which no targeted therapy is available, as these are only treated with traditional chemotherapeutics. Cancer, as well as bacterial, drug resistance can be intrinsic, acquired or adaptive. Adaptive cancer drug resistance is gaining attention as a mechanism for the generation of long-term drug resistance as is the case with bacterial antibiotic resistance. We have used a cellular model of triple negative breast cancer (CAL51) and its drug resistance derivative (CALDOX) to gain insight into genome-wide expression changes associated with long-term doxorubicin (a widely used anthracycline for cancer treatment) resistance and doxorubicin-induced stress. Previous work indicates that both naïve and resistance cells have a functional p53-p21 axis controlling cell cycle at G1, although this is not a driver for drug resistance, but down-regulation of TOP2A (topoisomerase IIα). As expected, CALDOX cells have a signature characterized, in addition to down-regulation of TOP2A, by genes and pathways associated with drug resistance, metastasis and stemness. Both CAL51 and CALDOX stress signatures share 12 common genes (TRIM22, FAS, SPATA18, SULF2, CDKN1A, GDF15, MYO6, CXCL5, CROT, EPPK1, ZMAT3 and CD44), with roles in the above-mentioned pathways, indicating that these cells have similar functional responses to doxorubicin relaying on the p53 control of apoptosis. Eight genes are shared by both drug stress signatures (in CAL51 and CALDOX cells) and CALDOX resistant cells (FAS, SULF2, CDKN1A, CXCL5, CD44, SPATA18, TRIM22 and CROT), many of them targets of p53. This corroborates experimental data indicating that CALDOX cells, even in the absence of drug, have activated, at least partially, the p53-p21 axis and DNA damage response. Although this eight-gene signature might be an indicator of adaptive resistance, as this transient phenomenon due to short-term stress may not revert to its original state upon withdrawal of the stressor, previous experimental data indicates that the p53-p21 axis is not responsible for doxorubicin resistance. Importantly, TOP2A is not responsive to doxorubicin treatment and thus absent in both drug stress signatures. This indicates that during the generation of doxorubicin resistance, cells acquire genetic changes likely to be random, leading to down regulation of TOP2A, but selected during the generation of cells due to the presence of drug in the culture medium. This poses a considerable constraint for the development of strategies aimed at avoiding the emergence of drug resistance in the clinic.
Collapse
Affiliation(s)
- Uttom Kumar
- Division of Cancer, Imperial College Faculty of Medicine, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Marcos Castellanos-Uribe
- Nottingham Arabidopsis Stock Centre, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, United Kingdom
| | - Sean T May
- Nottingham Arabidopsis Stock Centre, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, United Kingdom
| | - Ernesto Yagüe
- Division of Cancer, Imperial College Faculty of Medicine, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom.
| |
Collapse
|
43
|
Elmorsi RM, Kabel AM, El Saadany AA, Abou El-Seoud SH. The protective effects of topiramate and spirulina against doxorubicin-induced cardiotoxicity in rats. Hum Exp Toxicol 2023; 42:9603271231198624. [PMID: 37644674 DOI: 10.1177/09603271231198624] [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: 08/31/2023]
Abstract
Doxorubicin (DOX) is a widely used chemotherapy drug that can cause significant cardiotoxicity, limiting its clinical application. This study aimed to investigate the potential protective effects of topiramate (TPM) and spirulina (SP), either alone or in combination, in preventing DOX-induced cardiotoxicity. Adult Sprague Dawley rats were divided into five groups, including a normal control group and groups receiving DOX alone, DOX with TPM, DOX with SP, or DOX with a combination of TPM and SP. Cardiotoxicity was induced by administering DOX intraperitoneally at a cumulative dose of 16 mg/kg over 4 weeks. TPM and/or SP administration started 1 week before DOX treatment and continued for 35 days. Body weight, serum markers of cardiac damage, oxidative stress and inflammatory parameters were assessed. Histopathological and immunohistochemical examinations were performed on cardiac tissues. Results showed that TPM and SP monotherapy led to significant improvements in serum levels of cardiac markers, decreased oxidative stress, reduced fibrosis-related growth factor levels, increased antioxidant levels, and improved histopathological features. SP demonstrated more prominent effects in comparison to TPM, and the combination of TPM and SP exhibited even more pronounced effects. In conclusion, TPM and SP, either alone or in combination, hold promise as therapeutic interventions for mitigating DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Radwa M Elmorsi
