1
|
Finnigan D, Hajjaj OI, Othman M. Red blood cell changes due to cancer and cancer treatments: a narrative review. Curr Opin Hematol 2025; 32:93-103. [PMID: 39787019 DOI: 10.1097/moh.0000000000000859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
PURPOSE OF REVIEW To date, there is relatively limited research investigating changes in red blood cells (RBCs), particularly qualitative changes, in cancer patients and cancer patients receiving treatment. These changes may be important in better understanding cancer-associated anemia, which is the most prevalent hematological disorder in cancer patients with wide-ranging implications on patient care and quality of life. This review aims to summarize available evidence regarding qualitative and quantitative changes in RBCs in individuals with cancer prior to treatment and in patients undergoing treatment. RECENT FINDINGS The most commonly reported changes in RBCs in cancer patients were increased mean corpuscular volume (MCV) and decreased hemoglobin, RBC count, and hematocrit. There were increased lipid peroxidation products and decreased antioxidants. There were increased polyunsaturated fatty acids (PUFAs) and decreased monounsaturated fatty acids (MUFAs) and saturated fatty acids (FAs). Additionally, RBC shape alterations with various atypical morphologies, membrane structure abnormalities, and impaired fluidity were also reported. These and various other reported findings are discussed in depth. SUMMARY There are several reported quantitative and qualitative RBC changes in individuals with cancer, with some studies exhibiting conflicting results. Further research is needed to solidify the data and to better understand hematological-associated comorbidities in those patients.
Collapse
Affiliation(s)
- Deirdre Finnigan
- Department of Biomedical and Molecular Sciences, Queen's University
| | | | - Maha Othman
- Department of Biomedical and Molecular Sciences, Queen's University
- School of Baccalaureate Nursing, St. Lawrence College, Kingston, Ontario, Canada
- Clinical Pathology Department, Faculty of Medicine, Mansoura, Egypt
| |
Collapse
|
2
|
Bolliger M, Wasinger D, Brunmair J, Hagn G, Wolf M, Preindl K, Reiter B, Bileck A, Gerner C, Fitzal F, Meier-Menches SM. Mass spectrometry-based analysis of eccrine sweat supports predictive, preventive and personalised medicine in a cohort of breast cancer patients in Austria. EPMA J 2025; 16:165-182. [PMID: 39991101 PMCID: PMC11842658 DOI: 10.1007/s13167-025-00396-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/07/2025] [Indexed: 02/25/2025]
Abstract
Objective Metabolomics measurements of eccrine sweat may provide novel and relevant biomedical information to support predictive, preventive and personalised medicine (3PM). However, only limited data is available regarding metabolic alterations accompanying chemotherapy of breast cancer patients related to residual cancer burden (RCB) or therapy response. Here, we have applied Metabo-Tip, a non-invasive metabolomics assay based on the analysis of eccrine sweat from the fingertips, to investigate the feasibility of such an approach, especially with respect to drug monitoring, assessing lifestyle parameters and stratification of breast cancer patients. Methods Eccrine sweat samples were collected from breast cancer patients (n = 9) during the first cycle of neoadjuvant chemotherapy at four time points in this proof-of-concept study at a Tertiary University Hospital. Metabolites in eccrine sweat were analysed using mass spectrometry. Blood plasma samples from the same timepoints were also collected and analysed using a validated targeted metabolomics kit, in addition to proteomics and fatty acids/oxylipin analysis. Results A total of 247 exogenous small molecules and endogenous metabolites were identified in eccrine sweat of the breast cancer patients. Cyclophosphamide and ondansetron were successfully detected and monitored in eccrine sweat of individual patients and accurately reflected the administration schedule. The non-essential amino acids asparagine, serine and proline, as well as ornithine were significantly regulated in eccrine sweat and blood plasma over the therapy cycle. However, their distinct time-dependent profiles indicated compartment-specific distributions. Indeed, the metabolite composition of eccrine sweat seems to largely resemble the composition of the interstitial fluid. Plasma proteins and fatty acids/oxylipins were not affected by the first treatment cycle. Individual smoking habit was revealed by the simultaneous detection of nicotine and its primary metabolite cotinine in eccrine sweat. Stratification according to RCB revealed pronounced differences in the metabolic composition of eccrine sweat in these patients at baseline, e.g., essential amino acids, possibly due to the systemic contribution of breast cancer and its impact on metabolic turnover. Conclusion Mass spectrometry-based analysis of metabolites from eccrine sweat of breast cancer patients successfully qualified lifestyle parameters for risk assessment and allowed us to monitor drug treatment and systemic response to therapy. Moreover, eccrine sweat revealed a potentially predictive metabolic pattern stratifying patients by the extent of the metabolic activity of breast cancer tissue at baseline. Eccrine sweat is derived from the otherwise hardly accessible interstitial fluid and, thus, opens up a new dimension for biomonitoring of breast cancer in secondary and tertiary care. The simple sample collection without the need for trained personnel could also enable decentralised long-term biomonitoring to assess stable disease or disease progression. Eccrine sweat analysis may indeed significantly advance 3PM for the benefit of breast cancer patients. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-025-00396-6.