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ahmed M Kabel
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Amira A El Saadany
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | |
Collapse
|
44
|
Combination of Spirulina platensis, Ganoderma lucidum and Moringa oleifera Improves Cardiac Functions and Reduces Pro-Inflammatory Biomarkers in Preclinical Models of Short-Term Doxorubicin-Mediated Cardiotoxicity: New Frontiers in Cardioncology? J Cardiovasc Dev Dis 2022; 9:jcdd9120423. [PMID: 36547420 PMCID: PMC9780956 DOI: 10.3390/jcdd9120423] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022] Open
Abstract
Anthracyclines are essential adjuvant therapies for a variety of cancers, particularly breast, gastric and esophageal cancers. Whilst prolonging cancer-related survival, these agents can induce drug-related cardiotoxicity. Spirulina, Reishi (Ganoderma lucidum) and Moringa are three nutraceuticals with anti-inflammatory effects that are currently used in cancer patients as complementary and alternative medicines to improve quality of life and fatigue. We hypothesize that the nutraceutical combination of Spirulina, Reishi and Moringa (Singo) could reduce inflammation and cardiotoxicity induced by anthracyclines. Female C57Bl/6 mice were untreated (Sham, n = 6) or treated for 7 days with short-term doxorubicin (DOXO, n = 6) or Singo (Singo, n = 6), or pre-treated with Singo for 3 days and associated with DOXO for remaining 7 days (DOXO−Singo, n = 6). The ejection fraction and radial and longitudinal strain were analyzed through transthoracic echocardiography (Vevo 2100, Fujifilm, Tokyo, Japan). The myocardial expressions of NLRP3, DAMPs (galectin-3 and calgranulin S100) and 13 cytokines were quantified through selective mouse ELISA methods. Myocardial fibrosis, necrosis and hypertrophy were analyzed through immunohistochemistry (IHC). Human cardiomyocytes were exposed to DOXO (200 nM) alone or in combination with Singo (at 10, 25 and 50 µg/mL) for 24 and 48 h. Cell viability and inflammation studies were also performed. In preclinical models, Singo significantly improved ejection fraction and fractional shortening. Reduced expressions of myocardial NLRP3 and NF-kB levels in cardiac tissues were seen in DOXO−Singo mice vs. DOXO (p < 0.05). The myocardial levels of calgranulin S100 and galectin-3 were strongly reduced in DOXO−Singo mice vs. DOXO (p < 0.05). Immunohistochemistry analysis indicates that Singo reduces fibrosis and hypertrophy in the myocardial tissues of mice during exposure to DOXO. In conclusion, in the preclinical model of DOXO-induced cardiotoxicity, Singo is able to improve cardiac function and reduce biomarkers involved in heart failure and fibrosis.
Collapse
|
45
|
A nanodiamond chemotherapeutic folate receptor-targeting prodrug with triggerable drug release. Int J Pharm 2022; 630:122432. [PMID: 36435503 DOI: 10.1016/j.ijpharm.2022.122432] [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: 08/31/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/26/2022]
Abstract
Cancer chemotherapy is often accompanied by severe off-target effects that both damage quality of life and can decrease therapeutic compliance. This could be minimized through selective delivery of cytotoxic agents directly to the cancer cells. This would decrease the drug dose, consequently minimizing side effects and cost. With this goal in mind, a dual-gated folate-functionalized nanodiamond drug delivery system (NPFSSD) for doxorubicin with activatable fluorescence and cytotoxicity has been prepared. Both the cytotoxic activity and the fluorescence of doxorubicin (DOX) are quenched when it is covalently immobilized on the nanodiamond. The NPFSSD is preferentially uptaken by cancer cells overexpressing the folate receptor. Then, once inside a cell, the drug is preferentially released within tumor cells due to their high levels of endogenous of glutathione, required for releasing DOX through cleavage of a disulfide linker. Interestingly, once free DOX is loaded onto the nanodiamond, it can also evade resistance mechanisms that use protein pumps to remove drugs from the cytoplasm. This nanodrug, used in an in vivo model with local injection of drugs, effectively inhibits tumor growth with fewer side effects than direct injection of free DOX, providing a potentially powerful platform to improve therapeutic outcomes.
Collapse
|
46
|
Marocco L, Umrath F, Sachsenmaier S, Rabiner R, Wülker N, Danalache M. 5-Aminolevulinic Acid-Mediated Photodynamic Therapy Potentiates the Effectiveness of Doxorubicin in Ewing Sarcomas. Biomedicines 2022; 10:biomedicines10112900. [PMID: 36428464 PMCID: PMC9687703 DOI: 10.3390/biomedicines10112900] [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: 10/19/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Ewing sarcomas (ES) are aggressive primary bone tumors that require radical therapy. Promising low toxicity, 5-aminolevulinic acid (5-ALA)-mediated photodynamic therapy (PDT) could enhance the effectiveness of conventional treatment modalities (e.g., doxorubicin (DOX)), improving, thus, the anti-tumorigenic effects. In this study, we investigated the effects of DOX and 5-ALA PDT alone or in combination on three different human ES cell lines. Cell viability, reactive oxygen species (ROS) production, and cellular stiffness were measured 24 h after PDT (blue light-wavelength 436 nm with 5-ALA) with or without DOX. ES cell lines have a different sensitivity to the same doses and exposure of 5-ALA PDT. DOX in combination with 5-ALA PDT was found to be effective in impairing the viability of all ES cells while also increasing cytotoxic activity by high ROS production. The stiffness of the ES cells increased significantly (p < 0.05) post treatment. Overall, our results showed that across multiple ES cell lines, 5-ALA PDT can successfully and safely be combined with DOX to potentiate the therapeutic effect. The 5-ALA PDT has the potential to be a highly effective treatment when used alone or in conjunction with other treatments. More research is needed to assess the effectiveness of 5-ALA PDT in in vivo settings.
Collapse
Affiliation(s)
- Lea Marocco
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, 72072 Tübingen, Germany
- Correspondence:
| | - Felix Umrath
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, 72072 Tübingen, Germany
- Department of Oral and Maxillofacial Surgery, University Hospital of Tübingen, 72076 Tübingen, Germany
| | - Saskia Sachsenmaier
- Department of Orthopaedic Surgery, University Hospital of Tübingen, 72076 Tübingen, Germany
| | | | - Nikolaus Wülker
- Department of Orthopaedic Surgery, University Hospital of Tübingen, 72076 Tübingen, Germany
| | - Marina Danalache
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, 72072 Tübingen, Germany
| |
Collapse
|
47
|
Cao T, Liu Y, Jia Q, Wang X, Zhang S, Yu K, Zhou J. Rare earth fluorescent nanoprobes with minimal side effects enable tumor microenvironment activation for chemotherapy. J RARE EARTH 2022. [DOI: 10.1016/j.jre.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
48
|
Metabolomic Signatures in Doxorubicin-Induced Metabolites Characterization, Metabolic Inhibition, and Signaling Pathway Mechanisms in Colon Cancer HCT116 Cells. Metabolites 2022; 12:metabo12111047. [DOI: 10.3390/metabo12111047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/19/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent is used for various cancer cells. To characterize the chemical structural components and metabolic inhibition, we applied a DOX to HCT116 colon cancer cells using an independent metabolites profiling approach. Chemical metabolomics has been involved in the new drug delivery systems. Metabolomics profiling of DOX-applied HCT116 colon cancer cellular metabolisms is rare. We used 1H nuclear magnetic resonance (NMR) spectroscopy in this study to clarify how DOX exposure affected HCT116 colon cancer cells. Metabolomics profiling in HCT116 cells detects 50 metabolites. Tracking metabolites can reveal pathway activities. HCT116 colon cancer cells were evenly treated with different concentrations of DOX for 24 h. The endogenous metabolites were identified by comparison with healthy cells. We found that acetate, glucose, glutamate, glutamine, sn-glycero-3-phosphocholine, valine, methionine, and isoleucine were increased. Metabolic expression of alanine, choline, fumarate, taurine, o-phosphocholine, inosine, lysine, and phenylalanine was decreased in HCT116 cancer cells. The metabolic phenotypic expression is markedly altered during a high dose of DOX. It is the first time that there is a metabolite pool and phenotypic expression in colon cancer cells. Targeting the DOX-metabolite axis may be a novel strategy for improving the curative effect of DOX-based therapy for colon cancer cells. These methods facilitate the routine metabolomic analysis of cancer cells.