Collapse
Affiliation(s)
- Michael Bolliger
- Department of General Surgery (Division of Visceral Surgery), Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Department of Surgery, St. Francis Hospital, Nikolsdorfergasse 32, 1050 Vienna, Austria
| | - Daniel Wasinger
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Waehringer Str. 38-42, 1090 Vienna, Austria
| | - Julia Brunmair
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Gerhard Hagn
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Waehringer Str. 38-42, 1090 Vienna, Austria
| | - Michael Wolf
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, Waehringer Str. 38-42, 1090 Vienna, Austria
| | - Karin Preindl
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18–20, Vienna, 1090 Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Birgit Reiter
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18–20, Vienna, 1090 Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Andrea Bileck
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Christopher Gerner
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| | - Florian Fitzal
- Department of Surgery and Vascular Surgery, Hanusch Hospital, Heinrich-Collin-Str. 30, 1140 Vienna, Austria
| | - Samuel M. Meier-Menches
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Waehringer Str. 38, 1090 Vienna, Austria
- Faculty of Chemistry, Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 38, 1090 Vienna, Austria
| |
Collapse
|
3
|
Moomivand S, Nikbakht M, Majd A, Bikhof Torbati M, Mousavi SA. Combining Chemotherapy Agents and Autophagy Modulators for Enhanced Breast Cancer Cell Death. Adv Pharm Bull 2024; 14:908-917. [PMID: 40190668 PMCID: PMC11970493 DOI: 10.34172/apb.42733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 04/09/2025] Open
Abstract
Purpose Autophagy, governed by genes with dual roles in cell death and survival, plays a crucial role in cancer persistence. Arsenic trioxide (ATO), carboplatin (CP), and cyclophosphamide (CY) are used to treat various cancers. ATO impedes cell proliferation and triggers apoptosis in cancer cells. CP, a platinum-based drug, damages cancer cell DNA, while CY acts as an alkylating agent, disrupting cell proliferation. This study investigates the combined effects of ATO, CP, and CY on inducing apoptosis and modulating autophagy in triple-negative breast cancer (TNBC) cell lines, BT-20 and MDA-MB-231. Methods The cytotoxic effects of ATO, CP, and CY, alone and in combination, were evaluated using the MTT assay on BT-20 and MDA-MB-231 cells. Apoptosis and cell cycle progression were analyzed by annexin-V FITC/PI staining and flow cytometry. Gene expression of autophagy-and apoptosis-related markers, including Beclin 1, LC3, caspase 3, and BCL2, was quantified using RT-PCR. Data were analyzed using GraphPad Prism 4.0 with one-way ANOVA followed by Dunnett's test. Results The combination of ATO, CP, and CY significantly reduced cell viability and enhanced apoptosis, evidenced by increased caspase-3 activity and reduced BCL2 expression. Cell cycle arrest in the G1 phase was observed, alongside elevated autophagy markers Beclin 1 and LC3. Conclusion The combination of ATO, CP, and CY induces synergistic effects in promoting apoptosis and autophagy in TNBC cell lines. These findings suggest that this combination therapy could be a promising approach to enhancing treatment efficacy in aggressive breast cancers, offering new insights into potential therapeutic strategies.
Collapse
Affiliation(s)
- Soraya Moomivand
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
- Research Institute for Oncology, Hematology and Cell Therapy Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Nikbakht
- Research Institute for Oncology, Hematology and Cell Therapy Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | - Ahmad Majd
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Bikhof Torbati
- Department of Biology, Yadegar-e-Imam Khomeini (RAH) Shahre rey branch, Islamic Azad University, Tehran, Iran
| | - Seyed Asadoullah Mousavi
- Research Institute for Oncology, Hematology and Cell Therapy Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Alghareeb SA, Alsughayyir J, Alfhili MA. Ginsenoside Rh2 Regulates the Calcium/ROS/CK1α/MLKL Pathway to Promote Premature Eryptosis and Hemolysis in Red Blood Cells. Toxicol Pathol 2024; 52:284-294. [PMID: 39148410 DOI: 10.1177/01926233241268846] [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: 08/17/2024]
Abstract
Ginsenoside Rh2 (GRh2) exhibits significant potential as an anticancer agent; however, progress in developing chemotherapeutic drugs is impeded by their toxicity toward off-target tissues. Specifically, anemia caused by chemotherapy is a debilitating side effect and can be caused by red blood cell (RBC) hemolysis and eryptosis. Cells were exposed to GRh2 in the antitumor range and hemolytic and eryptotic markers were examined under different experimental conditions using photometric and cytofluorimetric methods. GRh2 caused Ca2+-independent, concentration-responsive hemolysis in addition to disrupted ion trafficking with K+ and Cl- leakage. Significant increases in cells positive for annexin-V-fluorescein isothiocyanate, Fluo4, and 2,7-dichlorofluorescein were noted upon GRh2 treatment coupled with a decrease in forward scatter and acetylcholinesterase activity. Importantly, the cytotoxic effects of GRh2 were mitigated by ascorbic acid and by blocking casein kinase 1α (CK1α) and mixed lineage kinase domain-like (MLKL) signaling. In contrast, Ca2+ omission, inhibition of KCl efflux, and isosmotic sucrose aggravated GRh2-induced RBC death. In whole blood, GRh2 selectively targeted reticulocytes and lymphocytes. Altogether, this study identified novel mechanisms underlying GRh2-induced RBC death involving Ca2+ buildup, loss of membrane phospholipid asymmetry and cellular volume, anticholinesterase activity, and oxidative stress. These findings shed light on the hematologic toxicity of GRh2 which is crucial for optimizing its utilization in cancer treatment.
Collapse
Affiliation(s)
- Sumiah A Alghareeb
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Jawaher Alsughayyir
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad A Alfhili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Moerland JA, Liby KT. The Triterpenoid CDDO-Methyl Ester Reduces Tumor Burden, Reprograms the Immune Microenvironment, and Protects from Chemotherapy-Induced Toxicity in a Preclinical Mouse Model of Established Lung Cancer. Antioxidants (Basel) 2024; 13:621. [PMID: 38929060 PMCID: PMC11201246 DOI: 10.3390/antiox13060621] [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: 04/04/2024] [Revised: 05/17/2024] [Accepted: 05/18/2024] [Indexed: 06/28/2024] Open
Abstract
NRF2 activation protects epithelial cells from malignancy, but cancer cells can upregulate the pathway to promote survival. NRF2 activators including CDDO-Methyl ester (CDDO-Me) inhibit cancer in preclinical models, suggesting NRF2 activation in other cell types may promote anti-tumor activity. However, the immunomodulatory effects of NRF2 activation remain poorly understood in the context of cancer. To test CDDO-Me in a murine model of established lung cancer, tumor-bearing wildtype (WT) and Nrf2 knockout (KO) mice were treated with 50-100 mg CDDO-Me/kg diet, alone or combined with carboplatin/paclitaxel (C/P) for 8-12 weeks. CDDO-Me decreased tumor burden in an Nrf2-dependent manner. The combination of CDDO-Me plus C/P was significantly (p < 0.05) more effective than either drug alone, reducing tumor burden by 84% in WT mice. CDDO-Me reduced the histopathological grade of WT tumors, with a significantly (p < 0.05) higher proportion of low-grade tumors and a lower proportion of high-grade tumors. These changes were augmented by combination with C/P. CDDO-Me also protected WT mice from C/P-induced toxicity and improved macrophage and T cell phenotypes in WT mice, reducing the expression of CD206 and PD-L1 on macrophages, decreasing immunosuppressive FoxP3+ CD4+ T cells, and increasing activation of CD8+ T cells in a Nrf2-dependent manner.