Collapse
|
49
|
Mirzayans R, Murray D. What Are the Reasons for Continuing Failures in Cancer Therapy? Are Misleading/Inappropriate Preclinical Assays to Be Blamed? Might Some Modern Therapies Cause More Harm than Benefit? Int J Mol Sci 2022; 23:13217. [PMID: 36362004 PMCID: PMC9655591 DOI: 10.3390/ijms232113217] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 07/30/2023] Open
Abstract
Over 50 years of cancer research has resulted in the generation of massive amounts of information, but relatively little progress has been made in the treatment of patients with solid tumors, except for extending their survival for a few months at best. Here, we will briefly discuss some of the reasons for this failure, focusing on the limitations and sometimes misunderstanding of the clinical relevance of preclinical assays that are widely used to identify novel anticancer drugs and treatment strategies (e.g., "synthetic lethality"). These include colony formation, apoptosis (e.g., caspase-3 activation), immunoblotting, and high-content multiwell plate cell-based assays, as well as tumor growth studies in animal models. A major limitation is that such assays are rarely designed to recapitulate the tumor repopulating properties associated with therapy-induced cancer cell dormancy (durable proliferation arrest) reflecting, for example, premature senescence, polyploidy and/or multinucleation. Furthermore, pro-survival properties of apoptotic cancer cells through phoenix rising, failed apoptosis, and/or anastasis (return from the brink of death), as well as cancer immunoediting and the impact of therapeutic agents on interactions between cancer and immune cells are often overlooked in preclinical studies. A brief review of the history of cancer research makes one wonder if modern strategies for treating patients with solid tumors may sometimes cause more harm than benefit.
Collapse
|
50
|
On the Reaction of 2-Alkanoylnaphthohydroquinones with Hydroxylamine: Access to Cytotoxic 2-(Hydroxyamino)-1,4-naphthoquinone and Their 3-(Hydroxyimino)alkyl Analogous. J CHEM-NY 2022. [DOI: 10.1155/2022/7664037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Oximes are known for their anti-inflammatory, antimicrobial, antioxidant, and anticancer activities. Frequently, modification of biologically active carbonyl compounds into oximes leads to increased activity. The present study reports the reactivity of 2-alkanoylnaphthohydroquinones against hydroxylamine under aerial conditions. Results show that, depending on the structure of the hydroquinones, the reaction proceeds through two different chemical pathways to produce 2-(hydroxyamino)-1,4-naphthoquinone and their C-3 (hydroxyimino)alkyl derivatives. Both the formation of the quinoid compounds under aerial oxidation and C-C cleavage reactions of hemiaminal intermediates are discussed. In vitro screening of the substituted 1,4-naphthoquinones on a panel of cancer cells reveals moderate cytotoxic activities. Compound 19, 2-(hydroxyamino)-1,4-naphthoquinone, stands out by its anticancer potency against prostate cancer cells as shown by the lowest IC50 value (8.08 μM) and the best selectivity index (3.90).
Collapse
|