Collapse
Affiliation(s)
- Jessica A. Moerland
- Department of Pharmacology and Toxicology, Michigan State University, 1355 Bogue Street, East Lansing, MI 48824, USA;
| | - Karen T. Liby
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA
| |
Collapse
|
6
|
Mikhailova DM, Skverchinskaya E, Sudnitsyna J, Butov KR, Koltsova EM, Mindukshev IV, Gambaryan S. Hematin- and Hemin-Induced Spherization and Hemolysis of Human Erythrocytes Are Independent of Extracellular Calcium Concentration. Cells 2024; 13:554. [PMID: 38534398 PMCID: PMC10969559 DOI: 10.3390/cells13060554] [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: 02/12/2024] [Revised: 03/10/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
Pathologies such as malaria, hemorrhagic stroke, sickle cell disease, and thalassemia are characterized by the release of hemoglobin degradation products from damaged RBCs. Hematin (liganded with OH-) and hemin (liganded with Cl-)-are the oxidized forms of heme with toxic properties due to their hydrophobicity and the presence of redox-active Fe3. In the present study, using the original LaSca-TM laser particle analyzer, flow cytometry, and confocal microscopy, we showed that both hematin and hemin induce dose-dependent RBC spherization and hemolysis with ghost formation. Hematin and hemin at nanomolar concentrations increased [Ca2+]i in RBC; however, spherization and hemolysis occurred in the presence and absence of calcium, indicating that both processes are independent of [Ca2+]i. Both compounds triggered acute phosphatidylserine exposure on the membrane surface, reversible after 60 min of incubation. A comparison of hematin and hemin effects on RBCs revealed that hematin is a more reactive toxic metabolite than hemin towards human RBCs. The toxic effects of heme derivatives were reduced and even reversed in the presence of albumin, indicating the presence in RBCs of the own recovery system against the toxic effects of heme derivatives.
Collapse
Affiliation(s)
- Diana M. Mikhailova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Ave., 194223 Saint Petersburg, Russia; (D.M.M.); (E.S.); (J.S.); (I.V.M.)
- Department of Cytology and Histology, Saint Petersburg State University, 7/9 Universitetskaya Emb., 199034 Saint Petersburg, Russia
| | - Elisaveta Skverchinskaya
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Ave., 194223 Saint Petersburg, Russia; (D.M.M.); (E.S.); (J.S.); (I.V.M.)
| | - Julia Sudnitsyna
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Ave., 194223 Saint Petersburg, Russia; (D.M.M.); (E.S.); (J.S.); (I.V.M.)
| | - Kirill R. Butov
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia;
| | - Ekaterina M. Koltsova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia;
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya st., 109029 Moscow, Russia
| | - Igor V. Mindukshev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Ave., 194223 Saint Petersburg, Russia; (D.M.M.); (E.S.); (J.S.); (I.V.M.)
| | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Ave., 194223 Saint Petersburg, Russia; (D.M.M.); (E.S.); (J.S.); (I.V.M.)
| |
Collapse
|
7
|
Lee J, Zhang W, Nguyen D, Zhou L, Amengual J, Zhai J, Cote T, Landolina M, Ahmadi E, Sands I, Mishra N, Yu H, Nieh MP, Wang K, Li Y, Chen Y. Computation-aided Design of Rod-Shaped Janus Base Nanopieces for Improved Tissue Penetration and Therapeutics Delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.577046. [PMID: 38328235 PMCID: PMC10849704 DOI: 10.1101/2024.01.24.577046] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Despite the development of various drug delivery technologies, there remains a significant need for vehicles that can improve targeting and biodistribution in "hard-to-penetrate" tissues. Some solid tumors, for example, are particularly challenging to penetrate due to their dense extracellular matrix (ECM). In this study, we have formulated a new family of rod-shaped delivery vehicles named Janus base nanopieces (Rod JBNps), which are more slender than conventional spherical nanoparticles, such as lipid nanoparticles (LNPs). These JBNp nanorods are formed by bundles of DNA-inspired Janus base nanotubes (JBNts) with intercalated delivery cargoes. To develop this novel family of delivery vehicles, we employed a computation-aided design (CAD) methodology that includes molecular dynamics and response surface methodology. This approach precisely and efficiently guides experimental designs. Using an ovarian cancer model, we demonstrated that JBNps markedly improve penetration into the dense ECM of solid tumors, leading to better treatment outcomes compared to FDA-approved spherical LNP delivery. This study not only successfully developed a rod-shaped delivery vehicle for improved tissue penetration but also established a CAD methodology to effectively guide material design.
Collapse
|
8
|
Katolkar UN, Surana SJ. Exploring the Potential Role of Phytopharmaceuticals in Alleviating Toxicities of Chemotherapeutic Agents. Curr Protein Pept Sci 2024; 25:753-779. [PMID: 38919003 DOI: 10.2174/0113892037307940240606075208] [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: 02/13/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Chemotherapy is the mainstay of cancer treatment, bringing patients optimism about recurrence and survival. However, the clinical effectiveness of chemotherapeutic drugs is frequently jeopardized by their intrinsic toxicity, resulting in side effects affecting the quality of life of cancer patients. This analysis explores the ethnopharmacological impact of phytopharmaceuticals, highlighting their traditional use in many cultures. The present study, which takes its cues from indigenous knowledge, aims to close the knowledge gap between traditional medicine and modern medicine in reducing the toxicities of chemotherapy treatments. AIM The present in-depth study aims to highlight the current research and upcoming developments in phytopharmaceuticals for reducing the toxicity of chemotherapeutic drugs. Further, we address the mechanisms through which phytopharmaceuticals may reduce chemotherapy-induced side effects that include nausea, vomiting, myelosuppression, nephropathy, neuropathy, and cardiotoxicity using data from a variety of preclinical and clinical investigations. MATERIALS AND METHODS The literature search was carried out by employing search engines such as PubMed and Google Scholar with keywords such as cancer, chemotherapy, CNS toxicity, hematopoietic toxicity, renal toxicity, GI toxicity, CNS toxicity, and phytopharmaceuticals. RESULTS Bioactive chemicals found in plants, such as fruits, vegetables, herbs, and spices, are being studied for their capacity to improve the safety and acceptability of chemotherapy regimens. The current review also dives into the investigation of phytopharmaceuticals as adjuvant medicines in cancer treatment, which is a viable path for addressing the pressing need to lessen chemotherapy-induced toxicities. CONCLUSION The present review revealed that the potential of phytopharmaceuticals in alleviating chemotherapeutic drug toxicities would pave the way for better cancer treatment and patient outcomes, harmonizing with the larger trend towards personalized and holistic approaches to chemotherapy.
Collapse
Affiliation(s)
- Ujwal N Katolkar
- Department of Pharmacology, R.C. Patel Institute of Pharmaceutical Education and Research, Karwand Naka, Shirpur Dist. Dhule Maharashtra 425405, India
| | - Sanjay J Surana
- Department of Pharmacology, R.C. Patel Institute of Pharmaceutical Education and Research, Karwand Naka, Shirpur Dist. Dhule Maharashtra 425405, India
| |
Collapse
|
9
|
Santulli G. Translational Aspects of Cardiovascular Biology: From Bench to Bedside. BIOLOGY 2023; 12:biology12050658. [PMID: 37237472 DOI: 10.3390/biology12050658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide, and the search for novel mechanisms and therapeutics is desperately needed [...].
Collapse
Affiliation(s)
- Gaetano Santulli
- Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, 80131 Naples, Italy
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|